Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
Oncotarget ; 8(43): 74006-74018, 2017 Sep 26.
Article in English | MEDLINE | ID: mdl-29088764

ABSTRACT

The anti-cancer agent CBP501 binds to calmodulin (CaM). Recent studies showed that migration and metastasis are inhibited by several CaM antagonists. However, there is no available evidence that CBP501 has similar effects. Here we found that CBP501 inhibits migration of non-small cell lung cancer (NSCLC) cells in vitro, even in the presence of migration inducing factors such as WNT, IL-6, and several growth factors. CBP501 also inhibited epidermal growth factor (EGF) enhanced invasion and the epithelial-to-mesenchymal transition (EMT), and this inhibition was accompanied by (i) suppression of Akt and ERK1/2 phosphorylation, and (ii) suppression of expression of transcription factor Zeb1 and the mesenchymal marker Vimentin. A pull down analysis performed using sepharose-immobilized CaM showed that CBP501 blocks the interaction between CaM and KRas. Furthermore, EGF induced Akt activation and cell migration was effectively suppressed by KRas down-regulation in NSCLC cells. Stable knockdown of KRas also made cells insensitive to CBP501's inhibition of growth factor-induced migration. Taken together, these results indicate that CBP501 inhibits binding of CaM with KRas and thereby suppresses the PI3K/AKT pathway, migration, invasion and EMT. These findings have identified a previously unrecognized effect of CBP501 on downstream KRas signaling mechanisms involving EMT and invasion, and provide support for the further clinical development of this agent.

2.
Oncotarget ; 8(38): 64015-64031, 2017 Sep 08.
Article in English | MEDLINE | ID: mdl-28969049

ABSTRACT

CBP501 is an anti-cancer drug candidate which has been shown to increase cis-diamminedichloro-platinum (II) (CDDP) uptake into cancer cell through calmodulin (CaM) inhibition. However, the effects of CBP501 on the cells in the tumor microenvironment have not been addressed. Here, we investigated new aspects of the potential anti-tumor mechanism of action of CBP501 by examining its effects on the macrophages. Macrophages contribute to cancer-related inflammation and sequential production of cytokines such as IL-6 and TNF-α which cause various biological processes that promote tumor initiation, growth and metastasis (1). These processes include the epithelial to mesenchymal transition (EMT) and cancer stem cell (CSC) formation, which are well-known, key events for metastasis. The present work demonstrates that CBP501 suppresses lipopolysaccharide (LPS)-induced production of IL-6, IL-10 and TNF-α by macrophages. CBP501 also suppressed formation of the tumor spheroids by culturing with conditioned medium from the LPS-stimulated macrophage cell line RAW264.7. Moreover, CBP501 suppressed expression of ABCG2, a marker for CSCs, by inhibiting the interaction between cancer cells expressing VCAM-1 and macrophages expressing VLA-4. Consistently with these results, CBP501 in vivo suppressed metastases of a tumor cell line, 4T1, one which is insensitive to combination treatment of CBP501 and CDDP in vitro. Taken together, these results offer potential new, unanticipated advantages of CBP501 treatment in anti-tumor therapy through a mechanism that entails the suppression of interactions between macrophages and cancer cells with suppression of sequential CSC-like cell formation in the tumor microenvironment.

3.
J Cell Sci ; 130(7): 1321-1332, 2017 04 01.
Article in English | MEDLINE | ID: mdl-28232522

ABSTRACT

Heparin-binding EGF-like growth factor (HB-EGF) plays an indispensable role in suppression of cell proliferation during mouse valvulogenesis. However, ligands of the EGF receptor (EGFR/ErbB1), including HB-EGF, are generally considered as growth-promoting factors, as shown in cancers. HB-EGF binds to and activates ErbB1 and ErbB4. We investigated the role of ErbB receptors in valvulogenesis in vivo using ErbB1- and ErbB4-deficient mice, and an ex vivo model of endocardial cushion explants. We show that HB-EGF suppresses valve mesenchymal cell proliferation through a heterodimer of ErbB1 and ErbB4, and an ErbB1 ligand (or ligands) promotes cell proliferation through a homodimer of ErbB1. Moreover, a rescue experiment with cleavable or uncleavable isoforms of ErbB4 in ERBB4-null cells indicates that the cleavable JM-A, but not the uncleavable JM-B, splice variant of ErbB4 rescues the defect of the null cells. These data suggest that the cytoplasmic intracellular domain of ErbB4, rather than the membrane-anchored tyrosine kinase, achieves this suppression. Our study demonstrates that opposing signals generated by different ErbB dimer combinations function in the same cardiac cushion mesenchymal cells for proper cardiac valve formation.


Subject(s)
ErbB Receptors/metabolism , Heart Valves/embryology , Heart Valves/metabolism , Mesoderm/cytology , Organogenesis , Receptor, ErbB-4/metabolism , Signal Transduction , Alleles , Animals , Cell Proliferation , Embryo, Mammalian/metabolism , Genes, Dominant , Heparin-binding EGF-like Growth Factor/metabolism , Ligands , Mice, Knockout , Models, Biological , Mutation/genetics , Protein Domains , Protein Isoforms/metabolism , Receptor, ErbB-4/chemistry , Up-Regulation
4.
Mol Cancer Ther ; 13(9): 2215-25, 2014 Sep.
Article in English | MEDLINE | ID: mdl-25053821

ABSTRACT

CBP501 is an anticancer drug candidate that was investigated in two randomized phase II clinical trials for patients with nonsquamous non-small cell lung cancer (NSCLC) and malignant pleural mesothelioma (MPM). CBP501 has been shown to have two mechanisms of action, namely calmodulin modulation and G2 checkpoint abrogation. Here, we searched for a biomarker to predict sensitivity to CBP501. Twenty-eight NSCLC cell lines were classified into two subgroups, CBP501-sensitive and -insensitive, by quantitatively analyzing the cis-diamminedichloro-platinum (II) (CDDP)-enhancing activity of CBP501 through treatments with short-term (1 hour) coexposure to CDDP and CBP501 or to either alone. Microarray analysis was performed on these cell lines to identify gene expression patterns that correlated with CBP501 sensitivity. We found that multiple nuclear factor erythroid-2-related factor 2 (Nrf2) target genes showed high expression in CBP501-insensitive cell lines. Western blot and immunocytochemical analysis for Nrf2 in NSCLC cell lines also indicated higher protein level in CBP501-insensitive cell lines. Moreover, CBP501 sensitivity is modulated by silencing or sulforaphane-induced overexpression of Nrf2. These results indicate that Nrf2 transcription factor is a potential candidate as a biomarker for resistance to CBP501. This study might help to identify those subpopulations of patients who would respond well to the CBP501 and CDDP combination treatment of NSCLC.


Subject(s)
Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/metabolism , Drug Resistance, Neoplasm , Lung Neoplasms/drug therapy , Lung Neoplasms/metabolism , NF-E2-Related Factor 2/metabolism , Peptide Fragments/chemistry , cdc25 Phosphatases/chemistry , Biomarkers, Tumor/chemistry , Calmodulin/chemistry , Cell Cycle , Cell Line, Tumor , G2 Phase , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Humans , Lentivirus/metabolism , Microscopy, Fluorescence , Oligonucleotide Array Sequence Analysis , RNA, Messenger/metabolism , RNA, Small Interfering/metabolism
5.
Mol Cancer Ther ; 10(10): 1929-38, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21831962

ABSTRACT

CBP501 is an anticancer drug currently in randomized phase II clinical trials for patients with non-small cell lung cancer and malignant pleural mesothelioma. CBP501 was originally described as a unique G(2) checkpoint-directed agent that binds to 14-3-3, inhibiting the actions of Chk1, Chk2, mitogen-activated protein kinase-activated protein kinase 2, and C-Tak1. However, unlike a G(2) checkpoint inhibitor, CBP501 clearly enhances the accumulation of tumor cells at G(2)-M phase that is induced by cisplatin or bleomycin at low doses and short exposure. By contrast, CBP501 does not similarly affect the accumulation of tumor cells at G(2)-M that is induced by radiation, doxorubicin, or 5-fluorouracil treatment. Our recent findings point to an additional mechanism of action for CBP501. The enhanced accumulation of tumor cells at G(2)-M upon combined treatment with cisplatin and CBP501 results from an increase in intracellular platinum concentrations, which leads to increased binding of platinum to DNA. The observed CBP501-enhanced platinum accumulation is negated in the presence of excess Ca(2+). Some calmodulin inhibitors behave similarly to, although less potently than, CBP501. Furthermore, analysis by surface plasmon resonance reveals a direct, high-affinity molecular interaction between CBP501 and CaM (K(d) = 4.62 × 10(-8) mol/L) that is reversed by Ca(2+), whereas the K(d) for the complex between CBP501 and 14-3-3 is approximately 10-fold weaker and is Ca(2+) independent. We conclude that CaM inhibition contributes to CBP501's activity in sensitizing cancer cells to cisplatin or bleomycin. This article presents an additional mechanism of action which might explain the clinical activity of the CBP501-cisplatin combination.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Bleomycin/pharmacology , Calmodulin/metabolism , Cisplatin/pharmacology , Neoplasms/drug therapy , Peptide Fragments/pharmacology , cdc25 Phosphatases/pharmacology , Bleomycin/administration & dosage , Calcium Chloride/pharmacology , Calmodulin/antagonists & inhibitors , Cell Line, Tumor , Cell Survival/drug effects , Cisplatin/administration & dosage , Cisplatin/pharmacokinetics , DNA Adducts/biosynthesis , Drug Synergism , Humans , Neoplasms/metabolism , Neoplasms/pathology , Peptide Fragments/pharmacokinetics , cdc25 Phosphatases/pharmacokinetics
6.
Development ; 137(13): 2205-14, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20530548

ABSTRACT

HB-EGF, a member of the EGF family of growth factors, plays an important role in cardiac valve development by suppressing mesenchymal cell proliferation. Here, we show that HB-EGF must interact with heparan sulfate proteoglycans (HSPGs) to properly function in this process. In developing valves, HB-EGF is synthesized in endocardial cells but accumulates in the mesenchyme by interacting with HSPGs. Disrupting the interaction between HB-EGF and HSPGs in an ex vivo model of endocardial cushion explants resulted in increased mesenchymal cell proliferation. Moreover, homozygous knock-in mice (HB(Delta)(hb/)(Delta)(hb)) expressing a mutant HB-EGF that cannot bind to HSPGs developed enlarged cardiac valves with hyperproliferation of mesenchymal cells; this resulted in a phenotype that resembled that of Hbegf-null mice. Interestingly, although Hbegf-null mice had abnormal heart chambers and lung alveoli, HB(Delta)(hb/)(Delta)(hb) mice did not exhibit these defects. These results indicate that interactions with HSPGs are essential for the function of HB-EGF, especially in cardiac valve development, in which HB-EGF suppresses mesenchymal cell proliferation.


Subject(s)
Endocardial Cushions/metabolism , Heart Valves/embryology , Heart Valves/metabolism , Heparan Sulfate Proteoglycans/metabolism , Intercellular Signaling Peptides and Proteins/metabolism , Animals , Cell Proliferation , Heparin-binding EGF-like Growth Factor , In Vitro Techniques , Mesoderm/metabolism , Mice
7.
Development ; 135(14): 2425-34, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18550712

ABSTRACT

In mouse, left-right (L-R) patterning depends on asymmetric expression of Nodal around the node, leading to Nodal expression specifically in the left lateral plate mesoderm (LPM). Bone morphogenetic protein (BMP) signaling is also involved, but the mechanistic relationship with Nodal expression remains unclear. We find that BMP signal transduction is higher in the right LPM, although Bmp4, which is required for L-R patterning, is expressed symmetrically. By contrast, the BMP antagonists noggin (Nog) and chordin (Chrd) are expressed at higher levels in the left LPM. In Chrd;Nog double mutants, BMP signaling is elevated on both sides, whereas Nodal expression is absent. Ectopic expression of Nog in the left LPM of double mutants restores Nodal expression. Ectopic Bmp4 expression in the left LPM of wild-type embryos represses Nodal transcription, whereas ectopic Nog in the right LPM leads to inappropriate Nodal expression. These data indicate that chordin and noggin function to limit BMP signaling in the left LPM, thereby derepressing Nodal expression. In the node, they promote peripheral Nodal expression and proper node morphology, potentially in concert with Notch signaling. These results indicate that BMP antagonism is required in both the node and LPM to facilitate L-R axis establishment in the mammalian embryo.


Subject(s)
Body Patterning/physiology , Bone Morphogenetic Proteins/antagonists & inhibitors , Mesoderm/metabolism , Animals , Animals, Outbred Strains , Body Patterning/genetics , Bone Morphogenetic Protein 4 , Bone Morphogenetic Proteins/genetics , Bone Morphogenetic Proteins/metabolism , Carrier Proteins/genetics , Carrier Proteins/metabolism , Embryo, Mammalian , Gene Expression Regulation, Developmental , Glycoproteins/genetics , Glycoproteins/metabolism , Immunohistochemistry , Intercellular Signaling Peptides and Proteins/genetics , Intercellular Signaling Peptides and Proteins/metabolism , Mice , Mice, Inbred ICR , Models, Biological , Mutation , Nodal Protein , Organ Culture Techniques , Signal Transduction , Transforming Growth Factor beta/genetics , Transforming Growth Factor beta/metabolism
8.
Dev Cell ; 11(4): 495-504, 2006 Oct.
Article in English | MEDLINE | ID: mdl-17011489

ABSTRACT

The bilateral symmetry of the mouse embryo is broken by leftward fluid flow in the node. However, it is unclear how this directional flow is then translated into the robust, left side-specific Nodal gene expression that determines and coordinates left-right situs throughout the embryo. While manipulating Nodal and Lefty gene expression, we have observed phenomena that are indicative of the involvement of a self-enhancement and lateral-inhibition (SELI) system. We constructed a mathematical SELI model that not only simulates, but also predicts, experimental data. As predicted by the model, Nodal expression initiates even on the right side. These results indicate that directional flow represents an initial small difference between the left and right sides of the embryo, but is insufficient to determine embryonic situs. Nodal and Lefty are deployed as a SELI system required to amplify this initial bias and convert it into robust asymmetry.


Subject(s)
Body Patterning/physiology , Embryo, Mammalian/embryology , Embryonic Induction , Animals , Body Patterning/genetics , Computer Simulation , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/metabolism , Genetic Vectors , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , In Situ Hybridization , Left-Right Determination Factors , Mice , Mice, Mutant Strains , Models, Biological , Models, Theoretical , Nodal Protein , Organ Culture Techniques , Transcription Factors/genetics , Transcription Factors/metabolism , Transforming Growth Factor beta/genetics , Transforming Growth Factor beta/metabolism , Homeobox Protein PITX2
9.
Development ; 132(19): 4317-26, 2005 Oct.
Article in English | MEDLINE | ID: mdl-16141218

ABSTRACT

Heparin-binding EGF-like growth factor (HB-EGF) is a member of the EGF family of growth factors that binds to and activates the EGF receptor (EGFR) and ERBB4. Here, we show that HB-EGF-EGFR signaling is involved in eyelid development. HB-EGF expression is restricted to the tip of the leading edge of the migrating epithelium during eyelid closure in late gestation mouse embryos. Both HB-EGF null (HB(del/del)) and secretion-deficient (HB(uc/uc)) mutant embryos exhibited delayed eyelid closure, owing to slower leading edge extension and reduced actin bundle formation in migrating epithelial cells. No changes in cell proliferation were observed in these embryos. In addition, activation of EGFR and ERK was decreased in HB(del/del) eyelids. Crosses between HB(del/del) mice and waved 2 mice, a hypomorphic EGFR mutant strain, indicate that HB-EGF and EGFR interact genetically in eyelid closure. Together with our data showing that embryos treated with an EGFR-specific kinase inhibitor phenocopy HB(del/del) embryos, these data indicate that EGFR mediates HB-EGF-dependent eyelid closure. Finally, analysis of eyelid closure in TGFalpha-null mice and in HB-EGF and TGFalpha double null mice revealed that HB-EGF and TGFalpha contribute equally to and function synergistically in this process. These results indicate that soluble HB-EGF secreted from the tip of the leading edge activates the EGFR and ERK pathway, and that synergy with TGFalpha is required for leading edge extension in epithelial sheet migration during eyelid closure.


Subject(s)
Epidermal Growth Factor/physiology , Epithelial Cells/physiology , Eyelids/metabolism , Actins/metabolism , Animals , Cell Movement , Cell Proliferation , Epidermal Growth Factor/biosynthesis , Epidermal Growth Factor/genetics , ErbB Receptors/metabolism , ErbB Receptors/physiology , Extracellular Signal-Regulated MAP Kinases/physiology , Eyelids/cytology , Eyelids/embryology , Heparin-binding EGF-like Growth Factor , Intercellular Signaling Peptides and Proteins , Mice , Mice, Knockout , Mutation , Signal Transduction , Transforming Growth Factor alpha/physiology
10.
Development ; 130(9): 1795-804, 2003 May.
Article in English | MEDLINE | ID: mdl-12642485

ABSTRACT

The transcription factor Foxh1 mediates Nodal signaling. The role of Foxh1 in left-right (LR) patterning was examined with mutant mice that lack this protein in lateral plate mesoderm (LPM). The mutant mice failed to express Nodal, Lefty2 and Pitx2 on the left side during embryogenesis and exhibited right isomerism. Ectopic introduction of Nodal into right LPM, by transplantation of left LPM or by electroporation of a Nodal vector, induced Nodal expression in wild-type embryos but not in the mutant. Ectopic Nodal expression in right LPM also induced Lefty1 expression in the floor plate. Nodal signaling thus initiates asymmetric Nodal expression in LPM and induces Lefty1 at the midline. Monitoring of Nodal activity in wild-type and Foxh1 mutant embryos suggested that Nodal activity travels from the node to left LPM, and from left LPM to the midline.


Subject(s)
DNA-Binding Proteins/metabolism , Mice/embryology , Signal Transduction/physiology , Transcription Factors/metabolism , Transforming Growth Factor beta/metabolism , Viscera/embryology , Animals , DNA-Binding Proteins/genetics , Forkhead Transcription Factors , Gene Deletion , Mutation , Nodal Protein , Transcription Factors/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...