Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 76
Filtrer
1.
Int J Mol Sci ; 25(11)2024 May 29.
Article de Anglais | MEDLINE | ID: mdl-38892143

RÉSUMÉ

Bladder cancer (BC) is a common genitourinary malignancy that exhibits silent morbidity and high mortality rates because of a lack of diagnostic markers and limited effective treatments. Here, we evaluated the role of the lncRNA brain cytoplasmic RNA 1 (BCYRN1) in BC. We performed loss-of-function assays to examine the effects of BCYRN1 downregulation in T24 and BOY BC cells. We found that BCYRN1 downregulation significantly inhibited the proliferation, migration, invasion, and three-dimensional spheroid formation ability and induced apoptosis in BC cells. Additionally, gene set enrichment analysis (GSEA) using RNA sequences from tumor fractions showed that BCYRN1 downregulation decreased the expression of mRNAs associated with the cell cycle. These findings were supported by observations of G2/M arrest in flow cytometry assays. Finally, we examined the expression of serum exosomal BCYRN1 as a biomarker. Clinically, BCYRN1 expression in serum exosomes from patients with BC (n = 31) was significantly higher than that in healthy donors (n = 19; mean difference: 4.1-fold higher, p < 0.01). Moreover, in patients who had undergone complete resection of BC, serum exosomal BCYRN1 levels were significantly decreased (n = 8). Thus, serum exosomal BCYRN1 may be a promising diagnostic marker and therapeutic target in patients with BC.


Sujet(s)
Apoptose , Marqueurs biologiques tumoraux , Prolifération cellulaire , Exosomes , Régulation de l'expression des gènes tumoraux , ARN long non codant , Tumeurs de la vessie urinaire , Humains , Tumeurs de la vessie urinaire/génétique , Tumeurs de la vessie urinaire/sang , Tumeurs de la vessie urinaire/diagnostic , Tumeurs de la vessie urinaire/anatomopathologie , ARN long non codant/génétique , ARN long non codant/sang , Exosomes/génétique , Exosomes/métabolisme , Marqueurs biologiques tumoraux/génétique , Marqueurs biologiques tumoraux/sang , Mâle , Lignée cellulaire tumorale , Prolifération cellulaire/génétique , Apoptose/génétique , Mouvement cellulaire/génétique , Femelle , Adulte d'âge moyen , Sujet âgé
2.
Mol Oncol ; 2024 Jun 14.
Article de Anglais | MEDLINE | ID: mdl-38874588

RÉSUMÉ

Gemcitabine plus cisplatin (GC) combination chemotherapy is the primary treatment for advanced bladder cancer (BC) with unresectable or metastatic disease. However, most cases develop resistance to this therapy. We investigated whether drug resistance could be targeted through metabolic reprogramming therapies. Metabolomics analyses in our lab's gemcitabine- and cisplatin-resistant cell lines revealed increased phosphoglycerate dehydrogenase (PHGDH) expression in gemcitabine-resistant cells compared with parental cells. Isocitrate dehydrogenase 2 (IDH2) gain of function stabilized hypoxia-inducible factor1α (HIF1α) expression, stimulating aerobic glycolysis. In gemcitabine-resistant cells, elevated fumaric acid suppressed prolyl hydroxylase domain-containing protein 2/Egl nine homolog 1 (PHD2) and stabilized HIF1α expression. PHGDH downregulation or inhibition in gemcitabine-resistant BC cells inhibited their proliferation, migration, and invasion. Cisplatin-resistant cells showed elevated fatty acid metabolism, upregulating fatty acid synthase (FASN) downstream of tyrosine kinase. Using the fibroblast growth factor receptor (FGFR) tyrosine kinase inhibitor erdafitinib, we inhibited malonyl-CoA production, which is crucial for fatty acid synthesis, and thereby suppressed upregulated HIF1α expression. Combination treatment with NCT503 and erdafitinib synergistically suppressed tumor cell proliferation and induced apoptosis in vitro and in vivo. Understanding these mechanisms could enable innovative BC therapeutic strategies to be developed.

3.
Biochem Biophys Res Commun ; 718: 149981, 2024 Jul 23.
Article de Anglais | MEDLINE | ID: mdl-38735134

RÉSUMÉ

In animal cells, vacuoles are absent, but can be induced by diseases and drugs. While phosphoinositides are critical for membrane trafficking, their role in the formation of these vacuoles remains unclear. The immunosuppressive KRP203/Mocravimod, which antagonizes sphingosine-1-phosphate receptors, has been identified as having novel multimodal activity against phosphoinositide kinases. However, the impact of this novel KRP203 activity is unknown. Here, we show that KRP203 disrupts the spatial organization of phosphoinositides and induces extensive vacuolization in tumor cells and immortalized fibroblasts. The KRP203-induced vacuoles are primarily from endosomes, and augmented by inhibition of PIKFYVE and VPS34. Conversely, overexpression of PTEN decreased KRP203-induced vacuole formation. Furthermore, V-ATPase inhibition completely blunted KRP203-induced vacuolization, pointing to a critical requirement of the endosomal maturation process. Importantly, nearly a half of KRP203-induced vacuoles are significantly decorated with PI4P, a phosphoinositide typically enriched at the plasma membrane and Golgi. These results suggest a model that noncanonical spatial reorganization of phosphoinositides by KRP203 alters the endosomal maturation process, leading to vacuolization. Taken together, this study reveals a previously unrecognized bioactivity of KRP203 as a vacuole-inducing agent and its unique mechanism of phosphoinositide modulation, providing a new insight of phosphoinositide regulation into vacuolization-associated diseases and their molecular pathologies.


Sujet(s)
Endosomes , Phosphohydrolase PTEN , Phosphatidyl inositols , Vacuoles , Vacuoles/métabolisme , Vacuoles/effets des médicaments et des substances chimiques , Endosomes/métabolisme , Endosomes/effets des médicaments et des substances chimiques , Humains , Phosphatidyl inositols/métabolisme , Animaux , Phosphohydrolase PTEN/métabolisme , Phosphohydrolase PTEN/génétique , Phosphatidylinositol 3-kinases/métabolisme , Phosphatidylinositol 3-kinases de classe III/métabolisme , Phosphatidylinositol 3-kinases de classe III/génétique , Souris , Morpholines/pharmacologie , Vacuolar Proton-Translocating ATPases/métabolisme , Vacuolar Proton-Translocating ATPases/antagonistes et inhibiteurs , Vacuolar Proton-Translocating ATPases/génétique , Cytoplasme/métabolisme , Cellules HeLa , Aminopyridines , Composés hétérocycliques 3 noyaux
4.
BMC Nephrol ; 25(1): 124, 2024 Apr 08.
Article de Anglais | MEDLINE | ID: mdl-38589827

RÉSUMÉ

BACKGROUND: Kidney transplant recipients (KTRs) are at risk of severe coronavirus disease 2019 (COVID-19), and even now that Omicron subvariants have become dominant, cases of severe disease are certain to occur. The aims of this retrospective study were to evaluate the efficacy of antiviral treatment for COVID-19 and to identify risk factors for severe disease in KTRs during Omicron subvariant-dominant periods. METHODS: A total of 65 KTRs diagnosed with COVID-19 who received antiviral treatment between July 2022 and September 2023 were analyzed. Mild cases received oral molnupiravir (MP) as outpatient therapy, while moderate or worse cases received intravenous remdesivir (RDV) as inpatient therapy. In principle, mycophenolate mofetil was withdrawn and switched to everolimus. We investigated the efficacy of antiviral treatment and compared the clinical parameters of mild/moderate and severe/critical cases to identify risk factors for severe COVID-19. RESULTS: Among 65 cases, 49 were mild, 6 were moderate, 9 were severe, and 1 was of critical severity. MP was administered to 57 cases; 49 (86%) improved and 8 (14%) progressed. RDV was administered to 16 cases; 14 (87%) improved and 2 (13%) progressed. Seventeen (26%) cases required hospitalization, and none died. Comparisons of the severe/critical group (n = 10) with the mild/moderate group (n = 55) demonstrated that the severe/critical group had a significantly higher median age (64 vs. 53 years, respectively; p = 0.0252), prevalence of diabetes (70% vs. 22%, respectively; p = 0.0047) and overweight/obesity (40% vs. 11%, respectively; p = 0.0393), as well as a significantly longer median time from symptom onset to initial antiviral therapy (3 days vs. 1 day, respectively; p = 0.0026). Multivariate analysis showed that a longer time from symptom onset to initial antiviral treatment was an independent risk factor for severe COVID-19 (p = 0.0196, odds ratio 1.625, 95% confidence interval 1.081-2.441). CONCLUSION: These findings suggest that a longer time from symptom onset to initial antiviral treatment is associated with a higher risk of severe COVID-19 in KTRs. Initiating antiviral treatment as early as possible is crucial for preventing severe outcomes; this represents a valuable insight into COVID-19 management in KTRs.


Sujet(s)
COVID-19 , Cytidine/analogues et dérivés , Hydroxylamines , Transplantation rénale , Humains , Études rétrospectives , Résultat thérapeutique , Facteurs de risque , Antiviraux/usage thérapeutique , Receveurs de transplantation
5.
PLoS One ; 18(10): e0287059, 2023.
Article de Anglais | MEDLINE | ID: mdl-37819994

RÉSUMÉ

The development of diabetes mellitus (DM) after living donor kidney transplantation (KT) is a risk factor for worsening transplant kidney function, cardiac disease, and cerebrovascular disease, which may affect prognosis after KT. At our institution, all patients' glucose tolerance is evaluated perioperatively by oral glucose tolerance tests (OGTTs) at pre-KT, and 3, 6, and 12 month (mo.) after KT. We analyzed the insulinogenic index (ISI) and homeostasis model assessment beta cell (HOMA-ß) based on the immunoreactive insulin (IRI) levels to determine how glucose tolerance changed after KT in 214 patients who had not been diagnosed with DM before KT. In addition, we analyzed the body mass index (BMI) which may also influence glucose tolerance after KT. The concentration of tacrolimus (TAC) in blood was also measured as the area under the curve (AUC) to examine its effects at each sampling point. The preoperative-OGTTs showed that DM was newly diagnosed in 22 of 214 patients (10.3%) who had not been given a diagnosis of DM by the pre-KT fasting blood sugar (FBS) tests. The glucose tolerance was improved in 15 of 22 DM patients at 12 mo. after KT. ISI and IRI deteriorated only at 3 mo. after KT but improved over time. There was a trend of an inverse correlation between HOMA-ß and TAC-AUC. We also found inverse correlations between IRI and an increase in BMI from 3 to 12 mo. after KT. Early corticosteroid withdrawal or the steroid minimization protocol with tacrolimus to maintain a low level of diabetogenic tacrolimus and BMI decrease after KT used by our hospital individualizes lifestyle interventions for each patient might contribute to an improvement in post-KT glucose tolerance.


Sujet(s)
Diabète de type 2 , Insulinorésistance , Transplantation rénale , Humains , Tacrolimus , Insuline , Transplantation rénale/effets indésirables , Peuples d'Asie de l'Est , Diabète de type 2/étiologie , Glucose , Stéroïdes , Poids , Glycémie
6.
Cancer Sci ; 114(10): 3946-3956, 2023 Oct.
Article de Anglais | MEDLINE | ID: mdl-37545017

RÉSUMÉ

Multitargeted receptor tyrosine kinase inhibitors, including vascular endothelial growth factor (VEGF) inhibitors, such as sunitinib, have been used as the primary targeted agents for patients with recurrent or distant metastasis of advanced renal cell carcinoma (RCC). However, endogenous or acquired sunitinib resistance has become a significant therapeutic problem. Therefore, we focused on mechanisms of sunitinib resistance in RCC. First, we undertook RNA sequencing analysis using previously established sunitinib-resistant RCC (SUR-Caki1, SUR-ACHN, and SUR-A498) cells. The results showed increased expression of secretogranin II (SCG2, chromogranin C) in SUR-RCC cells compared to parental cells. The Cancer Genome Atlas database showed that SCG2 expression was increased in RCC compared to normal renal cells. In addition, the survival rate of the SCG2 high-expression group was significantly lower than that of the RCC low-expression group. Thus, we investigated the involvement of SCG2 in sunitinib-resistant RCC. In vitro analysis showed that migratory and invasive abilities were suppressed by SCG2 knockdown SUR cells. As SCG2 was previously reported to be associated with angiogenesis, we undertook a tube formation assay. The results showed that suppression of SCG2 inhibited angiogenesis. Furthermore, coimmunoprecipitation assays revealed a direct interaction between SCG2 and hypoxia-inducible factor 1α (HIF1α). Expression levels of VEGF-A and VEGF-C downstream of HIF1α were found to be decreased in SCG2 knockdown SUR cells. In conclusion, SCG2 could be associated with sunitinib resistance through VEGF regulation in RCC cells. These findings could lead to a better understanding of the VHL/HIF/VEGF pathway and the development of new therapeutic strategies for sunitinib-resistant RCC.

7.
FEBS Open Bio ; 13(6): 1056-1066, 2023 06.
Article de Anglais | MEDLINE | ID: mdl-37079001

RÉSUMÉ

Combination chemotherapy with gemcitabine and cisplatin (GC) is recommended as the primary treatment for advanced bladder cancer (BC). However, the benefits of this approach are limited owing to the acquisition of drug resistance. Here, we found that gemcitabine-resistant and cisplatin-resistant BCs do not exhibit cross-resistance, and that these BCs exhibit different mRNA patterns, as revealed using RNA sequence analysis. To overcome drug resistance, we used the newly developed pan-RAS inhibitor Compound 3144. Compound 3144 inhibited cell viability through suppression of RAS-dependent signaling in gemcitabine- and cisplatin-resistant BCs. RNA sequencing revealed that several genes and pathways, particularly those related to the cell cycle, were significantly downregulated in Compound 3144-treated BCs. These findings provide insights into potential therapeutic strategies for treating BC.


Sujet(s)
Antinéoplasiques , Tumeurs de la vessie urinaire , Humains , , Cisplatine , Résistance aux médicaments antinéoplasiques/génétique , Antinéoplasiques/pharmacologie , Antinéoplasiques/usage thérapeutique , Tumeurs de la vessie urinaire/traitement médicamenteux , Tumeurs de la vessie urinaire/génétique , Désoxycytidine/pharmacologie , Désoxycytidine/usage thérapeutique
8.
Biochem Biophys Res Commun ; 630: 71-76, 2022 11 19.
Article de Anglais | MEDLINE | ID: mdl-36150242

RÉSUMÉ

Exosomes are 40-100 nm nano-sized extracellular vesicles and are receiving increasing attention as novel structures that participate in intracellular communication. We previously found that miRNA-1 (miR-1) functions as a tumor suppressor in renal cell carcinoma (RCC). In this study, we investigated the function of exosomal miR-1 and the possibility that the exosome constitutes a tumor maker in RCC. First, we established the method to collect exosomes from cell lysates and human serum by a spin column-based method. Next, we assessed exosomes using Nanosight nanoparticle tracking analysis and Western blot analysis with exosome marker CD63. We confirmed that exosomes labeled with PKH26 fused with recipient cells. Moreover, miR-1 expression was elevated in RCC cells treated with exosomes derived from miR-1-transfected cells. Functional analyses showed that exosomal miR-1 significantly inhibited cell proliferation, migration and invasion compared to control treatment. Our analyses with TCGA database of RCCs showed that miR-1 expression was significantly downregulated in clinical RCC samples compared to that in normal kidney samples, and patients with low miR-1 expression had poorer overall survival in comparison to patients with high expression. Furthermore, RNA sequence analyses showed that expression levels of several genes were altered by exposure to exosomal miR-1. The analyses with TCGA database indicated that high expression of MYO15A was associated with a poorer outcome in RCC. In addition, RT-qPCR analysis of exosomes from clinical patients' sera showed that MYO15A was significantly upregulated in RCC patients compared to that in healthy controls. This study showed that treatment with exosomal miR-1 might be an effective approach to treating RCCs. In addition, exosomal MYO15A could be a diagnostic tumor marker in RCCs.


Sujet(s)
Néphrocarcinome , Exosomes , Tumeurs du rein , microARN , Marqueurs biologiques tumoraux/métabolisme , Néphrocarcinome/diagnostic , Néphrocarcinome/génétique , Lignée cellulaire tumorale , Exosomes/métabolisme , Humains , Tumeurs du rein/diagnostic , Tumeurs du rein/génétique , microARN/métabolisme , Myosines/métabolisme
9.
Int J Nephrol ; 2022: 3060647, 2022.
Article de Anglais | MEDLINE | ID: mdl-35910424

RÉSUMÉ

Objectives: We analyzed the clinical outcomes of laparoscopic adrenalectomy for pheochromocytomas in hemodialysis compared with nonhemodialysis patients. Methods: Fifty-seven patients (7 hemodialysis and 50 nonhemodialysis) were included in the study. We analyzed the differences in clinical parameters and outcomes between the hemodialysis patient groups and nonhemodialysis patient groups as well as identified predictors for an intraoperative hypertensive spike. Results: The increasing intravascular volume before surgery in hemodialysis patients made perioperative hemodynamic management safer. No significant difference in clinical parameters between the two groups was observed except for the length of hospitalization that was significantly longer in the hemodialysis patients (9 vs. 6 days, P=0.005). An increase in systolic blood pressure at CO2 insufflation was an independent predictor of a hypertensive spike with a cutoff value of 22.5 mmHg (odds ratio 1.038, 95% confidence interval 1.012-1.078). Conclusion: Laparoscopic adrenalectomy for pheochromocytomas in hemodialysis was safe and feasible. An increase in systolic blood pressure at CO2 insufflation was a predictor of the intraoperative hypertensive spike. The research in this manuscript is not registered. This is a retrospective study.

10.
Biochem Biophys Res Commun ; 611: 99-106, 2022 06 30.
Article de Anglais | MEDLINE | ID: mdl-35487063

RÉSUMÉ

In recent years, cancer metabolism has attracted attention as a therapeutic target, and glutamine metabolism is considered one of the most important metabolic processes in cancer. Solute carrier family 1 member 5 (SLC1A5) is a sodium channel that functions as a glutamine transporter. In various cancer types, SLC1A5 gene expression is enhanced, and cancer cell growth is suppressed by inhibition of SLC1A5. However, the involvement of SLC1A5 in clear cell renal cell carcinoma (ccRCC) is unclear. Therefore, in this study, we evaluated the clinical importance of SLC1A5 in ccRCC using The Cancer Genome Atlas database. Our findings confirmed that SLC1A5 was a prognosis factor for poor survival in ccRCC. Furthermore, loss-of-function assays using small interfering RNAs or an SLC1A5 inhibitor (V9302) in human ccRCC cell lines (A498 and Caki1) showed that inhibition of SLC1A5 significantly suppressed tumor growth, invasion, and migration. Additionally, inhibition of SLC1A5 by V9302 in vivo significantly suppressed tumor growth, and the antitumor effects of SLC1A5 inhibition were related to cellular senescence. Our findings may improve our understanding of ccRCC and the development of new treatment strategies for ccRCC.


Sujet(s)
Système ASC de transport d'acides aminés , Néphrocarcinome , Vieillissement de la cellule , Tumeurs du rein , Antigènes mineurs d'histocompatibilité , Système ASC de transport d'acides aminés/génétique , Système ASC de transport d'acides aminés/métabolisme , Néphrocarcinome/génétique , Lignée cellulaire tumorale , Prolifération cellulaire , Régulation de l'expression des gènes tumoraux , Glutamine/métabolisme , Humains , Tumeurs du rein/génétique , Antigènes mineurs d'histocompatibilité/génétique , Petit ARN interférent/génétique
11.
Mol Oncol ; 16(6): 1329-1346, 2022 03.
Article de Anglais | MEDLINE | ID: mdl-35148461

RÉSUMÉ

Patients with advanced bladder cancer are generally treated with a combination of chemotherapeutics, including gemcitabine, but the effect is limited due to acquisition of drug resistance. Thus, in this study, we investigated the mechanism of gemcitabine resistance. First, gemcitabine-resistant cells were established and resistance confirmed in vitro and in vivo. Small RNA sequencing analyses were performed to search for miRNAs involved in gemcitabine resistance. miR-99a-5p, selected as a candidate miRNA, was downregulated compared to its parental cells. In gain-of-function studies, miR-99a-5p inhibited cell viabilities and restored sensitivity to gemcitabine. RNA sequencing analysis was performed to find the target gene of miR-99a-5p. SMARCD1 was selected as a candidate gene. Dual-luciferase reporter assays showed that miR-99a-5p directly regulated SMARCD1. Loss-of-function studies conducted with si-RNAs revealed suppression of cell functions and restoration of gemcitabine sensitivity. miR-99a-5p overexpression and SMARCD1 knockdown also suppressed gemcitabine-resistant cells in vivo. Furthermore, ß-galactosidase staining showed that miR-99a-5p induction and SMARCD1 suppression contributed to cellular senescence. In summary, tumor-suppressive miR-99a-5p induced cellular senescence in gemcitabine-resistant bladder cancer cells by targeting SMARCD1.


Sujet(s)
microARN , Tumeurs de la vessie urinaire , Lignée cellulaire tumorale , Prolifération cellulaire , Vieillissement de la cellule/génétique , Protéines chromosomiques nonhistones/génétique , Désoxycytidine/analogues et dérivés , Régulation de l'expression des gènes tumoraux , Humains , microARN/génétique , Tumeurs de la vessie urinaire/traitement médicamenteux , Tumeurs de la vessie urinaire/génétique ,
12.
Investig Clin Urol ; 62(2): 166-171, 2021 03.
Article de Anglais | MEDLINE | ID: mdl-33660443

RÉSUMÉ

PURPOSE: Preoperative deep vein thrombosis (pre-DVT) is a risk of symptomatic venous thromboembolism (VTE) and a serious postoperative surgical complication. However, little is known about pre-DVT in patients undergoing surgery. This study aimed to investigate the incidence and screening criteria of pre-DVT in patients undergoing urological surgery. MATERIALS AND METHODS: Between 2015 and 2017, 320 patients admitted to our hospital for urological surgery were included in this retrospective study. All patients underwent preoperative D-dimer testing. Patients with elevated D-dimer (≥1.0 µg/mL) levels underwent lower-limb compression ultrasonography (CUS). Clinical parameters were analyzed as predictors of pre-DVT, and modest cutoff value of D-dimer to predict pre-DVT were evaluated. RESULTS: Of 320 patients, preoperative elevated D-dimer levels and DVT were found in 81 (25.3%) and 20 (6.3%) patients, respectively. The positive predictive value (PPV) was 24.7% (20/81). ROC curve analysis revealed a cutoff D-dimer level of 1.8 µg/mL, yielding a PPV of 40.7% for pre-DVT among patients with elevated D-dimer levels. Preoperative DVT was detected in 16 (7.6%, n=210) patients with malignancy, 3 (5.7%, n=53) with adrenal tumors, and in 1 (1.8%, n=57) kidney donor. An age of >70 years was significantly associated with risk for pre-DVT (odds ratio, 2.81; 95% confidence interval, 1.12-7.19; p=0.0270). During a postoperative follow-up period of 90 days, no patient developed symptomatic VTE. CONCLUSIONS: The incidence of pre-DVT was 6.3% in patients undergoing urological surgery. Elderly patients and/or a cutoff D-dimer level of 1.8 µg/mL might be good indications for pre-DVT screening by CUS.


Sujet(s)
Procédures de chirurgie urologique , Thrombose veineuse/diagnostic , Adulte , Sujet âgé , Femelle , Humains , Incidence , Mâle , Adulte d'âge moyen , Valeur prédictive des tests , Période préopératoire , Études rétrospectives , Thrombose veineuse/épidémiologie
13.
BMC Cancer ; 21(1): 48, 2021 Jan 11.
Article de Anglais | MEDLINE | ID: mdl-33430801

RÉSUMÉ

BACKGROUND: Cisplatin-based chemotherapy is recommended as the primary treatment for advanced bladder cancer (BC) with unresectable or metastatic disease. However, the benefits are limited due to the acquisition of drug resistance. The mechanisms of resistance remain unclear. Although there are some reports that some molecules are associated with cisplatin resistance in advanced BC, those reports have not been fully investigated. Therefore, we undertook a new search for cisplatin resistance-related genes targeted by tumor suppressive microRNAs as well as genes that were downregulated in cisplatin-resistant BC cells and clinical BC tissues. METHODS: First, we established cisplatin-resistant BOY and T24 BC cell lines (CDDP-R-BOY, CDDP-R-T24). Then, Next Generation Sequence analysis was performed with parental and cisplatin-resistant cell lines to search for the microRNAs responsible for cisplatin resistance. We conducted gain-of-function analysis of microRNAs and their effects on cisplatin resistance, and we searched target genes comprehensively using Next Generation mRNA sequences. RESULTS: A total of 28 microRNAs were significantly downregulated in both CDDP-R-BOY and CDDP-R-T24. Among them, miR-486-5p, a tumor suppressor miRNA, was negatively correlated with the TNM classification of clinical BC samples in The Cancer Genome Atlas (TCGA) database. Transfection of miRNA-486-5p significantly inhibited cancer cell proliferation, migration, and invasion, and also improved the cells' resistance to cisplatin. Among the genes targeted by miRNA-486-5p, we focused on enoyl-CoA, hydratase/3-hydroxyacyl CoA dehydrogenase (EHHADH), which is involved in the degradation of fatty acids. EHHADH was directly regulated by miRNA-486-5p as determined by a dual-luciferase reporter assay. Loss-of-function study using EHHADH si-RNA showed significant inhibitions of cell proliferation, migration, invasion and the recovery of cisplatin sensitivity. CONCLUSION: Identification of EHHADH as a target of miRNA-486-5p provides novel insights into the potential mechanisms of cisplatin resistance in BC.


Sujet(s)
Marqueurs biologiques tumoraux/métabolisme , Cisplatine/pharmacologie , Résistance aux médicaments antinéoplasiques , Régulation de l'expression des gènes tumoraux , microARN/génétique , Enzyme bifonctionnelle péroxysomique/métabolisme , Tumeurs de la vessie urinaire/anatomopathologie , Animaux , Antinéoplasiques/pharmacologie , Apoptose , Marqueurs biologiques tumoraux/génétique , Mouvement cellulaire , Prolifération cellulaire , Femelle , Humains , Souris , Souris de lignée BALB C , Souris nude , Enzyme bifonctionnelle péroxysomique/génétique , Cellules cancéreuses en culture , Tumeurs de la vessie urinaire/génétique , Tumeurs de la vessie urinaire/métabolisme , Tests d'activité antitumorale sur modèle de xénogreffe
15.
PLoS One ; 15(7): e0236119, 2020.
Article de Anglais | MEDLINE | ID: mdl-32667929

RÉSUMÉ

The alcohol-abuse drug disulfiram has antitumor effects against diverse cancer types via inhibition of the ubiquitin-proteasome protein nuclear protein localization protein 4 (NPL4). However, the antitumor effects of NPL4 and disulfiram in clear cell renal cell carcinoma (ccRCC) are unclear. Here, we evaluated the therapeutic potential of targeting the ubiquitin-proteasome pathway using disulfiram and RNA interference and investigated the mechanisms underlying disulfiram in ccRCC. According to data from The Cancer Genome Atlas, NPL4 mRNA expression was significantly upregulated in clinical ccRCC samples compared with that in normal kidney samples, and patients with high NPL4 expression had poor overall survival compared with patients with low NPL4 expression. Disulfiram and NPL4 siRNA inhibited ccRCC cell proliferation in vitro, and disulfiram inhibited ccRCC tumor growth in a xenograft model. Synergistic antiproliferative effects were observed for combination treatment with disulfiram and sunitinib in vitro and in vivo. In RCC cells from mice treated with disulfiram and/or sunitinib, several genes associated with serine biosynthesis and aldose reductase were downregulated in cells treated with disulfiram or sunitinib alone and further downregulated in cells treated with both disulfiram and sunitinib. These findings provided insights into the mechanisms of disulfiram and suggested novel therapeutic strategies for RCC treatment.


Sujet(s)
Néphrocarcinome/traitement médicamenteux , Disulfirame/pharmacologie , Repositionnement des médicaments/méthodes , Résistance aux médicaments antinéoplasiques/effets des médicaments et des substances chimiques , Tumeurs du rein/traitement médicamenteux , Protéines nucléaires/antagonistes et inhibiteurs , Inhibiteurs de l'acétaldéhyde déshydrogénase/pharmacologie , Animaux , Apoptose , Marqueurs biologiques tumoraux/génétique , Marqueurs biologiques tumoraux/métabolisme , Néphrocarcinome/métabolisme , Néphrocarcinome/anatomopathologie , Prolifération cellulaire , Femelle , Régulation de l'expression des gènes tumoraux , Humains , Tumeurs du rein/métabolisme , Tumeurs du rein/anatomopathologie , Souris , Souris de lignée BALB C , Souris nude , Protéines nucléaires/génétique , Protéines nucléaires/métabolisme , Pronostic , Petit ARN interférent/génétique , Cellules cancéreuses en culture , Tests d'activité antitumorale sur modèle de xénogreffe
16.
Mol Oncol ; 14(9): 2190-2202, 2020 09.
Article de Anglais | MEDLINE | ID: mdl-32386122

RÉSUMÉ

d-3-Phosphoglycerate dehydrogenase (PHGDH) conducts an important step in the synthesis of serine. Importantly, the PHGDH gene is often amplified in certain cancers. Our previous studies revealed that PHGDH gene amplification was associated with poor overall survival in clear cell renal cell carcinoma (ccRCC) and that metabolic reprogramming of serine synthesis through PHGDH recruitment allowed ccRCC cells to survive in unfavorable environments. There have been no investigations of the role of PHGDH expression in bladder cancer (BC). In this investigation, we examined the clinical importance of PHDGH in BC. Furthermore, we asked whether PHGDH expression could be exploited for BC therapy. Finally, we investigated the regulatory mechanisms that modulated the expression of PHGDH. Using data from The Cancer Genome Atlas, we found that patients with high-grade BC had significantly higher PHGDH expression levels than did those with low-grade BC. In addition, patients with high PHGDH expression did not survive as long as those with low expression. PHGDH downregulation by si-RNAs or an inhibitor in BC cell lines significantly inhibited proliferative ability and induced apoptosis. Furthermore, combined treatment using a PHGDH inhibitor and gemcitabine/cisplatin achieved synergistic tumor suppression compared to use of a single agent both in vitro as well as in vivo. Mechanistic analyses of PHGDH regulation showed that PHGDH expression might be associated with DNA copy number and hypomethylation in BC. These findings suggest novel therapeutic strategies could be used in BC. Finally, our data enhance our understanding of the role of PHGDH in BC.


Sujet(s)
Cisplatine/usage thérapeutique , Méthylation de l'ADN/génétique , Désoxycytidine/analogues et dérivés , Régulation de l'expression des gènes tumoraux , Thérapie moléculaire ciblée , Phosphoglycerate dehydrogenase/génétique , Régions promotrices (génétique)/génétique , Tumeurs de la vessie urinaire/traitement médicamenteux , Tumeurs de la vessie urinaire/génétique , Animaux , Antinéoplasiques/pharmacologie , Antinéoplasiques/usage thérapeutique , Lignée cellulaire tumorale , Cisplatine/pharmacologie , Désoxycytidine/pharmacologie , Désoxycytidine/usage thérapeutique , Régulation négative/effets des médicaments et des substances chimiques , Régulation négative/génétique , Femelle , Régulation de l'expression des gènes codant pour des enzymes/effets des médicaments et des substances chimiques , Régulation de l'expression des gènes tumoraux/effets des médicaments et des substances chimiques , Humains , Souris de lignée BALB C , Souris nude , Phosphoglycerate dehydrogenase/antagonistes et inhibiteurs , Phosphoglycerate dehydrogenase/métabolisme , ARN messager/génétique , ARN messager/métabolisme , Tumeurs de la vessie urinaire/enzymologie ,
17.
PLoS One ; 15(5): e0232545, 2020.
Article de Anglais | MEDLINE | ID: mdl-32379831

RÉSUMÉ

Exosomes are 40-100 nm nano-sized extracellular vesicles. They are released from many cell types and move into the extracellular space, thereby transferring their components to recipient cells. Exosomes are receiving increasing attention as novel structures participating in intracellular communication. RAB27B is one of the leading proteins involved in exosome secretion, and oncogenic effects have been reported in several cancers. In recent years, molecularly targeted agents typified by sunitinib are widely used for the treatment of metastatic or recurrent renal cell carcinoma (RCC). However, intrinsic or acquired resistance to sunitinib has become a major issue. The present study aimed to elucidate the role of RAB27B in RCC including sunitinib-resistant and its role in exosomes. Bioinformatic analyses revealed that high expression of RAB27B correlates with progression of RCC. The expression of RAB27B protein in RCC cell lines was significantly enhanced compared with that in normal kidney cell lines. Furthermore, RAB27B protein expression was enhanced in all of the tested sunitinib-resistant RCC cell lines compared to parental cells. Although no specific effect of RAB27B on exosomes was identified in RCC cells, loss-of-function studies demonstrated that knockdown of RAB27B suppressed cell proliferation, migration and invasive activities. Moreover, anti-tumor effects of RAB27B downregulation were also observed in sunitinib-resistant RCC cells. RNA sequence and pathway analysis suggested that the oncogenic effects of RAB27B might be associated with MAPK and VEGF signaling pathways. These results showed that RAB27B is a prognostic marker and a novel therapeutic target in sunitinib-sensitive and -resistant RCCs. Further analyses should improve our understanding of sunitinib resistance in RCC.


Sujet(s)
Antinéoplasiques/usage thérapeutique , Néphrocarcinome/métabolisme , Exosomes/métabolisme , Tumeurs du rein/métabolisme , Sunitinib/usage thérapeutique , Protéines G rab/métabolisme , Technique de Western , Néphrocarcinome/traitement médicamenteux , Lignée cellulaire tumorale , Résistance aux médicaments antinéoplasiques , Humains , Rein/métabolisme , Tumeurs du rein/traitement médicamenteux
18.
Cancer Sci ; 111(5): 1607-1618, 2020 May.
Article de Anglais | MEDLINE | ID: mdl-32232883

RÉSUMÉ

Sunitinib, a multitargeted receptor tyrosine kinase inhibitor including vascular endothelial growth factor, has been widely used as a first-line treatment against metastatic renal cell carcinoma (mRCC). However, mRCC often acquires resistance to sunitinib, rendering it difficult to treat with this agent. Recently, Rapalink-1, a drug that links rapamycin and the mTOR kinase inhibitor MLN0128, has been developed with excellent therapeutic effects against breast cancer cells carrying mTOR resistance mutations. The aim of the present study was to evaluate the in vitro and in vivo therapeutic efficacy of Rapalink-1 against renal cell carcinoma (RCC) compared to temsirolimus, which is commonly used as a small molecule inhibitor of mTOR and is a derivative of rapamycin. In comparison with temsirolimus, Rapalink-1 showed significantly greater effects against proliferation, migration, invasion and cFolony formation in sunitinib-naïve RCC cells. Inhibition was achieved through suppression of the phosphorylation of substrates in the mTOR signal pathway, such as p70S6K, eukaryotic translation initiation factor 4E-binding protein 1 (4EBP1) and AKT. In addition, Rapalink-1 had greater tumor suppressive effects than temsirolimus against the sunitinib-resistant 786-o cell line (SU-R 786-o), which we had previously established, as well as 3 additional SU-R cell lines established here. RNA sequencing showed that Rapalink-1 suppressed not only the mTOR signaling pathway but also a part of the MAPK signaling pathway, the ErbB signaling pathway and ABC transporters that were associated with resistance to several drugs. Our study suggests the possibility of a new treatment option for patients with RCC that is either sunitinib-sensitive or sunitinib-resistant.


Sujet(s)
Néphrocarcinome/traitement médicamenteux , Résistance aux médicaments antinéoplasiques/effets des médicaments et des substances chimiques , Tumeurs du rein/traitement médicamenteux , Inhibiteurs de protéines kinases/pharmacologie , Sirolimus/analogues et dérivés , Sunitinib/pharmacologie , Sérine-thréonine kinases TOR/antagonistes et inhibiteurs , Animaux , Apoptose/effets des médicaments et des substances chimiques , Néphrocarcinome/métabolisme , Néphrocarcinome/anatomopathologie , Cycle cellulaire/effets des médicaments et des substances chimiques , Lignée cellulaire tumorale , Mouvement cellulaire/effets des médicaments et des substances chimiques , Prolifération cellulaire/effets des médicaments et des substances chimiques , Femelle , Humains , Tumeurs du rein/métabolisme , Tumeurs du rein/anatomopathologie , Souris , Souris nude , Inhibiteurs de protéines kinases/usage thérapeutique , Transduction du signal/effets des médicaments et des substances chimiques , Sirolimus/pharmacologie , Sirolimus/usage thérapeutique , Sunitinib/usage thérapeutique , Sérine-thréonine kinases TOR/métabolisme
20.
Jpn J Clin Oncol ; 50(1): 66-72, 2020 Jan 24.
Article de Anglais | MEDLINE | ID: mdl-31665352

RÉSUMÉ

BACKGROUND: Patients with advanced high-risk prostate cancer (PCa) are prone to have worse pathological diagnoses of positive surgical margins and/or lymph node invasion, resulting in early biochemical recurrence (BCR) despite having undergone radical prostatectomy (RP). Therefore, it is controversial whether patients with high-risk PCa should undergo RP. The purpose of this study was to evaluate the efficacy of neoadjuvant chemohormonal therapy (NAC) followed by "extended" RP. METHODS: A total of 87 patients with high-risk PCa prospectively underwent extended RP after NAC; most of the patients underwent 6 months of estramustine phosphate (EMP) 140 mg twice daily, along with a luteinizing hormone-releasing hormone agonist/antagonist. We developed our surgical technique to reduce the rate of positive surgical margins. We aimed to approach the muscle layer of the rectum by dissecting the mesorectal fascia and continuing the dissection through the mesorectum until the muscle layer of the rectum was exposed. RESULTS: More than 1 year had elapsed after surgery in all 86 patients, with a median follow-up period of 37.7 months. The 3-year BCR-free survival was 74.9%. Multivariate Cox-regression analysis revealed that a positive core ratio of 50% or greater and pathological stage of pT3 or greater were independent predictors for BCR. About 17 of 23 cases received salvage androgen deprivation therapy and concurrent external beam radiotherapy, and showed no progression after the salvage therapies. CONCLUSIONS: NAC concordant with extended RP is feasible and might provide good cancer control for patients with high-risk PCa.


Sujet(s)
Antinéoplasiques hormonaux/usage thérapeutique , Estramustine/usage thérapeutique , Hormone de libération des gonadotrophines/agonistes , Hormone de libération des gonadotrophines/antagonistes et inhibiteurs , Prostatectomie/méthodes , Tumeurs de la prostate/thérapie , Sujet âgé , Antagonistes des androgènes/usage thérapeutique , Humains , Japon , Noeuds lymphatiques/anatomopathologie , Métastase lymphatique/anatomopathologie , Mâle , Adulte d'âge moyen , Traitement néoadjuvant/méthodes , Études prospectives , Antigène spécifique de la prostate/sang , Tumeurs de la prostate/anatomopathologie , Études rétrospectives
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE