Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 6 de 6
1.
Int J Mol Sci ; 22(9)2021 Apr 29.
Article En | MEDLINE | ID: mdl-33946824

Red blood cells (RBCs) serve a variety of functions beyond mere oxygen transport both in health and pathology. Notably, RRx-001, a minimally toxic pleiotropic anticancer agent with macrophage activating and vascular normalization properties currently in Phase III trials, induces modification to RBCs which could promote vascular adhesion similar to sickle cells. This study assessed whether RBCs exposed to RRx-001 adhere to the tumor microvasculature and whether this adhesion alters tumor viability. We next investigated the biomechanics of RBC adhesion in the context of local inflammatory cytokines after treatment with RRx-001 as a potential mechanism for preferential tumor aggregation. Human HEP-G2 and HT-29 tumor cells were subcutaneously implanted into nu/nu mice and were infused with RRx-001-treated and Technetium-99m (99mTc)-labeled blood. RBC adhesion was quantified in an in vitro human umbilical vein endothelial cell (HUVEC) assay under both normoxic and hypoxic conditions with administration of either lipopolysaccharide (LPS) or Tumor necrosis alpha (TNFα) to mimic the known inflammation in the tumor microenvironment. One hour following administration of 99mTc labeled RBCs treated with 10 mg/kg RRx-001, we observed an approximate 2.0-fold and 1.5-fold increase in 99mTc-labeled RBCs compared to vehicle control in HEPG2 and HT-29 tumor models, respectively. Furthermore, we observed an approximate 40% and 36% decrease in HEP-G2 and HT-29 tumor weight, respectively, following treatment with RRx-001. To quantify RBC adhesive potential, we determined τ50, or the shear stress required for 50% disassociation of RBCs from HUVECs. After administration of TNF-α under normoxia, τ50 was determined to be 4.5 dynes/cm2 (95% CI: 4.3-4.7 dynes/cm2) for RBCs treated with 10 µM RRx-001, which was significantly different (p < 0.05) from τ50 in the absence of treatment. Under hypoxic conditions, the difference of τ50 with (4.8 dynes/cm2; 95% CI: 4.6-5.1 dynes/cm2) and without (2.6 dynes/cm2; 95% CI: 2.4-2.8 dynes/cm2) 10 µM RRx-001 treatment was exacerbated (p = 0.05). In conclusion, we demonstrated that RBCs treated with RRx-001 preferentially aggregate in HEP-G2 and HT-29 tumors, likely due to interactions between RRx-001 and cysteine residues within RBCs. Furthermore, RRx-001 treated RBCs demonstrated increased adhesive potential to endothelial cells upon introduction of TNF-α and hypoxia suggesting that RRx-001 may induce preferential adhesion in the tumor but not in other tissues with endothelial dysfunction due to conditions prevalent in older cancer patients such as heart disease or diabetic vasculopathy.


Antineoplastic Agents/pharmacology , Azetidines/pharmacology , Endothelial Cells/cytology , Erythrocyte Membrane/drug effects , Nitro Compounds/pharmacology , Animals , Antineoplastic Agents/therapeutic use , Azetidines/therapeutic use , Cell Adhesion/drug effects , Cell Hypoxia , Cysteine/chemistry , Cytokines/metabolism , Endothelial Cells/chemistry , Erythrocyte Aggregation/drug effects , Erythrocyte Membrane/chemistry , HT29 Cells/transplantation , Hep G2 Cells/transplantation , Human Umbilical Vein Endothelial Cells , Humans , Lipopolysaccharides/pharmacology , Membrane Lipids/biosynthesis , Mice , Mice, Nude , Neoplasms/blood supply , Neoplasms, Experimental/blood supply , Neoplasms, Experimental/drug therapy , Nitro Compounds/therapeutic use , Phosphatidylserines/biosynthesis , Receptors, Cell Surface/biosynthesis , Shear Strength , Tumor Microenvironment , Tumor Necrosis Factor-alpha/pharmacology
2.
J Cell Mol Med ; 24(18): 10525-10541, 2020 09.
Article En | MEDLINE | ID: mdl-32798252

Hepatocellular carcinoma (HCC) is the third leading cause of the cancer-related death in the world. Human amniotic mesenchymal stem cells (hAMSCs) have been characterized with a pluripotency, low immunogenicity and no tumorigenicity. Especially, the immunosuppressive and anti-inflammatory effects of hAMSCs make them suitable for treating HCC. Here, we reported that hAMSCs administrated by intravenous injection significantly inhibited HCC through suppressing cell proliferation and inducing cell apoptosis in tumour-bearing mice with Hepg2 cells. Cell tracking experiments with GFP-labelled hAMSCs showed that the stem cells possessed the ability of migrating to the tumorigenic sites for suppressing tumour growth. Importantly, both hAMSCs and the conditional media (hAMSC-CM) have the similar antitumour effects in vitro, suggesting that hAMSCs-derived cytokines might be involved in their antitumour effects. Antibody array assay showed that hAMSCs highly expressed dickkopf-3 (DKK-3), dickkopf-1 (DKK-1) and insulin-like growth factor-binding protein 3 (IGFBP-3). Furthermore, the antitumour effects of hAMSCs were further confirmed by applications of the antibodies or the specific siRNAs of DKK-3, DKK-1 and IGFBP-3 in vitro. Mechanically, hAMSCs-derived DKK-3, DKK-1 and IGFBP-3 markedly inhibited cell proliferation and promoted apoptosis of Hepg2 cells through suppressing the Wnt/ß-catenin signalling pathway and IGF-1R-mediated PI3K/AKT signalling pathway, respectively. Taken together, our study demonstrated that hAMSCs possess significant antitumour effects in vivo and in vitro and might provide a novel strategy for HCC treatment clinically.


Amnion/cytology , Carcinoma, Hepatocellular/therapy , Liver Neoplasms/therapy , Mesenchymal Stem Cell Transplantation , Adaptor Proteins, Signal Transducing/antagonists & inhibitors , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/physiology , Adipogenesis , Animals , Apoptosis , Carcinoma, Hepatocellular/pathology , Female , Genes, Reporter , Hep G2 Cells/transplantation , Humans , Insulin-Like Growth Factor Binding Protein 3/antagonists & inhibitors , Insulin-Like Growth Factor Binding Protein 3/genetics , Insulin-Like Growth Factor Binding Protein 3/physiology , Intercellular Signaling Peptides and Proteins/genetics , Intercellular Signaling Peptides and Proteins/physiology , Liver Neoplasms/pathology , Male , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/metabolism , Mice , Mice, Inbred BALB C , Mice, Inbred NOD , Mice, Nude , Mice, SCID , Neoplasm Proteins/antagonists & inhibitors , Neoplasm Proteins/genetics , Neoplasm Proteins/physiology , Osteogenesis , Paracrine Communication , Pregnancy , RNA Interference , RNA, Small Interfering/genetics , RNA, Small Interfering/pharmacology , Signal Transduction , Xenograft Model Antitumor Assays
3.
J Proteome Res ; 17(7): 2428-2439, 2018 07 06.
Article En | MEDLINE | ID: mdl-29750532

Targeted therapy of hepatocellular carcinoma (HCC) is essential for improved therapies. Therefore, identification of key targets specifically to HCC is an urgent requirement. Herein, an iTRAQ quantitative proteomic approach was employed to identify differentially expressed proteins in HCC tumor tissues. Of the upregulated tumor-related proteins, minichromosome maintenance 2 (MCM2), a DNA replication licensing factor, was one of the most significantly altered proteins, and its overexpression was confirmed using tissue microarray. Clinicopathological analysis of multiple cohorts of HCC patients indicated that overexpression of MCM2 was validated in 89.8% tumor tissues and strongly correlated with clinical stage. Furthermore, siRNA-mediated repression of MCM2 expression resulted in significant suppression of the HepG2 cell cycle and proliferation through the cyclin D-dependent kinases (CDKs) 2/7 pathway. Finally, the first small molecule-based MCM2-targeted NIR-II probe CH1055-MCM2 was concisely generated and subsequently evaluated in mice bearing HepG2 xenografts. The excellent imaging properties such as good tumor uptake and high tumor contrast and specificity were achieved in the small animal models. This analytical strategy can determine novel accessible targets of HCC useful for imaging and therapy.


Carcinoma, Hepatocellular/diagnostic imaging , Fluorescent Dyes/analysis , Minichromosome Maintenance Complex Component 2/analysis , Proteomics/methods , Animals , Carcinoma, Hepatocellular/chemistry , Cell Cycle , Cell Line, Tumor , Cell Proliferation , Cyclin-Dependent Kinases , Hep G2 Cells/transplantation , Heterografts , Humans , Liver Neoplasms/chemistry , Liver Neoplasms/diagnosis , Mice , Minichromosome Maintenance Complex Component 2/metabolism , Phenylpropionates/pharmacokinetics , Thiadiazoles/pharmacokinetics
4.
Transl Res ; 170: 89-98.e3, 2016 Apr.
Article En | MEDLINE | ID: mdl-26746804

Immune deficiency or suppression in host animals is an essential precondition for the success of cancer cell xenotransplantation because the host immune system has a tendency to reject implanted cells. However, in such animals, the typical tumor microenvironment seen in cancer subjects does not form because of the lack of normal immunity. Here, we developed a novel zebrafish (Danio rerio) model based on 2 rounds of cancer cell xenotransplantation that achieved cancer-specific immunologic tolerance without immunosuppression. We irradiated human cancer cells (PC-3, K562 and HepG2) to abolish their proliferative abilities and implanted them into zebrafish larvae. These cells survived for 2 weeks in the developing host. Three months after the first implantation, the zebrafish were implanted with the same, but nonirradiated, cell lines. These cancer cells proliferated and exhibited metastasis without immune suppression. To reveal the transcriptional mechanism of this immune tolerance, we conducted dual RNA-seq of the tumor with its surrounding tissues and identified several regulatory zebrafish genes that are involved in immunity; the expression of plasminogen activator, urokinase, and forkhead box P3 was altered in response to immunologic tolerance. In conclusion, this xenograft method has potential as a platform for zebrafish-based anticancer drug discovery because it can closely mimic human clinical cancers without inducing immune suppression.


Cell Line, Tumor/transplantation , Heterografts/immunology , Transplantation, Heterologous , Zebrafish Proteins/genetics , Zebrafish/immunology , Animals , Cell Line, Tumor/pathology , Cell Line, Tumor/radiation effects , Cell Proliferation , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/immunology , Graft Rejection/immunology , Hep G2 Cells/radiation effects , Hep G2 Cells/transplantation , Humans , Immune Tolerance/genetics , K562 Cells/radiation effects , K562 Cells/transplantation , Plasminogen Activators/genetics , Plasminogen Activators/metabolism , Zebrafish/genetics , Zebrafish Proteins/immunology
5.
Tumour Biol ; 37(5): 5821-8, 2016 May.
Article En | MEDLINE | ID: mdl-26581909

We examined the expression pattern and functional roles of microRNA 15a-5p (miR-15a-5p) in human hepatocellular carcinoma (HCC). Possible miR-15a-5p aberrant expression in HCC cell lines or clinical HCC specimens was examined by quantitative real-time PCR (qRT-PCR). In HCC HepG2 and SNU-182 cells, miR-15a-5p was ectopically overexpressed by lentiviral transduction. Its effect on HCC proliferation, cancer division, and in vivo tumor growth were examined by 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay, cell cycle assay, and tumorigenicity assay, respectively. The targeting of miR-15a-5p on its downstream gene, brain-derived neurotrophic factor (BDNF), was examined by dual-luciferase assay, qRT-PCR, and Western blot, respectively. BDNF was then overexpressed in HepG2 and SNU-182 cells to evaluate its selective effect on miR-15a-5p in HCC modulation. MiR-15a-5p is aberrantly downregulated in in vitro HCC cell lines and in vivo HCC clinical specimens. Ectopic overexpression of miR-15a-5p suppressed cancer proliferation, induced cell cycle arrest in HepG2 or SNU-182 cells in vitro, and inhibited HCC tumor growth in vivo. MiR-15a-5p selectively and negatively regulated BDNF at both gene and protein levels in HCC cells. Forced overexpression of BDNF effectively reversed the tumor suppressive functions of miR-15a-5p on HCC proliferation and cell division in vitro. Our study demonstrated that miR-15a-5p is a tumor suppressor in HCC and its regulation is through BDNF in HCC.


Brain-Derived Neurotrophic Factor/physiology , Carcinoma, Hepatocellular/pathology , Liver Neoplasms/pathology , MicroRNAs/genetics , Neoplasm Proteins/physiology , RNA, Neoplasm/genetics , Animals , Brain-Derived Neurotrophic Factor/genetics , Carcinoma, Hepatocellular/genetics , Cell Division , Cell Line, Tumor , Down-Regulation , Female , Gene Expression Regulation, Neoplastic , Genes, Tumor Suppressor , Genetic Vectors , Hep G2 Cells/transplantation , Humans , Lentivirus , Liver Neoplasms/genetics , Mice , Mice, Nude , Neoplasm Proteins/genetics , Recombinant Fusion Proteins/metabolism
6.
PLoS One ; 8(8): e71374, 2013.
Article En | MEDLINE | ID: mdl-23951150

Umbilical cord matrix stem cells (UCMSC) have generated great interest in various therapeutic approaches, including liver regeneration. This article aims to analyze the specific characteristics and the potential occurrence of premalignant alterations of UCMSC during long-term expansion, which are important issues for clinical applications. UCMSC were isolated from the umbilical cord of 14 full-term newborns and expanded in vitro until senescence. We examined the long-term growth potential, senescence characteristics, immunophenotype and multilineage differentiation capacity of these cells. In addition, their genetic stability was assessed through karyotyping, telomerase maintenance mechanisms and analysis of expression and functionality of cell cycle regulation genes. The tumorigenic potential was also studied in immunocompromised mice. In vitro, UCMSC reached up to 33.7 ± 2.1 cumulative population doublings before entering replicative senescence. Their immunophenotype and differentiation potential, notably into hepatocyte-like cells, remained stable over time. Cytogenetic analyses did not reveal any chromosomal abnormality and the expression of oncogenes was not induced. Telomere maintenance mechanisms were not activated. Just as UCMSC lacked transformed features in vitro, they could not give rise to tumors in vivo. UCMSC could be expanded in long-term cultures while maintaining stable genetic features and endodermal differentiation potential. UCMSC therefore represent safe candidates for liver regenerative medicine.


Cell Lineage/genetics , Cellular Senescence/genetics , Genomic Instability , Hepatocytes/cytology , Mesenchymal Stem Cells/cytology , Umbilical Cord/cytology , Animals , Biomarkers/metabolism , Cell Cycle/genetics , Cell Differentiation , Cell Proliferation , Cells, Cultured , Hep G2 Cells/transplantation , Hepatocytes/metabolism , Hepatocytes/transplantation , Humans , Immunophenotyping , Karyotyping , Mesenchymal Stem Cells/metabolism , Mice , Telomerase/genetics , Telomerase/metabolism , Umbilical Cord/metabolism
...