Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Language
Publication year range
1.
Transl Res ; 170: 89-98.e3, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26746804

ABSTRACT

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.


Subject(s)
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
2.
Front Pharmacol ; 6: 199, 2015.
Article in English | MEDLINE | ID: mdl-26441656

ABSTRACT

Both caloric restriction (CR) and resveratrol (RSV) have beneficial effects on obesity. However, the biochemical pathways that mediate these beneficial effects might be complex and interconnected and have not been fully elucidated. To reveal the common therapeutic mechanism of CR and RSV, we performed a comparative transcriptome analysis of adipose tissues from diet-induced obese (DIO) zebrafish and obese humans. We identified nine genes in DIO zebrafish and seven genes in obese humans whose expressions were regulated by CR and RSV. Although the gene lists did not overlap except for one gene, the gene ontologies enriched in the gene lists were highly overlapped, and included genes involved in adipocyte differentiation, lipid storage and lipid metabolism. Bioinformatic analysis of cis-regulatory sequences of these genes revealed that their transcriptional regulators also overlapped, including EP300, HDAC2, CEBPB, CEBPD, FOXA1, and FOXA2. We also identified 15 and 46 genes that were dysregulated in the adipose tissue of DIO zebrafish and obese humans, respectively. Bioinformatics analysis identified EP300, HDAC2, and CEBPB as common transcriptional regulators for these genes. EP300 is a histone and lysyl acetyltransferase that modulates the function of histone and various proteins including CEBPB, CEBPD, FOXA1, and FOXA2. We demonstrated that adiposity in larval zebrafish was significantly reduced by C646, an inhibitor of EP300 that antagonizes acetyl-CoA. The reduction of adiposity by C646 was not significantly different from that induced by RSV or co-treatment of C646 and RSV. These results indicate that the inhibition of EP300 might be a common therapeutic mechanism between CR and RSV in adipose tissues of obese individuals.

3.
Nutr Metab (Lond) ; 12: 17, 2015.
Article in English | MEDLINE | ID: mdl-26052340

ABSTRACT

BACKGROUND: Diet-induced hepatic steatosis is highly associated with nonalcoholic fatty liver disease, which is related to the development of metabolic syndrome. While advanced stage nonalcoholic hepatic steatosis and steatohepatitis (NASH) result ultimately in fibrosis and cirrhosis, the molecular basis for lipid droplet formation is poorly understood. Common pathways underlie the pathology of mammalian obesity and the zebrafish diet-induced obesity model (DIO-zebrafish) used in this study. METHODS: Our analysis involved a combination of transcriptome (DNA microarray) and proteome (two-dimensional electrophoresis) methods using liver tissue from DIO-zebrafish to find candidate genes involved in hepatic steatosis. We conducted intraperitoneal injection (i.p.) of morpholino antisense oligonucleotides (MOs) for each gene into DIO-zebrafish. We also conducted in vitro overexpression in human cells. Additionally, we examined gene expression during feeding experiments involving anti-obesity compounds, creatine and anserine. RESULTS: We found that fatty acid binding protein 3 (fabp3) and E2F transcription factors were upregulated in hepatic steatosis. E2f8 MO i.p. suppressed fabp3 expression in liver, and ameliorated hepatic steatosis. In human cells (HepG2), E2F8 overexpression promoted FABP3 expression. Additionally, co-administration of creatine and anserine suppressed obesity associated phenotypes including hepatic steatosis as indicated by e2f8 and fabp3 down regulation. CONCLUSION: We discovered that the e2f8-fabp3 axis is important in the promotion of hepatic steatosis in DIO-zebrafish. The combination of transcriptome and proteome analyses using the disease model zebrafish allow identification of novel pathways involved in human diseases.

4.
Biomaterials ; 52: 14-25, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25818410

ABSTRACT

Elimination of leukemia stem cells (LSCs) is necessary for the destruction of malignant cell populations. Owing to the very small number of LSCs in leukemia cells, xenotransplantation studies are difficult in terms of functionally and pathophysiologically replicating clinical conditions of cell culture experiments. There is currently a limited number of lead compounds that target LSCs. Using the LSC-xenograft zebrafish screening method we previously developed, we found that the fluorescent compound 3,3'-dipentyloxacarbocyanine iodide (DiOC5(3)) selectively marked LSCs and suppressed their proliferation in vivo and in vitro. DiOC5(3) had no obvious toxicity to human umbilical cord blood CD34+ progenitor cells and normal zebrafish. It accumulated in mitochondria through organic anion transporter polypeptides that are overexpressed in the plasma membrane of LSCs, and induced apoptosis via ROS overproduction. DiOC5(3) also inhibited the nuclear translocation of NF-κB through the downregulation of LSC-selective pathways, as indicated from DNA microarray analysis. In summary, DiOC5(3) is a new type of anti-LSC compound available for diagnostic imaging and therapeutics that has the advantage of being a single fluorescent chemical.


Subject(s)
Apoptosis/drug effects , Carbocyanines/pharmacology , Fluorescent Dyes/pharmacology , Leukemia, Myeloid, Acute/diagnosis , Leukemia, Myeloid, Acute/drug therapy , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/pathology , Animals , Carbocyanines/pharmacokinetics , Carbocyanines/therapeutic use , Cell Line , Cell Proliferation/drug effects , Fluorescent Dyes/pharmacokinetics , Fluorescent Dyes/therapeutic use , Humans , Leukemia, Myeloid, Acute/metabolism , Leukemia, Myeloid, Acute/pathology , Mitochondria/drug effects , Mitochondria/metabolism , Mitochondria/pathology , Reactive Oxygen Species/metabolism , Zebrafish
6.
FEBS Lett ; 588(18): 3409-16, 2014 09 17.
Article in English | MEDLINE | ID: mdl-25117453

ABSTRACT

Zinc finger, MYND-type containing 8 (ZMYND8) encodes a receptor for activated C-kinase protein. Here, we report that ZMYND8 promotes angiogenesis in prostate cancer xenografts in zebrafish, as well as tube formation in human umbilical vascular endothelial cell (HUVEC) cultures. Using transcriptome analyses, we found upregulation of ZMYND8 expression in both zebrafish prostate cancer xenografts and prostate cancer samples from patients. In vitro and in vivo ZMYND8 knockdown suppressed angiogenesis, whereas ZMYND8 overexpression enhanced angiogenesis. Notably, ZMYND8 induced vegfa mRNA expression selectively in prostate cancer xenografts. Integrated analysis of human and zebrafish transcriptomes, which identified ZMYND8, might be a powerful strategy to determine also other molecular targets for inhibiting prostate cancer progression.


Subject(s)
Neovascularization, Pathologic/metabolism , Receptors, Cell Surface/physiology , Transcriptional Activation , Vascular Endothelial Growth Factor A/genetics , Animals , Cell Line, Tumor , Female , Gene Expression Regulation, Neoplastic , Gene Knockdown Techniques , Human Umbilical Vein Endothelial Cells/physiology , Humans , Male , Neoplasm Transplantation , Neovascularization, Pathologic/genetics , Receptors for Activated C Kinase , Transcriptome , Tumor Suppressor Proteins , Vascular Endothelial Growth Factor A/metabolism , Zebrafish
SELECTION OF CITATIONS
SEARCH DETAIL
...