Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 168
Filtrar
1.
Int J Med Sci ; 21(12): 2414-2429, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39310261

RESUMEN

Background: CDK6 is linked to tumor progression and metastasis, although its molecular mechanism and prognostic value are unclear in bladder cancer. Materials and methods: In our study, raw data were obtained from public databases and Single-center retrospective case series. We conducted a bioinformatics analysis and immunohistochemistry to explore the biological landscape of CDK6 in tumors, with a particular focus on bladder cancer. We examined its expression characteristics and prognostic value and performed functional annotation analysis using gene function enrichment. We also assessed the association between bladder cancer molecular subtypes and mutation spectra and analyzed the landscape of the tumor immune microenvironment to predict treatment response sensitivity. Results: Our study found that CDK6 was a potential prognostic marker for bladder cancer. We discovered that bladder cancer patients with high CDK6 expression do not respond well to immunotherapy and have a poor prognosis. CDK6 regulates tumor immune status, metabolism, and cell cycle-related signaling pathways, thereby influencing tumor biological behavior. Furthermore, CDK6 mediated the suppression of the immune microenvironment to weaken anti-tumor immune responses. Finally, a comprehensive characterization of CDK6 was applied in the prognostic prediction of bladder cancer, suggesting that targeting CDK6 represents a potential therapeutic option. Conclusions: These results indicated that CDK6 is an independent biomarker for predicting prognosis and immunotherapy efficacy of bladder cancer. A deeper understanding of its specific molecular mechanisms may provide new treatment strategies.


Asunto(s)
Biomarcadores de Tumor , Biología Computacional , Quinasa 6 Dependiente de la Ciclina , Inmunohistoquímica , Inmunoterapia , Microambiente Tumoral , Neoplasias de la Vejiga Urinaria , Humanos , Quinasa 6 Dependiente de la Ciclina/antagonistas & inhibidores , Quinasa 6 Dependiente de la Ciclina/genética , Quinasa 6 Dependiente de la Ciclina/metabolismo , Neoplasias de la Vejiga Urinaria/tratamiento farmacológico , Neoplasias de la Vejiga Urinaria/genética , Neoplasias de la Vejiga Urinaria/patología , Neoplasias de la Vejiga Urinaria/inmunología , Neoplasias de la Vejiga Urinaria/mortalidad , Biomarcadores de Tumor/genética , Biomarcadores de Tumor/metabolismo , Pronóstico , Microambiente Tumoral/inmunología , Microambiente Tumoral/genética , Inmunoterapia/métodos , Estudios Retrospectivos , Masculino , Femenino , Regulación Neoplásica de la Expresión Génica , Mutación
2.
J Cell Mol Med ; 28(16): e70043, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-39205481

RESUMEN

Renal ischaemia-reperfusion injury (RIRI) is a primary cause of acute kidney damage, occurring frequently in situations like renal transplantation, yet the underlying mechanisms were not fully understood. Sentrin-specific protease 1 (SENP1) is an important member of the SENP family, which is widely involved in various diseases. However, the role of SENP1 in RIRI has been unclear. In our study, we discovered that SENP1 was involved in RIRI and reduced renal cell apoptosis and oxidative stress at elevated levels. Further mechanistic studies showed that hypoxia-inducible factor-1α (HIF-1α) was identified as a substrate of SENP1. Furthermore, SENP1 deSUMOylated HIF-1α, which reduced the degradation of HIF-1α, and exerted a renoprotective function. In addition, the protective function was lost after application of the HIF-1α specific inhibitor KC7F2. Briefly, our results fully demonstrated that SENP1 reduced the degradation of HIF-1α and attenuated oxidative stress and apoptosis in RIRI by regulating the deSUMOylation of HIF-1α, suggesting that SENP1 may serve as a potential therapeutic target for the treatment of RIRI.


Asunto(s)
Apoptosis , Cisteína Endopeptidasas , Subunidad alfa del Factor 1 Inducible por Hipoxia , Estrés Oxidativo , Daño por Reperfusión , Daño por Reperfusión/metabolismo , Daño por Reperfusión/patología , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Animales , Cisteína Endopeptidasas/metabolismo , Cisteína Endopeptidasas/genética , Sumoilación , Riñón/metabolismo , Riñón/patología , Humanos , Masculino , Ratones
3.
Oncogene ; 43(39): 2951-2969, 2024 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-39174859

RESUMEN

High invasive capacity and acquired tyrosine kinase inhibitors (TKI) resistance of kidney renal clear cell carcinoma (KIRC) cells remain obstacles to prolonging the survival time of patients with advanced KIRC. In the present study, we reported that sine oculis homeobox 1 (SIX1) was upregulated in sunitinib-resistant KIRC cells and metastatic KIRC tissues. Subsequently, we found that SIX1 mediated metastasis and sunitinib resistance via Focal adhesion (FA) signaling, and knockdown of SIX1 enhanced the antitumor efficiency of sunitinib in KIRC. Mechanistically, Integrin subunit beta 1 (ITGB1), an upstream gene of FA signaling, was a direct transcriptional target of SIX1. In addition, we showed that DExH-box helicase 9 (DHX9) was an important mediator for SIX1-induced ITGB1 transcription, and silencing the subunits of SIX1/DHX9 complex significantly reduced transcription of ITGB1. Downregulation of SIX1 attenuated nuclear translocation of DHX9 and abrogated the binding of DHX9 to ITGB1 promoter. Collectively, our results unveiled a new signal axis SIX1/ITGB1/FAK in KIRC and identified a novel therapeutic strategy for metastatic KIRC patients.


Asunto(s)
Carcinoma de Células Renales , ARN Helicasas DEAD-box , Resistencia a Antineoplásicos , Adhesiones Focales , Regulación Neoplásica de la Expresión Génica , Proteínas de Homeodominio , Integrina beta1 , Neoplasias Renales , Metástasis de la Neoplasia , Transducción de Señal , Sunitinib , Humanos , Resistencia a Antineoplásicos/genética , Carcinoma de Células Renales/genética , Carcinoma de Células Renales/patología , Carcinoma de Células Renales/tratamiento farmacológico , Carcinoma de Células Renales/metabolismo , Sunitinib/farmacología , Sunitinib/uso terapéutico , Neoplasias Renales/patología , Neoplasias Renales/genética , Neoplasias Renales/tratamiento farmacológico , Neoplasias Renales/metabolismo , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Línea Celular Tumoral , Integrina beta1/genética , Integrina beta1/metabolismo , Animales , Adhesiones Focales/genética , Adhesiones Focales/metabolismo , ARN Helicasas DEAD-box/genética , ARN Helicasas DEAD-box/metabolismo , Ratones , Transcripción Genética , Integrinas/metabolismo , Integrinas/genética , Quinasa 1 de Adhesión Focal
4.
Cancer Med ; 13(16): e70112, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-39166457

RESUMEN

BACKGROUND: Tumor mutation burden (TMB) and VHL mutation play a crucial role in the management of patients with clear cell renal cell carcinoma (ccRCC), such as guiding adjuvant chemotherapy and improving clinical outcomes. However, the time-consuming and expensive high-throughput sequencing methods severely limit their clinical applicability. Predicting intratumoral heterogeneity poses significant challenges in biology and clinical settings. Our aimed to develop a self-supervised attention-based multiple instance learning (SSL-ABMIL) model to predict TMB and VHL mutation status from hematoxylin and eosin-stained histopathological images. METHODS: We obtained whole slide images (WSIs) and somatic mutation data of 350 ccRCC patients from The Cancer Genome Atlas for developing SSL-ABMIL model. In parallel, 163 ccRCC patients from Clinical Proteomic Tumor Analysis Consortium cohort was used as independent external validation set. We systematically compared three different models (Wang-ABMIL, Ciga-ABMIL, and ImageNet-MIL) for their ability to predict TMB and VHL alterations. RESULTS: We first identified two groups of populations with high- and low-TMB (cut-off point = 0.9). In two independent cohorts, the Wang-ABMIL model achieved the highest performance with decent generalization performance (AUROC = 0.83 ± 0.02 and 0.8 ± 0.04 in predicting TMB and VHL, respectively). Attention heatmaps revealed that the Wang-ABMIL model paid the highest attention to tumor regions in high-TMB patients, while in VHL mutation prediction, non-tumor regions were also assigned high attention, particularly the stromal regions infiltrated by lymphocytes. CONCLUSIONS: Our results indicated that SSL-ABMIL can effectively extract histological features for predicting TMB and VHL mutation, demonstrating promising results in linking tumor morphology and molecular biology.


Asunto(s)
Carcinoma de Células Renales , Aprendizaje Profundo , Neoplasias Renales , Mutación , Proteína Supresora de Tumores del Síndrome de Von Hippel-Lindau , Humanos , Proteína Supresora de Tumores del Síndrome de Von Hippel-Lindau/genética , Neoplasias Renales/genética , Neoplasias Renales/patología , Carcinoma de Células Renales/genética , Carcinoma de Células Renales/patología , Masculino , Femenino , Persona de Mediana Edad , Biomarcadores de Tumor/genética , Anciano
5.
Biomedicines ; 12(7)2024 Jul 17.
Artículo en Inglés | MEDLINE | ID: mdl-39062155

RESUMEN

Epidermal growth factor receptor 2 (HER2) has been widely recognized as one of the targets for bladder cancer immunotherapy. The key to implementing personalized treatment for bladder cancer patients lies in achieving rapid and accurate diagnosis. To tackle this challenge, we have pioneered the application of deep learning techniques to predict HER2 expression status from H&E-stained pathological images of bladder cancer, bypassing the need for intricate IHC staining or high-throughput sequencing methods. Our model, when subjected to rigorous testing within the cohort from the People's Hospital of Wuhan University, which encompasses 106 cases, has exhibited commendable performance on both the validation and test datasets. Specifically, the validation set yielded an AUC of 0.92, an accuracy of 0.86, a sensitivity of 0.87, a specificity of 0.83, and an F1 score of 86.7%. The corresponding metrics for the test set were 0.88 for AUC, 0.67 for accuracy, 0.56 for sensitivity, 0.75 for specificity, and 77.8% for F1 score. Additionally, in a direct comparison with pathologists, our model demonstrated statistically superior performance, with a p-value less than 0.05, highlighting its potential as a powerful diagnostic tool.

6.
Ren Fail ; 46(2): 2378211, 2024 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-39082473

RESUMEN

OBJECTIVES: Transplant renal artery stenosis (TRAS) is now recognized as a curable disease with a good prognosis if intervention occurs in the early stage. However, the mid-term outcomes of TRAS when treated by percutaneous transluminal angioplasty with stent placement have yet to be fully elucidated. The purpose of this study was to compare mid-term graft and patient survival of TRAS group with a control group. PATIENTS AND METHODS: Ninety-two patients were diagnosed of TRAS between January 2016 and January 2022 in our center. Fifty-six pairs of recipients with grafts from the same donor were selected as a study group with TRAS and a control group without TRAS, respectively. All donor kidneys were from deceased organ donation rather than living donors. The primary endpoints were graft and patient survival. The secondary outcomes were changes in renal graft function. RESULTS: The mean follow-up time for the TRAS group was 43.6 months, while the mean follow-up time for the control group was 45.3 months. In the TRAS group, the age of patients ranged from 11 to 62 years with 39 males and 17 females. In the control group, the age of patients ranged from 18 to 67 years with 40 males and 16 females. In the TRAS group, there were more patients with diabetic nephropathy as the primary renal disease compared to the control group (5/56 vs 0/56), and the incidence of acute rejection was higher in the TRAS group than in the control group (12/56 vs 3/56). Eight patients in the TRAS group and one patient in the control group experienced graft loss (p = .019). Four patients in the TRAS group and four patients in the control group died with functional renal allograft during the follow-up time (p = .989). The levels of eGFR did not differ significantly between the two groups in the first three years after kidney transplant (p > .05). Patients in the TRAS group had worse graft functionality (eGFR, 44.96 ± 18.9 vs 54.9 ± 19.6 mL/min) in the fourth year when compared with the control group (p = .01). CONCLUSIONS: The graft function deteriorated faster, and graft survival was lower in the TRAS group treated by stent placement when compared with a control group without TRAS over the mid-term.


Asunto(s)
Supervivencia de Injerto , Trasplante de Riñón , Obstrucción de la Arteria Renal , Stents , Humanos , Masculino , Femenino , Obstrucción de la Arteria Renal/cirugía , Obstrucción de la Arteria Renal/etiología , Obstrucción de la Arteria Renal/terapia , Obstrucción de la Arteria Renal/mortalidad , Persona de Mediana Edad , Trasplante de Riñón/efectos adversos , Adulto , Estudios Retrospectivos , Anciano , Adulto Joven , Adolescente , Niño , Cadáver , Angioplastia/métodos , Tasa de Filtración Glomerular
7.
Sci Rep ; 14(1): 14667, 2024 06 25.
Artículo en Inglés | MEDLINE | ID: mdl-38918587

RESUMEN

Bladder urothelial carcinoma (BLCA) presents a persistent challenge in clinical management. Despite recent advancements demonstrating the BLCA efficacy of immune checkpoint inhibitors (ICI) in BLCA patients, there remains a critical need to identify and expand the subset of individuals who benefit from this treatment. Mitochondria, as pivotal regulators of various cell death pathways in eukaryotic cells, exert significant influence over tumor cell fate and survival. In this study, our objective was to investigate biomarkers centered around mitochondrial function and cell death mechanisms to facilitate prognostic prediction and guide therapeutic decision-making in BLCA. Utilizing ssGSEA and LASSO regression, we developed a prognostic signature termed mitochondrial function and cell death (mtPCD). Subsequently, we evaluated the associations between mtPCD score and diverse clinical outcomes, including prognosis, functional pathway enrichment, immune cell infiltration, immunotherapy response analysis and drug sensitivity, within high- and low-risk subgroups. Additionally, we employed single-cell level functional assays, RT-qPCR, and immunohistochemistry to validate the differential expression of genes comprising the mtPCD signature. The mtPCD signature comprises a panel of 10 highly influential genes, strongly correlated with survival outcomes in BLCA patients and exhibiting robust predictive capabilities. Importantly, individuals classified as high-risk according to mtPCD score displayed a subdued overall immune response, characterized by diminished immunotherapeutic efficacy. In summary, our findings highlight the development of a novel prognostic signature, which not only holds promise as a biomarker for BLCA prognosis but also offers insights into the immune landscape of BLCA. This paradigm may pave the way for personalized treatment strategies in BLCA management.


Asunto(s)
Biomarcadores de Tumor , Mitocondrias , Neoplasias de la Vejiga Urinaria , Neoplasias de la Vejiga Urinaria/genética , Neoplasias de la Vejiga Urinaria/patología , Neoplasias de la Vejiga Urinaria/mortalidad , Neoplasias de la Vejiga Urinaria/tratamiento farmacológico , Humanos , Pronóstico , Mitocondrias/genética , Mitocondrias/metabolismo , Biomarcadores de Tumor/genética , Regulación Neoplásica de la Expresión Génica , Muerte Celular/genética , Masculino , Perfilación de la Expresión Génica
8.
World J Urol ; 42(1): 238, 2024 Apr 16.
Artículo en Inglés | MEDLINE | ID: mdl-38627315

RESUMEN

BACKGROUND: Accurate estimation of the glomerular filtration rate (GFR) is clinically crucial for determining the status of obstruction, developing treatment strategies, and predicting prognosis in obstructive nephropathy (ON). We aimed to develop a deep learning-based system, named UroAngel, for non-invasive and convenient prediction of single-kidney function level. METHODS: We retrospectively collected computed tomography urography (CTU) images and emission computed tomography diagnostic reports of 520 ON patients. A 3D U-Net model was used to segment the renal parenchyma, and a logistic regression multi-classification model was used to predict renal function level. We compared the predictive performance of UroAngel with the Modification of Diet in Renal Disease (MDRD), Chronic Kidney Disease Epidemiology Collaboration (CKD-EPI) equations, and two expert radiologists in an additional 40 ON patients to validate clinical effectiveness. RESULTS: UroAngel based on 3D U-Net convolutional neural network could segment the renal cortex accurately, with a Dice similarity coefficient of 0.861. Using the segmented renal cortex to predict renal function stage had high performance with an accuracy of 0.918, outperforming MDRD and CKD-EPI and two radiologists. CONCLUSIONS: We proposed an automated 3D U-Net-based analysis system for direct prediction of single-kidney function stage from CTU images. UroAngel could accurately predict single-kidney function in ON patients, providing a novel, reliable, convenient, and non-invasive method.


Asunto(s)
Aprendizaje Profundo , Insuficiencia Renal Crónica , Riñón Único , Humanos , Estudios Retrospectivos , Riñón/diagnóstico por imagen , Insuficiencia Renal Crónica/diagnóstico , Tasa de Filtración Glomerular , Tomografía , Creatinina
9.
J Cell Mol Med ; 28(8): e18290, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38588015

RESUMEN

Growth hormone inducible transmembrane protein (GHITM), one member of Bax inhibitory protein-like family, has been rarely studied, and the clinical importance and biological functions of GHITM in kidney renal clear cell carcinoma (KIRC) still remain unknown. In the present study, we found that GHITM was downregulated in KIRC. Aberrant GHITM downregulation related to clinicopathological feature and unfavourable prognosis of KIRC patients. GHITM overexpression inhibited KIRC cell proliferation, migration and invasion in vitro and in vivo. Mechanistically, GHITM overexpression could induce the downregulation of Notch1, which acts as an oncogene in KIRC. Overexpression of Notch1 effectively rescued the inhibitory effect induced by GHITM upregulation. More importantly, GHITM could regulate PD-L1 protein abundance and ectopic overexpression of GHITM enhanced the antitumour efficiency of PD-1 blockade in KIRC, which provided new insights into antitumour therapy. Furthermore, we also showed that YY1 could decrease GHITM level via binding to its promoter. Taken together, our study revealed that GHITM was a promising therapeutic target for KIRC, which could modulate malignant phenotype and sensitivity to PD-1 blockade of renal cancer cells via Notch signalling pathway.


Asunto(s)
Carcinoma de Células Renales , Neoplasias Renales , Humanos , Carcinoma de Células Renales/tratamiento farmacológico , Carcinoma de Células Renales/genética , Riñón , Neoplasias Renales/tratamiento farmacológico , Neoplasias Renales/genética , Fenotipo , Receptor de Muerte Celular Programada 1
10.
Aging (Albany NY) ; 16(3): 2812-2827, 2024 02 05.
Artículo en Inglés | MEDLINE | ID: mdl-38319718

RESUMEN

BACKGROUND: Bladder cancer (BCa) is a common malignancy in the urinary system. Necroptosis, a recently discovered form of programmed cell death, is closely associated with the development and progression of various types of tumors. Targeting necroptosis through anti-cancer strategies has shown potential as a therapy for cancer. We aimed to develop a necroptosis-related lncRNAs (NRlncRNAs) risk model that can predict the survival and tumor immunity of BCa patients. METHODS: We analyzed sequencing data obtained from the TCGA database, and applied least absolute shrinkage and selection operator (LASSO) and Cox regression analysis to identify crucial NRlncRNAs for building a risk model. Using the risk score, we categorized patients into high- and low-risk groups, and assessed the accuracy with the area under the receiver operating characteristic (AUROC) and Kaplan-Meier curves. We performed the RT-qPCR to detect the expression differences of the genes based on the risk model. RESULTS: We identified a total of 296 NRlncRNAs, and 6 of them were included in the prognostic model. The AUC values for 1-, 3-, and 5-year predictions were 0.675, 0.726 and 0.734, respectively. Our risk model demonstrated excellent predictive performance and served as an independent predictor with high predictive power. Additionally, we performed PCA, TMB, GSEA analyses, and evaluated immune cell infiltration, to reveal significant differences between the high- and low-risk groups in functional signaling pathways, immunological status, and mutation profiles. Finally, we assessed the chemotherapeutic response of several drugs. According to the RT-qPCR results, we found that four NRlncRNAs of the risk model were more highly expressed in BCa cell lines than human immortalized uroepithelial cell line and regulated the occurrence and progression of bladder cancer. CONCLUSION: We constructed a novel NRlncRNAs-associated risk model, which could predict the prognosis and immune response of BCa patients.


Asunto(s)
ARN Largo no Codificante , Neoplasias de la Vejiga Urinaria , Humanos , ARN Largo no Codificante/genética , Necroptosis/genética , Neoplasias de la Vejiga Urinaria/genética , Pronóstico , Apoptosis , Microambiente Tumoral/genética
11.
Biomedicines ; 12(2)2024 Feb 04.
Artículo en Inglés | MEDLINE | ID: mdl-38397968

RESUMEN

BACKGROUND: This study aimed to develop a simple predictive model for early identification of the risk of adverse outcomes in kidney transplant-associated Pneumocystis carinii pneumonia (PCP) patients. METHODS: This study encompassed 103 patients diagnosed with PCP, who received treatment at our hospital between 2018 and 2023. Among these participants, 20 were categorized as suffering from severe PCP, and, regrettably, 13 among them succumbed. Through the application of machine learning techniques and multivariate logistic regression analysis, two pivotal variables were discerned and subsequently integrated into a nomogram. The efficacy of the model was assessed via receiver operating characteristic (ROC) curves and calibration curves. Additionally, decision curve analysis (DCA) and a clinical impact curve (CIC) were employed to evaluate the clinical utility of the model. The Kaplan-Meier (KM) survival curves were utilized to ascertain the model's aptitude for risk stratification. RESULTS: Hematological markers, namely Procalcitonin (PCT) and C-reactive protein (CRP)-to-albumin ratio (CAR), were identified through machine learning and multivariate logistic regression. These variables were subsequently utilized to formulate a predictive model, presented in the form of a nomogram. The ROC curve exhibited commendable predictive accuracy in both internal validation (AUC = 0.861) and external validation (AUC = 0.896). Within a specific threshold probability range, both DCA and CIC demonstrated notable performance. Moreover, the KM survival curve further substantiated the nomogram's efficacy in risk stratification. CONCLUSIONS: Based on hematological parameters, especially CAR and PCT, a simple nomogram was established to stratify prognostic risk in patients with renal transplant-related PCP.

12.
Cancer Biol Med ; 20(12)2024 02 05.
Artículo en Inglés | MEDLINE | ID: mdl-38318809

RESUMEN

OBJECTIVE: Real-word data on long-acting luteinizing hormone-releasing hormone (LHRH) agonists in Chinese patients with prostate cancer are limited. This study aimed to determine the real-world effectiveness and safety of the LHRH agonist, goserelin, particularly the long-acting 10.8-mg depot formulation, and the follow-up patterns among Chinese prostate cancer patients. METHODS: This was a multicenter, prospective, observational study in hormone treatment-naïve patients with localized or locally advanced prostate cancer who were prescribed goserelin 10.8-mg depot every 12 weeks or 3.6-mg depot every 4 weeks with or without an anti-androgen. The patients had follow-up evaluations for 26 weeks. The primary outcome was the effectiveness of goserelin in reducing serum testosterone and prostate-specific antigen (PSA) levels. The secondary outcomes included testosterone and PSA levels, attainment of chemical castration (serum testosterone <50 ng/dL), and goserelin safety. The exploratory outcome was the monitoring pattern for serum testosterone and PSA. All analyses were descriptive. RESULTS: Between September 2017 and December 2019, a total of 294 eligible patients received ≥ 1 dose of goserelin; 287 patients (97.6%) were treated with goserelin 10.8-mg depot. At week 24 ± 2, the changes from baseline [standard deviation (95% confidence interval)] in serum testosterone (n = 99) and PSA (n = 131) were -401.0 ng/dL [308.4 ng/dL (-462.5, -339.5 ng/dL)] and -35.4 ng/mL [104.4 ng/mL (-53.5, -17.4 ng/mL)], respectively. Of 112 evaluable patients, 100 (90.2%) achieved a serum testosterone level < 50 ng/dL. Treatment-emergent adverse events (TEAEs) and severe TEAEs occurred in 37.1% and 10.2% of patients, respectively. The mean testing frequency (standard deviation) was 1.6 (1.5) for testosterone and 2.2 (1.6) for PSA. CONCLUSIONS: Goserelin 10.8-mg depot effectively achieved and maintained castration and was well-tolerated in Chinese patients with localized and locally advanced prostate cancer.


Asunto(s)
Goserelina , Neoplasias de la Próstata , Masculino , Humanos , Goserelina/efectos adversos , Antígeno Prostático Específico/uso terapéutico , Antineoplásicos Hormonales/efectos adversos , Estudios Prospectivos , Neoplasias de la Próstata/tratamiento farmacológico , Testosterona/uso terapéutico , China
13.
Cell Death Discov ; 10(1): 17, 2024 Jan 09.
Artículo en Inglés | MEDLINE | ID: mdl-38195664

RESUMEN

Renal fibrosis is considered to be the ultimate pathway for various chronic kidney disease, with a complex etiology and great therapeutic challenges. Tripartite motif-containing (TRIM) family proteins have been shown to be involved in fibrotic diseases, but whether TRIM39 plays a role in renal fibrosis remain unexplored. In this study, we investigated the role of TRIM39 in renal fibrosis and its molecular mechanism. TRIM39 expression was analyzed in patients' specimens, HK-2 cells and unilateral ureteral obstruction (UUO) mice were used for functional and mechanistic studies. We found an upregulated expression of TRIM39 in renal fibrosis human specimens and models. In addition, TRIM39 knockdown was found efficient for alleviating renal fibrosis in both UUO mice and HK-2 cells. Mechanistically, we demonstrated that TRIM39 interacted with PRDX3 directly and induced ubiquitination degradation of PRDX3 at K73 and K149 through the K48 chain, which resulted in ROS accumulation and increased inflammatory cytokine generation, and further aggravated renal fibrosis. It provided an emerging potential target for the therapies of renal fibrosis.

14.
Biomedicines ; 11(10)2023 Sep 25.
Artículo en Inglés | MEDLINE | ID: mdl-37893001

RESUMEN

Long non-coding RNAs (lncRNAs), typically more than 200 nt long, cannot encode proteins, but can regulate gene expression. They play an indispensable role in the occurrence and progression of various cancers. The main purpose of this study is to discuss the role and mechanism of LNC-565686 in prostate cancer. First, we found an increased expression of LNC-565686 in prostate cancer cells using RNA sequencing, which was further verified using qRT-PCR. Then, catRAPID was used to find that LNC-565686 might regulate SND1. Furthermore, a protein half-life experiment was performed to verify that LNC-565686 could stabilize the expression of SND1. In order to further explore the effects of LNC-565686 and SND1 on prostate cancer cells, we knocked down LNC-565686 and SND1 in prostate cancer cells, and verified using CCK8 and flow cytometry and western blot for the detection of apoptosis-related indicators. Collectively, we have found that LNC-565686 can promote the proliferation of prostate cancer cells and inhibit apoptosis by stabilizing the expression of SND1. Therefore, targeting LNC-565686 might be a new treatment for prostate cancer.

15.
Aging (Albany NY) ; 15(20): 11654-11671, 2023 10 27.
Artículo en Inglés | MEDLINE | ID: mdl-37899170

RESUMEN

Protein L-isoaspartate (D-aspartate) O-methyltransferase (PCMT1) is a repair enzyme that catalyzes the conversion of isomerized aspartic acid (iso-Asp) residues into their normal structure, thereby restoring the configuration and function of proteins. Studies have shown that PCMT1 is overexpressed in several tumors and affects patients' prognosis. However, there are few reports on the role of PCMT1 in prostate cancer (PCa). In the present research, with the assistance of The Cancer Genome Atlas Program (TCGA) database, we found that PCMT1 was overexpressed in PCa tissues. The results of quantitative reverse transcription-polymerase chain reaction (qRT-PCR), western blot and immunohistochemistry staining also showed that PCMT1 expression was significantly increased in PCa tissues and cell lines. In PCa clinical samples, PCMT1 expression was closely related to Gleason score, clinical stage, lymph node metastasis and bone metastasis. The experiments of overexpression and knockdown of PCMT1 in vitro or in vivo showed that PCMT1 can significantly promote the proliferation, migration and invasion of PCa cells, inhibit cell apoptosis, and promote the growth of PCa. We furthermore confirmed that PCMT1 regulated the migration, invasion and apoptosis of PCa cells by modulating the phosphatidylinositol 3-kinase/AKT kinase/glycogen-synthase kinase-3ß (PI3K/AKT/GSK-3ß) signaling pathway. Collectively, PCMT1 plays a cancer-facilitative role in PCa by promoting the proliferation, migration and invasion of PCa cells, and inhibiting apoptosis. Therefore, PCMT1 is considered to represent a novel target for treating PCa.


Asunto(s)
Neoplasias de la Próstata , Proteínas Proto-Oncogénicas c-akt , Humanos , Masculino , Apoptosis/fisiología , Proliferación Celular/genética , Glucógeno Sintasa Quinasa 3 beta/genética , Fosfatidilinositol 3-Quinasas/metabolismo , Neoplasias de la Próstata/patología , Proteína D-Aspartato-L-Isoaspartato Metiltransferasa/genética , Proteína D-Aspartato-L-Isoaspartato Metiltransferasa/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo
16.
Eur J Pharmacol ; 960: 176110, 2023 Dec 05.
Artículo en Inglés | MEDLINE | ID: mdl-37838104

RESUMEN

Renal cell carcinoma (RCC) is the most common type of kidney cancer, and it appears to be highly susceptible to ferroptosis. Disulfiram, an alcoholism drug, has been shown to have anticancer properties in various studies, including those on RCC. However, the mechanism of the anticancer effect of disulfiram/copper on RCC remains unclear. In this study, we investigated the impact of disulfiram/copper on RCC treatment using both RCC cells and mouse subcutaneous tumor models. Our findings demonstrate that disulfiram/copper treatment reduced the viability of RCC cells, inhibited their invasion and migration, and disrupted mitochondrial homeostasis, ultimately leading to oxidative stress and ferroptosis. Mechanistically, disulfiram/copper treatment prolonged the half-life of NRF2 and reduced its degradation, but had no effect on transcription, indicating that the disulfiram/copper-induced increase in NRF2 was not related to transcription. Furthermore, we observed that disulfiram/copper treatment reduced the expression of NPL4, a ubiquitin protein-proteasome system involved in NRF2 degradation, while overexpression of NPL4 reversed NRF2 levels and enhanced disulfiram/copper-induced oxidative stress and ferroptosis. These results suggest that overcoming the compensatory increase in NRF2 induced by NPL4 inhibition enhances disulfiram/copper-induced oxidative stress and ferroptosis in RCC. In addition, our in vivo experiments revealed that disulfiram/copper synergized with sorafenib to inhibit the growth of RCC cells and induce ferroptosis. In conclusion, our study sheds light on a possible mechanism for disulfiram/copper treatment in RCC and provides a potential synergistic strategy to overcome sorafenib resistance.


Asunto(s)
Carcinoma de Células Renales , Ferroptosis , Neoplasias Renales , Ratones , Animales , Carcinoma de Células Renales/tratamiento farmacológico , Disulfiram/farmacología , Sorafenib/farmacología , Cobre/farmacología , Factor 2 Relacionado con NF-E2/metabolismo , Neoplasias Renales/tratamiento farmacológico , Estrés Oxidativo
17.
BMC Cancer ; 23(1): 1021, 2023 Oct 23.
Artículo en Inglés | MEDLINE | ID: mdl-37872487

RESUMEN

BACKGROUND: Bladder urothelial carcinoma (BLCA) is the most common genitourinary cancer and the prognosis of patients is often poor. However, studies of basement membrane-related genes (BM-related genes) in BLCA are less reported. Therefore, we established a BM-related genes signature to explore their functional and prognostic value in BLCA. METHODS: In this study, a BM-related genes signature was constructed by LASSO-Cox regression analysis, and then a series of bioinformatics methods was used to assess the accuracy and validity of the signature. We constructed a nomogram for clinical application and also screened for possible therapeutic drugs. To investigate the functions and pathways affected by BM-related genes in BLCA, we performed functional enrichment analyses. In addition, we analyzed the immune cell infiltration landscape and immune checkpoint-related genes in the high and low-risk groups. Finally, we confirmed the prognostic value of BM-related genes in BLCA in vitro. RESULTS: Combining multiple bioinformatics approaches, we identified a seven-gene signature. The accuracy and validity of this signature in predicting BLCA patients were confirmed by the test cohort. In addition, the risk score was strongly correlated with prognosis, immune checkpoint genes, drug sensitivity, and immune cell infiltration landscape. The risk score is an independent prognostic factor for BLCA patients. Further experiments revealed that all seven signature genes were differentially expressed between BLCA cell lines and normal bladder cells. Finally, overexpression of LAMA2 inhibited the migration and invasion ability of BLCA cell lines. CONCLUSIONS: In summary, the BM-related genes signature was able to predict the prognosis of BLCA patients accurately, indicating that the BM-related genes possess great clinical value in the diagnosis and treatment of BLCA. Moreover, LAMA2 could be a potential therapeutic target, which provides new insights into the application of the BM-related genes in BLCA patients.


Asunto(s)
Carcinoma de Células Transicionales , Neoplasias de la Vejiga Urinaria , Humanos , Carcinoma de Células Transicionales/genética , Neoplasias de la Vejiga Urinaria/genética , Vejiga Urinaria , Células Epiteliales , Membrana Basal , Pronóstico
18.
J Agric Food Chem ; 71(36): 13284-13303, 2023 Sep 13.
Artículo en Inglés | MEDLINE | ID: mdl-37646396

RESUMEN

Acute kidney injury and chronic renal fibrosis are intractable pathological processes to resolve, yet limited strategies are able to effectively address them. Cardamonin (CAD) is a flavonoid with talented antioxidant, anti-inflammatory capacity, and satisfactory biosafety. In our study, animal and cellular models of renal ischemia/reperfusion (I/R) and unilateral ureteral obstruction (UUO) were successfully constructed to confirm whether CAD confers protective effects and underlying mechanisms. Animal experiments demonstrated that CAD application (100 mg/kg) distinctly ameliorated tissue damage and improved renal function. Meanwhile, the continuous oral administration of CAD after UUO surgery efficiently inhibited renal fibrosis as confirmed by hematoxylin-eosin (H&E), Sirius red, and Masson staining as well as the downregulated mRNA and protein expression of collagen I, α-smooth muscle actin (α-SMA), collagen III, and fibronectin. Interestingly, in transforming growth factor ß1 (TGF-ß1)-stimulated and hypoxia/reoxygenation (H/R)-exposed human kidney-2 (HK-2) cells, protective effects of CAD were again authenticated. Meanwhile, we performed bioinformatics analysis and constructed the "ingredient-target-pathway-disease" network to conclude that the potential mechanisms of CAD protection may be through the regulation of oxidative stress, inflammation, apoptosis, and mitogen-activated protein kinase (MAPK) pathway. Furthermore, experimental data validated that CAD evidently decreased the reactive oxygen species (ROS) production and malondialdehyde (MDA) content while depressing the mRNA and protein expression of inflammatory markers (tumor necrosis factor-α (TNF-α), interleukin-6 (IL-6), and Il-1ß) and inhibiting apoptosis as evidenced by decreased levels of P53, BAX, cleaved caspase-3, and apoptotic rate in renal I/R and UUO models. In addition, the impact of CAD on restraining oxidative stress and inflammation was attributed to its ability to elevate antioxidant enzyme activities including catalase, superoxide dismutase 1 (SOD1), and superoxide dismutase 2 (SOD2) and to inhibit the inflammation-associated MARK/nuclear factor-κB (MAPK/NF-κB) signaling pathway. In conclusion, cardamonin restored the antioxidative capacity to block oxidative stress and suppressed the MAPK/NF-κB signaling pathway to alleviate inflammatory response, thus mitigating I/R-generated acute kidney injury/UUO-induced renal fibrosis in vivo and in vitro, which indicated the potential therapeutic advantage of cardamonin in attenuating acute and chronic kidney injuries.


Asunto(s)
Lesión Renal Aguda , Daño por Reperfusión , Obstrucción Ureteral , Animales , Humanos , Antioxidantes , FN-kappa B , Daño por Reperfusión/tratamiento farmacológico , Daño por Reperfusión/genética
19.
Front Immunol ; 14: 1205250, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37426643

RESUMEN

Disulfidptosis is a newly discovered mode of cell death induced by disulfide stress. However, the prognostic value of disulfidptosis-related genes (DRGs) in renal cell carcinoma (RCC) remains to be further elucidated. In this study, consistent cluster analysis was used to classify 571 RCC samples into three DRG-related subtypes based on changes in DRGs expression. Through univariate regression analysis and LASSO-Cox regression analysis of differentially expressed genes (DEGs) among three subtypes, we constructed and validated a DRG risk score to predict the prognosis of patients with RCC, while also identifying three gene subtypes. Analysis of DRG risk score, clinical characteristics, tumor microenvironment (TME), somatic cell mutations, and immunotherapy sensitivity revealed significant correlations between them. A series of studies have shown that MSH3 can be a potential biomarker of RCC, and its low expression is associated with poor prognosis in patients with RCC. Last but not least, overexpression of MSH3 promotes cell death in two RCC cell lines under glucose starvation conditions, indicating that MSH3 is a key gene in the process of cell disulfidptosis. In summary, we identify potential mechanism of RCC progression through DRGs -related tumor microenvironment remodeling. In addition, this study has successfully established a new disulfidptosis-related genes prediction model and discovered a key gene MSH3. They may be new prognostic biomarkers for RCC patients, provide new insights for the treatment of RCC patients, and may inspire new methods for the diagnosis and treatment of RCC patients.


Asunto(s)
Carcinoma de Células Renales , Neoplasias Renales , Humanos , Pronóstico , Carcinoma de Células Renales/genética , Microambiente Tumoral/genética , Muerte Celular , Neoplasias Renales/genética , Proteína 3 Homóloga de MutS
20.
Biomed Pharmacother ; 165: 115128, 2023 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-37429230

RESUMEN

Renal ischemia reperfusion injury (RIRI) is an inevitable complication during renal surgery. Histone deacetylase 6 (HDAC6), a key member of the histone deacetylase family, is associated with multiple pathologies, including renal diseases. However, whether HDAC6 could become a potential therapeutic target for clinical application of RIRI remained to be proven. Here, we found that HDAC6 expression was abnormally enhanced by the transcription factor OSR2 in RIRI. Moreover, we were the first to validate that a selective HDAC6 degrader, proteolysis-targeting chimeras (PROTAC) NP8, could significantly improve RIRI. Further in vivo and in vitro mechanism studies have found that the reduction of HDAC6 alleviated RIRI by inhibiting ROS mediated apoptosis. Remarkably, a renal protective protein, Klotho, has been proven to be a target of HDAC6, and the degradation of HDAC6 restored KL expression, thereby ameliorating ROS mediated apoptosis. Overall, our results illustrated that the degradation of HDAC6 restrained ROS mediated apoptosis by restoring Klotho expression during RIRI. PROTAC-NP8 might be a potential therapeutic strategy for clinical prevention of RIRI.


Asunto(s)
Riñón , Daño por Reperfusión , Humanos , Histona Desacetilasa 6/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Riñón/patología , Daño por Reperfusión/metabolismo , Apoptosis
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA