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










Publication year range
1.
Mol Oncol ; 16(4): 1009-1025, 2022 02.
Article in English | MEDLINE | ID: mdl-34482626

ABSTRACT

Sonic hedgehog (Shh)-driven medulloblastoma (Shh MB) cells are dependent on constitutive Shh signaling, but targeted treatment of Shh MB has been ineffective due to drug resistance. The purpose of this study was to address the critical role of signal transducer and activator of transcription 3 (STAT3) in Shh signaling and drug resistance in Shh MB cells. Herein, we show that STAT3 is required for Smoothened (Smo)-dependent Shh signaling and, in turn, is reciprocally regulated by Shh signaling, and demonstrate that STAT3 activity is critical for expression of HCK proto-oncogene, Src family tyrosine kinase (Hck) in Shh MB. We also demonstrate that maintained STAT3 activity suppresses p21 expression and promotes colony formation of Shh MB cells, whereas dual treatment with inhibitors of both Smo and STAT3 results in marked synergistic killing and overcomes drug resistance in vitro of Smo antagonist-resistant Shh MB cells. Finally, STAT3 inhibitor treatment significantly prevents in vivo tumor formation in genetically engineered Shh MB mice. Collectively, we show that STAT3 is necessary to maintain Shh signaling and thus is a potential therapeutic target to treat Shh MB and overcome anti-Smo drug resistance.


Subject(s)
Cerebellar Neoplasms , Medulloblastoma , Animals , Carcinogenesis/genetics , Cerebellar Neoplasms/pathology , Hedgehog Proteins/metabolism , Humans , Medulloblastoma/drug therapy , Medulloblastoma/genetics , Medulloblastoma/metabolism , Mice , STAT3 Transcription Factor/metabolism , Smoothened Receptor/genetics , Smoothened Receptor/metabolism
2.
J Neurooncol ; 121(1): 109-18, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25258252

ABSTRACT

Eph receptors and ephrin ligands are master regulators of oncogenic signaling required for proliferation, migration, and metastasis. Yet, Eph/ephrin expression and activity in medulloblastoma (MB), the most common malignant brain tumor of childhood, remains poorly defined. We hypothesized that Eph/ephrins are differentially expressed by sonic hedgehog (SHH) and non-SHH MB and that specific members contribute to the aggressive phenotype. Affymetrix gene expression profiling of 29 childhood MB, separated into SHH (N = 11) and non-SHH (N = 18), was performed followed by protein validation of selected Eph/ephrins in another 60 MB and two MB cell lines (DAOY, D556). Functional assays were performed using MB cells overexpressing or deleted for selected ephrins. We found EPHB4 and EFNA4 almost exclusively expressed by SHH MB, whereas EPHA2, EPHA8, EFNA1 and EFNA3 are predominantly expressed by non-SHH MB. The remaining family members, except EFNB1, are ubiquitously expressed by over 70-90 % MB, irrespective of subgroup. EFNB1 is the only member differentially expressed by 28 % of SHH and non-SHH MB. Corresponding protein expression for EphB/ephrinB1 and B2 was validated in MB. Only ephrinB2 was also detected in fetal cerebellum, indicating that EphB/ephrinB1 expression is MB-specific. EphrinB1 immunopositivity localizes to tumor cells within MB with the highest proliferative index. EphrinB1 overexpression promotes EphB activation, alters F-actin distribution and morphology, decreases adhesion, and significantly promotes proliferation. Either silencing or overexpression of ephrinB1 impairs migration. These results indicate that EphrinB1 is uniquely dysregulated in MB and promotes oncogenic responses in MB cells, implicating ephrinB1 as a potential target.


Subject(s)
Brain Neoplasms/metabolism , Ephrin-B1/metabolism , Hedgehog Proteins/metabolism , Medulloblastoma/metabolism , Actins/metabolism , Brain Neoplasms/pathology , Cell Adhesion/physiology , Cell Line, Tumor , Cell Proliferation/physiology , Cell Survival/physiology , Cerebellum/embryology , Cerebellum/metabolism , Cerebellum/pathology , Child , Ephrin-B1/genetics , Ephrin-B2/metabolism , Humans , Medulloblastoma/pathology , Proto-Oncogene Proteins pp60(c-src)/metabolism , RNA, Messenger/metabolism , Receptor, EphA2/metabolism , Receptor, EphA8/metabolism , Receptor, EphB4/metabolism
3.
Cancer Lett ; 354(1): 68-76, 2014 Nov 01.
Article in English | MEDLINE | ID: mdl-25107642

ABSTRACT

Medulloblastoma (MB) expresses Src kinase, while aurora kinase A overexpression correlates with poor survival. We thus investigated novel combination treatment with dasatinib and AT9283, inhibitors of Src and aurora kinase, respectively, on MB growth in vitro and in vivo. Treatment with each drug significantly reduced cell viability and combined treatment markedly potentiated this response. AT9283 induced p53 expression, autophagy, and G2/M cell-cycle arrest, while combined treatment induced S phase arrest. Dasatinib treatment caused tumor regression in vivo. Activated Src was detected in 44% MB analyzed. We conclude that further evaluation of this combination therapy for MB is highly warranted.


Subject(s)
Antineoplastic Agents/pharmacology , Aurora Kinase A/antagonists & inhibitors , Benzimidazoles/pharmacology , Enzyme Inhibitors/pharmacology , Medulloblastoma/drug therapy , Pyrimidines/pharmacology , Thiazoles/pharmacology , Urea/analogs & derivatives , Animals , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Autophagy , Cell Line, Tumor , Cell Movement , Cell Proliferation , Cell Survival , Dasatinib , G2 Phase , Humans , Mice , Mice, Inbred C57BL , Neoplasm Transplantation , Protein Kinase Inhibitors/pharmacology , Tumor Suppressor Protein p53/metabolism , Urea/pharmacology
4.
Mol Cancer ; 12: 18, 2013 Mar 05.
Article in English | MEDLINE | ID: mdl-23497290

ABSTRACT

BACKGROUND: Metastasis in medulloblastoma (MB) is associated with poor survival. Recent genetic studies revealed MB to comprise distinct molecular subgroups, including the sonic hedgehog (SHH) subgroup that exhibits a relatively high rate of progression. To identify targeted therapeutics against metastasis, a better understanding of the regulation of MB cell migration is needed. G protein-coupled receptor kinases (GRKs) have been implicated in cancer metastasis through their regulation of G-protein coupled receptors (GPCRs) involved in growth factor (GF)-mediated cell migration. However, the specific roles and regulation of GRKs in MB have not been investigated. METHODS: Microarray mRNA analysis was performed for GRKs, GPCRs, and GFs in 29 human MB, and real time RT-PCR was used to detect GRK6 expression in MB cells. Lenti- or retro-virus infection, and siRNA or shRNA transfection, of MB cells was used to overexpress and knockdown target genes, respectively. Western blot was used to confirm altered expression of proteins. The effect of altered target protein on cell migration was determined by Boyden chamber assay and xCELLigence migration assays. RESULTS: We observed co-overexpression of PDGFRA, CXCR4, and CXCL12 in the SHH MB subtype compared to non-SHH MB (5, 7, and 5-fold higher, respectively). GRK6, which typically acts as a negative regulator of CXCR4 signaling, is downregulated in MB, relative to other GRKs, while the percentage of GRK6 expression is lower in MB tumors with metastasis (22%), compared to those without metastasis (43%). In SHH-responsive MB cells, functional blockade of PDGFR abolished CXCR4-mediated signaling. shPDGFR transfected MB cells demonstrated increased GRK6 expression, while PDGF or 10% FBS treatment of native MB cells reduced the stability of GRK6 by inducing its proteosomal degradation. Overexpression or downregulation of Src, a key mediator of GF receptor/PDGFR signaling, similarly inhibited or induced GRK6 expression, respectively. siRNA downregulation of GRK6 enhanced CXCR4 signaling and promoted MB migration, while lentiviral-GRK6 overexpression suppressed CXCR4 signaling, potentiated the effect of AMD3100, a CXCR4 antagonist, and impaired migration. CONCLUSIONS: Our findings demonstrate a novel mechanism of GF receptor/PDGFR-Src-mediated dysregulation of CXCR4 signaling that promotes MB cell migration, which could potentially be exploited for therapeutic targeting in SHH MB.


Subject(s)
Cerebellar Neoplasms/metabolism , G-Protein-Coupled Receptor Kinases/genetics , Medulloblastoma/metabolism , Receptor, Platelet-Derived Growth Factor alpha/metabolism , Receptors, CXCR4/metabolism , Signal Transduction , src-Family Kinases/metabolism , Benzylamines , Cell Line, Tumor , Cell Movement , Cerebellar Neoplasms/pathology , Chemokine CXCL12/genetics , Chemokine CXCL12/metabolism , Cyclams , G-Protein-Coupled Receptor Kinases/metabolism , Gene Expression , Gene Expression Regulation, Neoplastic , Hedgehog Proteins/physiology , Heterocyclic Compounds/pharmacology , Humans , Platelet-Derived Growth Factor/physiology , Proteasome Endopeptidase Complex/metabolism , Proteolysis , Receptors, CXCR4/antagonists & inhibitors
5.
Mol Cancer ; 10: 121, 2011 Sep 24.
Article in English | MEDLINE | ID: mdl-21943176

ABSTRACT

BACKGROUND: The CCL2 chemokine is involved in promoting cancer angiogenesis, proliferation and metastasis by malignancies that express CCR2 receptor. Thus the CCL2/CCR2 axis is an attractive molecular target for anticancer drug development. METHODS: We have generated a novel fusion protein using GMCSF and an N-terminal truncated version of MCP1/CCL2 (6-76) [hereafter GMME1] and investigated its utility as a CCR2-specific tumoricidal agent. RESULTS: We found that distinct to full length CCL2 or its N-truncated derivative (CCL2 5-76), GMME1 bound to CCR2 on mouse lymphoma EG7, human multiple myeloma cell line U266, or murine and human medulloblastoma cell lines, and led to their death by apoptosis. We demonstrated that GMME1 specifically blocked CCR2-associated STAT3 phosphorylation and up-regulated pro-apoptotic BAX. Furthermore, GMME1 significantly inhibited EG7 tumor growth in C57BL/6 mice, and induced apoptosis of primary myeloma cells from patients. CONCLUSION: Our data demonstrate that GMME1 is a fusokine with a potent, CCR2 receptor-mediated pro-apoptotic effect on tumor cells and could be exploited as a novel biological therapy for CCR2+ malignancies including lymphoid and central nervous system malignancies.


Subject(s)
Antineoplastic Agents/pharmacology , Chemokine CCL2/genetics , Granulocyte-Macrophage Colony-Stimulating Factor/genetics , Receptors, CCR2/metabolism , Recombinant Fusion Proteins/pharmacology , Animals , Antigens, CD/metabolism , Antineoplastic Agents/therapeutic use , Apoptosis/drug effects , Cell Line, Tumor , Female , Humans , Lymphoma , Medulloblastoma , Mesenchymal Stem Cells/metabolism , Mice , Mice, Inbred C57BL , Multiple Myeloma/drug therapy , Multiple Myeloma/metabolism , Multiple Myeloma/pathology , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/therapeutic use , Tumor Burden , Xenograft Model Antitumor Assays
6.
Clin Exp Metastasis ; 27(7): 481-91, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20526801

ABSTRACT

We previously identified that overexpression of the platelet-derived growth factor receptor (PDGFR) is associated with metastatic medulloblastoma (MB) and showed that PDGF treatment increases ERK activity and promotes MB cell migration. In this study, we investigated whether ERK regulates Rac1/Pak1 signaling and is critically linked to MB cell migration. Herein we demonstrate that PDGF-BB treatment of MB cells induces concomitant activation of PDGFRß, MEK1/ERK, Rac1 and Pak1, but suppresses Rho activity, which together significantly promotes cell migration. Conversely, cells transfected with either PDGFRß or Pak1 siRNA or treated with an inhibitor of Rac1 (NSC23766) or N-myristoyltransferase-1 (Tris-dipalladium) are unable to activate Rac1 or Pak1 in response to PDGF, and consequently, are unable to undergo PDGF-mediated cell migration. Furthermore, we also demonstrate that either chemical inhibition of MEK/ERK (U0126) or stable downregulation of PDGFRß by shRNA similarly results in the loss of PDGF-induced ERK phosphorylation and abolishes Rac1/Pak1 activation and cell migration in response to PDGF. However, specific depletion of Pak1 by siRNA has no effect on PDGF-induced ERK phosphorylation, indicating that in MB cells ERK signaling is Pak1-independent, but PDGF-induced migration is dependent on ERK-mediated activation of Pak1. Finally, using tissue microarrays, we detect phosphorylated Pak1 in 53% of medulloblastomas and show that immunopositivity is associated with unfavorable outcome. We conclude that Rac1/Pak1 signaling is critical to MB cell migration and is functionally dependent on PDGFRß/ERK activity.


Subject(s)
Brain Neoplasms/pathology , Extracellular Signal-Regulated MAP Kinases/metabolism , Medulloblastoma/pathology , Neoplasm Metastasis , p21-Activated Kinases/metabolism , Blotting, Western , Brain Neoplasms/enzymology , Cell Line, Tumor , Enzyme Activation , Humans , Immunohistochemistry , Medulloblastoma/enzymology , Phosphorylation , Platelet-Derived Growth Factor/metabolism , RNA, Small Interfering , Receptors, Platelet-Derived Growth Factor/metabolism , Tissue Array Analysis , rac1 GTP-Binding Protein/metabolism
7.
Cancer Res ; 66(20): 9986-94, 2006 Oct 15.
Article in English | MEDLINE | ID: mdl-17047061

ABSTRACT

The cyclin D1 gene is amplified and overexpressed in human breast cancer, functioning as a collaborative oncogene. As the regulatory subunit of a holoenzyme phosphorylating Rb, cyclin D1 promotes cell cycle progression and a noncatalytic function has been described to sequester the cyclin-dependent kinase inhibitor protein p27. Cyclin D1 overexpression correlates with tumor metastasis and cyclin D1-deficient fibroblasts are defective in migration. The genetic mechanism by which cyclin D1 promotes migration and movement is poorly understood. Herein, cyclin D1 promoted cellular migration and cytokinesis of mammary epithelial cells. Cyclin D1 enhanced cellular migratory velocity. The induction of migration by cyclin D1 was abolished by mutation of K112 or deletion of NH(2)-terminal residues 46 to 90. These mutations of cyclin D1 abrogated physical interaction with p27(KIP1). Cyclin D1(-/-) cells were p27(KIP1) deficient and the defect in migration was rescued by p27(KIP1) reintroduction. Conversely, the cyclin D1 rescue of cyclin D1(-/-) cellular migration was reversed by p27(KIP1) small interfering RNA. Cyclin D1 regulated p27(KIP1) abundance at the posttranslational level, inhibiting the Skp2 promoter, Skp2 abundance, and induced p27(KIP1) phosphorylation at Ser(10). Together, these studies show cyclin D1 promotes mammary epithelial cell migration. p27(KIP1) is required for cyclin D1-mediated cellular migration.


Subject(s)
Cell Movement/physiology , Cyclin D1/physiology , Cyclin-Dependent Kinase Inhibitor p27/physiology , Actins/metabolism , Animals , Cell Line , Cell Line, Tumor , Cyclin D1/deficiency , Cyclin D1/genetics , Cyclin D1/metabolism , Cyclin-Dependent Kinase Inhibitor p27/metabolism , Epithelial Cells/cytology , Epithelial Cells/physiology , Humans , Mammary Glands, Animal/cytology , Mammary Glands, Animal/physiology , Mice , Mice, Inbred C57BL , Plasmids/genetics
8.
J Biol Chem ; 280(52): 42701-6, 2005 Dec 30.
Article in English | MEDLINE | ID: mdl-16260787

ABSTRACT

SHP-2, a tyrosine phosphatase implicated in diverse signaling pathways induced by growth factors and cytokines, is also involved in DNA damage-triggered signaling and cellular responses. We previously demonstrated that SHP-2 played an important role in DNA damage-induced apoptosis and G2/M cell cycle checkpoint. In the present studies, we have provided evidence that SHP-2 functions in DNA damage apoptosis and G2/M arrest in catalytically dependent and independent manners, respectively. Mutant embryonic fibroblasts with the Exon 3 deletion mutation in SHP-2 showed decreased apoptosis and diminished G2/M arrest in response to cisplatin treatment. Wild type (WT), but not catalytically inactive mutant SHP-2 (SHP-2 C459S), rescued the apoptotic response of the mutant cells. Interestingly, both WT and SHP-2 C459S efficiently restored the G2/M arrest response. Furthermore, inhibition of the catalytic activity of endogenous SHP-2 in WT cells by overexpression of SHP-2 C459S greatly decreased cell death but not G2/M arrest induced by cisplatin. Biochemical analyses revealed that activation of c-Abl kinase was decreased in SHP-2 C459S-overexpressing cells. However, DNA damage-induced translocation of Cdc25C from the nucleus to the cytoplasm was fully restored in both WT and SHP-2 C459S "rescued" cells. Additionally, we demonstrated that the role of SHP-2 in DNA damage-induced cellular responses was independent of the tumor suppressor p53. Embryonic stem cells with the SHP-2 deletion mutation showed markedly decreased sensitivity to cisplatin-induced apoptosis, attributed to impaired induction of p73 but not p53. In agreement with these results, DNA damage-induced apoptosis and G2/M arrest were also decreased in SHP-2/p53 double mutant embryonic fibroblasts. Collectively, these studies have further defined the mechanisms by which SHP-2 phosphatase regulates DNA damage responses.


Subject(s)
Apoptosis , Gene Expression Regulation, Enzymologic , Intracellular Signaling Peptides and Proteins/metabolism , Protein Tyrosine Phosphatases/metabolism , Animals , Catalysis , Cell Cycle , Cell Cycle Proteins/metabolism , Cell Division , Cell Nucleus/metabolism , Cell Separation , Cisplatin/pharmacology , Cytoplasm/metabolism , DNA Damage , DNA, Complementary/metabolism , DNA-Binding Proteins/metabolism , Exons , Fibroblasts/metabolism , Flow Cytometry , G2 Phase , Gene Deletion , Gene Transfer Techniques , Genes, Tumor Suppressor , Immunoblotting , Immunoprecipitation , Mice , Mitosis , Nuclear Proteins/metabolism , Protein Tyrosine Phosphatase, Non-Receptor Type 11 , Radiation, Ionizing , Retroviridae/genetics , Signal Transduction , Stem Cells/metabolism , Tumor Protein p73 , Tumor Suppressor Protein p53/metabolism , Tumor Suppressor Proteins , cdc25 Phosphatases/metabolism
9.
J Biol Chem ; 278(44): 42812-20, 2003 Oct 31.
Article in English | MEDLINE | ID: mdl-12937170

ABSTRACT

DNA damage induced by radiation or DNA-damaging agents leads to apoptosis and cell cycle arrest. However, DNA damage-triggered signal transduction involved in these cellular responses is not well understood. We previously demonstrated an important role for SHP-2, a ubiquitously expressed SH2 domain-containing tyrosine phosphatase, in the DNA damage-induced apoptotic response. Here we report a potential role for SHP-2 in a DNA damage-activated cell cycle checkpoint. Cell cycle analysis and the mitotic index assay showed that following DNA damage induced by cisplatin or gamma-irradiation, the G2 (but not S) arrest response was diminished in SV40 large T antigen-immortalized embryonic fibroblast cells lacking functional SHP-2. Notably, reintroduction of wild-type SHP-2 into the mutant cells fully restored the DNA damage-induced G2 arrest response, suggesting a direct role of SHP-2 in the G2/M checkpoint. Further biochemical analysis revealed that SHP-2 constitutively associated with 14-3-3beta, and that Cdc25C cytoplasmic translocation induced by DNA damage was essentially blocked in SHP-2 mutant cells. Additionally, we showed that following DNA damage, activation of p38 kinase was significantly elevated, while Erk kinase activation was decreased in mutant cells, and treatment of SHP-2 mutant cells with SB203580, a selective inhibitor for p38 kinase, partially restored the DNA damage-induced G2 arrest response. These results together provide the first evidence that SHP-2 tyrosine phosphatase enhances the DNA damage G2/M checkpoint in SV40 large T antigen immortalized murine embryonic fibroblast cells.


Subject(s)
Antigens, Polyomavirus Transforming/metabolism , DNA Damage , Fibroblasts/metabolism , G2 Phase , Mitosis , Protein Tyrosine Phosphatases/metabolism , Animals , Caffeine/pharmacology , Cell Cycle , Cell Cycle Proteins/metabolism , Cells, Cultured , Cisplatin/pharmacology , Cytoplasm/metabolism , DNA/drug effects , DNA/radiation effects , Enzyme Inhibitors/pharmacology , Gamma Rays , Immunoblotting , Intracellular Signaling Peptides and Proteins , Mice , Nocodazole/pharmacology , Precipitin Tests , Protein Transport , Protein Tyrosine Phosphatase, Non-Receptor Type 11 , SH2 Domain-Containing Protein Tyrosine Phosphatases , Time Factors , cdc25 Phosphatases/metabolism , src Homology Domains
10.
J Biol Chem ; 278(17): 15208-16, 2003 Apr 25.
Article in English | MEDLINE | ID: mdl-12594211

ABSTRACT

SHP-2, a ubiquitously expressed Src hmology 2 (SH2) domain-containing tyrosine phosphatase, plays a critical role in the regulation of growth factor and cytokine signal transduction. Here we report a novel function of this phosphatase in DNA damage-induced cellular responses. Mutant embryonic fibroblast cells lacking functional SHP-2 showed significantly decreased apoptosis in response to DNA damage. Following cisplatin treatment, induction of p73 and its downstream effector p21(Cip1) was essentially blocked in SHP-2 mutant cells. Further investigation revealed that activation of the nuclear tyrosine kinase c-Abl, an essential mediator in DNA damage induction of p73, was impaired in the mutant cells, suggesting a functional requirement of SHP-2 in c-Abl activation. Consistent with this observation, the effect of overexpression of c-Abl kinase in SHP-2 mutant cells on sensitizing the cells to DNA damage-induced death was abolished. Additionally, we found that in embryonic fibroblast cells 30-40% of SHP-2 was localized in the nuclei, and that a fraction of nuclear SHP-2 was constitutively associated with c-Abl via its SH3 domain. Phosphatase activity of nuclear but not cytoplasmic SHP-2 was significantly enhanced in response to DNA damage. These results together suggest a novel nuclear function for SHP-2 phosphatase in the regulation of DNA damage-induced apoptotic responses.


Subject(s)
Apoptosis , DNA Damage , Protein Tyrosine Phosphatases/physiology , Animals , Apoptosis/drug effects , Cisplatin/pharmacology , Cyclin-Dependent Kinase Inhibitor p21 , Cyclins/biosynthesis , DNA-Binding Proteins/biosynthesis , Embryo, Mammalian/cytology , Fibroblasts/cytology , Fibroblasts/enzymology , Gene Expression Regulation/drug effects , Genes, Tumor Suppressor , Intracellular Signaling Peptides and Proteins , Mice , Mice, Knockout , Mutagens/pharmacology , Nuclear Proteins/biosynthesis , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Nuclear Proteins/physiology , Protein Tyrosine Phosphatase, Non-Receptor Type 11 , Protein Tyrosine Phosphatases/genetics , Protein Tyrosine Phosphatases/metabolism , Proto-Oncogene Proteins c-abl/metabolism , SH2 Domain-Containing Protein Tyrosine Phosphatases , Signal Transduction , Tumor Protein p73 , Tumor Suppressor Proteins
SELECTION OF CITATIONS
SEARCH DETAIL
...