Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 108
Filtrar
1.
Biochem Genet ; 2024 Jun 20.
Artigo em Inglês | MEDLINE | ID: mdl-38902482

RESUMO

With the emergence of combined surgical treatments, complemented by radiotherapy and chemotherapy, survival rates for esophageal cancer patients have improved, but the overall 5-year survival rate remains low. Therefore, there is an urgent need for further research into the pathogenesis of esophageal cancer and the development of effective prevention, diagnosis, and treatment methods. We initially utilized the GeneCards and DisGeNET databases to identify the esophageal cancer-associated gene WWOX (WW domain containing oxidoreductase). Subsequently, we employed RT-qPCR (Reverse transcription-quantitative PCR) and WB (western blot) to investigate the differential expression of WWOX in HEEC (human esophageal endotheliocytes) and various ESCC (esophageal squamous cell carcinoma) cell lines. We further evaluated alterations in cell proliferation, migration and apoptosis via CCK8 (cell counting kit-8) and clonal formation, Transwell assays and flow cytometry. Additionally, we investigated changes in protein expressions related to the Hippo signaling pathway (YAP/TEAD) through RT-qPCR and WB. Lastly, to further elucidate the regulatory mechanism of WWOX in ESCC, we performed exogenous YAP rescue experiments in ESCC cells with WWOX overexpression to investigate the alterations in apoptosis and proliferation. Results indicated that the expression of WWOX in ESCC was significantly downregulated. Subsequently, upon overexpression of WWOX, ESCC cell proliferation and migration decreased, while apoptosis increased. Additionally, the expression of YAP and TEAD were reduced. However, the sustained overexpression of YAP attenuated the inhibitory effects of WWOX on ESCC cell malignancy. In conclusion, WWOX exerts inhibitory effects on the proliferation and migration of ESCC and promotes apoptosis by suppressing the Hippo signaling pathway. These findings highlight the potential of WWOX as a novel target for the diagnosis and treatment of esophageal cancer.

2.
BMC Med ; 21(1): 11, 2023 01 08.
Artigo em Inglês | MEDLINE | ID: mdl-36617560

RESUMO

BACKGROUND: Rezivertinib (BPI-7711) is a novel third-generation epidermal growth factor receptor (EGFR) tyrosine kinase inhibitor (TKI). This phase IIa study was part of a phase I/IIa study (NCT03386955), aimed to evaluate the efficacy and safety of rezivertinib as the first-line treatment for patients with locally advanced or metastatic/recurrent EGFR mutated non-small cell lung cancer (NSCLC). METHODS: Patients received the first-line treatment of 180 mg rezivertinib orally once daily until disease progression, unacceptable toxicity, or withdrawal of consent. The primary endpoint was the objective response rate (ORR) assessed by blinded independent central review (BICR). Secondary endpoints included disease control rate (DCR), duration of response (DoR), progression-free survival (PFS), overall survival (OS), and safety. RESULTS: From Jun 12, 2019, to Oct 17, 2019, 43 patients were enrolled. At the data cutoff date on Dec 23, 2021, the ORR by BICR was 83.7% (95% CI: 69.3-93.2%). The median DoR was 19.3 (95% CI: 15.8-25.0) months. The median PFS by BICR was 20.7 (95% CI: 13.8-24.8) months and 22.0 (95% CI: 16.8-26.3) months by investigators. Data on OS was immature. Totally, 40 (93.0%) patients had at least one treatment-related adverse event while 4 (9.3%) of them were grade ≥ 3. CONCLUSIONS: Rezivertinib (BPI-7711) showed promising efficacy and a favorable safety profile for the treatment among the locally advanced or metastatic/recurrent NSCLC patients with EGFR mutation in the first-line setting. TRIAL REGISTRATION: ClinicalTrials.gov, NCT03386955.


Assuntos
Antineoplásicos , Carcinoma Pulmonar de Células não Pequenas , Neoplasias Pulmonares , Humanos , Carcinoma Pulmonar de Células não Pequenas/tratamento farmacológico , Carcinoma Pulmonar de Células não Pequenas/genética , Neoplasias Pulmonares/tratamento farmacológico , Neoplasias Pulmonares/genética , Antineoplásicos/efeitos adversos , Inibidores de Proteínas Quinases/efeitos adversos , Receptores ErbB/genética , Receptores ErbB/uso terapêutico , Mutação
3.
J Cell Mol Med ; 26(18): 4859-4871, 2022 09.
Artigo em Inglês | MEDLINE | ID: mdl-35989423

RESUMO

RNA-binding Motif Protein39 (RBM39) is identified as a splicing factor and transcription coactivator. Despite mounting evidence that RBM39 plays a critical role in the development of specific malignancies, no systematic pan-cancer investigation of RBM39 has been conducted. As a result, we set out to investigate RBM39's prognostic significance and putative immunological activities in 33 different cancers. Based on TCGA and CCLE, GTEx, cBioportal and HPA, we used a series of bioinformatics approaches to explore the potential oncogenic role of RBM39, including analysis of the expression of the pan-cancer species RBM39, the prognostic relationship between RBM39 expression and overall survival (OS), disease-specific survival (DSS) and progression-free interval (PFI), the relationship between RBM39 expression and clinical phenotype, analysis of the relationship between RBM39 expression and tumour mutational burden (TMB), microsatellite instability (MSI), DNA methylation and immune cell infiltration. Our results showed that RBM39 is overexpressed in most cancers. RBM39 was positively or negatively correlated with the prognosis of different tumours. RBM39 expression was associated with TMB and MSI in 9 and 12 cancer types. In addition, RBM39 expression was associated with DNA methylation in almost all tumours. There are eight tumours were screened for further study, including BRCA, COAD, HNSC, LIHC, LUSC, SKCM, STAD, UCEC. In the screed tumours, RBM39 was found to be negatively correlated with the infiltration of most immune cells. In addition, the correlation with RBM39 expression varied by immune cell subtype. Based on RBM39's role in tumorigenesis and tumour immunity, we suggest it can serve as a surrogate prognostic marker.


Assuntos
Neoplasias , Proteínas de Ligação a RNA , Biomarcadores Tumorais/genética , Biomarcadores Tumorais/metabolismo , Humanos , Instabilidade de Microssatélites , Neoplasias/patologia , Prognóstico , Motivos de Ligação ao RNA , Proteínas de Ligação a RNA/genética , Proteínas de Ligação a RNA/metabolismo
4.
Childs Nerv Syst ; 38(10): 1867-1875, 2022 10.
Artigo em Inglês | MEDLINE | ID: mdl-35962792

RESUMO

OBJECTIVE: Therapeutic irradiation is commonly used to treat brain cancers but can induce cognitive dysfunction, especially in children. The mechanism is unknown but likely involves alterations in dendritic spine number and structure. METHODS: To explore the impact of radiation exposure on the alteration of dendritic spine morphology in the hippocampus of young brains, 21-day-old Sprague-Dawley rats received cranial irradiation (10 Gy), and changes in spine density and morphology in dentate gyrus (DG) granules and CA1 pyramidal neurons were detected 1 and 3 months later by using Golgi staining. Moreover, we analyzed synapse-associated proteins within dendritic spines after irradiation. RESULT: Our data showed that cognitive deficits were detected in young rats at both time points postirradiation, accompanied by morphological changes in dendritic spines. Our results revealed significant reductions in spine density in the DG at both 1 month (40.58%) and 3 months (28.92%) postirradiation. However, there was a decrease in spine density only at 1 month (33.29%) postirradiation in the basal dendrites of CA1 neurons and no significant changes in the apical dendrites of CA1 neurons at either time point. Notably, among our findings were the significant dynamic changes in spine morphology that persisted 3 months following cranial irradiation. Meanwhile, we found that depletion of the synapse-associated proteins PSD95 and Drebrin coincided with alterations in dendritic spines. CONCLUSION: These data suggest that the decreased levels of PSD95 and Drebrin after ionizing radiation may cause changes in synaptic plasticity by affecting the morphological structure of dendritic spines, blocking the functional connectivity pathways of the brain and leading to cognitive impairment. Although the mechanism involved is unclear, understanding how ionizing radiation affects young brain hippocampal tissue may be useful to gain new mechanistic insights into radiation-induced cognitive dysfunction.


Assuntos
Disfunção Cognitiva , Espinhas Dendríticas , Animais , Irradiação Craniana/efeitos adversos , Dendritos , Espinhas Dendríticas/efeitos da radiação , Hipocampo , Ratos , Ratos Sprague-Dawley
5.
Med Sci Monit ; 26: e923087, 2020 Jun 20.
Artigo em Inglês | MEDLINE | ID: mdl-32561704

RESUMO

BACKGROUND T-box transcription factor protein 21 (TBX21) is expressed in immune cells and some tumor cells. Defects in TBX21 gene can cause Th1/Th2 imbalance, which is closely related to tumorigenesis. The expression and clinical value of TBX21 in skin cutaneous melanoma (SKCM) are not clear. MATERIAL AND METHODS RNA-Seq expression and clinical information were downloaded from The Cancer Genome Atlas (TCGA) and Genotype-Tissue Expression (GTEx) databases. Wilcoxon signed-rank test and logistic regression were used to explore the relationship between TBX21 expression and clinical parameters such as gender, stage, etc. The correlation between clinicopathological characteristics and overall survival of SKCM patients was estimated by Cox regression and the Kaplan-Meier method. Gene set enrichment analysis (GSEA) and protein-protein interaction (PPI) were conducted to analyze the potential mechanism of TBX21 in the progression of SKCM. RESULTS Compared with normal samples, TBX21 was significantly upregulated in SKCM tissues. SKCM patients with lower TBX21 expression might have a worse prognosis than those with higher TBX21 expression according to Kaplan-Meier survival analysis. Cox analysis also reached the same conclusion: TBX21 was an independent prognostic indicator. GSEA showed that the highly expressed phenotypes in TBX21 were enriched to varying degrees with various signaling pathways. PPI network showed the top 10 proteins that were closely related to TBX21. CONCLUSIONS TBX21 expression was significantly correlated with the prognosis of SKCM patients and was found to be involved in a great many immunological pathways that affect the occurrence and development of tumors.


Assuntos
Melanoma/genética , Neoplasias Cutâneas/genética , Proteínas com Domínio T/genética , Bases de Dados Genéticas , Feminino , Expressão Gênica , Humanos , Estimativa de Kaplan-Meier , Modelos Logísticos , Metástase Linfática , Masculino , Melanoma/patologia , Pessoa de Meia-Idade , Metástase Neoplásica , Modelos de Riscos Proporcionais , Mapas de Interação de Proteínas , RNA-Seq , Neoplasias Cutâneas/patologia , Taxa de Sobrevida
6.
J Cell Mol Med ; 23(5): 3520-3529, 2019 05.
Artigo em Inglês | MEDLINE | ID: mdl-30821058

RESUMO

Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) can induce apoptosis in cancer cells while sparing normal cells, thereby leading to the development of TRAIL receptor agonists for cancer treatment. However, these agonist-based therapeutics exhibit little clinical benefits due to the lack of biomarkers to predict whether patients are responsive to the treatment, as well as determine the resistance of cancer cells to TRAIL-based agonists. Our previous study has demonstrated that ISG12a enhances TRAIL-induced apoptosis and might serve as a biomarker to predict the TRAIL response. The downstream mechanism by which ISG12a augments TRAIL-induced apoptosis remains to be elucidated. In this study, we found that ISG12a was localized in the mitochondria and nucleus and augmented TRAIL-induced apoptosis through intrinsic apoptotic pathway. In addition, ISG12a interacted with NR4A1 and promoted its nuclear-to-cytoplasm translocation. Upon translocate to cytoplasm, NR4A1 targeted mitochondria and induced Bcl2 conformational change, thereby exposing its BH3 domain. Moreover, TRAIL treatment can induce NR4A1 expression through the activation of NF-κB in TRAIL-resistant Huh7 hepatoma cells. Knockdown of NR4A1 could overcome TRAIL resistance. However, in TRAIL-sensitive LH86 liver cancer cells, TRAIL activated the Jun N-terminal kinases signalling pathway. Overall, these results showed that both ISG12a and its interaction partner NR4A1 are involved in TRAIL-mediated apoptosis in hepatoma cells.


Assuntos
Apoptose/efeitos dos fármacos , Proteínas de Membrana/metabolismo , Membro 1 do Grupo A da Subfamília 4 de Receptores Nucleares/metabolismo , Ligante Indutor de Apoptose Relacionado a TNF/farmacologia , Apoptose/genética , Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/metabolismo , Carcinoma Hepatocelular/patologia , Linhagem Celular Tumoral , Núcleo Celular/genética , Núcleo Celular/metabolismo , Células HEK293 , Humanos , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/patologia , Proteínas de Membrana/genética , Mitocôndrias/genética , Mitocôndrias/metabolismo , NF-kappa B/metabolismo , Membro 1 do Grupo A da Subfamília 4 de Receptores Nucleares/genética , Ligação Proteica , Proteínas Proto-Oncogênicas c-bcl-2/genética , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Interferência de RNA , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/genética
7.
Mol Pharm ; 16(3): 987-994, 2019 03 04.
Artigo em Inglês | MEDLINE | ID: mdl-30624945

RESUMO

A combination of different chemotherapy approaches can obtain the best response for many cancers. However, the greatest challenge is the development of a nanoparticle formulation that can encapsulate different chemotherapeutic agents to achieve the proper synergetic chemotherapy for the tumor. Here, amphiphilic ferrocenium-tetradecyl (Fe-C14) was constructed to form cationic micelles in an aqueous solution via self-assembly. Then, it was coated by hyaluronic acid (HA) through electrostatic interactions to generate HA-Fe-C14 micelles. The HA-Fe-C14 micelles were used to deliver doxorubicin (DOX), and it showed that the DOX could be released rapidly under a high-GSH tumor environment. The HA-Fe-C14/DOX micelles were able to accumulate efficiently in tumor and showed significant anticancer effect both in vitro and in vivo. These results suggest that HA-Fe-C14/DOX micelles are a useful drug delivery system that enhances synergic antitumor treatment effects.


Assuntos
Doxorrubicina/química , Doxorrubicina/uso terapêutico , Sistemas de Liberação de Medicamentos , Compostos Ferrosos/química , Glutationa/química , Ácido Hialurônico/química , Metalocenos/química , Micelas , Neoplasias/terapia , Alcanos/química , Animais , Sobrevivência Celular/efeitos dos fármacos , Terapia Combinada , Liberação Controlada de Fármacos , Compostos Ferrosos/síntese química , Humanos , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Neoplasias/patologia , Células PC-3 , Solubilidade , Resultado do Tratamento , Carga Tumoral
8.
Br J Cancer ; 118(4): 522-533, 2018 02 20.
Artigo em Inglês | MEDLINE | ID: mdl-29348487

RESUMO

BACKGROUND: The therapeutic resistance to ionising radiation (IR) and anti-angiogenesis mainly impair the prognosis of patients with glioblastoma. The primary and secondary MET aberrant activation is one crucial factor for these resistances. The kringle 1 domain of hepatocyte growth factor (HGFK1), an angiogenic inhibitor, contains a high-affinity binding domain of MET; however, its effects on glioblastoma remain elusive. METHODS: We formed the nanoparticles consisting of a folate receptor-targeted nanoparticle-mediated HGFK1 gene (H1/pHGFK1) and studied its anti-tumoural and radiosensitive activities in both subcutaneous and orthotopic human glioma cell-xenografted mouse models. We then elucidated its molecular mechanisms in human glioblastoma cell lines in vitro. RESULTS: We demonstrated for the first time that peritumoural injection of H1/pHGFK1 nanoparticles significantly inhibited tumour growth and prolonged survival in tumour-bearing mice, as well as enhanced the anti-tumoural efficacies of IR in vivo by reducing Ki-67 expression, enhancing TUNEL staining-indicated apoptotic indexes, reducing microvascular intensity and reversing IR-induced MET overexpression in tumour tissues. Furthermore, we showed that HGFK1 suppressed the proliferation and induced cell apoptosis and enhanced sensitivity to IR in glioblastoma cell lines, mainly by suppressing the activities of MET receptor, down-regulating ATM-Chk2 axis but up-regulating Chk1. CONCLUSIONS: H1/pHGFK1 exerts anti-tumoural and radiosensitive activities mainly through the inhibition and reversal of IR-induced MET and ATM-Chk2 axis activities in glioblastoma. H1/pHGFK1 nanoparticles are a potential radiosensitiser and angiogenic inhibitor for glioblastoma treatment.


Assuntos
Neoplasias Encefálicas/terapia , Glioblastoma/terapia , Fator de Crescimento de Hepatócito/genética , Plasmídeos/administração & dosagem , Proteínas Proto-Oncogênicas c-met/antagonistas & inibidores , Radiossensibilizantes/administração & dosagem , Animais , Neoplasias Encefálicas/genética , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Proliferação de Células/efeitos da radiação , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos da radiação , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Regulação Neoplásica da Expressão Gênica/efeitos da radiação , Glioblastoma/genética , Fator de Crescimento de Hepatócito/química , Humanos , Kringles , Camundongos , Nanopartículas/administração & dosagem , Plasmídeos/genética , Radiossensibilizantes/farmacologia , Ensaios Antitumorais Modelo de Xenoenxerto
9.
Nanomedicine ; 13(4): 1309-1321, 2017 05.
Artigo em Inglês | MEDLINE | ID: mdl-28185938

RESUMO

Androgen receptor (AR) plays a crucial role in the development and progression of prostate cancer (PCa). PCa patients typically receive androgen deprivation therapy; nonetheless, these patients eventually develop castration and radiation resistance. We hypothesized that we could further improve radiotherapeutic efficacy of hormone-independent PCa (HIPC) by silencing AR. In this study, nanoparticle (NP) AR-shRNA was formulated using folate-targeted H1 nanopolymer. We demonstrated that NP AR-shRNA enhances PCa radiosensitivity as indicated by the inhibition of cell growth, increased apoptosis, and increased cell cycle arrest in AR-dependent HIPC in vitro. The radiosensitizing effect of NP AR-shRNA could be validated in vivo, as NP AR-shRNA significantly suppressed tumor growth and prolonged the survival of HIPC tumor-bearing mice. Analysis at the molecular level revealed that NP AR-shRNA inhibits DNA damage repair signaling pathways. Our study supports further investigation of NP AR-shRNA for the improvement of radiotherapy efficacy in HIPC.


Assuntos
Antagonistas de Receptores de Andrógenos/uso terapêutico , Ácido Fólico/química , Nanopartículas/química , Neoplasias da Próstata/radioterapia , RNA Interferente Pequeno/uso terapêutico , Radiossensibilizantes/uso terapêutico , Animais , Linhagem Celular Tumoral , Humanos , Masculino , Camundongos , Camundongos Nus , Tolerância a Radiação , Ensaios Antitumorais Modelo de Xenoenxerto
10.
Sensors (Basel) ; 17(10)2017 Sep 21.
Artigo em Inglês | MEDLINE | ID: mdl-28934171

RESUMO

Because mobile ad hoc networks have characteristics such as lack of center nodes, multi-hop routing and changeable topology, the existing checkpoint technologies for normal mobile networks cannot be applied well to mobile ad hoc networks. Considering the multi-frequency hierarchy structure of ad hoc networks, this paper proposes a hybrid checkpointing strategy which combines the techniques of synchronous checkpointing with asynchronous checkpointing, namely the checkpoints of mobile terminals in the same cluster remain synchronous, and the checkpoints in different clusters remain asynchronous. This strategy could not only avoid cascading rollback among the processes in the same cluster, but also avoid too many message transmissions among the processes in different clusters. What is more, it can reduce the communication delay. In order to assure the consistency of the global states, this paper discusses the correctness criteria of hybrid checkpointing, which includes the criteria of checkpoint taking, rollback recovery and indelibility. Based on the designed Intra-Cluster Checkpoint Dependence Graph and Inter-Cluster Checkpoint Dependence Graph, the elimination rules for different kinds of checkpoints are discussed, and the algorithms for the same cluster checkpoints, different cluster checkpoints, and rollback recovery are also given. Experimental results demonstrate the proposed hybrid checkpointing strategy is a preferable trade-off method, which not only synthetically takes all kinds of resource constraints of Ad hoc networks into account, but also outperforms the existing schemes in terms of the dependence to cluster heads, the recovery time compared to the pure synchronous, and the pure asynchronous checkpoint advantage.

11.
Cancer Immunol Immunother ; 65(3): 305-14, 2016 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-26842126

RESUMO

T cell Ig and ITIM domain (TIGIT) is a newly identified inhibitory receptor expressed on T and natural killer (NK) cells. Cytokine-induced killer (CIK) cells express CD3 and CD56 molecules, and share functional properties with both NK and T cells. However, it remains unknown whether TIGIT is expressed in CIK cells. Here, we show that TIGIT is expressed by CIK cells and interacts with CD155. By blocking TIGIT using an anti-TIGIT functional antibody, we demonstrate that CIK cells display increased proliferation; higher cytotoxic targeting of tumor cells expressing CD155; and higher expression of interferon-γ (IFN-γ), interleukin-6 (IL-6), and tumor necrosis factor-α (TNF-α). Furthermore, increases in IFN-γ and cytotoxicity by blockade of TIGIT were reduced by blocking DNAX accessory molecule-1 (DNAM-1) signaling, implying that TIGIT exerts immunosuppressive effects by competing with DNAM-1 for the same ligand, CD155. Our results provide evidence that blockade of TIGIT may be a novel strategy to improve the cytotoxic activity of CIK cells.


Assuntos
Células Matadoras Induzidas por Citocinas/imunologia , Citotoxicidade Imunológica , Receptores Imunológicos/fisiologia , Receptores Virais/fisiologia , Antígenos de Diferenciação de Linfócitos T/fisiologia , Linhagem Celular Tumoral , Humanos , Transdução de Sinais/fisiologia
12.
Nanomedicine ; 12(8): 2261-2271, 2016 11.
Artigo em Inglês | MEDLINE | ID: mdl-27389144

RESUMO

Intensification of radiotherapy has been shown to improve prostate cancer (PCa) outcomes. We hypothesized that we could further improve radiotherapy efficacy through the use DNA repair inhibitors. In this study, we evaluated the use of a new class of DNA damage repair inhibitor, nanoparticle (NP) Dbait, in radiosensitization of PCa. NP Dbait was formulated using H1 nanopolymer (folate-polyethylenimine600-cyclodextrin). We demonstrated that NP Dbait was a potent radiosensitizer in vitro by colony forming assay using PCa cell lines. The result was validated in vivo using mouse xenograft models of PCa and we showed that NP Dbait significantly suppressed tumor growth and prolonged survival. Western blot, immunofluorescence and immunohistochemistry showed that NP Dbait inhibited DNA damage repair signaling pathways by mimicking DNA double-strand breaks. Our study supports further investigations of NP Dbait in improving the therapeutic efficacy of cancer radiotherapy.


Assuntos
Reparo do DNA , Nanopartículas , Neoplasias da Próstata/radioterapia , Radiossensibilizantes/administração & dosagem , Animais , DNA , Humanos , Masculino , Camundongos , Radioterapia/métodos
13.
Tumour Biol ; 35(9): 9387-94, 2014 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-24951956

RESUMO

The aim of our study was to elucidate the role of Rap2B in the development of human suprarenal epithelioma and to investigate the effect of Rap2B on suprarenal epithelioma cells migration and invasion. We use tissue microarray and immunohistochemistry to evaluate Rap2B staining in 75 suprarenal epithelioma tissues and 75 tumor-adjacent normal renal tissues. And the expression of Rap2B protein in human suprarenal epithelioma cells and tissues was detected by western blot simultaneously. The role of Rap2B in suprarenal epithelioma cells migration and invasion was detected by using wound healing assay, cell migration assay, and matrigel invasion assay. After that, we performed western blot analysis and gelatin zymography to detect MMP-2 protein expression and enzyme activity. Our research showed that Rap2B expression was increased in tumor tissues compared with tumor-adjacent normal renal tissues. But no correlation was found between Rap2B expression and clinicopathological parameters. In addition, we found that Rap2B promoted the cell migration and invasion abilities, and Rap2B increased MMP-2 expression and enzyme activity in suprarenal epithelioma cells. Our data indicated that Rap2B expression is significantly increased in human suprarenal epithelioma and Rap2B can promote the cell migration and invasion abilities, which may provide a new target for the treatment of suprarenal epithelioma.


Assuntos
Carcinoma de Células Renais/metabolismo , Movimento Celular , Neoplasias Renais/metabolismo , Proteínas rap de Ligação ao GTP/metabolismo , Western Blotting , Carcinoma de Células Renais/genética , Carcinoma de Células Renais/patologia , Linhagem Celular Tumoral , Feminino , Humanos , Imuno-Histoquímica , Neoplasias Renais/genética , Neoplasias Renais/patologia , Masculino , Metaloproteinase 2 da Matriz/metabolismo , Pessoa de Meia-Idade , Invasividade Neoplásica , Interferência de RNA , Análise Serial de Tecidos , Proteínas rap de Ligação ao GTP/genética
14.
Water Res ; 256: 121568, 2024 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-38593607

RESUMO

Constructed wetlands (CWs) are widely used in sewage treatment in rural areas, but there are only a few studies on field-scale CWs in treating wastewater-borne pesticides. In this study, the treatment and metabolic transformation of 29 pesticides in rural domestic sewage by 10 field-scale horizontal flow CWs (HF-CWs), each with a treatment scale of 36‒5000 m3/d and operated for 2‒10 years, in Guangzhou, Southern China was investigated. The risk of pesticides in treated effluent and main factors influencing such risk were evaluated. Results demonstrated that HF-CWs could remove pesticides in sewage and reduce their ecological risk in effluent, but the degree varied among types of pesticides. Herbicides had the highest mean removal rate (67.35 %) followed by insecticides (60.13 %), and the least was fungicides (53.22 %). In terms of single pesticide compounds, the mean removal rate of butachlor was the highest (73.32 %), then acetochlor (69.41 %), atrazine (68.28 %), metolachlor (58.40 %), and oxadixyl (53.28 %). The overall removal rates of targeted pesticides in each HF-CWs ranged from 11 %‒57 %, excluding two HF-CWs showing increases in pesticides in treated effluent. Residues of malathion, phorate, and endosulfan in effluent had high-risks (RQ > 5). The pesticide concentration in effluent was mainly affected by that in influent (P = 0.042), and source control was the key to reducing risk. The main metabolic pathways of pesticide in HF-CWs were oxidation, with hydroxyl group to carbonyl group or to form sulfones, the second pathways by hydrolysis, aerobic condition was conducive to the transformation of pesticides. Sulfones were generally more toxic than the metabolites produced by hydrolytic pathways. The present study provides a reference on pesticides for the purification performance improvement, long-term maintenance, and practical sustainable application of field-scale HF-CWs.


Assuntos
Praguicidas , Águas Residuárias , Poluentes Químicos da Água , Áreas Alagadas , Águas Residuárias/química , Medição de Risco , Eliminação de Resíduos Líquidos , China
16.
Biochem Pharmacol ; 219: 115950, 2024 01.
Artigo em Inglês | MEDLINE | ID: mdl-38043718

RESUMO

Metabolic network intertwines with cancerous signaling and drug responses. Malonate is a prevailing metabolite in cancer and a competitive inhibitor of succinate dehydrogenase (SDH). Recent studies showed that malonate induced reactive oxygen species (ROS)-dependent apoptosis in neuroblastoma cells, but protected cells from ischemia-reperfusion injury. We here revealed that malonate differentially regulated cell death and survival in cancer cells. While high-dose malonate triggered ROS-dependent apoptosis, the low-dose malonate induced autophagy and conferred resistance to multiple chemotherapeutic agents. Mechanistically, our results showed that malonate increased p53 stability and transcriptionally up-regulated autophagy modulator DRAM (damage-regulated autophagy modulator), thus promoting autophagy. We further proved that autophagy is required for malonate-associated chemoresistance. Collectively, our findings suggest that malonate plays a double-edge function in cancer response to stressors, and highlights a pro-cancer impact of p53-induced autophagy in response to malonate.


Assuntos
Neoplasias , Proteína Supressora de Tumor p53 , Humanos , Espécies Reativas de Oxigênio/metabolismo , Proteína Supressora de Tumor p53/genética , Proteína Supressora de Tumor p53/metabolismo , Sobrevivência Celular , Resistencia a Medicamentos Antineoplásicos , Apoptose , Autofagia , Malonatos/farmacologia , Linhagem Celular Tumoral
17.
Cancer Commun (Lond) ; 2024 Jul 17.
Artigo em Inglês | MEDLINE | ID: mdl-39016053

RESUMO

BACKGROUND: The initial randomized, double-blinded, actively controlled, phase III ANEAS study (NCT03849768) demonstrated that aumolertinib showed superior efficacy relative to gefitinib as first-line therapy in epidermal growth factor receptor (EGFR)-mutated advanced non-small cell lung cancer (NSCLC). Metastatic disease in the central nervous system (CNS) remains a challenge in the management of NSCLC. This study aimed to compare the efficacy of aumolertinib versus gefitinib among patients with baseline CNS metastases in the ANEAS study. METHODS: Eligible patients were enrolled and randomly assigned in a 1:1 ratio to orally receive either aumolertinib or gefitinib in a double-blinded fashion. Patients with asymptomatic, stable CNS metastases were included. Follow-up imaging of the same modality as the initial CNS imaging was performed every 6 weeks for 15 months, then every 12 weeks. CNS response was assessed by a neuroradiological blinded, independent central review (neuroradiological-BICR). The primary endpoint for this subgroup analysis was CNS progression-free survival (PFS). RESULTS: Of the 429 patients enrolled and randomized in the ANEAS study, 106 patients were found to have CNS metastases (CNS Full Analysis Set, cFAS) at baseline by neuroradiological-BICR, and 60 of them had CNS target lesions (CNS Evaluable for Response, cEFR). Treatment with aumolertinib significantly prolonged median CNS PFS compared with gefitinib in both cFAS (29.0 vs. 8.3 months; hazard ratio [HR] = 0.31; 95% confidence interval [CI], 0.17-0.56; P < 0.001) and cEFR (29.0 vs. 8.3 months; HR = 0.26; 95% CI, 0.11-0.57; P < 0.001). The confirmed CNS overall response rate in cEFR was 85.7% and 75.0% in patients treated with aumolertinib and gefitinib, respectively. Competing risk analysis showed that the estimated probability of CNS progression without prior non-CNS progression or death was consistently lower with aumolertinib than with gefitinib in patients with and without CNS metastases at baseline. No new safety findings were observed. CONCLUSIONS: These results indicate a potential advantage of aumolertinib over gefitinib in terms of CNS PFS and the risk of CNS progression in patients with EGFR-mutated advanced NSCLC with baseline CNS metastases. TRIAL REGISTRATION: ClinicalTrials.gov number, NCT03849768.

18.
JAMA Oncol ; 10(4): 448-455, 2024 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-38329745

RESUMO

Importance: The bioequivalence of denosumab biosimilar has yet to be studied in a 53-week, multicenter, large-scale, and head-to-head trial. A clinically effective biosimilar may help increase access to denosumab in patients with solid tumor-related bone metastases. Objectives: To establish the biosimilarity of MW032 to denosumab in patients with solid tumor-related bone metastases based on a large-scale head-to-head study. Design, Setting, and Participants: In this 53-week, randomized, double-blind, phase 3 equivalence trial, patients with solid tumors with bone metastasis were recruited from 46 clinical sites in China. Overall, 856 patients were screened and 708 eligible patients were randomly allocated to receive either MW032 or denosumab. Interventions: Patients were randomly assigned (1:1) to receive MW032 or reference denosumab subcutaneously every 4 weeks until week 49. Main Outcomes and Measures: The primary end point was percentage change from baseline to week 13 of natural logarithmic transformed urinary N-telopeptide/creatinine ratio (uNTx/uCr). Results: Among the 701 evaluable patients (350 in the MW032 group and 351 in the denosumab group), the mean (range) age was 56.1 (22.0-86.0) years and 460 patients were women (65.6%). The mean change of uNTx/uCr from baseline to week 13 was -72.0% (95% CI, -73.5% to -70.4%) in the MW032 group and -72.7% (95% CI, -74.2% to -71.2%) in the denosumab group. These percent changes corresponded to mean logarithmic ratios of -1.27 and -1.30, or a difference of 0.02. The 90% CI for the difference (-0.04 to 0.09) was within the equivalence margin (-0.13 to 0.13); the mean changes of uNTx/uCr and bone-specific alkaline phosphatase (s-BALP) at each time point were also similar during 53 weeks. The differences of uNTx/uCr change were 0.015 (95% CI, -0.06 to 0.09), -0.02 (95% CI, -0.09 to 0.06), -0.05 (95% CI, -0.13 to 0.03) and 0.001 (95% CI, -0.10 to 0.10) at weeks 5, 25, 37, and 53, respectively. The differences of s-BALP change were -0.006 (95% CI, 0.06 to 0.05), 0.00 (95% CI, -0.07 to 0.07), -0.085 (95% CI, -0.18 to 0.01), -0.09 (95% CI, -0.20 to 0.02), and -0.13 (95% CI, -0.27 to 0.004) at weeks 5, 13, 25, 37 and 53, respectively. No significant differences were observed in the incidence of skeletal-related events (-1.4%; 95% CI, -5.8% to 3.0%) or time to first on-study skeletal-related events (unadjusted HR, 0.86; P = .53; multiplicity adjusted HR, 0.87; P = .55) in the 2 groups. Conclusions and Relevance: MW032 and denosumab were biosimilar in efficacy, population pharmacokinetics, and safety profile. Availability of denosumab biosimilars may broaden the access to denosumab and reduce the drug burden for patients with advanced tumors. Trial Registration: ClinicalTrials.gov Identifier: NCT04812509.


Assuntos
Medicamentos Biossimilares , Neoplasias Ósseas , Humanos , Feminino , Pessoa de Meia-Idade , Idoso , Idoso de 80 Anos ou mais , Masculino , Denosumab , Anticorpos Monoclonais Humanizados , Neoplasias Ósseas/secundário , Creatinina , Método Duplo-Cego
19.
Fish Physiol Biochem ; 39(4): 1025-30, 2013 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-23248051

RESUMO

We studied the effects of salinity on survival, growth, food conversion efficiency and plasma osmolality of juvenile Siganus guttatus in two independent experiments. In the first experiment, fish were transferred from 30 ‰ salinity to freshwater, 5, 10, 20 and 30‰ salinities for 192 h. No fish died when transferred directly from 30‰ to salinities >5‰. However, all fish died in the freshwater treatment. In the second experiment, survival, growth, feeding rate, food conversion efficiency and plasma osmolality of fish were analyzed during 6 weeks in salinities of 5, 10, 20 and 30‰ (control). At the end of this experiment, the final weight and the specific growth rate of fish were significantly greater at 10‰ than fish in all other treatments. Feeding rate increased significantly with decreasing salinity: 10‰ > 20‰ > 30‰. However, the food conversion efficiency was not significantly different between fish in any treatment. Plasma osmolality of fish in 20 and 30‰ salinity was significantly greater than fish reared at 10 or 5‰. A salinity of 13.95‰ (411.88 mOsmol/kg) was the point of isosmolality for juvenile S. guttatus.


Assuntos
Digestão , Perciformes/crescimento & desenvolvimento , Salinidade , Animais , Aquicultura , Concentração Osmolar , Perciformes/sangue , Sobrevida
20.
J Clin Transl Hepatol ; 11(2): 369-381, 2023 Apr 28.
Artigo em Inglês | MEDLINE | ID: mdl-36643033

RESUMO

Background and Aims: Emerging evidence suggests that RNA-binding motif (RBM) proteins are involved in hepatocarcinogenesis and act either as oncogenes or tumor suppressors. The objective of this study was to investigate the role of RBM34, an RBM protein, in hepatocellular carcinoma (HCC). Methods: We first examined the expression of RBM34 across cancers. The correlation of RBM34 with clinicopathological features and the prognostic value of RBM34 for HCC was then investigated. Functional enrichment analysis of RBM34-related differentially expressed genes (DEGs) was performed to explore its biological function. RNA sequencing (RNA-seq) was applied to identify downstream genes and pathways affected upon RBM34 knockout. The correlation of RBM34 with immune characteristics was also analyzed. The oncogenic function of RBM34 was examined in in vitro and in vivo experiments. Results: RBM34 was highly expressed in hepatocellular carcinoma and correlated with poor clinicopathological features and prognosis. RBM34 was positively associated with tumor immune cell infiltration, biomarkers of immune cells, and immune checkpoint expression. A positive correlation was also observed between RBM34, T cell exhaustion, and regulatory T cell marker genes. Knockout of RBM34 significantly inhibited cell proliferation, migration, and xenograft tumor growth, and sensitized HCC cells to sorafenib treatment. RBM34 inhibition reduced FGFR2 expression and affected PI3K-AKT pathway activation in HCC cells. Conclusions: Our study suggests that RBM34 may serve as a new prognostic marker and therapeutic target of HCC.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA