Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 31
Filter
1.
J Cell Mol Med ; 28(7): e18212, 2024 04.
Article in English | MEDLINE | ID: mdl-38516826

ABSTRACT

SBFI26, an inhibitor of FABP5, has been shown to suppress the proliferation and metastasis of tumour cells. However, the underlying mechanism by which SBFI26 induces ferroptosis in breast cancer cells remains largely unknown. Three breast cancer cell lines were treated with SBFI26 and CCK-8 assessed cytotoxicity. Transcriptome was performed on the Illumina platform and verified by qPCR. Western blot evaluated protein levels. Malondialdehyde (MDA), total superoxide dismutase (T-SOD), Fe, glutathione (GSH) and oxidized glutathione (GSSG) were measured. SBFI26 induced cell death time- and dose-dependent, with a more significant inhibitory effect on MDA-MB-231 cells. Fer-1, GSH and Vitamin C attenuated the effects but not erastin. RNA-Seq analysis revealed that SBFI26 treatment significantly enriched differentially expressed genes related to ferroptosis. Furthermore, SBFI26 increased intracellular MDA, iron ion, and GSSG levels while decreasing T-SOD, total glutathione (T-GSH), and GSH levels.SBFI26 dose-dependently up-regulates the expression of HMOX1 and ALOX12 at both gene and protein levels, promoting ferroptosis. Similarly, it significantly increases the expression of SAT1, ALOX5, ALOX15, ALOXE3 and CHAC1 that, promoting ferroptosis while downregulating the NFE2L2 gene and protein that inhibit ferroptosis. SBFI26 leads to cellular accumulation of fatty acids, which triggers excess ferrous ions and subsequent lipid peroxidation for inducing ferroptosis.


Subject(s)
Ferroptosis , Triple Negative Breast Neoplasms , Humans , Triple Negative Breast Neoplasms/drug therapy , Triple Negative Breast Neoplasms/genetics , Glutathione Disulfide , Ferroptosis/genetics , Lipid Peroxidation , Glutathione , Iron , Superoxide Dismutase/genetics , Reactive Oxygen Species , Fatty Acid-Binding Proteins
2.
Am J Cancer Res ; 14(1): 300-323, 2024.
Article in English | MEDLINE | ID: mdl-38323289

ABSTRACT

Enzalutamide is a drug used to treat prostate cancer (PC) and docetaxel is a drug for chemotherapeutic treatment of diverse cancer types, including PC. The effectiveness of these drugs in treating castration-resistant prostate cancer (CRPC) is poor and therefore CRPC is still largely incurable. However, the bio-inhibitor of fatty acid-binding protein 5 (FABP5), dmrFABP5, which is a mutant form of FABP5 incapable of binding to fatty acids, has been shown recently to be able to suppress the tumorigenicity and metastasis of cultured CRPC cells. The present study investigated the possible synergistic effect of dmrFABP5 combined with either enzalutamide or docetaxel on suppressing the tumorigenic properties of PC cells, including cell viability, migration, invasion and colony proliferation in soft agar. A highly significant synergistic inhibitory effect on these properties was observed when dmrFABP5 was used in combination with enzalutamide on androgen-responsive PC 22RV1 cells. Moreover, a highly significant synergistic inhibitory effect was also observed when dmrFABP5 was combined with docetaxel, and added to 22RV1 cells and to the highly malignant, androgen-receptor (AR)-negative Du145 cells. DmrFABP5 alone failed to produce any suppressive effect when added to the FABP5-negative cell line LNCaP, although enzalutamide could significantly suppress LNCaP cells when used as a single agent. These synergistic inhibitory effects of dmrFABP5 were produced by interrupting the FABP5-related signal transduction pathway in PC cells. Thus, dmrFABP5 appears to be not only a potential single therapeutic agent, but it may also be used in combination with existing drugs to suppress both AR-positive and AR-negative PC.

3.
Int J Oncol ; 64(2)2024 Feb.
Article in English | MEDLINE | ID: mdl-38131188

ABSTRACT

Fatty acid­binding protein 5 (FABP5) and androgen receptor (AR) are critical promoters of prostate cancer. In the present study, the effects of knocking out the FABP5 or AR genes on malignant characteristics of prostate cancer cells were investigated, and changes in the expression of certain key proteins in the FABP5 (or AR)­peroxisome proliferator activated receptor­Î³ (PPARγ)­vascular endothelial growth factor (VEGF) signaling pathway were monitored. The results obtained showed that FABP5­ or AR­knockout (KO) led to a marked suppression of the malignant characteristics of the cells, in part, through disrupting this signaling pathway. Moreover, FABP5 and AR are able to interact with each other to regulate this pathway, with FABP5 controlling the dominant AR splicing variant 7 (ARV7), and AR, in return, regulates the expression of FABP5. Comparisons of the RNA profiles revealed the existence of numerous differentially expressed genes (DEGs) comparing between the parental and the FABP5­ or AR­KO cells. The six most abundant changes in DEGs were found to be attributable to the transition from androgen­responsive to androgen­unresponsive, castration­resistant prostate cancer (CRPC) cells. These findings have provided novel insights into the complex molecular pathogenesis of CRPC cells, and have demonstrated that interactions between FABP5 and AR contribute to the transition of prostate cancer cells to an androgen­independent state. Moreover, gene enrichment analysis revealed that the most highly enriched biological processes associated with the DEGs included those responsive to fatty acids, cholesterol and sterol biosynthesis, as well as to lipid and fatty acid transportation. Since these pathways regulated by FABP5 or AR may be crucial in terms of transducing signals for cancer cell progression, targeting FABP5, AR and their associated pathways, rather than AR alone, may provide a new avenue for the development of therapeutic strategies geared towards suppressing the malignant progression to CRPC cells.


Subject(s)
Prostatic Neoplasms, Castration-Resistant , Receptors, Androgen , Male , Humans , Receptors, Androgen/genetics , Receptors, Androgen/metabolism , Prostatic Neoplasms, Castration-Resistant/drug therapy , Androgens , Vascular Endothelial Growth Factor A/metabolism , Cell Line, Tumor , Gene Expression Regulation, Neoplastic , Fatty Acid-Binding Proteins/genetics , Fatty Acid-Binding Proteins/metabolism
4.
J Gastric Cancer ; 23(2): 340-354, 2023 Apr.
Article in English | MEDLINE | ID: mdl-37129157

ABSTRACT

PURPOSE: Gastric cancer (GC) is the second most lethal cancer globally and is associated with poor prognosis. Fatty acid-binding proteins (FABPs) can regulate biological properties of carcinoma cells. FABP5 is overexpressed in many types of cancers; however, the role and mechanisms of action of FABP5 in GC remain unclear. In this study, we aimed to evaluate the clinical and biological functions of FABP5 in GC. MATERIALS AND METHODS: We assessed FABP5 expression using immunohistochemical analysis in 79 patients with GC and evaluated its biological functions following in vitro and in vivo ectopic expression. FABP5 targets relevant to GC progression were determined using RNA sequencing (RNA-seq). RESULTS: Elevated FABP5 expression was closely associated with poor outcomes, and ectopic expression of FABP5 promoted proliferation, invasion, migration, and carcinogenicity of GC cells, thus suggesting its potential tumor-promoting role in GC. Additionally, RNA-seq analysis indicated that FABP5 activates immune-related pathways, including cytokine-cytokine receptor interaction pathways, interleukin-17 signaling, and tumor necrosis factor signaling, suggesting an important rationale for the possible development of therapies that combine FABP5-targeted drugs with immunotherapeutics. CONCLUSIONS: These findings highlight the biological mechanisms and clinical implications of FABP5 in GC and suggest its potential as an adverse prognostic factor and/or therapeutic target.

5.
Cell Biol Int ; 47(5): 929-942, 2023 May.
Article in English | MEDLINE | ID: mdl-36651331

ABSTRACT

Previous work showed that FABP5 inhibitors suppressed the malignant progression of prostate cancer cells, and this suppression might be achieved partially by promoting apoptosis. But the mechanisms involved were not known. Here, we investigated the effect of inhibitors on apoptosis and studied the relevant mechanisms. WtrFABP5 significantly reduced apoptotic cells in 22Rv1 and PC3 by 18% and 42%, respectively. In contrast, the chemical inhibitor SB-FI-26 produced significant increases in percentages of apoptotic cells in 22Rv1 and PC3 by 18.8% (±4.1) and 4.6% (±1.1), respectively. The bio- inhibitor dmrFABP5 also did so by 23.1% (±2.4) and 15.8% (±3.0), respectively, in these cell lines. Both FABP5 inhibitors significantly reduced the levels of the phosphorylated nuclear fatty acid receptor PPARγ, indicating that these inhibitors promoted apoptosis-induction sensitivity of the cancer cells by suppressing the biological activity of PPARγ. Thus, the phosphorylated PPARγ levels were reduced by FABP5 inhibitors, the levels of the phosphorylated AKT and activated nuclear factor kapper B (NFκB) were coordinately altered by additions of the inhibitors. These changes eventually led to the increased levels of cleaved caspase-9 and cleaved caspase-3; and thus, increase in the percentage of cells undergoing apoptosis. In untreated prostate cancer cells, increased FABP5 suppressed the apoptosis by increasing the biological activity of PPARγ, which, in turn, led to a reduced apoptosis by interfering with the AKT or NFκB signaling pathway. Our results suggested that the FABP5 inhibitors enhanced the apoptosis-induction of prostate cancer cells by reversing the biological effect of FABP5 and its related pathway.


Subject(s)
Prostatic Neoplasms , Proto-Oncogene Proteins c-akt , Male , Humans , Proto-Oncogene Proteins c-akt/metabolism , PPAR gamma/metabolism , Cell Line, Tumor , Apoptosis , Fatty Acid-Binding Proteins/metabolism
6.
J Interferon Cytokine Res ; 42(11): 592-593, 2022 11.
Article in English | MEDLINE | ID: mdl-36112111

ABSTRACT

Recently, messenger ribonucleic acid (mRNA) vaccine research and development became a hotspot in the field of prevention and treatment of Corona Virus Disease 2019 (COVID-19) and some other disorders. mRNA vaccine shows many advantages over other vaccines, including cost-effectiveness, safety, and rapid optimization of antigen-specific sequences and shorter development cycle. Cancer progression is significantly associated with immune response, and mRNA vaccine also shows obvious advantages for cancer immunotherapy. In this study, we briefly summarize the recent advances and discuss the perspectives on tumor mRNA vaccine development; particularly, these findings pave an avenue for effective cancer prevention and treatment.


Subject(s)
COVID-19 , Cancer Vaccines , Neoplasms , Humans , COVID-19/prevention & control , Cancer Vaccines/genetics , Neoplasms/genetics , Neoplasms/prevention & control , RNA, Messenger/genetics , mRNA Vaccines
8.
Genes Cancer ; 10(3-4): 80-96, 2019 May.
Article in English | MEDLINE | ID: mdl-31258834

ABSTRACT

Previous study has suggested that the FABP5-PPARγ-signalling transduction pathway gradually replaces the androgen receptor activated pathway in promoting malignant progression of castration-resistant prostate cancer (CRPC) cells. To interfere with this newly discovered FABP5-related signalling pathway, we have produced a highly efficient recombinant FABP5 inhibitor, named dmrFABP5. Treatment with dmrFABP5 significantly supressed the proliferation, migration, invasion and colony formation of the highly malignant prostate cancer cells PC3-M in vitro. To test dmrFABP5's suppressive effect in CRPC, the human PC3-M cells were implanted orthotopically into the prostate gland of immunosuppressed mice to produce tumours. These mice were then treated with dmrFABP5 and produced a highly significant reduction of 100% in metastatic rate and a highly significant reduction of 13-fold in the average size of primary tumours. Immunocytochemial staining showed that the staining intensity of dmrFABP5 treated tumours was reduced by 67%. When tested in vitro, dmrFABP5 suppressed the cancer cells by blocking fatty acid stimulation of PPARγ, and thereby prevented it activating down-stream cancer-promoting or inhibiting cancer-suppressing genes. Our results show that the FABP5 inhibitor dmrFABP5 is a novel molecule for treatment of experimental CRPC and its inhibitory effect is much greater than that produced by SB-FI-26 reported in our previous work.

9.
Precis Clin Med ; 2(3): 192-196, 2019 Oct.
Article in English | MEDLINE | ID: mdl-35694437

ABSTRACT

In this short communication, a novel fatty acid-binding protein 5 (FABP5)-related signal transduction pathway in prostate cancer is reviewed. In castration-resistant prostate cancer (CRPC) cells, the FABP5-related signal transduction pathway plays an important role during transformation of the cancer cells from androgen-dependent state to androgen-independent state. The detailed route of this signal transduction pathway can be described as follows: when FABP5 expression is increased as the increasing malignancy, excessive amounts of fatty acids from intra- and extra-cellular sources are transported into the nucleus of the cancer cells where they act as signalling molecules to stimulate their nuclear receptor peroxisome proliferator-activated receptor gamma (PPARγ). The phosphorylated or biologically activated PPARγ then modulates the expression of its downstream target regulatory genes to trigger a series of molecular events that eventually lead to enhanced tumour expansion and aggressiveness caused by an overgrowth of the cancer cells with a reduced apoptosis and an increased angiogenesis. Suppressing the FABP5-related pathway via RNA interference against FABP5 has produced a 63-fold reduction in the average size of the tumours developed from CRPC cells in nude mice, a seven-fold reduction of tumour incidence, and a 100% reduction of metastasis rate. Experimental treatments of CRPC with novel FABP5 inhibitors have successfully inhibited the malignant progression of CRPC cells both in vitro and in nude mouse. These studies suggest that FABP5-related signal transduction pathway is a novel target for therapeutic intervention of CRPC cells.

10.
Oncotarget ; 8(19): 31041-31056, 2017 May 09.
Article in English | MEDLINE | ID: mdl-28415688

ABSTRACT

Castration resistant-prostate cancer is largely impervious to feather hormonal therapy and hence the outlook for patients is grim. Here we use an approach to attach the recently discovered Achilles heel. The experimental treatment established in this study is based on the recent discovery that it is the FABP5-PPARγ-VEGF signalling axis, rather than the androgen receptor pathway, played a dominant role in promoting the malignant progression of castration resistant prostate cancer cells. Treatments have been established in mice by suppressing the biological activity of FABP5 using a chemical inhibitor SBFI26. The inhibitor significantly suppressed the proliferation, migration, invasiveness and colony formation of PC3-M cells in vitro. It also produced a highly significant suppression of both the metastases and the primary tumours developed from cancer cells implanted orthotopically into the prostate glands of the mice. The inhibitor SBFI26 interferes with the FABP5-PPARγ- signalling pathway at the initial stage of the signal transduction by binding competitively to FABP5 to inhibit cellular fatty acid uptake. This avoids the fatty-acid stimulation of PPARγ and prevents it activating the down-stream regulated cancer-promoting genes. This entirely novel experimental approach to treating castration- resistant prostate cancer is completely different from current treatments that are based on androgen-blockade therapy.


Subject(s)
Antineoplastic Agents/pharmacology , Cyclobutanes/pharmacology , Dicarboxylic Acids/pharmacology , Fatty Acid-Binding Proteins/antagonists & inhibitors , Fatty Acid-Binding Proteins/metabolism , Prostatic Neoplasms, Castration-Resistant/metabolism , Prostatic Neoplasms, Castration-Resistant/pathology , Animals , Binding, Competitive , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Cell Survival/drug effects , Disease Models, Animal , Disease Progression , Drug Discovery , Drug Screening Assays, Antitumor , Fatty Acids/metabolism , Humans , Ligands , Male , Mice , Neoplasm Metastasis , PPAR gamma/agonists , Prostatic Neoplasms, Castration-Resistant/drug therapy , Protein Binding , Xenograft Model Antitumor Assays
11.
Oncotarget ; 7(50): 82783-82797, 2016 Dec 13.
Article in English | MEDLINE | ID: mdl-27779102

ABSTRACT

In contrast to numerous studies conducted to investigate the crucial role of fatty acid binding protein 5 (FABP5) in prostate cancer, investigations on the possible involvement of other FABPs are rare. Here we first measured the mRNA levels of 10 FABPs in benign and malignant prostate cell lines and identified the differentially expressed FABP6 and FABP9 mRNAs whose levels in all malignant cell lines were higher than those in the benign cells. Thereafter we assessed the expression status of FABP6 and FABP9 in both prostate cell lines and in human tissues. FABP6 protein was overexpressed only in 1 of the 5 malignant cell lines and its immunostaining intensities were not significantly different between benign and malignant prostate tissues. In contrast, FABP9 protein was highly expressed in highly malignant cell lines PC-3 and PC3-M, but its level in the benign PNT-2 and other malignant cell lines was not detectable. When analysed in an archival set of human prostate tissues, immunohistochemical staining intensity for FABP9 was significantly higher in carcinomas than in benign cases and the increase in FABP9 was significantly correlated with reduced patient survival times. Moreover, the increased level of staining for FABP9 was significantly associated with the increased joint Gleason scores (GS) and androgen receptor index (AR). Suppression of FABP9 expression in highly malignant PC3-M cells inhibited their invasive potential. Our results suggest that FABP9 is a valuable prognostic marker to predict the outcomes of prostate cancer patients, perhaps by playing an important role in prostate cancer cell invasion.


Subject(s)
Biomarkers, Tumor/metabolism , Fatty Acid-Binding Proteins/metabolism , Prostatic Neoplasms/metabolism , Biomarkers, Tumor/genetics , Cell Line, Tumor , Cell Movement , Fatty Acid-Binding Proteins/genetics , Gastrointestinal Hormones/genetics , Gastrointestinal Hormones/metabolism , Humans , Immunohistochemistry , Kaplan-Meier Estimate , Male , Neoplasm Grading , Neoplasm Invasiveness , Predictive Value of Tests , Prostatic Neoplasms/genetics , Prostatic Neoplasms/pathology , Prostatic Neoplasms/therapy , RNA Interference , RNA, Messenger/genetics , Receptors, Androgen/metabolism , Time Factors , Up-Regulation
12.
FEBS Open Bio ; 6(3): 190-9, 2016 03.
Article in English | MEDLINE | ID: mdl-27047747

ABSTRACT

Fatty acid-binding proteins (FABPs) are responsible for binding and storing hydrophobic ligands such as long-chain fatty acids, and for transporting these ligands to the appropriate compartments within the cell. The present study demonstrates that the FABP5 gene is upregulated in colorectal cancer cells compared to normal colon cells in a manner that correlates with disease stage and that FABP5 significantly promotes colorectal cancer cell growth and metastatic potential. Thus, FABP5 might be a promising prognostic or therapeutic biomarker candidate in human colorectal cancer.

13.
Oncotarget ; 7(8): 9322-39, 2016 Feb 23.
Article in English | MEDLINE | ID: mdl-26814431

ABSTRACT

In previous work, it is suggested that the excessive amount of fatty acids transported by FABP5 may facilitate the malignant progression of prostate cancer cells through a FABP5-PPARγ-VEGF signal transduction axis to increase angiogenesis. To further functionally characterise the FABP5-PPARγ-VEGF signal transduction pathway, we have, in this work, investigated the molecular mechanisms involved in its tumorigenicity promoting role in prostate cancer. Suppression of PPARγ in highly malignant prostate cancer cells produced a significant reduction (up to 53%) in their proliferation rate, invasiveness (up to 89%) and anchorage-independent growth (up to 94%) in vitro. Knockdown of PPARγ gene in PC3-M cells by siRNA significantly reduced the average size of tumours formed in nude mice by 99% and tumour incidence by 90%, and significantly prolonged the latent period by 3.5 fold. Results in this study combined with some previous results suggested that FABP5 promoted VEGF expression and angiogenesis through PPARγ which was activated by fatty acids transported by FABP5. Further investigations showed that PPARγ up-regulated VEGF expression through acting with the PPAR-responsive elements in the promoter region of VEGF gene in prostate cancer cells. Although androgen can modulate VEGF expression through Sp1/Sp3 binding site on VEGF promoter in androgen-dependent prostate cancer cells, this route, disappeared as the cells gradually lost their androgen dependency; was replaced by the FABP5-PPARγ-VEGF signalling pathway. These results suggested that the FABP5-PPARγ-VEGF signal transduction axis, rather than androgen modulated route, may be a more important novel therapeutic target for angiogenesis-suppression treatment of castration resistant prostate cancer.


Subject(s)
Fatty Acid-Binding Proteins/metabolism , Fatty Acids/metabolism , PPAR gamma/metabolism , Promoter Regions, Genetic/genetics , Prostatic Neoplasms, Castration-Resistant/pathology , Vascular Endothelial Growth Factor A/biosynthesis , Animals , Binding Sites/genetics , Cell Line, Tumor , Cell Proliferation/genetics , Cell Transformation, Neoplastic , Gene Expression Regulation, Neoplastic/drug effects , Human Umbilical Vein Endothelial Cells , Humans , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Neoplasm Invasiveness/genetics , Neovascularization, Pathologic , PPAR gamma/antagonists & inhibitors , PPAR gamma/genetics , Prostate/pathology , Prostatic Neoplasms, Castration-Resistant/therapy , RNA Interference , RNA, Small Interfering/genetics , Signal Transduction , Vascular Endothelial Growth Factor A/genetics
14.
Biochem J ; 473(4): 449-61, 2016 Feb 15.
Article in English | MEDLINE | ID: mdl-26614767

ABSTRACT

FABPs (fatty-acid-binding proteins) are a family of low-molecular-mass intracellular lipid-binding proteins consisting of ten isoforms. FABPs are involved in binding and storing hydrophobic ligands such as long-chain fatty acids, as well as transporting these ligands to the appropriate compartments in the cell. FABP5 is overexpressed in multiple types of tumours. Furthermore, up-regulation of FABP5 is strongly associated with poor survival in triple-negative breast cancer. However, the mechanisms underlying the specific up-regulation of the FABP5 gene in these cancers remain poorly characterized. In the present study, we determined that FABP5 has a typical CpG island around its promoter region. The DNA methylation status of the CpG island in the FABP5 promoter of benign prostate cells (PNT2), prostate cancer cells (PC-3, DU-145, 22Rv1 and LNCaP) and human normal or tumour tissue was assessed by bisulfite sequencing analysis, and then confirmed by COBRA (combined bisulfite restriction analysis) and qAMP (quantitative analysis of DNA methylation using real-time PCR). These results demonstrated that overexpression of FABP5 in prostate cancer cells can be attributed to hypomethylation of the CpG island in its promoter region, along with up-regulation of the direct trans-acting factors Sp1 (specificity protein 1) and c-Myc. Together, these mechanisms result in the transcriptional activation of FABP5 expression during human prostate carcinogenesis. Importantly, silencing of Sp1, c-Myc or FABP5 expression led to a significant decrease in cell proliferation, indicating that up-regulation of FABP5 expression by Sp1 and c-Myc is critical for the proliferation of prostate cancer cells.


Subject(s)
Epigenesis, Genetic , Fatty Acid-Binding Proteins/genetics , Gene Expression Regulation, Neoplastic , Prostatic Neoplasms/genetics , Cell Line, Tumor , DNA Methylation , Humans , Male , Middle Aged , Promoter Regions, Genetic , Proto-Oncogene Proteins c-myc/metabolism , Sp1 Transcription Factor/metabolism
15.
Genes Cancer ; 5(5-6): 212-25, 2014 May.
Article in English | MEDLINE | ID: mdl-25061504

ABSTRACT

Constant deregulation of Id1 and Id3 has been implicated in a wide range of carcinomas. However, underlying molecular evidence for the joint role of Id1 and Id3 in the tumorigenicity of small cell lung cancer (SCLC) is sparse. Investigating the biological significance of elevated expression in SCLC cells, we found that Id1 and Id3 co-suppression resulted in significant reduction of proliferation rate, invasiveness and anchorage-independent growth. Suppressing both Id1 and Id3 expression also greatly reduced the average size of tumors produced by transfectant cells when inoculated subcutaneously into nude mice. Further investigation revealed that suppressed expression of Id1 and Id3 was accompanied by decreased angiogenesis and increased apoptosis. Therefore, the SCLC tumorigenicity suppression effect of double knockdown of Id1 and Id3 may be regulated through pathways of apoptosis and angiogenesis.

16.
Int J Oncol ; 44(1): 265-75, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24189640

ABSTRACT

The purpose of this study was to test the hypothesis that cooperative interaction between cutaneous fatty acid-binding protein (C-FABP) and peroxisome proliferator-activated receptors (PPAR) promotes the malignant progression of human prostate cancer. The expression of C-FABP, PPARß/δ and PPARγ was measured by western blot analysis in prostate cell lines and by immunohistochemical staining in tissue sections of benign prostatic hyperplasia (BPH) and prostatic carcinomas. The correlation between the expression of PPARs and C-FABP was assessed. The significance of increased expression of these proteins was analysed with respect to prognosis and compared with those of alternative biomarkers. The expression levels of C-FABP and PPARγ in prostate cancer cell lines and the cytoplasm and nuclei of carcinoma tissues were significantly (Student's t-test, p<0.05) higher compared to those in benign cell lines and BPH tissues. The raised expression level of C-FABP and PPARγ was significantly correlated with the increased combined Gleason scores (GS) of the carcinomas. Enhanced expression of cytoplasmic C-FABP significantly correlated with increased nuclear PPARγ (Student's t-test, p<0.005). While expression of PPARß/δ in carcinomas did not correlate with patient outcome, the increased levels of both C-FABP and PPARγ were associated with shorter patient survival. Multivariate analysis indicated that C-FABP was independently associated with patient survival, whereas PPARγ was confounded by C-FABP in predicting patient survival. Thus, the increased C-FABP may interact with PPARγ in a coordinated mechanism to facilitate malignant progression in prostatic cancer. Both C-FABP and PPARγ are suitable as prognostic factors to predict the clinical outcome of prostatic cancer patients.


Subject(s)
Fatty Acid-Binding Proteins/biosynthesis , Gene Expression Regulation, Neoplastic/genetics , PPAR gamma/biosynthesis , Prostatic Neoplasms/genetics , Aged , Fatty Acid-Binding Proteins/genetics , Humans , Kaplan-Meier Estimate , Male , Neoplasm Grading , PPAR gamma/genetics , Prognosis , Prostatic Neoplasms/pathology
17.
Genes Cancer ; 4(7-8): 297-314, 2013 Jul.
Article in English | MEDLINE | ID: mdl-24167657

ABSTRACT

Cutaneous fatty acid-binding protein (C-FABP), a cancer promoter and metastasis inducer, is overexpressed in the majority of prostatic carcinomas. Investigation of molecular mechanisms involved in tumor-promoting activity of C-FABP has established that there is a fatty acid-initiated signaling pathway leading to malignant progression of prostatic cancer cells. Increased C-FABP expression plays an important role in this novel signaling pathway. Thus, when C-FABP expression is increased, excessive amounts of fatty acids are transported into the nucleus where they act as signaling molecules to stimulate their nuclear receptor peroxisome proliferator-activated receptor gamma (PPARγ). The activated PPARγ then modulates the expression of its downstream target regulatory genes, which eventually lead to enhanced tumor expansion and aggressiveness caused by an overgrowth of cells with reduced apoptosis and an increased angiogenesis.

18.
PLoS One ; 6(7): e22672, 2011.
Article in English | MEDLINE | ID: mdl-21799931

ABSTRACT

We provide novel functional data that posttranscriptional silencing of gene RPL19 using RNAi not only abrogates the malignant phenotype of PC-3M prostate cancer cells but is selective with respect to transcription and translation of other genes. Reducing RPL19 transcription modulates a subset of genes, evidenced by gene expression array analysis and Western blotting, but does not compromise cell proliferation or apoptosis in-vitro. However, growth of xenografted tumors containing the knocked-down RPL19 in-vivo is significantly reduced. Analysis of the modulated genes reveals induction of the non-malignant phenotype principally to involve perturbation of networks of transcription factors and cellular adhesion genes. The data provide evidence that extra-ribosomal regulatory functions of RPL19, beyond protein synthesis, are critical regulators of cellular phenotype. Targeting key members of affected networks identified by gene expression analysis raises the possibility of therapeutically stabilizing a benign phenotype generated by modulating the expression of an individual gene and thereafter constraining a malignant phenotype while leaving non-malignant tissues unaffected.


Subject(s)
Gene Knockdown Techniques , Phenotype , Prostatic Neoplasms/genetics , Prostatic Neoplasms/pathology , RNA, Small Interfering/genetics , Ribosomal Proteins/deficiency , Ribosomal Proteins/genetics , Base Sequence , Cell Line, Tumor , Cell Proliferation , Gene Expression Profiling , Humans , Male , Molecular Targeted Therapy , Prostatic Neoplasms/therapy , RNA Interference , Ribosomal Proteins/metabolism , Transfection
19.
Int J Oncol ; 38(4): 1083-91, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21290088

ABSTRACT

Expression of osteopontin (OPN) is increased in prostate cancer cells. The possibility of utilising the increased OPN as a target to suppress the tumourigenicity was investigated in this study. Small interference RNAs against OPN were transfected into highly malignant DU145 prostate cancer cells, which express high level of OPN prior to the transfections, to establish OPN-suppressed clones. Compared with the control transfectants generated by scrambled RNA, suppressed expression of OPN significantly inhibited cell invasiveness and anchorage-independent growth. Similar results were obtained from in vivo experiments. OPN-suppressed transfectants produced significant reductions in average sizes of subcutaneous tumours after inoculation into nude mice. When the levels of OPN measured in transfectants before injection were related to tumour sizes, the reduction in tumour sizes was not propotionally related to the inhibition in OPN-levels. However, when the levels of OPN were analysed in the tumour tissues, it was found that the reduced OPN expression levels were significantly associated with the reducing tumour sizes. These results showed that changes in OPN levels had occurred after the transfectants were inoculated in mice. This study suggested while OPN can be an effective target for therapeutic suppression of prostate cancer, more effective way than RNAi is needed to inhibit OPN expression.


Subject(s)
Osteopontin/antagonists & inhibitors , Prostatic Neoplasms/pathology , RNA Interference , Animals , Cell Adhesion , Cell Line, Tumor , Cell Movement , Cell Proliferation , Humans , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Neoplasm Invasiveness , Osteopontin/genetics , Osteopontin/metabolism , Prostatic Neoplasms/genetics , RNA, Small Interfering/genetics , Tumor Burden , Xenograft Model Antitumor Assays
20.
Int J Oncol ; 37(3): 595-603, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20664928

ABSTRACT

Id3 is over-expressed in small cell lung cancer (SCLC). To test whether the tumourigenicity of SCLC cells can be inhibited by suppressing Id3 expression, we transfected siRNA into SCLC cell line GLC-19 and established two sublines (G-Id3-1 and G-Id3-7) which expressed only 30% of the level of Id3 measured in control transfectants. Suppression of Id3 expression in both G-Id3-1 and G-Id3-7 cells produced significant reductions in proliferation rates and in numbers of colonies formed in soft agar assay. When G-Id3-1, G-Id3-7 and the control transfectants were inoculated subcutaneously into 3 groups (8 each) of nude mice, respectively, all (100%) inoculated animals produced tumours. Although there was no difference in tumour incidents amongst the 3 groups, significant reductions were observed in both size and weight of tumours produced by either G-Id3-1 or G-Id3-7 cells. While the final average volume of tumours produced in control group was 1012.1+/-394 mm(3), it was significantly reduced (p<0.001, p<0.01) by 2.1- and 2.9-fold to 475.7+/-167 mm(3) and 354.3+/-218 mm(3) in groups inoculated with G-Id3-1 and G-Id3-7 cells, respectively. Similar differences were also observed in average weight of tumours. Upon induction of apoptosis by cytotoxin camptothecin, the percentages of apoptotic cells in G-Id3-1 and G-Id3-7 were, respectively >2.4-fold higher than that in control. The results in this study suggest that highly expressed Id3 in SCLC cells may be an important therapeutic target for tumour suppression.


Subject(s)
Inhibitor of Differentiation Proteins/biosynthesis , Lung Neoplasms/pathology , Neoplasm Proteins/biosynthesis , Small Cell Lung Carcinoma/pathology , Animals , Base Sequence , Camptothecin/pharmacology , Cell Growth Processes/genetics , Cell Line, Tumor , Gene Expression Regulation, Neoplastic , Humans , Inhibitor of Differentiation Proteins/antagonists & inhibitors , Inhibitor of Differentiation Proteins/genetics , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Lung Neoplasms/therapy , Mice , Mice, Nude , Molecular Sequence Data , Neoplasm Proteins/antagonists & inhibitors , Neoplasm Proteins/genetics , RNA, Small Interfering/administration & dosage , RNA, Small Interfering/genetics , Small Cell Lung Carcinoma/genetics , Small Cell Lung Carcinoma/metabolism , Small Cell Lung Carcinoma/therapy , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL
...