Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 1.106
Filtrer
1.
Cell Death Dis ; 15(8): 583, 2024 Aug 09.
Article de Anglais | MEDLINE | ID: mdl-39122708

RÉSUMÉ

In advanced hepatocellular carcinoma (HCC), RNA helicase DDX5 regulates the Wnt/ß-catenin-ferroptosis axis, influencing the efficacy of the multi-tyrosine kinase inhibitor (mTKI) sorafenib. DDX5 inhibits Wnt/ß-catenin signaling, preventing sorafenib-induced ferroptosis escape. Sorafenib/mTKIs reduce DDX5 expression, correlating with poor patient survival post-sorafenib treatment. Notably, DDX5-knockout in HCC cells activates Wnt/ß-catenin signaling persistently. Herein, we investigate the mechanistic impact of Wnt/ß-catenin activation resulting from DDX5 downregulation in the progression and treatment of HCC. RNAseq analyses identified shared genes repressed by DDX5 and upregulated by sorafenib, including Wnt signaling genes, NF-κB-inducing kinase (NIK) essential for non-canonical NF-κB (p52/RelB) activation, and cytoprotective transcription factor NRF2. We demonstrate, Wnt/ß-catenin activation induced NIK transcription, leading to non-canonical NF-κB activation, which subsequently mediated NRF2 transcription. Additionally, DDX5 deficiency extended NRF2 protein half-life by inactivating KEAP1 through p62/SQSTM1 stabilization. In a preclinical HCC mouse model, NRF2 knockdown or DDX5 overexpression restricted tumor growth upon sorafenib treatment, via induction of ferroptosis. Importantly, DDX5-knockout HCC cells exhibited elevated expression of Wnt signaling genes, NIK, p52/RelB, and NRF2-regulated genes, regardless of sorafenib treatment. Transcriptomic analyses of HCCs from TCGA and the Stelic Animal Model (STAM) of non-alcoholic steatohepatitis revealed elevated expression of these interconnected pathways in the context of DDX5 downregulation. In conclusion, DDX5 deficiency triggers Wnt/ß-catenin signaling, promoting p52/RelB and NRF2 activation, thereby enabling ferroptosis evasion upon sorafenib treatment. Similarly, independent of sorafenib, DDX5 deficiency in liver tumors enhances activation and gene expression of these interconnected pathways, underscoring the clinical relevance of DDX5 deficiency in HCC progression and therapeutic response.


Sujet(s)
Carcinome hépatocellulaire , DEAD-box RNA helicases , Évolution de la maladie , Tumeurs du foie , Facteur-2 apparenté à NF-E2 , Facteur de transcription NF-kappa B , Sorafénib , Sorafénib/pharmacologie , Carcinome hépatocellulaire/traitement médicamenteux , Carcinome hépatocellulaire/génétique , Carcinome hépatocellulaire/anatomopathologie , Carcinome hépatocellulaire/métabolisme , Tumeurs du foie/génétique , Tumeurs du foie/traitement médicamenteux , Tumeurs du foie/anatomopathologie , Tumeurs du foie/métabolisme , DEAD-box RNA helicases/métabolisme , DEAD-box RNA helicases/génétique , Facteur-2 apparenté à NF-E2/métabolisme , Facteur-2 apparenté à NF-E2/génétique , Animaux , Humains , Souris , Facteur de transcription NF-kappa B/métabolisme , Régulation de l'expression des gènes tumoraux/effets des médicaments et des substances chimiques , Lignée cellulaire tumorale , Voie de signalisation Wnt/effets des médicaments et des substances chimiques , Ferroptose/effets des médicaments et des substances chimiques , Ferroptose/génétique
2.
Cell Death Dis ; 15(8): 595, 2024 Aug 16.
Article de Anglais | MEDLINE | ID: mdl-39152108

RÉSUMÉ

Hepatocellular carcinoma (HCC) has risen as the villain of cancer-related death globally, with a usual cruel forecasting. Sorafenib was officially approved by the FDA as first-line treatment for advanced HCC. Despite the brilliant promise revealed in research, actual clinical results are limited due to the widespread appearance of drug resistance. The tumor microenvironment (TME) has been correlated to pharmacological resistance, implying that existing cellular level strategies may be insufficient to improve therapy success. The role of autophagy in cancer is a two-edged sword. On one hand, autophagy permits malignant cells to overcome stress, such as hypoxic TME and therapy-induced starvation. Autophagy, on the other hand, plays an important role in damage suppression, which can reduce carcinogenesis. As a result, controlling autophagy is certainly a viable technique in cancer therapy. The goal of this study was to investigate at the impact of autophagy manipulation with sorafenib therapy by analyzing autophagy induction and inhibition to sorafenib monotherapy in rats with HCC. Western blot, ELISA, immunohistochemistry, flow cytometry, and quantitative-PCR were used to investigate autophagy, apoptosis, and the cell cycle. Routine biochemical and pathological testing was performed. Ultracellular features and autophagic entities were observed using a transmission electron microscope (TEM). Both regimens demonstrated significant reductions in chemotherapeutic resistance and hepatoprotective effects. According to the findings, both autophagic inhibitors and inducers are attractive candidates for combating sorafenib-induced resistance in HCC.


Sujet(s)
Autophagie , Carcinome hépatocellulaire , Résistance aux médicaments antinéoplasiques , Tumeurs du foie , Sorafénib , Sorafénib/pharmacologie , Sorafénib/usage thérapeutique , Animaux , Autophagie/effets des médicaments et des substances chimiques , Carcinome hépatocellulaire/traitement médicamenteux , Carcinome hépatocellulaire/anatomopathologie , Carcinome hépatocellulaire/métabolisme , Carcinome hépatocellulaire/génétique , Résistance aux médicaments antinéoplasiques/effets des médicaments et des substances chimiques , Rats , Tumeurs du foie/traitement médicamenteux , Tumeurs du foie/anatomopathologie , Tumeurs du foie/métabolisme , Tumeurs du foie/génétique , Mâle , Apoptose/effets des médicaments et des substances chimiques , Antinéoplasiques/pharmacologie , Antinéoplasiques/usage thérapeutique , Humains , Microenvironnement tumoral/effets des médicaments et des substances chimiques
3.
J Nanobiotechnology ; 22(1): 473, 2024 Aug 09.
Article de Anglais | MEDLINE | ID: mdl-39135024

RÉSUMÉ

BACKGROUND: Hepatocellular carcinoma (HCC) is a highly malignant tumor known for its hypoxic environment, which contributes to resistance against the anticancer drug Sorafenib (SF). Addressing SF resistance in HCC requires innovative strategies to improve tumor oxygenation and effectively deliver therapeutics. RESULTS: In our study, we explored the role of KPNA4 in mediating hypoxia-induced SF resistance in HCC. We developed hemoglobin nanoclusters (Hb-NCs) capable of carrying oxygen, loaded with indocyanine green (ICG) and SF, named HPRG@SF. In vitro, HPRG@SF targeted HCC cells, alleviated hypoxia, suppressed KPNA4 expression, and enhanced the cytotoxicity of PDT against hypoxic, SF-resistant HCC cells. In vivo experiments supported these findings, showing that HPRG@SF effectively improved the oxygenation within the tumor microenvironment and countered SF resistance through combined photodynamic therapy (PDT). CONCLUSION: The combination of Hb-NCs with ICG and SF, forming HPRG@SF, presents a potent strategy to overcome drug resistance in hepatocellular carcinoma by improving hypoxia and employing PDT. This approach not only targets the hypoxic conditions that underlie resistance but also provides a synergistic anticancer effect, highlighting its potential for clinical applications in treating resistant HCC.


Sujet(s)
Carcinome hépatocellulaire , Hémoglobines , Vert indocyanine , Tumeurs du foie , Photothérapie dynamique , Sorafénib , Microenvironnement tumoral , Carcinome hépatocellulaire/traitement médicamenteux , Tumeurs du foie/traitement médicamenteux , Microenvironnement tumoral/effets des médicaments et des substances chimiques , Humains , Photothérapie dynamique/méthodes , Animaux , Hémoglobines/pharmacologie , Lignée cellulaire tumorale , Sorafénib/pharmacologie , Sorafénib/usage thérapeutique , Souris , Vert indocyanine/composition chimique , Vert indocyanine/pharmacologie , Vert indocyanine/usage thérapeutique , Souris nude , Souris de lignée BALB C , Antinéoplasiques/pharmacologie , Antinéoplasiques/composition chimique , Cariophérines alpha/métabolisme , Résistance aux médicaments antinéoplasiques/effets des médicaments et des substances chimiques , Nanoparticules/composition chimique
4.
Cell Commun Signal ; 22(1): 391, 2024 Aug 07.
Article de Anglais | MEDLINE | ID: mdl-39113090

RÉSUMÉ

BACKGROUND: Approximately 25-30% of patients with acute myeloid leukemia (AML) have FMS-like receptor tyrosine kinase-3 (FLT3) mutations that contribute to disease progression and poor prognosis. Prolonged exposure to FLT3 tyrosine kinase inhibitors (TKIs) often results in limited clinical responses due to diverse compensatory survival signals. Therefore, there is an urgent need to elucidate the mechanisms underlying FLT3 TKI resistance. Dysregulated sphingolipid metabolism frequently contributes to cancer progression and a poor therapeutic response. However, its relationship with TKI sensitivity in FLT3-mutated AML remains unknown. Thus, we aimed to assess mechanisms of FLT3 TKI resistance in AML. METHODS: We performed lipidomics profiling, RNA-seq, qRT-PCR, and enzyme-linked immunosorbent assays to determine potential drivers of sorafenib resistance. FLT3 signaling was inhibited by sorafenib or quizartinib, and SPHK1 was inhibited by using an antagonist or via knockdown. Cell growth and apoptosis were assessed in FLT3-mutated and wild-type AML cell lines via Cell counting kit-8, PI staining, and Annexin-V/7AAD assays. Western blotting and immunofluorescence assays were employed to explore the underlying molecular mechanisms through rescue experiments using SPHK1 overexpression and exogenous S1P, as well as inhibitors of S1P2, ß-catenin, PP2A, and GSK3ß. Xenograft murine model, patient samples, and publicly available data were analyzed to corroborate our in vitro results. RESULTS: We demonstrate that long-term sorafenib treatment upregulates SPHK1/sphingosine-1-phosphate (S1P) signaling, which in turn positively modulates ß-catenin signaling to counteract TKI-mediated suppression of FLT3-mutated AML cells via the S1P2 receptor. Genetic or pharmacological inhibition of SPHK1 potently enhanced the TKI-mediated inhibition of proliferation and apoptosis induction in FLT3-mutated AML cells in vitro. SPHK1 knockdown enhanced sorafenib efficacy and improved survival of AML-xenografted mice. Mechanistically, targeting the SPHK1/S1P/S1P2 signaling synergizes with FLT3 TKIs to inhibit ß-catenin activity by activating the protein phosphatase 2 A (PP2A)-glycogen synthase kinase 3ß (GSK3ß) pathway. CONCLUSIONS: These findings establish the sphingolipid metabolic enzyme SPHK1 as a regulator of TKI sensitivity and suggest that combining SPHK1 inhibition with TKIs could be an effective approach for treating FLT3-mutated AML.


Sujet(s)
Glycogen synthase kinase 3 beta , Leucémie aigüe myéloïde , Phosphotransferases (Alcohol Group Acceptor) , Protein Phosphatase 2 , bêta-Caténine , Tyrosine kinase-3 de type fms , Tyrosine kinase-3 de type fms/génétique , Tyrosine kinase-3 de type fms/métabolisme , Tyrosine kinase-3 de type fms/antagonistes et inhibiteurs , Humains , Leucémie aigüe myéloïde/traitement médicamenteux , Leucémie aigüe myéloïde/anatomopathologie , Leucémie aigüe myéloïde/génétique , Leucémie aigüe myéloïde/métabolisme , Glycogen synthase kinase 3 beta/métabolisme , Glycogen synthase kinase 3 beta/génétique , bêta-Caténine/métabolisme , bêta-Caténine/génétique , Phosphotransferases (Alcohol Group Acceptor)/métabolisme , Phosphotransferases (Alcohol Group Acceptor)/génétique , Phosphotransferases (Alcohol Group Acceptor)/antagonistes et inhibiteurs , Animaux , Souris , Protein Phosphatase 2/métabolisme , Protein Phosphatase 2/génétique , Protein Phosphatase 2/antagonistes et inhibiteurs , Lignée cellulaire tumorale , Sorafénib/pharmacologie , Apoptose/effets des médicaments et des substances chimiques , Inhibiteurs de protéines kinases/pharmacologie , Transduction du signal/effets des médicaments et des substances chimiques , Prolifération cellulaire/effets des médicaments et des substances chimiques , Synergie des médicaments , Tests d'activité antitumorale sur modèle de xénogreffe , Résistance aux médicaments antinéoplasiques/effets des médicaments et des substances chimiques , Résistance aux médicaments antinéoplasiques/génétique
5.
J Cell Mol Med ; 28(14): e18533, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-39034442

RÉSUMÉ

Hepatitis B Virus (HBV) infection significantly elevates the risk of hepatocellular carcinoma (HCC), with the HBV X protein (HBx) playing a crucial role in cancer progression. Sorafenib, the primary therapy for advanced HCC, shows limited effectiveness in HBV-infected patients due to HBx-related resistance. Numerous studies have explored combination therapies to overcome this resistance. Sodium diethyldithiocarbamate (DDC), known for its anticancer effects and its inhibition of superoxide dismutase 1 (SOD1), is hypothesized to counteract sorafenib (SF) resistance in HBV-positive HCCs. Our research demonstrates that combining DDC with SF significantly reduces HBx and SOD1 expressions in HBV-positive HCC cells and human tissues. This combination therapy disrupts the PI3K/Akt/mTOR signalling pathway and promotes apoptosis by increasing reactive oxygen species (ROS) levels. These cellular changes lead to reduced tumour viability and enhanced sensitivity to SF, as evidenced by the synergistic suppression of tumour growth in xenograft models. Additionally, DDC-mediated suppression of SOD1 further enhances SF sensitivity in HBV-positive HCC cells and xenografted animals, thereby inhibiting cancer progression more effectively. These findings suggest that the DDC-SF combination could serve as a promising strategy for overcoming SF resistance in HBV-related HCC, potentially optimizing therapy outcomes.


Sujet(s)
Carcinome hépatocellulaire , Virus de l'hépatite B , Tumeurs du foie , Phosphatidylinositol 3-kinases , Protéines proto-oncogènes c-akt , Espèces réactives de l'oxygène , Transduction du signal , Sorafénib , Superoxide dismutase-1 , Sérine-thréonine kinases TOR , Sorafénib/pharmacologie , Sorafénib/usage thérapeutique , Humains , Tumeurs du foie/traitement médicamenteux , Tumeurs du foie/virologie , Tumeurs du foie/métabolisme , Tumeurs du foie/anatomopathologie , Carcinome hépatocellulaire/traitement médicamenteux , Carcinome hépatocellulaire/virologie , Carcinome hépatocellulaire/métabolisme , Carcinome hépatocellulaire/anatomopathologie , Espèces réactives de l'oxygène/métabolisme , Protéines proto-oncogènes c-akt/métabolisme , Superoxide dismutase-1/métabolisme , Superoxide dismutase-1/génétique , Animaux , Sérine-thréonine kinases TOR/métabolisme , Sérine-thréonine kinases TOR/antagonistes et inhibiteurs , Phosphatidylinositol 3-kinases/métabolisme , Transduction du signal/effets des médicaments et des substances chimiques , Souris , Virus de l'hépatite B/effets des médicaments et des substances chimiques , Lignée cellulaire tumorale , Tests d'activité antitumorale sur modèle de xénogreffe , Apoptose/effets des médicaments et des substances chimiques , Hépatite B/complications , Hépatite B/traitement médicamenteux , Hépatite B/virologie , Acide diéthyl-dithiocarbamique/pharmacologie , Résistance aux médicaments antinéoplasiques/effets des médicaments et des substances chimiques , Souris nude , Prolifération cellulaire/effets des médicaments et des substances chimiques , Transactivateurs , Protéines virales régulatrices ou accessoires
6.
Signal Transduct Target Ther ; 9(1): 169, 2024 Jul 03.
Article de Anglais | MEDLINE | ID: mdl-38956074

RÉSUMÉ

More than 90% of hepatocellular carcinoma (HCC) cases develop in the presence of fibrosis or cirrhosis, making the tumor microenvironment (TME) of HCC distinctive due to the intricate interplay between cancer-associated fibroblasts (CAFs) and cancer stem cells (CSCs), which collectively regulate HCC progression. However, the mechanisms through which CSCs orchestrate the dynamics of the tumor stroma during HCC development remain elusive. Our study unveils a significant upregulation of Sema3C in fibrotic liver, HCC tissues, peripheral blood of HCC patients, as well as sorafenib-resistant tissues and cells, with its overexpression correlating with the acquisition of stemness properties in HCC. We further identify NRP1 and ITGB1 as pivotal functional receptors of Sema3C, activating downstream AKT/Gli1/c-Myc signaling pathways to bolster HCC self-renewal and tumor initiation. Additionally, HCC cells-derived Sema3C facilitated extracellular matrix (ECM) contraction and collagen deposition in vivo, while also promoting the proliferation and activation of hepatic stellate cells (HSCs). Mechanistically, Sema3C interacted with NRP1 and ITGB1 in HSCs, activating downstream NF-kB signaling, thereby stimulating the release of IL-6 and upregulating HMGCR expression, consequently enhancing cholesterol synthesis in HSCs. Furthermore, CAF-secreted TGF-ß1 activates AP1 signaling to augment Sema3C expression in HCC cells, establishing a positive feedback loop that accelerates HCC progression. Notably, blockade of Sema3C effectively inhibits tumor growth and sensitizes HCC cells to sorafenib in vivo. In sum, our findings spotlight Sema3C as a novel biomarker facilitating the crosstalk between CSCs and stroma during hepatocarcinogenesis, thereby offering a promising avenue for enhancing treatment efficacy and overcoming drug resistance in HCC.


Sujet(s)
Carcinome hépatocellulaire , Tumeurs du foie , Sémaphorines , Microenvironnement tumoral , Tumeurs du foie/génétique , Tumeurs du foie/anatomopathologie , Tumeurs du foie/métabolisme , Humains , Carcinome hépatocellulaire/génétique , Carcinome hépatocellulaire/anatomopathologie , Carcinome hépatocellulaire/métabolisme , Microenvironnement tumoral/génétique , Sémaphorines/génétique , Sémaphorines/métabolisme , Antigènes CD29/génétique , Antigènes CD29/métabolisme , Souris , Transduction du signal/génétique , Cellules étoilées du foie/métabolisme , Cellules étoilées du foie/anatomopathologie , Neuropiline 1/génétique , Neuropiline 1/métabolisme , Lignée cellulaire tumorale , Cellules souches tumorales/anatomopathologie , Cellules souches tumorales/métabolisme , Animaux , Régulation de l'expression des gènes tumoraux/génétique , Sorafénib/pharmacologie , Fibroblastes associés au cancer/métabolisme , Fibroblastes associés au cancer/anatomopathologie , Évolution de la maladie
7.
Theranostics ; 14(9): 3470-3485, 2024.
Article de Anglais | MEDLINE | ID: mdl-38948063

RÉSUMÉ

Background: Sorafenib is the standard treatment for advanced hepatocellular carcinoma (HCC), but acquired resistance during the treatment greatly limits its clinical efficiency. Lipid metabolic disorder plays an important role in hepatocarcinogenesis. However, whether and how lipid metabolic reprogramming regulates sorafenib resistance of HCC cells remains vague. Methods: Sorafenib resistant HCC cells were established by continuous induction. UHPLC-MS/MS, proteomics, and flow cytometry were used to assess the lipid metabolism. ChIP and western blot were used to reflect the interaction of signal transducer and activator of transcription 3 (STAT3) with glycerol-3-phosphate acyltransferase 3 (GPAT3). Gain- and loss-of function studies were applied to explore the mechanism driving sorafenib resistance of HCC. Flow cytometry and CCK8 in vitro, and tumor size in vivo were used to evaluate the sorafenib sensitivity of HCC cells. Results: Our metabolome data revealed a significant enrichment of triglycerides in sorafenib-resistant HCC cells. Further analysis using proteomics and genomics techniques demonstrated a significant increase in the expression of GPAT3 in the sorafenib-resistant groups, which was found to be dependent on the activation of STAT3. The restoration of GPAT3 resensitized HCC cells to sorafenib, while overexpression of GPAT3 led to insensitivity to sorafenib. Mechanistically, GPAT3 upregulation increased triglyceride synthesis, which in turn stimulated the NF-κB/Bcl2 signaling pathway, resulting in apoptosis tolerance upon sorafenib treatment. Furthermore, our in vitro and in vivo studies revealed that pan-GPAT inhibitors effectively reversed sorafenib resistance in HCC cells. Conclusions: Our data demonstrate that GPAT3 elevation in HCC cells reprograms triglyceride metabolism which contributes to acquired resistance to sorafenib, which suggests GPAT3 as a potential target for enhancing the sensitivity of HCC to sorafenib.


Sujet(s)
Carcinome hépatocellulaire , Résistance aux médicaments antinéoplasiques , Tumeurs du foie , Facteur de transcription STAT-3 , Sorafénib , Sorafénib/pharmacologie , Sorafénib/usage thérapeutique , Carcinome hépatocellulaire/traitement médicamenteux , Carcinome hépatocellulaire/métabolisme , Carcinome hépatocellulaire/anatomopathologie , Tumeurs du foie/traitement médicamenteux , Tumeurs du foie/métabolisme , Tumeurs du foie/anatomopathologie , Humains , Résistance aux médicaments antinéoplasiques/effets des médicaments et des substances chimiques , Lignée cellulaire tumorale , Animaux , Facteur de transcription STAT-3/métabolisme , Souris , Antinéoplasiques/pharmacologie , Antinéoplasiques/usage thérapeutique , Souris nude , Tests d'activité antitumorale sur modèle de xénogreffe , Métabolisme lipidique/effets des médicaments et des substances chimiques , Apoptose/effets des médicaments et des substances chimiques , Régulation de l'expression des gènes tumoraux/effets des médicaments et des substances chimiques , Transduction du signal/effets des médicaments et des substances chimiques
8.
Int J Biol Sci ; 20(9): 3442-3460, 2024.
Article de Anglais | MEDLINE | ID: mdl-38993554

RÉSUMÉ

In this study, we explored the oncogenic mechanism of cleavage and polyadenylation-specific factor 6 (CPSF6) in hepatocellular carcinoma (HCC). CPSF6 was overexpressed in HCC tissues with poor survival rates compared to normal tissues. Hence, CPSF6 depletion suppressed cell viability and colony formation, induced apoptosis via PARP cleavage, and increased the sub-G1 population of Hep3B and Huh7 cells. In addition, CPSF6 enhanced the stability of c-Myc via their binding through nuclear co-localization by binding to c-Myc at the site of 258-360. Furthermore, c-Myc degradation by CPSF6 depletion was disturbed by FBW7 depletion or treatment with the proteasomal inhibitor MG132. Additionally, CPSF6 depletion suppressed the Warburg effect by inhibiting glucose, HK2, PKM2, LDH, and lactate; showed a synergistic effect with Sorafenib in Hep3B cells; and inhibited angiogenesis by tube formation and CAM assays, along with decreased expression and production of vascular endothelial growth factor (VEGF). Notably, CPSF6 depletion attenuated PD-L1 expression and increased Granzyme B levels, along with an increase in the percentage of CD4/CD8 cells in the splenocytes of BALB/c nude mice bearing Hep3B cells. Consistently, immunohistochemistry showed that CPSF6 depletion reduced the growth of Hep3B cells in BALB/c mice in orthotopic and xenograft tumor models by inhibiting tumor microenvironment-associated proteins. Overall, these findings suggest that CPSF6 enhances the Warburg effect for immune escape and angiogenesis, leading to cancer progression via c-Myc, mediated by the HK, PD-L1, and VEGF networks, with synergistic potential with sorafenib as a molecular target for liver cancer therapy.


Sujet(s)
Carcinome hépatocellulaire , Tumeurs du foie , Protéines proto-oncogènes c-myc , Transduction du signal , Humains , Tumeurs du foie/métabolisme , Tumeurs du foie/traitement médicamenteux , Lignée cellulaire tumorale , Carcinome hépatocellulaire/métabolisme , Carcinome hépatocellulaire/traitement médicamenteux , Protéines proto-oncogènes c-myc/métabolisme , Animaux , Néovascularisation pathologique/métabolisme , Souris , Sorafénib/usage thérapeutique , Sorafénib/pharmacologie , Effet Warburg en oncologie , Souris nude , Souris de lignée BALB C , Apoptose ,
9.
Mol Biol Rep ; 51(1): 799, 2024 Jul 13.
Article de Anglais | MEDLINE | ID: mdl-39001931

RÉSUMÉ

BACKGROUND: Although DNA repair mechanisms function to maintain genomic integrity, in cancer cells these mechanisms may negatively affect treatment efficiency. The strategy of targeting cancer cells via inhibiting DNA damage repair has been successfully used in breast and ovarian cancer using PARP inhibitors. Unfortunately, such strategies have not yet yielded results in liver cancer. Hepatocellular carcinoma (HCC), the most common type of liver cancer, is a treatment-resistant malignancy. Despite the development of guided therapies, treatment regimens for advanced HCC patients still fall short of the current need and significant problems such as cancer relapse with resistance still exist. In this paper, we targeted telomeric replication protein CTC1, which is responsible for telomere maintenance. METHODS: CTC expression was analyzed using tumor and matched-tissue RNA-sequencing data from TCGA and GTEx. In HCC cell lines, q-RT-PCR and Western blotting were used to detect CTC1 expression. The knock-down of CTC1 was achieved using lentiviral plasmids. The effects of CTC1 silencing on HCC cells were analyzed by flow cytometry, MTT, spheroid and colony formation assays. RESULTS: CTC1 is significantly downregulated in HCC tumor samples. However, CTC1 protein levels were higher in sorafenib-resistant cell lines compared to the parental groups. CTC1 inhibition reduced cell proliferation in sorafenib-resistant HCC cell lines and diminished their spheroid and colony forming capacities. Moreover, these cells were more sensitive to single and combined drug treatment with G4 stabilizer RHPS4 and sorafenib. CONCLUSION: Our results suggest that targeting CTC1 might be a viable option for combinational therapies designed for sorafenib resistant HCC patients.


Sujet(s)
Carcinome hépatocellulaire , Prolifération cellulaire , Résistance aux médicaments antinéoplasiques , Tumeurs du foie , Sorafénib , Humains , Carcinome hépatocellulaire/génétique , Carcinome hépatocellulaire/traitement médicamenteux , Carcinome hépatocellulaire/anatomopathologie , Carcinome hépatocellulaire/métabolisme , Tumeurs du foie/génétique , Tumeurs du foie/traitement médicamenteux , Tumeurs du foie/métabolisme , Tumeurs du foie/anatomopathologie , Prolifération cellulaire/effets des médicaments et des substances chimiques , Prolifération cellulaire/génétique , Lignée cellulaire tumorale , Sorafénib/pharmacologie , Résistance aux médicaments antinéoplasiques/génétique , Résistance aux médicaments antinéoplasiques/effets des médicaments et des substances chimiques , Protéines télomériques/métabolisme , Protéines télomériques/génétique , Régulation de l'expression des gènes tumoraux/effets des médicaments et des substances chimiques
10.
Sci Rep ; 14(1): 15538, 2024 07 05.
Article de Anglais | MEDLINE | ID: mdl-38969729

RÉSUMÉ

Drug delivery is the process or method of delivering a pharmacological product to have therapeutic effects on humans or animals. The use of nanoparticles to deliver medications to cells is driving the present surge in interest in improving human health. Green nanodrug delivery methods are based on chemical processes that are acceptable for the environment or that use natural biomaterials such as plant extracts and microorganisms. In this study, zinc oxide-superparamagnetic iron oxide-silver nanocomposite was synthesized via green synthesis method using Fusarium oxysporum fungi mycelia then loaded with sorafenib drug. The synthesized nanocomposites were characterized by UV-visibile spectroscopy, FTIR, TEM and SEM techniques. Sorafenib is a cancer treatment and is also known by its brand name, Nexavar. Sorafenib is the only systemic medication available in the world to treat hepatocellular carcinoma. Sorafenib, like many other chemotherapeutics, has side effects that restrict its effectiveness, including toxicity, nausea, mucositis, hypertension, alopecia, and hand-foot skin reaction. In our study, 40 male albino rats were given a single dose of diethyl nitrosamine (DEN) 60 mg/kg b.wt., followed by carbon tetrachloride 2 ml/kg b.wt. twice a week for one month. The aim of our study is using the zinc oxide-superparamagnetic iron oxide-silver nanocomposite that was synthesized by Fusarium oxysporum fungi mycelia as nanocarrier for enhancement the sorafenib anticancer effect.


Sujet(s)
Antinéoplasiques , Carcinome hépatocellulaire , Tumeurs du foie , Argent , Sorafénib , Oxyde de zinc , Animaux , Sorafénib/pharmacologie , Sorafénib/composition chimique , Sorafénib/administration et posologie , Oxyde de zinc/composition chimique , Oxyde de zinc/pharmacologie , Argent/composition chimique , Rats , Antinéoplasiques/pharmacologie , Antinéoplasiques/composition chimique , Antinéoplasiques/administration et posologie , Carcinome hépatocellulaire/traitement médicamenteux , Carcinome hépatocellulaire/anatomopathologie , Mâle , Tumeurs du foie/traitement médicamenteux , Tumeurs du foie/anatomopathologie , Vecteurs de médicaments/composition chimique , Fusarium/effets des médicaments et des substances chimiques , Nanoparticules de magnétite/composition chimique , Nanocomposites/composition chimique , Humains , Nanoparticules magnétiques d'oxyde de fer/composition chimique
11.
Cell Rep ; 43(7): 114487, 2024 Jul 23.
Article de Anglais | MEDLINE | ID: mdl-38996071

RÉSUMÉ

The integrated stress response (ISR) is activated in response to intrinsic and extrinsic stimuli, playing a role in tumor progression and drug resistance. The regulatory role and mechanism of ISR in liver cancer, however, remain largely unexplored. Here, we demonstrate that OTU domain-containing protein 3 (OTUD3) is a deubiquitylase of eukaryotic initiation factor 2α (eIF2α), antagonizing ISR and suppressing liver cancer. OTUD3 decreases interactions between eIF2α and the kinase EIF2ΑK3 by removing K27-linked polyubiquitylation on eIF2α. OTUD3 deficiency in mice leads to enhanced ISR and accelerated progression of N-nitrosodiethylamine-induced hepatocellular carcinoma. Additionally, decreased OTUD3 expression associated with elevated eIF2α phosphorylation correlates with the progression of human liver cancer. Moreover, ISR activation due to decreased OTUD3 expression renders liver cancer cells resistant to sorafenib, while the combined use of the ISR inhibitor ISRIB significantly improves their sensitivity to sorafenib. Collectively, these findings illuminate the regulatory mechanism of ISR in liver cancer and provide a potential strategy to counteract sorafenib resistance.


Sujet(s)
Résistance aux médicaments antinéoplasiques , Tumeurs du foie , Sorafénib , Ubiquitin-specific proteases , Sorafénib/pharmacologie , Tumeurs du foie/anatomopathologie , Tumeurs du foie/traitement médicamenteux , Tumeurs du foie/métabolisme , Tumeurs du foie/génétique , Animaux , Humains , Résistance aux médicaments antinéoplasiques/effets des médicaments et des substances chimiques , Souris , Ubiquitin-specific proteases/métabolisme , Ubiquitin-specific proteases/génétique , Évolution de la maladie , Stress physiologique/effets des médicaments et des substances chimiques , Lignée cellulaire tumorale , Ubiquitination/effets des médicaments et des substances chimiques , Carcinome hépatocellulaire/anatomopathologie , Carcinome hépatocellulaire/traitement médicamenteux , Carcinome hépatocellulaire/génétique , Carcinome hépatocellulaire/métabolisme , Facteur-2 d'initiation eucaryote/métabolisme , Phosphorylation/effets des médicaments et des substances chimiques , Souris de lignée C57BL
12.
J Cancer Res Ther ; 20(3): 755-762, 2024 Apr 01.
Article de Anglais | MEDLINE | ID: mdl-39023579

RÉSUMÉ

BACKGROUND AND OBJECTIVES: Hepatocellular carcinoma (HCC) is a primary cancer that poorly responds to treatment. Molecular cancer studies led to the development of kinase inhibitors, among which sorafenib stands out as a multi-kinase inhibitor approved by FDA for first line use in HCC patients. However, the efficiency of sorafenib was shown to be counteracted by numerous subcellular pathways involving the effector kinase AKT, causing resistance and limiting its survival benefit. On the way of breaking such resistance mechanisms and increase the efficiency of sorafenib, deeper understanding of hepatocellular physiology is essential. Thyroid hormones were shown to be metabolized in liver and inevitably affect the molecular behaviour of hepatocytes. Interestingly, thyroid hormone T3 was also demonstrated to be potentially influential in liver regeneration and treatment with this hormone reportedly led to a decrease in HCC tumor growths. In this study, we aimed to uncover the impact of T3 hormone on the cytotoxic response to sorafenib in HCC in vitro. MATERIALS AND METHODS: We pre-treated the HCC cell line Huh-7 with T3 prior to sorafenib exposure both in 2D and 3D culture. We checked cell viability with MTT assay in 2D culture and measured the sizes of 3D spheroids with bright-field microscopy followed by a surface analysis with ImageJ. We also performed scratch assay to measure cell migration as well as western blot and qPCR to uncover affected pathways. RESULTS: We observed an additive effect to sorafenib's cytotoxicity both in 2D and 3D culture. Cell migration assay also confirmed our finding and pointed out a benefit of T3 hormone in HCC cell migration. Western blot experiments showed that T3 exerts its additive effect by suppressing AKT expression upon sorafenib treatment both at protein and gene expression levels. CONCLUSION: Our results open a promising new avenue in increasing sorafenib's cytotoxicity where thyroid hormone T3 is utilized to modulate AKT expression to combat resistance, and warrant further studies in the field.


Sujet(s)
Carcinome hépatocellulaire , Régulation de l'expression des gènes tumoraux , Tumeurs du foie , Protéines proto-oncogènes c-akt , Sorafénib , Tri-iodothyronine , Humains , Antinéoplasiques/pharmacologie , Carcinome hépatocellulaire/traitement médicamenteux , Carcinome hépatocellulaire/anatomopathologie , Carcinome hépatocellulaire/métabolisme , Carcinome hépatocellulaire/génétique , Lignée cellulaire tumorale , Prolifération cellulaire/effets des médicaments et des substances chimiques , Survie cellulaire/effets des médicaments et des substances chimiques , Résistance aux médicaments antinéoplasiques/effets des médicaments et des substances chimiques , Synergie des médicaments , Régulation de l'expression des gènes tumoraux/effets des médicaments et des substances chimiques , Tumeurs du foie/traitement médicamenteux , Tumeurs du foie/anatomopathologie , Tumeurs du foie/métabolisme , Tumeurs du foie/génétique , Inhibiteurs de protéines kinases/pharmacologie , Protéines proto-oncogènes c-akt/métabolisme , Sorafénib/pharmacologie , Tri-iodothyronine/pharmacologie
13.
Cell Mol Life Sci ; 81(1): 284, 2024 Jul 05.
Article de Anglais | MEDLINE | ID: mdl-38967794

RÉSUMÉ

Hepatocellular carcinoma (HCC) is a malignancy that occurs worldwide and is generally associated with poor prognosis. The development of resistance to targeted therapies such as sorafenib is a major challenge in clinical cancer treatment. In the present study, Ten-eleven translocation protein 1 (TET1) was found to be highly expressed in sorafenib-resistant HCC cells and knockdown of TET1 can substantially improve the therapeutic effect of sorafenib on HCC, indicating the potential important roles of TET1 in sorafenib resistance in HCC. Mechanistic studies determined that TET1 and Yes-associated protein 1 (YAP1) synergistically regulate the promoter methylation and gene expression of DNA repair-related genes in sorafenib-resistant HCC cells. RNA sequencing indicated the activation of DNA damage repair signaling was extensively suppressed by the TET1 inhibitor Bobcat339. We also identified TET1 as a direct transcriptional target of YAP1 by promoter analysis and chromatin-immunoprecipitation assays in sorafenib-resistant HCC cells. Furthermore, we showed that Bobcat339 can overcome sorafenib resistance and synergized with sorafenib to induce tumor eradication in HCC cells and mouse models. Finally, immunostaining showed a positive correlation between TET1 and YAP1 in clinical samples. Our findings have identified a previously unrecognized molecular pathway underlying HCC sorafenib resistance, thus revealing a promising strategy for cancer therapy.


Sujet(s)
Carcinome hépatocellulaire , Réparation de l'ADN , Résistance aux médicaments antinéoplasiques , Régulation de l'expression des gènes tumoraux , Tumeurs du foie , Transduction du signal , Sorafénib , Animaux , Humains , Souris , Protéines adaptatrices de la transduction du signal/métabolisme , Protéines adaptatrices de la transduction du signal/génétique , Antinéoplasiques/pharmacologie , Antinéoplasiques/usage thérapeutique , Carcinome hépatocellulaire/génétique , Carcinome hépatocellulaire/traitement médicamenteux , Carcinome hépatocellulaire/métabolisme , Carcinome hépatocellulaire/anatomopathologie , Lignée cellulaire tumorale , Méthylation de l'ADN/effets des médicaments et des substances chimiques , Réparation de l'ADN/effets des médicaments et des substances chimiques , Réparation de l'ADN/génétique , Résistance aux médicaments antinéoplasiques/génétique , Épigenèse génétique/effets des médicaments et des substances chimiques , Régulation de l'expression des gènes tumoraux/effets des médicaments et des substances chimiques , Voie de signalisation Hippo , Tumeurs du foie/génétique , Tumeurs du foie/traitement médicamenteux , Tumeurs du foie/métabolisme , Tumeurs du foie/anatomopathologie , Souris de lignée BALB C , Souris nude , Mixed function oxygenases/génétique , Mixed function oxygenases/métabolisme , Protéines proto-oncogènes/métabolisme , Protéines proto-oncogènes/génétique , Transduction du signal/effets des médicaments et des substances chimiques , Sorafénib/pharmacologie , Sorafénib/usage thérapeutique , Facteurs de transcription/métabolisme , Facteurs de transcription/génétique , Tests d'activité antitumorale sur modèle de xénogreffe , Protéines de signalisation YAP/métabolisme
14.
Aging (Albany NY) ; 16(14): 11339-11358, 2024 Jul 05.
Article de Anglais | MEDLINE | ID: mdl-39029955

RÉSUMÉ

BACKGROUND: Ferroptosis is associated with cancer progression and has a promising application for treating hepatocellular carcinoma (HCC). Long non-coding RNA (lncRNA) participates widely in the regulation of ferroptosis, but the key lncRNA regulators implicated in ferroptosis and their molecular mechanisms remain to be identified. METHODS: Bioinformatic analysis was performed in R based on The Cancer Genome Atlas Program (TCGA) public database. The relative expression of genes was detected by real-time quantitative PCR. Cell viability was assessed by the CCK8 assay. The cell cycle and apoptosis were detected by flow cytometry. Migration and invasion of HCC cells were detected by Transwell assay and wound healing assay. Expression of relevant proteins was detected by Western blotting. A dual-luciferase reporter assay was used to detect interactions between PART1 (or SLC7A11) and miR-490-3p. RESULTS: The PART1/miR-490-3p/SLC7A11 axis was identified as a potential regulatory pathway of ferroptosis in HCC. PART1 silencing reduced HCC cell proliferation, migration, and metastasis and promoted apoptosis and erastin-reduced ferroptosis. Further investigation revealed that PART1 acted as a competitive endogenous RNA (ceRNA) for miR-490-3p to enhance SLC7A11 expression. Overexpression of miR-490-3p downregulated the expression of SLC7A11, inhibiting the proliferation, invasion, and metastasis of HCC cells while promoting apoptosis and erastin-induced ferroptosis. Knockdown of PART1 in HCC cells significantly improved the sensitivity of HCC cells to sorafenib. CONCLUSION: Our results revealed that the PART1/miR-490-3p/SLC7A11 axis enhances HCC cell malignancy and suppresses ferroptosis, which provides a new perspective for understanding of the function of long chain non-coding RNAs in HCC. The PART1/miR-490-3p/SLC7A11 axis may be target for improving sorafenib sensitivity in HCC.


Sujet(s)
Système y+ de transport d'acides aminés , Carcinome hépatocellulaire , Ferroptose , Régulation de l'expression des gènes tumoraux , Tumeurs du foie , microARN , ARN long non codant , Ferroptose/génétique , Humains , microARN/génétique , microARN/métabolisme , Carcinome hépatocellulaire/génétique , Carcinome hépatocellulaire/anatomopathologie , Carcinome hépatocellulaire/métabolisme , Tumeurs du foie/génétique , Tumeurs du foie/anatomopathologie , Tumeurs du foie/métabolisme , Système y+ de transport d'acides aminés/génétique , Système y+ de transport d'acides aminés/métabolisme , Lignée cellulaire tumorale , ARN long non codant/génétique , ARN long non codant/métabolisme , Prolifération cellulaire/génétique , Carcinogenèse/génétique , Mouvement cellulaire/génétique , Apoptose/génétique , Sorafénib/pharmacologie
15.
Cell Death Dis ; 15(7): 493, 2024 Jul 10.
Article de Anglais | MEDLINE | ID: mdl-38987529

RÉSUMÉ

Lung cancer is a leading cause of cancer-related mortality globally, with a dismal 5-year survival rate, particularly for Lung Adenocarcinoma (LUAD). Mechanical changes within the tumor microenvironment, such as extracellular matrix (ECM) remodeling and fibroblast activity, play pivotal roles in cancer progression and metastasis. However, the specific impact of the basement membrane (BM) on the mechanical characteristics of LUAD remains unclear. This study aims to identify BM genes influencing internal mechanical stress in tumors, elucidating their effects on LUAD metastasis and therapy resistance, and exploring strategies to counteract these effects. Using Matrigel overlay and Transwell assays, we found that mechanical stress, mimicked by matrix application, augmented LUAD cell migration and invasion, correlating with ECM alterations and activation of the epithelial-mesenchymal transition (EMT) pathway. Employing machine learning, we developed the SVM_Score model based on relevant BM genes, which accurately predicted LUAD patient prognosis and EMT propensity across multiple datasets. Lower SVM_Scores were associated with worse survival outcomes, elevated cancer-related pathways, increased Tumor Mutation Burden, and higher internal mechanical stress in LUAD tissues. Notably, the SVM_Score was closely linked to COL5A1 expression in myofibroblasts, a key marker of mechanical stress. High COL5A1 expression from myofibroblasts promoted tumor invasiveness and EMT pathway activation in LUAD cells. Additionally, treatment with Sorafenib, which targets COL5A1 secretion, attenuated the tumor-promoting effects of myofibroblast-derived COL5A1, inhibiting LUAD cell proliferation, migration, and enhancing chemosensitivity. In conclusion, this study elucidates the complex interplay between mechanical stress, ECM alterations, and LUAD progression. The SVM_Score emerges as a robust prognostic tool reflecting tumor mechanical characteristics, while Sorafenib intervention targeting COL5A1 secretion presents a promising therapeutic strategy to mitigate LUAD aggressiveness. These findings deepen our understanding of the biomechanical aspects of LUAD and offer insights for future research and clinical applications.


Sujet(s)
Adénocarcinome pulmonaire , Collagène de type V , Transition épithélio-mésenchymateuse , Tumeurs du poumon , Myofibroblastes , Contrainte mécanique , Humains , Adénocarcinome pulmonaire/anatomopathologie , Adénocarcinome pulmonaire/génétique , Adénocarcinome pulmonaire/métabolisme , Adénocarcinome pulmonaire/traitement médicamenteux , Myofibroblastes/métabolisme , Myofibroblastes/effets des médicaments et des substances chimiques , Myofibroblastes/anatomopathologie , Tumeurs du poumon/anatomopathologie , Tumeurs du poumon/métabolisme , Tumeurs du poumon/génétique , Tumeurs du poumon/traitement médicamenteux , Transition épithélio-mésenchymateuse/effets des médicaments et des substances chimiques , Collagène de type V/métabolisme , Collagène de type V/génétique , Résistance aux médicaments antinéoplasiques/effets des médicaments et des substances chimiques , Lignée cellulaire tumorale , Animaux , Mouvement cellulaire/effets des médicaments et des substances chimiques , Métastase tumorale , Souris , Microenvironnement tumoral , Sorafénib/pharmacologie , Sorafénib/usage thérapeutique , Matrice extracellulaire/métabolisme
16.
Molecules ; 29(13)2024 Jul 04.
Article de Anglais | MEDLINE | ID: mdl-38999138

RÉSUMÉ

Cancer remains a leading cause of death worldwide, often resulting from uncontrolled growth in various organs. Protein kinase inhibitors represent an important class of targeted cancer therapies. Recently, the kinases BRAF and VEGFR-2 have shown synergistic effects on tumor progression. Seeking to develop dual BRAF/VEGFR-2 inhibitors, we synthesized 18 amino-benzothiazole derivatives with structural similarities to reported dual inhibitors. Four compounds-4a, 4f, 4l, and 4r-demonstrated remarkable cytotoxicity, with IC50 values ranging from 3.58 to 15.36 µM, against three cancer cell lines. Furthermore, these compounds showed IC50 values of 38.77-66.22 µM in the case of a normal cell line, which was significantly safer than the reference, sorafenib. Subsequent investigation revealed that compound 4f exhibited the capacity to inhibit the BRAF and VEGFR-2 enzymes, with IC50 values similar to sorafenib (0.071 and 0.194 µM, respectively). Moreover, compound 4f caused G2-M- and S-phase cycle arrest. Molecular modeling demonstrated binding patterns compatible with inhibition for both targets, where 4f exerted the critical interactions in the BRAF site and interacted in the VEGFR-2 site in a manner akin to sorafenib, demonstrating affinity similar to dabrafenib.


Sujet(s)
Antinéoplasiques , Benzothiazoles , Prolifération cellulaire , Simulation de docking moléculaire , Inhibiteurs de protéines kinases , Protéines proto-oncogènes B-raf , Thiadiazoles , Récepteur-2 au facteur croissance endothéliale vasculaire , Récepteur-2 au facteur croissance endothéliale vasculaire/antagonistes et inhibiteurs , Récepteur-2 au facteur croissance endothéliale vasculaire/métabolisme , Humains , Protéines proto-oncogènes B-raf/antagonistes et inhibiteurs , Protéines proto-oncogènes B-raf/métabolisme , Inhibiteurs de protéines kinases/pharmacologie , Inhibiteurs de protéines kinases/composition chimique , Inhibiteurs de protéines kinases/synthèse chimique , Benzothiazoles/composition chimique , Benzothiazoles/pharmacologie , Benzothiazoles/synthèse chimique , Thiadiazoles/composition chimique , Thiadiazoles/pharmacologie , Thiadiazoles/synthèse chimique , Lignée cellulaire tumorale , Prolifération cellulaire/effets des médicaments et des substances chimiques , Antinéoplasiques/pharmacologie , Antinéoplasiques/composition chimique , Antinéoplasiques/synthèse chimique , Conception de médicament , Relation structure-activité , Sorafénib/pharmacologie , Sorafénib/composition chimique , Structure moléculaire , Simulation numérique , Tests de criblage d'agents antitumoraux
17.
Int Immunopharmacol ; 139: 112807, 2024 Sep 30.
Article de Anglais | MEDLINE | ID: mdl-39068757

RÉSUMÉ

OBJECTIVE: Sorafenib is a chemotherapeutic agent used to treat hepatocellular carcinoma (HCC). However, its clinical response rates are often low. Tumour-associated macrophages (TAMs) have been implicated in tumour resistance. The relationship between TAMs-derived exosomes and primary resistance to sorafenib in hepatocellular carcinoma is unclear. METHODS: The study analysed RNA-SEQ data from TCGA-LIHC to explore the relationship between TAMs and sorafenib IC50. THP-1-induced M2 macrophages were used as a model to investigate the relationship between M2 macrophage exosomes and primary resistance to sorafenib in hepatocellular carcinoma cells using apoptosis, colony generation, cell viability and dual luciferase. RESULTS: M2 macrophage score and sorafenib IC50 were positively correlated in hepatocellular carcinoma patients, M2 macrophage exosomes promoted sorafenib resistance in hepatocellular carcinoma cells, and M2-exo-miR-200c-3p facilitated the development of sorafenib resistance in hepatocellular carcinoma cells by mediating the activation of PI3K/AKT. CONCLUSION: We propose and demonstrate for the first time that M2 macrophage exosomes promote sorafenib resistance in hepatocellular carcinoma, providing a new perspective for the clinical treatment of hepatocellular carcinoma patients.


Sujet(s)
Antinéoplasiques , Carcinome hépatocellulaire , Résistance aux médicaments antinéoplasiques , Exosomes , Tumeurs du foie , microARN , Sorafénib , Sorafénib/pharmacologie , Sorafénib/usage thérapeutique , Carcinome hépatocellulaire/traitement médicamenteux , Carcinome hépatocellulaire/génétique , Humains , Tumeurs du foie/traitement médicamenteux , Tumeurs du foie/génétique , Exosomes/métabolisme , Résistance aux médicaments antinéoplasiques/génétique , microARN/génétique , microARN/métabolisme , Antinéoplasiques/pharmacologie , Antinéoplasiques/usage thérapeutique , Lignée cellulaire tumorale , Macrophages/effets des médicaments et des substances chimiques , Macrophages/immunologie , Protéines proto-oncogènes c-akt/métabolisme , Macrophages associés aux tumeurs/immunologie , Macrophages associés aux tumeurs/effets des médicaments et des substances chimiques , Macrophages associés aux tumeurs/métabolisme , Phosphatidylinositol 3-kinases/métabolisme , Régulation de l'expression des gènes tumoraux/effets des médicaments et des substances chimiques , Apoptose/effets des médicaments et des substances chimiques , Cellules THP-1
18.
Exp Cell Res ; 441(2): 114169, 2024 Aug 15.
Article de Anglais | MEDLINE | ID: mdl-39029574

RÉSUMÉ

Advanced hepatocellular carcinoma (HCC) patients have poor prognosis. As an endogenous antioxidant enzyme involved in a variety of bioprocesses, sulfiredoxin-1 (SRXN1) plays an irreplaceable role in promoting the development of tumors. However, the role and working mechanism of SRXN1 in HCC remain unclear. In this study, we confirmed that SRXN1 promoted the cell proliferation of HCC at genetic and pharmacological level, respectively. Transcriptome sequencing analysis revealed SRXN1 knockdown had a significant effect on the expression of lysosome biogenesis related genes. Further experiments validated that lysosome biogenesis and autophagic flux were enhanced after SRXN1 inhibition and reduced as SRXN1 overexpression. Mechanism study revealed that ROS accumulation induced TFEB nuclear translocation, followed by increased autophagy. Following this rationale, the combination of SRXN1 inhibitor and sorafenib demonstrated noticeable synergistic antitumor effect through the boost of ROS both in vivo and in vitro. Taken together, SRXN1 could be a potential therapeutic target for HCC therapy.


Sujet(s)
Autophagie , Facteurs de transcription à motifs basiques hélice-boucle-hélice et à glissière à leucines , Carcinome hépatocellulaire , Prolifération cellulaire , Tumeurs du foie , Lysosomes , Oxidoreductases acting on sulfur group donors , Carcinome hépatocellulaire/anatomopathologie , Carcinome hépatocellulaire/métabolisme , Carcinome hépatocellulaire/génétique , Carcinome hépatocellulaire/traitement médicamenteux , Tumeurs du foie/anatomopathologie , Tumeurs du foie/métabolisme , Tumeurs du foie/génétique , Tumeurs du foie/traitement médicamenteux , Humains , Oxidoreductases acting on sulfur group donors/génétique , Oxidoreductases acting on sulfur group donors/métabolisme , Autophagie/effets des médicaments et des substances chimiques , Autophagie/génétique , Prolifération cellulaire/effets des médicaments et des substances chimiques , Lysosomes/métabolisme , Lysosomes/effets des médicaments et des substances chimiques , Animaux , Souris , Facteurs de transcription à motifs basiques hélice-boucle-hélice et à glissière à leucines/métabolisme , Facteurs de transcription à motifs basiques hélice-boucle-hélice et à glissière à leucines/génétique , Souris nude , Espèces réactives de l'oxygène/métabolisme , Lignée cellulaire tumorale , Régulation de l'expression des gènes tumoraux/effets des médicaments et des substances chimiques , Souris de lignée BALB C , Mâle , Sorafénib/pharmacologie
19.
Nan Fang Yi Ke Da Xue Xue Bao ; 44(7): 1425-1430, 2024 Jul 20.
Article de Chinois | MEDLINE | ID: mdl-39051089

RÉSUMÉ

OBJECTIVE: To investigate whether sodium butyrate (NaB) and sorafenib synergistically induces ferroptosis to suppress proliferation of hepatocellular carcinoma cells and the possible underlying mechanisms. METHODS: CCK8 assay and colony formation assay were used to assess the effects of NaB and sorafenib, alone or in combination, on proliferation of HepG2 cells, and ferroptosis of the treated cells was detected with GSH assay and C11-BODIPY 581/591 fluorescent probe. TCGA database was used to analyze differential YAP gene expression between liver cancer and normal tissues. The effects of NaB and sorafenib on YAP and p-YAP expressions in HepG2 cells were invesitigated using Western blotting. RESULTS: NaB (2 mmol/L) significantly reduced the IC50 of sorafenib in HepG2 cells, and combination index analysis confirmed the synergy between sorafenib and NaB. The ferroptosis inhibitor Fer-1 and the YAP activator (XMU) obviously reversed the growthinhibitory effects of the combined treatment with NaB and sorafenib in HepG2 cells. The combined treatment with NaB and sorafenib, as compared with the two agents used alone, significantly inhibited colony formation of HepG2 cells, further enhanced cellular shrinkage and dispersion, and decreased intracellular GSH and lipid ROS levels, and these effects were reversed by Fer-1 and XMU. TCGA analysis revealed a higher YAP mRNA expression in liver cancer tissues than in normal liver tissues. NaB combined with sorafenib produced significantly stronger effects than the individual agents for downregulating YAP protein expression and upregulating YAP phosphorylation level in HepG2 cells. CONCLUSION: NaB combined with sorafenib synergistically inhibit hepatocellular carcinoma cell proliferation possibly by inducing ferroptosis via inhibiting YAP expression.


Sujet(s)
Acide butyrique , Carcinome hépatocellulaire , Prolifération cellulaire , Synergie des médicaments , Ferroptose , Tumeurs du foie , Sorafénib , Protéines de signalisation YAP , Humains , Sorafénib/pharmacologie , Tumeurs du foie/métabolisme , Tumeurs du foie/anatomopathologie , Tumeurs du foie/traitement médicamenteux , Carcinome hépatocellulaire/métabolisme , Carcinome hépatocellulaire/anatomopathologie , Carcinome hépatocellulaire/traitement médicamenteux , Cellules HepG2 , Ferroptose/effets des médicaments et des substances chimiques , Prolifération cellulaire/effets des médicaments et des substances chimiques , Acide butyrique/pharmacologie , Facteurs de transcription/métabolisme , Protéines adaptatrices de la transduction du signal/métabolisme , Protéines du cycle cellulaire
20.
Cancer Biol Ther ; 25(1): 2382524, 2024 Dec 31.
Article de Anglais | MEDLINE | ID: mdl-39054566

RÉSUMÉ

Thioredoxin Reductase (TrxR) functions to recycle thioredoxin (Trx) during hydroperoxide metabolism mediated by peroxiredoxins and is currently being targeted using the FDA-approved anti-rheumatic drug, auranofin (AF), to selectively sensitize cancer cells to therapy. AF treatment decreased TrxR activity and clonogenic survival in small cell lung cancer (SCLC) cell lines (DMS273 and DMS53) as well as the H727 atypical lung carcinoid cell line. AF treatment also significantly sensitized DMS273 and H727 cell lines in vitro to sorafenib, an FDA-approved multi-kinase inhibitor that depleted intracellular glutathione (GSH). The pharmacokinetic, pharmacodynamic, and safety profile of AF was examined in nude mice with DMS273 xenografts administered AF intraperitoneally at 2 mg/kg or 4 mg/kg (IP) once (QD) or twice daily (BID) for 1-5 d. Plasma levels of AF were 10-20 µM (determined by mass spectrometry of gold), and the optimal inhibition of TrxR activity was obtained at 4 mg/kg once daily, with no effect on glutathione peroxidase 1 activity. This AF treatment extended for 14 d, inhibited TrxR (>75%), and resulted in a significant prolongation of median overall survival from 19 to 23 d (p = .04, N = 30 controls, 28 AF). In this experiment, there were no observed changes in animal bodyweight, complete blood counts (CBCs), bone marrow toxicity, blood urea nitrogen, or creatinine. These results support the hypothesis that AF effectively inhibits TrxR both in vitro and in vivo in SCLC, sensitizes NETs and SCLC to sorafenib, and could be repurposed as an adjuvant therapy with targeted agents that induce disruptions in thiol metabolism.


Sujet(s)
Auranofine , Tumeurs du poumon , Phénylurées , Carcinome pulmonaire à petites cellules , Sorafénib , Thioredoxin-disulfide reductase , Tests d'activité antitumorale sur modèle de xénogreffe , Auranofine/pharmacologie , Auranofine/usage thérapeutique , Animaux , Sorafénib/pharmacologie , Sorafénib/usage thérapeutique , Thioredoxin-disulfide reductase/antagonistes et inhibiteurs , Thioredoxin-disulfide reductase/métabolisme , Tumeurs du poumon/traitement médicamenteux , Tumeurs du poumon/anatomopathologie , Tumeurs du poumon/métabolisme , Carcinome pulmonaire à petites cellules/traitement médicamenteux , Carcinome pulmonaire à petites cellules/anatomopathologie , Carcinome pulmonaire à petites cellules/métabolisme , Humains , Souris , Lignée cellulaire tumorale , Phénylurées/pharmacologie , Phénylurées/usage thérapeutique , Tumeurs neuroendocrines/traitement médicamenteux , Tumeurs neuroendocrines/anatomopathologie , Tumeurs neuroendocrines/métabolisme , Souris nude , Nicotinamide/analogues et dérivés , Nicotinamide/pharmacologie , Nicotinamide/usage thérapeutique , Antinéoplasiques/pharmacologie , Antinéoplasiques/usage thérapeutique
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE