Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 59
Filtrar
1.
Front Oncol ; 14: 1350011, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38511140

RESUMO

Hepatocellular carcinoma (HCC) is a prevalent malignant tumor worldwide, characterized by high malignancy and rapid progression. Most cases are diagnosed at intermediate to advanced stages. Current treatment methods have limited efficacy, resulting in high recurrence rates and poor prognosis. Radical hepatectomy remains the primary treatment for HCC, complemented by radiotherapy, chemotherapy, targeted therapy, and immunotherapy. Despite significant improvement in patient prognosis with radical hepatectomy, the five-year survival rate post-surgery remains low; thus necessitating exploration of more effective therapeutic approaches. Ferroptosis is a recently discovered form of cell death that can modulate the occurrence and development of HCC through various mechanisms. This article aims to elucidate the mechanism of ferroptosis and its impact on HCC development to provide novel insights for diagnosis and treatment.

2.
Cell Death Dis ; 15(3): 209, 2024 Mar 13.
Artigo em Inglês | MEDLINE | ID: mdl-38480704

RESUMO

Metabolic reprogramming, a hallmark of cancer, is closely associated with tumor development and progression. Changes in glycolysis play a crucial role in conferring radiation resistance to tumor cells. How radiation changes the glycolysis status of cancer cells is still unclear. Here we revealed the role of TAB182 in regulating glycolysis and lactate production in cellular response to ionizing radiation. Irradiation can significantly stimulate the production of TAB182 protein, and inhibiting TAB182 increases cellular radiosensitivity. Proteomic analysis indicated that TAB182 influences several vital biological processes, including multiple metabolic pathways. Knockdown of TAB182 results in decreased lactate production and increased pyruvate and ATP levels in cancer cells. Moreover, knocking down TAB182 reverses radiation-induced metabolic changes, such as radioresistant-related lactate production. TAB182 is necessary for activating LDHA transcription by affecting transcription factors SP1 and c-MYC; its knockdown attenuates the upregulation of LDHA by radiation, subsequently suppressing lactate production. Targeted suppression of TAB182 significantly enhances the sensitivity of murine xenograft tumors to radiotherapy. These findings advance our understanding of glycolytic metabolism regulation in response to ionizing radiation, which may offer significant implications for developing new strategies to overcome tumor radioresistance.


Assuntos
L-Lactato Desidrogenase , Proteômica , Humanos , Animais , Camundongos , L-Lactato Desidrogenase/metabolismo , Lactato Desidrogenase 5/metabolismo , Linhagem Celular Tumoral , Glicólise , Lactatos , Tolerância a Radiação/genética
3.
J Oncol ; 2022: 7840606, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36532884

RESUMO

Primary liver carcinoma is the sixth most common cancer worldwide, while hepatocellular carcinoma (HCC) is the most dominant cancer type. Chronic hepatitis B and C virus infections and aflatoxin exposure are the main risk factors, while nonalcoholic fatty liver disease caused by obesity, diabetes, and metabolic syndrome are the more common risk factors for HCC. Metabolic disorders caused by these high-risk factors are closely related to the tumor microenvironment of HCC, revealing a possible cause-and-effect relationship between the two. These metabolic disorders involve many complex metabolic pathways, such as carbohydrate, lipid, lipid derivative, amino acid, and amino acid derivative metabolic processes. The resulting metabolites with significant abnormal changes in the concentration level in circulating blood may be used as biomarkers to guide the diagnosis, treatment, or prognosis of HCC. At present, there are high-throughput technologies that can quickly detect small molecular metabolites in many samples. Compared to tissue biopsy, blood samples are easier to obtain, and patients' willingness to participate is higher, which makes it possible to study blood HCC biomarkers. Over the past few years, a substantial body of research has been performed worldwide, and other potential biomarkers have been identified. Unfortunately, due to the limitations of each study, only a few markers have been widely verified and are suitable for clinical use. This review briefly summarizes the potential blood metabolic markers related to the diagnosis of HCC, mainly focusing on amino acids and their derivative metabolism, lipids and their derivative metabolism, and other possible related metabolisms.

5.
J Transl Med ; 20(1): 456, 2022 10 05.
Artigo em Inglês | MEDLINE | ID: mdl-36199069

RESUMO

BACKGROUND: Epithelial-to-mesenchymal transition (EMT) is a critical event contributing to more aggressive phenotypes in cancer cells. EMT is frequently activated in radiation-targeted cells during the course of radiotherapy, which often endows cancers with acquired radioresistance. However, the upstream molecules driving the signaling pathways of radiation-induced EMT have not been fully delineated. METHODS: In this study, RNA-seq-based transcriptome analysis was performed to identify the early responsive genes of HeLa cells to γ-ray irradiation. EMT-associated genes were knocked down by siRNA technology or overexpressed in HeLa cells and A549 cells, and the resulting changes in phenotypes of EMT and radiosensitivity were assessed using qPCR and Western blotting analyses, migration assays, colony-forming ability and apoptosis of flow cytometer assays. RESULTS: Through RNA-seq-based transcriptome analysis, we found that LPAR5 is downregulated in the early response of HeLa cells to γ-ray irradiation. Radiation-induced alterations in LPAR5 expression were further revealed to be a bidirectional dynamic process in HeLa and A549 cells, i.e., the early downregulating phase at 2 ~ 4 h and the late upregulating phase at 24 h post-irradiation. Overexpression of LPAR5 prompts EMT programing and migration of cancer cells. Moreover, increased expression of LPAR5 is significantly associated with IR-induced EMT and confers radioresistance to cancer cells. Knockdown of LPAR5 suppressed IR-induced EMT by attenuating the activation of ERK signaling and downstream Snail, MMP1, and MMP9 expression. CONCLUSIONS: LPAR5 is an important upstream regulator of IR-induced EMT that modulates the ERK/Snail pathway. This study provides further insights into understanding the mechanism of radiation-induced EMT and identifies promising targets for improving the effectiveness of cancer radiation therapy.


Assuntos
Metaloproteinase 1 da Matriz , Neoplasias , Linhagem Celular Tumoral , Transição Epitelial-Mesenquimal/genética , Células HeLa , Humanos , Metaloproteinase 9 da Matriz , RNA Interferente Pequeno , Receptores de Ácidos Lisofosfatídicos
6.
Cytokine ; 159: 156021, 2022 11.
Artigo em Inglês | MEDLINE | ID: mdl-36057231

RESUMO

BACKGROUND: Macrophages are highly enriched in renal cell carcinoma, and the inflammatory cytokines secreted by macrophages are remarkably associated with the survival rate of renal cell carcinoma. However, the relationship between gasdermin D (GSDMD) expression driven by macrophage and the invasion of renal cell carcinoma is not clear. METHODS: The Caki-2 and 786-O cells were co-cultured with monocytes cells (THP-1) derived macrophages, then the bio function changes of Caki-2 and 786-O cells and epithelial-mesenchymal transition of cancer cells were detected. Also, the role of IL-1ß in Caki-2 and 786-O cells and macrophage interaction were investigated. Then, the animal model was used to confirm the role of communication of GSDMD with renal cell carcinoma in the tumor microenvironment. RESULTS: CD68 and GSDMD were overexpressed in human renal cell carcinoma. GSDMD contributed to the secretion of IL­1ß in macrophages and was associated with the proliferation rate of renal cell carcinoma cells. Furthermore, silencing GSDMD elicited renal cell carcinoma cells motility through epithelial-mesenchymal transition change. The in vivo study confirmed that GSDMD promoted tumor progression and GSDMD knockout impaired renal cell carcinoma growth and metastases. Finally, the interactions between macrophages and renal cell carcinoma cells promoted renal cell carcinoma proliferation and metastasis, possibly mediated by IL-1ß. CONCLUSION: To our knowledge, this study showed that the GSDMD expressed by macrophages contributed to renal cell carcinoma cell growth, metastases, and epithelial-mesenchymal transition through regulating GSDMD/IL-1ß axis and may be a novel therapeutic target and a potential biomarker for treating and diagnosing renal cell carcinoma.


Assuntos
Carcinoma de Células Renais , Neoplasias Renais , Animais , Biomarcadores/metabolismo , Citocinas/metabolismo , Transição Epitelial-Mesenquimal , Humanos , Interleucina-1beta , Peptídeos e Proteínas de Sinalização Intracelular/genética , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Macrófagos/metabolismo , Proteínas de Ligação a Fosfato/metabolismo , Proteínas Citotóxicas Formadoras de Poros , Microambiente Tumoral
7.
Gen Physiol Biophys ; 41(3): 231-243, 2022 May.
Artigo em Inglês | MEDLINE | ID: mdl-35616003

RESUMO

We attempted to discover the biomarker associated with metastasis and prognosis of lung adenocarcinoma. The mRNA/lncRNA expression profiles (GSE101836) were downloaded from the publicly available database, which included three highly metastatic and three weakly metastatic samples. The differentially expressed genes and lncRNAs were analyzed and survival analysis were performed based on the TCGA database. The prognosis-associated PPI network and mRNA-lncRNA coexpression network were constructed followed by the function and pathway enrichment analysis. The expression levels of key genes were validated in other datasets. Difference in gender was analyzed. Total 256 differentially expressed genes and 2 lncRNAs were found to be closely related with prognosis. PPI network was constructed with 222 nodes and 1464 edges. Two modules were divided from PPI network. Genes in module A were significantly enriched in cell cycle checkpoint, chromosome segregation, and mitotic cell cycle checkpoint. The module B was closely related with pyridine nucleotide metabolic process, nicotinamide nucleotide metabolic process and carbon metabolism. Coexpression network revealed lncRNA H19 and lncRNA SNHG12 were significant nodes. SNHG12 was closely related with GO:0006260~DNA replication, GO:0055114~oxidation-reduction process and hsa00010: Glycolysis/Gluconeogenesis. H19 was enriched in GO:0006555~methionine metabolic process, and GO:0046655~folic acid metabolic process. The expression levels of TTK and CCNB1 were confirmed in other datasets. The expression of TTK and CCNB1 was significantly higher in the male group than in the female group. TTK, CCNB1 and lncRNA SNHG12 may be the biomarker associated with metastasis and prognosis of lung adenocarcinoma.


Assuntos
Adenocarcinoma , Neoplasias Pulmonares , RNA Longo não Codificante , Adenocarcinoma/genética , Biomarcadores , Biomarcadores Tumorais/genética , Biologia Computacional , Feminino , Regulação Neoplásica da Expressão Gênica , Humanos , Pulmão , Neoplasias Pulmonares/genética , Masculino , Nucleotídeos , RNA Longo não Codificante/genética , RNA Mensageiro/genética
8.
Acta Pharmacol Sin ; 43(11): 2895-2904, 2022 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-35468992

RESUMO

Angiotensin-converting enzyme 2 (ACE2) is the receptor of COVID-19 pathogen SARS-CoV-2, but the transcription factors (TFs) that regulate the expression of the gene encoding ACE2 (ACE2) have not been systematically dissected. In this study we evaluated TFs that control ACE2 expression, and screened for small molecule compounds that could modulate ACE2 expression to block SARS-CoV-2 from entry into lung epithelial cells. By searching the online datasets we found that 24 TFs might be ACE2 regulators with signal transducer and activator of transcription 3 (Stat3) as the most significant one. In human normal lung tissues, the expression of ACE2 was positively correlated with phosphorylated Stat3 (p-Stat3). We demonstrated that Stat3 bound ACE2 promoter, and controlled its expression in 16HBE cells stimulated with interleukin 6 (IL-6). To screen for medicinal compounds that could modulate ACE2 expression, we conducted luciferase assay using HLF cells transfected with ACE2 promoter-luciferase constructs. Among the 64 compounds tested, 6-O-angeloylplenolin (6-OAP), a sesquiterpene lactone in Chinese medicinal herb Centipeda minima (CM), represented the most potent ACE2 repressor. 6-OAP (2.5 µM) inhibited the interaction between Stat3 protein and ACE2 promoter, thus suppressed ACE2 transcription. 6-OAP (1.25-5 µM) and its parental medicinal herb CM (0.125%-0.5%) dose-dependently downregulated ACE2 in 16HBE and Beas-2B cells; similar results were observed in the lung tissues of mice following administration of 6-OAP or CM for one month. In addition, 6-OAP/CM dose-dependently reduced IL-6 production and downregulated chemokines including CXCL13 and CX3CL1 in 16HBE cells. Moreover, we found that 6-OAP/CM inhibited the entry of SARS-CoV-2 S protein pseudovirus into target cells. These results suggest that 6-OAP/CM are ACE2 inhibitors that may potentially protect lung epithelial cells from SARS-CoV-2 infection.


Assuntos
Enzima de Conversão de Angiotensina 2 , Tratamento Farmacológico da COVID-19 , Camundongos , Humanos , Animais , SARS-CoV-2 , Interleucina-6/metabolismo , Pulmão/metabolismo , Células Epiteliais
9.
Cell Calcium ; 102: 102527, 2022 03.
Artigo em Inglês | MEDLINE | ID: mdl-35026540

RESUMO

CACNA1E is a gene encoding the ion-conducting α1 subunit of R-type voltage-dependent calcium channels, whose roles in tumorigenesis remain to be determined. We previously showed that CACNA1E was significantly mutated in patients with non-small cell lung cancer (NSCLC) who were long-term exposed to household air pollution, with a mutation rate of 19% (15 of 79 cases). Here we showed that CACNA1E was also mutated in 207 (12.8%) of the 1616 patients with NSCLC in The Cancer Genome Atlas (TCGA) datasets. At mRNA and protein levels, CACNA1E was elevated in tumor tissues compared to counterpart non-tumoral lung tissues in NSCLCs of the public datasets and our settings, and its expression level was inversely associated with clinical outcome of the patients. Overexpression of wild type (WT) or A275S or R249G mutant CACNA1E transcripts promoted NSCLC cell proliferation with activation of epidermal growth factor receptor (EGFR) signaling pathway, whereas knockdown of this gene exerted inhibitory effects on NSCLC cells in vitro and in vivo. CACNA1E increased current density and Ca2+ entrance, whereas calcium channel blockers inhibited NSCLC cell proliferation. These data indicate that CACNA1E is required for NSCLC cell proliferation, and blockade of this oncoprotein may have therapeutic potentials for this deadly disease.


Assuntos
Carcinoma Pulmonar de Células não Pequenas , Neoplasias Pulmonares , Cálcio/metabolismo , Canais de Cálcio Tipo R , Carcinoma Pulmonar de Células não Pequenas/genética , Carcinoma Pulmonar de Células não Pequenas/metabolismo , Carcinoma Pulmonar de Células não Pequenas/patologia , Proteínas de Transporte de Cátions , Linhagem Celular Tumoral , Humanos , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patologia , Mutação/genética
10.
Acta Pharmacol Sin ; 43(3): 692-702, 2022 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-34035487

RESUMO

The tumor suppressor p53 is usually inactivated by somatic mutations in malignant neoplasms, and its reactivation represents an attractive therapeutic strategy for cancers. Here, we reported that a new quinolone compound RYL-687 significantly inhibited non-small cell lung cancer (NSCLC) cells which express wild type (wt) p53, in contract to its much weaker cytotoxicity on cells with mutant p53. RYL-687 upregulated p53 in cells with wt but not mutant p53, and ectopic expression of wt p53 significantly enhanced the anti-NSCLC activity of this compound. RYL-687 induced production of reactive oxygen species (ROS) and upregulation of Nrf2, leading to an elevation of the NAD(P)H:quinoneoxidoreductase-1 (NQO1) that can protect p53 by inhibiting its degradation by 20S proteasome. RYL-687 bound NQO1, facilitating the physical interaction between NQO1 and p53. NQO1 was required for RYL-687-induced p53 accumulation, because silencing of NQO1 by specific siRNA or an NQO1 inhibitor uridine, drastically suppressed RYL-687-induced p53 upregulation. Moreover, a RYL-687-related prodrug significantly inhibited tumor growth in NOD-SCID mice inoculated with NSCLC cells and in a wt p53-NSCLC patient-derived xenograft mouse model. These data indicate that targeting NQO1 is a rational strategy to reactivate p53, and RYL-687 as a p53 stabilizer bears therapeutic potentials in NSCLCs with wt p53.


Assuntos
Carcinoma Pulmonar de Células não Pequenas/patologia , Neoplasias Pulmonares/patologia , NAD(P)H Desidrogenase (Quinona)/efeitos dos fármacos , Quinolonas/farmacologia , Proteína Supressora de Tumor p53/efeitos dos fármacos , Apoptose/efeitos dos fármacos , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Humanos , Fator 2 Relacionado a NF-E2/efeitos dos fármacos , RNA Interferente Pequeno/farmacologia , Espécies Reativas de Oxigênio/metabolismo , Proteína Supressora de Tumor p53/genética , Regulação para Cima
11.
Front Genet ; 12: 733715, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34630524

RESUMO

Background: 5-Methylcytidine (m5C) is the most common RNA modification and plays an important role in multiple tumors including cervical cancer (CC). We aimed to develop a novel gene signature by identifying m5C modification subtypes of CC to better predict the prognosis of patients. Methods: We obtained the expression of 13 m5C regulatory factors from The Cancer Genome Atlas (TCGA all set, 257 patients) to determine m5C modification subtypes by the "nonnegative matrix factorization" (NMF). Then the "limma" package was used to identify differentially expressed genes (DEGs) between different subtypes. According to these DEGs, we performed Cox regression and Kaplan-Meier (KM) survival analysis to establish a novel gene signature in TCGA training set (128 patients). We also verified the risk prediction effect of gene signature in TCGA test set (129 patients), TCGA all set (257 patients) and GSE44001 (300 patients). Furthermore, a nomogram including this gene signature and clinicopathological parameters was established to predict the individual survival rate. Finally, the expression and function of these signature genes were explored by qRT-PCR, immunohistochemistry (IHC) and proliferation, colony formation, migration and invasion assays. Results: Based on consistent clustering of 13 m5C-modified genes, CC was divided into two subtypes (C1 and C2) and the C1 subtype had a worse prognosis. The 4-gene signature comprising FNDC3A, VEGFA, OPN3 and CPE was constructed. In TCGA training set and three validation sets, we found the prognosis of patients in the low-risk group was much better than that in the high-risk group. A nomogram incorporating the gene signature and T stage was constructed, and the calibration plot suggested that it could accurately predict the survival rate. The expression levels of FNDC3A, VEGFA, OPN3 and CPE were all high in cervical cancer tissues. Downregulation of FNDC3A, VEGFA or CPE expression suppressed the proliferation, migration and invasion of SiHa cells. Conclusions: Two m5C modification subtypes of CC were identified and then a 4-gene signature was established, which provide new feasible methods for clinical risk assessment and targeted therapies for CC.

12.
Cell Death Discov ; 7(1): 260, 2021 Sep 22.
Artigo em Inglês | MEDLINE | ID: mdl-34552057

RESUMO

To maintain genomic stability, the mammalian cells has evolved a coordinated response to DNA damage, including activation of DNA repair and cell cycle checkpoint processes. Exonuclease 1 (EXO1)-dependent excision of DNA ends is important for the initiation of homologous recombination (HR) repair of DNA breaks, which is thought to play a key role in activating the ATR-CHK1 pathway to induce G2/M cell cycle arrest. But the mechanism is still not fully understood. Here, we report that ZGRF1 forms complexes with EXO1 as well as other repair proteins and promotes DNA repair through HR. ZGRF1 is recruited to DNA damage sites in a MDC1-RNF8-BRCA1 dependent manner. Furthermore, ZGRF1 is important for the recruitment of RPA2 to DNA damage sites and the following ATR-CHK1 mediated G2/M checkpoint in response to irradiation. ZGRF1 null cells show increased sensitivity to many DNA-damaging agents, especially PARPi and irradiation. Collectively,our findings identify ZGRF1 as a novel regulator of DNA end resection and G2/M checkpoint. ZGRF1 is a potential target of radiation and PARPi cancer therapy.

13.
Chemistry ; 27(38): 9839-9849, 2021 Jul 07.
Artigo em Inglês | MEDLINE | ID: mdl-33878230

RESUMO

Targeting the low-oxygen (hypoxic) environments found in many tumours by using redox-active metal complexes is a strategy that can enhance efficacy and reduce the side effects of chemotherapies. We have developed a series of CuII complexes with tridentate pyridine aminophenolate-based ligands for preferential activation in the reduction window provided by hypoxic tissues. Furthermore, ligand functionalization with a pendant CF3 group provides a 19 F spectroscopic handle for magnetic-resonance studies of redox processes at the metal centre and behaviour in cellular environments. The phenol group in the ligand backbone was substituted at the para position with H, Cl, and NO2 to modulate the reduction potential of the CuII centre, giving a range of values below the window expected for hypoxic tissues. The NO2 -substituted complex, which has the highest reduction potential, showed enhanced cytotoxic selectivity towards HeLa cells grown under hypoxic conditions. Cell death occurs by apoptosis, as determined by analysis of the cell morphology. A combination of 19 F NMR and ICP-OES indicates localization of the NO2 complex in HeLa cell nuclei and increased cellular accumulation under hypoxia. This correlates with DNA nuclease activity being the likely origin of cytotoxic activity, as demonstrated by cleavage of DNA plasmids in the presence of the CuII nitro complex and a reducing agent. Selective detection of the paramagnetic CuII complexes and their diamagnetic ligands by 19 F MRI suggests hypoxia-targeting theranostic applications by redox activation.


Assuntos
Cobre , Compostos Organometálicos , Núcleo Celular , Citotoxinas , Células HeLa , Humanos , Hipóxia , Ligantes , Espectroscopia de Ressonância Magnética , Compostos Organometálicos/farmacologia
14.
Angew Chem Int Ed Engl ; 59(50): 22523-22530, 2020 12 07.
Artigo em Inglês | MEDLINE | ID: mdl-32790890

RESUMO

19 F magnetic resonance (MR) based detection coupled with well-designed inorganic systems shows promise in biological investigations. Two proof-of-concept inorganic probes that exploit a novel mechanism for 19 F MR sensing based on converting from low-spin (S=0) to high-spin (S=1) Ni2+ are reported. Activation of diamagnetic NiL1 and NiL2 by light or ß-galactosidase, respectively, converts them into paramagnetic NiL0 , which displays a single 19 F NMR peak shifted by >35 ppm with accelerated relaxation rates. This spin-state switch is effective for sensing light or enzyme expression in live cells using 19 F MR spectroscopy and imaging that differentiate signals based on chemical shift and relaxation times. This general inorganic scaffold has potential for developing agents that can sense analytes ranging from ions to enzymes, opening up diverse possibilities for 19 F MR based biosensing.

15.
Sci Adv ; 6(28): eaba7822, 2020 07.
Artigo em Inglês | MEDLINE | ID: mdl-32832608

RESUMO

Nonhomologous end joining (NHEJ) and homologous recombination (HR) are major repair pathways of DNA double-strand breaks (DSBs). The pathway choice of HR and NHEJ is tightly regulated in cellular response to DNA damage. Here, we demonstrate that the interaction of TIP60 with DNA-PKcs is attenuated specifically in S phase, which facilitates HR pathway activation. SUMO2 modification of TIP60 K430 mediated by PISA4 E3 ligase blocks its interaction with DNA-PKcs, whereas TIP60 K430R mutation recovers its interaction with DNA-PKcs, which results in abnormally increased phosphorylation of DNA-PKcs S2056 in S phase and marked inhibition of HR efficiency, but barely affects NHEJ activity. TIP60 K430R mutant cancer cells are more sensitive to radiation and PARP inhibitors in cancer cell killing and tumor growth inhibition. Collectively, coordinated regulation of TIP60 and DNA-PKcs facilitates HR pathway choice in S-phase cells. TIP60 K430R mutant is a potential target of radiation and PARPi cancer therapy.


Assuntos
Reparo do DNA , Neoplasias , DNA/genética , Quebras de DNA de Cadeia Dupla , Reparo do DNA por Junção de Extremidades , Recombinação Homóloga , Neoplasias/tratamento farmacológico , Neoplasias/genética , Sumoilação
16.
Mol Cell Probes ; 53: 101608, 2020 10.
Artigo em Inglês | MEDLINE | ID: mdl-32585183

RESUMO

Procollagen-lysine, 2-oxoglutarate 5-dioxygenase (PLOD3), also known as lysyl hydroxylase 3 (LH3) has been demonstrated to be overexpressed in several kinds of cancers and facilitate cell migration. Currently, we aimed to reveal the role of PLOD3 in renal cell carcinoma (RCC) progression, and explore whether TWIST1 (Twist family bHLH transcription factor 1) is involved in this process. Fifty-eight paired RCC tissues and normal tissues were collected and subjected to qPCR and immunohistochemistry (IHC) technology to detect the expression levels of PLOD3. The clinical value of PLOD3 in predicting RCC progression was then explored. Cell-Counting Kit-8 (CCK-8), wound healing, transwell chambers and tumor-bearing experiments were applied to monitor cell proliferation, migration, invasion and tumorigenesis. Protein levels were determined by using western blotting technology to assess cell apoptosis and epithelial to mesenchymal transition (EMT). PLOD3 expression was enhanced in RCC tissues and cells, which predicted higher T (tumor), N (lymph node) and M (metastasis) stages, histological grade and TNM (tumor, lymph node, metastasis) stage. PLOD3 downregulation in RCC A498 cells obviously inhibited cell proliferation, migration, invasion, EMT and tumorigenesis and increased cell apoptosis. PLOD3 overexpression led to opposite results in RCC A704 cells. PLOD3 downregulation reduced the expression levels of TWIST1, ß-catenin and p-AKT. In addition, TWIST1 overexpression rescued the repressions of cell proliferation, migration, invasion, EMT and the activation of ß-catenin and AKT signaling in addition to apoptosis promotion induced by PLOD3 downregulation. Collectively, this study illustrated that PLOD3 knockdown suppressed RCC malignance via inhibiting TWIST1-mediated activation of ß-catenin and AKT signaling.


Assuntos
Carcinoma de Células Renais/patologia , Neoplasias Renais/patologia , Proteínas Nucleares/metabolismo , Pró-Colágeno-Lisina 2-Oxoglutarato 5-Dioxigenase/genética , Pró-Colágeno-Lisina 2-Oxoglutarato 5-Dioxigenase/metabolismo , Proteína 1 Relacionada a Twist/metabolismo , Animais , Carcinoma de Células Renais/genética , Carcinoma de Células Renais/metabolismo , Linhagem Celular Tumoral , Progressão da Doença , Feminino , Regulação Neoplásica da Expressão Gênica , Técnicas de Silenciamento de Genes , Células Endoteliais da Veia Umbilical Humana , Humanos , Neoplasias Renais/genética , Neoplasias Renais/metabolismo , Metástase Linfática , Masculino , Camundongos , Estadiamento de Neoplasias , Transplante de Neoplasias , Transdução de Sinais , Regulação para Cima
17.
J Neurol Surg A Cent Eur Neurosurg ; 81(2): 155-162, 2020 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-31550736

RESUMO

BACKGROUND: Basal ganglia hemorrhage (BGH) is a devastating neurologic disease with high morbidity and mortality, and its management is still controversial. We evaluated the effectiveness of surgical treatments for BGH and investigated computed tomography (CT) imaging features affecting the hematoma evacuation rate (ER) in patients treated with neuroendoscopic surgery. MATERIALS AND METHODS: A total of 104 BGH patients who underwent craniotomy, burr-hole drainage, or neuroendoscopic surgery were analyzed retrospectively. Clinical characteristics, imaging features, and postoperative complications were compared. Univariate and multivariate regression analyses were applied to identify imaging factors associated with ER. RESULTS: A significant difference in ER was observed: 78.4% in patients treated with neuroendoscopic surgery, 33.6% in patients treated with burr-hole drainage, and 82.5% in patients treated with craniotomy (p < 0.001). Similar results were observed for operative time (p < 0.001). Five cases (12.5%) of rebleeding were found in patients treated with burr-hole drainage (p = 0.020). No significant difference was found for pneumonia, intracranial infection, gastrointestinal bleeding, hospital mortality, hospital stay, expenses, 3-day Glasgow Coma Scale (GCS) scores after surgery, or GCS at discharge. The CT imaging feature, the island sign (p = 0.004), was observed as an independent factor correlated with lower ER for neuroendoscopic surgery. CONCLUSIONS: The benefits and drawbacks of surgical treatments confirmed they have their own indications, and neuroendoscopic surgery may be relatively beneficial for BGH treatment. The island sign was an independent factor affecting ER for neuroendoscopic surgery. Therefore, comprehensive assessment of clinical data, especially the island sign, should be performed preoperatively in BGH patients.


Assuntos
Hemorragia dos Gânglios da Base/diagnóstico por imagem , Hemorragia dos Gânglios da Base/cirurgia , Drenagem/métodos , Endoscopia/métodos , Procedimentos Neurocirúrgicos/métodos , Idoso , Craniotomia , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Neuroendoscopia , Cuidados Pós-Operatórios , Complicações Pós-Operatórias/epidemiologia , Estudos Retrospectivos , Tomografia Computadorizada por Raios X , Resultado do Tratamento
18.
Cell Death Differ ; 27(4): 1383-1397, 2020 04.
Artigo em Inglês | MEDLINE | ID: mdl-31562368

RESUMO

End resection of DNA double-strand breaks (DSBs) to form 3' single-strand DNA (ssDNA) is critical to initiate the homologous recombination (HR) pathway of DSB repair. HR pathway is strictly limited in the G1-phase cells because of lack of homologous DNA as the templates. Exonuclease 1 (EXO1) is the key molecule responsible for 3' ssDNA formation of DSB end resection. We revealed that EXO1 is inactivated in G1-phase cells via ubiquitination-mediated degradation, resulting from an elevated expression level of RING-box protein 1 (RBX1) in G1 phase. The increased RBX1 significantly prompted the neddylation of Cullin1 and contributed to the G1 phase-specific degradation of EXO1. Knockdown of RBX1 remarkedly attenuated the degradation of EXO1 and increased the end resection and HR activity in γ-irradiated G1-phase cells, as demonstrated by the increased formation of RPA32, BrdU, and RAD51 foci. And EXO1 depletion mitigated DNA repair defects due to RBX1 reduction. Moreover, increased autophosphorylation of DNA-PKcs at S2056 was found to be responsible for the higher expression level of the RBX1 in the G1 phase. Inactivation of DNA-PKcs decreased RBX1 expression, and simultaneously increased EXO1 expression and DSB end resection in G1-phase cells. This study demonstrates a new mechanism for restraining the HR pathway of DNA DSB repair in G1 phase via RBX1-prompted inactivation of EXO1.


Assuntos
Proteínas de Transporte/metabolismo , Quebras de DNA de Cadeia Dupla , Enzimas Reparadoras do DNA/metabolismo , Reparo do DNA , Exodesoxirribonucleases/metabolismo , Fase G1 , Recombinação Homóloga , Proteólise , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos da radiação , Proteínas Culina/metabolismo , Proteína Quinase Ativada por DNA/metabolismo , Raios gama , Humanos , Modelos Biológicos , Rad51 Recombinase/metabolismo , Ubiquitinação
19.
Integr Cancer Ther ; 18: 1534735419890917, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31855073

RESUMO

Background: Current treatment of osteosarcoma is limited in part by side effects and low tolerability, problems generally avoided with traditional Chinese medicine. Ganoderma lucidum, a traditional Chinese medicine with antitumor effects, offers a potential alternative, but little is known about its molecular mechanisms in osteosarcoma cells. Objective: To investigate the effect of G lucidum on osteosarcoma cells and its mechanism. Methods: Osteosarcoma MG63 and U2-OS cells were treated with G lucidum, followed by assays for cell proliferation (Cell Counting Kit-8), colony formation, and apoptosis (Alexa Fluor 647-Annexin V/propidium iodide, flow cytometry). Migration and invasion of cells were assessed by wound healing and Transwell invasion assays, and the effect of G lucidum on Wnt/ß-catenin signal transduction was studied by real-time quantitative polymerase chain reaction, western blot, and dual-luciferase assay. Results:G lucidum inhibited the proliferation, migration, and invasion, and induced apoptosis of human osteosarcoma MG63 and U2-OS cells. Dual-luciferase assay showed that G lucidum suppressed the transcriptional activity of T-cell factor/lymphocyte enhancer factor in the Wnt/ß-catenin signaling pathway. Moreover, G lucidum blocked Wnt/ß-catenin signaling by inhibiting the Wnt co-receptor LRP5 and Wnt-related target genes, such as ß-catenin, cyclin D1, C-Myc, MMP-2, and MMP-9. At the same time, when Wnt/ß-catenin was inhibited, the expression of E-cadherin was upregulated. Conclusions: Our results suggest that G lucidum broadly suppresses osteosarcoma cell growth by inhibiting Wnt/ß-catenin signaling.


Assuntos
Produtos Biológicos/farmacologia , Osteossarcoma/tratamento farmacológico , Osteossarcoma/metabolismo , Reishi/química , Via de Sinalização Wnt/efeitos dos fármacos , beta Catenina/metabolismo , Apoptose/efeitos dos fármacos , Ciclo Celular/efeitos dos fármacos , Morte Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Movimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Metaloproteinase 2 da Matriz/metabolismo , Metaloproteinase 9 da Matriz/metabolismo
20.
Dose Response ; 17(1): 1559325819833474, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30833875

RESUMO

BACKGROUND: Radiation therapy induces acute and chronic radiological toxicity, in particular hematological toxicity (HT). This study aimed to explore the mechanistic clue and potential predictors at the messenger RNA (mRNA) level. MATERIALS AND METHODS: Peripheral blood was collected from 3 patients with cervical cancer (CC), nasopharynx cancer (NC), and tongue cancer (TC) after the first 2 Gy fraction of radiotherapy (RT). High-throughput sequencing was used to assess mRNA profiles. RESULTS: Eleven genes, such as ALAS2(5-aminolevulinate synthase), SLC4A1(solute carrier family 4 member 1), HBG2(hemoglobin subunit gamma 2), TNFAIP3 (TNF α-induced protein 3), PER1 (period circadian clock 1), CCDC136 (coiled-coil domain containing 136), C9orf84 (chromosome 9 open reading frame 84), IL1B (interleukin 1ß), FOSB (FosB protooncogene), NR4A2 (nuclear receptor subfamily 4), PARP15 (polymerase family member 15), had overlapping expression changes in all 3 cancers of which 3 (ALAS2, FOSB, and HBG2) are suggested as potential predictors for the early diagnosis of HT after RT. CONCLUSIONS: ALAS2, FOSB, and HBG2 may be useful predictors of HT in patients after RT. Eleven overlapping expression mRNAs among 3 cancers might be potential predictors for early diagnosis of radiation toxicity in patients.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA