Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 15.862
Filtrer
1.
Oncol Rep ; 52(2)2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-38963046

RÉSUMÉ

Arsenic trioxide (ATO) is expected to be a chemical drug with antitumor activity against acute promyelocytic leukemia (APL), a type of acute myeloid leukemia. In Japan, its antitumor effects were confirmed in clinical trials for APL, and it has been approved in various countries around the world. However, there have been no reports on ATO's antitumor effects on radioresistant leukemia cells, which can be developed during radiotherapy and in combination with therapeutic radiation beams. The present study sought to clarify the antitumor effect of ATO on APL cells with radiation resistance and determine its efficacy when combined with ionizing radiation (IR). The radiation­resistant HL60 (Res­HL60) cell line was generated by subjecting the native cells to 4­Gy irradiation every week for 4 weeks. The half­maximal inhibitory concentration (IC50) for cell proliferation by ATO on native cell was 0.87 µM (R2=0.67), while the IC50 for cell proliferation by ATO on Res­HL60 was 2.24 µM (R2=0.91). IR exposure increased the sub­G1 and G2/M phase ratios in both cell lines. The addition of ATO resulted in a higher population of G2/M after 24 h rather than 48 h. When the rate of change in the sub­G1 phase was examined in greater detail, the sub­G1 phase in both control cells without ATO significantly increased by exposure to IR at 24 h, but only under the condition of 2 Gy irradiation, it had continued to increase at 48 h. Res­HL60 supplemented with ATO showed a higher rate of sub­G1 change at 24 h; however, 2 Gy irradiation resulted in a decrease compared with the control. There was a significant increase in the ratio of the G2/M phase in cells after incubation with ATO for 24 h, and exposure to 2 Gy irradiation caused an even greater increase. To determine whether the inhibition of cell proliferation and cell cycle disruptions is related to reactive oxygen species (ROS) activity, intracellular ROS levels were measured with a flow cytometric assay. Although the ROS levels of Res­HL60 were higher than those of native cells in the absence of irradiation, they did not change after 0.5 or 2 Gy irradiation. Furthermore, adding ATO to Res­HL60 reduced intracellular ROS levels. These findings provide important information that radioresistant leukemia cells respond differently to the antitumor effect of ATO and the combined effect of IR.


Sujet(s)
Trioxyde d'arsenic , Composés de l'arsenic , Prolifération cellulaire , Leucémie aiguë promyélocytaire , Oxydes , Rayonnement ionisant , Humains , Trioxyde d'arsenic/pharmacologie , Leucémie aiguë promyélocytaire/traitement médicamenteux , Leucémie aiguë promyélocytaire/anatomopathologie , Leucémie aiguë promyélocytaire/radiothérapie , Prolifération cellulaire/effets des médicaments et des substances chimiques , Prolifération cellulaire/effets des radiations , Cellules HL-60 , Composés de l'arsenic/pharmacologie , Oxydes/pharmacologie , Apoptose/effets des médicaments et des substances chimiques , Apoptose/effets des radiations , Radiotolérance/effets des médicaments et des substances chimiques , Antinéoplasiques/pharmacologie , Espèces réactives de l'oxygène/métabolisme
2.
Cancer Med ; 13(13): e7332, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38967145

RÉSUMÉ

BACKGROUND: Radiotherapy (RT) is a widely utilized tumor treatment approach, while a significant obstacle in this treatment modality is the radioresistance exhibited by tumor cells. To enhance the effectiveness of RT, scientists have explored radiosensitization approaches, including the use of radiosensitizers and physical stimuli. Nevertheless, several approaches have exhibited disappointing results including adverse effects and limited efficacy. A safer and more effective method of radiosensitization involves low-intensity ultrasound (LIUS), which selectively targets tumor tissue and enhances the efficacy of radiation therapy. METHODS: This review summarized the tumor radioresistance reasons and explored LIUS potential radiosensitization mechanisms. Moreover, it covered diverse LIUS application strategies in radiosensitization, including the use of LIUS alone, ultrasound-targeted intravascular microbubble destruction, ultrasound-mediated targeted radiosensitizers delivery, and sonodynamic therapy. Lastly, the review presented the limitations and prospects of employing LIUS-RT combined therapy in clinical settings, emphasizing the need to connect research findings with practical applications. RESULTS AND CONCLUSION: LIUS employs cost-effective equipment to foster tumor radiosensitization, curtail radiation exposure, and elevate the quality of life for patients. This efficacy is attributed to LIUS's ability to utilize thermal, cavitation, and mechanical effects to overcome tumor cell resistance to RT. Multiple experimental analyses have underscored the effectiveness of LIUS in inducing tumor radiosensitization using diverse strategies. While initial studies have shown promising results, conducting more comprehensive clinical trials is crucial to confirm its safety and effectiveness in real-world situations.


Sujet(s)
Tumeurs , Radiosensibilisants , Ultrasonothérapie , Humains , Tumeurs/radiothérapie , Tumeurs/thérapie , Radiosensibilisants/usage thérapeutique , Radiosensibilisants/pharmacologie , Ultrasonothérapie/méthodes , Association thérapeutique , Animaux , Radiotolérance , Ondes ultrasonores
4.
Mol Cancer ; 23(1): 137, 2024 Jul 05.
Article de Anglais | MEDLINE | ID: mdl-38970074

RÉSUMÉ

BACKGROUND: The outcome of hepatocellular carcinoma (HCC) is limited by its complex molecular characteristics and changeable tumor microenvironment (TME). Here we focused on elucidating the functional consequences of Maternal embryonic leucine zipper kinase (MELK) in the tumorigenesis, progression and metastasis of HCC, and exploring the effect of MELK on immune cell regulation in the TME, meanwhile clarifying the corresponding signaling networks. METHODS: Bioinformatic analysis was used to validate the prognostic value of MELK for HCC. Murine xenograft assays and HCC lung metastasis mouse model confirmed the role of MELK in tumorigenesis and metastasis in HCC. Luciferase assays, RNA sequencing, immunopurification-mass spectrometry (IP-MS) and coimmunoprecipitation (CoIP) were applied to explore the upstream regulators, downstream essential molecules and corresponding mechanisms of MELK in HCC. RESULTS: We confirmed MELK to be a reliable prognostic factor of HCC and identified MELK as an effective candidate in facilitating the tumorigenesis, progression, and metastasis of HCC; the effects of MELK depended on the targeted regulation of the upstream factor miR-505-3p and interaction with STAT3, which induced STAT3 phosphorylation and increased the expression of its target gene CCL2 in HCC. In addition, we confirmed that tumor cell-intrinsic MELK inhibition is beneficial in stimulating M1 macrophage polarization, hindering M2 macrophage polarization and inducing CD8 + T-cell recruitment, which are dependent on the alteration of CCL2 expression. Importantly, MELK inhibition amplified RT-related immune effects, thereby synergizing with RT to exert substantial antitumor effects. OTS167, an inhibitor of MELK, was also proven to effectively impair the growth and progression of HCC and exert a superior antitumor effect in combination with radiotherapy (RT). CONCLUSIONS: Altogether, our findings highlight the functional role of MELK as a promising target in molecular therapy and in the combination of RT therapy to improve antitumor effect for HCC.


Sujet(s)
Carcinome hépatocellulaire , Chimiokine CCL2 , Régulation de l'expression des gènes tumoraux , Tumeurs du foie , Protein-Serine-Threonine Kinases , Microenvironnement tumoral , Tumeurs du foie/étiologie , Tumeurs du foie/anatomopathologie , Tumeurs du foie/métabolisme , Tumeurs du foie/radiothérapie , Carcinome hépatocellulaire/métabolisme , Carcinome hépatocellulaire/étiologie , Carcinome hépatocellulaire/anatomopathologie , Carcinome hépatocellulaire/radiothérapie , Humains , Animaux , Souris , Protein-Serine-Threonine Kinases/métabolisme , Protein-Serine-Threonine Kinases/génétique , Chimiokine CCL2/métabolisme , Lignée cellulaire tumorale , Radiotolérance , Pronostic , Facteur de transcription STAT-3/métabolisme , Tests d'activité antitumorale sur modèle de xénogreffe , microARN/génétique
5.
Mol Cancer ; 23(1): 139, 2024 Jul 05.
Article de Anglais | MEDLINE | ID: mdl-38970106

RÉSUMÉ

BACKGROUND: Radioresistance is the leading cause of death in advanced cervical cancer (CC). Dysregulation of RNA modification has recently emerged as a regulatory mechanism in radiation and drug resistance. We aimed to explore the biological function and clinical significance of 5-methylcytosine (m5C) in cervical cancer radiosensitivity. METHODS: The abundance of RNA modification in radiotherapy-resistant and sensitive CC specimens was quantified by liquid chromatography-tandem mass spectrometry. The essential RNA modification-related genes involved in CC radiosensitivity were screened via RNA sequencing. The effect of NSUN6 on radiosensitivity was verified in CC cell lines, cell-derived xenograft (CDX), and 3D bioprinted patient-derived organoid (PDO). The mechanisms of NSUN6 in regulating CC radiosensitivity were investigated by integrative m5C sequencing, mRNA sequencing, and RNA immunoprecipitation. RESULTS: We found a higher abundance of m5C modification in resistant CC samples, and NSUN6 was the essential m5C-regulating gene concerning radiosensitivity. NSUN6 overexpression was clinically correlated with radioresistance and poor prognosis in cervical cancer. Functionally, higher NSUN6 expression was associated with radioresistance in the 3D PDO model of cervical cancer. Moreover, silencing NSUN6 increased CC radiosensitivity in vivo and in vitro. Mechanistically, NDRG1 was one of the downstream target genes of NSUN6 identified by integrated m5C-seq, mRNA-seq, and functional validation. NSUN6 promoted the m5C modification of NDRG1 mRNA, and the m5C reader ALYREF bound explicitly to the m5C-labeled NDRG1 mRNA and enhanced NDRG1 mRNA stability. NDRG1 overexpression promoted homologous recombination-mediated DNA repair, which in turn led to radioresistance in cervical cancer. CONCLUSIONS: Aberrant m5C hypermethylation and NSUN6 overexpression drive resistance to radiotherapy in cervical cancer. Elevated NSUN6 expression promotes radioresistance in cervical cancer by activating the NSUN6/ALYREF-m5C-NDRG1 pathway. The low expression of NSUN6 in cervical cancer indicates sensitivity to radiotherapy and a better prognosis.


Sujet(s)
5-Méthyl-cytosine , Protéines du cycle cellulaire , Régulation de l'expression des gènes tumoraux , Protéines et peptides de signalisation intracellulaire , ARN messager , Radiotolérance , Tumeurs du col de l'utérus , Tumeurs du col de l'utérus/génétique , Tumeurs du col de l'utérus/métabolisme , Tumeurs du col de l'utérus/radiothérapie , Tumeurs du col de l'utérus/anatomopathologie , Humains , Femelle , Radiotolérance/génétique , 5-Méthyl-cytosine/métabolisme , 5-Méthyl-cytosine/analogues et dérivés , Protéines du cycle cellulaire/génétique , Protéines du cycle cellulaire/métabolisme , Protéines et peptides de signalisation intracellulaire/génétique , Protéines et peptides de signalisation intracellulaire/métabolisme , Animaux , Souris , ARN messager/génétique , ARN messager/métabolisme , Lignée cellulaire tumorale , Pronostic , Tests d'activité antitumorale sur modèle de xénogreffe , Methyltransferases/génétique , Methyltransferases/métabolisme
6.
Radiat Oncol ; 19(1): 84, 2024 Jun 26.
Article de Anglais | MEDLINE | ID: mdl-38926729

RÉSUMÉ

BACKGROUND: At present, it has been found that many patients have acquired resistance to radiotherapy, which greatly reduces the effect of radiotherapy and further affects the prognosis. CircRNAs is involved in the regulation of radiosensitivity of many kinds of tumor cells. Therefore, the main purpose of this study is to explore the regulatory effect of CircRNA_101491 on radiosensitivity of ESCC and its related mechanism. METHODS: We established ESCC radiation-resistant cell line (KYSE150R cell) by gradient dose method, and tested the difference of KYSE150 between KYSE150R cell and parent cell in vitro. Then, after knocking down the expression of CircRNA_101491, a series of in vitro experiments were conducted to verify the effects of CircRNA_101491 on the phenotype and radiosensitivity of KYSE150R cells, and further analyzed the related regulatory mechanism. In addition, we also used the model of transplanted tumor in nude mice to investigate the effect of CircRNA_101491 on the radiosensitivity of ESCC in vivo. RESULTS: According to a series of in vitro experiments, we confirmed that KYSE150R cells lost the epithelial phenotype and obtained interstitial cell-like phenotype, and found that CircRNA_101491 was highly expressed in KYSE150R cells. In addition, we found that knocking down the expression of CircRNA_101491 will lift the inhibition of miR-125a-5p, and then reverse the process of EMT, accelerate the process of apoptosis, thus play a role in radiosensitization. The in vivo experiment of transplanted tumor in nude mice also showed that knocking down the expression of CircRNA_101491 could enhance the radiosensitivity of ESCC. CONCLUSION: In conclusion, we confirmed that interfering with the expression of CircRNA_101491 can relieve the inhibition of miR-125a-5p, thus reverse the process of interstitial phenotype, accelerate the process of apoptosis, and enhance the radiosensitivity of ESCC.


Sujet(s)
Tumeurs de l'oesophage , Carcinome épidermoïde de l'oesophage , Souris nude , microARN , ARN circulaire , Radiotolérance , microARN/génétique , microARN/métabolisme , Animaux , ARN circulaire/génétique , Humains , Souris , Tumeurs de l'oesophage/génétique , Tumeurs de l'oesophage/anatomopathologie , Tumeurs de l'oesophage/radiothérapie , Tumeurs de l'oesophage/métabolisme , Carcinome épidermoïde de l'oesophage/génétique , Carcinome épidermoïde de l'oesophage/anatomopathologie , Carcinome épidermoïde de l'oesophage/radiothérapie , Carcinome épidermoïde de l'oesophage/métabolisme , Apoptose , Prolifération cellulaire , Régulation de l'expression des gènes tumoraux , Souris de lignée BALB C , Lignée cellulaire tumorale , Tests d'activité antitumorale sur modèle de xénogreffe , Cellules cancéreuses en culture
7.
Curr Med Sci ; 44(3): 545-553, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38900386

RÉSUMÉ

OBJECTIVE: Abnormal expression of T-lymphokine-activated killer cell-originated protein kinase (TOPK) was reported to be closely related to the resistance of prostate cancer to radiotherapy and to targeted drug resistance in lung cancer. However, the role of TOPK inhibition in enhancing radiosensitivity of colorectal cancer (CRC) cells is unclear. This study aimed to evaluate the radiosensitization of TOPK knockdown in CRC cells. METHODS: The expression of TOPK was detected in CRC tissues by immunohistochemistry, and the effect of TOPK knockdown was detected in CRC cells by Western blotting. CCK-8 and clonogenic assays were used to detect the growth and clonogenic ability of CRC cells after TOPK knockdown combined with radiotherapy in CRC cells. Furthermore, proteomic analysis showed that the phosphorylation of TOPK downstream proteins changed after radiotherapy. DNA damage was detected by the comet assay. Changes in the DNA damage response signaling pathway were analyzed by Western blotting, and apoptosis was detected by flow cytometry. RESULTS: The expression of TOPK was significantly greater in CRC tissues at grades 2-4 than in those at grade 1. After irradiation, CRC cells with genetically silenced TOPK had shorter comet tails and reduced expression levels of DNA damage response-associated proteins, including phospho-cyclin-dependent kinase 1 (p-CDK1), phospho-ataxia telangiectasia-mutated (p-ATM), poly ADP-ribose polymerase (PARP), and meiotic recombination 11 homolog 1 (MRE11). CONCLUSIONS: TOPK was overexpressed in patients with moderately to poorly differentiated CRC. Moreover, TOPK knockdown significantly enhanced the radiosensitivity of CRC cells by reducing the DNA damage response.


Sujet(s)
Apoptose , Tumeurs colorectales , Altération de l'ADN , Radiotolérance , Humains , Tumeurs colorectales/génétique , Tumeurs colorectales/radiothérapie , Tumeurs colorectales/anatomopathologie , Altération de l'ADN/effets des radiations , Radiotolérance/génétique , Radiotolérance/effets des médicaments et des substances chimiques , Lignée cellulaire tumorale , Mâle , Techniques de knock-down de gènes , Adulte d'âge moyen , Régulation de l'expression des gènes tumoraux/effets des médicaments et des substances chimiques , Transduction du signal , Femelle , Phosphorylation , Mitogen-Activated Protein Kinase Kinases
9.
Int J Mol Sci ; 25(11)2024 May 31.
Article de Anglais | MEDLINE | ID: mdl-38892269

RÉSUMÉ

We aimed to determine whether monitoring tumor-derived exosomal microRNAs (miRNAs) could be used to assess radiotherapeutic sensitivity in patients with locally advanced esophageal squamous cell carcinoma (ESCC). RNA sequencing was employed to conduct a comparative analysis of miRNA expression levels during radiotherapy, focusing on identifying miRNAs associated with progression. Electron microscopy confirmed the existence of exosomes, and co-cultivation assays and immunofluorescence validated their capacity to infiltrate macrophages. To determine the mechanism by which exosomal miR-143-3p regulates the interplay between ESCC cells and M2 macrophages, ESCC cell-derived exosomes were co-cultured with macrophages. Serum miR-143-3p and miR-223-3p were elevated during radiotherapy, suggesting resistance to radiation and an unfavorable prognosis for ESCC. Increased levels of both miRNAs independently predicted shorter progression-free survival (p = 0.015). We developed a diagnostic model for ESCC using serum microRNAs, resulting in an area under the curve of 0.751. Radiotherapy enhanced the release of miR-143-3p from ESCC cell-derived exosomes. Immune cell infiltration analysis at the Cancer Genome Atlas (TCGA) database revealed that ESCC cell-derived miR-143-3p triggered M2 macrophage polarization. Mechanistically, miR-143-3p upregulation affected chemokine activity and cytokine signaling pathways. Furthermore, ESCC cell exosomal miR-143-3p could be transferred to macrophages, thereby promoting their polarization. Serum miR-143-3p and miR-223-3p could represent diagnostic and prognostic markers for patients with ESCC undergoing radiotherapy. Unfavorable prognosis could be linked to the increased levels of ESCC cell-derived exosomal miR-143-3p, which might promote tumor progression by interacting with macrophages.


Sujet(s)
Tumeurs de l'oesophage , Carcinome épidermoïde de l'oesophage , Exosomes , Régulation de l'expression des gènes tumoraux , Macrophages , microARN , Radiotolérance , microARN/génétique , Humains , Exosomes/métabolisme , Carcinome épidermoïde de l'oesophage/génétique , Carcinome épidermoïde de l'oesophage/anatomopathologie , Carcinome épidermoïde de l'oesophage/radiothérapie , Carcinome épidermoïde de l'oesophage/métabolisme , Macrophages/métabolisme , Radiotolérance/génétique , Tumeurs de l'oesophage/génétique , Tumeurs de l'oesophage/radiothérapie , Tumeurs de l'oesophage/anatomopathologie , Tumeurs de l'oesophage/métabolisme , Lignée cellulaire tumorale , Mâle , Femelle , Adulte d'âge moyen , Pronostic , Sujet âgé , Activation des macrophages/génétique
10.
Aging (Albany NY) ; 16(11): 9727-9752, 2024 06 05.
Article de Anglais | MEDLINE | ID: mdl-38843383

RÉSUMÉ

This study explored the role of 14-3-3σ in carbon ion-irradiated pancreatic adenocarcinoma (PAAD) cells and xenografts and clarified the underlying mechanism. The clinical significance of 14-3-3σ in patients with PAAD was explored using publicly available databases. 14-3-3σ was silenced or overexpressed and combined with carbon ions to measure cell proliferation, cell cycle, and DNA damage repair. Immunoblotting and immunofluorescence (IF) assays were used to determine the underlying mechanisms of 14-3-3σ toward carbon ion radioresistance. We used the BALB/c mice to evaluate the biological behavior of 14-3-3σ in combination with carbon ions. Bioinformatic analysis revealed that PAAD expressed higher 14-3-3σ than normal pancreatic tissues; its overexpression was related to invasive clinicopathological features and a worse prognosis. Knockdown or overexpression of 14-3-3σ demonstrated that 14-3-3σ promoted the survival of PAAD cells after carbon ion irradiation. And 14-3-3σ was upregulated in PAAD cells during DNA damage (carbon ion irradiation, DNA damaging agent) and promotes cell recovery. We found that 14-3-3σ resulted in carbon ion radioresistance by promoting RPA2 and RAD51 accumulation in the nucleus in PAAD cells, thereby increasing homologous recombination repair (HRR) efficiency. Blocking the HR pathway consistently reduced 14-3-3σ overexpression-induced carbon ion radioresistance in PAAD cells. The enhanced radiosensitivity of 14-3-3σ depletion on carbon ion irradiation was also demonstrated in vivo. Altogether, 14-3-3σ functions in tumor progression and can be a potential target for developing biomarkers and treatment strategies for PAAD along with incorporating carbon ion irradiation.


Sujet(s)
Protéines 14-3-3 , Souris de lignée BALB C , Tumeurs du pancréas , Réparation de l'ADN par recombinaison , Protéines 14-3-3/métabolisme , Protéines 14-3-3/génétique , Tumeurs du pancréas/génétique , Tumeurs du pancréas/anatomopathologie , Tumeurs du pancréas/métabolisme , Tumeurs du pancréas/radiothérapie , Animaux , Humains , Souris , Lignée cellulaire tumorale , Régulation négative , Radiotolérance/génétique , Exoribonucleases/métabolisme , Exoribonucleases/génétique , Radiothérapie par ions lourds , Carbone , Prolifération cellulaire/génétique , Régulation de l'expression des gènes tumoraux , Mâle , Altération de l'ADN , Femelle
11.
Chem Biol Interact ; 398: 111089, 2024 Aug 01.
Article de Anglais | MEDLINE | ID: mdl-38823535

RÉSUMÉ

Radio-resistant triple negative breast cancer (TNBC) is resistant to conventional drugs and radiation therapy. ortho-topolin riboside (oTR) has been evaluated for its anticancer activity in several types of cancer cells. However, its anti-proliferative activity in radio-resistant TNBC cells has not yet been reported. Therefore, we investigated the anti-proliferative activity of oTR in radio-resistant TNBC cells, and performed metabolome, lipidome, transcriptome, and proteome profiling to reveal the mechanisms of the anticancer activity of oTR. oTR showed cytotoxicity against radio-resistant TNBC cells with an inhibitory concentration (IC50) value of 7.78 µM. Significantly decreased (p value < 0.05) basal and compensatory glycolysis were observed in the oTR-treated group than untreated group. Mitochondrial spare respiratory capacity, which is relevant to cell fitness and flexibility, was significantly decreased (p value < 0.05) in the oTR-treated group. The major metabolic pathways significantly altered by oTR according to metabolome, transcriptome, and proteome profiles were the glycerolipid/glycerophospholipid pathway (log2(FC) of MGLL = -0.13, log2(FC) of acylglycerol lipase = -1.35, log2(FC) of glycerol = -0.81), glycolysis (log2(FC) of EGLN1 = 0.16, log2(FC) of EGLN1 = 0.62, log2(FC) of glucose = -0.76, log2(FC) of lactate = -0.81), and kynurenine pathway (log2(FC) of KYNU = 0.29, log2(FC) of kynureninase = 0.55, log2(FC) of alanine = 0.72). Additionally, proline metabolism (log2(FC) of PYCR1 = -0.17, log2(FC) of proline = -0.73) was significantly altered in the metabolomic and transcriptomic profiles. The MAPK signaling pathway (log2(FC) of CCN1 = -0.15, log2(FC) of CCN family member 1 = -1.02) and Rap 1 signaling pathway (log2(FC) of PARD6B = -0.28, log2(FC) of PAR6B = -3.13) were also significantly altered in transcriptomic and proteomic profiles. The findings of this study revealed that oTR has anticancer activity in radio-resistant TNBC cells by affecting various metabolic pathways, suggesting the potential of oTR as a novel anticancer agent for radio-resistant TNBC patients.


Sujet(s)
Antinéoplasiques , Tumeurs du sein triple-négatives , Humains , Tumeurs du sein triple-négatives/métabolisme , Tumeurs du sein triple-négatives/traitement médicamenteux , Tumeurs du sein triple-négatives/anatomopathologie , Lignée cellulaire tumorale , Antinéoplasiques/pharmacologie , Voies et réseaux métaboliques/effets des médicaments et des substances chimiques , Femelle , Prolifération cellulaire/effets des médicaments et des substances chimiques , Transcriptome/effets des médicaments et des substances chimiques , Radiotolérance/effets des médicaments et des substances chimiques , Glycolyse/effets des médicaments et des substances chimiques , Métabolome/effets des médicaments et des substances chimiques , Multi-omique
12.
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
13.
Semin Radiat Oncol ; 34(3): 310-322, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38880540

RÉSUMÉ

Treating radioresistant and bulky tumors is challenging due to their inherent resistance to standard therapies and their large size. GRID and lattice spatially fractionated radiation therapy (simply referred to GRID RT and LRT) offer promising techniques to tackle these issues. Both approaches deliver radiation in a grid-like or lattice pattern, creating high-dose peaks surrounded by low-dose valleys. This pattern enables the destruction of significant portions of the tumor while sparing healthy tissue. GRID RT uses a 2-dimensional pattern of high-dose peaks (15-20 Gy), while LRT delivers a three-dimensional array of high-dose vertices (10-20 Gy) spaced 2-5 cm apart. These techniques are beneficial for treating a variety of cancers, including soft tissue sarcomas, osteosarcomas, renal cell carcinoma, melanoma, gastrointestinal stromal tumors (GISTs), pancreatic cancer, glioblastoma, and hepatocellular carcinoma. The specific grid and lattice patterns must be carefully tailored for each cancer type to maximize the peak-to-valley dose ratio while protecting critical organs and minimizing collateral damage. For gynecologic cancers, the treatment plan should align with the international consensus guidelines, incorporating concurrent chemotherapy for optimal outcomes. Despite the challenges of precise dosimetry and patient selection, GRID RT and LRT can be cost-effective using existing radiation equipment, including particle therapy systems, to deliver targeted high-dose radiation peaks. This phased approach of partial high-dose induction radiation therapy with standard fractionated radiation therapy maximizes immune modulation and tumor control while reducing toxicity. Comprehensive treatment plans using these advanced techniques offer a valuable framework for radiation oncologists, ensuring safe and effective delivery of therapy for radioresistant and bulky tumors. Further clinical trials data and standardized guidelines will refine these strategies, helping expand access to innovative cancer treatments.


Sujet(s)
Fractionnement de la dose d'irradiation , Tumeurs , Humains , Tumeurs/radiothérapie , Radiotolérance , Planification de radiothérapie assistée par ordinateur/méthodes
14.
Clinics (Sao Paulo) ; 79: 100415, 2024.
Article de Anglais | MEDLINE | ID: mdl-38897099

RÉSUMÉ

INTRODUCTION: Patients with Human Papillomavirus (HPV+)-associated Laryngeal Squamous Cell Carcinoma (LSCC) exhibit dramatically improved survival relative to those with HPV-Negative (HPV-) tumors. In this study, the authors aimed to investigate the radiosensitivity of all available confirmed HPV+ and HPV-LSCC cells in vitro and in vivo. METHODS: Primary LSCC cells were generated from tumor specimens obtained from patients. Real-time PCR was performed to confirm HPV infection and the expression of HPV-related genes (E6 and E7), p53, and pRB. Clonogenic survival assays, western blotting, and flow cytometry were used to assess radiation sensitivity, apoptosis, and the expression of p53 and pRB. p53 and pRB knockout cells were generated using CRISPR/Cas9 technology. RESULTS: HPV+ LSCC cells displayed enhanced radiation sensitivity compared to HPV- cells. Radiation-induced apoptosis in HPV+ LSCC cells, accompanied by increased levels of p53 and pRB. Knockout of p53 or pRB led to radiation resistance and attenuated radiation-induced apoptosis in HPV+ LSCC cells. In vivo experiments showed similar results, where knockout of p53 or pRB decreased radiosensitivity in tumor-bearing mice. CONCLUSION: The present findings demonstrated that HPV+ LSCC cells displayed obvious inherent radiation sensitivity, corresponding to increased apoptosis following radiation exposure. Mechanism study showed that the expression of p53 and pRB in HPV+ cells are required for radiation sensitivity. These findings highlight a novel mechanism by which p53 and pRB play key roles in the radiation sensitivity of HPV+ LSCC compared to HPV-LSCC.


Sujet(s)
Apoptose , Carcinome épidermoïde , Tumeurs du larynx , Infections à papillomavirus , Radiotolérance , Protéine p53 suppresseur de tumeur , Humains , Tumeurs du larynx/radiothérapie , Tumeurs du larynx/virologie , Carcinome épidermoïde/radiothérapie , Carcinome épidermoïde/virologie , Protéine p53 suppresseur de tumeur/métabolisme , Infections à papillomavirus/radiothérapie , Infections à papillomavirus/virologie , Infections à papillomavirus/complications , Apoptose/effets des radiations , Animaux , Lignée cellulaire tumorale , Réaction de polymérisation en chaine en temps réel , Mâle , Souris , Cytométrie en flux , Technique de Western , Protéine du rétinoblastome/métabolisme
15.
Cell Mol Life Sci ; 81(1): 247, 2024 Jun 03.
Article de Anglais | MEDLINE | ID: mdl-38829550

RÉSUMÉ

BACKGROUND: The high degree of intratumoral genomic heterogeneity is a major obstacle for glioblastoma (GBM) tumors, one of the most lethal human malignancies, and is thought to influence conventional therapeutic outcomes negatively. The proneural-to-mesenchymal transition (PMT) of glioma stem cells (GSCs) confers resistance to radiation therapy in glioblastoma patients. POLD4 is associated with cancer progression, while the mechanisms underlying PMT and tumor radiation resistance have remained elusive. METHOD: Expression and prognosis of the POLD family were analyzed in TCGA, the Chinese Glioma Genome Atlas (CGGA) and GEO datasets. Tumorsphere formation and in vitro limiting dilution assay were performed to investigate the effect of UCHL3-POLD4 on GSC self-renewal. Apoptosis, TUNEL, cell cycle phase distribution, modification of the Single Cell Gel Electrophoresis (Comet), γ-H2AX immunofluorescence, and colony formation assays were conducted to evaluate the influence of UCHL3-POLD4 on GSC in ionizing radiation. Coimmunoprecipitation and GST pull-down assays were performed to identify POLD4 protein interactors. In vivo, intracranial xenograft mouse models were used to investigate the molecular effect of UCHL3, POLD4 or TCID on GCS. RESULT: We determined that POLD4 was considerably upregulated in MES-GSCs and was associated with a meagre prognosis. Ubiquitin carboxyl terminal hydrolase L3 (UCHL3), a DUB enzyme in the UCH protease family, is a bona fide deubiquitinase of POLD4 in GSCs. UCHL3 interacted with, depolyubiquitinated, and stabilized POLD4. Both in vitro and in vivo assays indicated that targeted depletion of the UCHL3-POLD4 axis reduced GSC self-renewal and tumorigenic capacity and resistance to IR treatment by impairing homologous recombination (HR) and nonhomologous end joining (NHEJ). Additionally, we proved that the UCHL3 inhibitor TCID induced POLD4 degradation and can significantly enhance the therapeutic effect of IR in a gsc-derived in situ xenograft model. CONCLUSION: These findings reveal a new signaling axis for GSC PMT regulation and highlight UCHL3-POLD4 as a potential therapeutic target in GBM. TCID, targeted for reducing the deubiquitinase activity of UCHL3, exhibited significant synergy against MES GSCs in combination with radiation.


Sujet(s)
Cellules souches tumorales , Radiotolérance , Ubiquitin thiolesterase , Humains , Ubiquitin thiolesterase/métabolisme , Ubiquitin thiolesterase/génétique , Radiotolérance/génétique , Cellules souches tumorales/métabolisme , Cellules souches tumorales/anatomopathologie , Cellules souches tumorales/effets des radiations , Animaux , Souris , Lignée cellulaire tumorale , Gliome/anatomopathologie , Gliome/génétique , Gliome/radiothérapie , Gliome/métabolisme , Apoptose/génétique , Apoptose/effets des radiations , Ubiquitination , Tumeurs du cerveau/anatomopathologie , Tumeurs du cerveau/métabolisme , Tumeurs du cerveau/génétique , Tumeurs du cerveau/radiothérapie , Souris nude , Phénotype , Régulation de l'expression des gènes tumoraux , Pronostic
16.
PLoS One ; 19(6): e0304810, 2024.
Article de Anglais | MEDLINE | ID: mdl-38857267

RÉSUMÉ

This paper reports the results of gamma irradiation experiments and whole genome sequencing (WGS) performed on vegetative cells of two radiation resistant bacterial strains, Metabacillus halosaccharovorans (VITHBRA001) and Bacillus paralicheniformis (VITHBRA024) (D10 values 2.32 kGy and 1.42 kGy, respectively), inhabiting the top-ranking high background radiation area (HBRA) of Chavara-Neendakara placer deposit (Kerala, India). The present investigation has been carried out in the context that information on strategies of bacteria having mid-range resistance for gamma radiation is inadequate. WGS, annotation, COG and KEGG analyses and manual curation of genes helped us address the possible pathways involved in the major domains of radiation resistance, involving recombination repair, base excision repair, nucleotide excision repair and mismatch repair, and the antioxidant genes, which the candidate could activate to survive under ionizing radiation. Additionally, with the help of these data, we could compare the candidate strains with that of the extremely radiation resistant model bacterium Deinococccus radiodurans, so as to find the commonalities existing in their strategies of resistance on the one hand, and also the rationale behind the difference in D10, on the other. Genomic analysis of VITHBRA001 and VITHBRA024 has further helped us ascertain the difference in capability of radiation resistance between the two strains. Significantly, the genes such as uvsE (NER), frnE (protein protection), ppk1 and ppx (non-enzymatic metabolite production) and those for carotenoid biosynthesis, are endogenous to VITHBRA001, but absent in VITHBRA024, which could explain the former's better radiation resistance. Further, this is the first-time study performed on any bacterial population inhabiting an HBRA. This study also brings forward the two species whose radiation resistance has not been reported thus far, and add to the knowledge on radiation resistant capabilities of the phylum Firmicutes which are abundantly observed in extreme environment.


Sujet(s)
Rayons gamma , Génome bactérien , Radiotolérance , Radiotolérance/génétique , Rayonnement naturel , Séquençage du génome entier , Inde , Bacillus/génétique , Bacillus/effets des radiations , Bacillus/métabolisme , Réparation de l'ADN
18.
J Cell Mol Med ; 28(12): e18482, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38899556

RÉSUMÉ

Hypoxia poses a significant challenge to the effectiveness of radiotherapy in head and neck squamous cell carcinoma (HNSCC) patients, and it is imperative to discover novel approaches to overcome this. In this study, we investigated the underlying mechanisms contributing to x-ray radioresistance in HPV-negative HNSCC cells under mild hypoxic conditions (1% oxygen) and explored the potential for autophagy modulation as a promising therapeutic strategy. Our findings show that HNSCC cells exposed to mild hypoxic conditions exhibit increased radioresistance, which is largely mediated by the hypoxia-inducible factor (HIF) pathway. We demonstrate that siRNA knockdown of HIF-1α and HIF-1ß leads to increased radiosensitivity in HNSCC cells under hypoxia. Hypoxia-induced radioresistance was not attributed to differences in DNA double strand break repair kinetics, as these remain largely unchanged under normoxic and hypoxic conditions. Rather, we identify autophagy as a critical protective mechanism in HNSCC cells following irradiation under mild hypoxia conditions. Targeting key autophagy genes, such as BECLIN1 and BNIP3/3L, using siRNA sensitizes these cells to irradiation. Whilst autophagy's role in hypoxic radioresistance remains controversial, this study highlights the importance of autophagy modulation as a potential therapeutic approach to enhance the effectiveness of radiotherapy in HNSCC.


Sujet(s)
Autophagie , Hypoxie cellulaire , Radiotolérance , Carcinome épidermoïde de la tête et du cou , Humains , Autophagie/effets des radiations , Autophagie/génétique , Radiotolérance/génétique , Lignée cellulaire tumorale , Carcinome épidermoïde de la tête et du cou/radiothérapie , Carcinome épidermoïde de la tête et du cou/génétique , Carcinome épidermoïde de la tête et du cou/anatomopathologie , Carcinome épidermoïde de la tête et du cou/métabolisme , Hypoxie cellulaire/génétique , Sous-unité alpha du facteur-1 induit par l'hypoxie/métabolisme , Sous-unité alpha du facteur-1 induit par l'hypoxie/génétique , Bécline-1/métabolisme , Bécline-1/génétique , Tumeurs de la tête et du cou/radiothérapie , Tumeurs de la tête et du cou/génétique , Tumeurs de la tête et du cou/anatomopathologie , Tumeurs de la tête et du cou/métabolisme , Protéines membranaires/métabolisme , Protéines membranaires/génétique , Réparation de l'ADN/effets des radiations , Réparation de l'ADN/génétique , Petit ARN interférent/génétique , Petit ARN interférent/métabolisme , Protéines proto-oncogènes/métabolisme , Protéines proto-oncogènes/génétique , Rayons X , Cassures double-brin de l'ADN/effets des radiations , Protéines suppresseurs de tumeurs
19.
J Exp Clin Cancer Res ; 43(1): 163, 2024 Jun 11.
Article de Anglais | MEDLINE | ID: mdl-38863037

RÉSUMÉ

BACKGROUND: Triple-negative breast cancer (TNBC) is a highly aggressive form of breast cancer subtype often treated with radiotherapy (RT). Due to its intrinsic heterogeneity and lack of effective targets, it is crucial to identify novel molecular targets that would increase RT efficacy. Here we demonstrate the role of BUB1 (cell cycle Ser/Thr kinase) in TNBC radioresistance and offer a novel strategy to improve TNBC treatment. METHODS: Gene expression analysis was performed to look at genes upregulated in TNBC patient samples compared to other subtypes. Cell proliferation and clonogenic survivals assays determined the IC50 of BUB1 inhibitor (BAY1816032) and radiation enhancement ratio (rER) with pharmacologic and genomic BUB1 inhibition. Mammary fat pad xenografts experiments were performed in CB17/SCID. The mechanism through which BUB1 inhibitor sensitizes TNBC cells to radiotherapy was delineated by γ-H2AX foci assays, BLRR, Immunoblotting, qPCR, CHX chase, and cell fractionation assays. RESULTS: BUB1 is overexpressed in BC and its expression is considerably elevated in TNBC with poor survival outcomes. Pharmacological or genomic ablation of BUB1 sensitized multiple TNBC cell lines to cell killing by radiation, although breast epithelial cells showed no radiosensitization with BUB1 inhibition. Kinase function of BUB1 is mainly accountable for this radiosensitization phenotype. BUB1 ablation also led to radiosensitization in TNBC tumor xenografts with significantly increased tumor growth delay and overall survival. Mechanistically, BUB1 ablation inhibited the repair of radiation-induced DNA double strand breaks (DSBs). BUB1 ablation stabilized phospho-DNAPKcs (S2056) following RT such that half-lives could not be estimated. In contrast, RT alone caused BUB1 stabilization, but pre-treatment with BUB1 inhibitor prevented stabilization (t1/2, ~8 h). Nuclear and chromatin-enriched fractionations illustrated an increase in recruitment of phospho- and total-DNAPK, and KAP1 to chromatin indicating that BUB1 is indispensable in the activation and recruitment of non-homologous end joining (NHEJ) proteins to DSBs. Additionally, BUB1 staining of TNBC tissue microarrays demonstrated significant correlation of BUB1 protein expression with tumor grade. CONCLUSIONS: BUB1 ablation sensitizes TNBC cell lines and xenografts to RT and BUB1 mediated radiosensitization may occur through NHEJ. Together, these results highlight BUB1 as a novel molecular target for radiosensitization in women with TNBC.


Sujet(s)
Réparation de l'ADN par jonction d'extrémités , Protein-Serine-Threonine Kinases , Radiotolérance , Tumeurs du sein triple-négatives , Humains , Tumeurs du sein triple-négatives/radiothérapie , Tumeurs du sein triple-négatives/métabolisme , Tumeurs du sein triple-négatives/génétique , Tumeurs du sein triple-négatives/anatomopathologie , Animaux , Femelle , Souris , Lignée cellulaire tumorale , Protein-Serine-Threonine Kinases/métabolisme , Protein-Serine-Threonine Kinases/génétique , Tests d'activité antitumorale sur modèle de xénogreffe , Prolifération cellulaire , Régulation de l'expression des gènes tumoraux , Souris SCID
20.
Pediatr Allergy Immunol ; 35(6): e14171, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38860449

RÉSUMÉ

BACKGROUND: Artemis deficiency is an autosomal recessive disorder characterized by a combined immunodeficiency with increased cellular radiosensitivity. In this review, the clinical and genetic characteristics of 15 patients with DCLRE1C variants are presented. METHODS: The demographic, clinical, immunologic, and genetic characteristics of patients with confirmed DCLRE1C variants diagnosed between 2013 and 2023 were collected retrospectively. Three patients were evaluated for radiosensitivity by the Comet assay, compared with age- and sex-matched healthy control. RESULTS: Seven patients who had severe infections in the first 6 months of life were diagnosed with T-B-NK+ SCID (severe combined immunodeficiency). Among them, four individuals underwent transplantation, and one of those died due to post-transplant complications in early life. Eight patients had hypomorphic variants. Half of them were awaiting a suitable donor, while the other half had already undergone transplantation. The majority of patients were born into a consanguineous family (93.3%). Most patients had recurrent sinopulmonary infections (73.3%), and one patient had no other infection than an acute respiratory infection before diagnosis. Two patients (13.3%) had autoimmunity in the form of autoimmune hemolytic anemia. Growth retardation was observed in only one patient (6.6%), and no malignancy was detected in the surviving 11 patients during the median (IQR) of 21.5 (12-45) months of follow-up. Three patients who had novel variants exhibited increased radiosensitivity and compromised DNA repair, providing a potential vulnerability to malignant transformation. CONCLUSION: Early diagnosis, radiation avoidance, and careful preparation for transplantation contribute to minimizing complications, enhancing life expectancy, and improving the patient's quality of life.


Sujet(s)
Protéines de liaison à l'ADN , Radiotolérance , Immunodéficience combinée grave , Humains , Radiotolérance/génétique , Mâle , Femelle , Immunodéficience combinée grave/génétique , Immunodéficience combinée grave/thérapie , Nourrisson , Protéines de liaison à l'ADN/génétique , Enfant d'âge préscolaire , Études rétrospectives , Endonucleases/génétique , Protéines nucléaires/génétique , Enfant , Études de cohortes
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...