1
|
Speedy/RINGO protein interacts with ERK/MAPK and PI3K/AKT pathways in SH-SY5Y neuroblastoma cells. Mol Cell Biochem 2020; 473:133-141. [PMID: 32602013 DOI: 10.1007/s11010-020-03813-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2020] [Accepted: 06/19/2020] [Indexed: 10/24/2022]
Abstract
Abnormal activity of ERK/MAPK and PI3K/AKT pathways is one of the most important factors for the development of many cancer types including neuroblastoma cancer. Apart from these two pathways, some cell cycle regulators such as Speedy/RINGO also contribute to neuroblastoma development. There is data reinforcing the possible communication of the components of ERK/MAPK and PI3K/AKT pathways in carcinogenic process. In addition to this, there are studies about the direct/indirect interaction of Speedy/RINGO with these pathways in different cell types other than neuroblastoma. However, there is not any study available showing the interaction of Speedy/RINGO with both pathways in neuroblastoma cells. Therefore, the aim of this study is to determine the possible effect of Speedy/RINGO on PI3K/AKT and ERK/MAPK pathways in SH-SY5Y neuroblastoma cells. For this aim, Speedy/RINGO was silenced by siRNA technique to analyze the effects of direct inhibition of Speedy/RINGO on these pathways. Results showed that Speedy/RINGO silencing caused a significant decrease in MEK1/2 expression and AKT phosphorylation. Afterward, MEK1/2 was inhibited using a specific inhibitor U0126. Data reveal a corresponding decrease in the Speedy/RINGO expression and AKT phosphorylation indicating a reciprocal interaction between ERK/MAPK and Speedy/RINGO. In addition, MTS analysis showed that both ERK/MAPK inhibition and Speedy/RINGO silencing significantly reduced the viability of SH-SY5Y cells. This study provides information about a possible interaction of Speedy/RINGO with PI3K/AKT and ERK/MAPK pathways in SH-SY5Y cells for the first time. It will not only help to better understand the cancer-prone interactions of these pathways but also enable us to identify the appropriate molecular targets for developing efficient treatment strategies.
Collapse
|
2
|
Hua Z, Zhan Y, Zhang S, Dong Y, Jiang M, Tan F, Liu Z, Thiele CJ, Li Z. P53/PUMA are potential targets that mediate the protection of brain-derived neurotrophic factor (BDNF)/TrkB from etoposide-induced cell death in neuroblastoma (NB). Apoptosis 2019; 23:408-419. [PMID: 29959561 DOI: 10.1007/s10495-018-1467-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The over-expressions of brain-derived neurotrophic factor (BDNF) and its tyrosine kinase receptor TrkB have been reported to induce chemo-resistance in neuroblastoma (NB) cells. In this study, we investigated the roles of P53 and BCL2 family members in the protection of BDNF/TrkB from etoposide-induced NB cell death. TB3 and TB8, two tetracycline (TET)-regulated TrkB-expressing NB cell lines, were utilized. The expressions of P53 and BCL2 family members were detected by Western blot or RT-PCR. Transfection of siRNAs was used to knockdown P53 or PUMA. Activated lentiviral was used to over-express PUMA. Cell survival was performed by MTS assay, and the percentage of cell confluence was measured by IncuCyte ZOOM. Our results showed that etoposide treatment induced significant and time-dependent increase of P53, which could be blocked by pre-treatment with BDNF, and knockdown P53 by transfecting siRNA attenuated etoposide-induced TrkB-expressing NB cell death. PUMA was the most significantly changed BCL2 family member after treatment with etoposide, and pre-treatment with BDNF blocked the increased expression of PUMA. Transfection with siRNA inhibited etoposide-induced increased expression of PUMA, and attenuated etoposide-induced NB cell death. We also found that over-expression of PUMA by infection of activated lentiviral induced TrkB-expressing NB cell death in the absence of etoposide, and treatment of BDNF protected NB cells from PUMA-induced cell death. Our results suggested that P53 and PUMA may be potential targets that mediated the protection of BDNF/TrkB from etoposide-induced NB cell death.
Collapse
Affiliation(s)
- Zhongyan Hua
- Medical Research Center, Shengjing Hospital of China Medical University, #36 Sanhao Street, Heping District, Shenyang, 110004, China
| | - Yue Zhan
- Medical Research Center, Shengjing Hospital of China Medical University, #36 Sanhao Street, Heping District, Shenyang, 110004, China
| | - Simeng Zhang
- Medical Research Center, Shengjing Hospital of China Medical University, #36 Sanhao Street, Heping District, Shenyang, 110004, China
| | - Yudi Dong
- Medical Research Center, Shengjing Hospital of China Medical University, #36 Sanhao Street, Heping District, Shenyang, 110004, China
| | - Min Jiang
- Medical Research Center, Shengjing Hospital of China Medical University, #36 Sanhao Street, Heping District, Shenyang, 110004, China
| | - Fei Tan
- Department of Neurology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Zhihui Liu
- Cellular & Molecular Biology Section, Pediatric Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Carol J Thiele
- Cellular & Molecular Biology Section, Pediatric Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Zhijie Li
- Medical Research Center, Shengjing Hospital of China Medical University, #36 Sanhao Street, Heping District, Shenyang, 110004, China.
| |
Collapse
|
3
|
Nakagawara A, Li Y, Izumi H, Muramori K, Inada H, Nishi M. Neuroblastoma. Jpn J Clin Oncol 2018; 48:214-241. [PMID: 29378002 DOI: 10.1093/jjco/hyx176] [Citation(s) in RCA: 118] [Impact Index Per Article: 19.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2017] [Indexed: 02/07/2023] Open
Abstract
Neuroblastoma is one of the most common solid tumors in children and has a diverse clinical behavior that largely depends on the tumor biology. Neuroblastoma exhibits unique features, such as early age of onset, high frequency of metastatic disease at diagnosis in patients over 1 year of age and the tendency for spontaneous regression of tumors in infants. The high-risk tumors frequently have amplification of the MYCN oncogene as well as segmental chromosome alterations with poor survival. Recent advanced genomic sequencing technology has revealed that mutation of ALK, which is present in ~10% of primary tumors, often causes familial neuroblastoma with germline mutation. However, the frequency of gene mutations is relatively small and other aberrations, such as epigenetic abnormalities, have also been proposed. The risk-stratified therapy was introduced by the Japan Neuroblastoma Study Group (JNBSG), which is now moving to the Neuroblastoma Committee of Japan Children's Cancer Group (JCCG). Several clinical studies have facilitated the reduction of therapy for children with low-risk neuroblastoma disease and the significant improvement of cure rates for patients with intermediate-risk as well as high-risk disease. Therapy for patients with high-risk disease includes intensive induction chemotherapy and myeloablative chemotherapy, followed by the treatment of minimal residual disease using differentiation therapy and immunotherapy. The JCCG aims for better cures and long-term quality of life for children with cancer by facilitating new approaches targeting novel driver proteins, genetic pathways and the tumor microenvironment.
Collapse
Affiliation(s)
| | - Yuanyuan Li
- Laboratory of Molecular Biology, Life Science Research Institute, Saga Medical Center Koseikan
| | - Hideki Izumi
- Laboratory of Molecular Biology, Life Science Research Institute, Saga Medical Center Koseikan
| | | | - Hiroko Inada
- Department of Pediatrics, Saga Medical Center Koseikan
| | - Masanori Nishi
- Department of Pediatrics, Saga University, Saga 849-8501, Japan
| |
Collapse
|
4
|
Tajbakhsh A, Mokhtari-Zaer A, Rezaee M, Afzaljavan F, Rivandi M, Hassanian SM, Ferns GA, Pasdar A, Avan A. Therapeutic Potentials of BDNF/TrkB in Breast Cancer; Current Status and Perspectives. J Cell Biochem 2017; 118:2502-2515. [PMID: 28230291 DOI: 10.1002/jcb.25943] [Citation(s) in RCA: 64] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2017] [Accepted: 02/21/2017] [Indexed: 12/14/2022]
Abstract
Brain-derived neurotrophic factor (BDNF) is a potent neurotrophic factor that has been shown to stimulate breast cancer cell growth and metastasis via tyrosine kinase receptors TrkA, TrkB, and the p75NTR death receptor. The aberrant activation of BDNF/TrkB pathways can modulate several signaling pathways, including Akt/PI3K, Jak/STAT, NF-kB, UPAR/UPA, Wnt/β-catenin, and VEGF pathways as well as the ER receptor. Several microRNAs have been identified that are involved in the modulation of BDNF/TrkB pathways. These include miR-206, miR-204, MiR-200a/c, MiR-210, MiR-134, and MiR-191; and these may be of value as prognostic and predictive biomarkers for detecting patients at high risk of developing breast cancer. It has been also been demonstrated that a high expression of genes involved in the BDNF pathway in breast cancer is associated with poor clinical outcome and reduced survival of patients. Several approaches have been developed for targeting this pathway, for example TKr inhibitors (AZD6918, CEP-701) and RNA interference. The aim of the current review was to provide an overview of the role of BDNF/TrkB pathways in the pathogenesis of breast cancer and its value as a potential therapeutic target. J. Cell. Biochem. 118: 2502-2515, 2017. © 2017 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Amir Tajbakhsh
- Department of Modern Sciences and Technologies, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.,Student Research Committee, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Amin Mokhtari-Zaer
- Student Research Committee, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.,Neurogenic Inflammation Research Centre and Department of Physiology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mehdi Rezaee
- Student Research Committee, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.,Department of Medical Biotechnology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Fahimeh Afzaljavan
- Department of Modern Sciences and Technologies, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.,Student Research Committee, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mehdi Rivandi
- Department of Modern Sciences and Technologies, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.,Student Research Committee, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Seyed Mahdi Hassanian
- Department of Medical Biochemistry, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.,Metabolic Syndrome Research Center, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Gordon A Ferns
- Brighton & Sussex Medical School, Division of Medical Education, Falmer, Brighton, UK
| | - Alireza Pasdar
- Department of Modern Sciences and Technologies, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.,Division of Applied Medicine, Medical School, University of Aberdeen, Foresterhill, Aberdeen, UK.,Medical Genetics Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Amir Avan
- Metabolic Syndrome Research Center, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
5
|
Kushner BH, Cheung NKV, Modak S, Becher OJ, Basu EM, Roberts SS, Kramer K, Dunkel IJ. A phase I/Ib trial targeting the Pi3k/Akt pathway using perifosine: Long-term progression-free survival of patients with resistant neuroblastoma. Int J Cancer 2016; 140:480-484. [PMID: 27649927 DOI: 10.1002/ijc.30440] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2016] [Revised: 08/23/2016] [Accepted: 09/07/2016] [Indexed: 11/08/2022]
Abstract
AKT plays a pivotal role in driving the malignant phenotype of many cancers, including high-risk neuroblastoma (HR-NB). AKT signaling, however, is active in normal tissues, raising concern about excessive toxicity from its suppression. The oral AKT inhibitor perifosine showed tolerable toxicity in adults and in our phase I trial in children with solid tumors (clinicaltrials.gov NCT00776867). We now report on the HR-NB experience. HR-NB patients received perifosine 50-75 mg m-2 day-1 after a loading dose of 100-200 mg m-2 on day 1, and continued on study until progressive disease. The 27 HR-NB patients included three treated for primary refractory disease and 24 with disease resistant to salvage therapy after 1-5 (median 2) relapses; only one had MYCN-amplified HR-NB. Pharmacokinetic studies showed μM concentrations consistent with cytotoxic levels in preclinical models. Nine patients (all MYCN-non-amplified) remained progression-free through 43+ to 74+ (median 54+) months from study entry, including the sole patient to show a complete response and eight patients who had persistence of abnormal 123 I-metaiodobenzylguanidine skeletal uptake but never developed progressive disease. Toxicity was negligible in all 27 patients, even with the prolonged treatment (11-62 months, median 38) in the nine long-term progression-free survivors. The clinical findings (i) confirm the safety of therapeutic serum levels of an AKT inhibitor in children; (ii) support perifosine for MYCN-non-amplified HR-NB as monotherapy after completion of standard treatment or combined with other agents (based on preclinical studies) to maximize antitumor effects; and (iii) highlight the welcome possibility that refractory or relapsed MYCN-non-amplified HR-NB is potentially curable.
Collapse
Affiliation(s)
- Brian H Kushner
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Nai-Kong V Cheung
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Shakeel Modak
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Oren J Becher
- Department of Pediatrics, Duke University Medical Center, Durham, NC
| | - Ellen M Basu
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Stephen S Roberts
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Kim Kramer
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Ira J Dunkel
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, NY
| |
Collapse
|
6
|
Jiang Y, Li L, Ma J, Zhang L, Niu F, Feng T, Li C. Auricular vagus nerve stimulation promotes functional recovery and enhances the post-ischemic angiogenic response in an ischemia/reperfusion rat model. Neurochem Int 2016; 97:73-82. [DOI: 10.1016/j.neuint.2016.02.009] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2015] [Revised: 02/12/2016] [Accepted: 02/17/2016] [Indexed: 12/22/2022]
|
7
|
Togar B, Turkez H, Hacimuftuoglu A, Tatar A, Geyikoglu F. Guaiazulene biochemical activity and cytotoxic and genotoxic effects on rat neuron and N2a neuroblastom cells. JOURNAL OF COMPLEMENTARY MEDICINE RESEARCH 2015; 4:29-33. [PMID: 26401381 PMCID: PMC4566767 DOI: 10.5455/jice.20141124062203] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2014] [Accepted: 11/24/2014] [Indexed: 12/11/2022]
Abstract
AIM Neuroblastoma (NB)cells are often used in cancer researches such as glioblastoma cells since they have the potential of high mitotic activity, nuclear pleomorphism, and tumor necrosis. Guaiazulene (GYZ 1,4-dimethyl-7-isopropylazulene)is present in several essential oils of medicinal and aromatic plants. Many studies have reported the cytotoxic effect of GYZ; however, there are no studies that compare such effects between cancer cell lines and normal human cells after treatment with GYZ. MATERIALS AND METHODS In this study, we aimed to describe in vitro antiproliferative and/or cytotoxic properties (by 3-[4,5 dimetylthiazol -2-yl]-2,5 diphenlytetrazolium bromide [MTT] test), oxidative effects (by total antioxidant capacity [TAC] and total oxidative stress [TOS] analysis)and genotoxic damage potentials (by single cell gel electrophoresis)of GYZ. RESULT The results indicated that GYZ have anti-proliferative activity suppressing the proliferation of neuron and N2a-NB cells at high doses. In addition, GYZ treatments at higher doses led to decreases of TAC levels and increases of TOS levels in neuron and N2a-NB cells. On the other hand, the mean values of the total scores of cells showing DNA damage were not found different from the control values. CONCLUSION From this study, it is observed that GYZ has in vitro cytotoxic activity against neuron and N2a-NB cells.
Collapse
Affiliation(s)
- Basak Togar
- Department of Biology, Faculty of Science, Ataturk University, Erzurum, Turkey
| | - Hasan Turkez
- Department of Molecular Biology and Genetics, Faculty of Sciences, Erzurum Technical University, Erzurum, Turkey
| | - Ahmet Hacimuftuoglu
- Department of Medical Pharmacology, Faculty of Medicine, Ataturk University, Erzurum, Turkey
| | - Abdulgani Tatar
- Department of Medical Genetics, Faculty of Medicine, Ataturk University, Erzurum, Turkey
| | - Fatime Geyikoglu
- Department of Biology, Faculty of Science, Ataturk University, Erzurum, Turkey
| |
Collapse
|
8
|
Westhoff MA, Karpel-Massler G, Brühl O, Enzenmüller S, La Ferla-Brühl K, Siegelin MD, Nonnenmacher L, Debatin KM. A critical evaluation of PI3K inhibition in Glioblastoma and Neuroblastoma therapy. MOLECULAR AND CELLULAR THERAPIES 2014; 2:32. [PMID: 26056598 PMCID: PMC4452069 DOI: 10.1186/2052-8426-2-32] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/28/2014] [Accepted: 10/16/2014] [Indexed: 12/11/2022]
Abstract
Members of the PI3K/Akt/mTor signaling cascade are among the most frequently altered proteins in cancer, yet the therapeutic application of pharmacological inhibitors of this signaling network, either as monotherapy or in combination therapy (CT) has so far not been particularly successful. In this review we will focus on the role of PI3K/Akt/mTOR in two distinct tumors, Glioblastoma multiforme (GBM), an adult brain tumor which frequently exhibits PTEN inactivation, and Neuroblastoma (NB), a childhood malignancy that affects the central nervous system and does not harbor any classic alterations in PI3K/Akt signaling. We will argue that inhibitors of PI3K/Akt signaling can be components for potentially promising new CTs in both tumor entities, but further understanding of the signal cascade's complexity is essential for successful implementation of these CTs. Importantly, failure to do this might lead to severe adverse effects, such as treatment failure and enhanced therapy resistance.
Collapse
Affiliation(s)
- Mike-Andrew Westhoff
- Department of Pediatrics and Adolescent Medicine, University Medical Center Ulm, Ulm, Germany
| | - Georg Karpel-Massler
- Department of Neurosurgery, University Medical Center Ulm, Ulm, Germany ; Department of Pathology and Cell Biology, Columbia University Medical Center, New York, NY USA
| | - Oliver Brühl
- Laboratorio Analisi Sicilia Catania, Lentini, SR Italy
| | - Stefanie Enzenmüller
- Department of Pediatrics and Adolescent Medicine, University Medical Center Ulm, Ulm, Germany
| | | | - Markus D Siegelin
- Department of Pathology and Cell Biology, Columbia University Medical Center, New York, NY USA
| | - Lisa Nonnenmacher
- Department of Pediatrics and Adolescent Medicine, University Medical Center Ulm, Ulm, Germany
| | - Klaus-Michael Debatin
- Department of Pediatrics and Adolescent Medicine, University Medical Center Ulm, Ulm, Germany
| |
Collapse
|
9
|
Westhoff MA, Karpel-Massler G, Brühl O, Enzenmüller S, La Ferla-Brühl K, Siegelin MD, Nonnenmacher L, Debatin KM. A critical evaluation of PI3K inhibition in Glioblastoma and Neuroblastoma therapy. MOLECULAR AND CELLULAR THERAPIES 2014; 2:32. [PMID: 26056598 PMCID: PMC4452069] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Subscribe] [Scholar Register] [Received: 07/28/2014] [Accepted: 10/16/2014] [Indexed: 11/21/2023]
Abstract
Members of the PI3K/Akt/mTor signaling cascade are among the most frequently altered proteins in cancer, yet the therapeutic application of pharmacological inhibitors of this signaling network, either as monotherapy or in combination therapy (CT) has so far not been particularly successful. In this review we will focus on the role of PI3K/Akt/mTOR in two distinct tumors, Glioblastoma multiforme (GBM), an adult brain tumor which frequently exhibits PTEN inactivation, and Neuroblastoma (NB), a childhood malignancy that affects the central nervous system and does not harbor any classic alterations in PI3K/Akt signaling. We will argue that inhibitors of PI3K/Akt signaling can be components for potentially promising new CTs in both tumor entities, but further understanding of the signal cascade's complexity is essential for successful implementation of these CTs. Importantly, failure to do this might lead to severe adverse effects, such as treatment failure and enhanced therapy resistance.
Collapse
Affiliation(s)
- Mike-Andrew Westhoff
- />Department of Pediatrics and Adolescent Medicine, University Medical Center Ulm, Ulm, Germany
| | - Georg Karpel-Massler
- />Department of Neurosurgery, University Medical Center Ulm, Ulm, Germany
- />Department of Pathology and Cell Biology, Columbia University Medical Center, New York, NY USA
| | - Oliver Brühl
- />Laboratorio Analisi Sicilia Catania, Lentini, SR Italy
| | - Stefanie Enzenmüller
- />Department of Pediatrics and Adolescent Medicine, University Medical Center Ulm, Ulm, Germany
| | | | - Markus D Siegelin
- />Department of Pathology and Cell Biology, Columbia University Medical Center, New York, NY USA
| | - Lisa Nonnenmacher
- />Department of Pediatrics and Adolescent Medicine, University Medical Center Ulm, Ulm, Germany
| | - Klaus-Michael Debatin
- />Department of Pediatrics and Adolescent Medicine, University Medical Center Ulm, Ulm, Germany
| |
Collapse
|
10
|
Williams AJ, Umemori H. The best-laid plans go oft awry: synaptogenic growth factor signaling in neuropsychiatric disease. Front Synaptic Neurosci 2014; 6:4. [PMID: 24672476 PMCID: PMC3957327 DOI: 10.3389/fnsyn.2014.00004] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2013] [Accepted: 02/21/2014] [Indexed: 12/27/2022] Open
Abstract
Growth factors play important roles in synapse formation. Mouse models of neuropsychiatric diseases suggest that defects in synaptogenic growth factors, their receptors, and signaling pathways can lead to disordered neural development and various behavioral phenotypes, including anxiety, memory problems, and social deficits. Genetic association studies in humans have found evidence for similar relationships between growth factor signaling pathways and neuropsychiatric phenotypes. Accumulating data suggest that dysfunction in neuronal circuitry, caused by defects in growth factor-mediated synapse formation, contributes to the susceptibility to multiple neuropsychiatric diseases, including epilepsy, autism, and disorders of thought and mood (e.g., schizophrenia and bipolar disorder, respectively). In this review, we will focus on how specific synaptogenic growth factors and their downstream signaling pathways might be involved in the development of neuropsychiatric diseases.
Collapse
Affiliation(s)
- Aislinn J Williams
- Department of Psychiatry, University of Michigan Ann Arbor, MI, USA ; Molecular and Behavioral Neuroscience Institute, University of Michigan Ann Arbor, MI, USA
| | - Hisashi Umemori
- Molecular and Behavioral Neuroscience Institute, University of Michigan Ann Arbor, MI, USA ; Department of Neurology, F.M. Kirby Neurobiology Center, Harvard Medical School, Boston Children's Hospital Boston, MA, USA
| |
Collapse
|
11
|
Westhoff MA, Faham N, Marx D, Nonnenmacher L, Jennewein C, Enzenmüller S, Gonzalez P, Fulda S, Debatin KM. Sequential dosing in chemosensitization: targeting the PI3K/Akt/mTOR pathway in neuroblastoma. PLoS One 2013; 8:e83128. [PMID: 24391739 PMCID: PMC3877010 DOI: 10.1371/journal.pone.0083128] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2013] [Accepted: 10/31/2013] [Indexed: 01/14/2023] Open
Abstract
Breaking resistance to chemotherapy is a major goal of combination therapy in many tumors, including advanced neuroblastoma. We recently demonstrated that increased activity of the PI3K/Akt network is associated with poor prognosis, thus providing an ideal target for chemosensitization. Here we show that targeted therapy using the PI3K/mTOR inhibitor NVP-BEZ235 significantly enhances doxorubicin-induced apoptosis in neuroblastoma cells. Importantly, this increase in apoptosis was dependent on scheduling: while pretreatment with the inhibitor reduced doxorubicin-induced apoptosis, the sensitizing effect in co-treatment could further be increased by delayed addition of the inhibitor post chemotherapy. Desensitization for doxorubicin-induced apoptosis seemed to be mediated by a combination of cell cycle-arrest and autophagy induction, whereas sensitization was found to occur at the level of mitochondria within one hour of NVP-BEZ235 posttreatment, leading to a rapid loss of mitochondrial membrane potential with subsequent cytochrome c release and caspase-3 activation. Within the relevant time span we observed marked alterations in a ∼30 kDa protein associated with mitochondrial proteins and identified it as VDAC1/Porin protein, an integral part of the mitochondrial permeability transition pore complex. VDAC1 is negatively regulated by the PI3K/Akt pathway via GSK3β and inhibition of GSK3β, which is activated when Akt is blocked, ablated the sensitizing effect of NVP-BEZ235 posttreatment. Our findings show that cancer cells can be sensitized for chemotherapy induced cell death – at least in part – by NVP-BEZ235-mediated modulation of VDAC1. More generally, we show data that suggest that sequential dosing, in particular when multiple inhibitors of a single pathway are used in the optimal sequence, has important implications for the general design of combination therapies involving molecular targeted approaches towards the PI3K/Akt/mTOR signaling network.
Collapse
Affiliation(s)
- Mike-Andrew Westhoff
- Department of Pediatrics and Adolescent Medicine, University Medical Center Ulm, Ulm, Germany
| | - Najmeh Faham
- Department of Pediatrics and Adolescent Medicine, University Medical Center Ulm, Ulm, Germany
| | - Daniela Marx
- Department of Pediatrics and Adolescent Medicine, University Medical Center Ulm, Ulm, Germany
| | - Lisa Nonnenmacher
- Department of Pediatrics and Adolescent Medicine, University Medical Center Ulm, Ulm, Germany
| | - Claudia Jennewein
- Department of Pediatrics and Adolescent Medicine, University Medical Center Ulm, Ulm, Germany
| | - Stefanie Enzenmüller
- Department of Pediatrics and Adolescent Medicine, University Medical Center Ulm, Ulm, Germany
| | - Patrick Gonzalez
- Department of Pediatrics and Adolescent Medicine, University Medical Center Ulm, Ulm, Germany
| | - Simone Fulda
- Department of Pediatrics and Adolescent Medicine, University Medical Center Ulm, Ulm, Germany
- * E-mail: (SF); (KMD)
| | - Klaus-Michael Debatin
- Department of Pediatrics and Adolescent Medicine, University Medical Center Ulm, Ulm, Germany
- * E-mail: (SF); (KMD)
| |
Collapse
|
12
|
Ho WL, Che MI, Chou CH, Chang HH, Jeng YM, Hsu WM, Lin KH, Huang MC. B3GNT3 expression suppresses cell migration and invasion and predicts favorable outcomes in neuroblastoma. Cancer Sci 2013; 104:1600-8. [PMID: 24118321 DOI: 10.1111/cas.12294] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2013] [Revised: 09/05/2013] [Accepted: 09/08/2013] [Indexed: 12/19/2022] Open
Abstract
Aberrant expression of the simple mucin-type carbohydrate antigens such as T, Tn, sialyl-T and sialyl-Tn is associated with poor prognosis in several cancers. β1,3-N-acetylglucosaminyltransferase-3 (B3GNT3), a member of the β3GlcNAcT family, is responsible for forming extended core 1 (T antigen) oligosaccharides. The role of B3GNT3, which is expressed in various tissues including human fetal brain, in regulating neuroblastoma (NB) formation and cell behaviors remains unclear. Here, we showed that increased B3GNT3 expression evaluated using immunohistochemistry in NB tumor tissues correlated well with the histological grade of differentiation as well as a favorable Shimada's subset of pathology. Univariate and multivariate analyses revealed that positive B3GNT3 expression in tumor tissues predicted a favorable prognosis in NB patients independent of other prognostic markers. B3GNT3 overexpression suppresses T antigen formation and malignant phenotypes including migration and invasion of SK-N-SH cells, whereas B3GNT3 knockdown enhances these phenotypes of SK-N-SH cells. Moreover, B3GNT3 expression decreased phosphorylation of focal adhesion kinase (FAK), Src, paxillin, Akt and ERK1/2. We conclude that B3GNT3 predicts a favorable cancer behavior of NB and suppresses malignant phenotypes by modulating mucin-type O-glycosylation and signaling in NB cells.
Collapse
Affiliation(s)
- Wan-ling Ho
- Department of Pediatrics, National Taiwan University Hospital and College of Medicine, National Taiwan University, Taipei, Taiwan; Graduate Institute of Clinical Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan; Department of Pediatrics, Shin Kong Wu Ho-Su Memorial Hospital, Taipei, Taiwan; School of Medicine, Fu Jen Catholic University, New Taipei City, Taiwan
| | | | | | | | | | | | | | | |
Collapse
|
13
|
Yang X, Martin TA, Jiang WG. Biological influence of brain-derived neurotrophic factor (BDNF) on colon cancer cells. Exp Ther Med 2013; 6:1475-1481. [PMID: 24255678 PMCID: PMC3829751 DOI: 10.3892/etm.2013.1330] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2013] [Accepted: 08/23/2013] [Indexed: 12/13/2022] Open
Abstract
Brain-derived neurotrophic factor (BDNF) has been observed to be elevated in solid tumors including colorectal cancer. The present study aimed to investigate the effect of modulation of BDNF at the transcription level on the cellular function of colorectal cells and to increase our understanding of its biological role in human colon cancer. An investigation of a cohort of human colorectal tissues (tumor n=66; normal n=88) using quantitative PCR and immunohistochemistry demonstrated that BDNF is aberrantly expressed in human colon cancer and a significantly raised level of BDNF is associated with its stage at diagnosis. The expression profile of BDNF in human colon cancer cell lines was evaluated using RT-PCR. A set of anti-BDNF ribozymes were used to transfect colon cancer cells in order to generate BDNF knockdown cells to evaluate the effect on growth and apoptosis. BDNF gene transcripts were successfully detected in the colon cancer cell lines, Caco-2 and HRT18. BDNF knockdown in Caco-2 and HRT18 cell lines resulted in decreased rates of growth and proliferation. Analysis of apoptosis showed that cell apoptosis was increased. It is concluded that BDNF, a neurotrophic growth factor aberrantly expressed in cancers such as colon cancer, has a profound impact on the cellular behavior of colon cancer cells and that BDNF is associated with a reduction in the apoptosis of colon cancer. BDNF is therefore a potential therapeutic target in colon cancer and its effect in human colon cancer requires further investigation.
Collapse
Affiliation(s)
- Xiaomei Yang
- Metastasis and Angiogenesis Research Group, Cardiff University School of Medicine, Cardiff CF14 4XN, UK ; Cardiff University-Capital Medical University Joint Centre for Biomedical Research, School of Basic Medical Science, Capital Medical University, Beijing 100069, P.R. China ; Department of Biochemistry and Molecular Biology, Capital Medical University, Beijing 100069, P.R. China
| | | | | |
Collapse
|
14
|
Leitão A, Schramm A, Eggert A. Discovery of a new bioactive molecule for neuroblastoma. Chem Biol Drug Des 2013; 82:233-41. [PMID: 23601248 DOI: 10.1111/cbdd.12148] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2012] [Revised: 01/14/2013] [Accepted: 04/02/2013] [Indexed: 12/30/2022]
Abstract
Neuroblastoma, a common pediatric malignancy of neural crest origin, is unique in its wide spectrum of clinical and biological behavior, ranging from spontaneous regression or differentiation to rapid progression and metastasis. Overexpression of neurotrophin receptors of the tyrosine kinase (Trk) family has been identified as a major prognostic and biological factor for this disease. Novel molecules were selected using cheminformatics tools (structure-based virtual screening and ligand-based virtual screening) and screened in cell-based assays to modulate Trk receptor activity. One compound (C390-0031) had a potent antiproliferative activity in dose-response studies using neuroblastoma cell lines. The molecular effects of this molecule were further characterized by using cell cycle and Western blot analysis. Interestingly, despite the presence of the anchoring fragment to the Trk kinase domain composing its structure, this molecule does not inhibit TrkA like lestaurtinib, constituting a new chemical with a yet unknown mechanism of action.
Collapse
Affiliation(s)
- Andrei Leitão
- OncoLab - Hematological-Oncological Laboratory, University Children's Hospital, University of Duisburg-Essen, Essen, Germany.
| | | | | |
Collapse
|
15
|
Biological influence of brain-derived neurotrophic factor on breast cancer cells. Int J Oncol 2012; 41:1541-6. [PMID: 22895657 DOI: 10.3892/ijo.2012.1581] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2012] [Accepted: 06/15/2012] [Indexed: 01/29/2023] Open
Abstract
Brain-derived neurotrophic factor (BDNF) is a member of the neurotrophin superfamily which has been indicated in the pathophysiology of the nervous system and is important in a number of neurological and psychological conditions. Recently, BDNF was also shown to play a role in the development and progression of solid tumour myeloma. It has been reported that BDNF is aberrantly expressed in human breast cancer and that a raised level of BDNF is associated with poor clinical outcome and reduced survival. The present study investigated the role of BDNF in human breast cancer. A panel of human breast cancer cells was used and the expression profile of BDNF was evaluated using RT-PCR. We constructed a set of anti-BDNF transgenes which were used to transfect breast cancer cells in order to generate BDNF knocked down cells. The impact of BDNF knockdown on growth and apoptosis was evaluated. Statistical analysis was performed using SPSS. P<0.05 was considered statistically significant. BDNF gene transcripts were successfully detected in the breast cancer cell lines MCF-7, MDA-MB-231 and ZR75-1 MDA-MB-231 and MCF-7 wild-type cells were subject to transfection of anti-BDNF transgenes, followed by the establishment of BDNF knocked down sublines. Knockdown of BDNF in MDA-MB-231 and MCF-7 cell lines resulted in decreased rates of growth and proliferation. Analysis of apoptosis showed that cell apoptosis was increased in cells stably transfected with ribozymes for BDNF compared with the vector control. It is concluded that BDNF, a neurotrophic growth factor aberrantly expressed in cancers such as breast cancer, has a profound impact on the cellular behaviour of breast cancer cells and that BDNF is associated with a reduction of the apoptosis of breast cancer. BDNF is, therefore, a potential therapeutic target in breast cancer and its effect in human breast cancer requires further investigation.
Collapse
|
16
|
Ranjan S, Sood R, Dudas J, Glueckert R, Schrott-Fischer A, Roy S, Pyykkö I, Kinnunen PKJ. Peptide-mediated targeting of liposomes to TrkB receptor-expressing cells. Int J Nanomedicine 2012. [PMID: 22848172 DOI: 10.2147/ijn.s32367ijn-7-3475] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND The neurotrophic receptor tyrosine kinase B (TrkB) has diverse signaling roles in neurons and tumor cells. Accordingly, its suppressive targeting is of interest in neuroblastoma and other tumors, whereas its role in improving survival is focused in neurons. Here we describe targeting of TrkB-binding peptide-conjugated liposomes (PCL) to the TrkB-expressing mouse macrophage-like cell line RAW264, and to all-trans-retinoic acid-treated neuron-like TrkB⁺ SH-SY5Y human neuroblastoma cells. METHODS Binding and internalization of PCL was monitored by flow cytometry and confocal fluorescence microscopy. RESULTS Internalization of TrkB-targeted PCL by RAW264 cells was enhanced and faster when compared with PCL having the corresponding scrambled peptide. Likewise, binding and augmented uptake were confirmed for TrkB⁺ SH-SY5Y cells, with targeted PCL appearing in the cytoplasm after 20 minutes of incubation. CONCLUSION We demonstrate here the feasibility of targeting liposomes to TrkB-expressing cells by 18-mer peptides, promoting cellular uptake (at least partly into endosomes) via receptor-mediated pathways.
Collapse
Affiliation(s)
- Sanjeev Ranjan
- Helsinki Biophysics and Biomembrane Group, Department of Biomedical Engineering and Computational Science, Aalto University, Espoo, Finland
| | | | | | | | | | | | | | | |
Collapse
|
17
|
Ranjan S, Sood R, Dudas J, Glueckert R, Schrott-Fischer A, Roy S, Pyykkö I, Kinnunen PKJ. Peptide-mediated targeting of liposomes to TrkB receptor-expressing cells. Int J Nanomedicine 2012; 7:3475-85. [PMID: 22848172 PMCID: PMC3405886 DOI: 10.2147/ijn.s32367] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Background The neurotrophic receptor tyrosine kinase B (TrkB) has diverse signaling roles in neurons and tumor cells. Accordingly, its suppressive targeting is of interest in neuroblastoma and other tumors, whereas its role in improving survival is focused in neurons. Here we describe targeting of TrkB-binding peptide-conjugated liposomes (PCL) to the TrkB-expressing mouse macrophage-like cell line RAW264, and to all-trans-retinoic acid-treated neuron-like TrkB+ SH-SY5Y human neuroblastoma cells. Methods Binding and internalization of PCL was monitored by flow cytometry and confocal fluorescence microscopy. Results Internalization of TrkB-targeted PCL by RAW264 cells was enhanced and faster when compared with PCL having the corresponding scrambled peptide. Likewise, binding and augmented uptake were confirmed for TrkB+ SH-SY5Y cells, with targeted PCL appearing in the cytoplasm after 20 minutes of incubation. Conclusion We demonstrate here the feasibility of targeting liposomes to TrkB-expressing cells by 18-mer peptides, promoting cellular uptake (at least partly into endosomes) via receptor-mediated pathways.
Collapse
Affiliation(s)
- Sanjeev Ranjan
- Helsinki Biophysics and Biomembrane Group, Department of Biomedical Engineering and Computational Science, Aalto University, Espoo, Finland
| | | | | | | | | | | | | | | |
Collapse
|
18
|
Sun W, Modak S. Emerging treatment options for the treatment of neuroblastoma: potential role of perifosine. Onco Targets Ther 2012; 5:21-9. [PMID: 22419878 PMCID: PMC3299554 DOI: 10.2147/ott.s14578] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Achieving a cure for high-risk neuroblastoma, the most common extracranial solid tumor in children, remains a formidable task despite the recent addition of antibody-mediated anti-GD2 immunotherapy to established multimodality therapy. The PI3K/Akt pathway is a pivotal signaling pathway utilized by a plethora of receptor tyrosine kinases that contribute to the aggressive phenotype of high-risk neuroblastoma. Akt is aberrantly activated in high-risk neuroblastoma and is therefore an attractive therapeutic target. Perifosine is the best-characterized Akt inhibitor in preclinical studies and in clinical trials in adults, although safety in children is not yet confirmed. It is a synthetic third-generation alkylphospholipid with good oral bioavailability and modest side effects. Perifosine targets the lipid-binding PH domain of Akt and inhibits the translocation of Akt to the cell membrane, an essential step for Akt activation. It decreases Akt phosphorylation and increases caspase-dependent apoptosis in neuroblastoma cell lines, inhibits growth of neuroblastoma xenografts, and overcomes RTK/ligand-mediated chemoresistance. It is currently being studied in two Phase I clinical trials in children with recurrent or refractory solid tumors including neuroblastoma. In the single agent trial (ClinicalTrials.gov identifier NCT00776867), maximum tolerated dose has not yet been reached and pharmacokinetic data has been accrued. In the second study (ClinicalTrials.gov identifier NCT01049841), patients are treated with a combination of perifosine and the mTOR-inhibitor temsirolimus based on preclinical data showing synergy of the two agents, and the premise that direct Akt inhibition may overcome Akt activation secondary to mTOR inhibition. Based on results from adult trials, it is unlikely that perifosine alone will produce dramatic therapeutic effects against high-risk neuroblastoma. However, given the recent encouraging early-phase combination therapy results in adults with multiple myeloma and colorectal carcinoma, rational perifosine-containing combination regimens hold promise for neuroblastoma therapy. These will be explored after safety in children is established in Phase I studies.
Collapse
Affiliation(s)
- Weili Sun
- Department of Pediatrics, Keck School of Medicine, University of Southern California, Children's Hospital of Los Angeles, Los Angeles, CA, USA
| | | |
Collapse
|
19
|
Lee J, Jiffar T, Kupferman ME. A novel role for BDNF-TrkB in the regulation of chemotherapy resistance in head and neck squamous cell carcinoma. PLoS One 2012; 7:e30246. [PMID: 22276165 PMCID: PMC3262811 DOI: 10.1371/journal.pone.0030246] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2011] [Accepted: 12/15/2011] [Indexed: 02/06/2023] Open
Abstract
Mechanisms of resistance for HNSCC to cisplatin (CDDP), the foundational chemotherapeutic agent in the treatment of this disease, remain poorly understood. We previously demonstrated that cisplatin resistance (CR) can be overcome by targeting Trk receptor. In the current study, we explored the potential mechanistic role of the BDNF-TrkB signaling system in the development of CDDP resistance in HNSCC. Utilizing an in vitro system of acquired CR, we confirmed a substantial up-regulation of both BDNF and TrkB at the protein and mRNA levels in CR cells, suggesting an autocrine pathway dysregulation in this system. Exogenous BDNF stimulation led to an enhanced expression of the drug-resistance and anti-apoptotic proteins MDR1 and XiAP, respectively, in a dose-dependently manner, demonstrating a key role for BDNF-TrkB signaling in modulating the response to cytotoxic agents. In addition, modulation of TrkB expression induced an enhanced sensitivity of cells to CDDP in HNSCC. Moreover, genetic suppression of TrkB resulted in changes in expression of Bim, XiAP, and MDR1 contributing to HNSCC survival. To elucidate intracellular signaling pathways responsible for mechanisms underlying BDNF/TrkB induced CDDP-resistance, we analyzed expression levels of these molecules following inhibition of Akt. Inhibition of Akt eliminated BDNF effect on MDR1 and Bim expression in OSC-19P cells as well as modulated expressions of MDR1, Bim, and XiAP in OSC-19CR cells. These results suggest BDNF/TrkB system plays critical roles in CDDP-resistance development by utilizing Akt-dependent signaling pathways.
Collapse
MESH Headings
- ATP Binding Cassette Transporter, Subfamily B, Member 1/genetics
- ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism
- Antineoplastic Agents/pharmacology
- Apoptosis Regulatory Proteins/genetics
- Apoptosis Regulatory Proteins/metabolism
- Bcl-2-Like Protein 11
- Blotting, Western
- Brain-Derived Neurotrophic Factor/genetics
- Brain-Derived Neurotrophic Factor/metabolism
- Carcinoma, Squamous Cell/metabolism
- Cell Line, Tumor
- Cell Proliferation/drug effects
- Cisplatin/pharmacology
- Drug Resistance, Neoplasm/genetics
- Enzyme-Linked Immunosorbent Assay
- Head and Neck Neoplasms/metabolism
- Humans
- Immunohistochemistry
- Membrane Proteins/genetics
- Membrane Proteins/metabolism
- Proto-Oncogene Proteins/genetics
- Proto-Oncogene Proteins/metabolism
- Receptor, trkB/genetics
- Receptor, trkB/metabolism
- Reverse Transcriptase Polymerase Chain Reaction
- Squamous Cell Carcinoma of Head and Neck
- X-Linked Inhibitor of Apoptosis Protein/genetics
- X-Linked Inhibitor of Apoptosis Protein/metabolism
Collapse
Affiliation(s)
- Junegoo Lee
- Department of Head and Neck Surgery, MD Anderson Cancer Center, University of Texas, Houston, Texas, United States of America
| | - Tilahun Jiffar
- Department of Head and Neck Surgery, MD Anderson Cancer Center, University of Texas, Houston, Texas, United States of America
| | - Michael E. Kupferman
- Department of Head and Neck Surgery, MD Anderson Cancer Center, University of Texas, Houston, Texas, United States of America
- * E-mail:
| |
Collapse
|
20
|
Dokas LA, Malone AM, Williams FE, Nauli SM, Messer WS. Multiple protein kinases determine the phosphorylated state of the small heat shock protein, HSP27, in SH-SY5Y neuroblastoma cells. Neuropharmacology 2011; 61:12-24. [PMID: 21338617 PMCID: PMC3105189 DOI: 10.1016/j.neuropharm.2011.02.010] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2010] [Revised: 01/25/2011] [Accepted: 02/11/2011] [Indexed: 01/21/2023]
Abstract
In SH-SY5Y human neuroblastoma cells, the cholinergic agonist, carbachol, stimulates phosphorylation of the small heat shock protein 27 (HSP27). Carbachol increases phosphorylation of both Ser-82 and Ser-78 while the phorbol ester, phorbol-12, 13-dibutyrate (PDB) affects only Ser-82. Muscarinic receptor activation by carbachol was confirmed by sensitivity of Ser-82 phosphorylation to hyoscyamine with no effect of nicotine or bradykinin. This response to carbachol is partially reduced by inhibition of protein kinase C (PKC) with GF 109203X and p38 mitogen-activated protein kinase (MAPK) with SB 203580. In contrast, phosphorylation produced by PDB is completely reversed by GF 109203X or CID 755673, an inhibitor of PKD. Inhibition of phosphatidylinositol 3-kinase or Akt with LY 294002 or Akti-1/2 stimulates HSP27 phosphorylation while rapamycin, which inhibits mTORC1, does not. The stimulatory effect of Akti-1/2 is reversed by SB 203580 and correlates with increased p38 MAPK phosphorylation. SH-SY5Y cells differentiated with a low concentration of PDB and basic fibroblast growth factor to a more neuronal phenotype retain carbachol-, PDB- and Akti-1/2-responsive HSP27 phosphorylation. Immunofluorescence microscopy confirms increased HSP27 phosphorylation in response to carbachol or PDB. At cell margins, PDB causes f-actin to reorganize forming lamellipodial structures from which phospho-HSP27 is segregated. The resultant phenotypic change in cell morphology is dependent upon PKC, but not PKD, activity. The major conclusion from this study is that the phosphorylated state of HSP27 in SH-SY5Y cells results from integrated signaling involving PKC, p38 MAPK and Akt.
Collapse
Affiliation(s)
- Linda A. Dokas
- Department of Pharmacology, College of Pharmacy, 3000 Arlington Avenue, The University of Toledo, Toledo OH 43614 USA
| | - Amy M. Malone
- Department of Pharmacology, College of Pharmacy, 3000 Arlington Avenue, The University of Toledo, Toledo OH 43614 USA
| | - Frederick E. Williams
- Department of Pharmacology, College of Pharmacy, 3000 Arlington Avenue, The University of Toledo, Toledo OH 43614 USA
| | - Surya M. Nauli
- Department of Pharmacology, College of Pharmacy, 3000 Arlington Avenue, The University of Toledo, Toledo OH 43614 USA
- Department of Medicinal & Biological Chemistry, College of Pharmacy, 3000 Arlington Avenue, The University of Toledo, Toledo OH 43614 USA
| | - William S. Messer
- Department of Pharmacology, College of Pharmacy, 3000 Arlington Avenue, The University of Toledo, Toledo OH 43614 USA
- Department of Medicinal & Biological Chemistry, College of Pharmacy, 3000 Arlington Avenue, The University of Toledo, Toledo OH 43614 USA
| |
Collapse
|
21
|
Li J, Zhang Y, Zhao J, Kong F, Chen Y. Overexpression of miR-22 reverses paclitaxel-induced chemoresistance through activation of PTEN signaling in p53-mutated colon cancer cells. Mol Cell Biochem 2011; 357:31-8. [PMID: 21594648 DOI: 10.1007/s11010-011-0872-8] [Citation(s) in RCA: 73] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2011] [Accepted: 05/06/2011] [Indexed: 01/12/2023]
Abstract
Chemoresistance is a key cause of treatment failure in colon cancer. MiR-22 is a tumor-suppressing microRNA. To explore whether miR-22 is an important player in the development of chemoresistance in colon cancer, we overexpressed miR-22 and subsequently tested its role in cell proliferation, apoptosis, survival, and associated signaling in p53-mutated HT-29 and HCT-15 cells, and p53 wild-type HCT-116 cells. We further investigated the role of miR-22 on cytotoxicity of paclitaxel in both the p53-mutated and p53 wild-type colon cancer cells. Results showed that HT-29 and HCT-15 cells were resistant to paclitaxel-induced cytotoxicity, which normally inhibits cell proliferation and survival, and induces apoptosis. Conversely, HCT-116 was relatively sensitive to the cytotoxicity of paclitaxel. Overexpression of miR-22 significantly decreased cell proliferation and survival, and induced cell apoptosis in the p53-mutated colon cancer cells, but played no role in the p53 wild-type cells. Importantly, miR-22 overexpression enhanced the cytotoxic role of paclitaxel in p53-mutated HT-29 and HCT-15 cells, but not in p53 wild-type HCT-116 cell. We further demonstrated that the tumor-suppressive role of miR-22 in p53-mutated colon cancer cells was mediated by upregulating PTEN expression, which negatively regulated Akt phosphorylation at Ser(473) and MTDH expression, and subsequently increased Bax and active caspase-3 levels. Our study is the first to identify the tumor-suppressive role of miR-22 and its associated signaling in the p53-mutated colon cancer cells and highlighted the chemosensitive role of miR-22.
Collapse
Affiliation(s)
- Jian Li
- Hepatobiliary & Enteric Surgery Research Center, Central South University, Changsha 410008, China
| | | | | | | | | |
Collapse
|
22
|
Takei Y, Laskey R. Intracellular and Intercellular Cross Talk Between NGF and TNF. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2011; 691:559-65. [DOI: 10.1007/978-1-4419-6612-4_58] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
|
23
|
Stallings RL, Foley NH, Bryan K, Buckley PG, Bray I. Therapeutic targeting of miRNAs in neuroblastoma. Expert Opin Ther Targets 2010; 14:951-62. [PMID: 20658962 DOI: 10.1517/14728222.2010.510136] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
IMPORTANCE OF THE FIELD Neuroblastomas arise from precursor cells of the sympathetic nervous system and are noted for highly heterogeneous clinical behavior. These tumors currently account for approximately 15% of all childhood cancer related deaths in spite of intensive multimodal chemotherapy and are a major problem in pediatric oncology. The identification of novel therapeutic targets is urgently required to reduce patient morbidity. AREAS COVERED IN THIS REVIEW The purpose of this article is to review and synthesize all of the rapidly expanding evidence for the contribution of microRNAs (miRNAs) in neuroblastoma aggressive disease pathogenesis, along with the prospect of using small RNAs as therapeutics. WHAT THE READER WILL GAIN The reader will obtain insight on the miRNAs that are dysregulated in neuroblastoma along with potential therapeutic strategies and the most promising targets. TAKE HOME MESSAGE A number of miRNAs which are associated with aggressive disease pathogenesis in neuroblastoma patients have been demonstrated to contribute in major ways to cell proliferation rates, apoptosis, differentiation, invasiveness and tumor growth in vitro and in vivo. Directly or indirectly interfering with the function of these miRNAs may prove to be an important and novel form of therapy.
Collapse
|
24
|
Zhang S, Guo D, Luo W, Zhang Q, Zhang Y, Li C, Lu Y, Cui Z, Qiu X. TrkB is highly expressed in NSCLC and mediates BDNF-induced the activation of Pyk2 signaling and the invasion of A549 cells. BMC Cancer 2010; 10:43. [PMID: 20156366 PMCID: PMC2830183 DOI: 10.1186/1471-2407-10-43] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2009] [Accepted: 02/16/2010] [Indexed: 11/12/2022] Open
Abstract
Background Aberrant regulation in the invasion of cancer cells is closely associated with their metastatic potentials. TrkB functions as a receptor tyrosine kinase and is considered to facilitate tumor metastasis. Pyk2 is a non-receptor tyrosine kinase and integrates signals in cell invasion. However, little is known about the expression of TrkB in NSCLC and whether Pyk2 is involved in TrkB-mediated invasion of A549 cells. Methods The expression of TrkB was investigated in NSCLC by immunohistochemical staining. Both HBE and A549 cells were treated with BDNF. The expression of TrkB, Pyk2 and ERK phosphorylations were assessed by western blot. Besides, A549 cells were transfected with TrkB-siRNA or Pyk2-siRNA, or treated with ERK inhibitor where indicated. Transwell assay was performed to evaluate cell invasion. Results 40 cases (66.7%) of NSCLC were found higher expression of TrkB and patients with more TrkB expression had significant metastatic lymph nodes (p = 0.028). BDNF facilitated the invasion of A549 cells and the activations of Pyk2 in Tyr402 and ERK. However, the effects of BDNF were not observed in HBE cells with lower expression of TrkB. In addition, the increased Pyk2 and ERK activities induced by BDNF were significantly inhibited by blocking TrkB expression, so was the invasion of A549 cells. Knockdown studies revealed the essential role of Pyk2 for BDNF-induced cell invasion, since the invasion of A549 cells was abolished by Pyk2-siRNA. The application of ERK inhibitor also showed the suppressed ERK phosphorylation and cell invasion. Conclusion These data indicated that higher expression of TrkB in NSCLC was closely correlated with lymph node metastasis, and BDNF probably via TrkB/Pyk2/ERK promoted the invasion of A549 cells.
Collapse
Affiliation(s)
- Siyang Zhang
- Center of Laboratory Technology and Experimental Medicine, China Medical University, Shenyang, China.
| | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Macias M, Nowicka D, Czupryn A, Sulejczak D, Skup M, Skangiel-Kramska J, Czarkowska-Bauch J. Exercise-induced motor improvement after complete spinal cord transection and its relation to expression of brain-derived neurotrophic factor and presynaptic markers. BMC Neurosci 2009; 10:144. [PMID: 19961582 PMCID: PMC2802589 DOI: 10.1186/1471-2202-10-144] [Citation(s) in RCA: 58] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2009] [Accepted: 12/04/2009] [Indexed: 01/01/2023] Open
Abstract
BACKGROUND It has been postulated that exercise-induced activation of brain-derived neurotrophic factor (BDNF) may account for improvement of stepping ability in animals after complete spinal cord transection. As we have shown previously, treadmill locomotor exercise leads to up-regulation of BDNF protein and mRNA in the entire neuronal network of intact spinal cord. The questions arise: (i) how the treadmill locomotor training, supplemented with tail stimulation, affects the expression of molecular correlates of synaptic plasticity in spinal rats, and (ii) if a response is related to BDNF protein level and distribution. We investigated the effect of training in rats spinalized at low thoracic segments on the level and distribution of BDNF immunoreactivity (IR) in ventral quadrants of the lumbar segments, in conjunction with markers of presynaptic terminals, synaptophysin and synaptic zinc. RESULTS Training improved hindlimb stepping in spinal animals evaluated with modified Basso-Beattie-Bresnahan scale. Grades of spinal trained animals ranged between 5 and 11, whereas those of spinal were between 2 and 4. Functional improvement was associated with changes in presynaptic markers and BDNF distribution. Six weeks after transection, synaptophysin IR was reduced by 18% around the large neurons of lamina IX and training elevated its expression by over 30%. The level of synaptic zinc staining in the ventral horn was unaltered, whereas in ventral funiculi it was decreased by 26% postlesion and tended to normalize after the training. Overall BDNF IR levels in the ventral horn, which were higher by 22% postlesion, were unchanged after the training. However, training modified distribution of BDNF in the processes with its predominance in the longer and thicker ones. It also caused selective up-regulation of BDNF in two classes of cells (soma ranging between 100-400 microm2 and over 1000 microm2) of the ventrolateral and laterodorsal motor nuclei. CONCLUSION Our results show that it is not BDNF deficit that determines lack of functional improvement in spinal animals. They indicate selectivity of up-regulation of BDNF in distinct subpopulations of cells in the motor nuclei which leads to changes of innervation targeting motoneurons, tuned up by locomotor activity as indicated by a region-specific increase of presynaptic markers.
Collapse
Affiliation(s)
- Matylda Macias
- Department of Neurophysiology, Nencki Institute of Experimental Biology, (3 Pasteur Str), Warsaw (02-093), Poland
- Institute of Molecular and Cellular Biology, (4 Trojdena Str), Warsaw, (02-109), Poland
| | - Dorota Nowicka
- Department of Molecular Neurobiology, Nencki Institute of Experimental Biology, (3 Pasteur Str), Warsaw (02-093), Poland
| | - Artur Czupryn
- Department of Molecular Neurobiology, Nencki Institute of Experimental Biology, (3 Pasteur Str), Warsaw (02-093), Poland
| | - Dorota Sulejczak
- Department of Neurophysiology, Nencki Institute of Experimental Biology, (3 Pasteur Str), Warsaw (02-093), Poland
- Department of Experimental Pharmacology, Medical, Research Centre, Polish Academy of Sciences, (5 Pawińskiego Str), Warsaw, (02-106), Poland
| | - Małgorzata Skup
- Department of Neurophysiology, Nencki Institute of Experimental Biology, (3 Pasteur Str), Warsaw (02-093), Poland
| | - Jolanta Skangiel-Kramska
- Department of Molecular Neurobiology, Nencki Institute of Experimental Biology, (3 Pasteur Str), Warsaw (02-093), Poland
| | - Julita Czarkowska-Bauch
- Department of Neurophysiology, Nencki Institute of Experimental Biology, (3 Pasteur Str), Warsaw (02-093), Poland
| |
Collapse
|
26
|
Siu MKY, Wong OGW, Cheung ANY. TrkB as a therapeutic target for ovarian cancer. Expert Opin Ther Targets 2009; 13:1169-78. [PMID: 19694498 DOI: 10.1517/14728220903196787] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
BACKGROUND In many countries, ovarian cancer is the most lethal gynecological malignancy. Its poor prognosis is mainly due to the late stage of disease with metastasis at presentation. The significant failure rate of chemotherapy in patients with advanced stage disease is also a main concern. As such, developing novel therapeutic targets is essential to improve long-term survival. Overexpression of Tropomyosin-related kinase B (TrkB), a tyrosine kinase receptor, has been documented in ovarian cancer and is found to be correlated with poor prognosis. OBJECTIVE/METHODS We discuss the functional roles and the related downstream signaling pathways of TrkB and its ligand brain-derived neurotrophic factor (BDNF) in ovarian cancer. The possible crosstalk between TrkB/BDNF and other putative molecular targets in ovarian cancer is also discussed. RESULTS/CONCLUSIONS All these latest findings shed light on the application of TrkB as a therapeutic target for ovarian cancer.
Collapse
Affiliation(s)
- Michelle K Y Siu
- The University of Hong Kong, Queen Mary Hospital, Department of Pathology, Hong Kong, China
| | | | | |
Collapse
|
27
|
Li W, Xie Y, Sun RWY, Liu Q, Young J, Yu WY, Che CM, Tam PK, Ren Y. Inhibition of Akt sensitises neuroblastoma cells to gold(III) porphyrin 1a, a novel antitumour drug induced apoptosis and growth inhibition. Br J Cancer 2009; 101:342-9. [PMID: 19550420 PMCID: PMC2720197 DOI: 10.1038/sj.bjc.6605147] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
BACKGROUND Gold(III) porphyrin 1a is a new class of anticancer drug, which inhibits cell proliferation of wide range of human cancer cell lines and induces apoptosis in human nasopharyngeal carcinoma cells. However, the underlying signalling mechanism by which gold(III) porphyrin 1a modifies the intracellular apoptosis pathways in tumour cells has not been explained in detail in neuroblastoma cells. METHODS Cell proliferation and apoptosis were determined by measuring 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) and Annexin V binding, respectively. Western blot assay was used to detect proteins involved in apoptotic and Akt pathways. In vivo tumour growth was assessed by inoculating tumour cells to nude mice subcutaneously, and gold(III) porphyrin 1a was administrated intravenously. RESULTS This study assessed the antitumour effect and mechanism of gold(III) porphyrin 1a on neuroblastoma in vitro and in vivo. Gold(III) porphyrin 1a displayed a growth inhibition and induction of apoptosis in neuroblastoma cells effectively in vitro, which was accompanied with release of cytochrome c and Smac/DIABLO and caspases activation. Further studies indicated that gold(III) porphyrin 1a inhibited X-linked inhibitor of apoptosis (XIAP). However, we found that gold(III) porphyrin 1a can induce a survival signal, Akt activation within minutes and could last for at least 24 h. To further confirm association between activation of Akt and the effectiveness of gold(III) porphyrin 1a, neuroblastoma cells were treated with API-2, an Akt-specific inhibitor. API-2 sensitised cells to gold(III) porphyrin 1a-induced apoptosis and growth inhibition. CONCLUSION These results suggested that Akt may be considered as a molecular 'brake' that neuroblastoma cells rely on to slow down gold(III) porphyrin 1a-induced apoptosis and antiproliferation. Gold(III) porphyrin 1a is a mitochondrial apoptotic stimulus but also activates Akt, suggesting an involvement of Akt in mediating the effectiveness to growth inhibition and apoptosis by gold(III) porphyrin 1a and that inhibition of Akt can enhance the anticancer activity of gold(III) porphyrin 1a in neuroblastoma.
Collapse
Affiliation(s)
- W Li
- Department of Surgery, University of Hong Kong, Pokfulam Road, Hong Kong, PR China
| | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Abstract
Medulloblastoma and neuroblastoma are malignant embryonal childhood tumours of the central and peripheral nervous systems, respectively, which often show poor clinical prognosis due to resistance to current chemotherapy. Both these tumours have deficient apoptotic machineries adopted from their respective progenitor cells. This review focuses on the specific background for tumour development, and highlights biological pathways that present potential targets for novel therapeutic approaches.
Collapse
Affiliation(s)
- John Inge Johnsen
- Department of Woman and Child Health, Karolinska Institutet, Astrid Lindgren Children's Hospital, Childhood Cancer Research Unit, Karolinska University Hospital, Stockholm, Sweden
| | | | | | | |
Collapse
|
29
|
Servidei T, Riccardi A, Mozzetti S, Ferlini C, Riccardi R. Chemoresistant tumor cell lines display altered epidermal growth factor receptor and HER3 signaling and enhanced sensitivity to gefitinib. Int J Cancer 2009; 123:2939-49. [PMID: 18803287 DOI: 10.1002/ijc.23902] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
Deregulated signaling through the epidermal growth factor receptor (EGFR) is involved in chemoresistance. To identify the molecular determinants of sensitivity to the EGFR inhibitor gefitinib (Iressa, ZD1839) in chemoresistance, we compared the response of matched chemosensitive and chemoresistant glioma and ovarian cancer cell lines. We found that chemoresistant cell lines were 2- to 3-fold more sensitive to gefitinib growth-inhibitory effects, because of decreased proliferation rather than survival. Sensitivity to gefitinib correlated with overexpression and constitutive phosphorylation of HER2 and HER3, but not EGFR, altered HER ligand expression, and enhanced activation of EGF-triggered EGFR pathway. No activating mutations were found in EGFR. Gefitinib fully inhibited EGF-induced and constitutive Akt activation only in chemoresistant cells. In parallel, gefitinib downregulated constitutively phosphorylated HER2 and HER3, and activated GSK3beta with a concomitant degradation of cyclin D1. Ectopically overexpressed HER2 on its own was insufficient to sensitize chemonaive cells to gefitinib. pHER3 coimmunoprecipitated with p85-PI3K in chemoresistant cells and gefitinib dissociated these complexes. siRNA-mediated inhibition of HER3 decreased constitutive activation of Akt and sensitivity to gefitinib in chemoresistant cells. Our study indicates that in chemoresistant cells gefitinib inhibits both an enhanced EGF-triggered pathway and a constitutive HER3-mediated Akt activation, indicating that inhibition of HER3 together with that of EGFR could be relevant in chemorefractory tumors. Furthermore, in combination experiments gefitinib enhanced the effects of coadministered drugs more in chemoresistant than chemosensitive ovarian cancer cells. Combined treatment might be therapeutically beneficial in chemoresistant tumors from ovary and likely from other tissues.
Collapse
Affiliation(s)
- Tiziana Servidei
- Department of Pediatric Oncology, Catholic University, Rome, Italy.
| | | | | | | | | |
Collapse
|
30
|
Strub GM, Depcrynski A, Elmore LW, Holt SE. Recovery from stress is a function of age and telomere length. Cell Stress Chaperones 2008; 13:475-82. [PMID: 18491040 PMCID: PMC2673929 DOI: 10.1007/s12192-008-0047-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2008] [Revised: 04/07/2008] [Accepted: 04/14/2008] [Indexed: 10/22/2022] Open
Abstract
Cells are constantly exposed to a wide variety of stimuli and must be able to mount appropriate physiological responses in order to maintain proper form and function. Cells from every organism have evolved highly conserved mechanisms to cope with environmental changes, including the widely studied heat shock response (HSR), which is induced by a variety of cellular stresses such as heavy metal ion exposure. It has long been known that as organisms and individual cells age, their ability to appropriately cope with environmental stress is attenuated. Here, we examine the ability of two heavy metal ions (ZnCl(2), SnCl(2)) to induce the HSR in human fibroblasts by assessing the expression of heat shock proteins (Hsp90, Hsp70, and p23) and the ability of the cells to recover over time. We demonstrate that the induction and recovery of chaperone levels is attenuated with age and that cells immortalized with the human telomerase reverse transcriptase component of the telomerase enzyme do not attenuate their HSR as their replicative age increases. Our data suggest that the recovery of normal human cells from an HSR is related in part to age and the cell's overall telomere length.
Collapse
Affiliation(s)
- Graham M. Strub
- Department of Biochemistry, Medical College of Virginia Campus at Virginia Commonwealth University, 1101 E. Marshall St., Richmond, VA 23298-0662 USA
| | - Amy Depcrynski
- Department of Human and Molecular Genetics, Medical College of Virginia Campus at Virginia Commonwealth University, 1101 E. Marshall St., Richmond, VA 23298-0662 USA
| | - Lynne W. Elmore
- Department of Pathology, Medical College of Virginia Campus at Virginia Commonwealth University, 1101 E. Marshall St., Richmond, VA 23298-0662 USA
- Massey Cancer Center, Medical College of Virginia Campus at Virginia Commonwealth University, 1101 E. Marshall St., Richmond, VA 23298–0662 USA
| | - Shawn E. Holt
- Department of Human and Molecular Genetics, Medical College of Virginia Campus at Virginia Commonwealth University, 1101 E. Marshall St., Richmond, VA 23298-0662 USA
- Department of Pathology, Medical College of Virginia Campus at Virginia Commonwealth University, 1101 E. Marshall St., Richmond, VA 23298-0662 USA
- Department of Pharmacology and Toxicology, Medical College of Virginia Campus at Virginia Commonwealth University, 1101 E. Marshall St., Richmond, VA 23298–0662 USA
- Massey Cancer Center, Medical College of Virginia Campus at Virginia Commonwealth University, 1101 E. Marshall St., Richmond, VA 23298–0662 USA
| |
Collapse
|
31
|
Arscott WT, LaBauve AE, May V, Wesley UV. Suppression of neuroblastoma growth by dipeptidyl peptidase IV: relevance of chemokine regulation and caspase activation. Oncogene 2008; 28:479-91. [PMID: 18978811 PMCID: PMC2633428 DOI: 10.1038/onc.2008.402] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Imbalanced protease expression and activities may contribute to the development of cancers, including neuroblastoma (NB). NB is a fatal childhood cancer of the sympathetic nervous system that frequently overexpresses mitogenic peptides, chemokines and their receptors. Dipeptidyl peptidase IV (DPPIV), a cell surface serine protease, inactivates or degrades some of these bioactive peptides and chemokines, thereby regulating cell proliferation and survival. Our studies show that DPPIV is expressed in normal neural crest-derived structures, including superior cervical and dorsal root ganglion cells, sciatic nerve, and in adrenal glands, but its expression is greatly decreased or lost in cells derived from NB, their malignant counterpart. Restoration of DPPIV expression in NB cells led to their differentiation in association with increased expression of the neural marker MAP2 and decreased expression of chemokines, including stromal-derived factor 1 (SDF1) and its receptor CXCR4. Furthermore, DPPIV promoted apoptosis, and inhibited SDF1-mediated in vitro cell migration and angiogenic potential. These changes were accompanied by caspase activation and decreased levels of phospho-Akt and MMP9 activity, which are downstream effectors of SDF1-CXCR4 signaling. Importantly, DPPIV suppressed the tumorigenic potential of NB cells in a xenotransplantation mouse model. These data support a potential role for DPPIV in inhibiting NB growth and progression.
Collapse
Affiliation(s)
- W T Arscott
- Department of Microbiology and Molecular Genetics, Vermont Cancer Center, University of Vermont, Burlington, VT 05405, USA
| | | | | | | |
Collapse
|
32
|
Sartelet H, Oligny LL, Vassal G. AKT pathway in neuroblastoma and its therapeutic implication. Expert Rev Anticancer Ther 2008; 8:757-69. [PMID: 18471048 DOI: 10.1586/14737140.8.5.757] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
Neuroblastoma is a frequent pediatric tumor with a poor outcome in spite of aggressive treatment, even with autologous hematopoietic stem cell transplantation. The overall cure rate of 40% is unsatisfactory and new therapeutic strategies are urgently needed. AKT is a major mediator of survival signals that protect cells from apoptosis and regulate cell proliferation. The AKT signaling network is considered a key determinant of the biological aggressiveness of these tumors. In this article, the authors discuss the relation between activators of AKT in neuroblastoma, in particular, growth factors such as IGF-1, TRK, GDNF, VEGF and EGF, and their effects on tumoral proliferation, differentiation and apoptosis. Numerous other proteins interact with AKT in neuroblastoma. Several are relatively well characterized, such as PTEN and retinoic acid; others are new and potentially interesting, such as PKC and anaplastic lymphoma kinase. Specific inhibition of AKT has been studied, such as with LY249002, with significant effects on cell progression and apoptosis in tumoral cells. Moreover, a series of new drugs, such as geldanamycin and rapamycin, directly modify the expression of AKT in tumoral cells. Few specific inhibitors of AKT are available; less specific inhibitors are probably unsuitable therapeutic options in neuroblastoma. Drugs with a direct or indirect inhibitory effect on the AKT pathway, used alone or in combination with other drugs, seem to hold great promise as a new therapeutic modality in neuroblastoma.
Collapse
Affiliation(s)
- Hervé Sartelet
- Faculty of Medicine, University of Montreal and Pediatric Pathologist, Department of Pathology, CHU Sainte-Justine, 3175, Côte Sainte-Catherine, Montréal, Québec, H3T 1C5, Canada.
| | | | | |
Collapse
|