Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 6.744
Filtrer
1.
Invest Ophthalmol Vis Sci ; 65(8): 11, 2024 Jul 01.
Article de Anglais | MEDLINE | ID: mdl-38967943

RÉSUMÉ

Purpose: Ocular melanoma is a common primary malignant ocular tumor in adults with limited effective treatments. Epigenetic regulation plays an important role in tumor development. The switching/sucrose nonfermentation (SWI/SNF) chromatin remodeling complex and bromodomain and extraterminal domain family proteins are epigenetic regulators involved in several cancers. We aimed to screen a candidate small molecule inhibitor targeting these regulators and investigate its effect and mechanism in ocular melanoma. Methods: We observed phenotypes caused by knockdown of the corresponding gene and synergistic effects with BRD inhibitor treatment and SWI/SNF complex knockdown. The effect of JQ-1 on ocular melanoma cell cycle and apoptosis was analyzed with flow cytometry. Via RNA sequencing, we also explored the mechanism of BRD4. Results: The best tumor inhibitory effect was observed for the BRD4 inhibitor (JQ-1), although there were no statistically obvious changes in the shBRD4 and shBRD9 groups. Interestingly, the inhibitory effect of JQ-1 was decrease in the shBRD4 group. JQ-1 inhibits the growth of melanoma in various cell lines and in tumor-bearing mice. We found 17 of these 28 common differentially expressed genes were downregulated after MEL270 and MEL290 cells treated with JQ-1. Four of these 17 genes, TP53I11, SH2D5, SEMA5A, and MDGA1, were positively correlated with BRD4. In TCGA database, low expression of TP53I11, SH2D5, SEMA5A, and MDGA1 improved the overall survival rate of patients. Furthermore, the disease-free survival rate was increased in the groups with low expression of TP53I11, SH2D5, and SEMA5A. Conclusions: JQ-1 may act downstream of BRD4 and suppress ocular melanoma growth by inducing G1 cell cycle arrest.


Sujet(s)
Apoptose , Azépines , Points de contrôle du cycle cellulaire , Protéines du cycle cellulaire , Mélanome , Facteurs de transcription , Triazoles , Animaux , Mélanome/traitement médicamenteux , Mélanome/génétique , Mélanome/anatomopathologie , Mélanome/métabolisme , Souris , Humains , Facteurs de transcription/génétique , Facteurs de transcription/métabolisme , Azépines/pharmacologie , Triazoles/pharmacologie , Triazoles/usage thérapeutique , Points de contrôle du cycle cellulaire/effets des médicaments et des substances chimiques , Apoptose/effets des médicaments et des substances chimiques , Protéines du cycle cellulaire/génétique , Protéines du cycle cellulaire/métabolisme , Lignée cellulaire tumorale , Prolifération cellulaire/effets des médicaments et des substances chimiques , Régulation de l'expression des gènes tumoraux , Tumeurs de l'uvée/traitement médicamenteux , Tumeurs de l'uvée/génétique , Tumeurs de l'uvée/anatomopathologie , Tumeurs de l'uvée/métabolisme , Cytométrie en flux , Tests d'activité antitumorale sur modèle de xénogreffe , Souris nude , Protéines contenant un bromodomaine
2.
Sci Rep ; 14(1): 15406, 2024 07 04.
Article de Anglais | MEDLINE | ID: mdl-38965397

RÉSUMÉ

Patients with multiple myeloma (MM) experience relapse and drug resistance; therefore, novel treatments are essential. Clotrimazole (CTZ) is a wide-spectrum antifungal drug with antitumor activity. However, CTZ's effects on MM are unclear. We investigated CTZ's effect on MM cell proliferation and apoptosis induction mechanisms. CTZ's effects on MM.1S, NCI- H929, KMS-11, and U266 cell growth were investigated using Cell Counting Kit-8 (CCK-8) assay. The apoptotic cell percentage was quantified with annexin V-fluorescein isothiocyanate/7-amino actinomycin D staining. Mitochondrial membrane potential (MMP) and cell cycle progression were evaluated. Reactive oxygen species (ROS) levels were measured via fluorescence microscopy. Expression of apoptosis-related and nuclear factor (NF)-κB signaling proteins was analyzed using western blotting. The CCK-8 assay indicated that CTZ inhibited cell proliferation based on both dose and exposure time. Flow cytometry revealed that CTZ decreased apoptosis and MMP and induced G0/G1 arrest. Immunofluorescence demonstrated that CTZ dose-dependently elevated in both total and mitochondrial ROS production. Western blotting showed that CTZ enhanced Bax and cleaved poly ADP-ribose polymerase and caspase-3 while decreasing Bcl-2, p-p65, and p-IκBα. Therefore, CTZ inhibits MM cell proliferation by promoting ROS-mediated mitochondrial apoptosis, inducing G0/G1 arrest, inhibiting the NF-κB pathway, and has the potential for treating MM.


Sujet(s)
Apoptose , Prolifération cellulaire , Clotrimazole , Potentiel de membrane mitochondriale , Mitochondries , Myélome multiple , Espèces réactives de l'oxygène , Humains , Myélome multiple/anatomopathologie , Myélome multiple/traitement médicamenteux , Myélome multiple/métabolisme , Apoptose/effets des médicaments et des substances chimiques , Lignée cellulaire tumorale , Prolifération cellulaire/effets des médicaments et des substances chimiques , Mitochondries/effets des médicaments et des substances chimiques , Mitochondries/métabolisme , Espèces réactives de l'oxygène/métabolisme , Potentiel de membrane mitochondriale/effets des médicaments et des substances chimiques , Clotrimazole/pharmacologie , Phase G0/effets des médicaments et des substances chimiques , Points de contrôle de la phase G1 du cycle cellulaire/effets des médicaments et des substances chimiques , Transduction du signal/effets des médicaments et des substances chimiques , Facteur de transcription NF-kappa B/métabolisme , Antinéoplasiques/pharmacologie , Points de contrôle du cycle cellulaire/effets des médicaments et des substances chimiques
3.
Theranostics ; 14(9): 3565-3582, 2024.
Article de Anglais | MEDLINE | ID: mdl-38948069

RÉSUMÉ

Cancer therapy has moved from single agents to more mechanism-based targeted approaches. In recent years, the combination of HDAC inhibitors and other anticancer chemicals has produced exciting progress in cancer treatment. Herein, we developed a novel prodrug via the ligation of dichloroacetate to selenium-containing potent HDAC inhibitors. The effect and mechanism of this compound in the treatment of prostate cancer were also studied. Methods: The concerned prodrug SeSA-DCA was designed and synthesized under mild conditions. This compound's preclinical studies, including the pharmacokinetics, cell toxicity, and anti-tumor effect on prostate cancer cell lines, were thoroughly investigated, and its possible synergistic mechanism was also explored and discussed. Results: SeSA-DCA showed good stability in physiological conditions and could be rapidly decomposed into DCA and selenium analog of SAHA (SeSAHA) in the tumor microenvironment. CCK-8 experiments identified that SeSA-DCA could effectively inhibit the proliferation of a variety of tumor cell lines, especially in prostate cancer. In further studies, we found that SeSA-DCA could also inhibit the metastasis of prostate cancer cell lines and promote cell apoptosis. At the animal level, oral administration of SeSA-DCA led to significant tumor regression without obvious toxicity. Moreover, as a bimolecular coupling compound, SeSA-DCA exhibited vastly superior efficacy than the mixture with equimolar SeSAHA and DCA both in vitro and in vivo. Our findings provide an important theoretical basis for clinical prostate cancer treatment. Conclusions: Our in vivo and in vitro results showed that SeSA-DCA is a highly effective anti-tumor compound for PCa. It can effectively induce cell cycle arrest and growth suppression and inhibit the migration and metastasis of PCa cell lines compared with monotherapy. SeSA-DCA's ability to decrease the growth of xenografts is a little better than that of docetaxel without any apparent signs of toxicity. Our findings provide an important theoretical basis for clinical prostate cancer treatment.


Sujet(s)
Apoptose , Points de contrôle du cycle cellulaire , Inhibiteurs de désacétylase d'histone , Tumeurs de la prostate , cdc25 Phosphatases , Mâle , Tumeurs de la prostate/traitement médicamenteux , Tumeurs de la prostate/anatomopathologie , Humains , Animaux , Apoptose/effets des médicaments et des substances chimiques , Inhibiteurs de désacétylase d'histone/pharmacologie , Inhibiteurs de désacétylase d'histone/usage thérapeutique , Inhibiteurs de désacétylase d'histone/composition chimique , Lignée cellulaire tumorale , Points de contrôle du cycle cellulaire/effets des médicaments et des substances chimiques , cdc25 Phosphatases/métabolisme , Souris , Antinéoplasiques/pharmacologie , Antinéoplasiques/composition chimique , Prolifération cellulaire/effets des médicaments et des substances chimiques , Souris nude , Sélénium/pharmacologie , Sélénium/composition chimique , Sélénium/usage thérapeutique , Tests d'activité antitumorale sur modèle de xénogreffe , Promédicaments/pharmacologie , Promédicaments/composition chimique , Souris de lignée BALB C
4.
J Mater Chem B ; 12(25): 6257-6274, 2024 Jun 27.
Article de Anglais | MEDLINE | ID: mdl-38845545

RÉSUMÉ

Despite enormous advancements in its management, cancer is the world's primary cause of mortality. Therefore, tremendous strides were made to produce intelligent theranostics with mitigated side effects and improved specificity and efficiency. Thus, we developed a pH-sensitive theranostic platform composed of dextran immobilized zinc oxide nanoparticles, loaded with doxorubicin and radiolabeled with the technetium-99m radionuclide (99mTc-labelled DOX-loaded ZnO@dextran). The platform measured 11.5 nm in diameter with -12 mV zeta potential, 88% DOX loading efficiency and 98.5% radiolabeling efficiency. It showed DOX release in a pH-responsive manner, releasing 93.1% cumulatively at pH 5 but just 7% at pH 7.4. It showed improved intracellular uptake, which resulted in a high growth suppressive effect against MCF-7 cancer cells as compared to the free DOX. It boasted a 4 times lower IC50 than DOX, indicating its significant anti-proliferative potential (0.14 and 0.55 µg ml-1, respectively). The in vitro biological evaluation revealed that its molecular mode of anti-proliferative action included downregulating Cdk-2, which provoked G1/S cell cycle arrest, and upregulating both the intracellular ROS level and caspase-3, which induced apoptosis and necrosis. The in vivo experiments in Ehrlich-ascites carcinoma bearing mice demonstrated that DOX-loaded ZnO@dextran showed a considerable 4-fold increase in anti-tumor efficacy compared to DOX. Moreover, by utilizing the diagnostic radionuclide (99mTc), the radiolabeled platform (99mTc-labelled DOX-loaded ZnO@dextran) was in vivo monitored in tumor-bearing mice, revealing high tumor accumulation (14% ID g-1 at 1 h p.i.) and reduced uptake in non-target organs with a 17.5 T/NT ratio at 1 h p.i. Hence, 99mTc-labelled DOX-loaded ZnO@dextran could be recommended as a rectified tumor-targeted theranostic platform.


Sujet(s)
Apoptose , Points de contrôle du cycle cellulaire , Prolifération cellulaire , Doxorubicine , Nanomédecine théranostique , Oxyde de zinc , Doxorubicine/pharmacologie , Doxorubicine/composition chimique , Oxyde de zinc/composition chimique , Oxyde de zinc/pharmacologie , Humains , Animaux , Apoptose/effets des médicaments et des substances chimiques , Souris , Concentration en ions d'hydrogène , Prolifération cellulaire/effets des médicaments et des substances chimiques , Points de contrôle du cycle cellulaire/effets des médicaments et des substances chimiques , Cellules MCF-7 , Nanoparticules/composition chimique , Distribution tissulaire , Antibiotiques antinéoplasiques/pharmacologie , Antibiotiques antinéoplasiques/composition chimique , Dextrane/composition chimique , Vecteurs de médicaments/composition chimique , Technétium/composition chimique , Taille de particule
5.
Curr Med Sci ; 44(3): 623-632, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38853192

RÉSUMÉ

OBJECTIVE: Endometrial carcinoma (EC) is a prevalent gynecological malignancy characterized by increasing incidence and mortality rates. This underscores the critical need for novel therapeutic targets. One such potential target is cell division cycle 20 (CDC20), which has been implicated in oncogenesis. This study investigated the effect of the CDC20 inhibitor Apcin on EC and elucidated the underlying mechanism involved. METHODS: The effects of Apcin on EC cell proliferation, apoptosis, and the cell cycle were evaluated using CCK8 assays and flow cytometry. RNA sequencing (RNA-seq) was subsequently conducted to explore the underlying molecular mechanism, and Western blotting and coimmunoprecipitation were subsequently performed to validate the results. Animal studies were performed to evaluate the antitumor effects in vivo. Bioinformatics analysis was also conducted to identify CDC20 as a potential therapeutic target in EC. RESULTS: Treatment with Apcin inhibited proliferation and induced apoptosis in EC cells, resulting in cell cycle arrest. Pathways associated with apoptosis and the cell cycle were activated following treatment with Apcin. Notably, Apcin treatment led to the upregulation of the cell cycle regulator p21, which was verified to interact with CDC20 and consequently decrease the expression of downstream cyclins in EC cells. In vivo experiments confirmed that Apcin treatment significantly impeded tumor growth. Higher CDC20 expression was observed in EC tissue than in nonmalignant tissue, and increased CDC20 expression in EC patients was associated with shorter overall survival and progress free interval. CONCLUSION: CDC20 is a novel molecular target in EC, and Apcin could be developed as a candidate antitumor drug for EC treatment.


Sujet(s)
Apoptose , Protéines Cdc20 , Points de contrôle du cycle cellulaire , Prolifération cellulaire , Inhibiteur p21 de kinase cycline-dépendante , Tumeurs de l'endomètre , Femelle , Tumeurs de l'endomètre/traitement médicamenteux , Tumeurs de l'endomètre/anatomopathologie , Tumeurs de l'endomètre/génétique , Tumeurs de l'endomètre/métabolisme , Humains , Apoptose/effets des médicaments et des substances chimiques , Protéines Cdc20/génétique , Protéines Cdc20/métabolisme , Animaux , Points de contrôle du cycle cellulaire/effets des médicaments et des substances chimiques , Lignée cellulaire tumorale , Souris , Inhibiteur p21 de kinase cycline-dépendante/métabolisme , Inhibiteur p21 de kinase cycline-dépendante/génétique , Prolifération cellulaire/effets des médicaments et des substances chimiques , Tests d'activité antitumorale sur modèle de xénogreffe , Régulation de l'expression des gènes tumoraux/effets des médicaments et des substances chimiques , Antinéoplasiques/pharmacologie , Souris nude
6.
J Food Sci ; 89(7): 4469-4479, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38837700

RÉSUMÉ

This study aimed to evaluate the anti-cervical cancer activity of chondroitin sulfate-functionalized selenium nanoparticles (SeCS) and to elucidate their action mechanism. Cytotoxic effect of SeCS on HeLa cells was assessed by MTT assay. Further molecular mechanism of SeCS was analyzed by flow cytometric assay and western blotting. The results showed that treatment with SeCS resulted in a dose- and time-dependent inhibition in the proliferation of HeLa cells. The data obtained from flow cytometry demonstrated that SeCS inhibited HeLa cell growth via the induction of S-phase arrest and cell apoptosis. Further mechanism analysis found that SeCS down-regulated expression levels of cyclin A and CDK2 and up-regulated p21 expression, which contributed to S arrest. Moreover, SeCS increased the level of Bax and decreased the expression of Bcl-2, resulting in the release of cytochrome C from mitochondria and activating caspase-3/8/9 for caspase-dependent apoptosis. Meanwhile, intracellular reactive oxygen species (ROS) levels were elevated after SeCS treatment, suggesting that ROS might be upstream of SeCS-induced S-phase arrest and cell apoptosis. These data show that SeCS has anti-tumor effects and possesses the potential to become a new therapeutic agent or adjuvant therapy for cancer patients. PRACTICAL APPLICATION: In our previous study, we used chondroitin sulfate to stabilize nano-selenium to obtain SeCS to improve the bioactivity and stability of nano-selenium. We found that it possessed an inhibitory effect on HeLa cells. However, the molecular mechanism remains unclear. This study elucidated the mechanism of SeCS damage to HeLa cells. SeCS has the potential to become a new therapeutic agent or adjuvant therapy for cancer patients.


Sujet(s)
Apoptose , Chondroïtines sulfate , Nanoparticules , Espèces réactives de l'oxygène , Sélénium , Humains , Cellules HeLa , Chondroïtines sulfate/pharmacologie , Chondroïtines sulfate/composition chimique , Apoptose/effets des médicaments et des substances chimiques , Sélénium/pharmacologie , Sélénium/composition chimique , Nanoparticules/composition chimique , Espèces réactives de l'oxygène/métabolisme , Prolifération cellulaire/effets des médicaments et des substances chimiques , Points de contrôle de la phase S du cycle cellulaire/effets des médicaments et des substances chimiques , Points de contrôle du cycle cellulaire/effets des médicaments et des substances chimiques , Antinéoplasiques/pharmacologie
7.
Cell Cycle ; 23(6): 713-721, 2024 Mar.
Article de Anglais | MEDLINE | ID: mdl-38879812

RÉSUMÉ

Several breakthrough articles have recently confirmed the ability of tumor cells to escape the stable cell cycle arrest imposed by Therapy-Induced Senescence (TIS). Subsequently, accepting the hypothesis that TIS is escapable should encourage serious reassessments of the fundamental roles of senescence in cancer treatment. The potential for escape from TIS undermines the well-established tumor suppressor function of senescence, proposes it as a mechanism of tumor dormancy leading to disease recurrence and invites for further investigation of its unfavorable contribution to cancer therapy outcomes. Moreover, escaping TIS strongly indicates that the elimination of senescent tumor cells, primarily through pharmacological means, is a suitable approach for increasing the efficacy of cancer treatment, one that still requires further exploration. This commentary provides an overview of the recent evidence that unequivocally demonstrated the ability of therapy-induced senescent tumor cells in overcoming the terminal growth arrest fate and provides future perspectives on the roles of TIS in tumor biology.


Sujet(s)
Vieillissement de la cellule , Tumeurs , Humains , Tumeurs/anatomopathologie , Tumeurs/métabolisme , Animaux , Points de contrôle du cycle cellulaire/effets des médicaments et des substances chimiques , Antinéoplasiques/pharmacologie , Antinéoplasiques/usage thérapeutique
8.
Int J Mol Sci ; 25(11)2024 Jun 04.
Article de Anglais | MEDLINE | ID: mdl-38892366

RÉSUMÉ

In order to overcome the resistance to radiotherapy in human chondrosarcoma cells, the prevention from efficient DNA repair with a combined treatment with the DNA-dependent protein kinase catalytic subunit (DNA-PKcs) inhibitor AZD7648 was explored for carbon ion (C-ion) as well as reference photon (X-ray) irradiation (IR) using gene expression analysis, flow cytometry, protein phosphorylation, and telomere length shortening. Proliferation markers and cell cycle distribution changed significantly after combined treatment, revealing a prominent G2/M arrest. The expression of the G2/M checkpoint genes cyclin B, CDK1, and WEE1 was significantly reduced by IR alone and the combined treatment. While IR alone showed no effects, additional AZD7648 treatment resulted in a dose-dependent reduction in AKT phosphorylation and an increase in Chk2 phosphorylation. Twenty-four hours after IR, the key genes of DNA repair mechanisms were reduced by the combined treatment, which led to impaired DNA repair and increased radiosensitivity. A time-dependent shortening of telomere length was observed in both cell lines after combined treatment with AZD7648 and 8 Gy X-ray/C-ion IR. Our data suggest that the inhibition of DNA-PKcs may increase sensitivity to X-rays and C-ion IR by impairing its functional role in DNA repair mechanisms and telomere end protection.


Sujet(s)
Chondrosarcome , DNA-activated protein kinase , Radiothérapie par ions lourds , Télomère , Humains , DNA-activated protein kinase/antagonistes et inhibiteurs , DNA-activated protein kinase/métabolisme , DNA-activated protein kinase/génétique , Lignée cellulaire tumorale , Chondrosarcome/métabolisme , Chondrosarcome/génétique , Chondrosarcome/radiothérapie , Chondrosarcome/traitement médicamenteux , Télomère/effets des médicaments et des substances chimiques , Télomère/métabolisme , Points de contrôle du cycle cellulaire/effets des médicaments et des substances chimiques , Points de contrôle du cycle cellulaire/effets des radiations , Réparation de l'ADN/effets des médicaments et des substances chimiques , Radiotolérance/effets des médicaments et des substances chimiques , Pyrazoles/pharmacologie , Prolifération cellulaire/effets des médicaments et des substances chimiques , Tumeurs osseuses/métabolisme , Tumeurs osseuses/génétique , Tumeurs osseuses/anatomopathologie , Tumeurs osseuses/traitement médicamenteux , Points de contrôle de la phase G2 du cycle cellulaire/effets des médicaments et des substances chimiques , Points de contrôle de la phase G2 du cycle cellulaire/effets des radiations
9.
Int J Mol Sci ; 25(11)2024 May 24.
Article de Anglais | MEDLINE | ID: mdl-38891892

RÉSUMÉ

Recently, a compound derived from recent scientific advances named 34 has emerged as the focus of this research, the aim of which is to explore its potential impact on solid tumor cell lines. Using a combination of bioinformatics and biological assays, this study conducted an in-depth investigation of the effects of 34. The results of this study have substantial implications for cancer research and treatment. 34 has shown remarkable efficacy in inhibiting the growth of several cancer cell lines, including those representing prostate carcinoma (PC3) and cervical carcinoma (HeLa). The high sensitivity of these cells, indicated by low IC50 values, underscores its potential as a promising chemotherapeutic agent. In addition, 34 has revealed the ability to induce cell cycle arrest, particularly in the G2/M phase, a phenomenon with critical implications for tumor initiation and growth. By interfering with DNA replication in cancer cells, 34 has shown the capacity to trigger cell death, offering a new avenue for cancer treatment. In addition, computational analyses have identified key genes affected by 34 treatment, suggesting potential therapeutic targets. These genes are involved in critical biological processes, including cell cycle regulation, DNA replication and microtubule dynamics, all of which are central to cancer development and progression. In conclusion, this study highlights the different mechanisms of 34 that inhibit cancer cell growth and alter the cell cycle. These promising results suggest the potential for more effective and less toxic anticancer therapies. Further in vivo validation and exploration of combination therapies are critical to improve cancer treatment outcomes.


Sujet(s)
Acrylonitrile , Antinéoplasiques , Microtubules , Humains , Microtubules/effets des médicaments et des substances chimiques , Microtubules/métabolisme , Antinéoplasiques/pharmacologie , Antinéoplasiques/usage thérapeutique , Lignée cellulaire tumorale , Acrylonitrile/analogues et dérivés , Acrylonitrile/pharmacologie , Acrylonitrile/usage thérapeutique , Prolifération cellulaire/effets des médicaments et des substances chimiques , Tumeurs/traitement médicamenteux , Tumeurs/métabolisme , Tumeurs/anatomopathologie , Cellules HeLa , Apoptose/effets des médicaments et des substances chimiques , Triazoles/pharmacologie , Points de contrôle du cycle cellulaire/effets des médicaments et des substances chimiques , Modulateurs de la polymérisation de la tubuline/pharmacologie , Modulateurs de la polymérisation de la tubuline/usage thérapeutique , Cellules PC-3
10.
Cell Biol Toxicol ; 40(1): 41, 2024 Jun 04.
Article de Anglais | MEDLINE | ID: mdl-38833095

RÉSUMÉ

Hippocampal neurons maintain the ability of proliferation throughout life to support neurogenesis. Deoxynivalenol (DON) is a mycotoxin that exhibits brain toxicity, yet whether and how DON affects hippocampal neurogenesis remains unknown. Here, we use mouse hippocampal neuron cells (HT-22) as a model to illustrate the effects of DON on neuron proliferation and to explore underlying mechanisms. DON exposure significantly inhibits the proliferation of HT-22 cells, which is associated with an up-regulation of cell cycle inhibitor p21 at both mRNA and protein levels. Global and site-specific m6A methylation levels on the 3'UTR of p21 mRNA are significantly increased in response to DON treatment, whereas inhibition of m6A hypermethylation significantly alleviates DON-induced cell cycle arrest. Further mechanistic studies indicate that the m6A readers YTHDF1 and IGF2BP1 are responsible for m6A-mediated increase in p21 mRNA stability. Meanwhile, 3'UTR of E3 ubiquitin ligase TRIM21 mRNA is also m6A hypermethylated, and another m6A reader YTHDF2 binds to the m6A sites, leading to decreased TRIM21 mRNA stability. Consequently, TRIM21 suppression impairs ubiquitin-mediated p21 protein degradation. Taken together, m6A-mediated upregulation of p21, at both post-transcriptional and post-translational levels, contributes to DON-induced inhibition of hippocampal neuron proliferation. These results may provide new insights for epigenetic therapy of neurodegenerative diseases.


Sujet(s)
Prolifération cellulaire , Inhibiteur p21 de kinase cycline-dépendante , Hippocampe , Neurones , Trichothécènes , Régulation positive , Animaux , Trichothécènes/toxicité , Trichothécènes/pharmacologie , Hippocampe/métabolisme , Hippocampe/effets des médicaments et des substances chimiques , Hippocampe/cytologie , Souris , Neurones/effets des médicaments et des substances chimiques , Neurones/métabolisme , Inhibiteur p21 de kinase cycline-dépendante/métabolisme , Inhibiteur p21 de kinase cycline-dépendante/génétique , Régulation positive/effets des médicaments et des substances chimiques , Prolifération cellulaire/effets des médicaments et des substances chimiques , Protéines de liaison à l'ARN/métabolisme , Protéines de liaison à l'ARN/génétique , Lignée cellulaire , Régions 3' non traduites/génétique , Neurogenèse/effets des médicaments et des substances chimiques , ARN messager/métabolisme , ARN messager/génétique , Stabilité de l'ARN/effets des médicaments et des substances chimiques , Points de contrôle du cycle cellulaire/effets des médicaments et des substances chimiques , Ribonucléoprotéines/métabolisme , Ribonucléoprotéines/génétique , Méthylation/effets des médicaments et des substances chimiques
11.
Cell Death Dis ; 15(6): 431, 2024 Jun 19.
Article de Anglais | MEDLINE | ID: mdl-38898028

RÉSUMÉ

Non-small cell lung cancer (NSCLC) presents a global health challenge due to its low five-year survival rates, underscoring the need for novel therapeutic strategies. Our research explored the synergistic mechanisms of syrosingopine and UK-5099 in treating NSCLC. In vitro experiments showed that the combination of syrosingopine and UK-5099 significantly synergized to suppress NSCLC proliferation. Further experiments revealed that this combination induced cell cycle arrest and promoted apoptosis in NSCLC cells. In vivo experiments demonstrated that the combination of syrosingopine and UK-5099 markedly inhibited tumor growth. Mechanistic studies revealed that this drug combination promoted mitochondrial damage by inducing lactate accumulation and oxidative stress. Additionally, the combination triggered an integrated stress response (ISR) through the activation of heme-regulated inhibitor kinase (HRI). Importantly, our findings suggested that the synergistic suppression of NSCLC by syrosingopine and UK-5099 was dependent on ISR activation. In summary, our study proposed a promising therapeutic approach that involved the combination of Syrosingopine and UK-5099 to activate ISR, significantly hindering NSCLC growth and proliferation.


Sujet(s)
Carcinome pulmonaire non à petites cellules , Prolifération cellulaire , Synergie des médicaments , Tumeurs du poumon , Carcinome pulmonaire non à petites cellules/traitement médicamenteux , Carcinome pulmonaire non à petites cellules/anatomopathologie , Carcinome pulmonaire non à petites cellules/métabolisme , Humains , Tumeurs du poumon/anatomopathologie , Tumeurs du poumon/traitement médicamenteux , Tumeurs du poumon/métabolisme , Animaux , Prolifération cellulaire/effets des médicaments et des substances chimiques , Apoptose/effets des médicaments et des substances chimiques , Souris , Souris nude , Lignée cellulaire tumorale , Stress oxydatif/effets des médicaments et des substances chimiques , Tests d'activité antitumorale sur modèle de xénogreffe , Souris de lignée BALB C , Points de contrôle du cycle cellulaire/effets des médicaments et des substances chimiques
12.
Cancer Cell ; 42(6): 1126, 2024 Jun 10.
Article de Anglais | MEDLINE | ID: mdl-38866454
13.
Toxicon ; 245: 107788, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38823652

RÉSUMÉ

Ginkgo biloba L. is a valuable plant, which can be used for medicine, food and ornamental purposes. Despite the above benefits, the components of ginkgolic acids (GA) in ginkgo are considered to cause allergies, embryotoxicity, liver damage and some other adverse reactions. However, the mechanism of GA induced liver injury is still unclear. In this study, we developed an acute liver injury model induced by GA in mice, and investigated the mechanism of GA induced liver injury from the perspectives of oxidative stress, steatosis, apoptosis, and immune response. Intraperitoneal injection of GA (400 mg/kg) can cause liver damage. The levels of serum transaminase, oxidation and triglycerides were increased, liver fibrosis, hepatocyte apoptosis, G2/M phase arrest of the hepatic cell cycle and monocyte infiltration in the liver were detected in GA-treated mice. Flow cytometry analysis of cells separated from the spleen showed that the proportion of Th1 and Th17 cells were increased, and the proportion of Th2 cells were decreased in GA-treated mice. The rise in Th1/Th2 ratio and Th17 cell ratio usually cause inflammatory problems. At the same time, cleaved Caspase-8 and Caspase-3 were detected in hepatocytes, indicating that GA may induce apoptosis through FADD pathway. Although GA is capable of causing the above problems, the inflammation and damage in liver tissue are not severe and there are certain individual differences. Our study reveals the potential hepatotoxicity of GA in ginkgo and its mechanism of action, providing a new perspective for the intervention and prevention of ginkgo toxicity.


Sujet(s)
Apoptose , Lésions hépatiques dues aux substances , Salicylates , Animaux , Souris , Salicylates/toxicité , Apoptose/effets des médicaments et des substances chimiques , Ginkgo biloba , Stress oxydatif/effets des médicaments et des substances chimiques , Points de contrôle du cycle cellulaire/effets des médicaments et des substances chimiques , Foie/effets des médicaments et des substances chimiques , Foie/anatomopathologie , Mâle
14.
Mol Med Rep ; 30(2)2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-38904207

RÉSUMÉ

Montelukast and zafirlukast, cysteinyl leukotriene receptor antagonists (LTRAs), trigger apoptosis and inhibit cell proliferation of triple­negative breast cancer MDA­MB­231 cells. By contrast, only zafirlukast induces G0/G1 cell cycle arrest. The present study compared the effects of these drugs on proteins regulating cell proliferation, apoptosis, autophagy, and endoplasmic reticulum (ER) and oxidative stress using reverse transcription­quantitative PCR, western blotting and flow cytometry. The expression of proliferating markers, Ki­67 and proliferating cell nuclear antigen, was decreased by both drugs. Zafirlukast, but not montelukast, decreased the expression of cyclin D1 and CDK4, disrupting progression from G1 to S phase. Zafirlukast also increased the expression of p27, a cell cycle inhibitor. Both drugs decreased the expression of anti­apoptotic protein Bcl­2 and ERK1/2 phosphorylation, and increased levels of the autophagy marker LC3­II and DNA damage markers, including cleaved PARP­1, phosphorylated (p)­ATM and p­histone H2AX. The number of caspase 3/7­positive cells was greater in montelukast­treated cells compared with zafirlukast­treated cells. Montelukast induced higher levels of the ER stress marker CHOP compared with zafirlukast. Montelukast activated PERK, activating transcription factor 6 (ATF6) and inositol­requiring enzyme type 1 (IRE1) pathways, while zafirlukast only stimulated ATF6 and IRE1 pathways. GSK2606414, a PERK inhibitor, decreased apoptosis mediated by montelukast, but did not affect zafirlukast­induced cell death. The knockdown of CHOP by small interfering RNA reduced apoptosis triggered by montelukast and zafirlukast. In conclusion, the effects on cell cycle regulator proteins may contribute to cell cycle arrest caused by zafirlukast. The greater apoptotic effects of montelukast may be caused by the higher levels of activated caspase enzymes and the activation of three pathways of ER stress: PERK, ATF6, and IRE1.


Sujet(s)
Acétates , Apoptose , Autophagie , Cyclopropanes , Altération de l'ADN , Stress du réticulum endoplasmique , Indoles , Quinoléines , Sulfures , Sulfonamides , Humains , Sulfures/pharmacologie , Cyclopropanes/pharmacologie , Quinoléines/pharmacologie , Apoptose/effets des médicaments et des substances chimiques , Acétates/pharmacologie , Stress du réticulum endoplasmique/effets des médicaments et des substances chimiques , Lignée cellulaire tumorale , Autophagie/effets des médicaments et des substances chimiques , Sulfonamides/pharmacologie , Indoles/pharmacologie , Femelle , Altération de l'ADN/effets des médicaments et des substances chimiques , Phényl-carbamates/pharmacologie , Composés tosyliques/pharmacologie , Prolifération cellulaire/effets des médicaments et des substances chimiques , eIF-2 Kinase/métabolisme , eIF-2 Kinase/génétique , Endoribonucleases/métabolisme , Endoribonucleases/génétique , Points de contrôle du cycle cellulaire/effets des médicaments et des substances chimiques , Facteur de transcription CHOP/métabolisme , Facteur de transcription CHOP/génétique , Cycle cellulaire/effets des médicaments et des substances chimiques , Antagonistes des leucotriènes/pharmacologie , Protein-Serine-Threonine Kinases/métabolisme , Protein-Serine-Threonine Kinases/génétique
15.
Cell Biochem Funct ; 42(5): e4083, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38938150

RÉSUMÉ

Multidrug resistance (MDR) during clinical chemotherapy for cancer has been considered a major obstacle to treatment efficacy. The involvement of adenosine triphosphate-binding cassette (ABC) transporters in the MDR mechanism significantly reduces the efficacy of chemotherapeutics. This study investigates the potential of morin, a dietary bioflavonoid, to overcome colchicine resistance in KBChR-8-5 MDR cells. The P-gp inhibitory activity by morin was measured by calcein-AM drug efflux assay. Western blot analysis was employed to evaluate P-gp messenger RNA and protein expressions following morin treatment. Flow cytometry analysis and acridine orange/ethidium bromide fluorescence staining were utilised to investigate the induction of apoptosis and cell cycle arrest upon treatment with morin and paclitaxel in combination. Additionally, polymerase chain reaction (PCR) array analysis was conducted to study the gene expression profiles related to MDR, apoptosis and cell cycle arrest during treatment with morin, paclitaxel or their combination. Morin exhibited a strong binding interaction with human P-gp. This was corroborated by drug efflux assays, which showed a reduction in P-gp efflux function with increasing morin concentration. Furthermore, morin and paclitaxel combination potentiated the induction of apoptosis and G2/M phase cell cycle arrest. Morin treatment significantly downregulated the gene expression of ABCB1 and P-gp membrane expressions in MDR cells. Additionally, PCR array gene expression analysis revealed that the combination treatment with morin and paclitaxel upregulated proapoptotic and cell cycle arrest genes while downregulating ABCB1 gene and antiapoptotic genes. Thus, morin effectively reversed paclitaxel resistance in KBChR-8-5 drug-resistant cancer cells and concluded that morin resensitized the paclitaxel resistance in KBChR8-5 drug-resistant cancer cells.


Sujet(s)
Glycoprotéine P , Apoptose , Multirésistance aux médicaments , Résistance aux médicaments antinéoplasiques , Flavonoïdes , Paclitaxel , Humains , Flavonoïdes/pharmacologie , Multirésistance aux médicaments/effets des médicaments et des substances chimiques , Résistance aux médicaments antinéoplasiques/effets des médicaments et des substances chimiques , Apoptose/effets des médicaments et des substances chimiques , Paclitaxel/pharmacologie , Glycoprotéine P/métabolisme , Glycoprotéine P/génétique , Lignée cellulaire tumorale , Points de contrôle du cycle cellulaire/effets des médicaments et des substances chimiques , Sous-famille B de transporteurs à cassette liant l'ATP/métabolisme , Sous-famille B de transporteurs à cassette liant l'ATP/génétique , Sous-famille B de transporteurs à cassette liant l'ATP/antagonistes et inhibiteurs , Antinéoplasiques/pharmacologie , Antinéoplasiques/composition chimique , Flavones
16.
Int J Mol Sci ; 25(9)2024 May 03.
Article de Anglais | MEDLINE | ID: mdl-38732206

RÉSUMÉ

Breast cancer stands out as one of the most prevalent malignancies worldwide, necessitating a nuanced understanding of its molecular underpinnings for effective treatment. Hormone receptors in breast cancer cells substantially influence treatment strategies, dictating therapeutic approaches in clinical settings, serving as a guide for drug development, and aiming to enhance treatment specificity and efficacy. Natural compounds, such as curcumin, offer a diverse array of chemical structures with promising therapeutic potential. Despite curcumin's benefits, challenges like poor solubility and rapid metabolism have spurred the exploration of analogs. Here, we evaluated the efficacy of the curcumin analog NC2603 to induce cell cycle arrest in MCF-7 breast cancer cells and explored its molecular mechanisms. Our findings reveal potent inhibition of cell viability (IC50 = 5.6 µM) and greater specificity than doxorubicin toward MCF-7 vs. non-cancer HaCaT cells. Transcriptome analysis identified 12,055 modulated genes, most notably upregulation of GADD45A and downregulation of ESR1, implicating CDKN1A-mediated regulation of proliferation and cell cycle genes. We hypothesize that the curcumin analog by inducing GADD45A expression and repressing ESR1, triggers the expression of CDKN1A, which in turn downregulates the expression of many important genes of proliferation and the cell cycle. These insights advance our understanding of curcumin analogs' therapeutic potential, highlighting not just their role in treatment, but also the molecular pathways involved in their activity toward breast cancer cells.


Sujet(s)
Tumeurs du sein , Points de contrôle du cycle cellulaire , Curcumine , Inhibiteur p21 de kinase cycline-dépendante , Régulation de l'expression des gènes tumoraux , Humains , Curcumine/pharmacologie , Curcumine/analogues et dérivés , Tumeurs du sein/métabolisme , Tumeurs du sein/traitement médicamenteux , Tumeurs du sein/génétique , Tumeurs du sein/anatomopathologie , Cellules MCF-7 , Inhibiteur p21 de kinase cycline-dépendante/métabolisme , Inhibiteur p21 de kinase cycline-dépendante/génétique , Points de contrôle du cycle cellulaire/effets des médicaments et des substances chimiques , Femelle , Régulation de l'expression des gènes tumoraux/effets des médicaments et des substances chimiques , Régulation positive/effets des médicaments et des substances chimiques , Prolifération cellulaire/effets des médicaments et des substances chimiques , Survie cellulaire/effets des médicaments et des substances chimiques , Protéines du cycle cellulaire/métabolisme , Protéines du cycle cellulaire/génétique , Récepteur alpha des oestrogènes/métabolisme , Récepteur alpha des oestrogènes/génétique , Antinéoplasiques/pharmacologie ,
17.
Chem Commun (Camb) ; 60(48): 6150-6153, 2024 Jun 11.
Article de Anglais | MEDLINE | ID: mdl-38804255

RÉSUMÉ

Iron-binding strategies in anticancer drug design target the key role of iron in cancer growth. The incorporation of a quinoline moiety in the design of tetrazolium-based prochelators facilitates their intracellular reduction/activation to iron-binding formazans. The new prochelators are antiproliferative at submicromolar levels, induce apoptosis and cell cycle arrest, and impact iron signaling in cancer cells.


Sujet(s)
Antinéoplasiques , Apoptose , Prolifération cellulaire , Fer , Quinoléines , Humains , Quinoléines/composition chimique , Quinoléines/pharmacologie , Prolifération cellulaire/effets des médicaments et des substances chimiques , Antinéoplasiques/pharmacologie , Antinéoplasiques/composition chimique , Antinéoplasiques/synthèse chimique , Fer/composition chimique , Fer/métabolisme , Apoptose/effets des médicaments et des substances chimiques , Lignée cellulaire tumorale , Tests de criblage d'agents antitumoraux , Structure moléculaire , Points de contrôle du cycle cellulaire/effets des médicaments et des substances chimiques
18.
Exp Parasitol ; 262: 108773, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38723845

RÉSUMÉ

Giardiasis is a prevalent parasitic diarrheal disease caused by Giardia lamblia, affecting people worldwide. Recently, the availability of several drugs for its treatment has highlighted issues such as multidrug resistance, limited effectiveness and undesirable side effects. Therefore, it is necessary to develop alternative new drugs and treatment strategies that can enhance therapeutic outcomes and effectively treat giardiasis. Natural compounds show promise in the search for more potent anti-giardial agents. Our investigation focused on the effect of Andrographolide (ADG), an active compound of the Andrographis paniculata plant, on Giardia lamblia, assessing trophozoite growth, morphological changes, cell cycle arrest, DNA damage and inhibition of gene expression associated with pathogenic factors. ADG demonstrated anti-Giardia activity almost equivalent to the reference drug metronidazole, with an IC50 value of 4.99 µM after 24 h of incubation. In cytotoxicity assessments and morphological examinations, it showed significant alterations in trophozoite shape and size and effectively hindered the adhesion of trophozoites. It also caused excessive ROS generation, DNA damage, cell cycle arrest and inhibited the gene expression related to pathogenesis. Our findings have revealed the anti-giardial efficacy of ADG, suggesting its potential as an agent against Giardia infections. This could offer a natural and low-risk treatment option for giardiasis, reducing the risk of side effects and drug resistance.


Sujet(s)
Antiprotozoaires , Points de contrôle du cycle cellulaire , Altération de l'ADN , Diterpènes , Giardia lamblia , Concentration inhibitrice 50 , Espèces réactives de l'oxygène , Trophozoïtes , Diterpènes/pharmacologie , Giardia lamblia/effets des médicaments et des substances chimiques , Giardia lamblia/croissance et développement , Giardia lamblia/génétique , Trophozoïtes/effets des médicaments et des substances chimiques , Trophozoïtes/croissance et développement , Points de contrôle du cycle cellulaire/effets des médicaments et des substances chimiques , Espèces réactives de l'oxygène/métabolisme , Altération de l'ADN/effets des médicaments et des substances chimiques , Antiprotozoaires/pharmacologie , Humains , Animaux , Expression des gènes/effets des médicaments et des substances chimiques , Métronidazole/pharmacologie
19.
PLoS One ; 19(5): e0302662, 2024.
Article de Anglais | MEDLINE | ID: mdl-38748716

RÉSUMÉ

Kaab Dum, a prominent indigenous rice variety cultivated in the Pak Phanang Basin of Nakhon Si Thammarat, Thailand, is the focus of our study. We investigate the therapeutic potential of indigenous Kaab Dum rice extract in the context of chronic wounds. Our research encompasses an examination of the nutritional compositions and chemical profiles of Kaab Dum rice extract. Additionally, we assess how the extract affects chronic wounds in TGF-ß-induced HaCaT cells. Our evaluation methods include the detection of cellular oxidative stress, the examination of endoplasmic reticulum (ER) stress, wound healing assays, analysis of cell cycle arrest and the study of cellular senescence through senescence-associated ß-galactosidase (SA-ß-gal) staining. Our research findings demonstrate that TGF-ß induces oxidative stress in HaCaT cells, which subsequently triggers ER stress, confirmed by the expression of the PERK protein. This ER stress results in cell cycle arrest in HaCaT cells, characterized by an increase in p21 protein, a cyclin-dependent kinase inhibitor (CDKI). Ultimately, this leads to cellular senescence, as confirmed by SA-ß-gal staining. Importantly, our study reveals the effectiveness of Kaab Dum rice extract in promoting wound healing in the chronic wound model. The extract reduces ER stress and senescent cells. These beneficial effects are potentially linked to the antioxidant and anti-inflammatory properties of the rice extract. The findings of our study have the potential to make significant contributions to the development of enhanced products for both the prevention and treatment of chronic wounds.


Sujet(s)
Vieillissement de la cellule , Stress du réticulum endoplasmique , Kératinocytes , Oryza , Extraits de plantes , Cicatrisation de plaie , Humains , Oryza/composition chimique , Vieillissement de la cellule/effets des médicaments et des substances chimiques , Cicatrisation de plaie/effets des médicaments et des substances chimiques , Stress du réticulum endoplasmique/effets des médicaments et des substances chimiques , Kératinocytes/effets des médicaments et des substances chimiques , Kératinocytes/métabolisme , Extraits de plantes/pharmacologie , Thaïlande , Lignée cellulaire , Cellules HaCaT , Stress oxydatif/effets des médicaments et des substances chimiques , Facteur de croissance transformant bêta/métabolisme , Points de contrôle du cycle cellulaire/effets des médicaments et des substances chimiques , Peuples d'Asie du Sud-Est
20.
Genes (Basel) ; 15(5)2024 05 01.
Article de Anglais | MEDLINE | ID: mdl-38790208

RÉSUMÉ

T-2 toxin (T-2), an A-type mono mycotoxin produced by various Fusarium species, disrupts DNA/RNA and protein synthesis upon entering the body, resulting in pathological conditions in various tissues/organs and posing a significant threat to human and animal health. However, the mechanisms underlying its toxicity remain unclear. With the goal of learning how T-2 affects reproduction in animals, we utilized primary porcine ovarian granulosa cells (pGCs) as a carrier in vitro and constructed concentration models for analyzing cell morphology and RNA-sequencing (RNA-seq). Our findings showed that T-2 could influence pGCs morphology, induce cell cycle arrest, and promote apoptosis in a dose-dependent manner. The results of RNA-seq analyses indicated that a total of 8216 genes exhibited significant differential expression (DEG) following T-2 treatment, of which 4812 were observed to be down-regulated and 3404 were up-regulated. The DEGs following T-2 toxin treatment of pGCs had a notable impact on many metabolic pathways such as PI3K-Akt, Ras, MAPK, and apoptosis, which in turn altered important physiological processes. Gene set enrichment analysis (GSEA) indicated that the differences in the harmful effects of T-2 might be caused by the varying control of cellular processes and the pathway responsible for steroid metabolism. These results present further insights regarding the mechanism of T-2 action on sow reproductive toxicity, enhance our understanding of T-2 reproductive toxicological effects, and lay a theoretical foundation for the judicious prevention of T-2-induced reproductive toxicity.


Sujet(s)
Apoptose , Cellules de la granulosa , Toxine T-2 , Animaux , Toxine T-2/toxicité , Femelle , Cellules de la granulosa/effets des médicaments et des substances chimiques , Cellules de la granulosa/métabolisme , Apoptose/effets des médicaments et des substances chimiques , Suidae , Cellules cultivées , Points de contrôle du cycle cellulaire/effets des médicaments et des substances chimiques , Transduction du signal/effets des médicaments et des substances chimiques
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...