Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 113
Filter
2.
Neoplasia ; 21(10): 963-973, 2019 10.
Article in English | MEDLINE | ID: mdl-31437536

ABSTRACT

The transcriptional coactivator Amplified in Breast Cancer 1 (AIB1) plays a major role in the progression of hormone and HER2-dependent breast cancers but its role in triple negative breast cancer (TNBC) is undefined. Here, we report that established TNBC cell lines, as well as cells from a TNBC patient-derived xenograft (PDX) that survive chemotherapy treatment in vitro express lower levels of AIB1 protein. The surviving cell population has an impaired tube-formation phenotype when cultured onto basement membrane, a property shared with TNBC cells that survive shRNA-mediated depletion of AIB1 (AIB1LOW cells). DNA analysis by exome sequencing revealed that AIB1LOW cells represent a distinct subpopulation. Consistent with their in vitro phenotype AIB1LOW cells implanted orthotopically generated slower growing tumors with less capacity for pulmonary metastases. Gene expression analysis of cultured cells and tumors revealed that AIB1LOW cells display a distinct expression signature of genes in pro-inflammatory pathways, cell adhesion, proteolysis and tissue remodeling. Interestingly, the presence of this AIB1LOW expression signature in breast cancer specimens is associated with shorter disease free survival of chemotherapy treated patients. We concluded that TNBC cell lines contain heterogeneous populations with differential dependence on AIB1 and that the gene expression pattern of AIB1LOW cells may represent a signature indicative of poor response to chemotherapy in TNBC patients.


Subject(s)
Gene Expression Regulation, Neoplastic , Nuclear Receptor Coactivator 3/metabolism , Triple Negative Breast Neoplasms/genetics , Triple Negative Breast Neoplasms/metabolism , Animals , Cell Line, Tumor , Clonal Evolution/genetics , Disease Models, Animal , Female , Gene Expression Profiling , Heterografts , Humans , Mice , Phenotype , RNA, Small Interfering/genetics , Signal Transduction , Transcriptome , Triple Negative Breast Neoplasms/mortality , Triple Negative Breast Neoplasms/pathology , Exome Sequencing
3.
Oncogene ; 36(5): 593-605, 2017 02 02.
Article in English | MEDLINE | ID: mdl-27375028

ABSTRACT

Cancer cell vascular invasion is a crucial step in the malignant progression toward metastasis. Here we used a genome-wide RNA interference screen with E0771 mammary cancer cells to uncover drivers of endothelial monolayer invasion. We identified keratin-associated protein 5-5 (Krtap5-5) as a candidate. Krtap5-5 belongs to a large protein family that is implicated in crosslinking keratin intermediate filaments during hair formation, yet these Krtaps have no reported role in cancer. Depletion of Krtap5-5 from cancer cells led to cell blebbing and a loss of keratins 14 and 18, in addition to the upregulation of vimentin intermediate filaments. This intermediate filament subtype switching induced dysregulation of the actin cytoskeleton and reduced the expression of hemidesmosomal α6/ß4-integrins. We further demonstrate that knockdown of keratin 18 phenocopies the loss of Krtap5-5, suggesting that Krtap5-5 crosstalks with keratin 18 in E0771 cells. Disruption of the keratin cytoskeleton by perturbing Krtap5-5 function broadly altered the expression of cytoskeleton regulators and the localization of cell surface markers. Krtap5-5 depletion did not impact cell viability but reduced cell motility and extracellular matrix invasion, as well as extravasation of cancer cells into tissues in zebrafish and mice. We conclude that Krtap5-5 is a previously unknown regulator of cytoskeletal function in cancer cells that modulates motility and vascular invasion. Thus, in addition to its physiologic function, a Krtap can serve as a switch toward malignant progression.


Subject(s)
Cytoskeletal Proteins/metabolism , Cytoskeleton/metabolism , Mammary Neoplasms, Experimental/blood supply , Animals , Female , Human Umbilical Vein Endothelial Cells , Humans , Keratins/metabolism , Male , Mammary Neoplasms, Experimental/metabolism , Mammary Neoplasms, Experimental/pathology , Mice , Mice, Inbred C57BL , Neovascularization, Pathologic/metabolism , Neovascularization, Pathologic/pathology , Zebrafish
4.
Oncogene ; 34(48): 5879-89, 2015 Nov 26.
Article in English | MEDLINE | ID: mdl-25772246

ABSTRACT

Metastasis of cancer cells involves multiple steps, including their dissociation from the primary tumor and invasion through the endothelial cell barrier to enter the circulation and finding their way to distant organ sites where they extravasate and establish metastatic lesions. Deficient contact inhibition is a hallmark of invasive cancer cells, yet surprisingly the vascular invasiveness of commonly studied cancer cell lines is regulated by the density at which cells are propagated in culture. Cells grown at high density were less effective at invading an endothelial monolayer than cells grown at low density. This phenotypic difference was also observed in a zebrafish model of vascular invasion of cancer cells after injection into the yolk sac and extravasation of cancer cells into tissues from the vasculature. The vascular invasive phenotypes were reversible. A kinome-wide RNA interference screen was used to identify drivers of vascular invasion by panning small hairpin RNA (shRNA) library-transduced noninvasive cancer cell populations on endothelial monolayers. The selection of invasive subpopulations showed enrichment of shRNAs targeting the large tumor suppressor 1 (LATS1) kinase that inhibits the activity of the transcriptional coactivator yes-associated protein (YAP) in the Hippo pathway. Depletion of LATS1 from noninvasive cancer cells restored the invasive phenotype. Complementary to this, inhibition or depletion of YAP inhibited invasion in vitro and in vivo. The vascular invasive phenotype was associated with a YAP-dependent upregulation of the cytokines IL6, IL8 and C-X-C motif ligand 1, 2 and 3. Antibody blockade of cytokine receptors inhibited invasion and confirmed that they are rate-limiting drivers that promote cancer cell vascular invasiveness and could provide therapeutic targets.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Movement , Endothelium, Vascular/pathology , Gene Expression Regulation, Neoplastic , Phosphoproteins/metabolism , Protein Serine-Threonine Kinases/metabolism , Receptors, Interleukin-8B/metabolism , Adaptor Proteins, Signal Transducing/genetics , Animals , Apoptosis , Blotting, Western , Breast Neoplasms/genetics , Cell Cycle , Cell Proliferation , Cytokines/genetics , Cytokines/metabolism , Endothelium, Vascular/metabolism , Female , Human Umbilical Vein Endothelial Cells , Humans , Mice , Mice, Nude , Neoplasm Invasiveness , Phosphoproteins/genetics , Phosphorylation , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Serine-Threonine Kinases/genetics , RNA Interference , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Receptors, Interleukin-8B/genetics , Reverse Transcriptase Polymerase Chain Reaction , Transcription Factors , Tumor Cells, Cultured , Xenograft Model Antitumor Assays , YAP-Signaling Proteins , Zebrafish
5.
Oncogene ; 33(23): 3033-42, 2014 Jun 05.
Article in English | MEDLINE | ID: mdl-23851504

ABSTRACT

The key molecular events required for the formation of ductal carcinoma in situ (DCIS) and its progression to invasive breast carcinoma have not been defined. Here, we show that the nuclear receptor coactivator amplified in breast cancer 1 (AIB1) is expressed at low levels in normal breast but is highly expressed in DCIS lesions. This is of significance since reduction of AIB1 in human MCFDCIS cells restored a more normal three-dimensional mammary acinar structure. Reduction of AIB1 in MCFDCIS cells, both before DCIS development or in existing MCFDCIS lesions in vivo, inhibited tumor growth and led to smaller, necrotic lesions. AIB1 reduction in MCFDCIS cells was correlated with significant reduction in the CD24-/CD44+ breast cancer-initiating cell (BCIC) population, and a decrease in myoepithelial progenitor cells in the DCIS lesions in vitro and in vivo. The loss of AIB1 in MCFDCIS cells was also accompanied by a loss of expression of NOTCH 2, 3 and 4, JAG2, HES1, GATA3, human epidermal growth factor receptor 2 (HER2) and HER3 in vivo. These signaling molecules have been associated with differentiation of breast epithelial progenitor cells. These data indicate that AIB1 has a central role in the initiation and maintenance of DCIS and that reduction of AIB1 causes loss of BCIC, loss of components of the NOTCH, HER2 and HER3 signaling pathways and fewer DCIS myoepithelial progenitor cells in vivo. We propose that increased expression of AIB1, through the maintenance of BCIC, facilitates formation of DCIS, a necessary step before development of invasive disease.


Subject(s)
Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Carcinoma, Intraductal, Noninfiltrating/metabolism , Carcinoma, Intraductal, Noninfiltrating/pathology , Neoplastic Stem Cells/physiology , Nuclear Receptor Coactivator 3/metabolism , Animals , Cell Differentiation/genetics , Female , Gene Expression Regulation, Neoplastic , Humans , Mammary Neoplasms, Experimental , Mice , Mice, Nude , Neoplastic Stem Cells/pathology , Nuclear Receptor Coactivator 3/antagonists & inhibitors , Nuclear Receptor Coactivator 3/genetics , RNA, Small Interfering/pharmacology , Signal Transduction/genetics , Xenograft Model Antitumor Assays
7.
Oncogene ; 28(37): 3296-306, 2009 Sep 17.
Article in English | MEDLINE | ID: mdl-19633684

ABSTRACT

The tyrosine kinase receptor anaplastic lymphoma kinase (ALK) and its ligand, the growth factor pleiotrophin (PTN), are highly expressed during the development of the nervous system and have been implicated in the malignant progression of different tumor types. Here, we describe human single-chain variable fragment (scFv) antibodies that target the ligand-binding domain (LBD) in ALK and show the effect in vitro and in vivo. The ALK LBD was used as a bait in a yeast two-hybdrid system to select human scFv from a library with randomized complementarity-determining region 3 domains. Surface plasmon resonance showed high-affinity binding of the selected scFv. The anti-ALK scFv competed for binding of PTN to ALK in intact cells and inhibited PTN-dependent signal transduction through endogenous ALK. Invasion of an intact endothelial cell monolayer by U87MG human glioblastoma cells was inhibited by the anti-ALK scFv. In addition, the growth of established tumor xenografts in mice was reversed after the induction of the conditional expression of the anti-ALK scFv. In archival malignant brain tumors expression levels of ALK and PTN were found elevated and appear correlated with poor patient survival. This suggests a rate-limiting function of the PTN/ALK interaction that may be exploited therapeutically.


Subject(s)
Antibodies/immunology , Protein-Tyrosine Kinases/chemistry , Protein-Tyrosine Kinases/immunology , Amino Acid Sequence , Anaplastic Lymphoma Kinase , Animals , Bacteria/cytology , Bacteria/immunology , Binding, Competitive , Brain Neoplasms/genetics , Brain Neoplasms/immunology , Brain Neoplasms/pathology , Carrier Proteins/genetics , Carrier Proteins/metabolism , Cell Line, Tumor , Cell Proliferation , Computational Biology , Cytokines/genetics , Cytokines/metabolism , Endothelial Cells/pathology , Epitopes/immunology , Gene Expression Regulation, Neoplastic , Glioblastoma/pathology , Humans , Immunoglobulin Variable Region/immunology , Ligands , Mice , Midkine , Models, Molecular , Molecular Sequence Data , Protein Binding/immunology , Protein Structure, Tertiary , Protein Tyrosine Phosphatases/genetics , Protein Tyrosine Phosphatases/metabolism , Protein-Tyrosine Kinases/genetics , Protein-Tyrosine Kinases/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptor Protein-Tyrosine Kinases , Signal Transduction/immunology
8.
Oncogene ; 26(6): 859-69, 2007 Feb 08.
Article in English | MEDLINE | ID: mdl-16878150

ABSTRACT

Anaplastic lymphoma kinase (ALK) is a transmembrane receptor tyrosine kinase in the insulin receptor superfamily. We recently demonstrated that the growth factors pleiotrophin (PTN) and midkine (MK) are ligands for ALK and that upon ALK activation, insulin receptor substrate-1 (IRS-1) and other substrates are phosphorylated. Here, the role of IRS-1 in ligand-mediated ALK signaling is investigated in interleukin-3 (IL-3)-dependent 32D murine myeloid cells. These cells do not express ALK and IRS family members, and do not respond to exogenously added PTN or MK. We show that expression of ALK plus IRS-1 renders these cells independent of IL-3 owing to the activation of ALK by endogenous MK. Mutational analysis reveals that this transformed phenotype of 32D cells requires kinase-active ALK as well as the interaction of ALK with IRS-1. Furthermore, 32D/IRS-1/ALK cells display an enhanced activation of mitogen-activated protein kinase and PI3-kinase pathways, and a selective transcriptional activation of nuclear factor (NF)-kappaB. Small interfering RNA-mediated knockdown of the endogenous MK or p65/NF-kappaB revealed that both these are rate limiting for the transformed phenotype induced by ALK plus IRS-1. We conclude that the recruitment of IRS-1 to activated ALK and the activation of NF-kappaB are essential for the autocrine growth and survival signaling of MK.


Subject(s)
Cytokines/metabolism , NF-kappa B/metabolism , Phosphoproteins/metabolism , Protein-Tyrosine Kinases/metabolism , Signal Transduction , Anaplastic Lymphoma Kinase , Animals , Binding Sites , Cell Line , Cell Proliferation , Chlorocebus aethiops , Enzyme Activation , Humans , Insulin Receptor Substrate Proteins , Mice , Midkine , Mitogen-Activated Protein Kinases/metabolism , NF-kappa B/genetics , Phenotype , Phosphoproteins/genetics , Protein Subunits/genetics , Protein Subunits/metabolism , Protein-Tyrosine Kinases/genetics , Proto-Oncogene Proteins c-akt/metabolism , RNA, Small Interfering/genetics , Receptor Protein-Tyrosine Kinases/genetics , Receptor Protein-Tyrosine Kinases/metabolism , Transcription, Genetic/genetics
9.
Histol Histopathol ; 20(1): 225-37, 2005 01.
Article in English | MEDLINE | ID: mdl-15578441

ABSTRACT

Altered expression of genes in diseased tissues can prognosticate a distinct natural progression of the disease as well as predict sensitivity or resistance to particular therapies. Archival tissues from patients with a known medical history and treatments are an invaluable resource to validate the utility of candidate genes for prognosis and prediction of therapy outcomes. However, stored tissues with associated long-term follow-up information typically are formalin-fixed, paraffin-embedded specimen and this can severely restrict the methods applicable for gene expression analysis. We report here on the utility of tissue microarrays (TMAs) that use valuable tissues sparingly and provide a platform for simultaneous analysis of gene expression in several hundred samples. In particular, we describe a stable method applicable to mRNA expression screening in such archival tissues. TMAs are constructed from sections of small drill cores, taken from tissue blocks of archival tissues and multiple samples can thus be arranged on a single microscope slide. We used mRNA in situ hybridization (ISH) on >500 full sections and >100 TMAs for >10 different cDNAs that yielded >10,000 data points. We provide detailed experimental protocols that can be implemented without major hurdles in a molecular pathology laboratory and discuss quantitative analysis and the advantages and limitations of ISH. We conclude that gene expression analysis in archival tissues by ISH is reliable and particularly useful when no protein detection methods are available for a candidate gene.


Subject(s)
Gene Expression Profiling , In Situ Hybridization , RNA, Messenger , Tissue Array Analysis , Animals , Humans
10.
Gene Ther ; 12(4): 339-46, 2005 Feb.
Article in English | MEDLINE | ID: mdl-15496960

ABSTRACT

The growth and metastasis of solid tumors relies on the activities of polypeptide growth factors to recruit stromal tissue and expand the tumor mass. Pleiotrophin (PTN) is a secreted growth factor with angiogenic activity that has been found to contribute to the growth and metastasis of tumors including melanoma. Here, we present a gene therapy approach of targeting PTN in established tumors using ribozymes. Tetracycline-regulated ribozyme expression vectors were used to deplete conditionally PTN mRNA from melanoma xenograft tumors in vivo. We found that tetracycline-mediated initiation of ribozyme expression in established tumors reduced further tumor growth. Next, we generated synthetic anti-PTN ribozymes that inhibit PTN-dependent colony formation of cells in soft agar. Intraperitoneal administration of these synthetic ribozymes into nude mice inhibited growth of PTN-positive, subcutaneous melanoma. Furthermore, PTN released from the tumors into the circulation of mice was reduced after ribozyme treatment. These data show that ribozyme targeting of rate-limiting tumor growth factors could provide an efficient tool for cancer therapy and that the efficacy may be reflected in the reduction of the serum levels of the targeted protein, PTN.


Subject(s)
Carrier Proteins/genetics , Cytokines/genetics , Genetic Therapy/methods , Melanoma/therapy , RNA, Messenger/genetics , Skin Neoplasms/therapy , Animals , Gene Expression/drug effects , Genetic Vectors/genetics , Humans , Melanoma/blood supply , Mice , Mice, Nude , Neovascularization, Pathologic/prevention & control , RNA, Catalytic , Skin Neoplasms/blood supply , Tetracycline/therapeutic use , Transfection/methods
11.
Br J Cancer ; 86(6): 858-63, 2002 Mar 18.
Article in English | MEDLINE | ID: mdl-11953815

ABSTRACT

Pleiotrophin is a heparin-binding growth factor involved in the differentiation and proliferation of neuronal tissue during embryogenesis, and also secreted by melanoma and breast carcinoma cells. Pleiotrophin exhibits mitogenic and angiogenic properties and has been shown to influence the vascular supply, expansion and metastasis of tumour cells. Our aim was to study the serum and plasma concentrations of pleiotrophin and the classical angiogenic growth factor vascular endothelial growth factor. Using a specific ELISA-test we studied patients with small cell lung cancer (n=63), and patients with non-small cell lung cancer (n=22) in comparison to healthy control subjects (n=41). In most of the lung cancer patients (81%), we found serum levels of pleiotrophin above those of control subjects (P<0.001). Of the 63 small cell lung cancer patients in the study pleiotrophin serum levels were elevated in 55 cases (87%) and in 14 cases (63%) of the 22 non-small cell lung cancer patients. Pleiotrophin mean serum concentrations were 10.8-fold higher in the tumour patient group as compared to the control group (P<0.001). Furthermore, pleiotrophin serum levels correlated positively with the stage of disease and inversely with the response to therapy. Plasma vascular endothelial growth factor concentrations were elevated in only in 28.6% of small cell lung cancer and 45.5% of non-small cell lung cancer patients by an average of 2.3-fold. Quite strikingly, there was no apparent correlation between the plasma vascular endothelial growth factor concentration and the stage of disease. Our study suggests that pleiotrophin may be an early indicator of lung cancer and might be of use in monitoring the efficacy of therapy, which needs to be confirmed by larger studies.


Subject(s)
Carrier Proteins/blood , Cytokines/blood , Lung Neoplasms/blood , Carcinoma, Non-Small-Cell Lung/blood , Carcinoma, Small Cell/blood , Carrier Proteins/genetics , Cytokines/genetics , Endothelial Growth Factors/blood , Female , Humans , Lung Neoplasms/pathology , Lymphokines/blood , Male , Middle Aged , Neoplasm Staging , RNA, Messenger/analysis , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors
12.
Breast Cancer Res Treat ; 68(1): 21-8, 2001 Jul.
Article in English | MEDLINE | ID: mdl-11678305

ABSTRACT

The gene of the nuclear receptor coactivator AIB1 (amplified in breast cancer 1) is amplified in breast cancer cell lines as well as in breast tumor tissues. AIB1 mRNA is often highly expressed (>60%) in primary breast tumors and it has been shown that AIB1 enhances estrogen and progesterone dependent transcription in vitro. Therefore, it has been postulated that AIB1 contributes to the development of breast cancer. However, to date, it has not been shown that AIB1 amplification and overexpression correlates with elevated protein levels in breast cancer tissues. In this study we analyzed protein levels of AIB1 in normal and breast tumor tissues by immunohistochemistry. We compared 41 human breast tumor tissues with 24 normal breast tissue samples and found that AIB1 stained in the nuclei of approximately 46% of the tumors and 30% of the normal tissues. Overall, AIB1 protein levels were significantly higher in tumor tissue than in normal tissue and the highest levels of nuclear staining were found exclusively in breast tumor tissues in 9.8% of the cases. These data suggest that increased AIB1 mRNA expression does not always translate into elevated protein levels and that AIB1 most likely will be relevant to the etiology of a subset of about 10% of breast carcinomas.


Subject(s)
Breast Neoplasms/metabolism , Breast/metabolism , Gene Expression Regulation, Neoplastic , RNA, Messenger/metabolism , Transcription Factors/metabolism , Adult , Aged , Blotting, Northern , Breast/cytology , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Carcinoma, Ductal, Breast/genetics , Carcinoma, Ductal, Breast/metabolism , Carcinoma, Ductal, Breast/pathology , Carcinoma, Lobular/genetics , Carcinoma, Lobular/metabolism , Carcinoma, Lobular/pathology , Case-Control Studies , Cell Line , Female , Humans , Immunohistochemistry , In Situ Hybridization , Middle Aged , Neoplasms, Hormone-Dependent/genetics , Neoplasms, Hormone-Dependent/metabolism , Neoplasms, Hormone-Dependent/pathology , Nuclear Receptor Coactivator 3 , Transcription Factors/genetics , Tumor Cells, Cultured
13.
J Biol Chem ; 276(43): 39736-41, 2001 Oct 26.
Article in English | MEDLINE | ID: mdl-11502741

ABSTRACT

The AIB1 (amplified in breast cancer 1) protein is a coactivator that potentiates the transcriptional activity of nuclear hormone receptors, and its gene is amplified in a subset of human breast cancers. Here we report a splice variant of AIB1 mRNA that lacks the exon 3 sequence. We determined that the AIB-Delta3 mRNA encoded a 130-kDa protein that lacks the NH(2)-terminal basic helix-loop-helix and a portion of the PAS (Per-Arnt-Sim homology) dimerization domain. The 130-kDa protein was detected in MCF-7 breast cancer cells at levels that were 5-10% of the full-length protein, whereas in non-transformed mammary epithelium lines, the AIB-Delta3 protein was present at significantly lower levels compared with the full-length AIB1. Consistent with this finding, the abundance of AIB1-Delta3 mRNA was increased in human breast cancer specimens relative to that in normal breast tissue. To determine whether there were phenotypic changes associated with the overexpression of the AIB-Delta3 isoform, we performed functional reporter gene assays. These revealed that the ability of AIB1-Delta3 to promote transcription mediated by the estrogen or progesterone receptors was significantly greater than that of the full-length protein. Surprisingly, the AIB1-Delta3 isoform was also more effective than AIB1 in promoting transcription induced by epidermal growth factor. Overexpression of AIB1-Delta3 may thus play an important role in sensitizing breast tumor cells to hormone or growth factor stimulation.


Subject(s)
Acetyltransferases/genetics , Alternative Splicing , Breast Neoplasms/genetics , Epidermal Growth Factor/pharmacology , Estrogens/pharmacology , Progesterone/pharmacology , Saccharomyces cerevisiae Proteins , Acetyltransferases/metabolism , Dose-Response Relationship, Drug , Estrogen Receptor alpha , Female , Histone Acetyltransferases , Humans , Protein Biosynthesis , Protein Isoforms , RNA, Messenger/genetics , RNA, Messenger/isolation & purification , Receptors, Estrogen/metabolism , Receptors, Progesterone/metabolism , Signal Transduction
14.
J Biol Chem ; 276(43): 40247-53, 2001 Oct 26.
Article in English | MEDLINE | ID: mdl-11509569

ABSTRACT

Fibroblast growth factor-binding protein (FGF-BP) 1 is a secreted protein that can bind fibroblast growth factors (FGFs) 1 and 2. These FGFs are typically stored on heparan sulfate proteoglycans in the extracellular matrix in an inactive form, and it has been proposed that FGF-BP1 functions as a chaperone molecule that can mobilize locally stored FGF and present the growth factor to its tyrosine kinase receptor. FGF-BP1 is up-regulated in squamous cell, colon, and breast cancers and can act as an angiogenic switch during malignant progression of epithelial cells. For the present studies, we focused on FGF-1 and -2 and investigated interactions with recombinant human FGF-BP1 protein as well as effects on signal transduction, cell proliferation, and angiogenesis. We show that recombinant FGF-BP1 specifically binds FGF-2 and that this binding is inhibited by FGF-1, heparan sulfate, and heparinoids. Furthermore, FGF-BP1 enhances FGF-1- and FGF-2-dependent proliferation of NIH-3T3 fibroblasts and FGF-2-induced extracellular signal-regulated kinase 2 phosphorylation. Finally, in the chicken chorioallantoic membrane angiogenesis assay, FGF-BP1 synergizes with exogenously added FGF-2. We conclude that FGF-BP1 binds directly to FGF-1 and FGF-2 and positively modulates the biological activities of these growth factors.


Subject(s)
Carrier Proteins/metabolism , Fibroblast Growth Factors/metabolism , 3T3 Cells , Animals , Carrier Proteins/genetics , Carrier Proteins/pharmacology , Drug Synergism , Enzyme Activation , Fibroblast Growth Factor 1/metabolism , Fibroblast Growth Factor 1/pharmacology , Fibroblast Growth Factor 2/metabolism , Fibroblast Growth Factor 2/pharmacology , Fibroblast Growth Factors/pharmacology , Humans , Intercellular Signaling Peptides and Proteins , Intracellular Signaling Peptides and Proteins , Mice , Mitogen-Activated Protein Kinase 1/metabolism , Mitogens/metabolism , Mitogens/pharmacology , Molecular Sequence Data , Neovascularization, Physiologic/drug effects , Protein Binding , Recombinant Proteins/metabolism , Recombinant Proteins/pharmacology
15.
J Biol Chem ; 276(26): 23763-8, 2001 Jun 29.
Article in English | MEDLINE | ID: mdl-11328819

ABSTRACT

Human breast tumorigenesis is promoted by the estrogen receptor pathway, and nuclear receptor coactivators are thought to participate in this process. Here we studied whether one of these coactivators, AIB1 (amplified in breast cancer 1), was rate-limiting for hormone-dependent growth of human MCF-7 breast cancer cells. We developed MCF-7 breast cancer cell lines in which the expression of AIB1 can be modulated by regulatable ribozymes directed against AIB1 mRNA. We found that depletion of endogenous AIB1 levels reduced steroid hormone signaling via the estrogen receptor alpha or progesterone receptor beta on transiently transfected reporter templates. Down-regulation of AIB1 levels in MCF-7 cells did not affect estrogen-stimulated cell cycle progression but reduced estrogen-mediated inhibition of apoptosis and cell growth. Finally, upon reduction of endogenous AIB1 expression, estrogen-dependent colony formation in soft agar and tumor growth of MCF-7 cells in nude mice was decreased. From these findings we conclude that, despite the presence of different estrogen receptor coactivators in breast cancer cells, AIB1 exerts a rate-limiting role for hormone-dependent human breast tumor growth.


Subject(s)
Breast Neoplasms/etiology , Estrogens/pharmacology , RNA, Catalytic/genetics , Transcription Factors/physiology , Animals , Apoptosis , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Division , Down-Regulation , Female , Humans , Mice , Mice, Nude , Nuclear Receptor Coactivator 3 , Progesterone/pharmacology , RNA, Messenger/metabolism , Transcription Factors/antagonists & inhibitors , Transcription Factors/genetics , Transcriptional Activation , Transfection , Tumor Cells, Cultured
16.
Oncogene ; 20(14): 1730-8, 2001 Mar 29.
Article in English | MEDLINE | ID: mdl-11313920

ABSTRACT

The fibroblast growth factor-binding protein (FGF-BP) modulates FGF activity through binding and release from the extracellular matrix. Consequently, the expression of FGF-BP in certain tumor types is a rate-limiting regulator of FGF-mediated angiogenesis. FGF-BP is upregulated in squamous cell carcinoma by treatment with mitogens such as EGF or TPA. In this study, we investigated the regulation of FGF-BP gene expression by serum. Treatment of serum-starved ME-180 cells with fetal bovine serum (FBS) resulted in a rapid increase in steady-state levels of FGF-BP mRNA and in the rate of FGF-BP gene transcription. Serum induction of FGF-BP mRNA was not mediated through EGF receptor activation but was dependent on PKC, as well as ERK kinase (MEK) and p38 MAP kinase activation. Promoter analysis showed that C/EBP is the main promoter element required for the serum response. Unlike EGF-activation of FGF-BP, transcriptional induction by serum is not significantly regulated through the AP-1 or E-box sites in the promoter. These results illustrate differences between the mechanism of induction in response to serum and EGF.


Subject(s)
Blood Physiological Phenomena , CCAAT-Enhancer-Binding Proteins/physiology , Endopeptidases/genetics , Mitogen-Activated Protein Kinases/physiology , Transcription, Genetic , Cells, Cultured , Enzyme Activation , Epidermal Growth Factor/pharmacology , ErbB Receptors/physiology , Gene Expression Regulation , Humans , Phosphatidylinositol 3-Kinases/physiology , Promoter Regions, Genetic , Protein Kinase C/physiology , Transcription Factor AP-1/physiology , p38 Mitogen-Activated Protein Kinases
17.
Kidney Int ; 59(5): 1717-28, 2001 May.
Article in English | MEDLINE | ID: mdl-11318942

ABSTRACT

BACKGROUND: Basic fibroblast growth factor (bFGF) is an angiogenic growth factor that is involved in renal growth and the pathogenesis of renal diseases. We have detected high levels of bFGF accumulated in the kidney of HIV-transgenic mice and in children with HIV-associated renal diseases and the hemolytic uremic syndrome (HUS). However, the mechanism modulating the activity of bFGF under these circumstances is poorly understood. We carried out experiments to determine whether a secreted binding protein (FGF-BP) that modulates the activity of bFGF during the process of tumor growth was expressed in pediatric kidneys and to define whether the expression of FGF-BP was altered in pediatric renal diseases associated with high levels of bFGF. METHODS: Immunohistochemistry and in situ hybridization studies were done in 41 renal sections from children with HIV nephropathies, HUS, other pediatric renal diseases, controls, and fetal kidneys. Western blots and reverse transcriptase-polymerase chain reaction studies were done in selected urine samples and cultured renal cells. Recombinant FGF-BP was produced to study the mitogenic activity of FGF-BP in cultured human renal proximal tubular epithelial cells (RPTEcs). RESULTS: The expression of FGF-BP was up-regulated predominately in renal tubular epithelial cells in children with renal tubular injury, HIV-associated nephropathy (HIVAN), and HUS, and FGF-BP was secreted in the urine of these patients. FGF-BP was also abundantly expressed in developing fetal renal tubules. Recombinant FGF-BP enhanced the mitogenic effects of bFGF in cultured human RPTEcs. CONCLUSIONS: The localization of FGF-BP in renal tubular epithelial cells could provide a mechanism by which the activity of bFGF is modulated in developing and regenerating renal tubules of children.


Subject(s)
Carrier Proteins/metabolism , Kidney Diseases/metabolism , AIDS-Associated Nephropathy/metabolism , Animals , Base Sequence , Carrier Proteins/genetics , Carrier Proteins/pharmacology , Case-Control Studies , Cells, Cultured , Child , DNA Primers/genetics , Fetus/metabolism , Fibroblast Growth Factor 2/metabolism , Hemolytic-Uremic Syndrome/genetics , Hemolytic-Uremic Syndrome/metabolism , Humans , Immunohistochemistry , In Situ Hybridization , Intercellular Signaling Peptides and Proteins , Intracellular Signaling Peptides and Proteins , Kidney Diseases/genetics , Kidney Diseases/pathology , Kidney Tubules, Proximal/cytology , Kidney Tubules, Proximal/drug effects , Kidney Tubules, Proximal/metabolism , Mice , Models, Biological , RNA, Messenger/genetics , RNA, Messenger/metabolism , Recombinant Proteins/pharmacology , Thymidine/metabolism , Up-Regulation
18.
Int J Cancer ; 92(4): 510-7, 2001 May 15.
Article in English | MEDLINE | ID: mdl-11304685

ABSTRACT

Fibroblast growth factors FGF-1 (aFGF) and FGF-2 (bFGF) are found in most embryonic and adult normal and tumor tissues, where they are immobilized in the extracellular matrix (ECM). Mobilization of these FGFs is part of a tightly controlled process resulting in the activation of high-affinity FGF receptors. Recently, we have shown that a secreted FGF-binding protein (FGF-BP) binds non-covalently to FGF-2 and is able to release it from the ECM. This process of growth factor bioactivation seems to play a pivotal role in the growth of squamous cell carcinomas, especially through induction of tumor angiogenesis. Since previous studies provided only indirect evidence for the proposed mechanism of FGF-BP-mediated FGF-2 release, we decided to use recombinant purified FGF-BP to study further the underlying mechanism of FGF-BP action. Here we show that FGF-BP is able to bind directly to FGF-2 without additional cofactors and to exhibit bioactivity. The purified recombinant FGF-BP stimulates tumor cell growth as well as endothelial cell growth and chemotaxis, indicating a dual growth-supporting role of FGF-BP in tumors. We show that this paracrine FGF-BP effect is dependent on endogenously expressed FGF-2, since it can be completely blocked by anti-FGF-2 antibodies. In tumor xenografts and in tumor cells, we detected a pattern of specific FGF-BP-immunoreactive high molecular weight forms, which presumably represent stable covalent complexes of FGF-BP and show marked differences in their occurrence in different tumors and in their heparin binding affinity. By providing further insight into the mechanism of FGF-BP action, our results emphasize the relevance of FGF-BP and of FGF-2 in tumor growth.


Subject(s)
Carrier Proteins/metabolism , Endothelium/metabolism , Fibroblast Growth Factor 2/metabolism , Adrenal Gland Neoplasms/metabolism , Blotting, Western , Carcinoma, Squamous Cell/metabolism , Cell Division , Cell Movement , Cells, Cultured , Chemotaxis , Dose-Response Relationship, Drug , Endothelium, Vascular/cytology , Extracellular Matrix/metabolism , Glycosylation , Heparin/metabolism , Humans , Intercellular Signaling Peptides and Proteins , Male , Mass Spectrometry , Neoplasm Transplantation , Prostatic Neoplasms/metabolism , Protein Binding , Recombinant Proteins/metabolism , Sodium Chloride/pharmacology , Tumor Cells, Cultured , Umbilical Cord/cytology
19.
J Biol Chem ; 276(20): 16772-9, 2001 May 18.
Article in English | MEDLINE | ID: mdl-11278720

ABSTRACT

Pleiotrophin (PTN) is a secreted growth factor that induces neurite outgrowth and is mitogenic for fibroblasts, epithelial, and endothelial cells. During tumor growth PTN can serve as an angiogenic factor and drive tumor invasion and metastasis. To identify a receptor for PTN, we panned a phage display human cDNA library against immobilized PTN protein as a bait. From this we isolated a phage insert that was homologous to an amino acid sequence stretch in the extracellular domain (ECD) of the orphan receptor tyrosine kinase anaplastic lymphoma kinase (ALK). In parallel with PTN, ALK is highly expressed during perinatal development of the nervous system and down-modulated in the adult. Here we show in cell-free assays as well as in radioligand receptor binding studies in intact cells that PTN binds to the ALK ECD with an apparent Kd of 32 +/- 9 pm. This receptor binding is inhibited by an excess of PTN, by the ALK ECD, and by anti-PTN and anti-ECD antibodies. PTN added to ALK-expressing cells induces phosphorylation of both ALK and of the downstream effector molecules IRS-1, Shc, phospholipase C-gamma, and phosphatidylinositol 3-kinase. Furthermore, the growth stimulatory effect of PTN on different cell lines in culture coincides with the endogenous expression of ALK mRNA, and the effect of PTN is enhanced by ALK overexpression. From this we conclude that ALK is a receptor that transduces PTN-mediated signals and propose that the PTN-ALK axis can play a significant role during development and during disease processes.


Subject(s)
Carrier Proteins/metabolism , Cytokines/metabolism , Protein-Tyrosine Kinases/genetics , Protein-Tyrosine Kinases/metabolism , Adrenal Gland Neoplasms , Anaplastic Lymphoma Kinase , Animals , Base Sequence , Binding Sites , Brain/enzymology , Cell Division , Cell-Free System , Cloning, Molecular , Gene Library , Growth Substances/metabolism , Humans , Kinetics , Mice , Molecular Sequence Data , RNA, Messenger/genetics , Receptor Protein-Tyrosine Kinases , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Transcription, Genetic , Transfection , Tumor Cells, Cultured
20.
Oncol Rep ; 8(1): 161-4, 2001.
Article in English | MEDLINE | ID: mdl-11115590

ABSTRACT

The growth of supportive tissue during the progression of solid tumors is an absolute requirement for the nourishment of the tumor. The blockade of this proliferative response of normal tissues to the growing tumor should hence inhibit tumor progression. We have shown earlier, that the heparinoid pentosan polysulfate (PPS) can block tumor growth and neoangiogenesis induced by Kaposi's FGF as well as by other heparin-binding growth factors (HBGFs). We now report on the effects of a bacterial polysaccharide, tecogalan, on tumor xenografts of human breast cancer cells. Tecogalan inhibited FGF-dependent SW-13 cells in vitro very similarly to PPS. Growth factor-independent MDA-MB 231 cells were used in animal studies to assess the in vivo potential of tecogalan. Subcutaneous growth of tumors was inhibited by once weekly i.v. administration of tecogalan. PPS single weekly administration showed a similar effect. No gross side effects were observed. Based on our previous studies with these models, we conclude, that tecogalan acts by blocking HBGFs released from tumor cells. Interestingly, single weekly dosing of either PPS or tecogalan appears to be strikingly more efficacious than spreading the dose over several administrations. These findings with a novel compound, tecogalan, and a novel treatment regimen, PPS, suggests a different approach to planning of therapies with these types of drugs.


Subject(s)
Adenocarcinoma/pathology , Antineoplastic Agents/therapeutic use , Breast Neoplasms/pathology , Polysaccharides, Bacterial/therapeutic use , Adrenal Gland Neoplasms/pathology , Animals , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/pharmacology , Carcinoma/pathology , Drug Administration Schedule , Female , Humans , Mice , Mice, Nude , Pentosan Sulfuric Polyester/administration & dosage , Pentosan Sulfuric Polyester/pharmacology , Pentosan Sulfuric Polyester/therapeutic use , Polysaccharides, Bacterial/administration & dosage , Polysaccharides, Bacterial/pharmacology , Tumor Cells, Cultured/transplantation , Xenograft Model Antitumor Assays
SELECTION OF CITATIONS
SEARCH DETAIL
...