Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 32
Filter
1.
J Cell Mol Med ; 24(18): 10525-10541, 2020 09.
Article in English | MEDLINE | ID: mdl-32798252

ABSTRACT

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.


Subject(s)
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
2.
Sci Rep ; 10(1): 2735, 2020 02 17.
Article in English | MEDLINE | ID: mdl-32066763

ABSTRACT

Resistance to cancer therapy is a challenge because of innate tumor heterogeneity and constant tumor evolution. Since the pathway of resistance cannot be predicted, combination therapies may address this progression. We discovered that in addition to IGF1 and IGF2, IGFBP-3 binds bFGF, HGF, neuregulin, and PDGF AB with nanomolar affinity. Because growth factors drive resistance, simultaneous inhibition of multiple growth factor pathways may improve the efficacy of precision therapy. Growth factor sequestration by IGFBP-3-Fc enhances the activity of EGFR inhibitors by decreasing cell survival and inhibiting bFGF, HGF, and IGF1 growth factor rescue and also potentiates the activity of other cancer drugs. Inhibition of tumor growth in vivo with adjuvant IGFBP-3-Fc with erlotinib versus erlotinib after treatment cessation supports that the combination reduces cell survival. Inhibition of multiple growth factor pathways may postpone resistance and extend progression-free survival in many cancer indications.


Subject(s)
ErbB Receptors/genetics , Erlotinib Hydrochloride/pharmacology , Gene Expression Regulation, Neoplastic , Insulin-Like Growth Factor Binding Protein 3/genetics , Lung Neoplasms/drug therapy , Protein Kinase Inhibitors/pharmacology , Tumor Burden/drug effects , Animals , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Drug Resistance, Neoplasm/drug effects , Drug Resistance, Neoplasm/genetics , ErbB Receptors/antagonists & inhibitors , ErbB Receptors/metabolism , Fibroblast Growth Factors/genetics , Fibroblast Growth Factors/metabolism , HEK293 Cells , HT29 Cells , Hepatocyte Growth Factor/genetics , Hepatocyte Growth Factor/metabolism , Humans , Immunoglobulin Fc Fragments/pharmacology , Insulin-Like Growth Factor Binding Protein 3/antagonists & inhibitors , Insulin-Like Growth Factor Binding Protein 3/metabolism , Insulin-Like Growth Factor I/genetics , Insulin-Like Growth Factor I/metabolism , Insulin-Like Growth Factor II/genetics , Insulin-Like Growth Factor II/metabolism , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , MCF-7 Cells , Male , Mice , Mice, Inbred NOD , Neuregulin-1/genetics , Neuregulin-1/metabolism , Platelet-Derived Growth Factor/genetics , Platelet-Derived Growth Factor/metabolism , Signal Transduction , Xenograft Model Antitumor Assays
3.
J Neurosurg ; 132(1): 168-179, 2019 01 11.
Article in English | MEDLINE | ID: mdl-30641835

ABSTRACT

OBJECTIVE: Despite intensive medical treatment, patients with glioblastoma (grade IV glioma [GBM]) have a low 5-year survival rate of 5.5%. In this study, the authors tried to improve currently used therapies by identification of a therapeutic target, IGFBP3, for glioma treatment. METHODS: IGFBP3 RNA expression in 135 patients newly diagnosed with glioma was correlated with clinicopathological factors. Immunohistochemical analysis was performed to determine IGFBP3 protein expression in glioma specimens. The effect of IGFBP3 depletion on cell proliferation was examined using IGFBP3 knockdown glioma cells. Intracranial infusion of IGFBP3 siRNAs was performed to evaluate the effect of IGFBP3 depletion in mouse intracranial xenograft models. RESULTS: We demonstrated higher IGFBP3 expression in GBM than in tumor margin and grade II glioma. IGFBP3 expression was not only positively correlated with tumor grades but also associated with tumor histology and IDH1/2 mutation status. Additionally, higher IGFBP3 expression predicted shorter overall survival in glioma and GBM proneural subgroup patients. In vitro cell culture studies suggested IGFBP3 knockdown suppressed cell proliferation and induced cell cycle G2/M arrest as well as apoptosis in glioma cells. Also, accumulation of DNA double-strand breaks and γH2AX was observed in IGFBP3 knockdown cells. IGFBP3 knockdown delayed in vivo tumor growth in mouse subcutaneous xenograft models. Furthermore, convection-enhanced delivery of IGFBP3 siRNA to mouse brain suppressed intracranial tumor growth and prolonged survival of tumor-bearing mice. CONCLUSIONS: Our findings suggest IGFBP3 predicts poor outcome of glioma patients and is a potential therapeutic target for which depletion of its expression suppresses tumor growth through inducing apoptosis and accumulation of DNA damage in glioma cells.


Subject(s)
Brain Neoplasms/therapy , Glioma/therapy , Insulin-Like Growth Factor Binding Protein 3/antagonists & inhibitors , Molecular Targeted Therapy , Neoplasm Proteins/antagonists & inhibitors , RNA Interference , RNA, Small Interfering/therapeutic use , Animals , Apoptosis , Brain Neoplasms/chemistry , Brain Neoplasms/genetics , Brain Neoplasms/pathology , DNA Breaks, Double-Stranded , Female , Glioblastoma/chemistry , Glioblastoma/genetics , Glioblastoma/pathology , Glioblastoma/therapy , Glioma/chemistry , Glioma/genetics , Glioma/pathology , Histones/analysis , Humans , Insulin-Like Growth Factor Binding Protein 3/biosynthesis , Insulin-Like Growth Factor Binding Protein 3/genetics , Isocitrate Dehydrogenase/genetics , Male , Mice , Mice, Inbred NOD , Mice, SCID , Middle Aged , Mutation , Neoplasm Proteins/biosynthesis , Neoplasm Proteins/genetics , RNA, Messenger/biosynthesis , RNA, Neoplasm/biosynthesis , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
4.
Mol Vis ; 24: 727-732, 2018.
Article in English | MEDLINE | ID: mdl-30581279

ABSTRACT

Purpose: Inflammation is a key component of retinal disease. We previously reported that exchange protein for cAMP 1 (Epac1) reduced inflammatory mediators, including total levels of high mobility group box 1 (HMGB1) in retinal endothelial cells (RECs) and the mouse retina. The goal of this study was to determine intermediate pathways that allow Epac1 to reduce HMGB1, which could lead to novel targets for therapeutics. Methods: We used endothelial cell-specific conditional knockout mice for Epac1 and RECs to investigate whether Epac1 requires activation of insulin like growth factor binding protein 3 (IGFBP-3) and sirtuin 1 (SIRT1) to reduce acetylated HMGB1 levels with immunoprecipitation, western blot, and enzyme-linked immunosorbent assay (ELISA). Results: Data showed that high glucose reduced IGFBP-3 and SIRT1 levels, and increased acetylation of HMGB1 in RECs. An Epac1 agonist reduced acetylated HMGB1 levels in high glucose. The Epac1 agonist could not reduce HMGB1 or SIRT1 levels when IGFBP-3 siRNA was used. The agonist also could not reduce HMGB1 when SIRT1 siRNA was used. The mouse retina showed that loss of Epac1 increases acetylated HMGB1 levels and reduces IGFBP-3 and SIRT1 levels. Conclusions: Taken together, the data suggest that Epac1 activates IGFBP-3 to increase SIRT1, leading to a significant reduction in acetylated HMGB1. These findings provide novel therapeutic targets for reducing key inflammatory cascades in the retina.


Subject(s)
Endothelial Cells/metabolism , Guanine Nucleotide Exchange Factors/genetics , HMGB1 Protein/genetics , Insulin-Like Growth Factor Binding Protein 3/genetics , Retinal Vessels/metabolism , Sirtuin 1/genetics , Acetylation/drug effects , Animals , Cyclic AMP/analogs & derivatives , Cyclic AMP/pharmacology , Endothelial Cells/cytology , Endothelial Cells/drug effects , Female , Gene Expression Regulation , Glucose/pharmacology , Guanine Nucleotide Exchange Factors/agonists , Guanine Nucleotide Exchange Factors/deficiency , HMGB1 Protein/metabolism , Humans , Insulin-Like Growth Factor Binding Protein 3/antagonists & inhibitors , Insulin-Like Growth Factor Binding Protein 3/metabolism , Male , Mice , Mice, Knockout , Primary Cell Culture , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Retina/cytology , Retina/drug effects , Retina/metabolism , Retinal Vessels/cytology , Retinal Vessels/drug effects , Signal Transduction , Sirtuin 1/antagonists & inhibitors , Sirtuin 1/metabolism , Thionucleotides/pharmacology
5.
Acta Biomater ; 68: 125-136, 2018 03 01.
Article in English | MEDLINE | ID: mdl-29292168

ABSTRACT

Protein delivery is often used in tissue engineering applications to control differentiation processes, but is limited by protein instability and cost. An alternative approach is to control the cellular microenvironment through biomaterial-mediated sequestration of cell-secreted proteins important to differentiation. Thus, we utilized heparin-based microparticles to modulate cellular differentiation via protein sequestration in an in vitro model system of endochondral ossification. Heparin and poly(ethylene-glycol) (PEG; a low-binding material control)-based microparticles were incorporated into ATDC5 cell spheroids or incubated with ATDC5 cells in transwell culture. Reduced differentiation was observed in the heparin microparticle group as compared to PEG and no microparticle-containing groups. To determine if observed changes were due to sequestration of cell-secreted protein, the proteins sequestered by heparin microparticles were analyzed using SDS-PAGE and mass spectrometry. It was found that heparin microparticles bound insulin-like growth factor binding proteins (IGFBP)-3 and 5. When incubated with a small-molecule inhibitor of IGFBPs, NBI 31772, a similar delay in differentiation of ATDC5 cells was observed. These results indicate that heparin microparticles modulated chondrocytic differentiation in this system via sequestration of cell-secreted protein, a technique that could be beneficial in the future as a means to control cellular differentiation processes. STATEMENT OF SIGNIFICANCE: In this work, we present a proof-of-principle set of experiments in which heparin-based microparticles are shown to modulate cellular differentiation through binding of cell-secreted protein. Unlike existing systems that rely on expensive protein with limited half-lives to elicit changes in cellular behavior, this technique focuses on temporal modulation of cell-generated proteins. This technique also provides a biomaterials-based method that can be used to further identify sequestered proteins of interest. Thus, this work indicates that glycosaminoglycan-based biomaterial approaches could be used as substitutes or additions to traditional methods for modulating and identifying the cell-secreted proteins involved in directing cellular behavior.


Subject(s)
Cell Differentiation , Cell-Derived Microparticles/metabolism , Chondrocytes/cytology , Proteins/metabolism , Animals , Biomarkers/metabolism , Cell Line, Tumor , Chondrocytes/metabolism , Chondrogenesis , Gene Expression Regulation , Heparin/chemistry , Insulin-Like Growth Factor Binding Protein 3/antagonists & inhibitors , Insulin-Like Growth Factor Binding Protein 5/antagonists & inhibitors , Mice , Polyethylene Glycols/chemistry , Spheroids, Cellular/cytology , Staining and Labeling
6.
PLoS One ; 9(7): e103578, 2014.
Article in English | MEDLINE | ID: mdl-25073020

ABSTRACT

We recently reported a reciprocal relationship between tumor necrosis factor alpha (TNFα) and insulin-like receptor growth factor binding protein 3 (IGFBP-3) in whole retina of normal and IGFBP-3 knockout mice. A similar relationship was also observed in cultured retinal endothelial cells (REC). We found that TNFα significantly reduced IGFBP-3 levels and vice-versa, IGFBP-3 can lower TNFα and TNFα receptor expression. Since IGFBP-3 is protective to the diabetic retina and TNFα is causative in the development of diabetic retinopathy, we wanted to better understand the cellular mechanisms by which TNFα can reduce IGFBP-3 levels. For these studies, primary human retinal endothelial cells (REC) were used since these cells undergo TNFα-mediated apoptosis under conditions of high glucose conditions and contribute to diabetic retinopathy. We first cultured REC in normal or high glucose, treated with exogenous TNFα, then measured changes in potential signaling pathways, with a focus on P38 mitogen-activated protein kinase alpha (P38α) and casein kinase 2 (CK2) as these pathways have been linked to both TNFα and IGFBP-3. We found that TNFα significantly increased phosphorylation of P38α and CK2. Furthermore, specific inhibitors of P38α or CK2 blocked TNFα inhibition of IGFBP-3 expression, demonstrating that TNFα reduces IGFBP-3 through activation of P38α and CK2. Since TNFα and IGFBP-3 are key mediators of retinal damage and protection respectively in diabetic retinopathy, increased understanding of the relationship between these two proteins will offer new therapeutic options for treatment.


Subject(s)
Casein Kinase II/metabolism , Endothelial Cells/drug effects , Insulin-Like Growth Factor Binding Protein 3/metabolism , Mitogen-Activated Protein Kinase 14/metabolism , Retina/cytology , Tumor Necrosis Factor-alpha/pharmacology , Apoptosis/drug effects , Casein Kinase II/antagonists & inhibitors , Cells, Cultured , Diabetic Retinopathy/pathology , Endothelial Cells/cytology , Endothelial Cells/metabolism , Glucose/pharmacology , Humans , Insulin-Like Growth Factor Binding Protein 3/antagonists & inhibitors , Insulin-Like Growth Factor Binding Protein 3/genetics , Mitogen-Activated Protein Kinase 14/chemistry , Mutagenesis, Site-Directed , Phosphorylation/drug effects , RNA Interference , RNA, Small Interfering/metabolism , Signal Transduction/drug effects
7.
Endocrinology ; 154(10): 3739-44, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23836030

ABSTRACT

Humanin (HN) is a novel 24-amino acid mitochondrial-derived peptide that has demonstrated diverse cytoprotective effects, including an emerging role in diabetes. The purpose of this study was to examine the pharmacokinetics of humanin analogues, which show great potential as therapeutic agents (HNG and the non-IGFBP-3 binding, HNGF6A). 11-week-old male IGFBP-3(-/-) and wild type (WT) mice were divided into 3 groups: WT mice treated with HNG, WT mice treated with HNGF6A, and IGFBP-3(-/-) mice treated with HNG. Plasma was obtained from mice following ip injection with HN analogues, and HN levels were measured with ELISA. WT mice treated with HNGF6A and IGFBP-3(-/-) mice treated with HNG displayed a longer half-life of HN compared with WT mice treated with HNG. Following HNG injection, both IGF-1 and IGFBP-3 levels decreased over time. Adult male Sprague Dawley rats were also ip injected with HNG, and HN levels were measured in various tissues (plasma, liver, heart, and brain) by ELISA. The half-life of HN was found to be longer in rats compared with mice. In rats, HN levels were found to be highest in plasma, present in liver, and undetectable in brain or heart. The current study provides evidence of HN and IGFBP-3 association in the circulation and suggests that native HN may modulate the distribution of IGF-1 and IGFBP-3. The results also demonstrate varying kinetic profiles of HN analogues and interspecies variation in rodents. Sustainable levels of circulating HN measured in plasma underline the potential value of HN analogues as a new therapeutic intervention in the treatment of diabetes.


Subject(s)
Drugs, Investigational/pharmacokinetics , Hypoglycemic Agents/pharmacokinetics , Insulin-Like Growth Factor Binding Protein 3/antagonists & inhibitors , Intracellular Signaling Peptides and Proteins/pharmacokinetics , Animals , Biotransformation , Drugs, Investigational/administration & dosage , Drugs, Investigational/chemistry , Drugs, Investigational/metabolism , Enzyme-Linked Immunosorbent Assay , Half-Life , Hypoglycemic Agents/administration & dosage , Hypoglycemic Agents/chemistry , Hypoglycemic Agents/metabolism , Injections, Intraperitoneal , Insulin-Like Growth Factor Binding Protein 3/blood , Insulin-Like Growth Factor Binding Protein 3/genetics , Insulin-Like Growth Factor Binding Protein 3/metabolism , Insulin-Like Growth Factor I/metabolism , Intracellular Signaling Peptides and Proteins/administration & dosage , Intracellular Signaling Peptides and Proteins/chemistry , Intracellular Signaling Peptides and Proteins/metabolism , Liver/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Rats , Rats, Sprague-Dawley , Species Specificity , Tissue Distribution
8.
Zoolog Sci ; 26(2): 131-8, 2009 Feb.
Article in English | MEDLINE | ID: mdl-19341330

ABSTRACT

Insulin-like growth factor 1 (IGF1) is involved in the proliferation of mouse and rat endometrial cells in a paracrine or autocrine manner. Insulin-like growth factor binding protein-3 (IGFBP3) modulates actions of IGFs directly or indirectly. The present study aimed to determine whether IGFBP3 is involved in the regulation of proliferation of mouse endometrial cells. Mouse endometrial epithelial cells and stromal cells were isolated, and cultured in a serum free medium. IGF1 stimulated DNA synthesis by endometrial epithelial and stromal cells, and IGFBP3 inhibited IGF1-induced DNA synthesis. Estradiol-17beta (E2) decreased the Igfbp3 mRNA level in endometrial stromal cells, whereas it increased the Igf1 mRNA level. Transforming growth factor alpha (TGFalpha) significantly decreased IGFBP3 expression at both the mRNA and secreted protein levels in endometrial stromal cells. Progesterone (P4) did not affect the E2-induced down-regulation of Igfbp3 mRNA expression in endometrial stromal cells, although P4 alone increased Igfbp3 mRNA levels. The present findings suggest that in mouse endometrial stromal cells E2 enhances IGF1 action through enhancement of IGF1 synthesis and reduction of IGFBP3 synthesis, and that TGFalpha affects IGF1 actions through modulation of IGFBP3 levels.


Subject(s)
Endometrium/metabolism , Estradiol/pharmacology , Insulin-Like Growth Factor Binding Protein 3/biosynthesis , Progesterone/pharmacology , Transforming Growth Factor alpha/pharmacology , Animals , Blotting, Western , DNA/biosynthesis , Endometrium/cytology , Endometrium/drug effects , Enzyme Inhibitors/pharmacology , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Female , Insulin-Like Growth Factor Binding Protein 3/antagonists & inhibitors , Insulin-Like Growth Factor Binding Protein 3/genetics , Insulin-Like Growth Factor I/biosynthesis , Insulin-Like Growth Factor I/genetics , Mice , Mice, Inbred ICR , Quinazolines , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Receptor, IGF Type 1/biosynthesis , Receptor, IGF Type 1/genetics , Reverse Transcriptase Polymerase Chain Reaction , Stromal Cells/drug effects , Stromal Cells/metabolism , Transforming Growth Factor alpha/antagonists & inhibitors , Tyrphostins/pharmacology
9.
Virus Res ; 139(1): 14-21, 2009 Jan.
Article in English | MEDLINE | ID: mdl-18948152

ABSTRACT

Hepatitis B virus (HBV), a major causative agent of hepatocelluar carcinoma (HCC), encodes an oncogenic X-protein (HBx) which has been known as a transcriptional transactivator on multiple viral and celluar promoters. In the report, we verified that HBx transcriptionally repress insulin-like growth factor binding protein-3 (IGFBP-3) by promoting HBx/histone deacetylase 1 (HDAC1) complex formation. HBx recruited HDAC1 forms complex with Sp1 in a p53-independent manner) and deacetylates Sp1 which resulted in the diminished binding of Sp1 on targeted DNA during transcriptional repression. Deacetylation of Sp1 by HBx recruited HDAC1 likely to be a part of the mechanism that controls HBx induced IGFBP-3 repression and the modification of chromatin structure.


Subject(s)
Gene Expression Regulation, Neoplastic/physiology , Hepatitis B virus/physiology , Histone Deacetylases/metabolism , Insulin-Like Growth Factor Binding Protein 3/antagonists & inhibitors , Trans-Activators/metabolism , Transcription, Genetic , Cells, Cultured , Gene Expression Regulation, Viral , Hepatitis B virus/genetics , Histone Deacetylase 1 , Promoter Regions, Genetic , Protein Binding , Repressor Proteins/genetics , Repressor Proteins/metabolism , Viral Regulatory and Accessory Proteins
10.
Cancer Res ; 66(20): 9913-20, 2006 Oct 15.
Article in English | MEDLINE | ID: mdl-17047053

ABSTRACT

A disintegrin and metalloproteinases (ADAMs) are involved in various biological events including cell adhesion, cell fusion, membrane protein shedding, and proteolysis. In the present study, our reverse transcription-PCR analysis showed that among the 12 different ADAM species with a putative metalloproteinase motif, prototype membrane-anchored ADAM28m and secreted-type ADAM28s are selectively expressed in human breast carcinoma tissues. By real-time quantitative PCR, their expression levels were significantly higher in carcinomas than in nonneoplastic breast tissues. In situ hybridization, immunohistochemistry, and immunoblotting analyses indicated that ADAM28 is predominantly expressed in an active form by carcinoma cells within carcinoma tissues. A direct correlation was observed between mRNA expression levels and proliferative activity of the carcinoma cells. Treatment of ADAM28-expressing breast carcinoma cells (MDA-MB231) with insulin-like growth factor-I (IGF-I) increased cell proliferation, cleavage of IGF binding protein (IGFBP)-3, as well as IGF-I cell signaling; these processes were all significantly inhibited by treatment with ADAM inhibitor or anti-ADAM28 antibody. Down-regulation of ADAM28 expression in MDA-MB231 cells with small interfering RNA significantly reduced cell proliferation, IGFBP-3 cleavage, and growth of xenografts in mice. In addition, cleavage of IGFBP-3 in breast carcinoma tissues was correlated with ADAM28 expression levels and inhibited by treatment with ADAM inhibitor or anti-ADAM28 antibody. These results show that ADAM28 is overexpressed in an activated form in human breast carcinoma cells and suggest that ADAM28 is involved in cell proliferation through enhanced bioavailability of IGF-I released from the IGF-I/IGFBP-3 complex by selective IGFBP-3 cleavage in human breast carcinomas.


Subject(s)
ADAM Proteins/biosynthesis , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Insulin-Like Growth Factor Binding Protein 3/metabolism , ADAM Proteins/genetics , Animals , Breast Neoplasms/enzymology , Breast Neoplasms/genetics , Cell Growth Processes/physiology , Cell Line, Tumor , Female , Humans , Immunohistochemistry , In Situ Hybridization , Insulin-Like Growth Factor Binding Protein 3/antagonists & inhibitors , Insulin-Like Growth Factor I/pharmacology , Ki-67 Antigen/biosynthesis , Male , Mice , Mice, Inbred BALB C , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , RNA, Small Interfering/genetics , Signal Transduction/drug effects , Signal Transduction/physiology , Transplantation, Heterologous
11.
Cancer Sci ; 97(11): 1182-90, 2006 Nov.
Article in English | MEDLINE | ID: mdl-16965600

ABSTRACT

Insulin-like growth factor binding protein-3 (IGFBP-3) modulates cell proliferation of various cancer cell types. However, it remains unclear how IGF-IGFBP-3-signaling is involved in growth and progression of hepatocellular carcinoma (HCC). The aim of the present study was to evaluate the role of IGFBP-3 in HCC. Type 1 receptor for IGF (IGF-1R) was expressed at various levels in the seven lines examined, but IGF-2R was not expressed. Of the seven lines, the growth of HAK-1B, KIM-1, KYN-2 and HepG2 cells was stimulated in a dose-dependent manner by the exogenous addition of IGF-I or IGF-II, but the HAK-1A, KYN-1 and KYN-3 cell lines showed no growth. Exogenous addition of IGFBP-3 markedly blocked IGF-I and IGF-II-stimulated cell growth of KYN-2 and HepG2 cells, and moderately stimulated that of KIM-1 and HAK-1B cells, but no growth of the KYN-1, KYN-3 and HAK-1A cell lines was observed. IGF-I enhanced the phosphorylation of IGF-1R, Akt and Erk1/2 in KYN-2 cells, and coadministration of IGFBP-3 blocked all types of activation by IGF-I investigated here. In contrast, no such activation by IGF-I was detected in KYN-3 cells. IGFBP-3 also suppressed IGF-I-induced cell invasion by KYN-2 cells. Moreover, we were able to observe the apparent expression of IGFBP-3 in KYN-3 cells, but not in the other six cell lines. Furthermore reduced expression of IGFBP-3, but not that of IGF-1R, was significantly correlated with tumor size, histological differentiation, capsular invasion and portal venous invasion. Low expression of IGFBP-3 was independently associated with poor survival. IGFBP-3 could be a molecular target of intrinsic importance for further development of novel therapeutic strategy against HCC.


Subject(s)
Carcinoma, Hepatocellular/metabolism , Insulin-Like Growth Factor Binding Protein 3/metabolism , Liver Neoplasms/metabolism , Neoplasm Invasiveness/pathology , Portal Vein/pathology , Aged , Aged, 80 and over , Carcinoma, Hepatocellular/pathology , Female , Humans , Immunoenzyme Techniques , Insulin-Like Growth Factor Binding Protein 3/antagonists & inhibitors , Insulin-Like Growth Factor Binding Protein 3/genetics , Insulin-Like Growth Factor I/metabolism , Liver Neoplasms/pathology , Male , Middle Aged , Portal Vein/metabolism , Prognosis , Proto-Oncogene Proteins c-akt/metabolism , RNA, Small Interfering/pharmacology , Receptor, Fibroblast Growth Factor, Type 1/antagonists & inhibitors , Receptor, Fibroblast Growth Factor, Type 1/genetics , Receptor, Fibroblast Growth Factor, Type 1/metabolism , Survival Rate , Tumor Cells, Cultured
12.
Biochem Pharmacol ; 72(1): 53-61, 2006 Jun 28.
Article in English | MEDLINE | ID: mdl-16684509

ABSTRACT

The insulin-like growth factor binding proteins (IGFBPs) represent a unique class of IGF antagonists regulating the bioavailability of the IGFs extracellularly. Accordingly, they represent an important class of proteins for cancer therapeutics and chemoprevention. IGF-F1-1 is a cyclic hexadecapeptide identified by high throughput phage display that binds to the IGFBP-binding domain on IGF-1. It acts as an IGFBP-mimetic, capable of inhibiting IGF-1 binding to the IGFBPs. To further examine the utility of IGF-F1-1 as an IGF-1 antagonist we tested its ability to inhibit IGFBP-2 and IGFBP-3 binding to IGF-1, (125)I-IGF-1 binding to IGF-1Rs and to block IGF-1 induced Akt activation, cell cycle changes and [(3)H]thymidine incorporation in MCF-7 cells. These biological activities were inhibited by treatment with IGFBP-2, wortmannin or the IGF-1R tyrosine kinase inhibitor, NVP-AEW541, but not by IGF-F1-1. Our findings confirm previous studies indicating that IGF-F1-1 is a weak antagonist of IGF-1 binding to the IGFBPs and the IGF-1R and suggest that it does not effectively inhibit downstream events stimulated by IGF-1. We further demonstrated that IGF-F1-1 treatment of MCF-7 cells results in the paradoxical activation of Akt, S-phase transition and [(3)H]thymidine incorporation. These results suggest that IGF-F1-1 is a weak agonist, exhibiting mitogenic actions. IGF-F1-1 may act in conjunction with IGF-1 at the IGF-1R or independently of IGF-1 at the IGF-1R or another receptor.


Subject(s)
Insulin-Like Growth Factor I/antagonists & inhibitors , Peptides, Cyclic/pharmacology , Receptor, IGF Type 1/metabolism , Signal Transduction , Androstadienes/pharmacology , Cell Cycle/physiology , Cell Line, Tumor/drug effects , DNA Replication , Humans , Insulin-Like Growth Factor Binding Protein 2/antagonists & inhibitors , Insulin-Like Growth Factor Binding Protein 2/metabolism , Insulin-Like Growth Factor Binding Protein 2/pharmacology , Insulin-Like Growth Factor Binding Protein 3/antagonists & inhibitors , Insulin-Like Growth Factor Binding Protein 3/metabolism , Insulin-Like Growth Factor I/metabolism , Peptide Library , Phosphorylation , Protein Binding/drug effects , Proto-Oncogene Proteins c-akt/metabolism , Pyrimidines/pharmacology , Pyrroles/pharmacology , Wortmannin
13.
Cytokine ; 30(5): 248-53, 2005 Jun 07.
Article in English | MEDLINE | ID: mdl-15927849

ABSTRACT

The nature of erythropoietin (EPO)-dependent, erythroid cell regulatory factors secreted by endothelial cells is largely unknown. The production of thrombospondin 1 (TSP-1) and insulin-like growth factor binding protein 3 (IGFBP-3) is increased in cultures of human umbilical vein endothelial cells (HUVEC) incubated with erythropoietin (EPO). Simultaneous incubation of HUVEC with EPO and interleukin 3 (IL-3) resulted in a decreased production, suggesting that both TSP-1 and IGFBP-3 belong to the EPO- and IL-3-dependent erythroid regulatory factors previously described in cultures of bone marrow endothelial cells. TSP-1 and TSP-1 derived synthetic peptides based on the CD36 and CD47 binding sites of TSPs increased thymidine incorporation into bovine erythroid cells of fetal liver. IGBBP-3 inhibited thymidine incorporation in the same cells. Preincubation of erythroid cells with TSP-1 eliminated the inhibitory activity of IGFBP-3. We suggest that EPO-dependent, endothelial-derived TSP-1 may play a positive role in red cell production by acting directly on erythroid cells, stimulating DNA synthesis and preventing the inhibitory action of IGFBP-3.


Subject(s)
Endothelial Cells/metabolism , Erythroid Cells/metabolism , Erythropoietin/pharmacology , Insulin-Like Growth Factor Binding Protein 3/antagonists & inhibitors , Thrombospondin 1/metabolism , Thymidine/metabolism , Umbilical Cord/metabolism , Animals , Cattle , Cells, Cultured , Endothelial Cells/drug effects , Erythroid Cells/drug effects , Humans , Insulin-Like Growth Factor Binding Protein 3/metabolism , Interleukin-3/antagonists & inhibitors , Interleukin-3/metabolism , Peptide Fragments/pharmacology , Thrombospondin 1/pharmacology , Umbilical Cord/drug effects
14.
Cancer Res ; 65(6): 2314-20, 2005 Mar 15.
Article in English | MEDLINE | ID: mdl-15781645

ABSTRACT

DeltaNp63alpha is a nuclear transcription factor that maintains epithelial progenitor cell populations, is overexpressed in several epithelial cancers, and can negatively regulate apoptosis. However, the mechanisms by which DeltaNp63alpha promotes cell survival are unclear. DeltaNp63alpha has been reported to act as a transcriptional repressor, but specific target genes directly repressed by DeltaNp63alpha remain unidentified. Here, we present evidence that DeltaNp63alpha functions to negatively regulate the proapoptotic protein IGFBP-3. Disruption of p63 expression in squamous epithelial cells increases IGFBP-3 expression, whereas ectopic expression of DeltaNp63alpha down-regulates IGFBP-3. DeltaNp63alpha binds to sites in the IGFBP-3 gene in vivo and can modulate transcription through these sites. Furthermore, DeltaNp63alpha and IGFBP-3 expression patterns are inversely correlated in normal squamous epithelium and squamous cell carcinomas. These data suggest that IGFBP-3 is a target of transcriptional repression by DeltaNp63alpha and that this repression represents a mechanism by which tumors that overexpress p63 may be protected from apoptosis.


Subject(s)
Gene Expression Regulation, Neoplastic/physiology , Insulin-Like Growth Factor Binding Protein 3/genetics , Phosphoproteins/genetics , Trans-Activators/genetics , Apoptosis/genetics , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/metabolism , Cell Line, Tumor , DNA-Binding Proteins , Epithelial Cells/cytology , Epithelial Cells/metabolism , Epithelial Cells/physiology , Genes, Tumor Suppressor , Humans , Insulin-Like Growth Factor Binding Protein 3/antagonists & inhibitors , Insulin-Like Growth Factor Binding Protein 3/biosynthesis , Keratinocytes/cytology , Keratinocytes/metabolism , Keratinocytes/physiology , Phosphoproteins/biosynthesis , Trans-Activators/biosynthesis , Transcription Factors , Transcription, Genetic/physiology , Transfection , Tumor Suppressor Proteins , Up-Regulation
15.
Cancer Res ; 64(23): 8715-22, 2004 Dec 01.
Article in English | MEDLINE | ID: mdl-15574782

ABSTRACT

We earlier demonstrated that oral infusion of green tea polyphenols inhibits development and progression of prostate cancer in transgenic adenocarcinoma of the mouse prostate (TRAMP) model. Evidence indicates that elevated levels of IGF-I with concomitant lowering of IGF binding protein (IGFBP)-3 are associated with increased risk for prostate cancer development and progression. In this study, we examined the role of IGF/IGFBP-3 signaling and its downstream and other associated events during chemoprevention of prostate cancer by green tea polyphenols in TRAMP mice. Our data demonstrated an increase in the levels of IGF-I, phosphatidylinositol 3'-kinase, phosphorylated Akt (Thr-308), and extracellular signal-regulated kinase 1/2 with concomitant decrease in IGFBP-3 in dorso-lateral prostate of TRAMP mice during the course of cancer progression, i.e., as a function of age. Continuous green tea polyphenol infusion for 24 weeks to these mice resulted in substantial reduction in the levels of IGF-I and significant increase in the levels of IGFBP-3 in the dorso-lateral prostate. This modulation of IGF/IGFBP-3 was found to be associated with an inhibition of protein expression of phosphatidylinositol 3'-kinase, phosphorylated forms of Akt (Thr-308) and extracellular signal-regulated kinase 1/2. Furthermore, green tea polyphenol infusion resulted in marked inhibition of markers of angiogenesis and metastasis most notably vascular endothelial growth factor, urokinase plasminogen activator, and matrix metalloproteinases 2 and 9. Based on our data, we suggest that IGF-I/IGFBP-3 signaling pathway is a prime pathway for green tea polyphenol-mediated inhibition of prostate cancer that limits the progression of cancer through inhibition of angiogenesis and metastasis.


Subject(s)
Adenocarcinoma/drug therapy , Flavonoids/pharmacology , Insulin-Like Growth Factor I/antagonists & inhibitors , Phenols/pharmacology , Prostatic Neoplasms/drug therapy , Tea , Adenocarcinoma/blood supply , Adenocarcinoma/metabolism , Adenocarcinoma/pathology , Animals , Disease Progression , Extracellular Signal-Regulated MAP Kinases/biosynthesis , Female , Insulin-Like Growth Factor Binding Protein 3/antagonists & inhibitors , Insulin-Like Growth Factor Binding Protein 3/physiology , Insulin-Like Growth Factor I/physiology , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neoplasm Metastasis , Neovascularization, Pathologic/drug therapy , Neovascularization, Pathologic/pathology , Phosphatidylinositol 3-Kinases/biosynthesis , Polyphenols , Prostatic Neoplasms/blood supply , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Protein Serine-Threonine Kinases/biosynthesis , Proto-Oncogene Proteins/biosynthesis , Proto-Oncogene Proteins c-akt , Signal Transduction/drug effects
16.
Cancer Res ; 64(21): 7711-23, 2004 Nov 01.
Article in English | MEDLINE | ID: mdl-15520175

ABSTRACT

Epidermal growth factor receptor (EGFR) is frequently overexpressed in esophageal carcinoma and its precursor lesions. To gain insights into how EGFR overexpression affects cellular functions in primary human esophageal cells, we performed gene expression profiling and identified insulin-like growth factor-binding protein (IGFBP)-3 as the most up-regulated gene. IGFBP-3 regulates cell proliferation through both insulin-like growth factor-dependent and independent mechanisms. We found that IGFBP-3 mRNA and protein expression was increased in EGFR-overexpressing primary and immortalized human esophageal cells. IGFBP-3 was also up-regulated in EGFR-overexpressing cells in organotypic culture and in EGFR transgenic mice. Furthermore, IGFBP-3 mRNA was overexpressed in 80% of primary esophageal squamous cell carcinomas and 60% of primary esophageal adenocarcinomas. Concomitant up-regulation of EGFR and IGFBP-3 was observed in 60% of primary esophageal squamous cell carcinomas. Immunohistochemistry revealed cytoplasmic localization of IGFBP-3 in the preponderance of preneoplastic and neoplastic esophageal lesions. IGFBP-3 was also overexpressed in esophageal cancer cell lines at both mRNA (60%) and protein (40%) levels. IGFBP-3 secreted by cancer cells was capable of binding to insulin-like growth factor I. Functionally, epidermal growth factor appeared to regulate IGFBP-3 expression in esophageal cancer cell lines. Finally, suppression of IGFBP-3 by small interfering RNA augmented cell proliferation, suggesting that IGFBP-3 may inhibit tumor cell proliferation as a negative feedback mechanism. In aggregate, we have identified for the first time that IGFBP-3 is an aberrantly regulated gene through the EGFR signaling pathway and it may modulate EGFR effects during carcinogenesis.


Subject(s)
ErbB Receptors/physiology , Insulin-Like Growth Factor Binding Protein 3/genetics , Animals , Apoptosis , Cell Division , Cell Line, Tumor , DNA-Binding Proteins , Esophageal Neoplasms/metabolism , Esophagus/metabolism , Gene Expression Regulation, Neoplastic , Humans , Insulin-Like Growth Factor Binding Protein 3/antagonists & inhibitors , Insulin-Like Growth Factor Binding Protein 3/physiology , Mice , Mice, Transgenic , RNA, Small Interfering/pharmacology , Signal Transduction , Telomerase/physiology
17.
FASEB J ; 18(14): 1719-21, 2004 Nov.
Article in English | MEDLINE | ID: mdl-15371331

ABSTRACT

The type V TGF-beta receptor (TbetaR-V) mediates IGF-independent growth inhibition by IGFBP-3 and mediates growth inhibition by TGF-beta1 in concert with the other TGF-beta receptor types. TbetaR-V was recently found to be identical to LRP-1. Here we find that insulin and (Q3A4Y15L16) IGF-I (an IGF-I analog that has a low affinity for IGFBP-3) antagonize growth inhibition by IGFBP-3 in mink lung epithelial cells (Mv1Lu cells) stimulated by serum. In these cells, IGFBP-3 induces serine-specific dephosphorylation of IRS-1 and IRS-2. The IGFBP-3-induced dephosphorylation of IRS-2 is prevented by cotreatment of cells with insulin, (Q3A4Y15L16) IGF-I, or TbetaR-V/LRP-1 antagonists. The magnitude of the IRS-2 dephosphorylation induced by IGFBP-3 positively correlates with the degree of growth inhibition by IGFBP-3 in Mv1Lu cells and mutant cells derived from Mv1Lu cells. Stable transfection of murine 32D myeloid cells (which lack endogenous IRS proteins and are insensitive to growth inhibition by IGFBP-3) with IRS-1 or IRS-2 cDNA confers sensitivity to growth inhibition by IGFBP-3; this IRS-mediated growth inhibition can be completely reversed by insulin in 32D cells stably expressing IRS-2 and the insulin receptor. These results suggest that IRS-1 and IRS-2 are key molecules for the TbetaR-V/LRP-1-mediated growth inhibitory signaling cascade.


Subject(s)
Growth Inhibitors/pharmacology , Insulin-Like Growth Factor Binding Protein 3/pharmacology , Low Density Lipoprotein Receptor-Related Protein-1/metabolism , Phosphoproteins/physiology , Receptors, Transforming Growth Factor beta/metabolism , Animals , Cell Line , Cell Proliferation , Epithelial Cells/metabolism , Growth Inhibitors/antagonists & inhibitors , Insulin/pharmacology , Insulin Receptor Substrate Proteins , Insulin-Like Growth Factor Binding Protein 3/antagonists & inhibitors , Insulin-Like Growth Factor I/pharmacology , Intracellular Signaling Peptides and Proteins , Mice , Myeloid Cells/metabolism , Phosphoproteins/metabolism , Signal Transduction
19.
J Clin Endocrinol Metab ; 89(7): 3105-13, 2004 Jul.
Article in English | MEDLINE | ID: mdl-15240578

ABSTRACT

The catabolic state of critical illness has been linked to the suppressed somatotropic GH-IGF-binding protein (IGFBP) axis. In critically ill patients it has been demonstrated that, compared with the conventional approach, which only recommended insulin therapy when blood glucose levels exceeded 12 mmol/liter, strict maintenance of blood glucose levels below 6.1 mmol/liter with intensive insulin therapy almost halved intensive care mortality, acute renal failure, critical illness polyneuropathy, and bloodstream infections. Poor blood glucose control in diabetes mellitus has also been associated with low serum IGF-I levels, which can be increased by insulin therapy. We hypothesized that intensive insulin therapy would improve the IGF-I axis, possibly contributing to the clinical correlates of anabolism. Therefore, this study of 363 patients, requiring intensive care for more than 7 d and randomly assigned to either conventional or intensive insulin therapy, examines the effects of intensive insulin therapy on the somatotropic axis. Contrary to expectation, intensive insulin therapy suppressed serum IGF-I, IGFBP-3, and acid-labile subunit concentrations. This effect was independent of survival of the critically ill patient. Concomitantly, serum GH levels were increased by intensive insulin therapy. The suppression of IGF-I in association with the increased GH levels suggests GH resistance induced by intensive insulin therapy, which was reflected by the decreased serum GH-binding protein levels. Intensive insulin therapy did not affect IGFBP-3 proteolysis, which was markedly higher in protracted critically ill patients compared with healthy controls. Also, intensive insulin therapy did not suppress the urea/creatinine ratio, a clinical correlate of catabolism. In conclusion, our data suggest that intensive insulin therapy surprisingly suppressed the somatotropic axis despite its beneficial effects on patient outcome. GH resistance accompanied this suppression of the IGF-I axis. To what extent and through which mechanisms the changes in the GH-IGF-IGFBP axis contributed to the survival benefit under intensive insulin therapy remain elusive.


Subject(s)
Critical Care , Critical Illness/therapy , Growth Hormone/metabolism , Insulin/therapeutic use , Aged , Blood Glucose/drug effects , Blood Glucose/metabolism , Carrier Proteins/antagonists & inhibitors , Carrier Proteins/blood , Chronic Disease , Female , Gene Expression/drug effects , Glycoproteins/antagonists & inhibitors , Human Growth Hormone/metabolism , Humans , Insulin-Like Growth Factor Binding Protein 3/antagonists & inhibitors , Insulin-Like Growth Factor Binding Protein 3/metabolism , Insulin-Like Growth Factor I/antagonists & inhibitors , Insulin-Like Growth Factor I/genetics , Insulin-Like Growth Factor I/metabolism , Intensive Care Units , Length of Stay , Liver/metabolism , Male , Middle Aged , Peptide Hydrolases/metabolism , RNA, Messenger/metabolism , Receptors, Somatotropin/genetics , Survival Analysis
20.
Cancer Res ; 64(7): 2516-22, 2004 Apr 01.
Article in English | MEDLINE | ID: mdl-15059907

ABSTRACT

Autocrine motility factor/phosphoglucose isomerase (AMF/PGI) was identified as a binding partner for insulin-like growth factor binding protein-3 (IGFBP-3) in solubilized T47D and MCF-7 human breast cancer cell membranes. The interaction between AMF/PGI and IGFBP-3 was verified by cross-linking biotinylated IGFBP-3 to intact cells. After solubilization of the membranes, the biotinylated complexes were precipitated with streptavidin-agarose conjugate and analyzed by SDS-PAGE. A M(r) approximately 80,000 complex was identified when the nitrocellulose membranes were probed either with streptavidin-horseradish peroxidase conjugate or AMF/PGI antiserum confirming the cross-linking of IGFBP-3 to AMF/PGI. The interaction between IGFBP-3 and AMF/PGI was also further confirmed by ligand blotting of purified AMF/PGI using biotinylated IGFBP-3. Both glycosylated and nonglycosylated IGFBP-3 inhibited the catalytic activity of AMF/PGI in a dose-dependent fashion. In addition, IGFBP-3 inhibited the binding of AMF/PGI to breast cancer cells and AMF/PGI-induced migration of both T47D and MCF-7 human breast cancer cells. IGFBP-3 also decreased the phosphorylation of AMF/PGI and reduced the translocation of AMF/PGI to the cell membrane and AMF/PGI. AMF/PGI resulted in a dose-dependent inhibition of IGFBP-3 induced apoptosis in T47D and MCF-7 cells. In summary, we have identified AMF/PGI as a membrane-associated binding partner for IGFBP-3 in breast cancer cells. The ability of IGFBP-3 to bind and inhibit the actions of AMF/PGI may have some role in the antiproliferative proapoptotic effects of IGFBP-3.


Subject(s)
Glucose-6-Phosphate Isomerase/antagonists & inhibitors , Glucose-6-Phosphate Isomerase/metabolism , Insulin-Like Growth Factor Binding Protein 3/metabolism , Apoptosis/physiology , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Catalysis , Cell Line, Tumor , Cell Membrane/chemistry , Cell Membrane/metabolism , Cell Movement/drug effects , Glucose-6-Phosphate Isomerase/isolation & purification , Humans , Insulin-Like Growth Factor Binding Protein 3/antagonists & inhibitors , Insulin-Like Growth Factor Binding Protein 3/isolation & purification , Insulin-Like Growth Factor Binding Protein 3/pharmacology , Phosphorylation , Protein Binding
SELECTION OF CITATIONS
SEARCH DETAIL
...