Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
Curr Issues Mol Biol ; 45(2): 1113-1126, 2023 Jan 31.
Article in English | MEDLINE | ID: mdl-36826019

ABSTRACT

The tumor microenvironment (TME) plays a vital role in tumor invasion and metastasis and provides a rich environment for identifying novel therapeutic targets. The TME landscape consists of an extracellular matrix (ECM) and stromal cells. ECM is a major component of TME that mediates the interaction between cancer cells and stromal cells to promote invasion and metastasis. We have shown in published work that RASSF1C promotes cancer stem cell development, migration, and drug resistance, in part, by promoting EMT through a mechanism that involves up-regulation of the PIWIL1-piRNA axis. Consistent with this, in this study, we demonstrate that RASSF1C promotes lung cancer metastasis in vivo using an orthotopic mouse model. Interestingly, two target genes identified in a previously conducted microarray study to be up-regulated by RASSF1C in breast and non-small cell lung cancer (NSCLC) cells are prolyl 4-hydroxylase alpha-2 (P4HA2) and procollagen-lysine, 2-oxoglutarate 5-dioxygenase 2 (PLOD2). In cancer, P4H2A and PLOD2 are vital for collagen posttranslational modification and folding leading to the formation of a stiff ECM and induction of EMT and cancer stem cell marker gene expression, resulting in metastatic dissemination. Here, we also show that overexpression of RASSF1C up-regulates Collagen I, P4HA2, and PLOD2 in vitro. Up-regulation of P4HA2 and PLOD2 by RASSF1C was also confirmed in lung and breast cancer cells in vivo using mouse models. Further, we found that treatment of wildtype lung cancer cells or lung cancer cells overexpressing RASSF1C or PIWIL1 with piR-35127 and 46545 (both down-regulated by RASSF1C) decreased lung cancer cell invasion/migration. Taken together, our findings suggest that RASSF1C may promote lung cancer cell ECM remodeling to induce lung cancer cell stemness, invasion, and metastasis, in part, by up-regulating a previously unknown PIWIL1-P4HA2-PLOD2 pathway. Furthermore, piR-35127 and piR-46545 could potentially be important anti-metastatic tools.

2.
Oncotarget ; 11(45): 4082-4092, 2020 Nov 10.
Article in English | MEDLINE | ID: mdl-33227088

ABSTRACT

INTRODUCTION: Recently we have identified a novel RASSF1C-PIWIL1-piRNA pathway that promotes lung cancer cell progression and migration. PIWI-like proteins interact with piRNAs to form complexes that regulate gene expression at the transcriptional and translational levels. We have illustrated in previous work that RASSF1C modulates the expression of the PIWIL1-piRNA gene axis, suggesting the hypothesis that the RASSF1C-PIWI-piRNA pathway could potentially contribute to lung cancer stem cell development and progression, in part, through modulation of gene methylation of both oncogenic and tumor suppressor genes. Therefore, we tested this hypothesis using a non-small cell lung cancer (NSCLC) cell model to identify Candidate Differentially Methylated Regions (DMRs) modulated by the RASSF1C-PIWIL1-piRNA pathway. MATERIALS AND METHODS: We studied the impact of over-expressing RASSF1C and knocking down RASSF1C and PIWIL1 expression on global gene DNA methylation in the NSCLC cell line H1299 using the Reduced Representation Bisulfite Sequencing (RRBS) method. RESULTS: DMRs were identified by comparing DNA methylation profiles of experimental and control cells. Over-expression of RASSF1C and knocking down RASSF1C and PIWIL1 modulated DNA methylation of genomic regions; and statistically significant candidate genes residing DMR regions in lung cancer cells were identified, including oncogenes and tumor suppressors. One of the hypermethylated genes, Gem Interacting Protein (GMIP), displays tumor suppressor properties. GMIP expression attenuates lung cancer cell migration, and its over-expression is associated with longer survival of lung cancer patients. CONCLUSIONS: The RASSF1C-PIWI-piRNA pathway modulates key oncogenes and tumor suppressor genes. GMIP is hypermethylated by this pathway and has tumor suppressor properties.

3.
Oncotarget ; 10(2): 123-132, 2019 Jan 04.
Article in English | MEDLINE | ID: mdl-30719208

ABSTRACT

RASSF1C functions as an oncogene in lung cancer cells by stimulating proliferation and migration, and reducing apoptosis. Further, RASSF1C up-regulates important protein-coding and non-coding genes involved in lung cancer cell growth, including the stem cell self-renewal gene, piwil1, and small noncoding PIWI-interacting RNAs (piRNAs). In this article, we report the identification of microRNAs (miRNAs) that are modulated in lung cancer cells over-expressing RASSF1C. A lung cancer-specific miRNA PCR array screen was performed to identify RASSF1C target miRNA-coding genes using RNA isolated from the lung cancer cell line H1299 stably over-expressing RASSF1C and corresponding control. Several modulated miRNA genes were identified that are important in cancer cell proliferation and survival. Among the miRNAs down-regulated by RASSF1C is miRNA-33a-5p (miRNA-33a), which functions as a tumor suppressor in lung cancer cells. We validated that over-expression of RASSF1C down-regulates miR-33a expression and RASSF1C knockdown up-regulates miR-33a expression. We found that RASSF1C over-expression also increases ß-catenin, vimentin, and snail protein levels in cells over-expressing miR-33a. In addition, we found that RASSF1C up-regulates the expression of ABCA1 mRNA which is a known target of miR-33a. Our findings suggest that RASSF1C may promote lung epithelial mesenchymal transition (EMT), resulting in the development of a lung cancer stem cell phenotype, progression, and metastasis, in part, through modulation of miR-33a expression. Our findings reveal a new mechanistic insight into how RASSF1C functions as an oncogene.

4.
Oncotarget ; 8(21): 34268-34282, 2017 May 23.
Article in English | MEDLINE | ID: mdl-28423657

ABSTRACT

RASSF1C up-regulates important genes involved in lung cancer cell growth, including a stem cell self-renewal gene, piwil1. In this article, we report the identification of small noncoding PIWI-interacting RNAs (piRNAs) in lung cancer cells over-expressing RASSF1C. A piRNA microarray screen was performed using RNA isolated from the lung cancer cell line H1299 stably over-expressing RASSF1C and corresponding control. The piRNA microarray screen identified several piRNAs that are regulated by RASSF1C and we have validated the expression of two up-regulated piRNAs (piR-34871 and piR-52200) and two down-regulated piRNAs (piR-35127 and piR-46545) in lung cancer cells with silenced and over-expressed RASSF1C using RT-PCR. We also assessed the expression of these four piRNAs in lung tumor and matched normal tissues (n = 12). We found that piR-34871 and piR-52200 were up-regulated in 58% and 50%, respectively; while piR-35127 and piR-46545 were down-regulated in 50% in lung tumor tissues tested. The expression of piR-35127 was inversely correlated with RASSF1C expression in 10/12 tumor tissues. Over-expression of piR-35127 and piR-46545 and knock-down of piR-34871 and piR-52200 significantly reduced normal lung and breast epithelial cell proliferation and cell colony formation as well as proliferation of lung cancer cell lines (A549 and H1299) and breast cancer cell lines (Hs578T and MDA-MB-231). This suggests that these novel piRNAs may potentially be involved in regulating lung cell transformation and tumorigenesis. RASSF1C may potentially modulate the expression of its piRNA target genes through attenuation of the AMPK pathway, as over-expression of RASSF1C resulted in reduction of p-AMPK, p21, and p27 protein levels.


Subject(s)
Gene Regulatory Networks , Lung Neoplasms/genetics , RNA, Small Interfering/genetics , Tumor Suppressor Proteins/genetics , A549 Cells , Adenylate Kinase/genetics , Cell Line, Tumor , Cell Proliferation , Female , Gene Expression Regulation, Neoplastic , Humans , Oligonucleotide Array Sequence Analysis , Up-Regulation
5.
PLoS One ; 9(7): e101679, 2014.
Article in English | MEDLINE | ID: mdl-25007054

ABSTRACT

RASSF1C is a major isoform of the RASSF1 gene, and is emerging as an oncogene. This is in contradistinction to the RASSF1A isoform, which is an established tumor suppressor. We have previously shown that RASSF1C promotes lung cancer cell proliferation and have identified RASSF1C target genes with growth promoting functions. Here, we further report that RASSF1C promotes lung cancer cell migration and enhances lung cancer cell tumor sphere formation. We also show that RASSF1C over-expression reduces the inhibitory effects of the anti-cancer agent, betulinic acid (BA), on lung cancer cell proliferation. In previous work, we demonstrated that RASSF1C up-regulates piwil1 gene expression, which is a stem cell self-renewal gene that is over-expressed in several human cancers, including lung cancer. Here, we report on the effects of BA on piwil1 gene expression. Cells treated with BA show decreased piwil1 expression. Also, interaction of IGFBP-5 with RASSF1C appears to prevent RASSF1C from up-regulating PIWIL1 protein levels. These findings suggest that IGFBP-5 may be a negative modulator of RASSF1C/ PIWIL1 growth-promoting activities. In addition, we found that inhibition of the ATM-AMPK pathway up-regulates RASSF1C gene expression.


Subject(s)
Argonaute Proteins/metabolism , Gene Expression , Insulin-Like Growth Factor Binding Protein 5/metabolism , Tumor Suppressor Proteins/genetics , Antineoplastic Agents/pharmacology , Argonaute Proteins/genetics , Cell Movement , Cell Proliferation , Drug Resistance, Neoplasm , Gene Expression Regulation, Neoplastic , Humans , Hydroxamic Acids/pharmacology , Insulin-Like Growth Factor Binding Protein 5/genetics , Lung Neoplasms , Pentacyclic Triterpenes , Pyrazoles/pharmacology , Pyrimidines/pharmacology , Signal Transduction , Spheroids, Cellular , Triterpenes/pharmacology , Tumor Suppressor Proteins/metabolism , beta Catenin/genetics , beta Catenin/metabolism , Betulinic Acid
6.
BMC Res Notes ; 5: 239, 2012 May 16.
Article in English | MEDLINE | ID: mdl-22591718

ABSTRACT

BACKGROUND: RASSF1A and RASSF1C are two major isoforms encoded by the Ras association domain family 1 (RASSF1) gene through alternative promoter selection and mRNA splicing. RASSF1A is a well established tumor suppressor gene. Unlike RASSF1A, RASSF1C appears to have growth promoting actions in lung cancer. In this article, we report on the identification of novel RASSF1C target genes in non small cell lung cancer (NSCLC). METHODS: Over-expression and siRNA techniques were used to alter RASSF1C expression in human lung cancer cells, and Affymetrix-microarray study was conducted using NCI-H1299 cells over-expressing RASSF1C to identify RASSF1C target genes. RESULTS: The microarray study intriguingly shows that RASSF1C modulates the expression of a number of genes that are involved in cancer development, cell growth and proliferation, cell death, and cell cycle. We have validated the expression of some target genes using qRT-PCR. We demonstrate that RASSF1C over-expression increases, and silencing of RASSF1C decreases, the expression of PIWIL1 gene in NSCLC cells using qRT-PCR, immunostaining, and Western blot analysis. We also show that RASSF1C over-expression induces phosphorylation of ERK1/2 in lung cancer cells, and inhibition of the MEK-ERK1/2 pathway suppresses the expression of PIWIL1 gene expression, suggesting that RASSF1C may exert its activities on some target genes such as PIWIL1 through the activation of the MEK-ERK1/2 pathway. Also, PIWIL1 expression is elevated in lung cancer cell lines compared to normal lung epithelial cells. CONCLUSIONS: Taken together, our findings provide significant data to propose a model for investigating the role of RASSF1C/PIWIL1 proteins in initiation and progression of lung cancer.


Subject(s)
Argonaute Proteins/genetics , Gene Expression Regulation, Neoplastic/genetics , Neoplastic Stem Cells/metabolism , Tumor Suppressor Proteins/genetics , Apoptosis/genetics , Argonaute Proteins/metabolism , Benzamides/pharmacology , Blotting, Western , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/metabolism , Carcinoma, Non-Small-Cell Lung/pathology , Cell Cycle/genetics , Cell Line , Cell Line, Tumor , Cell Proliferation , Extracellular Signal-Regulated MAP Kinases/antagonists & inhibitors , Extracellular Signal-Regulated MAP Kinases/metabolism , Gene Expression Profiling , Gene Expression Regulation, Neoplastic/drug effects , Humans , Immunohistochemistry , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , MAP Kinase Signaling System/drug effects , Oligonucleotide Array Sequence Analysis , Phosphorylation/drug effects , RNA Interference , Reverse Transcriptase Polymerase Chain Reaction , Tumor Suppressor Proteins/metabolism
7.
BMC Cancer ; 10: 562, 2010 Oct 18.
Article in English | MEDLINE | ID: mdl-20955597

ABSTRACT

BACKGROUND: The Ras association domain family 1 (RASSF1) gene is a Ras effector encoding two major mRNA forms, RASSF1A and RASSF1C, derived by alternative promoter selection and alternative mRNA splicing. RASSF1A is a tumor suppressor gene. However, very little is known about the function of RASSF1C both in normal and transformed cells. METHODS: Gene silencing and over-expression techniques were used to modulate RASSF1C expression in human breast cancer cells. Affymetrix-microarray analysis was performed using T47D cells over-expressing RASSF1C to identify RASSF1C target genes. RT-PCR and western blot techniques were used to validate target gene expression. Cell invasion and apoptosis assays were also performed. RESULTS: In this article, we report the effects of altering RASSF1C expression in human breast cancer cells. We found that silencing RASSF1C mRNA in breast cancer cell lines (MDA-MB231 and T47D) caused a small but significant decrease in cell proliferation. Conversely, inducible over-expression of RASSF1C in breast cancer cells (MDA-MB231 and T47D) resulted in a small increase in cell proliferation. We also report on the identification of novel RASSF1C target genes. RASSF1C down-regulates several pro-apoptotic and tumor suppressor genes and up-regulates several growth promoting genes in breast cancer cells. We further show that down-regulation of caspase 3 via overexpression of RASSF1C reduces breast cancer cells' sensitivity to the apoptosis inducing agent, etoposide. Furthermore, we found that RASSF1C over-expression enhances T47D cell invasion/migration in vitro. CONCLUSION: Together, our findings suggest that RASSF1C, unlike RASSF1A, is not a tumor suppressor, but instead may play a role in stimulating metastasis and survival in breast cancer cells.


Subject(s)
Apoptosis , Breast Neoplasms/pathology , Gene Expression Regulation, Neoplastic , Tumor Suppressor Proteins/physiology , Breast Neoplasms/metabolism , Cell Line, Tumor , Cell Movement , Gene Silencing , Humans , Neoplasm Invasiveness , Neoplasm Metastasis , Oligonucleotide Array Sequence Analysis , Plasmids/metabolism , Treatment Outcome , Tumor Suppressor Proteins/metabolism
8.
Am J Physiol Lung Cell Mol Physiol ; 291(6): L1185-90, 2006 Dec.
Article in English | MEDLINE | ID: mdl-16891396

ABSTRACT

Recently, the Ras association domain family 1 gene (RASSF1) has been identified as a Ras effector encoding two major mRNA forms, RASSF1A and RASSF1C, derived by alternative promoter selection and alternative mRNA splicing. RASSF1A is a tumor suppressor gene. However, the function of RASSF1C, both in normal and cancer cells, is still unknown. To learn more about the function of RASSF1C in human cancer cells, we tested the effect of silencing RASSF1C mRNA with small interfering RNA on lung cancer cells (NCI H1299) that express RASSF1C but not RASSF1A. Small interfering RNA specific for RASSF1C reduced RASSF1C mRNA levels compared with controls. This reduction in RASSF1C expression caused a significant decrease in lung cancer cell proliferation. Furthermore, overexpression of RASSF1C increased cell proliferation in lung cancer cells. Finally, we found that RASSF1C, unlike RASSF1A, does not upregulate N-cadherin 2 and transglutaminase 2 protein expression in NCI H1299 lung cancer cells. This suggests that RASSF1C and RASSF1A have different effector targets. Together, our findings suggest that RASSF1C, unlike RASSF1A, is not a tumor suppressor but rather stimulates lung cancer cell proliferation.


Subject(s)
Tumor Suppressor Proteins/genetics , Cell Division , Cell Line, Tumor , Gene Expression Regulation, Neoplastic , Humans , Lung Neoplasms , Plasmids , RNA, Messenger/genetics , Recombinant Fusion Proteins/metabolism , Suppression, Genetic , Tumor Suppressor Proteins/metabolism
9.
Am J Physiol Cell Physiol ; 290(3): C900-6, 2006 Mar.
Article in English | MEDLINE | ID: mdl-16269403

ABSTRACT

Insulin-like growth factor binding protein-5 (IGFBP5) is a multifunctional protein, which acts not only as a traditional binding protein, but also functions as a growth factor independent of IGFs to stimulate bone formation. It has been predicted that the intrinsic growth factor action of IGFBP5 involves binding of IGFBP5 to a putative receptor to induce downstream signaling pathways and/or nuclear translocation of IGFBP5 to influence transcription of genes involved in osteoblast cell proliferation/differentiation. Our study indentified proteins that bound to IGFBP5 using IGFBP5 as bait in a yeast two-hybrid screen of the U2 human osteosarcoma cell cDNA library. One of the clones that interacted strongly with the bait under high-stringency conditions corresponded to a novel IGFBP5 interacting protein (IGFBP5-IP) encoded by a gene that resides in mouse chromosome 10. The interaction between IGFBP5-IP and IGFBP5 is confirmed by in vitro coimmunoprecipitation studies that used pFlag and IGFBP5 polyclonal antibody, and cell lysates overexpressing both IGFBP5-IP and IGFBP5. Northern blot and RT-PCR analysis showed that the IGFBP-IP is expressed in both untransformed normal human osteoblasts and in osteosarcoma cell lines, which are known to produce IGFBP5. To determine the roles of IGFBP5-IP, we evaluated the effect of blocking the expression of IGFBP5-IP on osteoblast proliferation. We found that using a IGFBP5-IP-specific small interfering-hairpin plasmid resulted in a decrease in both basal and IGFBP5-induced osteoblast cell proliferation. On the basis of these findings, we predict that IGFBP5-IP may act as intracellular mediator of growth promoting actions of IGFBP5 and perhaps other osteoregulatory agents in bone cells.


Subject(s)
Carrier Proteins/metabolism , Osteoblasts/cytology , Osteoblasts/metabolism , Amino Acid Sequence , Base Sequence , Carrier Proteins/chemistry , Cell Proliferation , DNA, Complementary , Gene Expression Regulation , Gene Silencing , Humans , Molecular Sequence Data
10.
J Bone Miner Res ; 20(8): 1430-9, 2005 Aug.
Article in English | MEDLINE | ID: mdl-16007340

ABSTRACT

UNLABELLED: The goal of this study was to identify downstream signaling molecules involved in mediating the IGF-independent effects of IGFBP-5 in osteoblasts. We identified RASSF1C, a member of the RASSF1 gene products, as a IGFBP-5 binding partner and as a potential mediator of IGFBP-5 effects on ERK phosphorylation and cell proliferation. INTRODUCTION: It has been predicted that the intrinsic growth factor action of insulin-like growth factor binding protein (IGFBP)-5 involves either the binding of IGFBP-5 to a putative receptor to induce downstream signaling pathways and/or intracellular translocation of IGFBP-5 to bind to potential signaling molecules involved in osteoblast cell regulation. This study reports the characterization of isoform C of the Ras association family 1 (RASSF1C) gene as an interacting partner of IGFBP-5. MATERIALS AND METHODS: IGFBP-5 was used as bait in a yeast two-hybrid screen of a human osteosarcoma cDNA library. Expression levels of RASSF1C were measured by RT-PCR and/or Northern blot. IGFBP-5 effects on ERK phosphorylation were evaluated by immunoblot analysis. The effect of RASSF1C siRNA on cell proliferation was measured by the AlamarBlue assay. RESULTS: One of the clones that interacted strongly with the bait under high stringency conditions corresponded to RASSF1C. The interaction between RASSF1C and IGFBP-5 was confirmed by in vitro co-immunoprecipitation studies. Northern blot and RT-PCR analysis showed that RASSF1C was expressed in a variety of osteoblast cell types that produce IGFBP-5. Addition of synthetic RASSF1C-specific small interfering (si) RNA duplex or use of a RASSF1C-specific si-hairpin plasmid caused a decrease in cell number and abolished IGFBP-5-induced extracellular signal-regulated kinase (ERK)-1/2 phosphorylation but had no effect on IGFBP-5-induced increases in alkaline phosphatase (ALP) activity. CONCLUSIONS: We have shown a novel interaction between IGFBP-5 and RASSF1C. Our findings that silencing of RASSF1C results in the reduction of osteoblast cell proliferation and that IGFBP-5 treatment increases phosphorylation of ERK-1/2 raise the possibility that RASSF1C, a Ras effector, could, in part, contribute to mediating the effects of IGFBP-5 on ERK phosphorylation and, consequently, cell proliferation.


Subject(s)
Insulin-Like Growth Factor Binding Protein 5/metabolism , Osteoblasts/physiology , Tumor Suppressor Proteins/metabolism , Amino Acid Sequence , Bone and Bones/cytology , Bone and Bones/metabolism , Cell Proliferation , Cells, Cultured , Gene Silencing , Humans , Immunoprecipitation , Insulin-Like Growth Factor Binding Protein 5/genetics , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Molecular Sequence Data , Osteoblasts/chemistry , Osteoblasts/metabolism , Phosphorylation , Protein Isoforms/genetics , Protein Isoforms/metabolism , RNA, Small Interfering/genetics , Tumor Suppressor Proteins/analysis , Tumor Suppressor Proteins/genetics
11.
Biochemistry ; 41(51): 15394-403, 2002 Dec 24.
Article in English | MEDLINE | ID: mdl-12484779

ABSTRACT

IGF binding protein-5 (BP-5) is an important bone formation regulator. Therefore, elucidation of the identity of IGF binding protein-5 (BP-5) protease produced by osteoblasts is important for our understanding of the molecular pathways that control the action of BP-5. In this regard, BP-5 protease purified by various chromatographic steps from a conditioned medium of U2 human osteosarcoma cells migrated as a single major band, which comigrated with the protease activity in native PAGE and yielded multiple bands in SDS-PAGE under reducing conditions. N-Terminal sequencing of these bands revealed that three of the bands yielded amino acid sequences that were identical to that of alpha2 macroglobulin (alpha2M). Although alpha2M was produced by human osteoblasts (OBs), it was not found to be a BP-5 protease. Because alpha2M had been shown to complex with ADAM proteases and because ADAM-12 was found to cleave BP-3 and BP-5, we evaluated if one of the members of ADAM family was the BP-5 protease. On the basis of the findings that (1) purified preparations of BP-5 protease from U2 cell CM contained ADAM-9, (2) ADAM-9 is produced and secreted in high abundance by various human OB cell types, (3) purified ADAM-9 cleaved BP-5 effectively while it did not cleave other IGFBPs or did so with less potency, and (4) purified ADAM-9 bound to alpha2M, we conclude that ADAM-9 is a BP-5 protease produced by human OBs.


Subject(s)
Disintegrins , Insulin-Like Growth Factor Binding Protein 5/metabolism , Membrane Proteins/biosynthesis , Membrane Proteins/chemistry , Metalloendopeptidases/biosynthesis , Metalloendopeptidases/chemistry , Muscle Proteins/biosynthesis , Muscle Proteins/chemistry , Osteoblasts/enzymology , Osteoblasts/metabolism , ADAM Proteins , Cell Line, Transformed , Cells, Cultured , Electrophoresis, Polyacrylamide Gel , Humans , Membrane Proteins/isolation & purification , Membrane Proteins/metabolism , Metalloendopeptidases/isolation & purification , Metalloendopeptidases/metabolism , Muscle Proteins/isolation & purification , Muscle Proteins/metabolism , Peptide Fragments/chemistry , Peptide Fragments/isolation & purification , Protein Binding , Recombinant Proteins/biosynthesis , Recombinant Proteins/chemistry , Recombinant Proteins/isolation & purification , Sequence Analysis, Protein , Substrate Specificity , Tumor Cells, Cultured , alpha-Macroglobulins/biosynthesis , alpha-Macroglobulins/chemistry
12.
J Biol Chem ; 277(14): 12053-60, 2002 Apr 05.
Article in English | MEDLINE | ID: mdl-11821401

ABSTRACT

Recent studies using insulin-like growth factor I (IGF-I) knockout mice demonstrate that IGF-binding protein (IGFBP)-5, an important bone formation regulator, itself is a growth factor with cellular effects not dependent on IGFs. Because IGFBP-5 contains a nuclear localization sequence that mediates transport of IGFBP-5 into the nucleus, we propose that IGFBP-5 interacts with nuclear proteins to affect transcription of genes involved in bone formation. We therefore undertook studies to identify proteins that bind to IGFBP-5 using IGFBP-5 as bait in a yeast two-hybrid screen of a U2 human osteosarcoma cDNA library. Five related clones that interacted strongly with the bait corresponded to the FHL2 gene, which contains four and a half LIM domains. Co-immunoprecipitation studies with lysates from U2 cells overexpressing FHL2 and IGFBP-5 confirmed that interaction between IGFBP-5 and FHL2 occurs in whole cells. In vitro interaction studies revealed that purified FHL2 interacted with IGFBP-5 but not with IGFBP-3, -4, or -6. Northern blot analysis showed that FHL2 was strongly expressed in human osteoblasts. Nuclear localization of both FHL2 and IGFBP-5 was evident from Western immunoblot analysis and immunofluorescence. The role of FHL2 as an intracellular mediator of the effects of IGFBP-5 and other osteoregulatory agents in osteoblasts will need to be verified in future studies.


Subject(s)
Homeodomain Proteins/chemistry , Homeodomain Proteins/metabolism , Insulin-Like Growth Factor Binding Protein 5/chemistry , Insulin-Like Growth Factor Binding Protein 5/metabolism , Muscle Proteins , Transcription Factors , Active Transport, Cell Nucleus , Animals , Blotting, Northern , Blotting, Western , Cell Nucleus/metabolism , Cytoplasm/metabolism , DNA, Complementary/metabolism , Gene Library , Genetic Vectors , Humans , Insulin-Like Growth Factor Binding Protein 3/metabolism , Insulin-Like Growth Factor Binding Protein 4/metabolism , Insulin-Like Growth Factor Binding Protein 6/metabolism , LIM-Homeodomain Proteins , Mice , Microscopy, Fluorescence , Plasmids/metabolism , Precipitin Tests , Protein Binding , Protein Structure, Tertiary , Recombinant Proteins/metabolism , Retroviridae/genetics , Transfection , Tumor Cells, Cultured , Two-Hybrid System Techniques
SELECTION OF CITATIONS
SEARCH DETAIL
...