Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 37
Filter
1.
Int J Med Sci ; 19(1): 47-64, 2022.
Article in English | MEDLINE | ID: mdl-34975298

ABSTRACT

Background: Clear cell renal cell carcinoma (ccRCC) is a cell metabolic disease with high metastasis rate and poor prognosis. Our previous studies demonstrate that glucose-6-phosphate dehydrogenase (G6PD), the first and rate-limiting enzyme of the pentose phosphate pathway, is highly expressed in ccRCC and predicts poor outcomes of ccRCC patients. The aims of this study were to confirm the oncogenic role of G6PD in ccRCC and unravels novel mechanisms involving Cyclin E1 and MMP9 in G6PD-mediated ccRCC progression. Methods: Real-time RT-PCR, Western blot and immunohistochemistry were used to determine the expression patterns of G6PD, Cyclin E1 and MMP9 in ccRCC. TCGA dataset mining was used to identify Cyclin E1 and MMP9 correlations with G6PD expression, relationships between clinicopathological characteristics of ccRCC and the genes of interest, as well as the prognosis of ccRCC patients. The role of G6PD in ccRCC progression and the regulatory effect of G6PD on Cyclin E1 and MMP9 expression were investigated by using a series of cytological function assays in vitro. To verify this mechanism in vivo, xenografted mice models were established. Results: G6PD, Cyclin E1 and MMP9 were overexpressed and positively correlated in ccRCC, and they were associated with poor prognosis of ccRCC patients. Moreover, G6PD changed cell cycle dynamics, facilitated cells proliferation, promoted migration in vitro, and enhanced ccRCC development in vivo, more likely through enhancing Cyclin E1 and MMP9 expression. Conclusion: These findings present G6PD, Cyclin E1 and MMP9, which contribute to ccRCC progression, as novel biomarkers and potential therapeutic targets for ccRCC treatment.


Subject(s)
Carcinoma, Renal Cell/genetics , Cyclin E/genetics , Gene Expression Regulation, Neoplastic , Glucosephosphate Dehydrogenase/physiology , Kidney Neoplasms/genetics , Matrix Metalloproteinase 9/genetics , Oncogene Proteins/genetics , Up-Regulation , Animals , Carcinoma, Renal Cell/metabolism , Carcinoma, Renal Cell/pathology , Cell Line, Tumor , Cyclin E/metabolism , Disease Models, Animal , Disease Progression , Female , Humans , Kidney Neoplasms/metabolism , Kidney Neoplasms/pathology , Male , Matrix Metalloproteinase 9/metabolism , Mice, Inbred BALB C , Mice, Nude , Middle Aged , Neoplasm Staging , Oncogene Proteins/metabolism
2.
Acta Biochim Biophys Sin (Shanghai) ; 54(2): 220-231, 2022 01 25.
Article in English | MEDLINE | ID: mdl-35130611

ABSTRACT

Clear cell renal carcinoma (ccRCC) is histologically defined by its cytoplasmic lipid deposits. Lipid metabolism disorder largely increases the risk of ccRCC. In this study, we aimed to investigate the biological functions and molecular mechanisms of carnitine palmitoyl transferase 1A (CPT1A) in ccRCC. Our results showed that CPT1A is decreased in ccRCC clinical samples and cell lines compared with that in normal samples. Lentivirus overexpressing CPT1A was used to investigate the neoplastic phenotypes of ccRCC, and the results showed that lipid accumulation and tumor growth are attenuated both and . In addition, CPT1A prevents cholesterol uptake and lipid accumulation by increasing the peroxisome proliferator-activated receptor α (PPARα) level through regulation of Class B scavenger receptor type 1 (SRB1) and cluster of differentiation 36 (CD36). Furthermore, PI3K/Akt signaling pathway promotes tumor cell proliferation in ccRCC, which is related to the enhanced expression of CD36. Functionally, weakened CPT1A expression is critical for lipid accumulation to promote ccRCC development. Collectively, our research unveiled a novel function of CPT1A in lipid metabolism via PPARα/CD36 axis, which provides a new theoretical explanation for the pathogenesis of ccRCC. Targeting CPT1A may be a potential therapeutic strategy to treat ccRCC.


Subject(s)
Carcinoma, Renal Cell , Carnitine O-Palmitoyltransferase/metabolism , Kidney Neoplasms , Carcinoma, Renal Cell/metabolism , Cell Line, Tumor , Cell Proliferation/genetics , Humans , Kidney Neoplasms/metabolism , Lipid Metabolism/genetics , Lipids , PPAR alpha/genetics , PPAR alpha/metabolism , Phosphatidylinositol 3-Kinases/metabolism
3.
Cancer Sci ; 112(10): 4075-4086, 2021 Oct.
Article in English | MEDLINE | ID: mdl-34310804

ABSTRACT

The regulatory relationship between silent information regulator 2 (SIRT2) and glucose 6-phosphate dehydrogenase (G6PD) in clear cell renal cell carcinoma (ccRCC) is still unclear. The present study aimed to explore the function of SIRT2 and its regulatory effect on G6PD in ccRCC. The Cancer Genome Atlas data mining of SIRT2 was first analyzed. Quantitative real-time PCR and western blot analyses were used to assess the mRNA and protein expression levels, respectively. Cell viability, colony formation, cell cycle, cell apoptosis, and TUNEL assays and EdU staining were used to investigate the roles of SIRT2 in ccRCC proliferation and apoptosis. The coimmunoprecipitation (Co-IP) assay was used to analyze the association between SIRT2 and G6PD in ccRCC cells. Quantitative Co-IP assay was used to detect the levels of G6PD ubiquitination and small ubiquitin-related modifier 1 (SUMO1). An in vivo experiment was also carried out to confirm in vitro findings. The results indicated that SIRT2 promoted ccRCC proliferation and inhibited apoptosis by regulating cell cycle and apoptosis related proteins. Silent information regulator 2 interacted with G6PD, facilitated its activity through deacetylation, and increased its stability by reducing its ubiquitination and enhancing its SUMO1 modification. Silent information regulator 2 also promoted ccRCC tumor development in vivo. Taken together, the present study indicated that SIRT2 promoted ccRCC progression by increasing G6PD activity and stability, and it could be a potential new diagnostic and therapeutic target for ccRCC.


Subject(s)
Carcinoma, Renal Cell/metabolism , Cysteine Endopeptidases/metabolism , Glucosephosphate Dehydrogenase/metabolism , Kidney Neoplasms/metabolism , Sirtuin 2/physiology , Acetylation , Animals , Apoptosis , Blotting, Western , Carcinoma, Renal Cell/pathology , Cell Cycle , Cell Cycle Proteins/metabolism , Cell Survival , Databases, Genetic , Disease Progression , Female , Humans , Immunoprecipitation , Kidney Neoplasms/pathology , Mice , Mice, Inbred BALB C , Mice, Nude , Neoplasm Proteins/metabolism , Protein Modification, Translational , RNA, Messenger/metabolism , Real-Time Polymerase Chain Reaction , Tumor Cells, Cultured , Tumor Stem Cell Assay , Ubiquitination
4.
Eur J Clin Invest ; 50(10): e13332, 2020 Oct.
Article in English | MEDLINE | ID: mdl-32589285

ABSTRACT

BACKGROUND: This study aimed to summarize the association between diabetes mellitus (DM) and the incidence of lung cancer using a meta-analysis of cohort studies. MATERIALS AND METHODS: We systematically searched PubMed, Embase and the Cochrane Library to identify potential cohort studies. Relative risk (RR) was used to calculate the association between DM and the risk of lung cancer. Subgroup analysis, sensitivity analysis and test for publication bias were performed. Twenty cohort studies were selected. RESULTS: The participants with DM showed little or no significant effect on the risk of lung cancer (RR: 1.10; 95% CI: 0.99-1.23; P = .087). DM was not associated with the risk of lung cancer in men (RR: 1.11; 95%CI: 0.92-1.35; P = .270), but a significant association was observed in women (RR: 1.18; 95%CI: 1.10-1.28; P < .001). Subgroup analysis suggested that smoker status was confounding variables that could bias the relationship between DM and the incidence of lung cancer. CONCLUSIONS: This meta-analysis suggests that DM has no significant impact on the incidence of lung cancer in men but has a harmful effect on women.


Subject(s)
Diabetes Mellitus/epidemiology , Lung Neoplasms/epidemiology , Humans
5.
Cancer Cell Int ; 20(1): 565, 2020 Nov 26.
Article in English | MEDLINE | ID: mdl-33292264

ABSTRACT

An amendment to this paper has been published and can be accessed via the original article.

6.
Cancer Cell Int ; 20: 483, 2020.
Article in English | MEDLINE | ID: mdl-33041664

ABSTRACT

BACKGROUND: Glucose 6-phosphate dehydrogenase (G6PD) serves key roles in cancer cell metabolic reprogramming, and has been reported to be involved in certain carcinogenesis. Previous results from our laboratory demonstrated that overexpressed G6PD was a potential prognostic biomarker in clear cell renal cell carcinoma (ccRCC), the most common subtype of kidney cancer. G6PD could stimulate ccRCC growth and invasion through facilitating reactive oxygen species (ROS)-phosphorylated signal transducer and activator of transcription 3 (pSTAT3) activation and ROS-MAPK-MMP2 axis pathway, respectively. However, the reasons for ectopic G6PD overexpression and the proliferation repressive effect of G6PD inhibition in ccRCC are still unclear. METHODS: The impact of ROS accumulation on NF-κB signaling pathway and G6PD expression was determined by real-time RT-PCR and Western blot in ccRCC cells following treatment with ROS stimulator or scavenger. The regulatory function of NF-κB signaling pathway in G6PD transcription was analyzed by real-time RT-PCR, Western blot, luciferase and ChIP assay in ccRCC cells following treatment with NF-κB signaling activator/inhibitor or lentivirus infection. ChIP and Co-IP assay was performed to demonstrate protein-DNA and protein-protein interaction of NF-κB and pSTAT3, respectively. MTS assay, human tissue detection and xenograft model were conducted to characterize the association between NF-κB, pSTAT3, G6PD expression level and proliferation functions. RESULTS: ROS-stimulated NF-κB and pSTAT3 signaling over-activation could activate each other, and exhibit cross-talks in G6PD aberrant transcriptional regulation. The underlying mechanism was that NF-κB signaling pathway facilitated G6PD transcription via direct DNA-protein interaction with p65 instead of p50. p65 and pSTAT3 formed a p65/pSTAT3 complex, occupied the pSTAT3-binding site on G6PD promoter, and contributed to ccRCC proliferation following facilitated G6PD overexpression. G6PD, pSTAT3, and p65 were highly expressed and positively correlated with each other in ccRCC tissues, confirming that NF-κB and pSTAT3 synergistically promote G6PD overexpression. Moreover, G6PD inhibitor exhibited tumor-suppressor activities in ccRCC and attenuated the growth of ccRCC cells both in vitro and in vivo. CONCLUSION: ROS-stimulated aberrations of NF-κB and pSTAT3 signaling pathway synergistically drive G6PD transcription through forming a p65/pSTAT3 complex. Moreover, G6PD activity inhibition may be a promising therapeutic strategy for ccRCC treatment.

7.
BMC Cancer ; 20(1): 682, 2020 Jul 22.
Article in English | MEDLINE | ID: mdl-32698787

ABSTRACT

BACKGROUND: Long intergenic non-coding RNA 00511 (LINC00511) is highly expressed in diverse cancers and has a correlation with poor clinical outcomes for cancer patients. In view of contradictory data among published data, we aim to evaluate the prognostic role of LINC00511 for cancer patients. METHODS: In the present study, a meta-analysis of related studies has been performed to investigate the prognostic significance of LINC00511 in cancer patients. Relevant studies published before December 22, 2019 were systematically searched online in PubMed, EMBASE, Web of Science, and the Cochrane Library databases. The relationship between LINC00511 expression and cancer patients' survival, including overall survival (OS), disease-free survival (DFS)/relapse-free survival (RFS) and progression-free survival (PFS), was evaluated using pooled hazard ratios (HRs) with their corresponding 95% confidence intervals (CIs). The association between LINC00511 expression and clinicopathological features was assessed using odd ratios (ORs) and their corresponding 95% CIs. RESULTS: A total of 14 eligible studies with 1883 patients were enrolled in the present meta-analysis. The results demonstrated that elevated expression of LINC00511 was significantly associated with poor OS (HR = 2.62; 95% CI: 2.00-3.45; p <  0.001), PFS (HR = 1.80; 95% CI: 1.29-2.51; p = 0.001) and DFS/RFS (HR = 2.90; 95% CI: 1.04-8.12; p = 0.04). Additionally, High LINC00511 expression was associated with large tumor size (OR = 3.10; 95% CI: 1.97-4.86; p <  0.00001), lymph node metastasis (OR = 3.11; 95% CI: 2.30-4.21; p <  0.00001), advanced clinical stage (OR = 3.95; 95% CI: 2.68-5.81; p <  0.00001), distant metastasis (OR = 2.39; 95% CI: 1.16-4.93; p = 0.02), and disease recurrence (OR = 4.62; 95% CI: 2.47-8.65; p <  0.00001). Meanwhile, no correlation was found between LINC00511 expression and age, gender, and histological grade. These findings were consolidated by the results of bioinformatics analysis. CONCLUSIONS: Based on our findings, LINC00511 may serve as a novel prognostic biomarker for cancer patients.


Subject(s)
Biomarkers, Tumor/metabolism , Neoplasms/metabolism , Neoplasms/mortality , RNA, Long Noncoding/metabolism , RNA, Neoplasm/metabolism , Humans , Prognosis , Publication Bias
8.
J Cell Biochem ; 120(11): 19124-19136, 2019 11.
Article in English | MEDLINE | ID: mdl-31310363

ABSTRACT

In this study, we aimed to explore the mechanism of glutathione peroxidase 3 (GPX3) in the growth of malignant melanoma (MM) cells by hypoxia-inducible factor-1α (HIF1-α) and HIF2-α regulating the metabolism through reactive oxygen species (ROS). The messenger RNA and protein expression of GPX3, HIF1-α, HIF2-α in tissues, and cell lines were measured by reverse transcription-quantitative PCR and Western blot analysis. A375 cells were transfected with GPX3 overexpression plasmid, small interfering RNA (siRNA) targeting GPX3, or siRNA targeting HIF1-α/HIF2-α to upregulate or downregulate the expression of GPX3 or HIF1-α/HIF2-α. The effects of H2 O2 and N-acetylcysteine (NAC) on the levels of HIF1-α and HIF2-α after overexpression of GPX3 were studied. The cell viability was detected by Cell Counting Kit-8. The levels of ROS, glucose uptake and lactic acid production, oxidative phosphorylation, and glycolysis of cells were measured for assessment of cellular metabolism. The expression of GPX3 decreased, while ROS, HIF1-α, and HIF2-α increased in MM tissues and cells. Overexpression of GPX3 inhibited the viability of MM cells and the growth of melanoma xenografts. The overexpression of GPX3 reduced the glucose uptake, extracellular lactic acid content, and extracellular acidification rate and increased the oxygen consumption rate level. Overexpression of GPX3 could reduce the levels of HIF1-α and HIF2-α, which could regulate metabolic levels. GPX3 reduced ROS level in MM to inhibit HIF1-α and HIF2-α. The addition of H2 O2 increased while NAC reduced the protein levels of HIF1-α and HIF2-α in the cells overexpressing GPX3. Our study demonstrates that GPX3 inhibits the growth of MM cells through its inhibitory effect on cell metabolic disorder by inhibiting HIF1-α via regulating ROS.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , Glutathione Peroxidase/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Melanoma/metabolism , Neoplasm Proteins/metabolism , Reactive Oxygen Species/metabolism , Aged , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Cell Line, Tumor , Female , Glutathione Peroxidase/genetics , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Male , Melanoma/genetics , Melanoma/pathology , Mice , Mice, Nude , Middle Aged , Neoplasm Proteins/genetics , Protein Stability
9.
J Hum Genet ; 64(7): 681-687, 2019 Jul.
Article in English | MEDLINE | ID: mdl-31019231

ABSTRACT

Weill-Marchesani syndrome (WMS) is a rare connective tissue disorder characterized by short stature, brachydactyly, joint stiffness, eye anomalies, including microspherophakia, ectopia of the lenses, severe myopia, glaucoma and occasionally heart defects. Given these complex clinical manifestations and genetic heterogeneity, WMS patients presented misdiagnosed as high myopia or angle closure glaucoma. Here, we report ADAMTS17 mutations, a member of the extracellular matrix protease family, from a Chinese family. Patients have features that fall within the WMS spectrum. The exome (protein-coding regions of the genome) makes up ~1 % of the genome, it contains about 85% of known disease-related variants. Whole exome sequencing (WES) has been performed to identify the disease-associated genes, including one patient, his healthy sister, and his asymptomatic wife. Genome-wide homozygosity map was used to identify the disease caused locus. SNVs and INDELs were further predicted with MutationTaster, LRT, SIFT and SiPhy and compared to dbSNP150 and 1000 Genomes project. Filtered mutation was confirmed with Sanger sequencing in whole family members. The Genome-wide homozygosity map based on WES identified a total of 20 locus which were possible pathogenic. Further, a novel nonsense mutation c.1051A >T result in p.(lys351Ter) in ADAMTS17 had been identified in a candidate loci. The Sanger sequencing data has verified two consanguineous WMS patients in the family pedigree and revealed autosomal recessive (AR) inheritance pattern. The nonsense mutation in ADAMTS17 was analyzed in silico to explore its effects on protein function. We predicted the mutation produced non-function protein sequence. A novel nonsense mutation c.1051 A > T in ADAMTS17 had been identified caused autosomal recessive WMS in the Chinese family.


Subject(s)
ADAMTS Proteins/genetics , Codon, Nonsense , Weill-Marchesani Syndrome/genetics , Adult , Child , China , Chromosome Mapping , Dwarfism/genetics , Eye Abnormalities/genetics , Female , Homozygote , Humans , Male , Pedigree , Weill-Marchesani Syndrome/diagnosis , Exome Sequencing , Young Adult
10.
Int J Biol Macromol ; 270(Pt 1): 131889, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38782624

ABSTRACT

This work aimed at building functional emulsions based on the linear dextrins (LDs) emulsion system. The gradient polyethylene glycol (PEG) precipitaion method was used to fractionate LDs into fractions with different degrees of polymerization (DP). A package, and co-precipitation procedure of LDs, and eicosapentaenoic acid (EPA) was used to fabricate LDs-EPA composites. The gas chromatograph, Fourier transform infrared spectroscopy, X-ray diffraction and differential scanning calorimetry analyses affirmed the formation of the LDs-EPA composites. The sizes of these composites were 38.55 nm, 59.14 nm to 80.62 nm, respectively, and they had good amphiphilicity. Compared with LDs, these LDs-EPA composites stabilized Pickering emulsion had higher stability and antioxidant capacity. Their emulsifying ability was positively correlated with the DP values of LDs. Furthermore, the oxidation stability results showed that LDsF10-EPA emulsion had the lowest lipid hydroperoxide (LHs) content, malondioxide (MDA) content and hexal concentration, which were 138.75 mmol kg-1 oil, 15.50 mmol kg-1 oil and 3.83 µmol kg-1 oil, respectively. The study provided a new idea and application values for the application of LDs in emulsion.


Subject(s)
Dextrins , Eicosapentaenoic Acid , Emulsions , Polymerization , Emulsions/chemistry , Eicosapentaenoic Acid/chemistry , Eicosapentaenoic Acid/analogs & derivatives , Dextrins/chemistry , Antioxidants/chemistry , Emulsifying Agents/chemistry , Polyethylene Glycols/chemistry , X-Ray Diffraction
11.
BMC Cancer ; 13: 251, 2013 May 22.
Article in English | MEDLINE | ID: mdl-23693134

ABSTRACT

BACKGROUND: Glucose-6-phosphate dehydrogenase (G6PD), elevated in tumor cells, catalyzes the first reaction in the pentose-phosphate pathway. The regulation mechanism of G6PD and pathological change in human melanoma growth remains unknown. METHODS: HEM (human epidermal melanocyte) cells and human melanoma cells with the wild-type G6PD gene (A375-WT), G6PD deficiency (A375-G6PD∆), G6PD cDNA overexpression (A375-G6PD∆-G6PD-WT), and mutant G6PD cDNA (A375-G6PD∆-G6PD-G487A) were subcutaneously injected into 5 groups of nude mice. Expressions of G6PD, STAT3, STAT5, cell cycle-related proteins, and apoptotic proteins as well as mechanistic exploration of STAT3/STAT5 were determined by quantitative real-time PCR (qRT-PCR), immunohistochemistry and western blot. RESULTS: Delayed formation and slowed growth were apparent in A375-G6PD∆ cells, compared to A375-WT cells. Significantly decreased G6PD expression and activity were observed in tumor tissues induced by A375-G6PD∆, along with down-regulated cell cycle proteins cyclin D1, cyclin E, p53, and S100A4. Apoptosis-inhibited factors Bcl-2 and Bcl-xl were up-regulated; however, apoptosis factor Fas was down-regulated, compared to A375-WT cells. Moderate protein expressions were observed in A375-G6PD∆-G6PD-WT and A375-G6PD∆-G6PD-G487A cells. CONCLUSIONS: G6PD may regulate apoptosis and expression of cell cycle-related proteins through phosphorylation of transcription factors STAT3 and STAT5, thus mediating formation and growth of human melanoma cells. Further study will, however, be required to determine potential clinical applications.


Subject(s)
Apoptosis , Glucosephosphate Dehydrogenase/genetics , Melanoma/genetics , Melanoma/metabolism , STAT3 Transcription Factor/metabolism , STAT5 Transcription Factor/metabolism , Signal Transduction , Animals , Apoptosis/genetics , Apoptosis Regulatory Proteins/genetics , Apoptosis Regulatory Proteins/metabolism , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Cell Line, Tumor , Cell Proliferation , Disease Models, Animal , Enzyme Activation , Gene Expression , Gene Knockdown Techniques , Genetic Variation , Glucosephosphate Dehydrogenase/metabolism , Heterografts , Humans , Melanoma/pathology , Mice , Mice, Nude
12.
Stem Cells Int ; 2023: 8934210, 2023.
Article in English | MEDLINE | ID: mdl-36798674

ABSTRACT

Long noncoding RNAs (lncRNAs) play crucial roles in melanoma initiation and development, serving as potential therapeutic targets and prognostic markers for melanoma. lncRNA survival-associated mitochondrial melanoma-specific oncogenic noncoding RNA (SAMMSON) is upregulated in many types of human cancers. However, the functions of SAMMSON in melanoma have not been fully elucidated. This study is aimed at investigating the expression and functions of SAMMSON in melanoma development. Bioinformatics analysis was performed to determine the expression of SAMMSON and its correlation with the 10-year overall survival (OS) in melanoma patients. Cell proliferation, migration, invasion, and tumorigenesis were detected by MTT, colony formation, Transwell assays, and mouse xenograft model. The expression of cell cycle-related factors, epithelial-to-mesenchymal transition (EMT) makers, and matrix metalloproteinases (MMPs) was assessed by RT-qPCR and western blotting analysis. The results demonstrated that SAMMSON expression was upregulated in melanoma tissues and cells, and lower SAMMSON expression was correlated with longer 10-year OS. SAMMSON knockdown decreased the proliferation, migration, and invasion of melanoma cells by regulating the expression of proliferation-related genes, EMT factors, and MMPs, respectively. Additionally, Forkhead box protein A2 (FOXA2) was confirmed to be a target of SAMMSON, and the biological effects induced by FOXA2 overexpression were similar to those induced by SAMMSON silencing in melanoma cells. Further studies showed that SAMMSON downregulated FOXA2 expression in melanoma cells by modulating the EZH2/H3K27me3 axis. Taken together, our data indicate that SAMMSON plays an important role in melanoma progression and can be a valuable biomarker and therapeutic target in melanoma.

13.
Front Immunol ; 13: 989263, 2022.
Article in English | MEDLINE | ID: mdl-36203561

ABSTRACT

Phosphoribosyl pyrophosphate synthetase 1 (PRPS1) is the first enzyme in the de novo purine nucleotide synthesis pathway and is essential for cell development. However, the effect of PRPS1 on melanoma proliferation and metastasis remains unclear. This study aimed to investigate the regulatory mechanism of PRPS1 in the malignant progression of melanoma. Here, we found PRPS1 was upregulated in melanoma and melanoma cells. In addition, our data indicated that PRPS1 could promote the proliferation and migration and invasion of melanoma both in vitro and in vivo. PRPS1 also could inhibit melanoma cell apoptosis. Furthermore, we found NRF2 is an upstream transcription factor of PRPS1 that drive malignant progression of melanoma.


Subject(s)
Melanoma , Ribose-Phosphate Pyrophosphokinase , Humans , NF-E2-Related Factor 2/genetics , NF-E2-Related Factor 2/metabolism , Purine Nucleotides , Ribose-Phosphate Pyrophosphokinase/genetics , Ribose-Phosphate Pyrophosphokinase/metabolism , Syndrome , Up-Regulation
14.
Metabolites ; 12(11)2022 Nov 17.
Article in English | MEDLINE | ID: mdl-36422271

ABSTRACT

A high-fat diet plays an important role in aggravating cancers. Palmitic acid (PA) is one of the components of saturated fatty acids; it has been reported to promote tumor proliferation in melanomas, but the signal transduction pathway mediated by palmitic acid remains unclear. This study showed that palmitic acid can promote the lung metastasis of melanomas. Moreover, the interaction between palmitic acid and toll-like receptor 4 (TLR4) was predicted by molecular docking. The experimental results proved that palmitic acid could promote the TLR4 and Toll/IL-1 receptor domain-containing adaptor-inducing IFN-ß (TRIF) expression. The expression of Pellino1 (Peli1) and the phosphorylation of NF-kappa B (pNF-κB) were downregulated after the suppression of TLR4 and the silencing of Peli1 also inhibited the phosphorylation of NF-κB. Therefore, we concluded that palmitic acid promoted the lung metastasis of melanomas through the TLR4/TRIF-Peli1-pNF-κB pathway.

15.
Indian J Biochem Biophys ; 48(5): 316-24, 2011 Oct.
Article in English | MEDLINE | ID: mdl-22165289

ABSTRACT

G6PD(Mahidol) enzyme is the most common variant in the Achang Chinese ethnic group and clinically manifests as class II. In this study, G6PD(Mahidol) enzyme was characterized by molecular modeling to understand its kinetics. G6PD(Mahidol), G6PD(G487A) and G6PD(WT) proteins were heterologously expressed in the G6PD-deficient DF213 E. coli strain, purified and their steady-state kinetic parameters were determined. Compared with G6PD(WT), the Km, and Vmax of NADP+ with G6PD(G487A) were about 28-fold and 12-fold lower, respectively. The Ki values of dehydroepiandrosterone (DHEA), NADPH and ATP with G6PD(G487A) showed 29.5-fold, 2.36-fold reduction and 1.83-fold increase, respectively. A molecular modeling of G6PD(G487A) was performed based on the X-ray structure of human G6PD (PDB: 2BH9). It is suggested that Ser-163 might affect the stability of G6PD(G487A) alpha-helix d and beta-strand E, besides the conformation of beta-strand D. In conclusion, the biochemical and structural properties of G6PD(G487A) and G6PD(WT) enzymes are significantly different, which may be responsible for clinical diversity of G6PD deficiencies.


Subject(s)
Anemia, Hemolytic/enzymology , Glucosephosphate Dehydrogenase Deficiency/enzymology , Glucosephosphate Dehydrogenase/chemistry , Glucosephosphate Dehydrogenase/pharmacokinetics , Molecular Dynamics Simulation , Acute Disease , Adolescent , Anemia, Hemolytic/etiology , Asian People , Computer Simulation , Female , Glucosephosphate Dehydrogenase/antagonists & inhibitors , Glucosephosphate Dehydrogenase/metabolism , Glucosephosphate Dehydrogenase Deficiency/complications , Humans , Kinetics , Mutation
16.
Indian J Pathol Microbiol ; 64(1): 161-164, 2021.
Article in English | MEDLINE | ID: mdl-33433431

ABSTRACT

A 60-year-old Chinese male with a hard mass, pressure pain, and ulcerous skin under his left axilla was first diagnosed with apocrine carcinoma, most likely metastasis from breast cancer. PET/CT scan detected multiple bone metastasis and enlarged lymph nodes at left axilla, mediastinal area 7, and left pulmonary hilus. Lumpectomy was performed to remove the mass followed by chemotherapy and radiotherapy against focal bone metastasis, left axillary lesion, and left subcutaneous chest wall. PET/CT examination showed progressive disease after the completion of the treatments. Two nontender hard nodules were noticed on the patient's left upper arm and multiple immobile nodules were palpated under his left axillary skin. Immunohistochemistry (HER2++, ER+, PR+, AR-) of the biopsy tissue combined with histopathology indicated invasive ductal carcinoma with neuroendocrine differentiation. Metastatic Luminal B subtype breast cancer was preferred. Anti-estrogen endocrine therapy was then performed and PET/CT scan showed partial remission after one month's fulvestrant administration. Two significant somatic mutations, AR R616H and GATA3 S408Afs*99, were detected in the biopsy tissue by next-generation sequencing. GATA3 is associated with estrogen receptor signaling and was identified as a driver gene of female breast cancer. However, the function of GATA3 in male breast cancer remains controversial. Report of this case hopefully will contribute to exploring the role of GATA3 mutation in molecular mechanisms and endocrine therapy of male breast cancer.


Subject(s)
Antineoplastic Agents, Hormonal/therapeutic use , Breast Neoplasms, Male/drug therapy , Breast Neoplasms, Male/genetics , GATA3 Transcription Factor/genetics , Mutation , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Biopsy , Breast/pathology , Breast Neoplasms, Male/diagnostic imaging , Breast Neoplasms, Male/secondary , Endocrine System/drug effects , Humans , Male , Middle Aged , Positron Emission Tomography Computed Tomography
17.
J Cancer ; 12(9): 2550-2559, 2021.
Article in English | MEDLINE | ID: mdl-33854616

ABSTRACT

Pancreatic cancer is associated with poor prognosis due to limited therapeutic options. Excision repair cross-complementing 3 (ERCC3) is an important member of nucleotide excision repair (NER) that is overexpressed in some cancers and may be regarded as a poor prognostic factor. Yet, its role in pancreatic cancer remains unclear. This study aimed to investigate the expression and functions of ERCC3 in pancreatic cancer patients and its relation with clinicopathological features. Our data suggested that the protein expression level of ERCC3 was higher in tumor tissues than in adjacent tissues. In addition, the expression of ERCC3 has shown to be associated with the tumor extent (p=0.035). Besides, analysis of the dataset in The Cancer Genome Atlas (TCGA) revealed that high expression of ERCC3 was associated with poor overall survival in pancreatic cancer patients (p=0.0136). In Cox regression analysis, ERCC3 was an independent prognostic factor for overall survival in pancreatic cancer (p<0.001). Furthermore, our in vitro data further suggested that the overexpression of ERCC3 significantly promoted pancreatic cancer (BxPC-3, CFPAC-1, and PANC-1 cells) proliferation, invasion, and migration. Taken together, this study suggested that high expression of ERCC3 might be a poor prognostic factor in human pancreatic cancer and might be used as a promising therapeutic target for pancreatic cancer treatment.

18.
J Exp Med ; 198(3): 369-77, 2003 Aug 04.
Article in English | MEDLINE | ID: mdl-12885868

ABSTRACT

The recruitment of activated T cell subsets to sites of effector immune responses is mediated by homing receptors induced upon activation in secondary lymphoid tissue. Using an adoptive transfer model, the intestinal recruitment of CD4+ T cells activated with intraperitoneal antigen in complete Freund's adjuvant was examined. The data demonstrate that activated CD4+ T cells recruited to intestinal Peyer's patches (PP) and lamina propria (LP) up-regulate functional P-selectin glycoprotein ligand 1 (PSGL-1). Blockade of IL-12 inhibited functional PSGL-1 expression and reduced PP and LP CD4+ T cell recruitment by >40%. P-selectin blockade reduced LP recruitment of activated cells by 56% without affecting PP recruitment. Studies of mice examined 3 d after adoptive transfer of differentiated T cell subsets revealed that Th1 but not Th2 cells were recruited to small intestine PP and LP. Mucosal addressin cell adhesion molecule blockade reduced Th1 recruitment to PP by 90% and to LP by >72%, whereas P-selectin blockade reduced Th1 recruitment to PP by 18% and Th1 recruitment to LP by 84%. These data suggest that IL-12-induced functional PSGL-1 expression is a major determinant for the recruitment of Th1 effector cells to noninflamed as well as inflamed intestine.


Subject(s)
Cell Movement/physiology , Intestine, Small/metabolism , Membrane Glycoproteins/metabolism , P-Selectin/metabolism , Th1 Cells/metabolism , Adoptive Transfer , Animals , Cells, Cultured , Interleukin-12/metabolism , Intestine, Small/anatomy & histology , Intestine, Small/immunology , Ligands , Lymphocyte Activation , Membrane Glycoproteins/immunology , Mice , Mice, Inbred BALB C , P-Selectin/immunology , Th1 Cells/cytology , Th1 Cells/immunology , Th2 Cells/cytology , Th2 Cells/immunology , Th2 Cells/metabolism
19.
Int J Oncol ; 57(1): 197-212, 2020 07.
Article in English | MEDLINE | ID: mdl-32319593

ABSTRACT

Glucose­6­phosphate dehydrogenase (G6PD) is crucial rate­limiting enzyme of the pentose phosphate pathway (PPP). G6PD dysregulation has been reported in various types of human cancer, and the role of G6PD in cancer progression was demonstrated in numerous studies. A previous study from our laboratory described the prognostic significance of G6PD in clear cell renal cell carcinoma (ccRCC), and demonstrated its proliferative role through positive feedback regulation of the phosphorylated form of signal transducer and activator of transcription 3. However, the role of G6PD in ccRCC invasion remains unclear. In the present study, reverse transcription­quantitative (RT­q) PCR, western blotting, enzyme activity assay, transwell assay and immunohistochemistry analysis in cell model, xenograft mice model and human specimen studies were performed to evaluate the role of G6PD in ccRCC invasion. The results from the present study demonstrated that G6PD may promote ccRCC cell invasive ability by increasing matrix metalloproteinase 2 (MMP2) mRNA and protein expression both in vitro and in vivo. In addition, a positive correlation between G6PD and MMP2 expression was demonstrated by RT­qPCR and western blotting in twenty pairs of ccRCC tumor specimens and matched adjacent normal tissues. Furthermore, G6PD promoted reactive oxygen species (ROS) generation and activated the MAPK signaling pathway in ccRCC cells. In addition, ROS significantly promoted the MAPK signaling pathway activation, which in turn contributed to MMP2 overexpression in ccRCC cells. In conclusion, the present study demonstrated that G6PD may facilitate ccRCC cell invasive ability by enhancing MMP2 expression through ROS­MAPK axis pathway.


Subject(s)
Carcinoma, Renal Cell/pathology , Glucosephosphate Dehydrogenase/metabolism , Kidney Neoplasms/pathology , Matrix Metalloproteinase 2/metabolism , Cell Line, Tumor , Cell Proliferation/genetics , Gene Expression Regulation, Neoplastic , Gene Knockdown Techniques , Glucosephosphate Dehydrogenase/genetics , Humans , Kidney/pathology , MAP Kinase Signaling System/genetics , Neoplasm Invasiveness/pathology , Pentose Phosphate Pathway/genetics , Reactive Oxygen Species/metabolism , Specific Pathogen-Free Organisms , Xenograft Model Antitumor Assays
20.
Nutr Res ; 73: 1-14, 2020 01.
Article in English | MEDLINE | ID: mdl-31835095

ABSTRACT

The purpose of this review is to discuss the molecular mechanisms underlying the anticancer properties of S-allylcysteine (SAC). Over the decades, evidence derived from in vitro and in vivo studies has shown that this predominant organosulfur component of aged garlic extract has multiple anticancer properties; hence, some potential mechanisms responsible for the anticarcinogenic action have been suggested. These mechanisms include induction of carcinogen detoxification, inhibition of cell proliferation and growth, mediation of cell cycle arrest, induction of cell death, inhibition of epithelial-mesenchymal transition and cell invasion, suppression of metastasis, and induction of immunomodulation in cancer cells. However, the actions and mechanisms are not comprehensive, and important aspects of the anticancer activities of SAC still need to be explored. In light of the current evidence, more specific studies, specifically clinical and epidemiological, are required to advance the promising use of SAC as a chemopreventive and therapeutic agent in cancer.


Subject(s)
Antineoplastic Agents/pharmacology , Cysteine/analogs & derivatives , Garlic , Neoplasms/drug therapy , Animals , Apoptosis/drug effects , Cell Cycle Checkpoints/drug effects , Cell Proliferation/drug effects , Cysteine/pharmacology , Disease Models, Animal , Humans , Mice , Rats
SELECTION OF CITATIONS
SEARCH DETAIL