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










Publication year range
1.
PLoS One ; 5(3): e9646, 2010 Mar 19.
Article in English | MEDLINE | ID: mdl-20333297

ABSTRACT

The AKT/PKB kinase is a key signaling component of one of the most frequently activated pathways in cancer and is a major target of cancer drug development. Most studies have focused on its activation by Receptor Tyrosine Kinase (RTK) mediated Phosphatidylinositol-3-OH kinase (PI3K) activation or loss of Phosphatase and Tensin homolog (PTEN). We have uncovered that growth factors binding to RTKs lead to activation of a non-receptor tyrosine kinase, Ack1 (also known as ACK or TNK2), which directly phosphorylates AKT at an evolutionarily conserved tyrosine 176 in the kinase domain. Tyr176-phosphorylated AKT localizes to the plasma membrane and promotes Thr308/Ser473-phosphorylation leading to AKT activation. Mice expressing activated Ack1 specifically in the prostate exhibit AKT Tyr176-phosphorylation and develop murine prostatic intraepithelial neoplasia (mPINs). Further, expression levels of Tyr176-phosphorylated-AKT and Tyr284-phosphorylated-Ack1 were positively correlated with the severity of disease progression, and inversely correlated with the survival of breast cancer patients. Thus, RTK/Ack1/AKT pathway provides a novel target for drug discovery.


Subject(s)
Breast Neoplasms/pathology , Protein-Tyrosine Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Tyrosine/chemistry , Animals , Cell Membrane/metabolism , Disease Progression , Female , Humans , Male , Mice , Mice, Transgenic , Phosphorylation , Prostatic Intraepithelial Neoplasia/pathology , Protein Structure, Tertiary
2.
Mol Cell Biol ; 29(18): 4935-48, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19596786

ABSTRACT

Data concerning the prognostic value of ErbB4 in breast cancer and effects on cell growth have varied in published reports, perhaps due to the unknown signaling consequences of expression of the intracellular proteolytic ErbB4 s80(HER4) fragment or due to differing signaling capabilities of alternatively spliced ErbB4 isoforms. One isoform (Cyt1) contains a 16-residue intracellular sequence that is absent from the other (Cyt2). We expressed s80(Cyt1) and s80(Cyt2) in HC11 mammary epithelial cells, finding diametrically opposed effects on the growth and organization of colonies in three-dimensional matrices. Whereas expression of s80(Cyt1) decreased growth and increased the rate of three-dimensional lumen formation, that of s80(Cyt2) increased proliferation without promoting lumen formation. These results were recapitulated in vivo, using doxycycline-inducible, mouse breast-transgenic expression of s80(Cyt1) amd s80(Cyt2). Expression of s80(Cyt1) decreased growth of the mammary ductal epithelium, caused precocious STAT5a activation and lactogenic differentiation, and increased cell surface E-cadherin levels. Remarkably, ductal growth inhibition by s80(Cyt1) occurred simultaneously with lobuloalveolar growth that was unimpeded by s80(Cyt1), suggesting that the response to ErbB4 may be influenced by the epithelial subtype. In contrast, expression of s80(Cyt2) caused epithelial hyperplasia, increased Wnt and nuclear beta-catenin expression, and elevated expression of c-myc and cyclin D1 in the mammary epithelium. These results demonstrate that the Cyt1 and Cyt2 ErbB4 isoforms, differing by only 16 amino acids, exhibit markedly opposing effects on mammary epithelium growth and differentiation.


Subject(s)
Alternative Splicing/genetics , Amino Acids/metabolism , Epithelium/metabolism , ErbB Receptors/metabolism , Mammary Glands, Animal/metabolism , Amino Acid Motifs , Animals , Cell Line , Cell Nucleus/metabolism , Cell Proliferation , ErbB Receptors/chemistry , Female , Gene Expression Regulation , Humans , Mammary Glands, Animal/cytology , Mice , Milk Proteins/genetics , Milk Proteins/metabolism , Phosphorylation , Pregnancy , Protein Isoforms/chemistry , Protein Isoforms/metabolism , Protein Structure, Tertiary , Puberty/metabolism , Receptor, ErbB-4 , STAT5 Transcription Factor/metabolism , Signal Transduction , beta Catenin/metabolism
3.
Mol Cell Biol ; 29(3): 892-906, 2009 Feb.
Article in English | MEDLINE | ID: mdl-19047365

ABSTRACT

In general, epidermal growth factor receptor family members stimulate cell proliferation. In contrast, at least one HER4 isoform, JM-a/Cyt1, inhibits cell growth after undergoing a two-step proteolytic cleavage that first produces a membrane-anchored 80-kDa fragment (m80(HER4)) and subsequently liberates a soluble 80-kDa fragment, s80(HER4). Here we report that s80(HER4) Cyt1 action increased the expression of WWP1 (for WW domain-containing protein 1), an E3 ubiquitin ligase, but not other members of the Nedd4 E3 ligase family. The HER4 Cyt1 isoform contains three proline-rich tyrosine (PY) WW binding motifs, while Cyt2 has only two. WWP1 binds to all three Cyt1 PY motifs; the interaction with PY2 found exclusively in Cyt1 was strongest. WWP1 ubiquitinated and caused the degradation of HER4 but not of EGFR, HER2, or HER3. The HER4-WWP1 interaction also accelerated WWP1 degradation. Membrane HER4 (full length and m80(HER4), the product of the first proteolytic cleavage) were the preferred targets of WWP1, correlating with the membrane localization of WWP1. Conversely s80(HER4), a poorer WWP1 substrate, was found in the cell nucleus, while WWP1 was not. Deletion of the C2 membrane association domain of WWP1 allowed more efficient s80(HER4) degradation, suggesting that WWP1 is normally part of a membrane complex that regulates HER4 membrane species levels, with a predilection for the growth-inhibitory Cyt1 isoform. Finally, WWP1 expression diminished HER4 biologic activity in MCF-7 cells. We previously showed that nuclear s80(HER4) is ubiquitinated and degraded by the anaphase-promoting complex, suggesting that HER4 ubiquitination within specific cellular compartments helps regulate the unique HER4 signaling capabilities.


Subject(s)
ErbB Receptors/metabolism , Protein Processing, Post-Translational , Signal Transduction , Ubiquitin-Protein Ligases/metabolism , Animals , COS Cells , Cell Line, Tumor , Cell Membrane/enzymology , Chlorocebus aethiops , Enzyme Stability , ErbB Receptors/chemistry , Gene Expression Regulation , Humans , Mice , Molecular Weight , Proteasome Endopeptidase Complex/metabolism , Protein Binding , Protein Isoforms/metabolism , Protein Structure, Tertiary , Protein Transport , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptor, ErbB-4 , Solubility , Subcellular Fractions/enzymology , Substrate Specificity , Ubiquitin-Protein Ligases/genetics , Ubiquitination
4.
Mol Endocrinol ; 22(10): 2307-21, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18653779

ABSTRACT

Differentiation of mammary epithelium in vivo requires signaling through prolactin and ErbB4/HER4-dependent mechanisms. Although stimulation of either the prolactin receptor or ErbB4/HER4 results in activation of the transcription factor signal transducer and activator of transcription 5A (STAT5A) and induction of lactogenic differentiation, how these pathways intersect is unknown. We show herein that prolactin signaling in breast cells cooperates with and is substantially enhanced by the receptor tyrosine kinase ErbB4/HER4. Prolactin and the ErbB4/HER4 ligand heparin-binding epidermal growth factor each induced STAT5A tyrosine phosphorylation and nuclear translocation; each pathway required the intracellular tyrosine kinase Janus kinase 2 (JAK2). We found that full prolactin-mediated STAT5A activation and binding to the endogenous beta-casein promoter required ErbB4/HER4 but did not require ErbB1/epidermal growth factor receptor. For example, prolactin-induced STAT5A activity was markedly diminished in cells overexpressing kinase inactive HER4, in cells transfected with small interfering RNAs to specifically knock down endogenous ErbB4/HER4 expression and in cells treated with a small molecule inhibitor that targets ErbB4 kinase. Interestingly, prolactin caused ErbB4/HER4 tyrosine phosphorylation in a JAK2 kinase-dependent manner. Finally, prolactin receptor, ErbB4/HER4, and JAK2 were coimmunoprecipitated from prolactin-treated but not untreated cells. These results suggest that prolactin signaling engages the ErbB4 pathway via JAK2 and that ErbB4 provides an important component of STAT5A-dependent lactogenic differentiation; this pathway integration may help explain the similar deficit in mammary development observed in gene-targeted mice deficient in prolactin receptor, JAK2, ErbB4, or STAT5A.


Subject(s)
Cell Differentiation , Epithelial Cells/physiology , ErbB Receptors/metabolism , Janus Kinase 2/metabolism , Mammary Glands, Animal/cytology , Prolactin/metabolism , Signal Transduction/physiology , Animals , Cell Differentiation/genetics , Cell Differentiation/physiology , Cell Line , Enzyme Activation , Epithelial Cells/cytology , ErbB Receptors/genetics , Female , Gene Expression Regulation , Heparin-binding EGF-like Growth Factor , Intercellular Signaling Peptides and Proteins/metabolism , Janus Kinase 2/genetics , Mice , Phosphorylation , Pregnancy , Receptor, ErbB-4 , Receptors, Prolactin/genetics , Receptors, Prolactin/metabolism , STAT5 Transcription Factor/genetics , STAT5 Transcription Factor/metabolism , Tyrosine/metabolism
5.
J Mammary Gland Biol Neoplasia ; 13(2): 235-46, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18437540

ABSTRACT

The ErbB receptor tyrosine kinase family has often been associated with increased growth of breast epithelial cells, as well as malignant transformation and progression. In contrast, ErbB4/HER4 exhibits unique attributes from a two step proteolytic cleavage which releases an 80 kilodalton, nuclear localizing, tyrosine kinase to a signal transduction mechanism that slows growth and stimulates differentiation of breast cells. This review provides an overview of ErbB4/HER4 in growth and differentiation of the mammary epithelium, including its physiologic role in development, the contrasting growth inhibition/tumor suppression and growth acceleration of distinct ErbB4/HER4 isoforms and a description of the unique cell cycle regulated pattern of nuclear HER4 ubiquitination and destruction.


Subject(s)
Biomarkers, Tumor/metabolism , Breast Neoplasms/metabolism , ErbB Receptors/metabolism , Mammary Glands, Human/growth & development , Animals , Breast Neoplasms/physiopathology , Cell Differentiation , Female , Growth Inhibitors/metabolism , Humans , Mammary Glands, Animal/cytology , Mammary Glands, Animal/growth & development , Mammary Glands, Animal/metabolism , Mammary Glands, Human/cytology , Mammary Glands, Human/metabolism , Mice , Protein Isoforms , Receptor, ErbB-4 , Signal Transduction , Tumor Cells, Cultured
6.
J Clin Invest ; 118(1): 64-78, 2008 Jan.
Article in English | MEDLINE | ID: mdl-18079969

ABSTRACT

Overexpression of the receptor tyrosine kinase EPH receptor A2 (EphA2) is commonly observed in aggressive breast cancer and correlates with a poor prognosis. However, while EphA2 has been reported to enhance tumorigenesis, proliferation, and MAPK activation in several model systems, other studies suggest that EphA2 activation diminishes these processes and inhibits the activity of MAPK upon ligand stimulation. In this study, we eliminated EphA2 expression in 2 transgenic mouse models of mammary carcinoma. EphA2 deficiency impaired tumor initiation and metastatic progression in mice overexpressing ErbB2 (also known as Neu) in the mammary epithelium (MMTV-Neu mice), but not in mice overexpressing the polyomavirus middle T antigen in mammary epithelium (MMTV-PyV-mT mice). Histologic and ex vivo analyses of MMTV-Neu mouse mammary epithelium indicated that EphA2 enhanced tumor proliferation and motility. Biochemical analyses revealed that EphA2 formed a complex with ErbB2 in human and murine breast carcinoma cells, resulting in enhanced activation of Ras-MAPK signaling and RhoA GTPase. Additionally, MMTV-Neu, but not MMTV-PyV-mT, tumors were sensitive to therapeutic inhibition of EphA2. These data suggest that EphA2 cooperates with ErbB2 to promote tumor progression in mice and may provide a novel therapeutic target for ErbB2-dependent tumors in humans. Moreover, EphA2 function in tumor progression appeared to depend on oncogene context, an important consideration for the application of therapies targeting EphA2.


Subject(s)
Adenocarcinoma/metabolism , Cell Transformation, Neoplastic/metabolism , MAP Kinase Signaling System , Mammary Neoplasms, Experimental/metabolism , Receptor, EphA2/metabolism , Receptor, ErbB-2/metabolism , Adenocarcinoma/genetics , Adenocarcinoma/pathology , Adenocarcinoma/therapy , Animals , Antigens, Polyomavirus Transforming/genetics , Antigens, Polyomavirus Transforming/metabolism , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Breast Neoplasms/therapy , Cell Movement/genetics , Cell Proliferation , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/pathology , Female , Humans , MAP Kinase Signaling System/genetics , Male , Mammary Glands, Animal/metabolism , Mammary Glands, Animal/pathology , Mammary Glands, Human/metabolism , Mammary Glands, Human/pathology , Mammary Neoplasms, Experimental/genetics , Mammary Neoplasms, Experimental/pathology , Mammary Neoplasms, Experimental/therapy , Mice , Mice, Transgenic , Neoplasm Metastasis , Receptor, EphA2/genetics , Receptor, ErbB-2/genetics , rho GTP-Binding Proteins/genetics , rho GTP-Binding Proteins/metabolism , rhoA GTP-Binding Protein
7.
Cancer Res ; 67(14): 6582-90, 2007 Jul 15.
Article in English | MEDLINE | ID: mdl-17638867

ABSTRACT

Heregulin-mediated activation of HER4 initiates receptor cleavage (releasing an 80-kDa HER4 intracellular domain, s80(HER4), containing nuclear localization sequences) and results in G(2)-M delay by unknown signaling mechanisms. We report herein that s80(HER4) contains a functional cyclin B-like sequence known as a D-box, which targets proteins for degradation by anaphase-promoting complex (APC)/cyclosome, a multisubunit ubiquitin ligase. s80(HER4) ubiquitination and proteasomal degradation occurred during mitosis but not during S phase. Inhibition of an APC subunit (APC2) using short interfering RNA knockdown impaired s80(HER4) degradation. Mutation of the s80(HER4) D-box sequence stabilized s80(HER4) during mitosis, and s80(HER4)-dependent growth inhibition via G(2)-M delay was significantly greater with the D-box mutant. Polyomavirus middle T antigen-transformed HC11 cells expressing s80(HER4) resulted in smaller, less proliferative, more differentiated tumors in vivo than those expressing kinase-dead s80(HER4) or the empty vector. Cells expressing s80(HER4) with a disrupted D-box did not form tumors, instead forming differentiated ductal structures. These results suggest that cell cycle-dependent degradation of s80(HER4) limits its growth-inhibitory action, and stabilization of s80(HER4) enhances tumor suppression, thus providing a link between HER4-mediated growth inhibition and cell cycle control.


Subject(s)
Cell Nucleus/metabolism , ErbB Receptors/physiology , Mitosis , Neuregulin-1/metabolism , Amino Acid Motifs , Antigens, Polyomavirus Transforming/metabolism , Cell Division , Cell Line, Transformed , Cell Line, Tumor , G2 Phase , Green Fluorescent Proteins/metabolism , HeLa Cells , Humans , Protein Structure, Tertiary , Receptor, ErbB-4 , Ubiquitin/metabolism
8.
Mol Endocrinol ; 21(8): 1861-76, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17505063

ABSTRACT

Unlike the proliferative action of other epidermal growth factor (EGF) receptor family members, HER4/ErbB4 is often associated with growth-inhibitory and differentiation signaling. These actions may involve HER4 two-step proteolytic processing by intramembraneous gamma-secretase, releasing the soluble, intracellular 80-kDa HER4 cytoplasmic domain, s80HER4. We demonstrate that pharmacological inhibition of either gamma-secretase activity or HER4 tyrosine kinase activity blocked heregulin-dependent growth inhibition of SUM44 breast cancer cells. We next generated breast cell lines stably expressing GFP-s80HER4 [green fluorescent protein (GFP) fused to the N terminus of the HER4 cytoplasmic domain, residues 676-1308], GFP-CT(HER4) (GFP fused to N terminus of the HER4 C-terminus distal to the tyrosine kinase domain, residues 989-1308), or GFP alone. Both GFP-s80HER4 and GFP-CTHER4 were found in the nucleus, but GFP-s80HER4 accumulated to a greater extent and sustained its nuclear localization. s80HER4 was constitutively tyrosine phosphorylated, and treatment of cells with a specific HER family tyrosine kinase inhibitor 1) blocked tyrosine phosphorylation; 2) markedly diminished GFP-s80HER4 nuclear localization; and 3) reduced signal transducer and activator of transcription (STAT)5A tyrosine phosphorylation and nuclear localization as well as GFP-s80HER4:STAT5A interaction. Multiple normal mammary and breast cancer cell lines, stably expressing GFP-s80HER4 (SUM44, MDA-MB-453, MCF10A, SUM102, and HC11) were growth inhibited compared with the same cell line expressing GFP-CTHER4 or GFP alone. The s80HER4-induced cell number reduction was due to slower growth because rates of apoptosis were equivalent in GFP-, GFP-CTHER4-, and GFP-s80HER4-expressing cells. Lastly, GFP-s80HER4 enhanced differentiation signaling as indicated by increased basal and prolactin-dependent beta-casein expression. These results indicate that surface HER4 tyrosine phosphorylation and ligand-dependent release of s80HER4 are necessary, and s80HER4 signaling is sufficient for HER4-dependent growth inhibition.


Subject(s)
Cell Proliferation , Cytoplasm/physiology , ErbB Receptors/physiology , Growth Inhibitors/physiology , Mammary Glands, Human/cytology , Mammary Glands, Human/metabolism , Peptide Fragments , Amyloid Precursor Protein Secretases/antagonists & inhibitors , Cell Line, Tumor , Female , Humans , Mammary Glands, Human/enzymology , Neuregulin-1/antagonists & inhibitors , Neuregulin-1/physiology , Peptide Fragments/physiology , Protein Structure, Tertiary , Receptor, ErbB-4
9.
Mol Cell Biol ; 26(17): 6412-24, 2006 Sep.
Article in English | MEDLINE | ID: mdl-16914727

ABSTRACT

HER4 expression in human breast cancers correlates with a positive prognosis. While heregulin inhibits the growth of HER4-positive breast cancer cells, it does so by undefined mechanisms. We demonstrate that heregulin-induced HER4 activity inhibits cell proliferation and delays G(2)/M progression of breast cancer cells. While investigating pathways of G(2)/M delay, we noted that heregulin increased the expression of BRCA1 in a HER4-dependent, HER2-independent manner. Induction of BRCA1 by HER4 occurred independently of the cell cycle. Moreover, BRCA1 expression was elevated in HER4-postive human breast cancer specimens. Heregulin stimulated c-Jun N-terminal kinase (JNK), and pharmacologic inhibition of JNK impaired heregulin-enhanced expression of BRCA1 and mitotic delay; inhibition of Erk1/2 did not. Knockdown of BRCA1 with small interfering RNA in a human breast cancer cell line interfered with HER4-mediated mitotic delay. Heregulin/HER4-dependent mitotic delay was examined further with an isogenic pair of mouse mammary epithelial cells (MECs) derived from mice harboring homozygous LoxP sites flanking exon 11 of BRCA1, such that one cell line expressed BRCA1 while the other cell line, after Cre-mediated excision, did not. BRCA1-positive MECs displayed heregulin-dependent mitotic delay; however, the isogenic BRCA1-negative MECs did not. These results suggest that heregulin-mediated growth inhibition in HER4-postive breast cancer cells requires BRCA1.


Subject(s)
BRCA1 Protein/metabolism , ErbB Receptors/metabolism , Mitosis/drug effects , Neuregulin-1/pharmacology , Animals , BRCA1 Protein/deficiency , BRCA1 Protein/genetics , Breast Neoplasms/pathology , Epithelial Cells/cytology , Epithelial Cells/drug effects , ErbB Receptors/genetics , Exons/genetics , G2 Phase/drug effects , Gene Expression Regulation, Neoplastic/drug effects , HeLa Cells , Humans , Mammary Glands, Animal/cytology , Mice , Mitogen-Activated Protein Kinase 8/metabolism , Mitogen-Activated Protein Kinase 9/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptor, ErbB-2/metabolism , Receptor, ErbB-4 , Tumor Cells, Cultured
10.
Mol Biol Cell ; 17(9): 4118-29, 2006 Sep.
Article in English | MEDLINE | ID: mdl-16837552

ABSTRACT

Differentiation of mammary epithelium in vivo requires signaling through prolactin- and ErbB4/HER4-dependent mechanisms; how these pathways intersect is unknown. We show herein that HC11 mouse mammary cells undergo ErbB4-dependent lactational differentiation. Prolactin and the ErbB4 ligand HB-EGF each induced STAT5A activation, expression of lactogenic differentiation markers, and lumen formation in three-dimensional Matrigel cultures in HC11 cells. ErbB4 undergoes ligand-dependent transmembrane domain cleavage at Val-675, releasing a soluble 80-kDa intracellular domain (s80(HER4)) that localizes to nuclei; the physiological relevance of s80(HER4) is unknown. A HER4(V675A) mutant abolishing transmembrane cleavage impaired STAT5A activity, lactogenic gene expression, and lumen formation. Kinase-dead HER4(KD) was neither cleaved nor able to induce differentiation of HC11 cells. Without treating HC11 cells with prolactin or HB-EGF, s80(HER4) (expressed from a cDNA construct) localized to the nucleus, activated STAT5A, and induced three-dimensional lumen formation. Nuclear localization of exogenous s80(HER4) required intact kinase activity of s80(HER4), as did activation of STAT5A. In contrast, nuclear localization of s80(HER4) and STAT5A activation did not require the 16-amino acid region of the ErbB4 intracellular domain specific to the Cyt-1 isoform of ErbB4, and absent in the Cyt-2 isoform. These results suggest that s80(HER4) formation contributes to ErbB4-dependent differentiation of mammary epithelial cells.


Subject(s)
Cell Differentiation , Epithelial Cells/cytology , ErbB Receptors/chemistry , ErbB Receptors/metabolism , Mammary Glands, Animal/cytology , Alternative Splicing/genetics , Amino Acid Sequence , Animals , Cell Membrane/metabolism , Cell Nucleus/metabolism , Cells, Cultured , ErbB Receptors/genetics , Ligands , Mice , Molecular Sequence Data , Phosphorylation , Phosphotransferases/metabolism , Protein Processing, Post-Translational , Protein Structure, Tertiary , Protein Transport , Receptor, ErbB-4 , STAT5 Transcription Factor/metabolism , Solubility , Transcriptional Activation/genetics
11.
Clin Cancer Res ; 11(2 Pt 2): 937s-43s, 2005 Jan 15.
Article in English | MEDLINE | ID: mdl-15701890

ABSTRACT

It is generally accepted that transforming growth factor beta (TGFbeta) is both a tumor suppressor and tumor promoter. Whereas loss or attenuation of TGFbeta signal transduction is permissive for transformation, introduction of dominant-negative TGFbeta receptors into metastatic breast cancer cells has been shown to inhibit epithelial-to-mesenchymal transition, motility, invasiveness, survival, and metastases. In addition, there is evidence that excess production and/or activation of TGFbeta by cancer cells can contribute to tumor progression by paracrine mechanisms involving neoangiogenesis, production of stroma and proteases, and subversion of immune surveillance mechanisms in tumor hosts. These data provide a rationale in favor of blockade of autocrine/paracrine TGFbeta signaling in human mammary tumors with therapeutic intent. Several treatment approaches are currently in early clinical development and have been the focus of our laboratory. These include (1) ligand antibodies or receptor-containing fusion proteins aimed at blocking ligand binding to cognate receptors and (2) small-molecule inhibitors of the type I TGFbeta receptor serine/threonine kinase. Many questions remain about the viability of anti-TGFbeta treatment strategies, the best molecular approach (or combinations) for inhibition of TGFbeta function in vivo, the biochemical surrogate markers of tumor response, the molecular profiles in tumors for selection into clinical trials, and potential toxicities, among others.


Subject(s)
Breast Neoplasms/metabolism , Transforming Growth Factor beta/physiology , Animals , Breast Neoplasms/secondary , Female , Humans
12.
Cancer Res ; 64(24): 9002-11, 2004 Dec 15.
Article in English | MEDLINE | ID: mdl-15604265

ABSTRACT

To address the role of transforming growth factor (TGF) beta in the progression of established tumors while avoiding the confounding inhibitory effects of TGF-beta on early transformation, we generated doxycycline (DOX)-inducible triple transgenic mice in which active TGF-beta1 expression could be conditionally regulated in mouse mammary tumor cells transformed by the polyomavirus middle T antigen. DOX-mediated induction of TGF-beta1 for as little as 2 weeks increased lung metastases >10-fold without a detectable effect on primary tumor cell proliferation or tumor size. DOX-induced active TGF-beta1 protein and nuclear Smad2 were restricted to cancer cells, suggesting a causal association between autocrine TGF-beta and increased metastases. Antisense-mediated inhibition of TGF-beta1 in polyomavirus middle T antigen-expressing tumor cells also reduced basal cell motility, survival, anchorage-independent growth, tumorigenicity, and metastases. Therefore, induction and/or activation of TGF-beta in hosts with established TGF-beta-responsive cancers can rapidly accelerate metastatic progression.


Subject(s)
Mammary Neoplasms, Experimental/metabolism , Mammary Neoplasms, Experimental/pathology , Transforming Growth Factor beta/biosynthesis , Animals , Cell Movement/physiology , DNA, Antisense/genetics , DNA-Binding Proteins/physiology , Female , Mammary Neoplasms, Experimental/genetics , Mice , Mice, Transgenic , Neoplasm Metastasis , Oncogenes , Smad Proteins , Trans-Activators/physiology , Transforming Growth Factor beta/genetics , Transforming Growth Factor beta/pharmacology , Transforming Growth Factor beta1
SELECTION OF CITATIONS
SEARCH DETAIL
...