Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
Add more filters










Publication year range
1.
J Ethnopharmacol ; 307: 116226, 2023 May 10.
Article in English | MEDLINE | ID: mdl-36739926

ABSTRACT

ETHNOPHARMACOLOGICAL RELEVANCE: Ganoderma lucidum (Curtis) P. Karst., a bioactive mushroom with medicinal properties, is known to exert immunomodulatory, anti-inflammatory, hypocholesterolemic, hypoglycemic, and hepatoprotective effects. AIM OF THE STUDY: In this study, the effects of the G. lucidum fruiting body dry extract (GLE) on human liver (HepG2/C3A) and kidney (786-O) tumor cells and peripheral blood lymphocytes were evaluated. MATERIALS AND METHODS: MTT-based cytotoxicity, trypan blue-based cell viability, comet, and cytokinesis-block micronucleus cytome assays were performed, and the production of reactive oxygen species was evaluated in vitro. RESULTS: GLE was toxic to the tumor cells, decreasing their viability by increasing their production of reactive oxygen species and inducing damage to their DNA. By contrast, only high concentrations of GLE were toxic to lymphocytes and decreased their viability, whereas low concentrations increased lymphocyte viability. Moreover, primary DNA damage was induced by GLE only at the highest concentration tested. CONCLUSIONS: G. lucidum shows potential antitumor effects against cancerous kidney and liver cells, exhibiting cytotoxic and genotoxic activity at low concentrations, whereas the same effects in lymphocytes are mediated only at high concentrations. This mushroom has the potential to be biotechnologically developed into a therapeutic agent for diseases, such as cancer.


Subject(s)
Agaricales , Kidney Neoplasms , Reishi , Humans , Reactive Oxygen Species , Kidney , Liver , Lymphocytes
2.
Front Pharmacol ; 13: 1053184, 2022.
Article in English | MEDLINE | ID: mdl-36532756

ABSTRACT

Renal cell carcinoma (RCC) is a common malignant tumor of the urinary system, which is highly invasive, metastatic, and insensitive to radiotherapy and chemotherapy. Chinese herbal medicine has always been an important source of anti-tumor drug development. Reineckia carnea Kunth is a traditional herb commonly used by the Miao nationality in southwest China. In this study, the extract of Reineckia carnea was isolated and purified by reverse phase preparative chromatography and other chromatographic techniques. According to the physicochemical properties and spectral data, the structure of the compound was identified, and a novel biflavone compound named Reineckia-biflavone A (RFA) was obtained. The result of antiproliferative activity showed that RFA had cytotoxicity on 786-O cells with an IC50 value of 19.34 µmol/L. The results of CCK-8 and hemolysis assays showed that RFA was not significantly cytotoxic to both red blood cells (RBC) and peripheral blood mononuclear cells (PBMC). By Hoechst 33258 apoptosis staining, typical apoptotic morphology was observed under fluorescence microscope. RFA could induce the apoptosis of 786-O cells with the increase of apoptosis rate. The cell cycle tests showed that the cell proportion was obviously arrested in the S phase. At the same time, RFA could decrease the mitochondrial membrane potential and increase the intracellular free Ca2+ concentration. Western blot showed that the expression levels of pro-apoptotic proteins (Bax, Caspase-3, Cleaved Caspase-3, and Cytochrome c) in cells rose, while the expression level of anti-apoptotic proteins (Bcl-2) declined significantly. In conclusion, this study suggests that the RFA is a new biflavone determined by SciFinder retrieval. The apoptosis may be triggered by RFA through the mitochondrial pathway, which is mediated by up-regulating the intracellular calcium ion, down-regulating the mitochondrial membrane potential, and changing the apoptosis-related proteins.

3.
Cells ; 11(3)2022 01 29.
Article in English | MEDLINE | ID: mdl-35159281

ABSTRACT

The inactivation of von Hippel-Lindau (VHL) is critical for clear cell renal cell carcinoma (ccRCC) and VHL syndrome. VHL loss leads to the stabilization of hypoxia-inducible factor α (HIFα) and other substrate proteins, which, together, drive various tumor-promoting pathways. There is inadequate molecular characterization of VHL restoration in VHL-defective ccRCC cells. The identities of HIF-independent VHL substrates remain elusive. We reinstalled VHL expression in 786-O and performed transcriptome, proteome and ubiquitome profiling to assess the molecular impact. The transcriptome and proteome analysis revealed that VHL restoration caused the downregulation of hypoxia signaling, glycolysis, E2F targets, and mTORC1 signaling, and the upregulation of fatty acid metabolism. Proteome and ubiquitome co-analysis, together with the ccRCC CPTAC data, enlisted 57 proteins that were ubiquitinated and downregulated by VHL restoration and upregulated in human ccRCC. Among them, we confirmed the reduction of TGFBI (ubiquitinated at K676) and NFKB2 (ubiquitinated at K72 and K741) by VHL re-expression in 786-O. Immunoprecipitation assay showed the physical interaction between VHL and NFKB2. K72 of NFKB2 affected NFKB2 stability in a VHL-dependent manner. Taken together, our study generates a comprehensive molecular catalog of a VHL-restored 786-O model and provides a list of putative VHL-dependent ubiquitination substrates, including TGFBI and NFKB2, for future investigation.


Subject(s)
Carcinoma, Renal Cell , Kidney Neoplasms , Carcinoma, Renal Cell/metabolism , Cell Line, Tumor , Humans , Hypoxia , Kidney Neoplasms/pathology , Proteome , Von Hippel-Lindau Tumor Suppressor Protein/genetics
4.
Molecules ; 26(24)2021 Dec 11.
Article in English | MEDLINE | ID: mdl-34946600

ABSTRACT

Molecule interacting with CasL 2 (MICAL2), a cytoskeleton dynamics regulator, are strongly expressed in several human cancer types, especially at the invasive front, in metastasizing cancer cells and in the neo-angiogenic vasculature. Although a plethora of data exist and stress a growing relevance of MICAL2 to human cancer, it is worth noting that only one small-molecule inhibitor, named CCG-1423 (1), is known to date. Herein, with the aim to develop novel MICAL2 inhibitors, starting from CCG-1423 (1), a small library of new compounds was synthetized and biologically evaluated on human dermal microvascular endothelial cells (HMEC-1) and on renal cell adenocarcinoma (786-O) cells. Among the novel compounds, 10 and 7 gave interesting results in terms of reduction in cell proliferation and/or motility, whereas no effects were observed in MICAL2-knocked down cells. Aside from the interesting biological activities, this work provides the first structure-activity relationships (SARs) of CCG-1423 (1), thus providing precious information for the discovery of new MICAL2 inhibitors.


Subject(s)
Anilides , Benzamides , Enzyme Inhibitors , Microfilament Proteins , Oxidoreductases , Small Molecule Libraries , Humans , Anilides/chemistry , Anilides/pharmacology , Benzamides/chemistry , Benzamides/pharmacology , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Microfilament Proteins/antagonists & inhibitors , Microfilament Proteins/metabolism , Molecular Structure , Oxidoreductases/antagonists & inhibitors , Oxidoreductases/metabolism , Small Molecule Libraries/chemistry , Small Molecule Libraries/pharmacology
5.
Cancers (Basel) ; 13(11)2021 May 24.
Article in English | MEDLINE | ID: mdl-34073906

ABSTRACT

Genes associated with the DEAD-box helicase DDX11 are significant biomarkers of aggressive renal cell carcinoma (RCC), but their molecular function is poorly understood. We analyzed the molecular pathways through which DDX11 is involved in RCC cell survival and poly (ADP-ribose) polymerase (PARP) inhibitor sensitivity. Immunohistochemistry and immunoblotting determined DDX11 expression in normal kidney tissues, benign renal tumors, and RCC tissues and cell lines. Quantitative polymerase chain reaction validated the downregulation of DDX11 in response to transfection with DDX11-specific small interfering RNA. Proliferation analysis and apoptosis assays were performed to determine the impact of DDX11 knockdown on RCC cells, and the relevant effects of sunitinib, olaparib, and sunitinib plus olaparib were evaluated. DDX11 was upregulated in high-grade, advanced RCC compared to low-grade, localized RCC, and DDX11 was not expressed in normal kidney tissues or benign renal tumors. DDX11 knockdown resulted in the inhibition of RCC cell proliferation, segregation defects, and rapid apoptosis. DDX11-deficient RCC cells exhibited significantly increased sensitivity to olaparib compared to sunitinib alone or sunitinib plus olaparib combination treatments. Moreover, DDX11 could determine PARP inhibitor sensitivity in RCC. DDX11 could serve as a novel therapeutic biomarker for RCC patients who are refractory to conventional targeted therapies and immunotherapies.

6.
Bioorg Med Chem Lett ; 43: 128058, 2021 07 01.
Article in English | MEDLINE | ID: mdl-33895276

ABSTRACT

The protein kinase R (PKR)-like endoplasmic reticulum kinase (PERK) is one of the three endoplasmic reticulum (ER) transmembrane sensors of the unfolded protein response (UPR) that regulates protein synthesis, alleviates cellular ER stress and has been implicated in tumorigenesis and prolonged cancer cell survival. In this study, we report a series of 2-amino-3-amido-5-aryl-pyridines that we have identified as potent, selective, and orally bioavailable PERK inhibitors. Amongst the series studied herein, compound (28) a (R)-2-Amino-5-(4-(2-(3,5-difluorophenyl)-2-hydroxyacetamido)-2-ethylphenyl)-N-isopropylnicotinamide has demonstrated potent biochemical and cellular activity, robust pharmacokinetics and 70% oral bioavailability in mice. Given these data, this compound (28) was studied in the 786-O renal cell carcinoma xenograft model. We observed dose-dependent, statistically significant tumor growth inhibition, supporting the use of this tool compound in additional mechanistic studies.


Subject(s)
Drug Discovery , Pyridines/pharmacology , eIF-2 Kinase/antagonists & inhibitors , Administration, Oral , Biological Availability , Dose-Response Relationship, Drug , Humans , Molecular Structure , Pyridines/administration & dosage , Pyridines/chemistry , Structure-Activity Relationship , eIF-2 Kinase/metabolism
7.
Article in Chinese | WPRIM (Western Pacific) | ID: wpr-912115

ABSTRACT

Objective:To investigate the effects of B7-H3 molecule on clear cell renal cell carcinoma (786-O) metastasis.Methods:Lentiviral transfection method was used to construct 786-O cells stably expressing low level of B7-H3 (shB7-H3 group) and a negative control cell line (shNC group). RT-qPCR, flow cytometry and Western blot were used to assess the efficiency of lentiviral transfection. CCK-8 method was used to detect the proliferation of 786-O cells in the two groups. Flow cytometry was performed to detect the changes in cell cycle. Cell scratch test and Transwell assay were used to detect the differences in cell migration and invasion. Western blot was used to detect the expression of marker proteins in the process of epithelial-mesenchymal transition (epithelial-mesenchymal transition, EMT). Changes in the expression of chemokines and their receptors were analyzed by flow cytometry and RT-qPCR. Effects of anti-CCL4 antibody on cell migration and invasion were analyzed by Transwell assay.Results:Flow cytometry showed that 786-O cells highly expressed B7-H3 molecules and the lentiviral transfection method successfully constructed the cell line with lower expression of B7-H3 (786-O-shB7-H3) and control cell line (786-O-shNC). B7-H3 molecule had no significant effect on the proliferation of 786-O cells. No significant difference in cell cycle was found between the two groups. Compared with 786-O-shNC cells, the migration and invasion ability of 786-O-shB7-H3 cells was suppressed. Moreover, the expression of EMT-related marker proteins (fibronectin and N-cadherin) was reduced and the expression of E-cadherin was increased in 786-O-shB7-H3 cells. The expression of CCL4 and its receptor CCR5 in the shB7-H3 group was lower than that in the shNC group. After intervention with anti-CCL4 antibody, the migration and invasion ability of 786-O-shNC cells was reduced, while that of 786-O-shB7-H3 cells had no significant change.Conclusions:Knocking down the expression of B7-H3 molecule had no significant effect on the proliferation of 786-O cells, but could affect the EMT process of 786-O cells and reduce tumor migration and invasion ability, thereby inhibiting tumor progression.

8.
Chin Med ; 15: 77, 2020.
Article in English | MEDLINE | ID: mdl-32760434

ABSTRACT

BACKGROUND: Cisplatin (CDDP) is a chemotherapeutic drug which also causes adverse side effects. Glechoma hederacea is a traditional Chinese herb belonging to the Labiatae family and has many biological activities. Our previous study indicated that rosmarinic acid (RA) was the most abundant phytochemical in G. hederacea. However, the antioxidant or anti-inflammatory effects of the combined treatment of G. hederacea, RA and CDDP on human renal cell carcinoma (RCC) 786-O cells have not been clearly demonstrated. We aimed to investigate the bioefficacy of hot water extracts of G. hederacea (HWG) and RA in inhibiting RCC 786-O cell activity and its synergism with CDDP against metastatic renal cancer cell. METHODS: Bioactivities of the combination treatment of HWG, RA, HWG/CDDP and RA/CDDP were assessed using the MTT assay and transwell migration, and the crude extract/compound efficacy was evaluated using wound healing migration assays, flow cytometry and western blotting. RESULTS: Our study indicates that CDDP inhibits 786-O cell proliferation and migration and HWG and RA protect against these effects. On the other hand, HWG and RA demonstrate a low cytotoxic effect in human renal proximal tubular epithelial cell line -2 (HK-2 cells). Cell cycle analysis found that HWG/CDDP and RA/CDDP combined treatment exerted cytotoxicity by inducing G2/M arrest and apoptosis. RA in combined with CDDP significantly inhibiting the expression of p-FAK (Tyr 925) in RCC 786-O cells in vitro. CONCLUSION: We propose that the inhibition of RA on RCC 786-O cell invasion and migration may partly occur through the downregulation of FAK phosphorylation. The HWG/CDDP and RA/CDDP combined treatments may be effective strategies for intervention of RCC 786-O cell activity.

9.
Zhonghua Yi Xue Za Zhi ; 100(24): 1890-1894, 2020 Jun 23.
Article in Chinese | MEDLINE | ID: mdl-32575934

ABSTRACT

Objective: To investigate the role of Wnt/ß-catenin/TCF-4 pathway in renal cancer cells and to analyze its possible mechanism. Methods: ß-catenin and TCF-4 were inhibited by siRNA in 786-O cells. The proliferation of transfected cells was detected by CCK8. The cell death of transfected cells was detected by acridine orange -ethidium bromide staining. The expressions of TCF-4, bcl-2, bax and Caspase-3 were detected in transfection group, empty vector group and negative control groups by western blot. Results: The cell proliferation ability of the ß-catenin transfection group was significantly lower than that of the control group (0.443±0.145 vs 0.910±0.721), meanwhile, the cell death rate was significantly increased (16.38±5.32 vs 6.61±1.04), the expression level of Caspase 3 and bax was increased, and the expression of anti-apoptotic protein Bcl-2 was decreased. Decreased TCF-4 led to the same results as inhibition of ß-catenin (all P<0.05). Conclusion: The Wnt/ß-catenin/TCF-4 pathway may play a role in the regulation of proliferation and apoptosis in 786-O renal cancer cells. The mechanism might through regulating of the downstream apoptosis proteins Caspase 3, bax and anti-apoptotic protein Bcl-2.


Subject(s)
Carcinoma, Renal Cell , Kidney Neoplasms , Apoptosis , Cell Line, Tumor , Cell Proliferation , Humans , Wnt Signaling Pathway , beta Catenin
10.
Biomolecules ; 10(2)2020 02 04.
Article in English | MEDLINE | ID: mdl-32033222

ABSTRACT

The renal cell carcinoma (RCC) is the most common type of kidney cancer. Identifying novel and more effective therapies, while minimizing toxicity, continues to be fundamental in curtailing RCC. Rutin, a bioflavonoid widely found in nature, has shown promising anticancer properties, but with limited applicability due to its poor water solubility and pharmacokinetics. Thus, the potential anticancer effects of rutin toward a human renal cancer cell line (786-O), while considering its safety in Vero kidney cells, was assessed, as well as the applicability of ionic liquids (ILs) to improve drug delivery. Rutin (up to 50 µM) did not show relevant cytotoxic effects in Vero cells. However, in 786-O cells, a significant decrease in cell viability was already observed at 50 µM. Moreover, exposure to rutin caused a significant increase in the sub-G1 population of 786-O cells, reinforcing the possible anticancer activity of this biomolecule. Two choline-amino acid ILs, at non-toxic concentrations, enhanced rutin's solubility/loading while allowing the maintenance of rutin's anticancer effects. Globally, our findings suggest that rutin may have a beneficial impact against RCC and that its combination with ILs ensures that this poorly soluble drug is successfully incorporated into ILs-nanoparticles hybrid systems, allowing controlled drug delivery.


Subject(s)
Antineoplastic Agents/administration & dosage , Carcinoma, Renal Cell/drug therapy , Drug Carriers/chemistry , Ionic Liquids/chemistry , Kidney Neoplasms/drug therapy , Rutin/administration & dosage , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Carcinoma, Renal Cell/pathology , Cell Cycle/drug effects , Cell Line, Tumor , Cell Survival/drug effects , Chlorocebus aethiops , Drug Liberation , Humans , Kidney Neoplasms/pathology , Nanoparticles/chemistry , Rutin/chemistry , Rutin/pharmacology , Solubility , Vero Cells
11.
Article in Chinese | WPRIM (Western Pacific) | ID: wpr-821004

ABSTRACT

@# Objective: To investigate the effect of long non-coding RNA (lncRNA) lung cancer associated transcript 1 (LUCAT1) on proliferation and migration of clear cell renal cell carcinoma (ccRCC) 786-O cells and the underlying mechanism. Methods: A total of 40 pairs of pathologically confirmed tumor tissues and corresponding adjacent normal tissues from ccRCC patients, who underwent surgical resection in the Department of Urology, the First People's Hospital of Yichang during June 2013 and June 2017, were selected for this study. ccRCC cell lines (786-O, ACHN, UM-RC-2) and normal renal epithelial KiMA cells were also used in this study. qPCR was used to detect the mRNA expressions of LUCAT1, miR-199a-5p and hypoxia inducible fator 1α (HIF-1α) in above mentioned tissues and cell lines; CCK-8 assay was used to evaluate the proliferation of 786-O cells; Transwell assay was used to evaluate the migration of 786-O cells; Dual luciferase reporter gene assay was performed to validate the relationship between LUCAT1 and miR-199a-5p; and Western blotting was conducted to detect the effect of LUCAT1 and miR-199a-5p on the protein expression of HIF-1α. Results: LUCAT1 was significantly up-regulated in ccRCC tissues and cell lines (all P<0.01), and its knockdown significantly inhibited the proliferation and migration of 786-O cells (all P<0.01). miR-199a-5p was low-expressed in ccRCC tissues and cell lines (all P<0.01), StarBase analysis showed that LUCAT1 contained a conserved target site for miR-199a-5p. miR-199a-5p exerted significant suppression on the luciferase activity of LUCAT1-Wt (P<0.01), and LUCAT1 knockdown significantly reduced miR-199a-5p expression (P< 0.01). LUCAT1 was low-expressed in 786-O cells transfected with miR-199a-5p mimics, however, it was attenuated after co-transfection with LUCAT1. The mRNA and protein expressions of HIF-1α in 786-O cells transfected with miR-199a-5p mimics were up-regulated, which was then reversed by LUCAT1 over-expression (P<0.05 or P<0.01). miR-199a-5p over-expression suppressed the proliferation and migration of 786-O cells, which was partially attenuated by LUCAT1 transfection (P<0.05 or P<0.01). Conclusion: LUCAT1 exerts oncogenic function in ccRCC via regulating miR-199a-5p/HIF-1α axis.·

12.
Nat Prod Res ; 33(21): 3044-3051, 2019 Nov.
Article in English | MEDLINE | ID: mdl-30362364

ABSTRACT

Five new lanostane-type triterpenoids, named piptolinic acids F - J (1-5), as well as seven known analogues (6-12), were isolated from methanolic extract of the fruiting bodies of Piptoporus betulinus. Compounds 1-4 were 24-methyl-lanostane triterpenoids, while compound 5 was a 3,4-seco-lanostane derivative. Their structures were established on the basis of extensive spectroscopic analysis (1D, 2D NMR, and HRESIMS). Cytotoxicity evaluation indicated that compound 6 exhibited moderate cytotoxic activity against human melanoma cell line A-375 (IC50 = 42.8 µM) and human renal carcinoma cell line 786-O (IC50 = 56.5 µM).


Subject(s)
Fruiting Bodies, Fungal/chemistry , Ganoderma/chemistry , Lanosterol/analogs & derivatives , Triterpenes/isolation & purification , Cell Line, Tumor , Drug Screening Assays, Antitumor , Humans , Inhibitory Concentration 50 , Molecular Structure , Spectrum Analysis , Triterpenes/chemistry
13.
Article in Chinese | WPRIM (Western Pacific) | ID: wpr-793114

ABSTRACT

@# Objective: To explore the mechanism of miR-19a-3p regulating cell adhesion molecule 2 (CADM2) to inhibit the proliferation and metastasis of renal carcinoma cells via the AKT signaling pathway. Methods: A total of 42 patients with renal cancer admitted to Department of Nephrology, the First Affiliated Hospital of Suzhou University from April 2012 to November 2017 were enrolled to collect samples of surgically resected renal carcinoma tissues and paracancerous tissues. Expression of miR-19a-3p was detected in renal carcinoma tissues and 4 types of renal carcinoma cell lines such as 786-O by quantitative Real-time polymerase chain reaction (qPCR). The effects of miR-19a-3p knockdown on proliferation, invasion and epithelial mesenchymal transition (EMT) of renal carcinoma 786-O cells were evaluated by CCK-8 assay, Transwell assay and immunofluorescence, respectively. Subsequently, dual luciferase reporter assay was used to verify whether CADM2 was a target gene of miR-19a-3p. Furthermore, Wb was applied to detect the regulatory effect of miR-19a-3p onAKT signaling pathway through CADM2. Results: miR-19a-3p expression was significantly up-regulated in renal carcinoma tissues and cell lines (all P<0.01). Knockdown of miR-19a-3p could inhibit proliferation, invasion and EMT process of 786-O cells; furthermore, the results indicated that CADM2 was a direct target of miR-19a-3p and its expression was down-regulated (P <0.05 or P<0.01). Additionally, knockdown of miR-19a-3p obviously suppressed proliferation, migration and EMT process of 786-O cells via up-regulating CADM2 and blocking AKT pathway (all P<0.05 or P<0.01), thus alleviating the occurrence and development of renal carcinoma. Conclusion: The study demonstrates that miR-19a-3p has a high expression level in renal carcinoma tissues; knockdown of miR-19a-3p could significantly inhibit the proliferation, migration and EMT process of renal carcinoma tissues, and its mechanism may be associated with miR-19a-3p/CADM2/AKT axis.

14.
Article in Chinese | WPRIM (Western Pacific) | ID: wpr-802237

ABSTRACT

Objective: To investigate the anticancer effect of isoliquiritigenin (ISL) on human clear cell renal cell carcinoma 786-O cells, and explore its possible molecular mechanism. Method: Thiazolyl blue tetrazolium bromide (MTT) assay was used to detect effect of ISL (0, 10, 25,50, 75, 100 μmol·L-1) on proliferation of 786-O cells. The effect of ISL on migration and invasion of 786-O cells was detected by cell scratch test and Transwell assay. The autophagy was observed under the fluorescence microscope through acridine orange staining and Ad-GFP-LC3 transfection experiment. Western blot was used to detect the expression of autophagy related protein and analyze the changes of phosphatidylinositol-3-kinase (PI3K)/protein kinase B(Akt)/mammalian target of rapamycin (mTOR) signaling pathway to explore the possible mechanism. Result: MTT results showed that ISL could significantly inhibit the proliferation of 786-O cells in a time-dose dependent manner (PPPPPPPPConclusion: ISL can inhibit the proliferation, migration and invasion of clear cell renal carcinoma 786-O cells, and induce autophagy by inhibiting the PI3K/Akt/mTOR signaling pathway.

15.
Oncol Lett ; 16(1): 1285-1290, 2018 Jul.
Article in English | MEDLINE | ID: mdl-30061948

ABSTRACT

Previous reports have indicated that the abundance of specific microRNAs (miRNA) contained within the exosome/microvesicle compartment of patient biofluids may be useful in diagnosing specific types of cancer. In the present study, the 786-O cell line, which is derived from a clear cell renal cell carcinoma (ccRCC), was used as an in vitro ccRCC tumor model and the human renal proximal tubule cell line HK-2 was used as its normal renal tissue control to investigate the similarities of exosomal content of selected ccRCC miRNA biomarkers in the supernatant with the content of those markers in the cells themselves. A PCR array identified miRNA biomarkers of solid RCC tumors (miR-210, MiR-34a, miR-155-5p and miR-150-5p) that were increased by 2-8 fold in 786-O exosomes compared with the control. These were subsequently chosen for further investigation using TaqMan RT-qPCR in addition to miR-15a and miR-205, which were selected based on prior interest as RCC biomarkers. MiR-15a, -34a, -210 and -155 levels were significantly lower in exosomes when compared with that in whole cells but did not differ between the HK-2 and 786-O cells in either the cytoplasmic, exosome or exosome-free supernatant fractions. By contrast, cytoplasmic miR-150 and miR-205 exhibited significant differences in concentration between the two cell lines. In addition, the cytoplasmic content of miR-150 and miR-205 was mirrored in the exosomal content of these miRNAs. Furthermore, the difference in exosomal miR-205 content was statistically significant. The present study indicated that measurements of the exosomal content of miR-205 and possibly miR-150, but not those of the other examined miRNAs, are proportional to their respective contents in the cells that secreted them. These findings suggest that in vitro RCC systems may be useful in identifying miRNAs with sufficiently high levels of exportation into exosomes; and with sufficiently different expression levels between tumor and normal cells to serve as ccRCC biomarkers in vivo.

16.
Talanta ; 175: 235-242, 2017 Dec 01.
Article in English | MEDLINE | ID: mdl-28841985

ABSTRACT

Renal cell carcinoma (RCC) is the most common form of kidney cancer with poor prognosis. Early diagnosis of RCC would significantly improve patient prognosis and quality of life. In this work, we developed new aptamer probes for RCC by using cell-SELEX (systematic evolution of ligands by exponential enrichment) only after 12 rounds of selection, in which a clear cell renal cell carcinoma (ccRCC) cell line 786-O was used as target cell, and embryonic kidney cell line 293T as negative control cell. The selected aptamers were subjected to flow cytometry and laser confocal fluorescence microscopy to evaluate their binding affinity and selectivity. The dissociation constant Kd values of four selected aptamers are all in the nanomolar range. Aptamer W786-1 with the best binding affinity and a Kd value of 9.4 ± 2.0nM was further optimized and its truncated sequence W786-1S showed considerable affinity to 786-O cells. The proteinase and temperature treatment experiment indicated that W786-1 could recognize the target 786-O cells through surface proteins, and remain good binding affinity and excellent selectivity under physiological conditions. Therefore, on the basis of its excellent targeting properties and functional versatility, W786-1 holds great potential to be used as a molecular probe for identifying and targeting RCC.


Subject(s)
Aptamers, Nucleotide/chemistry , Biosensing Techniques/methods , Carcinoma, Renal Cell/diagnosis , Kidney Neoplasms/diagnosis , SELEX Aptamer Technique/methods , Base Sequence , Binding Sites , Carcinoma, Renal Cell/chemistry , Carcinoma, Renal Cell/pathology , Cell Line, Tumor , Flow Cytometry/methods , HEK293 Cells , Humans , Kidney Neoplasms/chemistry , Kidney Neoplasms/pathology , Microscopy, Confocal/methods , Microscopy, Fluorescence/methods
17.
Med Mol Morphol ; 50(4): 220-226, 2017 Dec.
Article in English | MEDLINE | ID: mdl-28631038

ABSTRACT

T-cell immunoglobulin and mucin domain (TIMD) family genes are related to innate immune responses. TIMD4 is a receptor for phosphatidylserine and is involved in the phagocytosis of apoptotic cells by macrophages. In the present study, we found that TIMD4 is expressed on the cancer cells of patients with clear cell renal cell carcinoma (ccRCC). TIMD4 was immunostained in the resected samples of 89 patients diagnosed as ccRCC. High expression of TIMD4 in cancer cells was closely related to short progression free survival time; however, it was not correlated with other clinicopathological factors. Intracellular expression of TIMD4 was observed in the RCC cell line, 786-O. In vitro studies using 786-O cells and shRNA targeting TIMD4 indicated that TIMD4 expression was associated with resistance to sorafenib but not with cell proliferation. TIMD4 might be useful as a prognostic factor and may also be a new target for therapy of ccRCC.


Subject(s)
Carcinoma, Renal Cell/genetics , Drug Resistance, Neoplasm/genetics , Kidney Neoplasms/genetics , Membrane Proteins/genetics , Aged , Antineoplastic Agents/pharmacology , Carcinoma, Renal Cell/drug therapy , Carcinoma, Renal Cell/mortality , Carcinoma, Renal Cell/surgery , Cell Line, Tumor , Cell Proliferation/drug effects , Disease-Free Survival , Female , Gene Expression , Humans , Kidney Neoplasms/drug therapy , Kidney Neoplasms/mortality , Kidney Neoplasms/surgery , Male , Membrane Proteins/antagonists & inhibitors , Membrane Proteins/metabolism , Middle Aged , Niacinamide/analogs & derivatives , Niacinamide/pharmacology , Phenylurea Compounds/pharmacology , Prognosis , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Sorafenib
18.
Article in Chinese | WPRIM (Western Pacific) | ID: wpr-658161

ABSTRACT

Objective To investigate the expression of miR-21 in renal clear cell carcinoma and its clinical significance as well as how miR-21 regulates the proliferation and apoptosis of 786-O renal clear cell carcinoma cell line through regulating programmed cell death 4 (PDCD4).Methods By analyzing the data of renal clear cells cancer in The Cancer Genome Atlas (TCGA)database,we compared the expression of miR-21 in renal cancer tissues and adjacent normal tissues and explored the differences in miR-21 level in renal cancer at different clinicopathological stage,T stage,N stage and M stage.We also analyzed the association between miR-21 level and survival of patients by Kaplan-Meier method and Log-rank test.786-O cells were transfected with AS-miR-21 to deplete miR-21. MTT assay and flow cytometry were applied to measure cell proliferation and apoptosis, respectively.We then measured the mRNA and protein levels of PDCD4 in 786-O cells depleted for miR-21 by qRT-PCR and Western blot,respectively,and performed a dual-luciferase assay to detect the direct regulation of PDCD4 by miR-21.Results Expression of miR-21 was significantly higher in renal cancer tissues than in adjacent tissues(P <0.0001).The expression levels of miR-21 at stage Ⅲ and stage Ⅳ renal cancer were significantly higher than that at stage Ⅰ (both P <0.0001).Moreover,miR-21 expression was positively correlated with clinicopathological stages of renal cancer by correlation analysis (r =0.262,P <0.0001 ).The correlation test indicated that miR-21 level was also positively correlated with T stage of renal cancer (r =0.250,P <0.0001 ),lymph node metastasis (N1)and distant metastasis (all P <0.0002).Patients with high miR-21 expression had significantly shorter median survival time than those with low miR-21 expression (Log-rank P < 0.001 ).Compared with control cells,786-O cells depleted for miR-21 showed significantly decreased cell proliferation (P <0.05 )and increased cell apoptosis rate (P =0.005 ).PDCD4 mRNA (P = 0.002 )and protein levels were significantly elevated in 786-O cells with down-regulated miR-21 levels.In addition,the dual-luciferase reporter assay showed that the relative luciferase intensity of PDCD4 reporter in cells transfected with AS-miR-21 was significantly higher than that of control cells (P =0.003).Conclusion miR-21 expression was up-regulated in renal cancer and correlated with clinicopathological stage and survival of patients.miR-21 promoted 786-O cell proliferation and inhibited apoptosis probably through regulating PDCD4 expression. These results indicate that miR-21 plays an important role in formation and development of renal cancer.

19.
Article in Chinese | WPRIM (Western Pacific) | ID: wpr-660955

ABSTRACT

Objective To investigate the expression of miR-21 in renal clear cell carcinoma and its clinical significance as well as how miR-21 regulates the proliferation and apoptosis of 786-O renal clear cell carcinoma cell line through regulating programmed cell death 4 (PDCD4).Methods By analyzing the data of renal clear cells cancer in The Cancer Genome Atlas (TCGA)database,we compared the expression of miR-21 in renal cancer tissues and adjacent normal tissues and explored the differences in miR-21 level in renal cancer at different clinicopathological stage,T stage,N stage and M stage.We also analyzed the association between miR-21 level and survival of patients by Kaplan-Meier method and Log-rank test.786-O cells were transfected with AS-miR-21 to deplete miR-21. MTT assay and flow cytometry were applied to measure cell proliferation and apoptosis, respectively.We then measured the mRNA and protein levels of PDCD4 in 786-O cells depleted for miR-21 by qRT-PCR and Western blot,respectively,and performed a dual-luciferase assay to detect the direct regulation of PDCD4 by miR-21.Results Expression of miR-21 was significantly higher in renal cancer tissues than in adjacent tissues(P <0.0001).The expression levels of miR-21 at stage Ⅲ and stage Ⅳ renal cancer were significantly higher than that at stage Ⅰ (both P <0.0001).Moreover,miR-21 expression was positively correlated with clinicopathological stages of renal cancer by correlation analysis (r =0.262,P <0.0001 ).The correlation test indicated that miR-21 level was also positively correlated with T stage of renal cancer (r =0.250,P <0.0001 ),lymph node metastasis (N1)and distant metastasis (all P <0.0002).Patients with high miR-21 expression had significantly shorter median survival time than those with low miR-21 expression (Log-rank P < 0.001 ).Compared with control cells,786-O cells depleted for miR-21 showed significantly decreased cell proliferation (P <0.05 )and increased cell apoptosis rate (P =0.005 ).PDCD4 mRNA (P = 0.002 )and protein levels were significantly elevated in 786-O cells with down-regulated miR-21 levels.In addition,the dual-luciferase reporter assay showed that the relative luciferase intensity of PDCD4 reporter in cells transfected with AS-miR-21 was significantly higher than that of control cells (P =0.003).Conclusion miR-21 expression was up-regulated in renal cancer and correlated with clinicopathological stage and survival of patients.miR-21 promoted 786-O cell proliferation and inhibited apoptosis probably through regulating PDCD4 expression. These results indicate that miR-21 plays an important role in formation and development of renal cancer.

20.
Oncol Lett ; 12(3): 2201-2209, 2016 Sep.
Article in English | MEDLINE | ID: mdl-27602164

ABSTRACT

The chemokine monocyte chemoattractant protein-1 [MCP-1; also known as chemokine (C-C motif) ligand 2] is an important mediator of monocyte recruitment during inflammatory processes. Pathologically high expression levels of MCP-1 by tumor cells have been observed in a variety of cancer types. In the majority of cases, high MCP-1 expression is associated with a poor prognosis, as infiltration of the tumor with inflammatory monocytes promotes tumor progression and metastasis. MCP-1 is also expressed in renal cell carcinoma (RCC). In the present study, the function and the regulation of MCP-1 was investigated in two RCC cell lines, CaKi-1 and 786-O. In both cell lines, expression of MCP-1 was significantly enhanced compared with non-cancerous control cells. As expected, secretion of MCP-1 into the medium facilitated the recruitment of peripheral blood monocytes via the chemokine (C-C motif) receptor type 2 (CCR2). As expression of CCR2 was also detected in 786-O and CaKi-1 cells, the effect of autocrine MCP-1/CCR2 signaling was evaluated in these cells. In proliferation assays, administration of an MCP-1 neutralizing antibody or of a CCR2 antagonist to CaKi-1 and 786-O cells significantly decreased cell growth; supplementation of the growth medium with recombinant human MCP-1 had no additional effect on proliferation. The migration ability of RCC cells was impaired by MCP-1 neutralization or pharmacological CCR2 inhibition, while it was stimulated by the addition of recombinant human MCP-1, compared with untreated control cells. Finally, substantial differences in the regulation of MCP-1 expression were observed between RCC cell lines. In CaKi-1 cells, expression of MCP-1 appears to be largely mediated by the transcription factor nuclear factor of activated T cells 5, while in 786-O cells, deletion of the tumor suppressor gene Von-Hippel-Lindau appeared to be responsible for MCP-1 upregulation, as suggested by previous studies. Taken together, the results of the current study indicate that expression of MCP-1 in RCC cells promotes tumor progression and metastasis not only by paracrine, but also by autocrine, MCP-1/CCR2 signaling events, enhancing cell proliferation and migration ability. Therefore, the present findings suggest the MCP-1/CCR2 axis is a potential target for future therapeutic strategies in the treatment of metastatic RCC.

SELECTION OF CITATIONS
SEARCH DETAIL
...