1
|
Ribeiro M, Campos J, Pinho TS, Sampaio-Marques B, Barata-Antunes S, Cibrão JR, Araújo R, Duarte-Silva S, Moreira E, Sousa RA, Costa PM, Salgado AJ. Human platelet lysate supports SH-SY5Y neuroblastoma cell proliferation and differentiation into a dopaminergic-like neuronal phenotype under xenogeneic-free culture conditions. Biotechnol J 2024; 19:e2400068. [PMID: 38987218 DOI: 10.1002/biot.202400068] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Revised: 05/24/2024] [Accepted: 06/03/2024] [Indexed: 07/12/2024]
Abstract
SH-SY5Y is a human neuroblastoma cell line that can be differentiated into several neuronal phenotypes, depending on culture conditions. For this reason, this cell line has been widely used as an in vitro model of neurodegenerative conditions, such as Parkinson's disease (PD). However, most studies published to date used fetal bovine serum (FBS) as culture medium supplement for SH-SY5Y cell differentiation. We report on the testing of human platelet lysate (hPL) as a culture medium supplement to support SH-SY5Y cell culture. Both standard hPL and a fibrinogen-depleted hPL (FD-hPL) formulation, which does not require the addition of anticoagulants to culture media, promoted an increase in SH-SY5Y cell proliferation in comparison to FBS, without compromising metabolic activity. SH-SY5Y cells cultured in hPL or FD-hPL also displayed a higher number of neurite extensions and stained positive for MAP2 and synaptophysin, in the absence of differentiation stimuli; reducing hPL or FD-hPL concentration to 1% v/v did not affect cell proliferation or metabolic activity. Furthermore, following treatment with retinoic acid (RA) and further stimulation with brain-derived neurotrophic factor (BDNF) and nerve growth factor beta (NGF-β), the percentage of SH-SY5Y cells stained positive for dopaminergic neuronal differentiation markers (tyrosine hydroxylase [TH] and Dopamine Transporter [DAT]) was higher in hPL or FD-hPL than in FBS, and gene expression of dopaminergic markers TH, DAT, and DR2 was also detected. Overall, the data herein presented supports the use of hPL to differentiate SH-SY5Y cells into a neuronal phenotype with dopaminergic features, and the adoption of FD-hPL as a fully xenogeneic free alternative to FBS to support the use of SH-SY5Y cells as a neurodegeneration model.
Collapse
Affiliation(s)
- Miguel Ribeiro
- Life and Health Sciences Research Institute (ICVS), Universidade do Minho, Braga, Portugal
- ICVS/3B's--PT Government Associate Laboratory, Braga/, Guimarães, Braga, Portugal
| | - Jonas Campos
- Life and Health Sciences Research Institute (ICVS), Universidade do Minho, Braga, Portugal
- ICVS/3B's--PT Government Associate Laboratory, Braga/, Guimarães, Braga, Portugal
| | - Tiffany S Pinho
- Life and Health Sciences Research Institute (ICVS), Universidade do Minho, Braga, Portugal
- ICVS/3B's--PT Government Associate Laboratory, Braga/, Guimarães, Braga, Portugal
| | - Belém Sampaio-Marques
- Life and Health Sciences Research Institute (ICVS), Universidade do Minho, Braga, Portugal
- ICVS/3B's--PT Government Associate Laboratory, Braga/, Guimarães, Braga, Portugal
| | - Sandra Barata-Antunes
- Life and Health Sciences Research Institute (ICVS), Universidade do Minho, Braga, Portugal
- ICVS/3B's--PT Government Associate Laboratory, Braga/, Guimarães, Braga, Portugal
| | - Jorge Ribeiro Cibrão
- Life and Health Sciences Research Institute (ICVS), Universidade do Minho, Braga, Portugal
- ICVS/3B's--PT Government Associate Laboratory, Braga/, Guimarães, Braga, Portugal
| | - Ricardo Araújo
- Stemmatters, Biotecnologia e Medicina Regenerativa SA, Guimarães, Portugal
| | - Sara Duarte-Silva
- Life and Health Sciences Research Institute (ICVS), Universidade do Minho, Braga, Portugal
- ICVS/3B's--PT Government Associate Laboratory, Braga/, Guimarães, Braga, Portugal
| | - Elsa Moreira
- Stemmatters, Biotecnologia e Medicina Regenerativa SA, Guimarães, Portugal
| | - Rui A Sousa
- Stemmatters, Biotecnologia e Medicina Regenerativa SA, Guimarães, Portugal
| | - Pedro M Costa
- Stemmatters, Biotecnologia e Medicina Regenerativa SA, Guimarães, Portugal
| | - António J Salgado
- Life and Health Sciences Research Institute (ICVS), Universidade do Minho, Braga, Portugal
- ICVS/3B's--PT Government Associate Laboratory, Braga/, Guimarães, Braga, Portugal
| |
Collapse
|
2
|
Takatori A, Hossain MS, Ogura A, Akter J, Nakamura Y, Nakagawara A. NLRR1 Is a Potential Therapeutic Target in Neuroblastoma and MYCN-Driven Malignant Cancers. Front Oncol 2021; 11:669667. [PMID: 34277416 PMCID: PMC8279747 DOI: 10.3389/fonc.2021.669667] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Accepted: 06/07/2021] [Indexed: 12/28/2022] Open
Abstract
Receptor tyrosine kinases (RTKs) receive different modulation before transmitting proliferative signals. We previously identified neuronal leucine-rich repeat 1 (NLRR1) as a positive regulator of EGF and IGF-1 signals in high-risk neuroblastoma cells. Here, we show that NLRR1 is up-regulated in various adult cancers and acts as a key regulator of tumor cell proliferation. In the extracellular domains of NLRR1, fibronectin type III (FNIII) domain is responsible for its function to promote cell proliferation. We generated monoclonal antibodies against the extracellular domains of NLRR1 (N1mAb) and screened the positive N1mAbs for growth inhibitory effect. The treatment of N1mAbs reduces tumor cell proliferation in vitro and in vivo, and sensitizes the cells to EGFR inhibitor, suggesting that NLRR1 is a novel regulatory molecule of RTK function. Importantly, epitope mapping analysis has revealed that N1mAbs with growth inhibitory effect recognize immunoglobulin-like and FNIII domains of NLRR1, which also indicates the importance of FNIII domain in the function of NLRR1. Thus, the present study provides a new insight into the development of a cancer therapy by targeting NLRR1 as a modulator of proliferative signals on cellular membrane of tumor cells.
Collapse
Affiliation(s)
- Atsushi Takatori
- Division of Innovative Cancer Therapeutics, Chiba Cancer Center Research Institute, Chiba, Japan
| | - Md Shamim Hossain
- Division of Innovative Cancer Therapeutics, Chiba Cancer Center Research Institute, Chiba, Japan
| | - Atsushi Ogura
- Division of Innovative Cancer Therapeutics, Chiba Cancer Center Research Institute, Chiba, Japan
| | - Jesmin Akter
- Division of Innovative Cancer Therapeutics, Chiba Cancer Center Research Institute, Chiba, Japan
| | - Yohko Nakamura
- Division of Innovative Cancer Therapeutics, Chiba Cancer Center Research Institute, Chiba, Japan
| | - Akira Nakagawara
- Division of Innovative Cancer Therapeutics, Chiba Cancer Center Research Institute, Chiba, Japan
| |
Collapse
|
3
|
Subramonian D, Phanhthilath N, Rinehardt H, Flynn S, Huo Y, Zhang J, Messer K, Mo Q, Huang S, Lesperance J, Zage PE. Regorafenib is effective against neuroblastoma in vitro and in vivo and inhibits the RAS/MAPK, PI3K/Akt/mTOR and Fos/Jun pathways. Br J Cancer 2020; 123:568-579. [PMID: 32457362 PMCID: PMC7434894 DOI: 10.1038/s41416-020-0905-8] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2019] [Revised: 03/26/2020] [Accepted: 04/29/2020] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND Regorafenib is an inhibitor of multiple kinases with aberrant expression and activity in neuroblastoma tumours that have potential roles in neuroblastoma pathogenesis. METHODS We evaluated neuroblastoma cells treated with regorafenib for cell viability and confluence, and analysed treated cells for apoptosis and cell cycle progression. We evaluated the efficacy of regorafenib in vivo using an orthotopic xenograft model. We evaluated regorafenib-mediated inhibition of kinase targets and performed reverse-phase protein array (RPPA) analysis of neuroblastoma cells treated with regorafenib. Lastly, we evaluated the efficacy and effects of the combination of regorafenib and 13-cis-retinoic acid on intracellular signalling. RESULTS Regorafenib treatment resulted in reduced neuroblastoma cell viability and confluence, with both induction of apoptosis and of cell cycle arrest. Regorafenib treatment inhibits known receptor tyrosine kinase targets RET and PDGFRβ and intracellular signalling through the RAS/MAPK, PI3K/Akt/mTOR and Fos/Jun pathways. Regorafenib is effective against neuroblastoma tumours in vivo, and the combination of regorafenib and 13-cis-retinoic acid demonstrates enhanced efficacy compared with regorafenib alone. CONCLUSIONS The effects of regorafenib on multiple intracellular signalling pathways and the potential additional efficacy when combined with 13-cis-retinoic acid represent opportunities to develop treatment regimens incorporating regorafenib for children with neuroblastoma.
Collapse
Affiliation(s)
- Divya Subramonian
- Department of Pediatrics, Division of Hematology-Oncology, University of California San Diego, La Jolla, CA, USA
| | - Nikki Phanhthilath
- Department of Pediatrics, Division of Hematology-Oncology, University of California San Diego, La Jolla, CA, USA
| | - Hannah Rinehardt
- Department of Pediatrics, Division of Hematology-Oncology, University of California San Diego, La Jolla, CA, USA
| | - Sean Flynn
- Department of Pediatrics, Division of Hematology-Oncology, University of California San Diego, La Jolla, CA, USA
| | - Yuchen Huo
- Department of Pediatrics, Division of Hematology-Oncology, University of California San Diego, La Jolla, CA, USA
| | - Jing Zhang
- Department of Family Medicine and Public Health, University of California San Diego, La Jolla, CA, USA
| | - Karen Messer
- Department of Family Medicine and Public Health, University of California San Diego, La Jolla, CA, USA
| | - Qianxing Mo
- Department of Medicine, Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, TX, USA
- Department of Biostatistics & Bioinformatics, H. Lee Moffitt Cancer Center & Research Institute, 12902 Magnolia Drive, Tampa, FL, 33612, USA
| | - Shixia Huang
- Department of Medicine, Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, TX, USA
| | - Jacqueline Lesperance
- Department of Pediatrics, Division of Hematology-Oncology, University of California San Diego, La Jolla, CA, USA
| | - Peter E Zage
- Department of Pediatrics, Division of Hematology-Oncology, University of California San Diego, La Jolla, CA, USA.
- Peckham Center for Cancer and Blood Disorders, Rady Children's Hospital, San Diego, CA, USA.
| |
Collapse
|
4
|
Mechanisms of Efficacy of the FGFR1–3 Inhibitor AZD4547 in Pediatric Solid Tumor Models. Invest New Drugs 2020; 38:1677-1686. [DOI: 10.1007/s10637-020-00933-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2020] [Accepted: 04/07/2020] [Indexed: 02/07/2023]
|
5
|
Kefalakes E, Sarikidi A, Bursch F, Ettcheto M, Schmuck M, Rumpel R, Grothe C, Petri S. Isoform-selective as opposed to complete depletion of fibroblast growth factor 2 (FGF-2) has no major impact on survival and gene expression in SOD1 G93A amyotrophic lateral sclerosis mice. Eur J Neurosci 2019; 50:3028-3045. [PMID: 30883949 DOI: 10.1111/ejn.14405] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2018] [Revised: 01/29/2019] [Accepted: 03/05/2019] [Indexed: 12/27/2022]
Abstract
We have previously shown that total knockout of fibroblast growth factor-2 (FGF-2) results in prolonged survival and improved motor performance in superoxide dismutase 1 (SOD1G93A ) mutant mice, the most widely used animal model of the fatal adult onset motor neuron disease amyotrophic lateral sclerosis (ALS). Moreover, we found differential expression of growth factors in SOD1G93A mice, with distinct regulation patterns of FGF-2 in spinal cord and muscle tissue. Within the present study we aimed to characterize FGF-2-isoform specific effects on survival, motor performance as well as gene expression patterns predominantly in muscle tissue by generating double mutant SOD1G93A FGF-2 high molecular weight- and SOD1G93A FGF-2 low molecular weight-knockout mice. While isoform specific depletion was not beneficial regarding survival or motor performance of double mutant mice, we found isoform-dependent differential gene expression of epidermal growth factor (EGF) in the muscle of SOD1G93A FGF-2 low molecular weight knockout mice compared to single mutant SOD1G93A mice. This significant downregulation of EGF in the muscle tissue of double mutant SOD1G93A FGF-2 low molecular weight knockout mice implies that FGF-2 low molecular weight knockout (or the presence of the FGF-2 high molecular weight isoform) selectively impacts EGF gene expression in ALS muscle tissue.
Collapse
Affiliation(s)
- Ekaterini Kefalakes
- Department of Neurology, Hannover Medical School, Hannover, Germany.,Center for Systems Neuroscience (ZSN), Hannover, Germany
| | | | - Franziska Bursch
- Department of Neurology, Hannover Medical School, Hannover, Germany.,Center for Systems Neuroscience (ZSN), Hannover, Germany
| | - Miren Ettcheto
- Department of Pharmacology, Toxicology and Therapeutical Chemistry, Faculty of Pharmacy and Food Science, University of Barcelona, Barcelona, Spain.,Biomedical Research Networking Center in Neurodegenerative Diseases (CIBERNED), Madrid, Spain.,Institute of Neuroscience, University of Barcelona, Barcelona, Spain.,Department of Biochemistry, Faculty of Medicine and Life Science, University of Rovira i Virgili, Reus, Spain
| | - Martin Schmuck
- University of California, DAVIS School of Veterinary Medicine, Davis, CA
| | - Regina Rumpel
- Institute of Neuroanatomy and Cell Biology, Hannover Medical School, Hannover, Germany
| | - Claudia Grothe
- Center for Systems Neuroscience (ZSN), Hannover, Germany.,Institute of Neuroanatomy and Cell Biology, Hannover Medical School, Hannover, Germany
| | - Susanne Petri
- Department of Neurology, Hannover Medical School, Hannover, Germany.,Center for Systems Neuroscience (ZSN), Hannover, Germany
| |
Collapse
|
6
|
A Polymorphism in the FGFR4 Gene Is Associated With Risk of Neuroblastoma and Altered Receptor Degradation. J Pediatr Hematol Oncol 2016; 38:131-8. [PMID: 26840079 PMCID: PMC4758892 DOI: 10.1097/mph.0000000000000506] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
BACKGROUND Outcomes for children with high-risk neuroblastoma are poor, and improved understanding of the mechanisms underlying neuroblastoma pathogenesis, recurrence, and treatment resistance will lead to improved outcomes. Aberrant growth factor receptor expression and receptor tyrosine kinase signaling are associated with the pathogenesis of many malignancies. A germline polymorphism in the FGFR4 gene is associated with increased receptor expression and activity and with decreased survival, treatment resistance, and aggressive disease for many malignancies. We therefore investigated the role of this FGFR4 polymorphism in neuroblastoma pathogenesis. MATERIALS AND METHODS Germline DNA from neuroblastoma patients and matched controls was assessed for the FGFR4 Gly/Arg388 polymorphism by RT-PCR. Allele frequencies were assessed for association with neuroblastoma patient outcomes and prognostic features. Degradation rates of the FGFR4 Arg388 and Gly388 receptors and rates of receptor internalization into the late endosomal compartment were measured. RESULTS Frequency of the FGFR4 AA genotype and the prevalence of the A allele were significantly higher in patients with neuroblastoma than in matched controls. The Arg388 receptor demonstrated slower degradation than the Gly388 receptor in neuroblastoma cells and reduced internalization into multivesicular bodies. CONCLUSIONS The FGFR4 Arg388 polymorphism is associated with an increased prevalence of neuroblastoma in children, and this association may be linked to differences in FGFR4 degradation rates. Our study provides the first evidence of a role for FGFR4 in neuroblastoma, suggesting that FGFR4 genotype and the pathways regulating FGFR4 trafficking and degradation may be relevant for neuroblastoma pathogenesis.
Collapse
|
7
|
Haas AJ, Le Page Y, Zhadobov M, Sauleau R, Le Dréan Y. Effects of 60-GHz millimeter waves on neurite outgrowth in PC12 cells using high-content screening. Neurosci Lett 2016; 618:58-65. [PMID: 26921450 DOI: 10.1016/j.neulet.2016.02.038] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2016] [Accepted: 02/21/2016] [Indexed: 01/06/2023]
Abstract
Technologies for wireless telecommunication systems using millimeter waves (MMW) will be widely deployed in the near future. Forthcoming applications in this band, especially around 60GHz, are mainly developed for high data-rate local and body-centric telecommunications. At those frequencies, electromagnetic radiations have a very shallow penetration into biological tissues, making skin keratinocytes, and free nerve endings of the upper dermis the main targets of MMW. Only a few studies assessed the impact of MMW on neuronal cells, and none of them investigated a possible effect on neuronal differentiation. We used a neuron-like cell line (PC12), which undergoes neuronal differentiation when treated with the neuronal growth factor (NGF). PC12 cells were exposed at 60.4GHz for 24h, at an incident power density averaged over the cell monolayer of 10mW/cm(2). Using a large scale cell-by-cell analysis based on high-content screening microscopy approach, we assessed potential effects of MMW on PC12 neurite outgrowth and cytoskeleton protein expression. No differences were found in protein expression of the neuronal marker β3-tubulin nor in internal expression control β-tubulin. On the other hand, our data showed a slight increase, although insignificant, in neurite outgrowth, induced by MMW exposure. However, experimental controls demonstrated that this increase was related to heating.
Collapse
Affiliation(s)
- Alexis J Haas
- Transcription, Environment and Cancer Group, Institute of Research in Environmental and Occupational Health (IRSET), INSERM, University of Rennes 1, Rennes, France
| | - Yann Le Page
- Transcription, Environment and Cancer Group, Institute of Research in Environmental and Occupational Health (IRSET), INSERM, University of Rennes 1, Rennes, France
| | - Maxim Zhadobov
- Institute of Electronics and Telecommunications of Rennes (IETR), University of Rennes 1, UMR CNRS, Rennes, France
| | - Ronan Sauleau
- Institute of Electronics and Telecommunications of Rennes (IETR), University of Rennes 1, UMR CNRS, Rennes, France
| | - Yves Le Dréan
- Transcription, Environment and Cancer Group, Institute of Research in Environmental and Occupational Health (IRSET), INSERM, University of Rennes 1, Rennes, France.
| |
Collapse
|
8
|
Sensitivity of neuroblastoma to the novel kinase inhibitor cabozantinib is mediated by ERK inhibition. Cancer Chemother Pharmacol 2015; 76:977-87. [DOI: 10.1007/s00280-015-2871-z] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2015] [Accepted: 09/08/2015] [Indexed: 10/23/2022]
|
9
|
Maioli M, Rinaldi S, Migheli R, Pigliaru G, Rocchitta G, Santaniello S, Basoli V, Castagna A, Fontani V, Ventura C, Serra PA. Neurological morphofunctional differentiation induced by REAC technology in PC12. A neuro protective model for Parkinson's disease. Sci Rep 2015; 5:10439. [PMID: 25976344 PMCID: PMC4432565 DOI: 10.1038/srep10439] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2014] [Accepted: 04/13/2015] [Indexed: 12/12/2022] Open
Abstract
Research for the use of physical means, in order to induce cell differentiation for new therapeutic strategies, is one of the most interesting challenges in the field of regenerative medicine, and then in the treatment of neurodegenerative diseases, Parkinson's disease (PD) included. The aim of this work is to verify the effect of the radio electric asymmetric conveyer (REAC) technology on the PC12 rat adrenal pheochromocytoma cell line, as they display metabolic features of PD. PC12 cells were cultured with a REAC regenerative tissue optimization treatment (TO-RGN) for a period ranging between 24 and 192 hours. Gene expression analysis of specific neurogenic genes, as neurogenin-1, beta3-tubulin and Nerve growth factor, together with the immunostaining analysis of the specific neuronal protein beta3-tubulin and tyrosine hydroxylase, shows that the number of cells committed toward the neurogenic phenotype was significantly higher in REAC treated cultures, as compared to control untreated cells. Moreover, MTT and Trypan blue proliferation assays highlighted that cell proliferation was significantly reduced in REAC TO-RGN treated cells. These results open new perspectives in neurodegenerative diseases treatment, particularly in PD. Further studies will be needed to better address the therapeutic potential of the REAC technology.
Collapse
Affiliation(s)
- Margherita Maioli
- Department of Biomedical Sciences, University of Sassari, 07100, Sassari, Italy
- Laboratory of Molecular Biology and Stem Cell Engineering - National Institute of Biostructures and Biosystems, 40121, Bologna, Italy
- Department of Regenerative Medicine, Rinaldi Fontani Institute, 50144, Florence, Italy
| | - Salvatore Rinaldi
- Department of Regenerative Medicine, Rinaldi Fontani Institute, 50144, Florence, Italy
- Research Department, Rinaldi Fontani Foundation - NPO, 50144, Florence, Italy
| | - Rossana Migheli
- Department of Clinical and Experimental Medicine, University of Sassari, 07100, Sassari, Italy
| | - Gianfranco Pigliaru
- Department of Biomedical Sciences, University of Sassari, 07100, Sassari, Italy
- Laboratory of Molecular Biology and Stem Cell Engineering - National Institute of Biostructures and Biosystems, 40121, Bologna, Italy
| | - Gaia Rocchitta
- Department of Clinical and Experimental Medicine, University of Sassari, 07100, Sassari, Italy
| | - Sara Santaniello
- Department of Biomedical Sciences, University of Sassari, 07100, Sassari, Italy
- Laboratory of Molecular Biology and Stem Cell Engineering - National Institute of Biostructures and Biosystems, 40121, Bologna, Italy
| | - Valentina Basoli
- Department of Biomedical Sciences, University of Sassari, 07100, Sassari, Italy
| | - Alessandro Castagna
- Department of Regenerative Medicine, Rinaldi Fontani Institute, 50144, Florence, Italy
- Research Department, Rinaldi Fontani Foundation - NPO, 50144, Florence, Italy
| | - Vania Fontani
- Department of Regenerative Medicine, Rinaldi Fontani Institute, 50144, Florence, Italy
- Research Department, Rinaldi Fontani Foundation - NPO, 50144, Florence, Italy
| | - Carlo Ventura
- Laboratory of Molecular Biology and Stem Cell Engineering - National Institute of Biostructures and Biosystems, 40121, Bologna, Italy
- Stem Wave Institute for Tissue Healing (SWITH), Gruppo Villa Maria and Ettore Sansavini Health Science Foundation NPO, 48022, Lugo, Italy
| | - Pier Andrea Serra
- Department of Clinical and Experimental Medicine, University of Sassari, 07100, Sassari, Italy
| |
Collapse
|
10
|
The novel kinase inhibitor EMD1214063 is effective against neuroblastoma. Invest New Drugs 2014; 32:815-24. [PMID: 24832869 DOI: 10.1007/s10637-014-0107-4] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2014] [Accepted: 04/23/2014] [Indexed: 12/16/2022]
Abstract
BACKGROUND Children with high-risk neuroblastoma have poor survival rates, and novel therapies are needed. Previous studies have identified a role for the HGF/c-Met pathway in neuroblastoma pathogenesis. We hypothesized that EMD1214063 would be effective against neuroblastoma tumor cells and tumors in preclinical models via inhibition of HGF/c-Met signaling. Methods We determined the expression of c-Met protein by Western blots in a panel of neuroblastoma tumor cell lines and neuroblastoma cell viability after treatment with EMD1214063 using MTT assays. TUNEL assays and assays for DNA ladder formation, were performed to measure the induction of apoptosis after EMD1214063 treatment. Inhibition of intracellular signaling was measured by Western blot analysis of treated and untreated cells. To investigate the efficacy of EMD1214063 against neuroblastoma tumors in vivo, neuroblastoma cells were injected orthotopically into immunocompromised mice, and mice were treated with oral EMD1214063. Tumors were evaluated for growth, histologic appearance, and induction of apoptosis by immunohistochemistry. Results All neuroblastoma cell lines were sensitive to EMD1214063, and IC50 values ranged from 2.4 to 8.5 μM. EMD1214063 treatment inhibited HGF-mediated c-Met phosphorylation and MEK phosphorylation in neuroblastoma cells. EMD1214063 induced apoptosis in all tested cell lines. In mice with neuroblastoma xenograft tumors, EMD1214063 treatment reduced tumor growth. Conclusions Treatment of neuroblastoma tumor cells with EMD1214063 inhibits HGF-induced c-Met phosphorylation and results in cell death. EMD1214063 treatment is also effective in reducing tumor growth in vivo. EMD1214063 therefore represents a novel therapeutic agent for neuroblastoma, and further preclinical studies of EMD1214063 are warranted.
Collapse
|
11
|
Heo Y, Lee HJ, Kim EH, Kim MK, Ito Y, Son TI. Regeneration effect of visible light-curing furfuryl alginate compound by release of epidermal growth factor for wound healing application. J Appl Polym Sci 2013. [DOI: 10.1002/app.40113] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Affiliation(s)
- Yun Heo
- Department of Systems Biotechnology; Chung-Ang University; Anseong Gyeonggi-Do 456-756 Republic of Korea
| | - Hyung-Jae Lee
- Department of Systems Biotechnology; Chung-Ang University; Anseong Gyeonggi-Do 456-756 Republic of Korea
| | - Eun-Hye Kim
- Department of Systems Biotechnology; Chung-Ang University; Anseong Gyeonggi-Do 456-756 Republic of Korea
| | - Mi-Kyung Kim
- Department of Pathology; College of Medicine, Chung-Ang University; Dongjak-Gu Seoul 156-756 Republic of Korea
| | - Yoshihiro Ito
- Nano Medical Engineering Laboratory; RIKEN; Wako Saitama 351-0198 Japan
| | - Tae-Il Son
- Department of Systems Biotechnology; Chung-Ang University; Anseong Gyeonggi-Do 456-756 Republic of Korea
| |
Collapse
|
12
|
Koves IH, Russo VC, Higgins S, Mishra A, Pitt J, Cameron FJ, Werther GA. An in vitro paradigm for diabetic cerebral oedema and its therapy: a critical role for taurine and water channels. Neurochem Res 2011; 37:182-92. [PMID: 21935730 DOI: 10.1007/s11064-011-0598-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2011] [Revised: 07/15/2011] [Accepted: 09/08/2011] [Indexed: 11/29/2022]
Abstract
The pathophysiology of cerebral oedema (CE) in diabetic ketoacidosis (DKA) remains enigmatic. We investigated the role of the idiogenic osmol taurine and aquaporin channels in an in vitro model, the SH-SY5Y neuroblastoma cell line, by sequentially mimicking DKA-like hyperglycemia/hypertonicity and hypotonic fluid therapy. Exposure to DKA-like hyperosmolarity led to shrinkage, while hypotonic fluid exposure led to cell swelling and impaired viability. Low sodium compensated in part for elevated glucose, pointing to a critical role for overall osmolality. Taurine, was synthesized and retained intracellularly during DKA-like hypertonicity, and released during hypotonicity, in part mitigating neuronal swelling. Metabolic labeling showed that the rate of taurine release was inadequate to fully prevent neuronal swelling during hypotonic fluid therapy following DKA-like hypertonicity. Under these conditions, Aquaporin4 & 9 channels were respectively down and up-regulated. Our study provides further novel insights into molecular mechanisms contributing to CE in DKA and its therapy.
Collapse
Affiliation(s)
- Ildi H Koves
- Centre for Hormone Research, Murdoch Childrens Research Institute, Royal Children's Hospital, Flemington Rd., Parkville, VIC 3052, Australia
| | | | | | | | | | | | | |
Collapse
|
13
|
Wheldon LM, Haines BP, Rajappa R, Mason I, Rigby PW, Heath JK. Critical role of FLRT1 phosphorylation in the interdependent regulation of FLRT1 function and FGF receptor signalling. PLoS One 2010; 5:e10264. [PMID: 20421966 PMCID: PMC2858647 DOI: 10.1371/journal.pone.0010264] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2009] [Accepted: 03/25/2010] [Indexed: 01/03/2023] Open
Abstract
BACKGROUND Fibronectin leucine rich transmembrane (FLRT) proteins have dual properties as regulators of cell adhesion and potentiators of fibroblast growth factor (FGF) mediated signalling. The mechanism by which the latter is achieved is still unknown and is the subject of this investigation. PRINCIPAL FINDINGS Here we show that FLRT1 is a target for tyrosine phosphorylation mediated by FGFR1 and implicate a non-receptor Src family kinase (SFK). We identify the target tyrosine residues in the cytoplasmic domain of FLRT1 and show that these are not direct substrates for Src kinase suggesting that the SFK may exert effects via potentiation of FGFR1 kinase activity. We show that whilst FLRT1 expression results in a ligand-dependent elevation of MAP kinase activity, a mutant version of FLRT1, defective as an FGFR1 kinase substrate (Y3F-FLRT1), has the property of eliciting ligand-independent chronic activation of the MAP kinase pathway which is suppressed by pharmacological inhibition of either FGFR1 or Src kinase. Functional investigation of FGFR1 and FLRT1 signalling in SH-SY5Y neuroblastoma cells reveals that FLRT1 alone acts to induce a multi-polar phenotype whereas the combination of FLRT1 and FGFR activation, or expression of Y3F-FLRT1, acts to induce neurite outgrowth via MAPK activation. Similar results were obtained in a dendrite outgrowth assay in primary hippocampal neurons. We also show that FGFR1, FLRT1 and activated Src are co-localized and this complex is trafficked toward the soma of the cell. The presence of Y3F-FLRT1 rather than FLRT1 resulted in prolonged localization of this complex within the neuritic arbour. CONCLUSIONS This study shows that the phosphorylation state of FLRT1, which is itself FGFR1 dependent, may play a critical role in the potentiation of FGFR1 signalling and may also depend on a SFK-dependent phosphorylation mechanism acting via the FGFR. This is consistent with an 'in vivo' role for FLRT1 regulation of FGF signalling via SFKs. Furthermore, the phosphorylation-dependent futile cycle mechanism controlling FGFR1 signalling is concurrently crucial for regulation of FLRT1-mediated neurite outgrowth.
Collapse
Affiliation(s)
- Lee M. Wheldon
- Molecular Bacteriology and Immunology Group (MBIG), Centre for Biomolecular Sciences, University of Nottingham, Nottingham, United Kingdom
| | - Bryan P. Haines
- Section of Gene Function and Regulation, The Institute of Cancer Research, Chester Beatty Laboratories, London, United Kingdom
| | - Rajit Rajappa
- MRC Centre for Developmental Neurobiology, King's College London, London, United Kingdom
| | - Ivor Mason
- MRC Centre for Developmental Neurobiology, King's College London, London, United Kingdom
| | - Peter W. Rigby
- Section of Gene Function and Regulation, The Institute of Cancer Research, Chester Beatty Laboratories, London, United Kingdom
| | - John K. Heath
- Cancer Research UK Growth Factor Group, School of Biosciences, University of Birmingham, Birmingham, United Kingdom
| |
Collapse
|
14
|
Botting RA, McLachlan CS. Does cellular heterogeneity influence neuroblastoma cell line proliferation and invasiveness with granulocyte colony-stimulating factor? J Pediatr Surg 2009; 44:2436-7; author reply 2437. [PMID: 20006048 DOI: 10.1016/j.jpedsurg.2009.08.029] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/26/2009] [Accepted: 08/27/2009] [Indexed: 11/28/2022]
|
15
|
Thress K, Macintyre T, Wang H, Whitston D, Liu ZY, Hoffmann E, Wang T, Brown JL, Webster K, Omer C, Zage PE, Zeng L, Zweidler-McKay PA. Identification and preclinical characterization of AZ-23, a novel, selective, and orally bioavailable inhibitor of the Trk kinase pathway. Mol Cancer Ther 2009; 8:1818-27. [PMID: 19509272 DOI: 10.1158/1535-7163.mct-09-0036] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Tropomyosin-related kinases (TrkA, TrkB, and TrkC) are receptor tyrosine kinases that, along with their ligands, the neurotrophins, are involved in neuronal cell growth, development, and survival. The Trk-neurotrophin pathway may also play a role in tumorigenesis through oncogenic fusions, mutations, and autocrine signaling, prompting the development of novel Trk inhibitors as agents for cancer therapy. This report describes the identification of AZ-23, a novel, potent, and selective Trk kinase inhibitor. In vitro studies with AZ-23 showed improved selectivity over previous compounds and inhibition of Trk kinase activity in cells at low nanomolar concentrations. AZ-23 showed in vivo TrkA kinase inhibition and efficacy in mice following oral administration in a TrkA-driven allograft model and significant tumor growth inhibition in a Trk-expressing xenograft model of neuroblastoma. AZ-23 represents a potent and selective Trk kinase inhibitor from a novel series with the potential for use as a treatment for cancer.
Collapse
Affiliation(s)
- Kenneth Thress
- Cancer and Infection Research, AstraZeneca, Mereside, Alderley Park, Macclesfield, Cheshire, UK.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
16
|
|
17
|
Michaelis M, Bliss J, Arnold SC, Hinsch N, Rothweiler F, Deubzer HE, Witt O, Langer K, Doerr HW, Wels WS, Cinatl J. Cisplatin-resistant neuroblastoma cells express enhanced levels of epidermal growth factor receptor (EGFR) and are sensitive to treatment with EGFR-specific toxins. Clin Cancer Res 2008; 14:6531-7. [PMID: 18927293 DOI: 10.1158/1078-0432.ccr-08-0821] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE Neuroblastomas frequently show expression of the epidermal growth factor receptor (EGFR) and may therefore be susceptible to EGFR-targeted therapies. Here, EGFR expression and functionality was investigated in parental chemosensitive neuroblastoma cell lines (UKF-NB-3, IMR-32, NLF, SH-SY5Y) and their cisplatin-resistant sublines (UKF-NB-3(r)CDDP(1000), IMR-32(r)CDDP(1000), NLF(r)CDDP(1000), and SH-SY5Y(r)CDDP(500)). Moreover, the EGFR antibody cetuximab, the EGFR tyrosine kinase inhibitor Tyrphostin B46, and recombinant EGFR-targeted toxins were investigated for their influence on the viability and growth of neuroblastoma cells. EXPERIMENTAL DESIGN EGFR expression and function was measured by flow cytometry or Western blot. Cell viability was detected by 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide assay. Apoptosis was examined by immunostaining for active caspase-3 or cleaved poly(ADP-ribose) polymerase. Cellular binding of FITC-labeled immunotoxins was studied by flow cytometry, and cellular uptake was studied by confocal laser scanning microscopy. RESULTS The EGFR-targeted antibody and growth factor toxins scFv(14E1)- Pseudomonas exotoxin A (ETA) and TGF-alpha-ETA exerted anti-cancer effects in neuroblastoma cell lines that were insensitive to cetuximab or EGFR tyrosine kinase inhibitors. Furthermore, adaptation of chemosensitive neuroblastoma cells to cisplatin increased EGFR expression and sensitivity to both recombinant toxins. Treatment of chemosensitive neuroblastoma cells with cisplatin reversibly increased EGFR expression, whereas cisplatin-resistant cells showed enhanced EGFR expression independent of the presence of cisplatin. Combination treatment with scFv(14E1)-ETA or TGF-alpha-ETA and cisplatin exerted significantly improved anticancer effects compared with either single treatment in parental neuroblastoma cells, cisplatin-resistant sublines, and primary cultures. CONCLUSIONS EGFR-targeted cytotoxic reagents such as scFv(14E1)-ETA and TGF-alpha-ETA represent promising candidates for further development as antineuroblastoma agents, especially in combination with cisplatin.
Collapse
Affiliation(s)
- Martin Michaelis
- Institut für Medizinische Virologie, Klinikum der J.W. Goethe Universität, Germany
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
18
|
Kobayashi K, Xin Y, Ymer SI, Werther GA, Russo VC. Subtractive hybridisation screen identifies genes regulated by glucose deprivation in human neuroblastoma cells. Brain Res 2007; 1170:129-39. [PMID: 17719568 DOI: 10.1016/j.brainres.2007.07.042] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2007] [Revised: 07/24/2007] [Accepted: 07/25/2007] [Indexed: 02/06/2023]
Abstract
Glucose is the major source of energy for the brain and inadequate glucose supply causes damage of neuronal cells. In this study we employed the human neuroblastoma cell line SH-SY5Y, as an in vitro model for neuronal cells, to identify genes regulated by glucose deprivation. Using subtractive hybridisation screen, validated by Northern analysis, we identify for the first time specific targets of the glucopenic response. These genes are involved in key cellular process including gene transcription, protein synthesis, mitochondrial metabolism, neuronal development, neuroprotection and neuronal apoptosis. Our findings suggest that the fate of neuronal cells undergoing glucose starvation relies on complex gene interactions. Modulation of the expression of these genes in vivo will enable determination of the precise role of each gene and possibly identify key elements and potential therapeutic targets of the glucopenic response.
Collapse
Affiliation(s)
- Kisho Kobayashi
- Centre for Hormone Research, Murdoch Children's Research Institute, Department of Paediatrics, University of Melbourne, Royal Children's Hospital, 3052 Parkville, Australia
| | | | | | | | | |
Collapse
|
19
|
Chiu B, Mirkin B, Madonna MB. Mitogenic and apoptotic actions of epidermal growth factor on neuroblastoma cells are concentration-dependent. J Surg Res 2006; 135:209-12. [PMID: 16872636 DOI: 10.1016/j.jss.2006.04.018] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2006] [Revised: 04/20/2006] [Accepted: 04/21/2006] [Indexed: 11/17/2022]
Abstract
BACKGROUND In previous studies we have shown that epidermal growth factor (EGF) at concentrations between 50 and 100 ng/mL induced apoptosis in wild-type SK-N-SH neuroblastoma cells. We hypothesize that this apoptotic event separates EGF-induced neuroblastoma cell growth into a biphasic concentration-dependent process, due to activation of different signaling cascades. METHODS Cells were incubated in concentrations of EGF ranging from 5 to 250 ng/mL for 3 days, and cell proliferation was determined by the MTT assay. Cells incubated with EGF 5, 100, or 250 ng/mL for 17 h were also assayed for apoptosis by DNA laddering. Western immunoblots were performed on whole cell lysates prepared from cells incubated with EGF (5-250 ng/mL) for 17 h. Antibodies against cleaved caspase3, p-AKT, p-GSK-3beta, p-BAD, p-RAF, p-ERK, and p-P38 were used as probes. RESULTS A triphasic, concentration-dependent response was observed following incubation of cells with EGF. Cell proliferation was increased by EGF 5 ng/mL (P < 0.05), decreased by EGF 100 ng/mL, and increased when incubated with EGF 250 ng/mL (P < 0.05). DNA laddering only occurred after treatment with EGF 100 ng/mL. The expressions of p-ERK, p-RAF, p-BAD, and p-GSK-3beta were increased at EGF concentrations of 5-10 ng/mL. At 50-100 ng/mL EGF, the expression of cleaved caspase3 was increased. Maximal p-P38 expression was at 50 ng/mL EGF. At EGF concentrations of 150-250 ng/mL, the expressions of p-AKT and p-GSK-3beta were elevated. CONCLUSIONS Neuroblastoma cell growth induced by EGF exhibited a triphasic pattern; cell growth was increased at EGF concentrations 5-20 and 150-250 ng/mL, but decreased at 50-100 mg/mL. Apoptosis was induced at 50-100 ng/mL EGF. Each growth phase activated different signaling molecules.
Collapse
Affiliation(s)
- Bill Chiu
- Department of Surgery, Children's Memorial Hospital, Northwestern University Feinberg School of Medicine, Chicago, Illinois 60611, USA
| | | | | |
Collapse
|
20
|
Ho R, Minturn JE, Hishiki T, Zhao H, Wang Q, Cnaan A, Maris J, Evans AE, Brodeur GM. Proliferation of human neuroblastomas mediated by the epidermal growth factor receptor. Cancer Res 2005; 65:9868-75. [PMID: 16267010 DOI: 10.1158/0008-5472.can-04-2426] [Citation(s) in RCA: 108] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Neuroblastoma is a common solid tumor of childhood that is derived from the neural crest. Expression of epidermal growth factor (EGF) receptors (EGFRs) has been associated with enhanced cell growth and aggressive behavior in other tumors. Here, we examined the expression profile of EGFRs in neuroblastoma cell lines and primary tumors. We found that all 13 neuroblastoma cell lines examined expressed EGFR1 (HER1), most at readily detectable levels. Low levels of other human EGFR family receptors were also detected in almost all cell lines. All primary tumors examined expressed readily detectable levels of HER1 and HER3 and lower levels of HER2 and HER4. EGF had a significant effect on the proliferation of neuroblastoma cell lines in vitro. EGF treatment (100 ng/mL) of the cell lines SY5Y and NLF significantly increased cell number (P < 0.01). EGF stimulated more cells to enter S and G2-M phase, as suggested by flow cytometry, indicating that EGF increases cell number by increasing proliferation, with no appreciable change in apoptosis. EGF exposure resulted in receptor autophosphorylation and activation of both the mitogen-activated protein kinase (MAPK) and phosphoinositide 3-kinase (PI3K)/AKT pathways. Exposure to 0.5 micromol/L ZD1839, a HER1-specific inhibitor, caused a 40% to 50% reduction in the number of SY5Y and NLF cells grown in medium containing 10% fetal bovine serum (P < 0.01). Even at 0.01 micromol/L, ZD1839 inhibited autophosphorylation of HER1 by EGF. At 0.1 micromol/L, it also blocked phosphorylation of AKT, but not MAPK, in NLF cells. Additional studies showed that the PI3K/AKT-specific inhibitor LY294002 had a more profound effect than the MAPK-specific inhibitor U0126 in blocking EGF-induced cell proliferation. This suggests that the PI3K/AKT pathway is the main signaling pathway responsible for the proliferation effects of EGF in neuroblastomas. Our results also indicate that ZD1839 is a potent inhibitor of neuroblastoma cell proliferation; therefore, it may be a useful, biologically based therapeutic agent for these tumors.
Collapse
Affiliation(s)
- Ruth Ho
- Division of Oncology, Children's Hospital of Philadelphia and University of Pennsylvania, Philadelphia, Pennsylvania 19104-4318, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
21
|
Russo VC, Schütt BS, Andaloro E, Ymer SI, Hoeflich A, Ranke MB, Bach LA, Werther GA. Insulin-like growth factor binding protein-2 binding to extracellular matrix plays a critical role in neuroblastoma cell proliferation, migration, and invasion. Endocrinology 2005; 146:4445-55. [PMID: 15994346 DOI: 10.1210/en.2005-0467] [Citation(s) in RCA: 87] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
IGF binding proteins (IGFBPs) modulate IGF cellular bioavailability and may directly regulate tumor growth and invasion. We have previously shown that IGFBP-2 binds and localizes IGF-I to the pericellular matrix and have provided some evidence suggesting that the heparin binding domain (HBD) or the arginine-glycine-aspartic acid (RGD) integrin binding motif may be involved in these interactions. However, the precise mechanisms involved remain to be elucidated. We therefore mutated the HBD or RGD sequence of IGFBP-2 and investigated consequent effects on extracellular matrix (ECM) binding, IGF-induced proliferation, and migration of neuroblastoma cells. IGFBP-2 and its arginine-glycine-glutamic acid (RGE) mutant similarly bound ECM components, whereas binding of mutant HBD-IGFBP-2 to each of the ECM substrates was markedly reduced by 70-80% (P < 0.05). IGF-I (100 ng/ml) increased incorporation of 3H-thymidine in neuroblastoma SK-N-SHEP cells by approximately 30%, an effect blunted by exogenously added native or either mutant IGFBP-2. Overexpression of IGFBP-2 and its RGE mutant potently promoted SHEP cell proliferation (5-fold), whereas SHEP cell proliferation was negligible when HBD-IGFBP-2 was overexpressed. Addition or overexpression of IGFBP-2 and its RGE mutant potently (P < 0.05) enhanced SHEP cell migration/invasion through the ECM. However, overexpression of the HBD-IGFBP-2 mutant potently inhibited (50-60%) SHEP cell invasion through ECM. Thus, IGFBP-2, which binds to the ECM, enhances proliferation and metastatic behavior of neuroblastoma cells, functions that directly or indirectly use the HBD but not the integrin binding sequence. Our novel findings thus point to a key role for the HBD of IGFBP-2 in the control and regulation of neuroblastoma growth and invasion.
Collapse
Affiliation(s)
- V C Russo
- Murdoch Childrens Research Institute, Centre for Hormone Research and Department of Paediatrics, University of Melbourne, Parkville 3052, Victoria, Australia.
| | | | | | | | | | | | | | | |
Collapse
|
22
|
Motegi A, Fujimoto J, Kotani M, Sakuraba H, Yamamoto T. ALK receptor tyrosine kinase promotes cell growth and neurite outgrowth. J Cell Sci 2005; 117:3319-29. [PMID: 15226403 DOI: 10.1242/jcs.01183] [Citation(s) in RCA: 121] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
Anaplastic lymphoma kinase (ALK) is a receptor-type protein tyrosine kinase that is expressed preferentially in neurons of the central and peripheral nervous systems at late embryonic stages. To elucidate the role of ALK in neurons, we developed an agonist monoclonal antibody (mAb) against the extracellular domain of ALK. Here we show that mAb16-39 elicits tyrosine phosphorylation of endogenously expressed ALK in human neuroblastoma (SK-N-SH) cells. Stimulation of these cells with mAb16-39 markedly induces the tyrosine phosphorylation of insulin receptor substrate-1 (IRS-1), Shc, and c-Cbl and also their interaction with ALK and activation of ERK1/2. Furthermore, we show that continuous incubation with mAb16-39 induces the cell growth and neurite outgrowth of SK-N-SH cells. These responses are completely blocked by MEK inhibitor PD98059 but not by the phosphatidylinositol 3-kinase (PI 3-kinase) inhibitor wortmannin, indicating an essential role of the mitogen-activated protein kinase (MAP kinase) signaling cascade in ALK-mediated growth and differentiation of neurons.
Collapse
Affiliation(s)
- Akira Motegi
- Division of Oncology, Institute of Medical Science, University of Tokyo, 4-6-1 Shirokanedai, Minato-ku, 108-8639, Japan
| | | | | | | | | |
Collapse
|
23
|
Hettmer S, Ladisch S, Kaucic K. Low complex ganglioside expression characterizes human neuroblastoma cell lines. Cancer Lett 2005; 225:141-9. [PMID: 15922866 PMCID: PMC2866625 DOI: 10.1016/j.canlet.2004.11.036] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2004] [Accepted: 11/17/2004] [Indexed: 11/23/2022]
Abstract
Low (< or = 35%) or absent expression of the complex 'b' pathway gangliosides GD1b, GT1b and GQ1b (CbG) correlates with an aggressive biological phenotype in human neuroblastoma tumors. To develop an in vitro model to probe mechanisms by which CbG may contribute to neuroblastoma behavior, we have comprehensively evaluated ganglioside expression in nine well-established human neuroblastoma cell lines, all derived from poor prognosis tumors. Total cellular ganglioside content ranged from 8 to 69 nmol/10(8) cells. High performance thin layer chromatography revealed that the simple disialoganglioside GD2 was prominent in eight of the cell lines (up to 60% of total gangliosides), whereas CbG were low (1-21%) in all nine cell lines. The structurally most complex 'b' pathway species, GQ1b, was not detected in any of the cell lines. The prominence of GD2 in neuroblastoma cell lines mirrors the high expression of GD2 that characterizes human neuroblastoma tumors, and the low CbG expression in the cell lines is analogous to that found in clinically and biologically unfavorable neuroblastoma tumors, thus establishing these neuroblastoma cell lines as valuable model systems for study of the role of CbG in the pathobiology of human neuroblastoma.
Collapse
Affiliation(s)
| | | | - Karen Kaucic
- Corresponding author. Tel.: +1 202 884 3217; fax: +1 202 884 3929. (K. Kaucic)
| |
Collapse
|
24
|
Russo VC, Metaxas S, Kobayashi K, Harris M, Werther GA. Antiapoptotic effects of leptin in human neuroblastoma cells. Endocrinology 2004; 145:4103-12. [PMID: 15166121 DOI: 10.1210/en.2003-1767] [Citation(s) in RCA: 108] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Many factors regulate nervous system development, including complex cross-talk between local neuroendocrine systems. The adipocyte-secreted hormone leptin, mainly known for its key roles in nutrition and reproductive balance, may also be involved in neuroanatomical organization, myelination processes, and neuronal/glia maturation. SK-N-SH-SY5Y neuroblastoma cells were employed as an in vitro model of human neuronal cells to determine whether leptin exerts neuroprotective activities. We show that SH-SY5Y cells express leptin, the long and short isoforms of the leptin receptor (ObRl, ObRs). In SH-SY5Y cells, leptin induced signal transducer and activator of transcription (STAT)-3 phosphorylation and suppressor of cytokine signaling-3 mRNA expression. Leptin dose-dependently increased cell number (up to 200% at 1 microm by 48 h, P < 0.01), and at 24-48 h, leptin at 100 nm increased SH-SY5Y cell number by 30-50%, respectively. SH-SY5Y cell viability was reduced in serum-free conditions at 24 h, and addition of leptin at 100 nm significantly reduced apoptosis by approximately 20% (P < 0.001). Leptin's antiapoptotic activity required Janus kinase/STAT, MAPK, and phosphatidylinositol-3-kinase activation because the antiapoptotic effects of leptin were abolished, and caspase-3 immunoreactivity increased in the presence of the specific blockers AG490, U0126, or LY294002. Gene array demonstrated that leptin inhibits apoptosis via potent down-regulation of caspase-10 and TNF-related apoptosis-inducing ligand. Our data thus demonstrate, for the first time, that leptin stimulates, in a time- and dose-dependent manner, neuroblastoma cell proliferation and that the underlying mechanisms involve suppression of apoptosis via the Janus kinase-STAT, phosphatidylinositol-3 kinase, and MAPK pathways that culminate altogether in the down-regulation of the apoptotic factors caspase-10 and TNF-related apoptosis-inducing ligand.
Collapse
Affiliation(s)
- V C Russo
- Centre for Hormone Research, Murdoch Childrens Research Institute, Parkville 3052, Victoria, Australia.
| | | | | | | | | |
Collapse
|
25
|
Russo VC, Andaloro E, Fornaro SA, Najdovska S, Newgreen DF, Bach LA, Werther GA. Fibroblast growth factor-2 over-rides insulin-like growth factor-I induced proliferation and cell survival in human neuroblastoma cells. J Cell Physiol 2004; 199:371-80. [PMID: 15095284 DOI: 10.1002/jcp.10416] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
The insulin-like growth factor (IGF) system is a key regulator of cell growth, survival and differentiation, and these functions are co-modulated by other growth factors including fibroblast growth factor-2 (FGF-2). To investigate IGF/FGF interactions in neuronal cells, we employed neuroblastoma cells (SK-N-MC). In serum free conditions proliferation of the SK-N-MC cells was promoted by IGF-I (25 ng/ml), but blunted by FGF-2 (50 ng/ml). IGF-I-induced proliferation was abolished in the presence of FGF-2 even when IGF-I was used at 100 ng/ml. In addition to our previously described FGF-2 induced proteolytic cleavage of IGFBP-2, we found that FGF-2 increased IGFBP-6 levels in conditioned medium (CM) without affecting IGFBP-6 mRNA abundance. Modulation of IGFBP-2 and -6 levels were not significant mechanisms involved in the blockade of IGF-I action since the potent IGF-I analogues [QAYL]IGF-I and des(1-3)IGF-I (minimal IGFBP affinity) were unable to overcome FGF-2 inhibition of cell proliferation. FGF-2 treated cells showed morphological differentiation expressing the TUJ1 neuronal marker while cells treated with IGF-I alone showed no morphological change. When IGF-I was combined with FGF-2, however, cell morphology was indistinguishable from that seen with FGF-2 alone. FGF-2 inhibited proliferation and enhanced differentiation was also associated with a 70% increase in cell death. Although IGF-I alone was potently anti-apoptotic (60% decreased), IGF-I was unable to prevent apoptosis when administrated in combination with FGF-2. Gene-array analysis confirmed FGF-2 activation of the intrinsic and extrinsic apoptotic pathways and blockade of IGF anti-apoptotic signaling. FGF-2, directly and indirectly, overcomes the proliferative and anti-apoptotic activity of IGF-I by complex mechanisms, including enhancement of differentiation and apoptotic pathways, and inhibition of IGF-I induced anti-apoptotic signalling. Modulation of IGF binding protein abundance by FGF-2 does not play a significant role in inhibition of IGF-I induced mitogenesis.
Collapse
|
26
|
Russo VC, Kobayashi K, Najdovska S, Baker NL, Werther GA. Neuronal protection from glucose deprivation via modulation of glucose transport and inhibition of apoptosis: a role for the insulin-like growth factor system. Brain Res 2004; 1009:40-53. [PMID: 15120582 DOI: 10.1016/j.brainres.2004.02.042] [Citation(s) in RCA: 64] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/26/2004] [Indexed: 11/23/2022]
Abstract
Glucose is the brain's major energy source; therefore, loss of neuronal cells is a potential consequence of hypoglycaemia. Since apoptosis is a major mechanism of neuronal loss following a range of insults, we explored potent anti-apoptotic systems (IGF-I and bcl-2) as means of enhancing neuronal survival in the face of glucose deprivation. Human neuroblastoma cells (SH-SY5Y, SHEP and SHEP-bcl-2) were exposed to low glucose as a model of glucopenia-induced neuronal damage. Administration of IGF-I and/or over-expression of the survival gene bcl-2 were exploited to attempt to limit neuronal loss. Neuronal survival mechanisms and interactions between these systems were investigated. Low glucose (0.25-2.5 mM) adversely affected cell growth and survival; however, IGF-I ameliorated these outcomes. Over-expression of bcl-2 blunted low glucose-induced apoptosis and up-regulated IGF-I receptor, with the effect of IGF-I addition being negligible on apoptosis, while significantly enhancing mitochondrial activity. In SH-SY5Y cells, IGF-I significantly changed >two-fold mRNA levels of the apoptosis-related genes gadd45, fas, iNOS, NFkB, TRAIL, without further affecting bcl-2 expression. In low glucose, IGF-I acutely enhanced glucose transport and translocation of GLUT1 protein to the cell membrane. GLUT1 mRNA expression was up-regulated by both IGF-I and bcl-2. The potent anti-apoptotic systems IGF-I and bcl-2 are both thus able to enhance cell survival in a glucose-deprived human neuronal model. Although we clearly show evidence of positive cross-talk via bcl-2 modulation of IGF-I receptor, IGF-I also has enhancing effects on mitochondrial function outside the bcl-2 pathway. The common effect of both systems on enhancement of GLUT-1 expression suggests that this is a key mechanism for enhanced survival. These studies also point to the potential use of IGF-I therapy in prevention or amelioration of hypoglycaemic brain injury.
Collapse
Affiliation(s)
- V C Russo
- Centre for Hormone Research, Murdoch Childrens Research Institute, Royal Children's Hospital, Flemington Road, Parkville 3052, Victoria, Australia.
| | | | | | | | | |
Collapse
|
27
|
Barthlen W, Flaadt D, Girgert R, Conzelmann J, Schweizer P, Zugmaier G, Buck M, Knabbe C. Significance of heparin-binding growth factor expression on cells of solid pediatric tumors. J Pediatr Surg 2003; 38:1296-304. [PMID: 14523809 DOI: 10.1016/s0022-3468(03)00385-3] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
BACKGROUND The heparin-binding growth factors pleiotrophin (PTN), midkine (MK), vascular endothelial growth factor (VEGF), and basic fibroblast growth factor (bFGF) stimulate tumor cell proliferation and angiogenesis. In this study the authors wanted to know if these growth factors are expressed by cell lines and tumor tissue of solid pediatric tumors, growth factor expression is influenced by proinflammatory cytokines, and local growth factor concentration has an influence on experimental tumor growth. METHODS Growth factor mRNA expression was analyzed by reverse transcriptase polymerase chain reaction (RT-PCR) and protein secretion by enzyme-linked immunosorbent assay (ELISA). Neuroblastoma cells were suspended in solutions containing different growth factor concentrations before injection into the nude mice, which were given pentosan polysulfate (PPS) for antagonism. RESULTS The analyzed growth factors were expressed by most cells of solid malignant pediatric tumors. Their expression was not influenced by proinflammatory cytokines. The inhibition of tumor growth by PPS in the nude mouse model was dependent on the local growth factor concentration. High concentration excluded significant tumor suppression. CONCLUSIONS Because of the redundancy of growth factor expression and the abolishment of PPS efficacy by a high local growth factor concentration, the authors conclude that overall targeting of growth factors is a promising approach to cancer therapy in childhood.
Collapse
Affiliation(s)
- Winfried Barthlen
- Department of Pediatric Surgery, University Tübingen, Tübingen, Germany
| | | | | | | | | | | | | | | |
Collapse
|
28
|
Bian X, Opipari AW, Ratanaproeksa AB, Boitano AE, Lucas PC, Castle VP. Constitutively active NFkappa B is required for the survival of S-type neuroblastoma. J Biol Chem 2002; 277:42144-50. [PMID: 12198114 DOI: 10.1074/jbc.m203891200] [Citation(s) in RCA: 55] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The NFkappaB transcription factors can both promote cell survival and induce apoptosis depending on cell type and context. Neuroblastoma (NB) cells display two predominant culture phenotypes identified as N- and S-types. Malignant S-type cells express neither high levels of MYCN nor Bcl-2, suggesting that other survival mechanisms are important. We characterized NFkappaB activity in S-type cells and determined its role in their survival. S-type lines (SH-EP1 and SK-N-AS) were treated with pyrrolidine dithiocarbamate (PDTC), a NFkappaB inhibitor, or l-1-tosylamido-2-phenylethyl chloromethyl ketone (TPCK), a serine protease inhibitor that blocks IkappaBalpha degradation. Both agents induced cell death, suggesting that constitutive NFkappaB activity is required for survival. The transient expression of a super-repressor IkappaBalpha mutant killed S-type cells. The inhibition of NFkappaB produced an apoptotic response characterized by the collapse of the mitochondrial transmembrane electrochemical gradient, caspase-9 activation, and apoptotic DNA changes. Constitutive NFkappaB DNA binding activity specifically involving p65 and p50 was demonstrated in S- but not N-type cells by electromobility supershift and gene reporter assays. This study demonstrates a role for NFkappaB in the survival of S-type NB tumor cells and suggests that NFkappaB activity and function differ according to NB tumor cell phenotype.
Collapse
Affiliation(s)
- Xin Bian
- Department of Pediatrics, University of Michigan, Ann Arbor 48109, USA
| | | | | | | | | | | |
Collapse
|
29
|
Gallo R, Zazzeroni F, Alesse E, Mincione C, Borello U, Buanne P, D'Eugenio R, Mackay AR, Argenti B, Gradini R, Russo MA, Maroder M, Cossu G, Frati L, Screpanti I, Gulino A. REN: a novel, developmentally regulated gene that promotes neural cell differentiation. J Cell Biol 2002; 158:731-40. [PMID: 12186855 PMCID: PMC2174014 DOI: 10.1083/jcb.200202024] [Citation(s) in RCA: 44] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Expansion and fate choice of pluripotent stem cells along the neuroectodermal lineage is regulated by a number of signals, including EGF, retinoic acid, and NGF, which also control the proliferation and differentiation of central nervous system (CNS) and peripheral nervous system (PNS) neural progenitor cells. We report here the identification of a novel gene, REN, upregulated by neurogenic signals (retinoic acid, EGF, and NGF) in pluripotent embryonal stem (ES) cells and neural progenitor cell lines in association with neurotypic differentiation. Consistent with a role in neural promotion, REN overexpression induced neuronal differentiation as well as growth arrest and p27Kip1 expression in CNS and PNS neural progenitor cell lines, and its inhibition impaired retinoic acid induction of neurogenin-1 and NeuroD expression. REN expression is developmentally regulated, initially detected in the neural fold epithelium of the mouse embryo during gastrulation, and subsequently throughout the ventral neural tube, the outer layer of the ventricular encephalic neuroepithelium and in neural crest derivatives including dorsal root ganglia. We propose that REN represents a novel component of the neurogenic signaling cascade induced by retinoic acid, EGF, and NGF, and is both a marker and a regulator of neuronal differentiation.
Collapse
Affiliation(s)
- Rita Gallo
- Department of Experimental Medicine, University of L'Aquila, 67100 L'Aquila, Italy
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Tanaka A, Kamiakito T, Hakamata Y, Fujii A, Kuriki K, Fukayama M. Extensive neuronal localization and neurotrophic function of fibroblast growth factor 8 in the nervous system. Brain Res 2001; 912:105-15. [PMID: 11532426 DOI: 10.1016/s0006-8993(01)02726-3] [Citation(s) in RCA: 22] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Fibroblast growth factor (FGF) 8 has been well established to play a critical role in the early development of the central nervous system (CNS). We report here extensive neuronal localization and neurotrophic function of FGF8 in the nervous system. In sections of mouse embryos at E10.5, FGF8 was immunohistochemically found in neurons at the marginal zones of the CNS and in the dorsal root ganglia (DRG). Neuronal localization of FGF8 was marked at later embryonic stages and in adults, involving most of the central and peripheral neurons, including intermuscular enteric neurons, DRGs, and paraaortic sympathetic ganglia. Functionally, FGF8 promoted neurite outgrowth in human neuroblastoma SK-N-MC cells as well as in rat pheochromocytoma PC12 cells, suggesting that FGF8 acts as a neurotrophic factor. FGF8 also supported neuronal survival and differentiation in cultured human neural progenitor cells. In a cell growth assay, treatment with 50 ng/ml FGF8 on human cultured neuroblastoma SK-N-MC and IMR32 cells attenuated the growth of both. In accordance with these in vitro findings, the immunohistochemical analysis on human neurological diseases showed that FGF8 expression is evident in differentiating histological types of neuroblastoma and ganglioneuroblastoma, and that the levels of FGF8 immunoreactivity in the substantia nigra from Parkinson's disease are significantly lower than those in age-matched controls. Taken together, the present findings strongly suggest that FGF8 acts as a more generalized neurotrophic factor than previously reported.
Collapse
Affiliation(s)
- A Tanaka
- Department of Pathology, Jichi Medical School, 3311-1 Yakushiji, Minamikawachi, Kawachi, 329-0498, Tochigi, Japan.
| | | | | | | | | | | |
Collapse
|
31
|
Abstract
Neuroblastomas are heterogeneous tumors arising from sympathetic precursors in the neural crest. Growth factor stimulation of neuroblastomas promote diverse biological responses (mitogenesis, differentiation, cell death) depending on the particular tumor studied. Here we show that brief treatment with retinoic acid (RA) rendered the human neuroblastoma lines SY5Y, NGP, SMS-KCNR, and SK-N-SH dependent on brain-derived neurotrophic factor (BDNF) for survival. The BDNF- and trkB-expressing line SMS-KCN was dependent on an autocrine BDNF/trkB survival without exposure to RA. We conclude that the BDNF/trkB pathway plays an important role in neuroblastoma survival and speculate on a possible role in tumor pathogenesis.
Collapse
Affiliation(s)
- X Feng
- Department of Neurology, University of Chicago, Chicago, Illinois, USA
| | | | | | | | | |
Collapse
|
32
|
Beierle EA, Strande LF, Berger AC, Chen MK. VEGF is upregulated in a neuroblastoma and hepatocyte coculture model. J Surg Res 2001; 97:34-40. [PMID: 11319877 DOI: 10.1006/jsre.2001.6097] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
BACKGROUND We hypothesize that angiogenic factors are altered by the interaction between neuroblastoma cells and host tissues. MATERIALS AND METHODS Human Chang hepatocytes and human neuroblastoma cells are cultured separately and in a noncontact, coculture system. Immunostaining for VEGF is performed on the cells. ELISA is used to detect vascular endothelial growth factor (VEGF), basic fibroblast growth factor, and interleukin-8 in the conditioned media. Human umbilical vein endothelial cells (HUVEC) are cultured with standard medium (control) and hepatocyte, neuroblastoma, and coculture conditioned media. After 48 and 72 h, cells are counted to determine proliferation. Finally, VEGF-blocking antibody is added to the HUVEC cultures with the conditioned media. RESULTS VEGF is markedly elevated in the coculture medium compared to the media from hepatocytes or neuroblastoma grown alone [412.2 +/- 52 vs 235 +/- 35 or 74.5 +/- 28.5 (pg/10(6) cells), P < 0.05]. Other growth factors are almost undetectable in any of the media. Immunostaining for VEGF in the cocultured hepatocytes is decreased by almost 50%, but VEGF immunostaining is increased fourfold in the cocultured neuroblastoma cells. A significant increase in cell proliferation is seen at both 48 and 72 h when HUVEC are cultured with the coculture media. Cell proliferation is blocked with the addition of anti-VEGF antibody. CONCLUSION The interaction of neuroblastoma with hepatocytes results in an increased production of VEGF. It stimulates endothelial cell proliferation and may enhance the tumor's metastatic potential in an autocrine fashion.
Collapse
Affiliation(s)
- E A Beierle
- Division of Pediatric Surgery, University of Florida, Gainesville, Florida, USA.
| | | | | | | |
Collapse
|
33
|
Haïk S, Gauthier LR, Granotier C, Peyrin JM, Lages CS, Dormont D, Boussin FD. Fibroblast growth factor 2 up regulates telomerase activity in neural precursor cells. Oncogene 2000; 19:2957-66. [PMID: 10871847 DOI: 10.1038/sj.onc.1203596] [Citation(s) in RCA: 54] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
During brain development, neuronal and glial cells are generated from neural precursors on a precise schedule involving steps of proliferation, fate commitment and differentiation. We report that telomerase activity is highly expressed during embryonic murine cortical neurogenesis and early steps of gliogenesis and progressively decreases thereafter during cortex maturation to be undetectable in the normal adult brain. We evidenced neural precursor cells (NPC) as the principal telomerase-expressing cells in primary cultures from E15 mouse embryo cortices. Their differentiation either in neurons or in glial cells lead to a down regulation of telomerase activity that was directly correlated to the decrease of telomerase core protein (mTERT) mRNA synthesis. Furthermore, we show that FGF2 (fibroblast growth factor 2), one of the main regulators of CNS development, induces a dose-dependant increase of both the proliferation of NPC and telomerase activity in primary cortical cultures without affecting the mTERT mRNA synthesis compared to that of glyceraldehyde-3-phosphate dehydrogenase (mGAPDH). Finally, we evidenced that AZT (3'-azido-2', 3'-dideoxythymidine), known to inhibit telomerase activity, blocks in a dose dependant manner the FGF2-induced proliferation of NPC. Altogether, our results are in favor of an important role of telomerase activity during brain organogenesis. Oncogene (2000).
Collapse
Affiliation(s)
- S Haïk
- CEA, Service de Neurovirologie DSV/DRM, CRSSA, IPSC, BP 6, 92 265 Fontenay-aux-Roses cedex, France
| | | | | | | | | | | | | |
Collapse
|
34
|
Langer I, Vertongen P, Perret J, Fontaine J, Atassi G, Robberecht P. Expression of vascular endothelial growth factor (VEGF) and VEGF receptors in human neuroblastomas. MEDICAL AND PEDIATRIC ONCOLOGY 2000; 34:386-93. [PMID: 10842244 DOI: 10.1002/(sici)1096-911x(200006)34:6<386::aid-mpo2>3.0.co;2-3] [Citation(s) in RCA: 58] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
BACKGROUND Vascular endothelial growth factor (VEGF) is a specific endothelial cell mitogen that stimulates angiogenesis and plays a crucial role in tumor growth. The aim of the present study was to evaluate the expression of VEGF and of its two high-affinity tyrosine kinase receptors (KDR and Flt-1) in neuroblastoma surgical samples and cell lines. PROCEDURE The VEGF, KDR, and Flt-1 mRNA expression in neuroblastoma surgical samples and cell lines was studied by RT-PCR. The receptors were identified in [(125)I]VEGF binding and in functional studies (effect on cell growth). VEGF production by neuroblastomas was investigated by the ELISA method. RESULTS It was possible to observe the mRNAs encoding for VEGF and its two receptors in some of the surgical specimens examined, including most of the high-grade tumors. It was also possible to demonstrate that the SK-N-BE cell line expressed VEGF, KDR, and Flt-1 mRNAs as well as biologically active receptors: The cells bound [(125)I]-VEGF, and their growth was stimulated by exogenous VEGF. Moreover, VEGF protein could be detected in their culture conditioned medium. CONCLUSIONS These results suggest that, in addition to its effect on angiogenesis, VEGF may affect neuroblastoma cell growth directly and could be an autocrine growth factor.
Collapse
MESH Headings
- Child
- Child, Preschool
- DNA, Neoplasm/biosynthesis
- Endothelial Growth Factors/biosynthesis
- Endothelial Growth Factors/genetics
- Female
- Gene Expression Regulation, Neoplastic
- Humans
- Infant
- Lymphokines/biosynthesis
- Lymphokines/genetics
- Male
- Neuroblastoma/metabolism
- Proto-Oncogene Proteins/biosynthesis
- Proto-Oncogene Proteins/genetics
- RNA
- RNA, Messenger/analysis
- RNA, Neoplasm/analysis
- Receptor Protein-Tyrosine Kinases/biosynthesis
- Receptor Protein-Tyrosine Kinases/genetics
- Receptors, Growth Factor/biosynthesis
- Receptors, Growth Factor/genetics
- Receptors, Mitogen/biosynthesis
- Receptors, Mitogen/genetics
- Receptors, Vascular Endothelial Growth Factor
- Reverse Transcriptase Polymerase Chain Reaction
- Tumor Cells, Cultured
- Vascular Endothelial Growth Factor A
- Vascular Endothelial Growth Factor Receptor-1
- Vascular Endothelial Growth Factors
Collapse
Affiliation(s)
- I Langer
- Department of Pharmacology, Institute of Pharmacy, Université Libre de Bruxelles, Brussels, Belgium
| | | | | | | | | | | |
Collapse
|
35
|
Davy A, Feuerstein C, Robbins SM. Signaling within a caveolae-like membrane microdomain in human neuroblastoma cells in response to fibroblast growth factor. J Neurochem 2000; 74:676-83. [PMID: 10646519 DOI: 10.1046/j.1471-4159.2000.740676.x] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
It is now clear that the plasma membrane is not homogeneous but contains specific subcompartments characterized by their unique lipid and protein composition. Based on their enrichment in various signaling molecules, these microcompartments are now recognized to be sites of localized signal transduction for several extracellular stimuli. At least two different types of microdomains can be identified, largely based on the presence or absence of the caveolin proteins. The generic name of caveolae-like domains is commonly used to refer to both domains indistinguishably. Although caveolin proteins were long thought to be absent from the brain, we have shown that the human neuroblastoma cell line LAN-1 expresses both caveolin-1 and caveolin-2. Basic fibroblast growth factor (FGF)-2 induced a specific signaling response within the caveolae-like domain of LAN-1 cells, characterized by the tyrosine phosphorylation of a 75-80-kDa protein. This protein present in the caveolae-like domains has properties suggesting that it is a member of the SNT family of adapter proteins. The signaling event originating in the caveolae-like domains in response to FGF-2 appeared to require the activation of at least Fyn and Lyn, two members of the Src family of tyrosine kinases. This work suggests that compartmentalized signaling within caveolae-like domains may create a level of specificity for certain growth factors such as FGF.
Collapse
Affiliation(s)
- A Davy
- Department of Oncology, University of Calgary, Alberta, Canada
| | | | | |
Collapse
|
36
|
Van Golen CM, Feldman EL. Insulin-like growth factor I is the key growth factor in serum that protects neuroblastoma cells from hyperosmotic-induced apoptosis. J Cell Physiol 2000; 182:24-32. [PMID: 10567913 DOI: 10.1002/(sici)1097-4652(200001)182:1<24::aid-jcp3>3.0.co;2-6] [Citation(s) in RCA: 52] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Neuroblastoma is a childhood tumor of the peripheral nervous system that remains largely uncurable by conventional methods. Mannitol induces apoptosis in neuroblastoma cell types and insulin-like growth factor I (IGF-I) protects these cells from hyperosmotic-induced apoptosis by affecting apoptosis-regulatory proteins. In the current study, we investigate factors that enable SH-SY5Y neuroblastoma cells to survive in the presence of an apoptotic stimulus. When SH-SY5Y cells are exposed to high mannitol concentrations, more than 60% of the cells are apoptotic within 48 h. Normal CS prevents hyperosmotic-induced apoptosis in a dose-dependent manner, with 0.6% CS protecting 50% of the cells, and 3% CS rescuing more than 70% of the cells from apoptosis. Serum also delays the commitment point for SH-SY5Y cells from 9 h to 35 h. A survey of several growth factors, including epidermal growth factor (EGF), platelet-derived growth factor (PDGF), nerve growth factor (NGF), fibroblast growth factor (FGF), and IGF-I reveals that IGF-I is a component of serum necessary for protection of neuroblastoma cells from death. Mitochondrial membrane depolarization occurs in greater than 40% of the cells after mannitol exposure and caspase-3 activation is increased in high mannitol conditions after 9 h. IGF-I blocks both the mitochondrial membrane depolarization and caspase-3 activation normally induced by hyperosmotic treatment in neuroblastoma cells. Our results suggest that (1) IGF-I is a key factor in serum necessary for protection from death and (2) IGF-I acts upstream from the mitochondria and the caspases to prevent apoptosis in human neuroblastoma.
Collapse
Affiliation(s)
- C M Van Golen
- Department of Neurology, University of Michigan, Ann Arbor, Michigan, USA
| | | |
Collapse
|
37
|
Fredriksson A, Johnström P, Thorell JO, von Heijne G, Hassan M, Eksborg S, Kogner P, Borgström P, Ingvar M, Stone-Elander S. In vivo evaluation of the biodistribution of 11C-labeled PD153035 in rats without and with neuroblastoma implants. Life Sci 1999; 65:165-74. [PMID: 10416822 DOI: 10.1016/s0024-3205(99)00233-7] [Citation(s) in RCA: 45] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The biodistribution of 11C-labeled 4-(3-bromoanilino)-6,7-dimethoxyquinazoline, an inhibitor of the epidermal growth factor (EGF) receptor tyrosine kinase, has been evaluated in vivo in rats using positron emission tomography (PET). Time-activity data obtained after i.v. administration in one rat revealed that the radiotracer rapidly cleared from plasma with subsequent uptake in major organs of the body (brain, heart, liver, gastrointestinal tract and bladder). Uptake in proliferating tissue in rats with human neuroblastoma xenografts indicate that [O-11C-methyl]PD153035 shows promise as a new agent for in vivo imaging of tumors with PET.
Collapse
Affiliation(s)
- A Fredriksson
- Karolinska Pharmacy, and Clinical Neuroscience Department, Karolinska Hospital and Institute, Stockholm, Sweden.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Russo VC, Rekaris G, Baker NL, Bach LA, Werther GA. Basic fibroblast growth factor induces proteolysis of secreted and cell membrane-associated insulin-like growth factor binding protein-2 in human neuroblastoma cells. Endocrinology 1999; 140:3082-90. [PMID: 10385400 DOI: 10.1210/endo.140.7.6771] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Insulin-like growth factor (IGF) action in the brain is modulated by IGF-binding proteins (IGFBPs) whose abundance can be altered by other locally expressed growth factors. However, the mechanisms involved are unclear. We here employed the neuroblastoma cell line SK-N-MC as a model to define the mechanisms involved in modulation of IGFBPs in neuronal cells. Western ligand blotting analysis and immunoprecipitation of conditioned media (CM) from SK-N-MC cells showed that in these cells, as in the brain, the most abundantly expressed IGFBP was IGFBP-2. However, IGFBP-2 was barely detectable in CM from cells treated with basic fibroblast growth factor (bFGF) without a change in IGFBP-2 messenger RNA (mRNA) abundance. These CM contained specific IGFBP-2 proteolytic activity, resulting in two IGFBP-2 fragments of 14 and 22 kDa. The activity was inhibited by EDTA/phenylmethylsulfonyl fluoride or aprotinin. Competitive binding studies indicated that IGFBP-2 fragments had reduced binding affinity for IGF-I. bFGF induced IGFBP-3 mRNA and protein. Affinity cross-linking of [125I]IGF-I to neuroblastoma cell membranes followed by immunoprecipitation revealed a approximately 38 kDa [125I]IGF-I/IGFBP-2 complex. Cell surface-associated IGFBP-2 was also susceptible to bFGF-induced proteolysis, with the appearance of a single cross-linked 21-kDa complex with low affinity for IGF-I. These findings indicate that intact IGFBP-2 and the 14-kDa, but not the 22-kDa fragment, bind to the cell surface. Our data suggest that induction of IGFBP-2 proteolysis on neuronal cell surface is a novel mechanism whereby IGF availability is modulated by the local growth factor bFGF.
Collapse
Affiliation(s)
- V C Russo
- Centre for Hormone Research, Royal Children's Hospital Research Institute, Parkville, Victoria, Australia
| | | | | | | | | |
Collapse
|
39
|
Girgert R, Vogt Y, Becke D, Bruchelt G, Schweizer P. Growth inhibition of neuroblastoma cells by lovastatin and L-ascorbic acid is based on different mechanisms. Cancer Lett 1999; 137:167-72. [PMID: 10374838 DOI: 10.1016/s0304-3835(98)00355-3] [Citation(s) in RCA: 20] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Hydroxymethyl-glutaryl-CoA-reductase (HMG-CoA-reductase), the key enzyme for cholesterol synthesis and essential for the synthesis of the precursor for p21ras farnesylation, was inhibited in neuroblastoma cells by lovastatin or L-ascorbic acid. Both compounds inhibited clonogenic colony formation of neuroblastoma cells in soft agar. However, while the addition of mevalonate, the product of HMG-CoA-reductase, circumvented the inhibition by lovastatin it had no reversing effect on the inhibition by L-ascorbic acid. The role of reactive oxygen compounds generated by the degradation of catecholamines, and the pro-oxidative effects of L-ascorbic acid are discussed as mechanisms of action of L-ascorbic acid.
Collapse
Affiliation(s)
- R Girgert
- Department of Pediatric Surgery, Klinikum Schmarrenberg, University of Tübingen, Germany
| | | | | | | | | |
Collapse
|
40
|
Bale TL, Dorsa DM. Transcriptional regulation of the oxytocin receptor gene. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 1999; 449:307-15. [PMID: 10026818 DOI: 10.1007/978-1-4615-4871-3_38] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/10/2023]
Affiliation(s)
- T L Bale
- Department of Pharmacology, University of Washington, Seattle 98195, USA
| | | |
Collapse
|
41
|
Verity AN, Wyatt TL, Lee W, Hajos B, Baecker PA, Eglen RM, Johnson RM. Differential regulation of glial cell line-derived neurotrophic factor (GDNF) expression in human neuroblastoma and glioblastoma cell lines. J Neurosci Res 1999; 55:187-97. [PMID: 9972821 DOI: 10.1002/(sici)1097-4547(19990115)55:2<187::aid-jnr6>3.0.co;2-t] [Citation(s) in RCA: 54] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Human SK-N-AS neuroblastoma and U-87MG glioblastoma cell lines were found to secrete relatively high levels of glial cell line-derived neurotrophic factor (GDNF). In response to growth factors, cytokines, and pharmacophores, the two cell lines differentially regulated GDNF release. A 24-hr exposure to tumor necrosis factor-alpha (TNFalpha; 10 ng/ml) or interleukin-1beta (IL-1,; 10 ng/ml) induced GDNF release in U-87MG cells, but repressed GDNF release from SK-N-AS cells. Fibroblast growth factors (FGF)-1, -2, and -9 (50 ng/ml), the prostaglandins PGA2, PGE2, and PGI2 (10 microM), phorbol 12,13-didecanoate (PDD; 10 nM), okadaic acid (10 nM), dexamethasone (1 microM), and vitamin D3 (1 microm) also differentially effected GDNF release from U-87MG and SK-N-AS cells. A result shared by both cell lines, was a two- to threefold increase in GDNF release by db-cAMP (1 mM), or forskolin (10 microM). In general, analysis of steady-state GDNF mRNA levels correlated with changes in extracellular GDNF levels in U-87MG cells but remained static in SK-N-AS cells. The data suggest that human GDNF synthesis/release can be regulated by numerous factors, signaling through multiple and diverse secondary messenger systems. Furthermore, we provide evidence of differential regulation of human GDNF synthesis/release in cells of glial (U-87MG) and neuronal (SK-N-AS) origin.
Collapse
Affiliation(s)
- A N Verity
- Department of Molecular and Cellular Biochemistry, Center for Biological Research, Roche Bioscience, Palo Alto, California 94304-1397, USA.
| | | | | | | | | | | | | |
Collapse
|
42
|
Cassoni P, Sapino A, Stella A, Fortunati N, Bussolati G. Presence and significance of oxytocin receptors in human neuroblastomas and glial tumors. Int J Cancer 1998; 77:695-700. [PMID: 9688301 DOI: 10.1002/(sici)1097-0215(19980831)77:5<695::aid-ijc6>3.0.co;2-q] [Citation(s) in RCA: 54] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
To determine whether oxytocin (OT) could be added to the list of growth factors acting on neoplastic cells of nervous origin, we investigated the presence of oxytocin receptors (OTR) in human primary neuroblastomas and glioblastomas and related cell lines. OTR were demonstrated both at mRNA level (using a RT-PCR procedure) and at protein level (using immunocytochemical and immunofluorescence procedures). In order to clarify whether OT exerts any biological effect on these tumors through OTR, we also studied cell proliferation in 3 human neuroblastoma cell lines (SK-N-SH, SH-SY5Y, IMR-32) and one human anaplastic astrocytoma cell line (MOG-G-UVW) treated with OT 1 nM to 100 nM for 48 and 96 hr. At these doses, a dose-dependent inhibitory effect on cell proliferation was demonstrated. This inhibition was accompanied by a significant increase in the intracellular concentration of cAMP, which we have reported to be the intracellular mediator of the OT anti-proliferative effect in breast-carcinoma cell lines. Our data indicate that specific OTR are present in human neuroblastomas and glioblastomas. Through these receptors, OT could inhibit cell proliferation and modulate tumor growth.
Collapse
Affiliation(s)
- P Cassoni
- Department of Biomedical Sciences and Oncology, University of Turin, Italy
| | | | | | | | | |
Collapse
|
43
|
Chen MK, Strande L, Kain M, Whalen TV, Doolin EJ. Induction of apoptosis in a neuroblastoma and hepatocyte coculture model. J Surg Res 1998; 78:123-30. [PMID: 9733629 DOI: 10.1006/jsre.1998.5363] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
BACKGROUND The dysregulation of apoptosis may alter the progression of tumor growth and explain the clinical dichotomy observed in children with neuroblastoma (NB). An overexpression of the bcl-2 proto-oncogene induces resistance to apoptosis and has been observed in unfavorable NB. We hypothesized that alterations in apoptosis may be a result of the interactions between NB and the tissues surrounding it. MATERIALS AND METHODS Human Chang liver cells (HCL, 10(4) cells/cm2) were plated in two-chamber slides for 3 days. Human NB cells (10(5) cells/cm2) were added to one of the chambers and incubated for 3 more days. Control NB were plated under identical conditions in its own medium and in the HCL medium with growth curves measured. DNA fragmentation was detected via the TUNEL method (TdT-mediated nick end-labeling) and bcl-2 expression was determined by immunostaining. RESULTS NB growth was unaltered by the change in medium. NB stained mildly positive for bcl-2 when plated alone but became markedly positive in coculture. Histologically, HCL and NB appeared healthy when plated alone, but a halo of apoptotic HCL was seen around NB in the coculture. When plated alone, both NB and HCL demonstrated minimal apoptotic activity as detected via the TUNEL method. In the coculture, a halo of HCL surrounding the NB exhibited markedly increased DNA fragmentation and this intensity diminished in cells distant from the NB. CONCLUSIONS The regulation of apoptosis was altered in this coculture model of NB and HCL. HCL stimulated NB to overexpress bcl-2 and presumably become resistant to apoptosis. Conversely, NB induced the surrounding HCL to undergo apoptosis. The interaction between the local tissue and NB induced alterations in apoptosis in both cell types and resulted in a survival advantage for NB.
Collapse
Affiliation(s)
- M K Chen
- Department of Surgery, University of Medicine and Dentistry of New Jersey, Camden, New Jersey, 08103, USA
| | | | | | | | | |
Collapse
|
44
|
Missale C, Codignola A, Sigala S, Finardi A, Paez-Pereda M, Sher E, Spano PF. Nerve growth factor abrogates the tumorigenicity of human small cell lung cancer cell lines. Proc Natl Acad Sci U S A 1998; 95:5366-71. [PMID: 9560282 PMCID: PMC20267 DOI: 10.1073/pnas.95.9.5366] [Citation(s) in RCA: 38] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Nerve growth factor (NGF) has antiproliferative and differentiating effects on adenomas of neuroendocrine origin. Cell lines derived from small-cell lung carcinoma (SCLC), a very aggressive neuroendocrine tumor, express NGF receptors. The role of NGF in the control of proliferation and progression of this carcinoma, however, has never been investigated. Chronic exposure of NCI-N-592 and GLC8 SCLC cell lines to NGF remarkably inhibited their proliferation rate both in vitro and in vivo, prevented their anchorage-independent clonal growth in soft agar, impaired their invasive capacity in vitro, and abolished their tumorigenic potential in nude mice. The proliferative response of SCLC cell lines to nicotine was also remarkably impaired by in vitro NGF treatment. Furthermore, NGF treatment activates in SCLC cell lines the expression and secretion of NGF. NGF thus reverts SCLC cell lines to a noninvasive, nontumorigenic phenotype that does not respond to nicotine and produces NGF.
Collapse
Affiliation(s)
- C Missale
- Department of Biomedical Sciences and Biotechnology, Division of Pharmacology, University of Brescia, Via Valsabbina 19, 25124, Brescia, Italy.
| | | | | | | | | | | | | |
Collapse
|
45
|
Bale TL, Dorsa DM. NGF, cyclic AMP, and phorbol esters regulate oxytocin receptor gene transcription in SK-N-SH and MCF7 cells. BRAIN RESEARCH. MOLECULAR BRAIN RESEARCH 1998; 53:130-7. [PMID: 9473629 DOI: 10.1016/s0169-328x(97)00287-8] [Citation(s) in RCA: 24] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Oxytocin receptor (OTR) gene transcription has predominantly been thought to be regulated by estrogen. However, the continuous presence of receptors in certain brain regions after gonadectomy suggests the existence of alternate mechanisms of regulation. We have cloned and sequenced 4 kb of 5'-flanking DNA of the rat OTR gene and identified an internal segment which was absent in the initial publication of this promoter sequence. Sequence analysis of this segment, as well as of a novel upstream region, revealed the presence of a CRE as well as several other potential regulatory elements, including AP-1, AP-2, AP-3, AP-4 sites, an ERE, and a half-SRE (SRE/2). The effects of phorbol 12-myristate 13-acetate (PMA), forskolin, and NGF treatment on this promoter were tested in transfection experiments in MCF7 and SK-N-SH cells. Transcription of the full-length OTR promoter was induced by forskolin and by the phorbol ester PMA, and a synergistic (17-fold) effect was observed in MCF7 cells treated with both agents. Receptor binding studies using the OTR antagonist 125I-labeled ornithine vasotocin, and Western blot analyses of OTRs in MCF7 cells, showed that PMA and forskolin also increased the density of endogenous human oxytocin receptors. Mutational analyses of the CRE and half-SRE sites in this promoter indicated that these elements function as enhancers and support forskolin and NGF effects, respectively, on transcription. These studies have identified a novel region of the rat OTR promoter containing elements which impart cAMP and/or phorbol ester inducibility of OTR gene transcription. A potential role of the PKA and/or PKC pathways in OTR gene regulation is suggested.
Collapse
Affiliation(s)
- T L Bale
- Department of Pharmacology, University of Washington, Box 357280, Seattle, WA 98195, USA.
| | | |
Collapse
|
46
|
Luo J, Miller MW. Differential Sensitivity of Human Neuroblastoma Cell Lines to Ethanol: Correlations with Their Proliferative Responses to Mitogenic Growth Factors and Expression of Growth Factor Receptors. Alcohol Clin Exp Res 1997. [DOI: 10.1111/j.1530-0277.1997.tb04436.x] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
47
|
Torcia M, Bracci-Laudiero L, Lucibello M, Nencioni L, Labardi D, Rubartelli A, Cozzolino F, Aloe L, Garaci E. Nerve growth factor is an autocrine survival factor for memory B lymphocytes. Cell 1996; 85:345-56. [PMID: 8616890 DOI: 10.1016/s0092-8674(00)81113-7] [Citation(s) in RCA: 293] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Production of nerve growth factor (NGF) was assessed in cultures of human T and B lymphocytes and macrophages. NGF was constitutively produced by B cells only, which also expressed surface p140trk-A and p75NGFR molecules and hence efficiently bound and internalized the cytokine. Neutralization of endogenous NGF caused disappearance of Bcl-2 protein and apoptotic death of resting lymphocytes bearing surface IgG or IgA, a population comprising memory cells, while surface IgM/IgD "virgin" B lymphocytes were not affected. In vivo administration of neutralizing anti-NGF antibodies caused strong reduction in the titer of specific IgG in mice immunized with tetanus toxoid, nitrophenol, or arsonate and reduced numbers of surface IgG or IgA B lymphocytes. Thus, NGF is an autocrine survival factor for memory B lymphocytes.
Collapse
Affiliation(s)
- M Torcia
- Department of Experimental Medicine, University of Rome "Tor Vergata", Italy
| | | | | | | | | | | | | | | | | |
Collapse
|