Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
1.
Pediatr Blood Cancer ; 68(11): e29046, 2021 11.
Article in English | MEDLINE | ID: mdl-33939263

ABSTRACT

BACKGROUND: The biotherapeutic asparaginase is a cornerstone of therapy in acute lymphoblastic leukaemia (ALL). With limited access to the original native Escherichia coli-derived asparaginase (EcASNase), a variety of EcASNase biogenerics are used in low-middle-income countries (LMICs). The variable quality of these biogenerics potentially influences clinical outcomes. PROCEDURE: Seven biogeneric EcASNases (P1-P7) marketed widely in India were evaluated, with P2 as an exemplar for in vivo monitoring. Therapeutic activity of P2 (10,000 IU/m2 /dose, intramuscular, every 72 hours) was monitored during induction therapy, and drug-related toxicities recorded. Molecular identity, purity and in vitro drug activity of seven biogenerics were characterised using multimodal analyses, and findings compared with reference EcASNase (R). RESULTS: In patients (N = 62) receiving P2, subtherapeutic asparaginase activity (<100 U/L) was observed in 66% (46/70) of trough timepoints (72 hours postdose) during induction. Twelve patients (19%), 11 with high-risk ALL, developed hypersensitivity. Isoforms of EcASNase were identified in all seven biogenerics. All generic products contained impurities with batch-to-batch variability. These included high levels of protein aggregates and host cell protein contamination. In vitro assays of EcASNase activity and leukaemia cell line cytotoxicity were not discriminatory. CONCLUSIONS: Our findings confirm widespread concerns over the unsatisfactory quality and therapeutic activity of native EcASNase biogenerics marketed in LMICs. Appropriate use of these products requires monitored studies to identify clinical suitability and determine appropriate dosing and schedule. For large parts of the world, assured access to high-quality asparaginases remains an unmet therapeutic need.


Subject(s)
Antineoplastic Agents , Asparaginase , Biological Products/therapeutic use , Drugs, Generic/therapeutic use , Precursor Cell Lymphoblastic Leukemia-Lymphoma , Antineoplastic Agents/therapeutic use , Asparaginase/therapeutic use , Escherichia coli/enzymology , Humans , India , Precursor Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Therapeutic Equivalency
2.
Lab Invest ; 93(6): 639-45, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23608754

ABSTRACT

Gastrin-releasing peptide (GRP) is a proangiogenic ligand secreted by tumors and acts directly upon binding to GRP receptor in endothelial cells. Angiogenesis plays a critical role in the pathology of various diseases, including cancer, as the formation of new blood vessels potentiates the rate of tumor growth and dissemination. GRP increases the migration of endothelial cells, but much is unknown about its role on endothelial cell proliferation and survival, as well as the signaling pathways involved. In the present study, we showed that GRP increases endothelial cell proliferation and tubule formation. There was a time-dependent increase in the levels of phosphorylated AKT, mammalian target of rapamycin (mTOR), and S6R in human umbilical vein endothelial cells treated with GRP. Interestingly, GRP treatment decreased the expression of proautophagic factors, ATG5, BECN1, and LC3 proteins. GRP also attenuated rapamycin-induced formation of autophagosomes. Moreover, overexpression of ATG5 or BECN1 significantly decreased tubule formation induced by exogenous GRP, whereas siRNA against ATG5 or BECN1 resulted in increased tubule formation with GRP treatment. Our results show that GRP inhibits the process of autophagy in vascular endothelial cells, thereby increasing endothelial cell proliferation and tubule formation. Here, we describe a novel role of GRP in the regulation of autophagy of endothelial cells, thereby providing a potential new therapeutic strategy in targeting angiogenesis during cancer progression.


Subject(s)
Autophagy , Cell Proliferation , Endothelial Cells/physiology , Gastrin-Releasing Peptide/physiology , Neovascularization, Pathologic , Apoptosis Regulatory Proteins/metabolism , Autophagy-Related Protein 5 , Beclin-1 , Coculture Techniques , Endothelium, Vascular/growth & development , Gene Expression Regulation , Human Umbilical Vein Endothelial Cells , Humans , Membrane Proteins/metabolism , Microtubule-Associated Proteins/metabolism , Paracrine Communication , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction , TOR Serine-Threonine Kinases/metabolism
3.
Biochem Biophys Res Commun ; 435(2): 295-9, 2013 May 31.
Article in English | MEDLINE | ID: mdl-23618860

ABSTRACT

Gastrin-releasing peptide (GRP) and its receptor (GRP-R) are highly expressed in undifferentiated neuroblastoma, and they play critical roles in oncogenesis. We previously reported that GRP activates the PI3K/AKT signaling pathway to promote DNA synthesis and cell cycle progression in neuroblastoma cells. Conversely, GRP-R silencing induces cell cycle arrest. Here, we speculated that GRP/GRP-R signaling induces neuroblastoma cell proliferation via regulation of cyclin-dependent kinase (CDK) inhibitors. Surprisingly, we found that GRP/GRP-R differentially induced expressions of p21 and p27. Silencing GRP/GRP-R decreased p21, but it increased p27 expressions in neuroblastoma cells. Furthermore, we found that the intracellular localization of p21 and p27 in the nuclear and cytoplasmic compartments, respectively. In addition, we found that GRP/GRP-R silencing increased the expression and accumulation of PTEN in the cytoplasm of neuroblastoma cells where it co-localized with p27, thus suggesting that p27 promotes the function of PTEN as a tumor suppressor by stabilizing PTEN in the cytoplasm. GRP/GRP-R regulation of CDK inhibitors and tumor suppressor PTEN may be critical for tumoriogenesis of neuroblastoma.


Subject(s)
Cyclin-Dependent Kinase Inhibitor p27/metabolism , Gastrin-Releasing Peptide/metabolism , Gene Expression Regulation, Neoplastic , Neuroblastoma/enzymology , PTEN Phosphohydrolase/metabolism , Receptors, Bombesin/metabolism , Cell Line, Tumor , Cyclin-Dependent Kinase Inhibitor p21 , Humans
4.
Biochem Biophys Res Commun ; 424(3): 421-6, 2012 Aug 03.
Article in English | MEDLINE | ID: mdl-22766505

ABSTRACT

Neuroblastoma, the most common extra-cranial solid tumor in infants and children, is characterized by a high rate of spontaneous remissions in infancy. Retinoic acid (RA) has been known to induce neuroblastoma differentiation; however, the molecular mechanisms and signaling pathways that are responsible for RA-mediated neuroblastoma cell differentiation remain unclear. Here, we sought to determine the cell signaling processes involved in RA-induced cellular differentiation. Upon RA administration, human neuroblastoma cell lines, SK-N-SH and BE(2)-C, demonstrated neurite extensions, which is an indicator of neuronal cell differentiation. Moreover, cell cycle arrest occurred in G1/G0 phase. The protein levels of cyclin-dependent kinase inhibitors, p21 and p27(Kip), which inhibit cell proliferation by blocking cell cycle progression at G1/S phase, increased after RA treatment. Interestingly, RA promoted cell survival during the differentiation process, hence suggesting a potential mechanism for neuroblastoma resistance to RA therapy. Importantly, we found that the PI3K/AKT pathway is required for RA-induced neuroblastoma cell differentiation. Our results elucidated the molecular mechanism of RA-induced neuroblastoma cellular differentiation, which may be important for developing novel therapeutic strategy against poorly differentiated neuroblastoma.


Subject(s)
Extracellular Signal-Regulated MAP Kinases/metabolism , Neuroblastoma/enzymology , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Tretinoin/pharmacology , Cell Differentiation , G1 Phase Cell Cycle Checkpoints , Humans , Neuroblastoma/pathology , Phosphorylation , Resting Phase, Cell Cycle
5.
Org Biomol Chem ; 7(1): 94-102, 2009 Jan 07.
Article in English | MEDLINE | ID: mdl-19081951

ABSTRACT

The development of new antibiotics is of increasing importance due to the growing resistance power of microbes against conventional drugs. To this end, cationic peptides are emerging as clinically potent antimicrobial agents. In the present study, we have synthesized six dipeptide-based cationic amphiphiles with different head group structures by varying combinations of l-amino acid residues. These amphiphiles showed remarkable growth inhibiting activity on several Gram-positive (minimum inhibitory concentration (MIC)=0.1-10 microg/mL) and Gram-negative (MIC=5-150 microg/mL) bacteria as well as on fungus (MIC=1-50 microg/mL). The inherent antimicrobial efficacies of these cationic dipeptides were influenced by the head group architecture of the amphiphiles. Encouragingly, these amphiphiles selectively attacked microbial cells, while showing biocompatibility toward mammalian cells. The results show that the rational designing of short peptide-based cationic amphiphiles might serve as a promising strategy in the development of antimicrobial agents with greater cell specificities. In addition, the amphiphiles showed water gelation ability at room temperature. The formation of non-covalent supramolecular networks in gelation was established by microscopic and spectroscopic studies.


Subject(s)
Anti-Infective Agents/chemical synthesis , Anti-Infective Agents/pharmacology , Biocompatible Materials/chemistry , Animals , Cations , Cell Line, Tumor , Gram-Negative Bacteria/drug effects , Gram-Positive Bacteria/drug effects , HeLa Cells , Humans , Kinetics , Models, Chemical , Peptides/chemistry , Temperature , Time Factors , Water/chemistry
6.
Anticancer Res ; 38(2): 647-654, 2018 02.
Article in English | MEDLINE | ID: mdl-29374686

ABSTRACT

BACKGROUND/AIM: Sirtuins (SIRTs) play crucial roles in various signaling pathways that modulate differentiation and proliferation. We sought to elucidate the role of SIRTs in differentiation and proliferation of human neuroblastoma (NB). MATERIALS AND METHODS: NB cells were treated with nicotinamide (NAM), a non-specific SIRT inhibitor, SIRT-targeted short hairpin RNAs, and retinoic acid to assess cell growth and differentiation. RESULTS: SIRTs are involved in proliferation and differentiation using NAM in BE(2)-C cells. Specifically, SIRT6 knockdown in BE(2)-C cells reduced cell proliferation, induced neurite extension, corresponding with induction of p21CIP1 expression and G1 cell-cycle arrest. These effects were rescued by forced re-overexpression of SIRT6. SIRT6 expression was reduced in differentiated human NB sections, and RA-induced differentiation in BE(2)-C cells. CONCLUSION: SIRTs have important oncogenic properties in NB beyond its established functions in aging and genome stability. SIRT6 may represent a novel target for developing future therapeutics for the treatment of aggressive NBs.


Subject(s)
Cell Differentiation/drug effects , Neuroblastoma/drug therapy , Neuroblastoma/metabolism , Sirtuins/antagonists & inhibitors , Cell Line, Tumor , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , Gene Knockdown Techniques , Humans , Neuroblastoma/pathology , Niacinamide/pharmacology , RNA, Small Interfering/pharmacology , Sirtuins/genetics , Tretinoin/administration & dosage , Tretinoin/pharmacology
7.
Oncotarget ; 8(47): 82609-82620, 2017 Oct 10.
Article in English | MEDLINE | ID: mdl-29137288

ABSTRACT

Approximately two-thirds of patients with neuroblastoma are found to have metastatic disease at time of diagnosis with frequent skeletal, lymph node, central nervous system, and liver involvement. Using a serial in vivo splenic injection model, we have isolated an aggressive subclone (BE(2)-C/LM2) from MYCN-amplified neuroblastomas that demonstrate an enhanced propensity to develop metastatic liver lesions. BE(2)-C/LM2 subclone cells demonstrate increased adherent, soft agar colony and tumorsphere growth in vitro. Components of the tumor microenvironment regulate cancer progression, via networks of cytokines and growth factors. Cytokine array analysis identified increased TIMP-1 in the plasma of mice injected with BE(2)-C/LM2 subclone cells, leading us to hypothesize that TIMP-1 may play a role in our observed prometastatic phenotype. Immunoblotting and ELISA demonstrated enhanced endogenous TIMP-1 expression in our isolated neuroblastoma subclone. Silencing endogenous TIMP-1 successfully blocked in vitro proliferation, soft agar colony formation and tumorsphere formation by BE(2)-C/LM2 cells. Stable RNA interference of endogenous TIMP-1 failed to reverse the prometastatic phenotype of our BE(2)-C/LM2 subclone in our liver metastasis model, suggesting that endogenous TIMP-1 levels may not be an essential component of this in vivo behavior. Notably, tissue microarray analysis and Kaplan-Meier by gene expression demonstrates that elevated TIMP-1 expression is correlated with increased disease relapse and mortality in patients with neuroblastoma. Taken together, our study identifies TIMP-1 as a novel soluble factor that is associated with a prometastatic phenotype in our in vivo model and adverse outcomes in patients with neuroblastoma.

8.
Surgery ; 158(3): 827-36, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26088922

ABSTRACT

BACKGROUND: Reactive oxygen species (ROS) contribute to adult tumorigenesis; however, their roles in pediatric solid tumors are unknown. Here, we sought to define the steady-state ROS levels in neuroblastoma and to examine whether aggressive cellular behavior, which may predict treatment failure, is regulated by ROS. METHODS: Neuroblastoma sections were assessed for 4-hydroxynonenal (4-HNE), a marker of intracellular lipid peroxidation and a byproduct of increased levels of ROS. Human neuroblastoma cell lines, MYCN-amplified BE(2)-C and MYCN-nonamplified SK-N-SH, were examined in our study. Superoxide and hydroperoxide oxidation products were detected by staining for dihydroethidium (DHE) and 5, 6-carboxy-2', 7'-dichlorodihydrofluorescein diacetate (CDCFH2), using the oxidation-insensitive analog CDCF as a negative control. Cells were treated with N-acetylcysteine (NAC; 10 mmol/L) daily for 5 days and analyzed. RESULTS: Greater expression of 4-HNE was observed in undifferentiated tumor sections as compared with the more differentiated tumors. Interestingly, increased levels of ROS were detected in MYCN-amplified BE(2)-C cells. Moreover, gastrin-releasing peptide receptor-induced ROS production stimulated upregulation of the hypoxia inducible factor (HIF)-1α/vascular endothelial growth factor (VEGF) pathway and an increase in cell growth. Antioxidant NAC decreased HIF-1α/VEGF expression and inhibited BE(2)-C cell growth. CONCLUSION: We report a novel observation that shifting the redox balance toward greater ROS levels results in a more aggressive neuroblastoma phenotype. Our data suggest that ROS play a critical role in refractory neuroblastoma.


Subject(s)
Acetylcysteine/pharmacology , Antioxidants/pharmacology , Biomarkers, Tumor/metabolism , Cell Proliferation/drug effects , Neuroblastoma/metabolism , Reactive Oxygen Species/antagonists & inhibitors , Blotting, Western , Cell Line, Tumor , Humans , Immunohistochemistry , Reactive Oxygen Species/metabolism
9.
Anticancer Res ; 34(5): 2269-74, 2014 May.
Article in English | MEDLINE | ID: mdl-24778030

ABSTRACT

BACKGROUND/AIM: It is unknown whether hypoxia regulates aurora kinase A (AURKA), a serine/threonine kinase, in neuroblastoma to stimulate cell growth or migration. We sought to determine whether AURKA mediates hypoxia-induced regulation of neuroblastoma tumorigenicity. MATERIALS AND METHODS: Human neuroblastoma BE(2)-C cells were treated with CoCl2, a chemical hypoxia mimetic, and MLN8237, a pharmalogical inhibitor of AURKA, to assess cell viability, colony formation and transwell migration. Focal adhesion kinase (FAK) expression was analyzed after silencing of AURKA under normoxic vs. hypoxic conditions. RESULTS: Hypoxia up-regulated expression of AURKA mRNA and protein. CoCl2 stimulated cell proliferation and migration, while inhibiting colony formation. MLN8237 reduced colony formation and cell migration. Silencing of AURKA reduced expression of FAK and pFAK under normoxia and hypoxia. CONCLUSION: Hypoxia positively regulates AURKA expression. Hypoxia-induced stimulation of colony formation and migration is, in part, mediated by AURKA. These findings establish that AURKA is a critical regulator of hypoxia-mediated tumor progression in neuroblastoma.


Subject(s)
Aurora Kinase A/metabolism , Carcinogenesis/metabolism , Neuroblastoma/metabolism , Cell Hypoxia/physiology , Cell Line, Tumor , Cell Movement/physiology , Cell Proliferation , Humans , Immunoblotting , RNA Interference , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Transfection
10.
J Pediatr Surg ; 49(1): 159-65, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24439602

ABSTRACT

PURPOSE: Aurora kinase A (AURKA) overexpression is associated with poor prognosis in neuroblastoma and has been described to upregulate VEGF in gastric cancer cells. However, the exact role of AURKA in the regulation of neuroblastoma tumorigenesis remains unknown. We hypothesize that AURKA-mediated stabilization of N-Myc may affect VEGF expression and angiogenesis in neuroblastoma. Therefore, we sought to determine whether inhibition of AURKA modulates neuroblastoma angiogenesis. METHODS: Cell viability and anchorage-independent growth were determined after silencing AURKA or after treatment with MLN8237, AURKA inhibitor. Immunofluorescence was used to determine N-Myc localization. Human umbilical vein endothelial cells (HUVECs) were used to assess angiogenesis in vitro. Real time-PCR and ELISA were performed to determine VEGF transcription and secretion, respectively. RESULTS: Knockdown of AURKA significantly reduced cell proliferation and inhibited anchorage-independent growth. It also decreased N-Myc protein levels and nuclear localization. AURKA inhibition also decreased HUVECs tubule formation along with VEGF transcription and secretion. Similarly, MLN8237 treatment decreased neuroblastoma tumorigenicity in vitro. CONCLUSIONS: Our findings demonstrate that AURKA plays a critical role in neuroblastoma angiogenesis. AURKA regulates nuclear translocation of N-Myc in neuroblastoma cells, thus potentially affecting cell proliferation, anchorage-independent cell growth, and angiogenesis. Targeting AURKA might provide a novel therapeutic strategy in treating aggressive neuroblastomas.


Subject(s)
Antineoplastic Agents/pharmacology , Aurora Kinase A/antagonists & inhibitors , Azepines/pharmacology , Molecular Targeted Therapy , Neoplasm Proteins/antagonists & inhibitors , Neovascularization, Pathologic/drug therapy , Neuroblastoma/pathology , Protein Kinase Inhibitors/pharmacology , Pyrimidines/pharmacology , RNA, Small Interfering/pharmacology , Aurora Kinase A/physiology , Cell Adhesion/drug effects , Cell Division/drug effects , Cell Line, Tumor , Gene Expression Regulation, Neoplastic/drug effects , Human Umbilical Vein Endothelial Cells , Humans , In Vitro Techniques , Neoplasm Proteins/physiology , Neovascularization, Pathologic/enzymology , Neovascularization, Pathologic/etiology , Neuroblastoma/blood supply , Neuroblastoma/drug therapy , Neuroblastoma/enzymology , Protein Transport/drug effects , Proto-Oncogene Proteins c-myc/physiology , RNA, Neoplasm/biosynthesis , Tumor Stem Cell Assay , Vascular Endothelial Growth Factor A/biosynthesis , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor A/metabolism
11.
Surgery ; 154(2): 369-75, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23889963

ABSTRACT

BACKGROUND: Gastrin-releasing peptide (GRP) and its receptor, GRP-R, are critically involved in neuroblastoma tumorigenesis; however, the molecular mechanisms and signaling pathways that are responsible for GRP/GRP-R-induced cell migration and invasion remain unclear. In this study, we sought to determine the cell signals involved in GRP/GRP-R-mediated neuroblastoma cell migration and invasion. METHODS: Human neuroblastoma cell lines SK-N-SH, LAN-1, and IMR-32 were used for our study. Transwell migration and invasion assays were performed after GRP (10(-7) M) stimulation. The cDNA GEArray Microarray kit was used to determine GRP-R-induced gene expression changes. Protein and membrane expression of integrin subunits were confirmed by Western blotting and flow cytometry analysis. siRNA transfection was performed using Lipofectamine 2000. For scratch assay, a confluent monolayer of cells in 6-well plates were wounded with micropipette tip and observed microscopically at 24 to 72 h. RESULTS: GRP increased neuroblastoma cell migration and expressions of MMP-2 whereas the TIMP-1 level decreased. GRP-R overexpression stimulated SK-N-SH cell migration and upregulated integrin α2, α3, and ß1 protein as well as mRNA expression. Targeted silencing of integrin ß1 inhibited cell migration. CONCLUSION: GRP/GRP-R signaling contributes to neuroblastoma cell migration and invasion. Moreover, the integrin ß1 subunit critically regulates GRP-R-mediated neuroblastoma cell migration and invasion.


Subject(s)
Cell Movement , Integrin beta1/physiology , Neuroblastoma/pathology , Receptors, Bombesin/physiology , Cell Line, Tumor , Gastrin-Releasing Peptide/pharmacology , Humans , Integrin alpha2/analysis , Integrin alpha3/analysis , Neoplasm Invasiveness
12.
Oncotarget ; 4(8): 1149-57, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23900341

ABSTRACT

Activation of the Hedgehog (Hh) signaling pathway has been implicated in a variety of malignancies including neuroblastoma. Expression of Gli1, a downstream effector of Hh, correlates with a favorable prognosis in patients with neuroblastoma. Moreover, Gli1 overexpression reduces mitotic index and induces transcription of genes involved in the differentiation of neuroblastoma cells; however, much remains unknown regarding the regulation of Gli1 transcriptional activity. Here, we report a novel negative regulation of Gli1 transcriptional activity by PI3K/AKT2 signal transduction pathway. Constitutively active PI3K subunit, p110α, inhibited Gli1 transcriptional activity in neuroblastoma cells, whereas, overexpression of an inactive form of PI3K subunit, p85, enhanced its activity. Specifically, the AKT2 isoform inhibited Gli1 luciferase activity. Silencing AKT2 using siRNA increased Gli1 transcriptional activity and conversely, overexpression of constitutively active AKT2 (myr-AKT2) decreased Gli1 transcriptional activity. Furthermore, Gli1 overexpression-mediated decrease in anchorage-independent growth was rescued by AKT2 overexpression. We also demonstrated that AKT2 overexpression regulates the nuclear-cytoplasmic distribution of exogenous Gli1 protein in neuroblastoma cells by relieving a GSK3ß-mediated destabilization of SUFU, a negative regulator of Gli1 nuclear translocation. Inhibition of nuclear Gli1 accumulation may explain for the suppression of the tumor-suppressive function of Gli1. Collectively, our findings suggest an important role of Gli1 as a tumor suppressor in neuroblastoma, and offer a mechanism by which AKT2 regulates the subcellular localization, and in turn, inhibits the tumor-suppressive function of Gli1 in neuroblastoma.


Subject(s)
Neuroblastoma/genetics , Proto-Oncogene Proteins c-akt/genetics , Transcription Factors/genetics , Cell Differentiation/physiology , Cell Line, Tumor , Humans , Neuroblastoma/enzymology , Neuroblastoma/metabolism , Phosphatidylinositol 3-Kinases/genetics , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction , Transcription Factors/metabolism , Zinc Finger Protein GLI1
13.
Autophagy ; 9(10): 1579-90, 2013 Oct.
Article in English | MEDLINE | ID: mdl-24108003

ABSTRACT

Neuroblastoma is characterized by florid vascularization leading to rapid tumor dissemination to distant organs; angiogenesis contributes to tumor progression and poor clinical outcomes. We have previously demonstrated an increased expression of gastrin-releasing peptide (GRP) and its receptor, GRPR, in neuroblastoma and that GRP activates the PI3K-AKT pathway as a proangiogenic factor during tumor progression. Interestingly, AKT activation phosphorylates MTOR, a critical negative regulator of autophagy, a cellular process involved in the degradation of key proteins. We hypothesize that inhibition of GRPR enhances autophagy-mediated degradation of GRP and subsequent inhibition of angiogenesis in neuroblastoma. Here, we demonstrated a novel phenomenon where targeting GRPR using shRNA or a specific antagonist, RC-3095, decreased GRP secretion by neuroblastoma cells and tubule formation by endothelial cells in vitro. Furthermore, shGRPR or RC-3095 treatment enhanced expression of proautophagic proteins in human neuroblastoma cell lines, BE(2)-C, and BE(2)-M17. Interestingly, rapamycin, an inhibitor of MTOR, enhanced the expression of the autophagosomal marker LC3-II and GRP was localized within LC3-II-marked autophagosomes in vitro as well as in vivo, indicating autophagy-mediated degradation of GRP. Moreover, overexpression of ATG5 or BECN1 attenuated GRP secretion and tubule formation, whereas opposite effects were observed with siRNA silencing of ATG5 and BECN1. Our data supported the role of autophagy in the degradation of GRP and subsequent inhibition of angiogenesis. Therefore, activation of autophagy may lead to novel antivascular therapeutic strategies in the treatment of highly vascular neuroblastomas.


Subject(s)
Autophagy/physiology , Gastrin-Releasing Peptide/metabolism , Neovascularization, Pathologic/metabolism , Neuroblastoma/metabolism , Autophagy/genetics , Cell Line, Tumor , Cell Proliferation/physiology , Endothelial Cells/metabolism , Gene Expression Regulation, Neoplastic , Humans , Neovascularization, Pathologic/genetics , Neuroblastoma/blood supply , Neuroblastoma/pathology , Proteolysis , Proto-Oncogene Proteins c-akt/metabolism , RNA, Small Interfering/metabolism
14.
PLoS One ; 8(2): e56382, 2013.
Article in English | MEDLINE | ID: mdl-23468863

ABSTRACT

Activation of PI3K/AKT pathway correlates with poor prognosis in patients with neuroblastoma. Our previous studies have demonstrated that PI3K/AKT signaling is critical for the oncogenic transformations induced by gastrin-releasing peptide (GRP) and its receptor, GRP-R, in neuroblastoma. Moreover, PI3K/AKT-dependent oncogenic transformations require N-myc, an extensively studied oncogene in neuroblastoma. Whether AKT directly regulates the expression of N-myc oncogene is yet to be determined. Here, we report a novel finding that of the three AKT isoforms, AKT2 specifically regulated N-myc expression in neuroblastoma cells. We also confirmed that GRP-R is upstream of AKT2 and in turn, regulated N-myc expression via AKT2 in neuroblastoma cells. Functional assays demonstrated that attenuation of AKT2 impaired cell proliferation and anchorage-independent cell growth, and decreased the secretion of angiogenic factor VEGF in vitro. Furthermore, silencing AKT2 inhibited migration and invasion of neuroblastoma cells in vitro. Xenografts established by injecting AKT2 silenced human neuroblastoma cells into murine spleen expressed decreased levels of AKT2 and resulted in fewer liver metastases compared to controls in vivo. Hence, our study highlights the potential molecular mechanism(s) mediating the oncogenic role of GRP/GRP-R and demonstrates a novel role for AKT2 in neuroblastoma tumorigenesis, indicating that targeting the GRP/GRP-R/AKT2 axis may be important for developing novel therapeutics in the treatment of clinically aggressive neuroblastoma.


Subject(s)
Neuroblastoma/genetics , Neuroblastoma/pathology , Proto-Oncogene Proteins c-akt/genetics , Animals , Cell Line, Tumor , Cell Transformation, Neoplastic/genetics , Disease Models, Animal , Gene Expression Regulation, Neoplastic , Gene Silencing , Genes, myc , Humans , Liver Neoplasms/genetics , Liver Neoplasms/secondary , Male , Mice , Neoplasm Metastasis , Neuroblastoma/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Receptors, G-Protein-Coupled
15.
Surgery ; 154(2): 226-33, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23806264

ABSTRACT

BACKGROUND: microRNA (miRNA) functions broadly as post-transcriptional regulators of gene expression, and disproportionate miRNAs can result in dysregulation of oncogenes in cancer cells. We have previously shown that gastrin-releasing peptide receptor (GRP-R) signaling regulates tumorigenicity of neuroblastoma cells. Herein, we sought to characterize miRNA profile in GRP-R silenced neuroblastoma cells, and to determine the role of miRNAs on tumorigenicity and metastatic potential. METHODS: Human neuroblastoma cell lines, BE(2)-C and SK-N-SH, were used for our study. Stably transfected GRP-R silenced cells were assessed for miRNA profiles. Cells were transfected with miR-335, miR-363, or miR-CON, a nontargeting control, and in vitro assays were performed. In vivo functions of miR-335 and miR-363 were also assessed in a spleen-liver metastasis murine model. RESULTS: GRP-R silencing significantly increased expression of miR-335 and miR-363 in BE(2)-C cells. Overexpression of miR-335 and miR-363 decreased tumorigenicity as measured by clonogenicity, anchorage-independent growth, and metastasis determined by cell invasion assay and liver metastasis in vivo. CONCLUSION: We report, for the first time, that GRP-R-mediated tumorigenicity and increased metastatic potential in neuroblastoma are regulated, in part, by miR-335 and miR-363. A better understanding of the anti-tumor functions of miRNAs could provide valuable insights to discerning molecular mechanisms responsible for neuroblastoma metastasis.


Subject(s)
Cell Transformation, Neoplastic , MicroRNAs/physiology , Neuroblastoma/etiology , ADAM Proteins/genetics , Animals , Cell Line, Tumor , Humans , Liver Neoplasms, Experimental/secondary , Male , Membrane Proteins/genetics , Mice , Neoplasm Invasiveness , Neuroblastoma/pathology , Neuroblastoma/secondary , Receptors, Bombesin/analysis , Receptors, Bombesin/physiology
16.
PLoS One ; 8(9): e72570, 2013.
Article in English | MEDLINE | ID: mdl-24039782

ABSTRACT

We have previously demonstrated the role of gastrin-releasing peptide (GRP) as an autocrine growth factor for neuroblastoma. Here, we report that GRP silencing regulates cell signaling involved in the invasion-metastasis cascade. Using a doxycycline inducible system, we demonstrate that GRP silencing decreased anchorage-independent growth, inhibited migration and neuroblastoma cell-mediated angiogenesis in vitro, and suppressed metastasis in vivo. Targeted inhibition of GRP decreased the mRNA levels of oncogenes responsible for neuroblastoma progression. We also identified PTEN/AKT signaling as a key mediator of the tumorigenic properties of GRP in neuroblastoma cells. Interestingly, PTEN overexpression decreased GRP-mediated migration and angiogenesis; a novel role for this, otherwise, understated tumor suppressor in neuroblastoma. Furthermore, activation of AKT (pAKT) positively correlated with neuroblastoma progression in an in vivo tumor-metastasis model. PTEN expression was slightly decreased in metastatic lesions. A similar phenomenon was observed in human neuroblastoma sections, where, early-stage localized tumors had a higher PTEN expression relative to pAKT; however, an inverse expression pattern was observed in liver lesions. Taken together, our results argue for a dual purpose of targeting GRP in neuroblastoma--1) decreasing expression of critical oncogenes involved in tumor progression, and 2) enhancing activation of tumor suppressor genes to treat aggressive, advanced-stage disease.


Subject(s)
Gastrin-Releasing Peptide/genetics , Liver Neoplasms/metabolism , Neuroblastoma/metabolism , PTEN Phosphohydrolase/metabolism , Animals , Cell Adhesion , Cell Line, Tumor , Cell Movement , Disease Progression , Enzyme Activation , Gastrin-Releasing Peptide/metabolism , Gene Knockdown Techniques , Human Umbilical Vein Endothelial Cells/physiology , Humans , Liver Neoplasms/secondary , Male , Mice , Mice, Nude , Neoplasm Transplantation , Neovascularization, Pathologic/metabolism , Neuroblastoma/pathology , Phenotype , Proto-Oncogene Proteins c-akt/metabolism , RNA Interference , RNA, Small Interfering/genetics , Signal Transduction
17.
Anticancer Res ; 32(11): 4691-6, 2012 Nov.
Article in English | MEDLINE | ID: mdl-23155231

ABSTRACT

BACKGROUND: Intracellular signaling responsible for gastrin-releasing peptide (GRP) receptor-mediated neovascularization is not clearly understood. We sought to determine the cellular mechanisms involved in the GRP receptor regulation of vascular endothelial growth factor (VEGF) release in neuroblastoma cells. MATERIALS AND METHODS: BE(2)-C cells were treated with bombesin (BBS), the amphibian equivalent of GRP, Phorbol myristate acetate (PMA) a PKC agonist, or GF109293X (GFX), and analyses were performed for VEGF secretion, phosphorylated protein kinase B (AKT), extracellular signal-regulated kinases (ERK) and protein kinase D (PKD) expression. RESULTS: BBS rapidly increased VEGF secretion at 30 min. Pre-treatment with PMA alone produced similar results; this effect was synergistic with the addition of GRP. Conversely, GFX blocked PMA-stimulated increase in VEGF secretion. Immunofluorescent staining for VEGF correlated to BBS, PMA and GFX. CONCLUSION: PKC is critically responsible for rapid VEGF secretion by GRP receptor signaling in neuroblastoma cells. Inhibition of VEGF significantly reduced GRP-mediated cell proliferation, suggesting its crucial role in neuroblastoma tumorigenesis.


Subject(s)
Bombesin/pharmacology , Neuroblastoma/enzymology , Neuroblastoma/metabolism , Protein Kinase C/metabolism , Vascular Endothelial Growth Factor A/metabolism , Blotting, Western , Cell Line, Tumor , Enzyme-Linked Immunosorbent Assay , Fluorescent Antibody Technique , Gastrin-Releasing Peptide/metabolism , Humans , Neovascularization, Pathologic/metabolism , Neuroblastoma/pathology , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/drug effects
18.
Oncotarget ; 3(12): 1576-87, 2012 Dec.
Article in English | MEDLINE | ID: mdl-23211542

ABSTRACT

Neuroblastomas express increased levels of gastrin-releasing peptide receptor (GRP-R). However, the exact molecular mechanisms involved in GRP-R-mediated cell signaling in neuroblastoma growth and metastasis are unknown. Here, we report that focal adhesion kinase (FAK), as a critical downstream target of GRP-R, is an important regulator of neuroblastoma tumorigenicity. We found that FAK expression correlates with GRP-R expression in human neuroblastoma sections and cell lines. GRP-R overexpression in SK-N-SH cells increased FAK, integrin α3 and ß1 expressions and cell migration. These cells demonstrated flatter cell morphology with broad lamellae, in which intense FAK expression was localized to the leading edges of lamellipodia. Interestingly, FAK activation was, in part, dependent on integrin α3 and ß1 expression. Conversely, GRP-R silencing decreased FAK as well as Mycn levels in BE(2)-C cells, which displayed a denser cellular morphology. Importantly, rescue experiments in GRP-R silenced BE(2)-C cells showed FAK overexpression significantly enhanced cell viability and soft agar colony formation; similarly, FAK overexpression in SK-N-SH cells also resulted in increased cell growth. These effects were reversed in FAK silenced BE(2)-C cells in vitro as well as in vivo. Moreover, we evaluated the effect of FAK inhibition in vivo. FAK inhibitor (Y15) suppressed GRP-induced neuroblastoma growth and metastasis. Our results indicate that FAK is a critical downstream regulator of GRP-R, which mediates tumorigenesis and metastasis in neuroblastoma.


Subject(s)
Cell Movement , Focal Adhesion Kinase 1/metabolism , Integrin alpha3/metabolism , Liver Neoplasms/enzymology , Liver Neoplasms/secondary , Neuroblastoma/enzymology , Neuroblastoma/secondary , Animals , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation , Cell Shape , Enzyme Activation , Focal Adhesion Kinase 1/antagonists & inhibitors , Focal Adhesion Kinase 1/genetics , Humans , Integrin beta1/metabolism , Liver Neoplasms/drug therapy , Liver Neoplasms/genetics , Male , Mice , Mice, Nude , N-Myc Proto-Oncogene Protein , Neoplasm Invasiveness , Neuroblastoma/drug therapy , Neuroblastoma/genetics , Nuclear Proteins/metabolism , Oncogene Proteins/metabolism , Protein Kinase Inhibitors/pharmacology , Pseudopodia/enzymology , RNA Interference , Receptors, Bombesin/genetics , Receptors, Bombesin/metabolism , Signal Transduction , Time Factors , Transfection , Xenograft Model Antitumor Assays
19.
Surgery ; 149(3): 425-32, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21035156

ABSTRACT

BACKGROUND: The overall survival for neuroblastoma remains dismal, in part due to the emergence of resistance to chemotherapeutic drugs. We have demonstrated that gastrin-releasing peptide (GRP), a gut peptide secreted by neuroblastoma, acts as an autocrine growth factor. We hypothesized that knockdown of GRP will induce apoptosis in neuroblastoma cells and potentiate the cytotoxic effects of chemotherapeutic agents. METHODS: The human neuroblastoma cell lines (JF, SK-N-SH) were transfected with small interfering (si) RNA targeted at GRP. Apoptosis was assessed by DNA fragmentation assay. Immunoblotting was used to confirm molecular markers of apoptosis, and flow cytometry was performed to determine cell cycle arrest after GRP knockdown. RESULTS: siGRP resulted in an increase in apoptosis in the absence of chemotherapeutic interventions. A combination of GRP silencing and chemotherapeutic drugs resulted in enhanced apoptosis when compared to either of the treatments alone. GRP silencing led to increased expression of proapoptotic proteins, p53 and p21. CONCLUSION: Silencing of GRP induces apoptosis in neuroblastoma cells; it acts synergistically with chemotherapeutic effects of etoposide and vincristine. GRP knockdown-mediated apoptosis appears to be associated with upregulation of p53 in neuroblastoma cells. Targeting GRP may be postulated as a potential novel agent for combinational treatment to treat aggressive neuroblastomas.


Subject(s)
Gastrin-Releasing Peptide/antagonists & inhibitors , Neuroblastoma/therapy , RNA, Small Interfering/genetics , Apoptosis/drug effects , Caspase 3/metabolism , Cell Cycle , Cell Line, Tumor , Cell Proliferation , Etoposide/pharmacology , Gastrin-Releasing Peptide/genetics , Humans , Neuroblastoma/pathology , Poly(ADP-ribose) Polymerases/metabolism , Transfection , Tumor Suppressor Protein p53/physiology , Vincristine/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL