1
|
Abstract
14-3-3 proteins are mostly expressed in the brain and are closely involved in numerous brain functions and various brain disorders. Among the isotypes of the 14-3-3 proteins, 14-3-3γ is mainly expressed in neurons and is highly produced during brain development, which could indicate that it has a significance in neural development. Furthermore, the distinctive levels of temporally and locally regulated 14-3-3γ expression in various brain disorders suggest that it could play a substantial role in brain plasticity of the diseased states. In this review, we introduce the various brain disorders reported to be involved with 14-3-3γ, and summarize the changes of 14-3-3γ expression in each brain disease. We also discuss the potential of 14-3-3γ for treatment and the importance of research on specific 14-3-3 isotypes for an effective therapeutic approach.
Collapse
Affiliation(s)
- Eunsil Cho
- School of Biosystem and Biomedical Science, College of Health Science, Korea University, Seoul 02708, Korea
| | - Jae-Yong Park
- School of Biosystem and Biomedical Science, College of Health Science, Korea University, Seoul 02708, Korea
| |
Collapse
|
2
|
Targeting 14-3-3ε-CDC25A interactions to trigger apoptotic cell death in skin cancer. Oncotarget 2020; 11:3267-3278. [PMID: 32934772 PMCID: PMC7476737 DOI: 10.18632/oncotarget.27700] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2019] [Accepted: 07/21/2020] [Indexed: 11/25/2022] Open
Abstract
Non-melanoma skin cancer is the most common form of cancer worldwide. We previously documented an anti-apoptotic role for CDC25A in cutaneous squamous cell carcinoma (SCC), an activity dependent on its association with 14-3-3 proteins. We hypothesized that targeting CDC25A-14-3-3ε interactions may be an effective strategy for inducing skin cancer cell apoptosis. Co-immunoprecipitation revealed that CDC25A associated with 14-3-3ε, 14-3-3γ and 14-3-3ζ in SCC cells but not normal keratinocytes. 14-3-3ε and CDC25A activated Akt/BAD/Survivin pro-survival signaling. To target the interaction of 14-3-3ε with CDC25A for cancer therapy, we developed two novel phospho-peptides, pS and pT, corresponding to each of the 14-3-3 binding sites of CDC25A, to specifically interfere with 14-3-3ε binding to CDC25A. Peptides pT (IC50 = 22.1 μM), and pS (IC50 = 29 μM) induced SCC cell death and blocked 14-3-3ε binding to CDC25A. pS or pT treatment of SCC xenografts increased apoptotic cell death and decreased pro-survival P-Akt (S473) and Survivin, demonstrating the effectiveness of the peptides in vivo. These findings lay a framework for the further development of peptides to target 14-3-3ε-CDC25A interactions for skin cancer treatment.
Collapse
|
3
|
Hashemi M, Zali A, Hashemi J, Oraee-Yazdani S, Akbari A. Down-regulation of 14-3-3 zeta sensitizes human glioblastoma cells to apoptosis induction. Apoptosis 2019; 23:616-625. [PMID: 30101359 DOI: 10.1007/s10495-018-1476-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
Strong 14-3-3 zeta protein expression plays an important role in tumorigenesis, including in the maintenance of cell growth, resistance increase, and the prevention of apoptosis. In this study, we focus on two targets: (1) the expression of 14-3-3 zeta in the different grades of human astrocytoma (II-IV), (2) suppression of 14-3-3 zeta protein expression in glioblastoma derived astrocytes by 14-3-3 zeta shRNA lentiviral particles. The tissues of human astrocytoma were provided from 30 patients (ten of each grade of astrocytoma). Control tissues were obtained from the peritumoral brain zone of those patients with glioblastoma. The protein and mRNA expression levels of each astrocytoma grade were assessed via western blotting and RT-PCR, respectively. Results indicated that 14-3-3 zeta was significantly expressed in glioblastoma multiforme (grade IV) and 14-3-3 zeta expression levels enhanced according to the increase of astrocytoma malignancy. In the cellular study for knock down of the 14-3-3 zeta protein, surgical biopsy of glioblastoma was used to isolate primary astrocyte. Astrocytes were transduced with 14-3-3 zeta shRNA or non-targeted shRNA lentiviral particles. Furthermore, reduction of the 14-3-3 zeta protein expression in the astrocytes evaluated through qRT-PCR and western blot after transduction of 14-3-3 zeta shRNA lentiviral particles. Moreover, apoptosis properties, including DNA fragmentation and ratio increase of Bax/Bcl-2 were observed in astrocytes following reduction of 14-3-3 zeta protein expression. Further observation indicated that the mitochondrial pathway through release of cytochorome c and caspase-3 activity was involved in the apoptosis induction. Hence, this study demonstrates a key role of the 14-3-3 zeta protein in tumorigenesis but also indicates that 14-3-3 zeta can be considered as a target for the astrocytoma treatment specially glioblastoma.
Collapse
Affiliation(s)
- Mansoureh Hashemi
- Functional Neurosurgery Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran. .,Shohada Tajrish Hospital, Functional Neurosurgery Research Center, Shahrdari St, Tajrish Square, Tehran, 1989934148, Iran.
| | - Alireza Zali
- Functional Neurosurgery Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Javad Hashemi
- Department of Chemical Engineering, University of Louisville, Louisville, KY40292, USA
| | - Saeed Oraee-Yazdani
- Functional Neurosurgery Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Akhtar Akbari
- Functional Neurosurgery Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
4
|
Down-regulation of 14-3-3zeta reduces proliferation and
increases apoptosis in human glioblastoma. Cancer Gene Ther 2019; 27:399-411. [DOI: 10.1038/s41417-019-0097-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2019] [Revised: 04/08/2019] [Accepted: 04/13/2019] [Indexed: 11/08/2022]
|
5
|
Yun D, Wang H, Wang Y, Chen Y, Zhao Z, Ma J, Ji Y, Huang Q, Chen J, Chen H, Lu D. Shuttling SLC2A4RG is regulated by 14-3-3θ to modulate cell survival via caspase-3 and caspase-6 in human glioma. EBioMedicine 2019; 40:163-175. [PMID: 30686753 PMCID: PMC6413354 DOI: 10.1016/j.ebiom.2019.01.030] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2018] [Revised: 01/10/2019] [Accepted: 01/11/2019] [Indexed: 12/27/2022] Open
Abstract
Background Glioma is the most common and aggressive primary brain tumor with polygenic susceptibility. The cytoplasmic/nuclear shuttling protein, SLC2A4RG (SLC2A4 regulator), has been identified in the 20q13.33 region influencing glioma susceptibility by genome-wide association studies (GWAS) and fine mapping analyses. Methods To discover the expression of SLC2A4RG and its relationship with patient prognosis, tissue microarray containing glioma samples and normal brains was constructed followed by immunohistochemical staining. The role of SLC2A4RG on cell proliferation, cell cycle, and apoptosis was evaluated by gain- and loss-of-function assays in vivo, and subcutaneous and intracranial xenografts were performed to assess its functional effects. The mechanism underlying SLC2A4RG was further investigated via luciferase reporter analyses, ChIP, mass spectrometry, Co-IP, immunofluorescence, etc. Findings The potential tumor suppressor role of SLC2A4RG was further validated by in vitro and in vivo experiments that SLC2A4RG could attenuate cell proliferation via G2/M phase arrest and induce glioma cell apoptosis by direct transactivation of caspase-3 and caspase-6. Moreover, its function displaying showed to depend on the nuclear transportation of SLC2A4RG, however, bound with 14-3-3θ, it would be sequestered in the cytoplasm followed by reversal effect. Interpretation We identify a new pro-oncogenic mechanism whereby 14-3-3θ negatively regulates the nuclear function of the tumor suppressor SLC2A4RG, with significant therapeutic implications for the intervention of human glioma. Fund This work was supported by the National Natural Science Foundation of China (81372706, 81572501, and 81372235).
Collapse
Affiliation(s)
- Dapeng Yun
- State Key Laboratory of Genetic Engineering, School of Life Sciences and Zhongshan Hospital, Fudan University, 2005 Songhu Road, Shanghai 200438, China
| | - Hongxiang Wang
- Department of Neurosurgery, Shanghai Institute of Neurosurgery, Changzheng Hospital, Second Military Medical University, Shanghai 200003, China
| | - Yuqi Wang
- State Key Laboratory of Genetic Engineering, School of Life Sciences and Zhongshan Hospital, Fudan University, 2005 Songhu Road, Shanghai 200438, China
| | - Yuanyuan Chen
- Department of Critical Care Medicine, Wuxi No'2 People's Hospital, Wuxi, Jiangsu Province, China
| | - Zhipeng Zhao
- State Key Laboratory of Genetic Engineering, School of Life Sciences and Zhongshan Hospital, Fudan University, 2005 Songhu Road, Shanghai 200438, China
| | - Jiawei Ma
- Division of Molecular Thoracic Oncology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Yuanyuan Ji
- State Key Laboratory of Genetic Engineering, School of Life Sciences and Zhongshan Hospital, Fudan University, 2005 Songhu Road, Shanghai 200438, China
| | - Qilin Huang
- Department of Neurosurgery, Shanghai Institute of Neurosurgery, Changzheng Hospital, Second Military Medical University, Shanghai 200003, China
| | - Juxiang Chen
- Department of Neurosurgery, Shanghai Institute of Neurosurgery, Changzheng Hospital, Second Military Medical University, Shanghai 200003, China.
| | - Hongyan Chen
- State Key Laboratory of Genetic Engineering, School of Life Sciences and Zhongshan Hospital, Fudan University, 2005 Songhu Road, Shanghai 200438, China.
| | - Daru Lu
- State Key Laboratory of Genetic Engineering, School of Life Sciences and Zhongshan Hospital, Fudan University, 2005 Songhu Road, Shanghai 200438, China.
| |
Collapse
|
6
|
Combination Treatment with PPAR γ Ligand and Its Specific Inhibitor GW9662 Downregulates BIS and 14-3-3 Gamma, Inhibiting Stem-Like Properties in Glioblastoma Cells. BIOMED RESEARCH INTERNATIONAL 2017. [PMID: 28642874 PMCID: PMC5470001 DOI: 10.1155/2017/5832824] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
PPARγ is a nuclear receptor that regulates differentiation and proliferation and is highly expressed in many cancer cells. Its synthetic ligands, such as rosiglitazone and ciglitazone, and its inhibitor GW9662, were shown to induce cellular differentiation, inhibit proliferation, and lead to apoptosis. Glioblastoma is a common brain tumor with poor survival prospects. Recently, glioblastoma stem cells (GSCs) have been examined as a potential target for anticancer therapy; however, little is known about the combined effect of various agents on GSCs. In this study, we found that cotreatment with PPARγ ligands and GW9662 inhibited stem-like properties in GSC-like spheres, which significantly express SOX2. In addition, this treatment decreased the activation of STAT3 and AKT and decreased the amounts of 14-3-3 gamma and BIS proteins. Moreover, combined administration of small-interfering RNA (siRNA) transfection with PPARγ ligands induced downregulation of SOX2 and MMP2 activity together with inhibition of sphere-forming activity regardless of poly(ADP-ribose) polymerase (PARP) cleavage. Taken together, our findings suggest that a combination therapy using PPARγ ligands and its inhibitor could be a potential therapeutic strategy targeting GSCs.
Collapse
|
7
|
Seo SB, Lee JJ, Yun HH, Im CN, Kim YS, Ko JH, Lee JH. 14-3-3β Depletion Drives a Senescence Program in Glioblastoma Cells Through the ERK/SKP2/p27 Pathway. Mol Neurobiol 2017; 55:1259-1270. [PMID: 28116547 DOI: 10.1007/s12035-017-0407-8] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2016] [Accepted: 01/12/2017] [Indexed: 12/12/2022]
Abstract
The induction of senescence in cancer cells has recently been implicated as a mechanism of tumor regression in response to various modes of stress. 14-3-3 proteins are conserved scaffolding molecules that are involved in various cellular functions. Among the seven isoforms, 14-3-3β is specifically expressed in astrocytoma in correlation with the malignancy grade. We investigated the possible role of 14-3-3β in the regulation of senescence induction in A172 glioblastoma cells. The knockdown of 14-3-3β by specific small interfering RNA resulted in a significant change in cellular phenotypes and an increase in cells staining positive for senescence-associated β-galactosidase. Western blotting of the 14-3-3β-depleted A172 cells revealed increased p27 expression and decreased SKP2 expression, while the expression of p53 and p21 was not altered. Subsequently, we demonstrated that ERK is a key modulator of SKP2/p27 axis activity in 14-3-3β-mediated senescence based on the following: (1) 14-3-3β knockdown decreased p-ERK levels; (2) treatment with U0126, an MEK inhibitor, completely reproduced the senescence morphology as well as the expression profiles of p27 and SKP2; and (3) the senescence phenotypes induced by 14-3-3β depletion were considerably recovered by constitutively active ERK expression. Our results indicate that 14-3-3β negatively regulates senescence in glioblastoma cells via the ERK/SKP2/p27 pathway. Furthermore, 14-3-3β depletion also resulted in senescence phenotypes in U87 glioblastoma cells, suggesting that 14-3-3β could be targeted to induce premature senescence as a therapeutic strategy against glioblastoma progression.
Collapse
Affiliation(s)
- Sung Bin Seo
- Department of Biochemistry, College of Medicine, The Catholic University of Korea, 222, Banpo-daero, Seocho-gu, Seoul, 06591, Republic of Korea.,The Institute for Aging and Metabolic Diseases, College of Medicine, The Catholic University of Korea, Seoul, 06591, Republic of Korea
| | - Je-Jung Lee
- Tunneling Nanotube Research Center, Korea University, Seoul, 02841, Republic of Korea
| | - Hye Hyeon Yun
- Department of Biochemistry, College of Medicine, The Catholic University of Korea, 222, Banpo-daero, Seocho-gu, Seoul, 06591, Republic of Korea.,The Institute for Aging and Metabolic Diseases, College of Medicine, The Catholic University of Korea, Seoul, 06591, Republic of Korea
| | - Chang-Nim Im
- Department of Biochemistry, College of Medicine, The Catholic University of Korea, 222, Banpo-daero, Seocho-gu, Seoul, 06591, Republic of Korea.,The Institute for Aging and Metabolic Diseases, College of Medicine, The Catholic University of Korea, Seoul, 06591, Republic of Korea
| | - Yong-Sam Kim
- Genome Editing Research Center, KRIBB, Daejeon, 34141, Republic of Korea
| | - Jeong-Heon Ko
- Genome Editing Research Center, KRIBB, Daejeon, 34141, Republic of Korea
| | - Jeong-Hwa Lee
- Department of Biochemistry, College of Medicine, The Catholic University of Korea, 222, Banpo-daero, Seocho-gu, Seoul, 06591, Republic of Korea. .,The Institute for Aging and Metabolic Diseases, College of Medicine, The Catholic University of Korea, Seoul, 06591, Republic of Korea.
| |
Collapse
|
8
|
Bryukhovetskiy I, Shevchenko V. Molecular mechanisms of the effect of TGF-β1 on U87 human glioblastoma cells. Oncol Lett 2016; 12:1581-1590. [PMID: 27446475 DOI: 10.3892/ol.2016.4756] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2015] [Accepted: 05/26/2016] [Indexed: 12/17/2022] Open
Abstract
Glioblastoma multiforme (GBM) is the most widespread and aggressive type of primary brain tumor. The prognosis following diagnosis with GBM is poor, with a median survival time of 14 months. Tumor cell invasion, metastasis and proliferation are the major causes of mortality in patients with GBM. In order to develop effective GBM treatment methods it is necessary to identify novel targets involved in these processes. Recently, there has been increasing interest in investigating the signaling pathways involved in GBM development, and the transforming growth factor-β (TGF-β) signaling pathway is understood to be significant for regulating the behavior of GBM, as well as stimulating its invasion and metastatic development. Particular interest has been given to investigating the modulation of TGF-β-induced epithelial-to-mesenchymal transition (EMT); during this process, epithelial cells transdifferentiate into mobile cells with a mesenchymal phenotype. The induction of EMT increases the invasiveness of various types of carcinoma; however, the role of TGF-β in this process remains to be elucidated, particularly in the case of GBM. The current study presents a comparative proteome mapping of the U87 human glioblastoma cell line, with and without TGF-β1 treatment. Proteome analysis identified numerous proteins involved in the molecular mechanisms of GBM oncogenesis and TGF-β1 signaling in glioblastoma. The results of the present study facilitated the identification of novel potential markers of metastasis and candidates for targeted glioblastoma therapy, which may potentially be validated and used in clinical medicine to develop improved approaches for GBM diagnosis and treatment.
Collapse
Affiliation(s)
- Igor Bryukhovetskiy
- Laboratory of Molecular and Cellular Neurobiology, School of Biomedicine, Far Eastern Federal University, Vladivostok 690091, Russian Federation
| | - Valeriy Shevchenko
- Laboratory of Molecular and Cellular Neurobiology, School of Biomedicine, Far Eastern Federal University, Vladivostok 690091, Russian Federation; Laboratory of Onco Proteomics, NN Blokhin Russian Cancer Research Center of The Ministry of Health of The Russian Federation, Moscow 115478, Russian Federation
| |
Collapse
|
9
|
Subramanian S, Mahadevan A, Satishchandra P, Shankar SK. Development of a dot blot assay with antibodies to recombinant "core" 14-3-3 protein: Evaluation of its usefulness in diagnosis of Creutzfeldt-Jakob disease. Ann Indian Acad Neurol 2016; 19:205-10. [PMID: 27293331 PMCID: PMC4888683 DOI: 10.4103/0972-2327.176867] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
Abstract
BACKGROUND AND PURPOSE Definitive diagnosis of Creutzfeldt-Jakob disease (CJD) requires demonstration of infective prion protein (PrP(Sc)) in brain tissues by immunohistochemistry or immunoblot, making antemortem diagnosis of CJD difficult. The World Health Organization (WHO) recommends detection of 14-3-3 protein in cerebrospinal fluid (CSF) in cases of dementia, with clinical correlation, as a useful diagnostic marker for CJD, obviating the need for brain biopsy. This facility is currently available in only a few specialized centers in the West and no commercial kit is available for clinical diagnostic use in India. Hence the objective of this study was to develop an in-house sensitive assay for quantitation of 14-3-3 protein and to evaluate its diagnostic potential to detect 14-3-3 proteins in CSF as a biomarker in suspected cases of CJD. MATERIALS AND METHODS A minigene expressing the "core" 14-3-3 protein was synthesized by overlapping polymerase chain reaction (PCR) and the recombinant protein was produced by employing a bacterial expression system. Polyclonal antibodies raised in rabbit against the purified recombinant protein were used for developing a dot blot assay with avidin-biotin technology for signal amplification and quantitation of 14-3-3 protein in CSF. RESULTS The results in the present study suggest the diagnostic potential of the dot blot method with about 10-fold difference (P< 0.001) in the CSF levels of 14-3-3 protein between the CJD cases (N= 50) and disease controls (N= 70). The receiver operating characteristic (ROC) analysis of the results suggested an optimal cutoff value of 2 ng/mL. CONCLUSIONS We have developed an indigenous, economical, and sensitive dot blot method for the quantitation of 14-3-3 protein in CSF.
Collapse
Affiliation(s)
- Sarada Subramanian
- Department of Neurochemistry, National Institute of Mental Health and Neurosciences, Bangalore, Karnataka, India
| | - Anita Mahadevan
- Department of Neuropathology, National Institute of Mental Health and Neurosciences, Bangalore, Karnataka, India
| | | | - Susarla Krishna Shankar
- Department of Neuropathology, National Institute of Mental Health and Neurosciences, Bangalore, Karnataka, India
| |
Collapse
|
10
|
Cao L, Lei H, Chang MZ, Liu ZQ, Bie XH. Down-regulation of 14-3-3β exerts anti-cancer effects through inducing ER stress in human glioma U87 cells: Involvement of CHOP-Wnt pathway. Biochem Biophys Res Commun 2015; 462:389-95. [PMID: 25982477 DOI: 10.1016/j.bbrc.2015.05.003] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2015] [Accepted: 05/01/2015] [Indexed: 12/22/2022]
Abstract
We previously identified 14-3-3β as a tumor-specific isoform of 14-3-3 protein in astrocytoma, but its functional role in glioma cells and underlying mechanisms are poorly understood. In the present study, we investigated the effects of 14-3-3β inhibition in human glioma U87 cells using specific targeted small interfering RNA (siRNA). The results showed that 14-3-3β is highly expressed in U87 cells but not in normal astrocyte SVGp12 cells. Knockdown of 14-3-3β by Si-14-3-3β transfection significantly decreased the cell viability but increased the LDH release in a time-dependent fashion in U87 cells, and these effects were accompanied with G0/G1 cell cycle arrest and apoptosis. In addition, 14-3-3β knockdown induced ER stress in U87 cells, as evidenced by ER calcium release, increased expression of XBP1S mRNA and induction of ER related pro-apoptotic factors. Down-regulation of 14-3-3β significantly decreased the nuclear localization of β-catenin and inhibited Topflash activity, which was shown to be reversely correlated with CHOP. Furthermore, Si-CHOP and sFRP were used to inhibit CHOP and Wnt, respectively. The results showed that the anti-cancer effects of 14-3-3β knockdown in U87 cells were mediated by increased expression of CHOP and followed inhibition of Wnt/β-catenin pathway. In summary, the remarkable efficiency of 14-3-3β knockdown to induce apoptotic cell death in U87 cells may find therapeutic application for the treatment of glioma patients.
Collapse
Affiliation(s)
- Lei Cao
- Department of Neurological Disease, Xi'an Central Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi 710000, China
| | - Hui Lei
- Department of Neurological Disease, Xi'an Central Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi 710000, China
| | - Ming-Ze Chang
- Department of Neurological Disease, Xi'an Central Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi 710000, China
| | - Zhi-Qin Liu
- Department of Neurological Disease, Xi'an Central Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi 710000, China
| | - Xiao-Hua Bie
- Department of Functional Neurosurgery, Xi'an Red Cross Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi 710054, China.
| |
Collapse
|
11
|
Kawai N, Lin W, Cao WD, Ogawa D, Miyake K, Haba R, Maeda Y, Yamamoto Y, Nishiyama Y, Tamiya T. Correlation between ¹⁸F-fluoromisonidazole PET and expression of HIF-1α and VEGF in newly diagnosed and recurrent malignant gliomas. Eur J Nucl Med Mol Imaging 2014; 41:1870-8. [PMID: 24781871 DOI: 10.1007/s00259-014-2776-9] [Citation(s) in RCA: 54] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2014] [Accepted: 03/28/2014] [Indexed: 01/05/2023]
Abstract
PURPOSE Hypoxia and its consequences at the molecular level promote tumour progression and affect patient prognosis. One of the main early cellular events evoked by hypoxia is induction of hypoxia-inducible factor 1 (HIF-1) and subsequent upregulation of vascular endothelial growth factor (VEGF). In this study we sought to determine whether hypoxia detected by (18)F-fluoromisonidazole (FMISO) PET accurately reflects the expression of HIF-1α and VEGF in the tumour and can be used as a biomarker of antiangiogenic treatment and as a prognostic factor in newly diagnosed and recurrent malignant gliomas. METHODS Enrolled in this study were 32 patients with newly diagnosed glioma and 16 with recurrent glioma of grade III or grade IV. All the patients had undergone FMISO PET preoperatively. The maximum tumour-to-blood FMISO activity ratio (T/Bmax) was used to evaluate the degree of tumour hypoxia and the hypoxic volume (HV) was calculated using a tumour-to-blood FMISO uptake ratio of ≥1.2. Immunohistochemical expressions of HIF-1α and VEGF were evaluated semiquantitatively using the immunoreactivity score (IRS, scores 0 to 12) and the correlation was examined between IRS of HIF-1α or VEGF and FMISO uptake of the tumour (SUVtumour) using navigation-based sampling. Survival was estimated using the Kaplan-Meier method in relation to the T/Bmax and the HV. RESULTS The T/Bmax and the HV in grade IV gliomas were significantly higher than in grade III gliomas (P < 0.01 and P < 0.01, respectively). Moderate to strong HIF-1α and VEGF expression was observed in the majority of malignant gliomas. The IRS of HIF-1α and VEGF in the tumour were not significantly different between grade III and grade IV gliomas. The IRS of HIF-1α in the tumour did not correlate with the SUVtumour of FMISO in either newly diagnosed or recurrent glioma. There was a significant but weak correlation between the IRS of VEGF and the SUVtumour of FMISO in newly diagnosed glioma, but not in recurrent glioma. The overall survival time in patients with a small HV and a low FMISO T/Bmax was significantly longer than in those with a large HV and a high FMISO T/Bmax (P < 0.01 and P < 0.05, respectively). CONCLUSION Preoperative FMISO uptake is significantly correlated with the expression of VEGF in the tumour and might be used as a biomarker of antiangiogenic treatment in newly diagnosed malignant gliomas. However, caution is required because the correlation was weak and there was a large overlap of FMISO uptake between glioma with high and low VEGF expression. In addition, hypoxia determined by FMISO PET appears to be a suitable biomarker for predicting a highly malignant tumour and a poor prognosis in patients with malignant glioma.
Collapse
Affiliation(s)
- Nobuyuki Kawai
- Department of Neurological Surgery, Faculty of Medicine, Kagawa University, 1750-1 Miki-cho, Kita-gun, Kagawa, 761-0793, Japan,
| | | | | | | | | | | | | | | | | | | |
Collapse
|
12
|
Kasinski A, Dong X, Khuri FR, Boss J, Fu H. Transcriptional regulation of YWHAZ, the gene encoding 14-3-3ζ. PLoS One 2014; 9:e93480. [PMID: 24690670 PMCID: PMC3972145 DOI: 10.1371/journal.pone.0093480] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2012] [Accepted: 03/06/2014] [Indexed: 12/14/2022] Open
Abstract
Aberrant expression of oncogenic 14-3-3 proteins is correlated with poor survival of cancer patients. While the underlying mechanism of the abnormal expression in tumors remains elusive for the six oncogenic 14-3-3 isoforms; the potential involvement of a transcriptional component has been suggested. Unfortunately, little experimental data has been reported to support this hypothesis. In this study we describe the genetic structure of YWHAZ, the gene encoding 14-3-3ζ, including the identification of previously unreported transcript variants. In total, five transcript variants were revealed and their expressions confirmed in a panel of cell lines. Expressed sequence tag (EST) database mining and in vitro rapid-amplification of cDNA ends (RACE) confirmed that one variant, 1c, represents >80% of the expressed transcripts, which is also the most efficiently translated. An analysis of the proximal promoter of this variant revealed a functional Cyclic-AMP Response Element (CRE). Factors that bound to the CRE element were recognized through fractionation and subsequent EMSAs. This analysis identified CREB and ATF-1 as the trans-interacting factors. Cell-based assays confirm that ATF-1, and to a lesser extent CREB, bind the endogenous YWHAZ promoter especially under TNF-α stimulating conditions. In support of a role of ATF-1 in the regulation of YWHAZ, silencing of ATF-1 resulted in a marked reduction in two of the five YWHAZ transcripts. These data suggest a novel mechanism for the transcriptional regulation of a major pro-survival gene, YWHAZ, by ATF-1.
Collapse
Affiliation(s)
- Andrea Kasinski
- Program in Genetics and Molecular Biology, Emory University, Atlanta, Georgia, United States of America
- Department of Pharmacology, Emory University, Atlanta, Georgia, United States of America
| | - Xueyuan Dong
- Department of Pharmacology, Emory University, Atlanta, Georgia, United States of America
| | - Fadlo R. Khuri
- Department of Hematology and Medical Oncology, Emory University, Atlanta, Georgia, United States of America
| | - Jeremy Boss
- Department of Microbiology and Immunology, Emory University, Atlanta, Georgia, United States of America
| | - Haian Fu
- Department of Pharmacology, Emory University, Atlanta, Georgia, United States of America
- * E-mail:
| |
Collapse
|
13
|
Ruiz Esparza-Garrido R, Velázquez-Flores MÁ, Diegopérez-Ramírez J, López-Aguilar E, Siordia-Reyes G, Hernández-Ortiz M, Martínez-Batallar AG, Encarnación-Guevara S, Salamanca-Gómez F, Arenas-Aranda DJ. A proteomic approach of pediatric astrocytomas: MiRNAs and network insight. J Proteomics 2013; 94:162-75. [PMID: 24060999 DOI: 10.1016/j.jprot.2013.09.009] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2013] [Revised: 07/23/2013] [Accepted: 09/12/2013] [Indexed: 12/13/2022]
Abstract
UNLABELLED Pediatric astrocytomas, a leading cause of death associated with cancer, are the most common primary central nervous system tumors found in children. Most studies of these tumors focus on adults, not on children. We examined the global protein and microRNA expression pattern by 2D SDS-PAGE, mass spectrometry (MALDI-TOF), and RT(2) miRNA PCR Array System. Proteomic studies revealed 49 proteins with changes on the expression. Interactome showed that vimentin, calreticulin, and 14-3-3 epsilon protein are hub proteins in these neoplasms. MicroRNA analyses demonstrated for the first time novel microRNAs involved in the astrocytoma biology. In conclusion, our results show that novel proteins and microRNAs with expression changes on pediatric astrocytoma could serve as biomarkers of tumor progression. BIOLOGICAL SIGNIFICANCE Astrocytomas are tumors that progress rapidly and that invade surrounding tissues. Although some drugs have been developed to treat these neoplasms, the mortality of patients is still very high. In this study, we describe for the first time, to our knowledge, some proteins and miRNAs associated with the biology of astrocytic tumors that could be postulated as possible diagnostic or prognostic biomarkers. Altogether, our results indicate that large-scale analyses allow making a fairly accurate prediction of different cellular processes altered in astrocytic tumors.
Collapse
Affiliation(s)
- Ruth Ruiz Esparza-Garrido
- Unidad de Investigación Médica en Genética Humana, Hospital de Pediatría, Centro Médico Nacional Siglo XXI, IMSS, 06720 México, D.F., Mexico; Posgrado en Ciencias Biológicas, Universidad Nacional Autónoma de México, Av. Universidad 3000, C.P. 04510 Coyoacán, D. F., Mexico
| | | | | | | | | | | | | | | | | | | |
Collapse
|
14
|
Gong F, Wang G, Ye J, Li T, Bai H, Wang W. 14-3-3β regulates the proliferation of glioma cells through the GSK3β/β-catenin signaling pathway. Oncol Rep 2013; 30:2976-82. [PMID: 24065186 DOI: 10.3892/or.2013.2740] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2013] [Accepted: 08/20/2013] [Indexed: 11/05/2022] Open
Abstract
We previously demonstrated that 14-3-3β is overexpressed in astrocytomas; however, the underlying mechanisms are poorly understood. Based on the reported multiple functions of 14-3-3β, we hypothesized that it interacts with glycogen synthase kinase 3 β (GSK3β), which regulates β-catenin-mediated oncogene expression and contributes to tumorigenesis and astrocytoma progression. To test these hypotheses, we used 14-3-3β overexpression vectors and small interfering RNA (siRNA) transfection in the human normal astrocyte cell line SVGp12 and the glioma cell line U87, respectively. The results showed that overexpression of 14-3-3β promoted the proliferation of SVGp12 cells, while knockdown of 14-3-3β inhibited the proliferation of U87 cells as analyzed by the 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) and bromodeoxyuridine (BrdU) assays. In Flag-tagged 14-3-3β-overexpressing cells, GSK3β was co-immunoprecipitated with 14-3-3β using a Flag antibody. Knockdown of β-catenin by siRNA blocked cell proliferation induced by overexpression of 14-3-3β. Furthermore, overexpression of 14-3-3β suppressed the phosphorylation of β-catenin leading to its accumulation and nuclear translocation as revealed by western blot analysis. In addition, β-catenin nuclear translocation induced by overexpression of 14-3-3β activated the transcription of oncogenes including c-myc and cyclin D1. Collectively, these results revealed that 14-3-3β regulates the proliferation of astrocytes and glioma cells through the GSK3β/β-catenin signaling pathway. The delineated mechanism of 14-3-3β may be responsible for the tumorigenesis and progression of human astrocytomas. Thus, new therapeutic strategies or drugs aimed at 14-3-3β may have potential for the treatment of human astrocytomas.
Collapse
Affiliation(s)
- Fanghe Gong
- Department of Neurosurgery, Guangzhou General Hospital of Guangzhou Military Command, Guangzhou 510010, P.R. China
| | | | | | | | | | | |
Collapse
|
15
|
Cao WD, Kawai N, Miyake K, Zhang X, Fei Z, Tamiya T. Relationship of 14-3-3zeta (ζ), HIF-1α, and VEGF expression in human brain gliomas. Brain Tumor Pathol 2013; 31:1-10. [PMID: 23358800 DOI: 10.1007/s10014-013-0135-3] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2012] [Accepted: 01/06/2013] [Indexed: 12/11/2022]
Abstract
Accumulating evidence suggests that tissue hypoxia and apoptosis play important roles in the malignant progression of brain tumors. We investigated the relationship of 14-3-3zeta (an apoptosis-related protein), HIF-1α, and VEGF immunohistochemistry, and evaluated the prognostic value of their expression in human brain gliomas. A semiquantitative analysis of the immunoreactivity scores (IRSs) of the 14-3-3zeta, HIF-1α, and VEGF proteins was performed in 27 patients with various grades of gliomas. The IRS of 14-3-3zeta increased with tumor grade, with grade IV gliomas having the highest score (P < 0.05). Similar results were found for the IRSs of HIF-1α and VEGF (P < 0.05). A significant positive correlation was found between the IRSs of 14-3-3zeta and HIF-1α, 14-3-3zeta and VEGF, and HIF-1α and VEGF (P < 0.001 for all). The survival time of HIF-1α in grade III and grade IV glioma patients with low IRSs (0-6) was significantly longer than that in such glioma patients with high IRSs (8-12) (P < 0.05). These data indicate that 14-3-3zeta, HIF-1α, and VEGF are involved in the same cascade of the malignant progression of gliomas. Further studies will elucidate their detailed role in the malignant progression of glioma, and will contribute to the development of a new treatment strategy for this lethal disease.
Collapse
Affiliation(s)
- Wei-Dong Cao
- Department of Neurological Surgery, Faculty of Medicine, Kagawa University, 1750-1, Miki-cho, Kita-gun, Kagawa, 761-0793, Japan
| | | | | | | | | | | |
Collapse
|
16
|
Yan Y, Xu Y, Gao YY, Zong ZH, Zhang Q, Li C, Wang HQ. Implication of 14-3-3ε and 14-3-3θ/τ in proteasome inhibition-induced apoptosis of glioma cells. Cancer Sci 2012; 104:55-61. [PMID: 23020756 DOI: 10.1111/cas.12033] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2012] [Revised: 09/24/2012] [Accepted: 09/25/2012] [Indexed: 12/18/2022] Open
Abstract
Proteasome inhibitors represent a novel class of anticancer agents that are used in the treatment of hematologic malignancies and various solid tumors. However, mechanisms underlying their anticancer actions were not fully understood. It has been reported that strong 14-3-3 protein expression is observed and associated with tumor genesis and progression of astrocytoma. In addition, global inhibition of 14-3-3 functions with a general 14-3-3 antagonist difopein induces apoptosis of human astrocytoma cells, validating 14-3-3 as a potential molecular target for anticancer therapeutic management. In the current study, for the first time we demonstrated that proteasome inhibitors downregulated 14-3-3ε and 14-3-3θ/τ in U87 and SF295 glioma cells. Overexpression of 14-3-3ε and 14-3-3θ/τ significantly suppressed apoptosis of human glioma cells induced by proteasome inhibitors. We also demonstrated that MG132 activated ASK1 and siASK1 compromised the MG132-induced apoptosis of glioma cells. Furthermore, overexpression of 14-3-3ε and 14-3-3θ/τ markedly suppressed activation of ASK1. Collectively, the current study supported that proteasome inhibitors, at least in part, caused cytotoxicity of glioma cells via downregulation of 14-3-3ε and 14-3-3θ/τ and subsequent activation of ASK1.
Collapse
Affiliation(s)
- Ying Yan
- Department of Radiotherapy, Shenyang Northern Hospital, Shenyang, China
| | | | | | | | | | | | | |
Collapse
|
17
|
Liang S, Shen G, Liu Q, Xu Y, Zhou L, Xiao S, Xu Z, Gong F, You C, Wei Y. Isoform-specific expression and characterization of 14-3-3 proteins in human glioma tissues discovered by stable isotope labeling with amino acids in cell culture-based proteomic analysis. Proteomics Clin Appl 2012; 3:743-53. [PMID: 21136983 DOI: 10.1002/prca.200800198] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Human 14-3-3 proteins have isoform-specific expression and functions in different kinds of normal or tumor cells and tissues. However, the expression profiling of 14-3-3 proteins and isoform-specific biological functions are unclear in human glioma so far. In our study, the expression levels and characterization of 14-3-3 isoforms in human glioma tissues were investigated by a sensitive, accurate stable isotope labeling with amino acids in cell culture-based quantitative proteomic strategy. As a result, except unexpressed 14-3-3σ, the other six isoforms, with different expression levels, were existed in glioma tissues and para-cancerous brain tissues (PBTs). 14-3-3β and η were upregulated, whereas 14-3-3ζ was downregulated in glioma tissues compared with that in PBTs. And the other three isoforms 14-3-3ε, θ, and γ had similar expression levels in human glioma tissues and PBTs. Western blot and immunohistochemistry analysis were both consistent with the quantitative proteomic data. The loss of expression of 14-3-3σ was further discovered due to DNA high methylation in its coding region in glioma by methylation-specific PCR analysis. These results indicated that the four isoforms, including 14-3-3β, η, ζ, and σ, may play important roles in tumorigenesis of human glioma, which is probably used as potential biomarkers for diagnosis and targets for treatment of human gliomas in future.
Collapse
Affiliation(s)
- Shufang Liang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University, Chengdu, P.R. China.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
18
|
Yang X, Cao W, Zhou J, Zhang W, Zhang X, Lin W, Fei Z, Lin H, Wang B. 14-3-3ζ positive expression is associated with a poor prognosis in patients with glioblastoma. Neurosurgery 2012; 68:932-8; discussion 938. [PMID: 21242845 DOI: 10.1227/neu.0b013e3182098c30] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
BACKGROUND When identifying clinical markers predicting clinical outcome, disease recurrence and resistance to therapies often determine the diagnosis and therapy of some cancer types. OBJECTIVE To investigate whether 14-3-3zeta positive expression is an indicator of prognosis in patients with glioblastoma. METHODS Forty-seven patients treated with surgery, radiotherapy, and adjuvant chemotherapy between 2005 and 2007 were divided into 2 groups according to 14-3-3zeta expression in an immunohistochemical study: the 14-3-3zeta negative group (n = 12 patients) and the 14-3-3zeta positive group (n = 35 patients). The clinicopathologic features and survival data for patients in the 14-3-3zeta positive group were compared with data from the patients in the 14-3-3zeta negative group. Kaplan-Meier survival analysis and univariate and multivariate analyses were performed to determine the prognostic factors that influenced patient survival. RESULTS 14-3-3zeta positive expression was observed in approximately 74.5% of patients with glioblastoma. Patients in the 14-3-3zeta positive group had lower overall survival rates and median survival time than those in the 14-3-3zeta negative group (overall 2-year actuarial survival rates, 8.6% for the 14-3-3zeta positive group vs 16.7% for the 14-3-3zeta negative group; overall 2-year median survival time, 12.9 months for the 14-3-3zeta positive group vs 17.9 months for the 14-3-3zeta negative group, P = .019). 14-3-3zeta positive expression in tumor cells also was correlated with a shorter interval to tumor recurrence (median interval to recurrence, 5.9 months in the 14-3-3zeta positive group vs 8.3 months in the 14-3-3zeta negative group, P = .002). Univariate and multivariate analyses showed that 14-3-3zeta positive expression was an independent prognostic factor. CONCLUSION 14-3-3zeta positive expression can be used as a potential molecular risk factor in patients with glioblastoma.
Collapse
Affiliation(s)
- Xiaoliang Yang
- Institute of Neurosurgery, Xijing Hospital, Fourth Military Medical University, Xían, Shaanxi Province, People's Republic of China
| | | | | | | | | | | | | | | | | |
Collapse
|
19
|
Leal MF, Calcagno DQ, Demachki S, Assumpção PP, Chammas R, Burbano RR, Smith MDAC. Clinical implication of 14-3-3 epsilon expression in gastric cancer. World J Gastroenterol 2012; 18:1531-7. [PMID: 22509086 PMCID: PMC3319950 DOI: 10.3748/wjg.v18.i13.1531] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/24/2011] [Revised: 12/23/2011] [Accepted: 12/31/2011] [Indexed: 02/06/2023] Open
Abstract
AIM: To evaluate for the first time the protein and mRNA expression of 14-3-3ε in gastric carcinogenesis.
METHODS: 14-3-3ε protein expression was determined by western blotting, and mRNA expression was examined by real-time quantitative RT-PCR in gastric tumors and their matched non-neoplastic gastric tissue samples.
RESULTS: Authors observed a significant reduction of 14-3-3ε protein expression in gastric cancer (GC) samples compared to their matched non-neoplastic tissue. Reduced levels of 14-3-3ε were also associated with diffuse-type GC and early-onset of this pathology. Our data suggest that reduced 14-3-3ε may have a role in gastric carcinogenesis process.
CONCLUSION: Our results reveal that the reduced 14-3-3ε expression in GC and investigation of 14-3-3ε interaction partners may help to elucidate the carcinogenesis process.
Collapse
|
20
|
Uçar E, Arda N, Aitken A. Extract from mistletoe, Viscum album L., reduces Hsp27 and 14-3-3 protein expression and induces apoptosis in C6 rat glioma cells. GENETICS AND MOLECULAR RESEARCH 2012; 11:2801-13. [DOI: 10.4238/2012.august.24.5] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
|
21
|
|
22
|
Di Sanzo M, Gaspari M, Misaggi R, Romeo F, Falbo L, De Marco C, Agosti V, Quaresima B, Barni T, Viglietto G, Larsen MR, Cuda G, Costanzo F, Faniello MC. H Ferritin Gene Silencing in a Human Metastatic Melanoma Cell Line: A Proteomic Analysis. J Proteome Res 2011; 10:5444-53. [DOI: 10.1021/pr200705z] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Affiliation(s)
- Maddalena Di Sanzo
- Dipartimento di Medicina Sperimentale e Clinica “G. Salvatore”, Università degli Studi di Catanzaro “Magna Græcia”, viale Europa, Campus Universitario, “S. Venuta” - 88100 Catanzaro, Italy
| | - Marco Gaspari
- Dipartimento di Medicina Sperimentale e Clinica “G. Salvatore”, Università degli Studi di Catanzaro “Magna Græcia”, viale Europa, Campus Universitario, “S. Venuta” - 88100 Catanzaro, Italy
| | - Roberta Misaggi
- Dipartimento di Medicina Sperimentale e Clinica “G. Salvatore”, Università degli Studi di Catanzaro “Magna Græcia”, viale Europa, Campus Universitario, “S. Venuta” - 88100 Catanzaro, Italy
| | - Francesco Romeo
- Dipartimento di Medicina Sperimentale e Clinica “G. Salvatore”, Università degli Studi di Catanzaro “Magna Græcia”, viale Europa, Campus Universitario, “S. Venuta” - 88100 Catanzaro, Italy
| | - Lucia Falbo
- Dipartimento di Medicina Sperimentale e Clinica “G. Salvatore”, Università degli Studi di Catanzaro “Magna Græcia”, viale Europa, Campus Universitario, “S. Venuta” - 88100 Catanzaro, Italy
| | - Carmela De Marco
- Laboratorio di Oncologia Molecolare, BioGem s.c. a r.l., Ariano Irpino (AV), Italy
| | - Valter Agosti
- Dipartimento di Medicina Sperimentale e Clinica “G. Salvatore”, Università degli Studi di Catanzaro “Magna Græcia”, viale Europa, Campus Universitario, “S. Venuta” - 88100 Catanzaro, Italy
| | - Barbara Quaresima
- Dipartimento di Medicina Sperimentale e Clinica “G. Salvatore”, Università degli Studi di Catanzaro “Magna Græcia”, viale Europa, Campus Universitario, “S. Venuta” - 88100 Catanzaro, Italy
| | - Tullio Barni
- Dipartimento di Medicina Sperimentale e Clinica “G. Salvatore”, Università degli Studi di Catanzaro “Magna Græcia”, viale Europa, Campus Universitario, “S. Venuta” - 88100 Catanzaro, Italy
| | - Giuseppe Viglietto
- Dipartimento di Medicina Sperimentale e Clinica “G. Salvatore”, Università degli Studi di Catanzaro “Magna Græcia”, viale Europa, Campus Universitario, “S. Venuta” - 88100 Catanzaro, Italy
| | - Martin Røssel Larsen
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, Odense, Denmark
| | - Giovanni Cuda
- Dipartimento di Medicina Sperimentale e Clinica “G. Salvatore”, Università degli Studi di Catanzaro “Magna Græcia”, viale Europa, Campus Universitario, “S. Venuta” - 88100 Catanzaro, Italy
| | - Francesco Costanzo
- Dipartimento di Medicina Sperimentale e Clinica “G. Salvatore”, Università degli Studi di Catanzaro “Magna Græcia”, viale Europa, Campus Universitario, “S. Venuta” - 88100 Catanzaro, Italy
| | - Maria Concetta Faniello
- Dipartimento di Medicina Sperimentale e Clinica “G. Salvatore”, Università degli Studi di Catanzaro “Magna Græcia”, viale Europa, Campus Universitario, “S. Venuta” - 88100 Catanzaro, Italy
| |
Collapse
|
23
|
Broadbelt KG, Rivera KD, Paterson DS, Duncan JR, Trachtenberg FL, Paulo JA, Stapels MD, Borenstein NS, Belliveau RA, Haas EA, Stanley C, Krous HF, Steen H, Kinney HC. Brainstem deficiency of the 14-3-3 regulator of serotonin synthesis: a proteomics analysis in the sudden infant death syndrome. Mol Cell Proteomics 2011; 11:M111.009530. [PMID: 21976671 DOI: 10.1074/mcp.m111.009530] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Impaired brainstem responses to homeostatic challenges during sleep may result in the sudden infant death syndrome (SIDS). Previously we reported a deficiency of serotonin (5-HT) and its key biosynthetic enzyme, tryptophan hydroxylase (TPH2), in SIDS infants in the medullary 5-HT system that modulates homeostatic responses during sleep. Yet, the underlying basis of the TPH2 and 5-HT deficiency is unknown. In this study, we tested the hypothesis that proteomics would uncover previously unrecognized abnormal levels of proteins related to TPH2 and 5-HT regulation in SIDS cases compared with controls, which could provide novel insight into the basis of their deficiency. We first performed a discovery proteomic analysis of the gigantocellularis of the medullary 5-HT system in the same data set with deficiencies of TPH2 and 5-HT levels. Analysis in 6 SIDS cases and 4 controls revealed a 42-75% reduction in abundance in 5 of the 6 isoforms identified of the 14-3-3 signal transduction family, which is known to influence TPH2 activity (p < 0.07). These findings were corroborated in an additional SIDS and control sample using an orthogonal MS(E)-based quantitative proteomic strategy. To confirm these proteomics results in a larger data set (38 SIDS, 11 controls), we applied Western blot analysis in the gigantocellularis and found that 4/7 14-3-3 isoforms identified were significantly reduced in SIDS cases (p ≤ 0.02), with a 43% reduction in all 14-3-3 isoforms combined (p < 0.001). Abnormalities in 5-HT and TPH2 levels and 5-HT(1A) receptor binding were associated with the 14-3-3 deficits in the same SIDS cases. These data suggest a potential molecular defect in SIDS related to TPH2 regulation, as 14-3-3 is critical in this process.
Collapse
Affiliation(s)
- Kevin G Broadbelt
- Department of Pathology, Children's Hospital Boston and Harvard Medical School, Boston, Massachusetts; Proteomics Center, Children's Hospital Boston, Boston, Massachusetts.
| | - Keith D Rivera
- Department of Pathology, Children's Hospital Boston and Harvard Medical School, Boston, Massachusetts
| | - David S Paterson
- Department of Pathology, Children's Hospital Boston and Harvard Medical School, Boston, Massachusetts
| | - Jhodie R Duncan
- Department of Pathology, Children's Hospital Boston and Harvard Medical School, Boston, Massachusetts
| | | | - Joao A Paulo
- Department of Pathology, Children's Hospital Boston and Harvard Medical School, Boston, Massachusetts; Proteomics Center, Children's Hospital Boston, Boston, Massachusetts
| | | | - Natalia S Borenstein
- Department of Pathology, Children's Hospital Boston and Harvard Medical School, Boston, Massachusetts
| | - Richard A Belliveau
- Department of Pathology, Children's Hospital Boston and Harvard Medical School, Boston, Massachusetts
| | - Elisabeth A Haas
- Rady Children's Hospital San Diego and University of California, San Diego School of Medicine, La Jolla, California
| | | | - Henry F Krous
- Rady Children's Hospital San Diego and University of California, San Diego School of Medicine, La Jolla, California
| | - Hanno Steen
- Department of Pathology, Children's Hospital Boston and Harvard Medical School, Boston, Massachusetts; Proteomics Center, Children's Hospital Boston, Boston, Massachusetts
| | - Hannah C Kinney
- Department of Pathology, Children's Hospital Boston and Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
24
|
Wang Z, Nesland JM, Suo Z, Trope CG, Holm R. The prognostic value of 14-3-3 isoforms in vulvar squamous cell carcinoma cases: 14-3-3β and ε are independent prognostic factors for these tumors. PLoS One 2011; 6:e24843. [PMID: 21935479 PMCID: PMC3174199 DOI: 10.1371/journal.pone.0024843] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2010] [Accepted: 08/22/2011] [Indexed: 12/13/2022] Open
Abstract
Background The 14-3-3 family is comprised of highly conserved proteins that are functionally important in the maintenance of homeostasis. Their involvement with the cell cycle, their association with proto-oncogenes and oncogenes, and their abnormal expression in various tumors has linked this family of proteins to the etiology of human cancer. Mounting evidence now indicates that 14-3-3σ is a cancer suppressor gene but the roles of the other 14-3-3 isoforms and their interactions in tumorigenesis have not yet been elucidated. In our current study, we examined the expression of 14-3-3β, γ, ε, ζ, η and τ in a large series of vulvar squamous cell carcinomas to evaluate any clinical significance. Methods Tumor biopsies from 298 vulvar carcinomas were examined by immunohistochemistry for the expression of 14-3-3β, γ, ε, ζ, η and τ. Statistical analyses were employed to validate any associations between the expression of any 14-3-3 isoform and clinicopathologic variables for this disease. Results High cytoplasmic levels of 14-3-3β, γ, ζ, ε and η were observed in 79%, 58%, 50%, 86% and 54% of the vulvar carcinomas analyzed, respectively, whereas a low nuclear expression of 14-3-3τ was present in 80% of these cases. The elevated cytoplasmic expression of 14-3-3β, γ, ε, ζ and η was further found to be associated with advanced disease and aggressive features of these cancers. The overexpression of cytoplasmic 14-3-3β and ε significantly correlated with a poor disease-specific survival by univariate analysis (P = 0.007 and P = 0.04, respectively). The independent prognostic significance of these factors was confirmed by multivariate analysis (P = 0.007 and P = 0.009, respectively). Conclusions We reveal for the first time that the 14-3-3β, γ, ε, ζ, η and τ isoforms may be involved in the progression of vulvar carcinomas. Furthermore, our analyses show that high cytoplasmic levels of 14-3-3β and ε independently correlate with poor disease-specific survival.
Collapse
Affiliation(s)
- Zhihui Wang
- Department of Pathology, The Norwegian Radium Hospital, Oslo University Hospital and University of Oslo, Oslo, Norway
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Medical College of Zhengzhou University, Zhengzhou, China
| | - Jahn M. Nesland
- Department of Pathology, The Norwegian Radium Hospital, Oslo University Hospital and University of Oslo, Oslo, Norway
| | - Zhenhe Suo
- Department of Pathology, The Norwegian Radium Hospital, Oslo University Hospital and University of Oslo, Oslo, Norway
| | - Claes G. Trope
- Department of Obstetrics and Gynecology, The Norwegian Radium Hospital, Oslo University Hospital and University of Oslo, Oslo, Norway
| | - Ruth Holm
- Department of Pathology, The Norwegian Radium Hospital, Oslo University Hospital, Oslo, Norway
- * E-mail:
| |
Collapse
|
25
|
Neal CL, Yu D. 14-3-3ζ as a prognostic marker and therapeutic target for cancer. Expert Opin Ther Targets 2011; 14:1343-54. [PMID: 21058923 DOI: 10.1517/14728222.2010.531011] [Citation(s) in RCA: 107] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
IMPORTANCE OF THE FIELD The ubiquitously expressed 14-3-3ζ protein is involved in numerous important cellular pathways involved in cancer. Recent research suggests 14-3-3ζ may play a central role regulating multiple pathways responsible for cancer initiation and progression. This review will provide an overview of 14-3-3 proteins and address the role of 14-3-3ζ overexpression in cancer. AREAS COVERED IN THIS REVIEW The review covers the basic role of 14-3-3 in regulation of multiple pathways with a focus on 14-3-3ζ as a clinically relevant biomarker for cancer recurrence. WHAT THE READER WILL GAIN 14-3-3ζ overexpression has been found in multiple cancers; however, the clinical implications were unclear. Recently, 14-3-3ζ has been identified as a biomarker for poor prognosis and chemoresistance in multiple tumor types, indicating a potential clinical application for using 14-3-3ζ in selecting treatment options and predicting cancer patients' outcome. TAKE HOME MESSAGE 14-3-3ζ is a potential prognostic marker of cancer recurrence and predictive marker for therapeutic resistance. The overexpression of 14-3-3ζ in multiple cancers suggests that it may be a common target to intervene tumor progression; therefore, more efforts are needed for the development of 14-3-3 inhibitors.
Collapse
Affiliation(s)
- Christopher L Neal
- The University of Texas M. D. Anderson Cancer Center, Department of Molecular and Cellular Oncology, Houston, TX 77030, USA
| | | |
Collapse
|
26
|
Mathie A, Rees KA, El Hachmane MF, Veale EL. Trafficking of neuronal two pore domain potassium channels. Curr Neuropharmacol 2010; 8:276-86. [PMID: 21358977 PMCID: PMC3001220 DOI: 10.2174/157015910792246146] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2009] [Revised: 01/18/2010] [Accepted: 01/18/2010] [Indexed: 01/05/2023] Open
Abstract
The activity of two pore domain potassium (K2P) channels regulates neuronal excitability and cell firing. Post-translational regulation of K2P channel trafficking to the membrane controls the number of functional channels at the neuronal membrane affecting the functional properties of neurons. In this review, we describe the general features of K channel trafficking from the endoplasmic reticulum (ER) to the plasma membrane via the Golgi apparatus then focus on established regulatory mechanisms for K2P channel trafficking. We describe the regulation of trafficking of TASK channels from the ER or their retention within the ER and consider the competing hypotheses for the roles of the chaperone proteins 14-3-3, COP1 and p11 in these processes and where these proteins bind to TASK channels. We also describe the localisation of TREK channels to particular regions of the neuronal membrane and the involvement of the TREK channel binding partners AKAP150 and Mtap2 in this localisation. We describe the roles of other K2P channel binding partners including Arf6, EFA6 and SUMO for TWIK1 channels and Vpu for TASK1 channels. Finally, we consider the potential importance of K2P channel trafficking in a number of disease states such as neuropathic pain and cancer and the protection of neurons from ischemic damage. We suggest that a better understanding of the mechanisms and regulations that underpin the trafficking of K2P channels to the plasma membrane and to localised regions therein may considerably enhance the probability of future therapeutic advances in these areas.
Collapse
Affiliation(s)
- Alistair Mathie
- Medway School of Pharmacy, Universities of Kent and Greenwich at Medway, Central Avenue, Chatham Maritime, Kent ME4 4TB, UK
| | | | | | | |
Collapse
|
27
|
Liang S, Xu Y, Shen G, Liu Q, Zhao X, Xu Z, Xie X, Gong F, Li R, Wei Y. Quantitative protein expression profiling of 14-3-3 isoforms in human renal carcinoma shows 14-3-3 epsilon is involved in limitedly increasing renal cell proliferation. Electrophoresis 2010; 30:4152-62. [PMID: 19960480 DOI: 10.1002/elps.200900249] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
14-3-3 proteins regulate many cellular processes that are implicated in cancer development, and the seven 14-3-3 isoforms have different expression level and isoform-specific roles in different tumors. However, the biological functions of 14-3-3 proteins and their correlations with renal carcinoma have not been investigated so far. In our study, the expression profiles and functional characterization of 14-3-3 proteins were discovered by a sensitive stable isotope labeling with amino acids in cell culture based quantitative proteomics analysis in human renal carcinoma tissues. We found that 14-3-3epsilon was up-regulated with 1.44-fold changes in renal cancerous tissues compared with that in counterpart kidney tissues, and 14-3-3sigma was almost not detected in both tissues due to its DNA highly methylated in our previous reports. The other five isoforms almost have similar expression level in two states of renal tissues. The following RT-PCR, Western blot and immunohistochemistry analysis for specific 14-3-3 isoform expression were all consistent with the quantitative proteomic data. Furthermore, the overexpression of 14-3-3epsilon in vitro can limitedly prompt the abnormal growth of renal tumor cells.
Collapse
Affiliation(s)
- Shufang Liang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University, Chengdu 610041, PR China.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Ratan RR, Noble M. Novel multi-modal strategies to promote brain and spinal cord injury recovery. Stroke 2008; 40:S130-2. [PMID: 19064774 DOI: 10.1161/strokeaha.108.534933] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Stroke is the leading cause of disability in the United States, and yet no definitive interventions can drive the nervous system beyond its measurable but often limited spontaneous recovery. Treatment to limit injury progression and enhance repair after stroke or other types of central nervous system injury is complicated by the heterogeneous nature of cell death and wound healing mechanisms and the multiple barriers to functional recovery. The heterogeneity of injury and repair mechanisms requires interventions that are broad and multi-modal, but also intrinsically safe. We describe a process to identify such interventions by screening multiple individual targets in the historically separate realms of neuroprotection, repair, and regeneration against a library of FDA-approved compounds with known safety. We have identified nearly 10 compounds that are able to activate simultaneously protective and reparative genes. These compounds have a theoretical therapeutic window that spans from evolving injury (minutes to hours) to stable injury (days to months to years). It is our hypothesis that these compounds will be most efficacious when paired with training. The notion is that drugs will alter the propensity of the nervous system toward recovery, whereas specific training will engage the needed instructive cues to achieve this goal. Indeed, robotic training can provide a level of motor learning that seems to enhance the salutary effects of training. In a system that depends heavily on cells that do not easily replenish themselves, cellular therapies could also ultimately be an important part of the cocktail. We conclude that combinations of interventions will be needed to surmount the multiple barriers to recovery in stroke and other types of brain and spinal cord injury recovery.
Collapse
Affiliation(s)
- Rajiv R Ratan
- Winifred Masterson Burke Medical Research Institute, 785 Mamaroneck Avenue, White Plains, NY 10605, USA.
| | | |
Collapse
|