1
|
Chernov A, Kudryavtsev I, Komlev A, Alaverdian D, Tsapieva A, Galimova E, Shamova O. Nerve Growth Factor, Antimicrobial Peptides and Chemotherapy: Glioblastoma Combination Therapy to Improve Their Efficacy. Biomedicines 2023; 11:3009. [PMID: 38002009 PMCID: PMC10669874 DOI: 10.3390/biomedicines11113009] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Revised: 10/07/2023] [Accepted: 10/24/2023] [Indexed: 11/26/2023] Open
Abstract
Glioblastoma (GBM) is an aggressive and lethal malignancy of the central nervous system with a median survival rate of 15 months. We investigated the combined anticancer effects of nerve growth factor (NGF), cathelicidin (LL-37), and protegrin-1 (PG-1) with chemotherapy (temozolomide, doxorubicin, carboplatin, cisplatin, and etoposide) in the glioblastoma U251 cell line to overcome the limitations of conventional chemotherapy and to guarantee specific treatments to succeed. The MTT (3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide) assay was used to study cell viability and to determine the cytotoxic effects of NGF, LL-37, and PG-1 and their combination with chemotherapy in U251 cells. Synergism or antagonism was determined using the combination index (CI) method. Caspase-3 activity was evaluated spectrophotometrically using a caspase-3 activity assay kit. Apoptosis was analyzed with flow cytometry using propidium iodide (PI) and YO-PRO-1. NGF and the peptides showed a strong cytotoxic effect on U251 glioma cells in the MTT test (IC50 0.0214, 3.1, and 26.1 μM, respectively) compared to chemotherapy. The combination of PG-1 + etoposide had a synergistic effect on apoptosis of U251 glioma cells. It should be noted that the cells were in the early and late stages of apoptosis, respectively, compared with the control cells. The caspase-3 activation analysis revealed that the caspase-3 level was not significantly (p > 0.05) increased in U251 cells following PG-1 with etoposide treatment compared with that in the untreated cells, suggesting that the combination of PG-1 and etoposide may induce caspase-independent apoptosis in U251 cells. NGF, LL-37, and PG-1 represent promising drug candidates as the treatment regimen for GBM. Furthermore, the synergistic efficacy of the combined protocol using PG-1 and etoposide may overcome some of the typical limitations of the conventional therapeutic protocols, thus representing a promising approach for GBM therapy.
Collapse
Affiliation(s)
- Alexandr Chernov
- Institute of Experimental Medicine, WCRC “Center for Personalized Medicine”, Saint-Petersburg 197022, Russia; (I.K.); (A.K.); (A.T.); (O.S.)
| | - Igor Kudryavtsev
- Institute of Experimental Medicine, WCRC “Center for Personalized Medicine”, Saint-Petersburg 197022, Russia; (I.K.); (A.K.); (A.T.); (O.S.)
| | - Aleksei Komlev
- Institute of Experimental Medicine, WCRC “Center for Personalized Medicine”, Saint-Petersburg 197022, Russia; (I.K.); (A.K.); (A.T.); (O.S.)
| | - Diana Alaverdian
- Medical Genetics, Department of Medical Biotechnologies, University of Siena, 53100 Siena, Italy;
| | - Anna Tsapieva
- Institute of Experimental Medicine, WCRC “Center for Personalized Medicine”, Saint-Petersburg 197022, Russia; (I.K.); (A.K.); (A.T.); (O.S.)
| | - Elvira Galimova
- Institute of Experimental Medicine, WCRC “Center for Personalized Medicine”, Saint-Petersburg 197022, Russia; (I.K.); (A.K.); (A.T.); (O.S.)
- Sechenov Institute of Evolutionary Physiology and Biochemistry, Russian Academy of Sciences, Saint-Petersburg 194223, Russia
| | - Olga Shamova
- Institute of Experimental Medicine, WCRC “Center for Personalized Medicine”, Saint-Petersburg 197022, Russia; (I.K.); (A.K.); (A.T.); (O.S.)
- Department of Biochemistry, Saint Petersburg State University, Saint-Petersburg 199034, Russia
| |
Collapse
|
2
|
Gao L, Wu J, Wang H, Yang Y, Zheng Z, Ni B, Wang X, Peng Y, Li Y. LMO1 Plays an Oncogenic Role in Human Glioma Associated With NF-kB Pathway. Front Oncol 2022; 12:770299. [PMID: 35280742 PMCID: PMC8907846 DOI: 10.3389/fonc.2022.770299] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2021] [Accepted: 01/24/2022] [Indexed: 12/15/2022] Open
Abstract
Background LIM domain only protein1(LMO1), a nuclear transcription coregulator, is implicated in the pathogenesis of T-cell acute lymphoblastic leukemia and neuroblastoma. However, the clinical significance and potential mechanism of LMO1 in human gliomas remain to be determined. Methods In this study, expression level data and clinical information were obtained via three databases. The Cox proportional hazards regression model was used to predict outcomes for glioma patients. In vitro and in vivo assays were used to explore the function of LMO1 in human glioma. Gene set enrichment analysis (GSEA), RNA-seq and western blot were used to explore the potential molecular mechanisms. A prognostic model was built for predicting the overall survival(OS) of human glioma patients. Results High LMO1 expression was associated with a high tumor grade and a poor prognosis in patients. High levels of LMO1 mRNA were correlated with poor prognosis in patients with isocitrate dehydrogenase (IDH)-wild-type (wt) and 1p/19q non-codeletion gliomas. Gene silencing of LMO1 significantly inhibited tumor growth, invasion and migration in vitro. In contrast, LMO1 over-expression promoted tumor growth, invasion and migration. Mechanically, LMO1 may positively regulate the level of NGFR mRNA and protein. NGFR mediated the regulation between LMO1 and NF-kB activation. Consistently, the nude mice study further confirmed that knockdown of LMO1 blocked tumor growth via NGFR-NF-kB axis. Finally, The nomogram based on the LMO1 signature for overall survival (OS) prediction in human glioma patients exhibited good performance in the individual mortality risk. Conclusion This study provides new insights and evidences that high level expression of LMO1 is significantly correlated with progression and prognosis in human gliomas. LMO1 played a critical role in tumorigenesis and progression. The present study first investigated the LMO1–NGFR–NF-kB axis regulate cell growth and invasion in human glioma cells, whereby targeting this pathway may be a therapeutic target for glioma.
Collapse
Affiliation(s)
- Lei Gao
- Department of Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou, China.,Laboratory for Precision Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Jia Wu
- Department of Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou, China.,Laboratory for Precision Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Hai Wang
- Department of Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou, China.,Laboratory for Precision Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Yongyu Yang
- Department of Hepatobiliary Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Zongliao Zheng
- Department of Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou, China.,Laboratory for Precision Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Bowen Ni
- Department of Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou, China.,Laboratory for Precision Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Xiran Wang
- Department of Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou, China.,Laboratory for Precision Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Yuping Peng
- Department of Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou, China.,Laboratory for Precision Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Yaomin Li
- Department of Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou, China.,Laboratory for Precision Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou, China
| |
Collapse
|
3
|
Parmigiani E, Scalera M, Mori E, Tantillo E, Vannini E. Old Stars and New Players in the Brain Tumor Microenvironment. Front Cell Neurosci 2021; 15:709917. [PMID: 34690699 PMCID: PMC8527006 DOI: 10.3389/fncel.2021.709917] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Accepted: 08/04/2021] [Indexed: 12/12/2022] Open
Abstract
In recent years, the direct interaction between cancer cells and tumor microenvironment (TME) has emerged as a crucial regulator of tumor growth and a promising therapeutic target. The TME, including the surrounding peritumoral regions, is dynamically modified during tumor progression and in response to therapies. However, the mechanisms regulating the crosstalk between malignant and non-malignant cells are still poorly understood, especially in the case of glioma, an aggressive form of brain tumor. The presence of unique brain-resident cell types, namely neurons and glial cells, and an exceptionally immunosuppressive microenvironment pose additional important challenges to the development of effective treatments targeting the TME. In this review, we provide an overview on the direct and indirect interplay between glioma and neuronal and glial cells, introducing new players and mechanisms that still deserve further investigation. We will focus on the effects of neural activity and glial response in controlling glioma cell behavior and discuss the potential of exploiting these cellular interactions to develop new therapeutic approaches with the aim to preserve proper brain functionality.
Collapse
Affiliation(s)
- Elena Parmigiani
- Embryology and Stem Cell Biology, Department of Biomedicine, University of Basel, Basel, Switzerland
| | - Marta Scalera
- Neuroscience Institute, Consiglio Nazionale delle Ricerche (CNR), Pisa, Italy
| | | | - Elena Tantillo
- Neuroscience Institute, Consiglio Nazionale delle Ricerche (CNR), Pisa, Italy
| | - Eleonora Vannini
- Neuroscience Institute, Consiglio Nazionale delle Ricerche (CNR), Pisa, Italy
| |
Collapse
|
4
|
Franzese O, Di Francesco AM, Meco D, Graziani G, Cusano G, Levati L, Riccardi R, Ruggiero A. hTERT Transduction Extends the Lifespan of Primary Pediatric Low-Grade Glioma Cells While Preserving the Biological Response to NGF. Pathol Oncol Res 2021; 27:612375. [PMID: 34257579 PMCID: PMC8262147 DOI: 10.3389/pore.2021.612375] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Accepted: 02/10/2021] [Indexed: 12/21/2022]
Abstract
The neurotrophin nerve growth factor (NGF) modulates the growth of human gliomas and is able to induce cell differentiation through the engagement of tropomyosin receptor kinase A (TrkA) receptor, although the role played in controlling glioma survival has proved controversial. Unfortunately, the slow growth rate of low-grade gliomas (LGG) has made it difficult to investigate NGF effects on these tumors in preclinical models. In fact, patient-derived low-grade human astrocytoma cells duplicate only a limited number of times in culture before undergoing senescence. Nevertheless, replicative senescence can be counteracted by overexpression of hTERT, the catalytic subunit of telomerase, which potentially increases the proliferative potential of human cells without inducing cancer-associated changes. We have extended, by hTERT transduction, the proliferative in vitro potential of a human LGG cell line derived from a pediatric pilocytic astrocytoma (PA) surgical sample. Remarkably, the hTERT-transduced LGG cells showed a behavior similar to that of the parental line in terms of biological responses to NGF treatment, including molecular events associated with induction of NGF-related differentiation. Therefore, transduction of LGG cells with hTERT can provide a valid approach to increase the in vitro life-span of patient-derived astrocytoma primary cultures, characterized by a finite proliferative potential.
Collapse
Affiliation(s)
- Ornella Franzese
- Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy
| | - Angela M Di Francesco
- Institute of Internal Medicine, Periodic Fever and Rare Diseases Center, Fondazione Policlinico A. Gemelli, IRCCS, Rome, Italy
| | - Daniela Meco
- UOC di Oncologia Pediatrica, "Fondazione Policlinico Universitario A. Gemelli", IRCCS, Rome, Italy
| | - Grazia Graziani
- Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy
| | - Gabriella Cusano
- UOC di Oncologia Pediatrica, "Fondazione Policlinico Universitario A. Gemelli", IRCCS, Rome, Italy
| | | | - Riccardo Riccardi
- UOC di Oncologia Pediatrica, "Fondazione Policlinico Universitario A. Gemelli", IRCCS, Rome, Italy
| | - Antonio Ruggiero
- UOC di Oncologia Pediatrica, "Fondazione Policlinico Universitario A. Gemelli", IRCCS, Rome, Italy
| |
Collapse
|
5
|
Chernov AN, Alaverdian DA, Glotov OS, Talabaev MV, Urazov SP, Shcherbak SG, Renieri A, Frullanti E, Shamova O. Related expression of TRKA and P75 receptors and the changing copy number of MYC-oncogenes determine the sensitivity of brain tumor cells to the treatment of the nerve growth factor in combination with cisplatin and temozolomide. Drug Metab Pers Ther 2020; 0:/j/dmdi.ahead-of-print/dmdi-2020-0109/dmdi-2020-0109.xml. [PMID: 32887179 DOI: 10.1515/dmdi-2020-0109] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2020] [Accepted: 07/29/2020] [Indexed: 02/07/2023]
Abstract
Objectives Oncological diseases are an urgent medical and social problem. The chemotherapy induces not only the death of the tumor cells but also contributes to the development of their multidrug resistance and death of the healthy cells and tissues. In this regard, the search for the new pharmacological substances with anticancer activity against drug-resistant tumors is of utmost importance. In the present study we primarily investigated the correlation between the expression of TrkA and p75 receptors with the nerve growth factor (NGF) and cisplatin or temozolomide sensitivity of anaplastic astrocytoma (AA), glioblastoma (GB) and medulloblastoma (MB) cell cultures. We then evaluated the changing of copy numbers of MYCC and MYCN and its correlation with cytotoxicity index (CI) in MB cells under NGF exposition. Methods The primary cell cultures were obtained from the tumor biopsy samples of the patients with AA (n=5), GB (n=7) or MB (n=25) prior to radiotherapy and chemotherapy. The cytotoxicity effect of NGF and its combinations with cisplatin or temozolomide, the relative expression of TrkA and p75 receptors, its correlations with CI in AA, GB and MB primary cell cultures were studied by trypan blue cytotoxicity assay and immunofluorescence staining respectively. The effect of NGF on MYCC and MYCN copy numbers in MB cell cultures was studied by fluorescence in situ hybridization. Results We found that the expression of TrkA and p75 receptors (p=0.03) and its ratio (p=0.0004) depends on the sensitivity of AA and GB cells to treatment with NGF and its combinations with cisplatin or temozolomide. NGF reduces (p<0.05) the quantity of MB cells with six or eight copies of MYCN and three or eight copies of MYCC. Besides, NGF increases (p<0.05) the quantity of MB cells containing two copies of both oncogenes. The negative correlation (r=-0.65, p<0.0001) is established between MYCC average copy numbers and CI of NGF in MB cells. Conclusions The relative expression of NGF receptors (TrkA/p75) and its correlation with CI of NGF and its combinations in AA and GB cells point to the mechanism involving a cell death signaling pathway. NGF downregulates (p<0.05) some increased copy numbers of MYCC and MYCN in the human MB cell cultures, and upregulates normal two copies of both oncogenes (p<0.05).
Collapse
Affiliation(s)
- Alexandr N Chernov
- Department of Clinics and Genetics Investigations, Saint Petersburg City Hospital No40 of Resort District, Saint Petersburg, Russian Federation
- Laboratory of Design and Synthesis of Biologically Active Peptides, Department of General Pathology and Pathophysiology, Institute of Experimental Medicine, Saint-Petersburg, Russian Federation
| | - Diana A Alaverdian
- Medical Genetics, Department of Medical Biotechnologies, University of Siena, Siena, Italy
| | - Oleg S Glotov
- Department of Clinics and Genetics Investigations, Saint Petersburg City Hospital No40 of Resort District, Saint Petersburg, Russian Federation
| | - Michael V Talabaev
- Department of Pediatric Neurosurgery, Republican Center for Neurology and Neurosurgery, Minsk, The Republic of Belarus
| | - Stanislav P Urazov
- Department of Clinics and Genetics Investigations, Saint Petersburg City Hospital No40 of Resort District, Saint Petersburg, Russian Federation
| | - Sergei G Shcherbak
- Department of Clinics and Genetics Investigations, Saint Petersburg City Hospital No40 of Resort District, Saint Petersburg, Russian Federation
| | - Alessandra Renieri
- Medical Genetics, Department of Medical Biotechnologies, University of Siena, Siena, Italy
- Genetica Medica, Azienda Ospedaliera Universitaria Senese, Siena, Italy
| | - Elisa Frullanti
- Medical Genetics, Department of Medical Biotechnologies, University of Siena, Siena, Italy
| | - Olga Shamova
- Laboratory of Design and Synthesis of Biologically Active Peptides, Department of General Pathology and Pathophysiology, Institute of Experimental Medicine, Saint-Petersburg, Russian Federation
| |
Collapse
|
6
|
Chernov AN, Alaverdian DA, Glotov OS, Talabaev MV, Urazov SP, Shcherbak SG, Renieri A, Frullanti E, Shamova O. Related expression of TRKA and P75 receptors and the changing copy number of MYC-oncogenes determine the sensitivity of brain tumor cells to the treatment of the nerve growth factor in combination with cisplatin and temozolomide. Drug Metab Pers Ther 2020; 35:dmpt-2020-0109. [PMID: 34704697 DOI: 10.1515/dmpt-2020-0109] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2020] [Accepted: 07/29/2020] [Indexed: 12/24/2022]
Abstract
OBJECTIVES Oncological diseases are an urgent medical and social problem. The chemotherapy induces not only the death of the tumor cells but also contributes to the development of their multidrug resistance and death of the healthy cells and tissues. In this regard, the search for the new pharmacological substances with anticancer activity against drug-resistant tumors is of utmost importance. In the present study we primarily investigated the correlation between the expression of TrkA and p75 receptors with the nerve growth factor (NGF) and cisplatin or temozolomide sensitivity of anaplastic astrocytoma (AA), glioblastoma (GB) and medulloblastoma (MB) cell cultures. We then evaluated the changing of copy numbers of MYCC and MYCN and its correlation with cytotoxicity index (CI) in MB cells under NGF exposition. METHODS The primary cell cultures were obtained from the tumor biopsy samples of the patients with AA (n=5), GB (n=7) or MB (n=25) prior to radiotherapy and chemotherapy. The cytotoxicity effect of NGF and its combinations with cisplatin or temozolomide, the relative expression of TrkA and p75 receptors, its correlations with CI in AA, GB and MB primary cell cultures were studied by trypan blue cytotoxicity assay and immunofluorescence staining respectively. The effect of NGF on MYCC and MYCN copy numbers in MB cell cultures was studied by fluorescence in situ hybridization. RESULTS We found that the expression of TrkA and p75 receptors (p=0.03) and its ratio (p=0.0004) depends on the sensitivity of AA and GB cells to treatment with NGF and its combinations with cisplatin or temozolomide. NGF reduces (p<0.05) the quantity of MB cells with six or eight copies of MYCN and three or eight copies of MYCC. Besides, NGF increases (p<0.05) the quantity of MB cells containing two copies of both oncogenes. The negative correlation (r=-0.65, p<0.0001) is established between MYCC average copy numbers and CI of NGF in MB cells. CONCLUSIONS The relative expression of NGF receptors (TrkA/p75) and its correlation with CI of NGF and its combinations in AA and GB cells point to the mechanism involving a cell death signaling pathway. NGF downregulates (p<0.05) some increased copy numbers of MYCC and MYCN in the human MB cell cultures, and upregulates normal two copies of both oncogenes (p<0.05).
Collapse
Affiliation(s)
- Alexandr N Chernov
- Department of Clinics and Genetics Investigations, Saint Petersburg City Hospital No40 of Resort District, Saint Petersburg, Russian Federation.,Laboratory of Design and Synthesis of Biologically Active Peptides, Department of General Pathology and Pathophysiology, Institute of Experimental Medicine, Saint-Petersburg, Russian Federation
| | - Diana A Alaverdian
- Medical Genetics, Department of Medical Biotechnologies, University of Siena, Siena, Italy
| | - Oleg S Glotov
- Department of Clinics and Genetics Investigations, Saint Petersburg City Hospital No40 of Resort District, Saint Petersburg, Russian Federation
| | - Michael V Talabaev
- Department of Pediatric Neurosurgery, Republican Center for Neurology and Neurosurgery, Minsk, The Republic of Belarus
| | - Stanislav P Urazov
- Department of Clinics and Genetics Investigations, Saint Petersburg City Hospital No40 of Resort District, Saint Petersburg, Russian Federation
| | - Sergei G Shcherbak
- Department of Clinics and Genetics Investigations, Saint Petersburg City Hospital No40 of Resort District, Saint Petersburg, Russian Federation
| | - Alessandra Renieri
- Medical Genetics, Department of Medical Biotechnologies, University of Siena, Siena, Italy.,Genetica Medica, Azienda Ospedaliera Universitaria Senese, Siena, Italy
| | - Elisa Frullanti
- Medical Genetics, Department of Medical Biotechnologies, University of Siena, Siena, Italy
| | - Olga Shamova
- Laboratory of Design and Synthesis of Biologically Active Peptides, Department of General Pathology and Pathophysiology, Institute of Experimental Medicine, Saint-Petersburg, Russian Federation
| |
Collapse
|
7
|
Meco D, Di Francesco AM, Melotti L, Ruggiero A, Riccardi R. Ectopic nerve growth factor prevents proliferation in glioma cells by senescence induction. J Cell Physiol 2018; 234:6820-6830. [PMID: 30417351 DOI: 10.1002/jcp.27430] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2018] [Accepted: 08/27/2018] [Indexed: 12/30/2022]
Abstract
OBJECTIVE The neurotrophin nerve growth factor (NGF) affects survival, regulation and differentiation of both central and peripheral nervous system neurons. NGF exerts its effects primarily through tropomyosin receptor kinase A (TrkA), inducing a cascade of tyrosine kinase-initiated responses. In spite of its importance, the general behavior of NGF looks contradictory: its effects can be both stimulatory and inhibitory. The present study aims to explore the molecular mechanisms induced by NGF in glioma cancer cells. METHODS The effects of NGF were investigated in high grade glioma and low grade pediatric glioma (PLGG) cell lines through comparative studies. In particular, we investigated TrkA-mediated cellular pathways, molecular signaling, proliferation, cell cycle and cellular senescence. RESULTS We found that exposure of PLGG cells to NGF produced stable growth arrest with the features of a senescence phenotype but without the expression of anti-poly(ADP-ribose) polymerase cleavage, a marker of apoptosis. Moreover, NGF treatment promoted the phosphorylation of extracellular signal-regulated kinase1/2 (ERK1/2), signal transducer and activator of transcription 3 (STAT3), and phosphatidylinositol-3-kinase (PI3K)/protein kinase B (AKT) signaling. In addition, K252a, a TrkA inhibitor, significantly reduced the phosphorylation of the aforementioned signaling pathways, suggesting that NGF-activated ERK1/2 and AKT signaling take place downstream of TrkA-neurotrophin interaction. CONCLUSIONS These findings provide the first evidence that NGF can induce senescence of PLGG cells in a receptor-mediated fashion, thus supporting the hypothesis that in the clinical setting NGF might be beneficial to pediatric glioma patients.
Collapse
Affiliation(s)
- Daniela Meco
- Oncologia Pediatrica, Fondazione Policlinico Universitario A. Gemelli, IRCSS, Rome, Italy
| | | | | | - Antonio Ruggiero
- Oncologia Pediatrica, Fondazione Policlinico Universitario A. Gemelli, IRCSS, Rome, Italy.,Università Cattolica del Sacro Cuore, Rome, Italy
| | - Riccardo Riccardi
- Oncologia Pediatrica, Fondazione Policlinico Universitario A. Gemelli, IRCSS, Rome, Italy
| |
Collapse
|
8
|
Alshehri MM, Robbins SM, Senger DL. The Role of Neurotrophin Signaling in Gliomagenesis: A Focus on the p75 Neurotrophin Receptor (p75 NTR/CD271). VITAMINS AND HORMONES 2017; 104:367-404. [PMID: 28215302 DOI: 10.1016/bs.vh.2016.11.001] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
The p75 neurotrophin receptor (p75NTR, a.k.a. CD271), a transmembrane glycoprotein and a member of the tumor necrosis family (TNF) of receptors, was originally identified as a nerve growth factor receptor in the mid-1980s. While p75NTR is recognized to have important roles during neural development, its presence in both neural and nonneural tissues clearly supports the potential to mediate a broad range of functions depending on cellular context. Using an unbiased in vivo selection paradigm for genes underlying the invasive behavior of glioma, a critical characteristic that contributes to poor clinical outcome for glioma patients, we identified p75NTR as a central regulator of glioma invasion. Herein we review the expanding role that p75NTR plays in glioma progression with an emphasis on how p75NTR may contribute to the treatment refractory nature of glioma. Based on the observation that p75NTR is expressed and functional in two critical glioma disease reservoirs, namely, the highly infiltrative cells that evade surgical resection, and the radiation- and chemotherapy-resistant brain tumor-initiating cells (also referred to as brain tumor stem cells), we propose that p75NTR and its myriad of downstream signaling effectors represent rationale therapeutic targets for this devastating disease. Lastly, we provide the provocative hypothesis that, in addition to the well-documented cell autonomous signaling functions, the neurotrophins, and their respective receptors, contribute in a cell nonautonomous manner to drive the complex cellular and molecular composition of the brain tumor microenvironment, an environment that fuels tumorigenesis.
Collapse
Affiliation(s)
- M M Alshehri
- Arnie Charbonneau Cancer Centre, University of Calgary, Calgary, AB, Canada
| | - S M Robbins
- Arnie Charbonneau Cancer Centre, University of Calgary, Calgary, AB, Canada
| | - D L Senger
- Arnie Charbonneau Cancer Centre, University of Calgary, Calgary, AB, Canada.
| |
Collapse
|
9
|
Aloe L, Rocco ML, Balzamino BO, Micera A. Nerve growth factor: role in growth, differentiation and controlling cancer cell development. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2016; 35:116. [PMID: 27439311 PMCID: PMC4955168 DOI: 10.1186/s13046-016-0395-y] [Citation(s) in RCA: 58] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/18/2016] [Accepted: 07/12/2016] [Indexed: 02/01/2023]
Abstract
Recent progress in the Nerve Growth Factor (NGF) research has shown that this factor acts not only outside its classical domain of the peripheral and central nervous system, but also on non-neuronal and cancer cells. This latter observation has led to divergent hypothesis about the role of NGF, its specific distribution pattern within the tissues and its implication in induction as well as progression of carcinogenesis. Moreover, other recent studies have shown that NGF has direct clinical relevance in certain human brain neuron degeneration and a number of human ocular disorders. These studies, by suggesting that NGF is involved in a plethora of physiological function in health and disease, warrant further investigation regarding the true role of NGF in carcinogenesis. Based on our long-lasting experience in the physiopathology of NGF, we aimed to review previous and recent in vivo and in vitro NGF studies on tumor cell induction, progression and arrest. Overall, these studies indicate that the only presence of NGF is unable to generate cell carcinogenesis, both in normal neuronal and non-neuronal cells/tissues. However, it cannot be excluded the possibility that the co-expression of NGF and pro-carcinogenic molecules might open to different consequence. Whether NGF plays a direct or an indirect role in cell proliferation during carcinogenesis remains to demonstrate.
Collapse
Affiliation(s)
- Luigi Aloe
- Institute of Cell Biology and Neurobiology, CNR, Via Del Fosso di Fiorano, 64 I-00143, Rome, Italy.
| | - Maria Luisa Rocco
- Institute of Cell Biology and Neurobiology, CNR, Via Del Fosso di Fiorano, 64 I-00143, Rome, Italy
| | | | - Alessandra Micera
- IRCCS - G.B. Bietti Foundation, Via Santo Stefano Rotondo, 6 I-00184, Rome, Italy
| |
Collapse
|
10
|
Demir IE, Tieftrunk E, Schorn S, Friess H, Ceyhan GO. Nerve growth factor & TrkA as novel therapeutic targets in cancer. Biochim Biophys Acta Rev Cancer 2016; 1866:37-50. [PMID: 27264679 DOI: 10.1016/j.bbcan.2016.05.003] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2016] [Revised: 05/24/2016] [Accepted: 05/28/2016] [Indexed: 12/11/2022]
Abstract
In the past 20years, nerve growth factor (NGF) and its receptors TrkA & p75NTR were recognized to be overexpressed in the overwhelming majority of human solid cancers. Recent studies discovered the presence of overactive TrkA signaling due to TrkA rearrangements or TrkA fusion products in frequent cancers like colorectal cancer, thyroid cancer, or acute myeloid leukemia. Thus, targeting TrkA/NGF via selective small-molecule-inhibitors or antibodies has gained enormous attention in the drug discovery sector. Clinical studies on the anti-cancer impact of NGF-blocking antibodies are likely to be accelerated after the recent removal of clinical holds on these agents by regulatory authorities. Based on these current developments, the present review provides not only a broad overview of the biological effects of NGF-TrkA-p75NTR on cancer cells and their microenvironment, but also explains why NGF and its receptors are going to evoke major interest as promising therapeutic anti-cancer targets in the coming decade.
Collapse
Affiliation(s)
- Ihsan Ekin Demir
- Department of Surgery, Klinikum rechts der Isar, Technische Universität München, 81675 Munich, Germany.
| | - Elke Tieftrunk
- Department of Surgery, Klinikum rechts der Isar, Technische Universität München, 81675 Munich, Germany
| | - Stephan Schorn
- Department of Surgery, Klinikum rechts der Isar, Technische Universität München, 81675 Munich, Germany
| | - Helmut Friess
- Department of Surgery, Klinikum rechts der Isar, Technische Universität München, 81675 Munich, Germany
| | - Güralp O Ceyhan
- Department of Surgery, Klinikum rechts der Isar, Technische Universität München, 81675 Munich, Germany
| |
Collapse
|
11
|
Falsini B, Chiaretti A, Rizzo D, Piccardi M, Ruggiero A, Manni L, Soligo M, Dickmann A, Federici M, Salerni A, Timelli L, Guglielmi G, Lazzareschi I, Caldarelli M, Galli-Resta L, Colosimo C, Riccardi R. Nerve growth factor improves visual loss in childhood optic gliomas: a randomized, double-blind, phase II clinical trial. ACTA ACUST UNITED AC 2016; 139:404-14. [PMID: 26767384 DOI: 10.1093/brain/awv366] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2015] [Accepted: 10/26/2015] [Indexed: 11/15/2022]
Abstract
Paediatric optic pathway gliomas are low-grade brain tumours characterized by slow progression and invalidating visual loss. Presently there is no strategy to prevent visual loss in this kind of tumour. This study evaluated the effects of nerve growth factor administration in protecting visual function in patients with optic pathway glioma-related visual impairment. A prospective randomized double-blind phase II clinical trial was conducted in 18 optic pathway glioma patients, aged from 2 to 23 years, with stable disease and severe visual loss. Ten patients were randomly assigned to receive a single 10-day course of 0.5 mg murine nerve growth factor as eye drops, while eight patients received placebo. All patients were evaluated before and after treatment, testing visual acuity, visual field, visual-evoked potentials, optic coherence tomography, electroretinographic photopic negative response, and magnetic resonance imaging. Post-treatment evaluations were repeated at 15, 30, 90, and 180 days Brain magnetic resonance imaging was performed at baseline and at 180 days. Treatment with nerve growth factor led to statistically significant improvements in objective electrophysiological parameters (electroretinographic photopic negative response amplitude at 180 days and visual-evoked potentials at 30 days), which were not observed in placebo-treated patients. Furthermore, in patients in whom visual fields could still be measured, visual field worsening was only observed in placebo-treated cases, while three of four nerve growth factor-treated subjects showed significant visual field enlargement. This corresponded to improved visually guided behaviour, as reported by the patients and/or the caregivers. There was no evidence of side effects related to nerve growth factor treatment. Nerve growth factor eye drop administration appears a safe, easy and effective strategy for the treatment of visual loss associated with optic pathway gliomas.
Collapse
Affiliation(s)
- Benedetto Falsini
- 1 Institute of Ophthalmology, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| | - Antonio Chiaretti
- 2 Paediatric Oncology, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| | - Daniela Rizzo
- 2 Paediatric Oncology, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| | - Marco Piccardi
- 1 Institute of Ophthalmology, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| | - Antonio Ruggiero
- 2 Paediatric Oncology, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| | - Luigi Manni
- 3 Institute of Translational Pharmacology, CNR, 00142 Rome, Italy
| | - Marzia Soligo
- 3 Institute of Translational Pharmacology, CNR, 00142 Rome, Italy
| | - Anna Dickmann
- 1 Institute of Ophthalmology, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| | - Matteo Federici
- 1 Institute of Ophthalmology, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| | - Annabella Salerni
- 1 Institute of Ophthalmology, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| | | | - Gaspare Guglielmi
- 5 Pharmacy Gemelli Hospital, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| | - Ilaria Lazzareschi
- 2 Paediatric Oncology, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| | - Massimo Caldarelli
- 6 Paediatric Neurosurgery, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| | | | - Cesare Colosimo
- 8 Institute of Radiology, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| | - Riccardo Riccardi
- 2 Paediatric Oncology, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| |
Collapse
|
12
|
Nijaguna MB, Patil V, Hegde AS, Chandramouli BA, Arivazhagan A, Santosh V, Somasundaram K. An Eighteen Serum Cytokine Signature for Discriminating Glioma from Normal Healthy Individuals. PLoS One 2015; 10:e0137524. [PMID: 26390214 PMCID: PMC4577083 DOI: 10.1371/journal.pone.0137524] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2015] [Accepted: 08/18/2015] [Indexed: 02/04/2023] Open
Abstract
Glioblastomas (GBM) are largely incurable as they diffusely infiltrate adjacent brain tissues and are difficult to diagnose at early stages. Biomarkers derived from serum, which can be obtained by minimally invasive procedures, may help in early diagnosis, prognosis and treatment monitoring. To develop a serum cytokine signature, we profiled 48 cytokines in sera derived from normal healthy individuals (n = 26) and different grades of glioma patients (n = 194). We divided the normal and grade IV glioma/GBM serum samples randomly into equal sized training and test sets. In the training set, the Prediction Analysis for Microarrays (PAM) identified a panel of 18 cytokines that could discriminate GBM sera from normal sera with maximum accuracy (95.40%) and minimum error (4.60%). The 18-cytokine signature obtained in the training set discriminated GBM sera from normal sera in the test set as well (accuracy 96.55%; error 3.45%). Interestingly, the 18-cytokine signature also differentiated grade II/Diffuse Astrocytoma (DA) and grade III/Anaplastic Astrocytoma (AA) sera from normal sera very efficiently (DA vs. normal–accuracy 96.00%, error 4.00%; AA vs. normal–accuracy 95.83%, error 4.17%). Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analysis using 18 cytokines resulted in the enrichment of two pathways, cytokine-cytokine receptor interaction and JAK-STAT pathways with high significance. Thus our study identified an 18-cytokine signature for distinguishing glioma sera from normal healthy individual sera and also demonstrated the importance of their differential abundance in glioma biology.
Collapse
Affiliation(s)
- Mamatha B. Nijaguna
- Department of Microbiology and Cell Biology, Indian Institute of Science, Bangalore 560012, India
| | - Vikas Patil
- Department of Microbiology and Cell Biology, Indian Institute of Science, Bangalore 560012, India
| | - Alangar S. Hegde
- Department of Neurosurgery, Sri Satya Sai Institute of Higher Medical Sciences, Bangalore 560066, India
| | - Bangalore A. Chandramouli
- Department of Neurosurgery, National Institute of Mental Health and Neuro Sciences, Bangalore 560029, India
| | - Arimappamagan Arivazhagan
- Department of Neurosurgery, National Institute of Mental Health and Neuro Sciences, Bangalore 560029, India
| | - Vani Santosh
- Department of Neuropathology, National Institute of Mental Health and Neuro Sciences, Bangalore 560029, India
| | - Kumaravel Somasundaram
- Department of Microbiology and Cell Biology, Indian Institute of Science, Bangalore 560012, India
- * E-mail:
| |
Collapse
|
13
|
Swartling FJ, Čančer M, Frantz A, Weishaupt H, Persson AI. Deregulated proliferation and differentiation in brain tumors. Cell Tissue Res 2015; 359:225-54. [PMID: 25416506 PMCID: PMC4286433 DOI: 10.1007/s00441-014-2046-y] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2014] [Accepted: 10/22/2014] [Indexed: 01/24/2023]
Abstract
Neurogenesis, the generation of new neurons, is deregulated in neural stem cell (NSC)- and progenitor-derived murine models of malignant medulloblastoma and glioma, the most common brain tumors of children and adults, respectively. Molecular characterization of human malignant brain tumors, and in particular brain tumor stem cells (BTSCs), has identified neurodevelopmental transcription factors, microRNAs, and epigenetic factors known to inhibit neuronal and glial differentiation. We are starting to understand how these factors are regulated by the major oncogenic drivers in malignant brain tumors. In this review, we will focus on the molecular switches that block normal neuronal differentiation and induce brain tumor formation. Genetic or pharmacological manipulation of these switches in BTSCs has been shown to restore the ability of tumor cells to differentiate. We will discuss potential brain tumor therapies that will promote differentiation in order to reduce treatment resistance, suppress tumor growth, and prevent recurrence in patients.
Collapse
Affiliation(s)
- Fredrik J Swartling
- Department of Immunology, Genetics and Pathology, Science for Life Laboratory, Rudbeck Laboratory, Uppsala University, Uppsala, SE-751 85, Sweden
| | - Matko Čančer
- Department of Immunology, Genetics and Pathology, Science for Life Laboratory, Rudbeck Laboratory, Uppsala University, Uppsala, SE-751 85, Sweden
| | - Aaron Frantz
- Departments of Neurology and Neurological Surgery, Sandler Neurosciences Center, University of California, San Francisco, CA, 94158, USA
- Brain Tumor Research Center, University of California, San Francisco, CA, 94158, USA
| | - Holger Weishaupt
- Department of Immunology, Genetics and Pathology, Science for Life Laboratory, Rudbeck Laboratory, Uppsala University, Uppsala, SE-751 85, Sweden
| | - Anders I Persson
- Departments of Neurology and Neurological Surgery, Sandler Neurosciences Center, University of California, San Francisco, CA, 94158, USA
- Brain Tumor Research Center, University of California, San Francisco, CA, 94158, USA
| |
Collapse
|
14
|
Investigation on The Inhibition Effects of Bone Marrow Mesenchymal Stem Cells With CD Gene on Glioma in C6 Rat*. PROG BIOCHEM BIOPHYS 2012. [DOI: 10.3724/sp.j.1206.2011.00157] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
15
|
Kosaka H, Ichikawa T, Kurozumi K, Kambara H, Inoue S, Maruo T, Nakamura K, Hamada H, Date I. Therapeutic effect of suicide gene-transferred mesenchymal stem cells in a rat model of glioma. Cancer Gene Ther 2012; 19:572-8. [DOI: 10.1038/cgt.2012.35] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
|
16
|
Eskandary H, Basiri M, Nematollahi-Mahani SN, Mehravaran S. The role of stem cells in tumor targeting and growth suppression of gliomas. Biologics 2011; 5:61-70. [PMID: 21637731 PMCID: PMC3104605 DOI: 10.2147/btt.s17838] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2011] [Indexed: 01/14/2023]
Abstract
Glioma remains the most challenging solid organ tumor to treat successfully. Based on the capacity of stem cells to migrate extensively and target invading glioma cells, the transplantation of stem cells as a cell-based delivery system may provide additional tools for the treatment of gliomas. In addition to the use of modified stem cells for the delivery of therapeutic agents, unmodified stem cells have been shown to have growth-suppressing effects on tumors in vitro and in vivo. This review outlines the probable factors involved in tumor tropism and tumor growth suppression, with a specific focus on the use of unmodified stem cells in the treatment of gliomas. Based on these and further future data, clinical trials may be justified.
Collapse
|
17
|
Roesler R, Brunetto AT, Abujamra AL, de Farias CB, Brunetto AL, Schwartsmann G. Current and emerging molecular targets in glioma. Expert Rev Anticancer Ther 2011; 10:1735-51. [PMID: 21080801 DOI: 10.1586/era.10.167] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Gliomas are the most common and lethal neurological cancers. Despite research efforts, the prognosis for patients with malignant gliomas remains poor. Advances in the understanding of cellular and molecular alterations in gliomas have led to the emergence of experimental molecularly targeted therapies. This article summarizes recent progress in the development of targeted therapies for glioma, focusing on emerging molecular targets, including neuropeptide and neurotrophin pathways, glutamate receptors, epigenetic mechanisms and glioma stem cell targets. Recent clinical trials of small molecules and antibodies targeted at growth factor pathways and intracellular signaling cascades are also discussed.
Collapse
Affiliation(s)
- Rafael Roesler
- Laboratory of Molecular Neuropharmacology, Department of Pharmacology, Institute for Basic Health Sciences, Federal University of Rio Grande do Sul, 90050-170 Porto Alegre, RS, Brazil.
| | | | | | | | | | | |
Collapse
|
18
|
Chiaretti A, Falsini B, Servidei S, Marangoni D, Pierri F, Riccardi R. Nerve growth factor eye drop administration improves visual function in a patient with optic glioma. Neurorehabil Neural Repair 2011; 25:386-90. [PMID: 21343523 DOI: 10.1177/1545968310395601] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND Spontaneous visual improvement in people with an optic glioma (OG) of the anterior or retrochiasmatic optic pathways is rare. OBJECTIVE To evaluate the effects on visual function of nerve growth factor (NGF) eye drop administration in a patient with severe visual impairment due to a low-grade OG. METHODS A 45-year-old woman with OG and long-standing optic nerve atrophy was assessed before and after 2 NGF treatment courses. The drug used was 2.5S murine NGF. One milligram of NGF, diluted in saline solution, was administered onto the conjunctiva of both eyes for 10 consecutive days 3 times a day for each treatment. The follow-up was performed by clinical, neuroradiologic, and electrophysiological tests (electroretinogram and visual evoked potentials [VEPs]) at the end of each treatment and 30 and 60 days later. RESULTS A repeated subjective and objective improvement of visual function (>3 lines visual acuity; >40° visual field; >50% VEP amplitude increase, Wilcoxon test P < .01) was recorded after NGF treatment. These measures tended to deteriorate toward baseline values 60 days from the end of each NGF treatment. No ocular or systemic side effects were observed throughout treatment. CONCLUSIONS Conjunctival NGF may be a beneficial adjunct therapy for visual loss in patients with OG, possibly exerting its effects on residual viable optic pathways.
Collapse
Affiliation(s)
- Antonio Chiaretti
- Department of Pediatric Neuroscience, Catholic University of Rome, Rome, Italy.
| | | | | | | | | | | |
Collapse
|
19
|
Le Hericium erinaceus: des propriétés essentiellement dépendantes du neuronal growth factor. ACTA ACUST UNITED AC 2011. [DOI: 10.1007/s10298-010-0601-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
|
20
|
Cragnolini AB, Huang Y, Gokina P, Friedman WJ. Nerve growth factor attenuates proliferation of astrocytes via the p75 neurotrophin receptor. Glia 2009; 57:1386-92. [PMID: 19229990 DOI: 10.1002/glia.20857] [Citation(s) in RCA: 66] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The p75 neurotrophin receptor has been implicated in the regulation of multiple cellular functions that differ depending on the cell context. We have observed that p75(NTR) is strongly induced on astrocytes as well as neurons in the hippocampal CA3 region after seizures; however, the function of this receptor on these glial cells has not been defined. We have employed a primary culture system to investigate the effects of neurotrophins on astrocytes. Treatment of hippocampal astrocytes with nerve growth factor (NGF) caused a reduction in cell number, but did not elicit an apoptotic response, in contrast to hippocampal neurons. Instead, activation of p75(NTR) by NGF attenuated proliferation induced by mitogens such as EGF or serum. These studies demonstrate the cell type specificity of neurotrophin functions in the brain.
Collapse
Affiliation(s)
- Andrea B Cragnolini
- Department of Biological Sciences, Rutgers University, Newark, New Jersey 07102, USA
| | | | | | | |
Collapse
|
21
|
Brown MC, Staniszewska I, Lazarovici P, Tuszynski GP, Del Valle L, Marcinkiewicz C. Regulatory effect of nerve growth factor in alpha9beta1 integrin-dependent progression of glioblastoma. Neuro Oncol 2009; 10:968-80. [PMID: 19074980 DOI: 10.1215/15228517-2008-047] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
In the present study we described the role of alpha9beta1 integrin in glioblastoma progression following its interaction with nerve growth factor (NGF). The level of expression of alpha9beta1 on astrocytomas is correlated with increased grade of this brain tumor and is highest on glioblastoma, whereas normal astrocytes do not express this integrin. Two glioblastoma cell lines, LN229 and LN18, that are alpha9beta1 integrin positive and negative, respectively, were used for alpha9beta1 integrin-dependent NGF-induced tumor progression. NGF was a significant promoter of promigratory and pro-proliferative activities of glioblastoma cells through direct interaction with alpha9beta1 integrin and activation of MAPK Erk1/2 pathway. The level of NGF increases approximately threefold in the most malignant glioma tissue when compared with normal brain. This increase is related to secretion of NGF by tumor cells. Specific inhibitors of alpha9beta1 integrin or gene silencing inhibited NGF-induced proliferation of LN229 cell line to the level shown by LN18 cells. VLO5 promoted alpha9beta1-dependent programmed cell death by induction of intrinsic apoptosis pathway in cancer cells. LN229 cells were rescued from proapoptotic effect of VLO5 by the presence of NGF. This disintegrin significantly inhibited tumor growth induced by implantation of LN229 cells to the chorioallantoic membrane (CAM) of quail embryonic model, and this inhibitory effect was significantly abolished by the presence of NGF. alpha9beta1 integrin appears to be an interesting target for blocking the progression of malignant gliomas, especially in light of the stimulatory effect of NGF on the development of these tumors and its ability to transfer proapoptotic signals in cancer cells.
Collapse
Affiliation(s)
- Meghan C Brown
- Department of Neuroscience, Center for Neurovirology and Cancer Biology, School of Medicine, Temple University, Philadelphia, PA, USA
| | | | | | | | | | | |
Collapse
|
22
|
Brown MC, Staniszewska I, Lazarovici P, Tuszynski GP, Del Valle L, Marcinkiewicz C. Regulatory effect of nerve growth factor in α9β1 integrin–dependent progression of glioblastoma. Neuro Oncol 2008. [DOI: 10.1215/15228517-2008-0047] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023] Open
|
23
|
Kang SG, Jeun SS, Lim JY, Kim SM, Yang YS, Oh WI, Huh PW, Park CK. Cytotoxicity of human umbilical cord blood-derived mesenchymal stem cells against human malignant glioma cells. Childs Nerv Syst 2008; 24:293-302. [PMID: 17968556 DOI: 10.1007/s00381-007-0515-2] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/03/2007] [Indexed: 01/14/2023]
Abstract
BACKGROUND Mesenchymal stem cells (MSCs) represent a potential useful source for cell-based glioma therapies because these cells evidence both orthodox and unorthodox plasticity and also show tropism for cancer. In this study, the authors attempted to access the cytotoxicity of human umbilical cord blood (hUCB)-derived MSCs, with or without cytokine activations against malignant glioma cells. MATERIALS AND METHODS hUCB-derived MSCs were activated by interleukin-2, interleukin-15, granulocyte macrophage colony-stimulating factor, and combinations. The hUCB-derived MSCs and activated hUCB-derived MSCs were effector cells. The cytotoxicity of the unactivated hUCB-derived MSCs and activated hUCB-derived MSCs against the target cells (human malignant glioma cells) was estimated via visual survival cell assays and transwell inserts. Phenotypic changes occurring in these hUCB-derived MSCs before and after cytokine activation were determined via flow cytometry. The secreted proteins from these effector cells were estimated via enzyme-linked immunosorbent assays. RESULTS We noted a significant cytotoxicity of hUCB-derived MSCs against malignant glioma cells. In addition, the hUCB-derived MSCs activated with cytokines evidenced significantly higher cytotoxicity than that observed with unactivated hUCB-derived MSCs. Differentiated immune effectors cells from the hUCB-derived MSCs after cytokine activation were not shown to have increased in number. However, the activated hUCB-derived MSCs secreted more immune response-related proteins (interleukin 4, interferon-gamma) than did the unactivated hUCB-derived MSCs. CONCLUSION The data collected herein confirm for the first time that hUCB-derived MSCs, with or without activation, evidence significant cytotoxicity against human malignant glioma cells, and the immune response-related proteins secreted in this process may perform relevant functions.
Collapse
Affiliation(s)
- Seok-Gu Kang
- Department of Neurosurgery, The Catholic University of Korea College of Medicine, Uijeongbu St. Mary's Hospital, 65-1, Kumoh-dong, Uijeongbu 480-130, South Korea
| | | | | | | | | | | | | | | |
Collapse
|
24
|
Cragnolini AB, Friedman WJ. The function of p75NTR in glia. Trends Neurosci 2008; 31:99-104. [PMID: 18199491 DOI: 10.1016/j.tins.2007.11.005] [Citation(s) in RCA: 78] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2007] [Revised: 11/20/2007] [Accepted: 11/21/2007] [Indexed: 11/19/2022]
Abstract
The p75 neurotrophin receptor (p75(NTR)) is expressed on many cell types and can influence a variety of cellular functions. This receptor can mediate cell survival or cell death, can promote or inhibit axonal growth and can facilitate or attenuate proliferation, depending on the cell context. The emerging picture regarding p75(NTR) indicates that it can partner with different coreceptors to dictate specific responses. It then signals by recruiting intracellular binding proteins to activate different signaling pathways. The function of p75(NTR) has mainly been studied in neurons; however, it is also expressed in a variety of glial populations, especially during development and after injury, where its roles have been poorly defined. In this review, we will examine the potential roles for p75(NTR) in glial function.
Collapse
Affiliation(s)
- Andrea B Cragnolini
- Department of Biological Sciences, Rutgers University, Newark, NJ 07102, USA
| | | |
Collapse
|
25
|
Expression of cannabinoid receptors and neurotrophins in human gliomas. Neurol Sci 2008; 28:304-10. [DOI: 10.1007/s10072-007-0843-8] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2007] [Accepted: 12/22/2007] [Indexed: 10/22/2022]
|
26
|
Hansen K, Wagner B, Hamel W, Schweizer M, Haag F, Westphal M, Lamszus K. Autophagic cell death induced by TrkA receptor activation in human glioblastoma cells. J Neurochem 2007; 103:259-75. [PMID: 17635673 DOI: 10.1111/j.1471-4159.2007.04753.x] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
The neurotrophin receptor tropomyosin-related kinase A (TrkA) and its ligand nerve growth factor (NGF) are expressed in astrocytomas, and an inverse association of TrkA expression with malignancy grade was described. We hypothesized that TrkA expression might confer a growth disadvantage to glioblastoma cells. To analyze TrkA function and signaling, we transfected human TrkA cDNA into the human glioblastoma cell line G55. We obtained three stable clones, all of which responded with striking cytoplasmic vacuolation and subsequent cell death to NGF. Analyzing the mechanism of cell death, we could exclude apoptosis and cellular senescence. Instead, we identified several indications of autophagy: electron microscopy showed typical autophagic vacuoles; acridine orange staining revealed acidic vesicular organelles; acidification of acidic vesicular organelles was prevented using bafilomycin A1; cells displayed arrest in G2/M; increased processing of LC3 occurred; vacuolation was prevented by the autophagy inhibitor 3-methyladenine; no caspase activation was detected. We further found that both activation of ERK and c-Jun N-terminal kinase but not p38 were involved in autophagic vacuolation. To conclude, we identified autophagy as a novel mechanism of NGF-induced cell death. Our findings suggest that TrkA activation in human glioblastomas might be beneficial therapeutically, especially as several of the currently used chemotherapeutics also induce autophagic cell death.
Collapse
Affiliation(s)
- Katharina Hansen
- Department of Neurosurgery, University Medical Center Hamburg-Eppendorf, Martinistrasse, Hamburg, Germany
| | | | | | | | | | | | | |
Collapse
|
27
|
Kang SG, Jeun SS, Lim JY, Yoo DS, Huh PW, Cho KS, Kim DS, Shin HJ, Kim JH, Kim MC, Kang JK. Cytotoxicity of rat marrow stromal cells against malignant glioma cells. Childs Nerv Syst 2005; 21:528-38. [PMID: 15933882 DOI: 10.1007/s00381-005-1216-3] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/22/2005] [Indexed: 01/14/2023]
Abstract
OBJECTS Marrow stromal cells (MSCs) have been shown to have the capacity of orthodox and unorthodox plasticity. In this study, the authors tried to access in vitro cytotoxicity of MSCs from rat and also to differentiate MSCs into immune effector cell. METHODS Rat MSCs (rMSCs) were isolated by standard methodology and were activated by interleukin-2 (IL-2), interleukin-15 (IL-15), granulocyte macrophage colony stimulating factor, and combinations, which were effector cells. Cytotoxicity of rMSCs and activated rMSCs against the target cells (9L rat glioma cell line) was estimated using visual survival cell assay. Phenotypes of these various activated cells were determined using flow cytometry. The secreted protein from effector cells was estimated by enzyme-linked immunosorbent assay. The expression of immune response-related genes in activated cells was measured. RESULTS There was a significant cytotoxicity of rMSCs activated with various cytokine combinations. After various cytokine activations of rMSCs, the population of immune effector cells (CD8, CD161a) and immune reaction-related proteins (IL-4, gamma-INF) might increase. Apoptosis may be one of the lysis mechanisms of target cells by activated rMSCs. The contributing genes could be gamma-INF, FasL, and perforin. CONCLUSION This study suggests that rMSC may be used as adoptive transfer therapy in patients suffering from malignant brain tumor, but we have to investigate orthotopic animal study for the proper translation.
Collapse
Affiliation(s)
- Seok-Gu Kang
- Department of Neurosurgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, Gangnam-gu, Seoul, Republic of Korea
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Nakamura K, Ito Y, Kawano Y, Kurozumi K, Kobune M, Tsuda H, Bizen A, Honmou O, Niitsu Y, Hamada H. Antitumor effect of genetically engineered mesenchymal stem cells in a rat glioma model. Gene Ther 2004; 11:1155-64. [PMID: 15141157 DOI: 10.1038/sj.gt.3302276] [Citation(s) in RCA: 439] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The prognosis of patients with malignant glioma is extremely poor, despite the extensive surgical treatment that they receive and recent improvements in adjuvant radio- and chemotherapy. In the present study, we propose the use of gene-modified mesenchymal stem cells (MSCs) as a new tool for gene therapy of malignant brain neoplasms. Primary MSCs isolated from Fischer 344 rats possessed excellent migratory ability and exerted inhibitory effects on the proliferation of 9L glioma cell in vitro. We also confirmed the migratory capacity of MSCs in vivo and showed that when they were inoculated into the contralateral hemisphere, they migrated towards 9L glioma cells through the corpus callosum. MSCs implanted directly into the tumor localized mainly at the border between the 9L tumor cells and normal brain parenchyma, and also infiltrated into the tumor bed. Intratumoral injection of MSCs caused significant inhibition of 9L tumor growth and increased the survival of 9L glioma-bearing rats. Gene-modification of MSCs by infection with an adenoviral vector encoding human interleukin-2 (IL-2) clearly augmented the antitumor effect and further prolonged the survival of tumor-bearing rats. Thus, gene therapy employing MSCs as a targeting vehicle would be promising as a new therapeutic approach for refractory brain tumor.
Collapse
Affiliation(s)
- K Nakamura
- Department of Molecular Medicine, Sapporo Medical University, Sapporo, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|