1
|
Lopez A, Bellagamba F, Moretti VM. Nutritional quality traits of Mediterranean mussels ( Mytilus galloprovincialis): A sustainable aquatic food product available on Italian market all year round. FOOD SCI TECHNOL INT 2023; 29:718-728. [PMID: 35790391 DOI: 10.1177/10820132221109582] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
In this study, Mediterranean mussels (Mytilus galloprovincialis) coming from Italian production were purchased in the most important Italian wholesale fish market in different seasons. Biometrical parameters and chemical composition were investigated, with a particular focus on lipid quality and fatty acids (FAs) composition. Results showed a valuable nutritional profile independently by the season of production, represented by high protein and low-fat content, with the lipid portion represented by high amounts of beneficial FAs, particularly the long chain of the n-3 series. Some differences (p < 0.05) were found in carbohydrates and fat content of mussels edible tissues and in FAs profile of specimens collected in different seasons. The most favourable composition in terms of lipid quality was found in mussels collected during spring, corresponding to the moment of the year when mussels store energy reserves in the form of carbohydrates and fat (preparing for the spawning events) and when seawater is enriched in phytoplankton. The lipid health indices calculated (n6/n3, AI, TI) showed optimal values independently by the season of production. The outcomes obtained in this study could help supporting the appreciation of Mediterranean mussels as nutritional valuable seafood product, thus helping encouraging their consumption and promoting the appraisal of this farming sector essential for Italian aquaculture and related to low environmental impact.
Collapse
Affiliation(s)
- Annalaura Lopez
- Department of Veterinary Medicine and Animal Science, Università degli Studi di Milano, Lodi, Italy
| | - Federica Bellagamba
- Department of Veterinary Medicine and Animal Science, Università degli Studi di Milano, Lodi, Italy
| | - Vittorio Maria Moretti
- Department of Veterinary Medicine and Animal Science, Università degli Studi di Milano, Lodi, Italy
| |
Collapse
|
2
|
Tripodi L, Sasso E, Feola S, Coluccino L, Vitale M, Leoni G, Szomolay B, Pastore L, Cerullo V. Systems Biology Approaches for the Improvement of Oncolytic Virus-Based Immunotherapies. Cancers (Basel) 2023; 15:1297. [PMID: 36831638 PMCID: PMC9954314 DOI: 10.3390/cancers15041297] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Revised: 02/06/2023] [Accepted: 02/10/2023] [Indexed: 02/22/2023] Open
Abstract
Oncolytic virus (OV)-based immunotherapy is mainly dependent on establishing an efficient cell-mediated antitumor immunity. OV-mediated antitumor immunity elicits a renewed antitumor reactivity, stimulating a T-cell response against tumor-associated antigens (TAAs) and recruiting natural killer cells within the tumor microenvironment (TME). Despite the fact that OVs are unspecific cancer vaccine platforms, to further enhance antitumor immunity, it is crucial to identify the potentially immunogenic T-cell restricted TAAs, the main key orchestrators in evoking a specific and durable cytotoxic T-cell response. Today, innovative approaches derived from systems biology are exploited to improve target discovery in several types of cancer and to identify the MHC-I and II restricted peptide repertoire recognized by T-cells. Using specific computation pipelines, it is possible to select the best tumor peptide candidates that can be efficiently vectorized and delivered by numerous OV-based platforms, in order to reinforce anticancer immune responses. Beyond the identification of TAAs, system biology can also support the engineering of OVs with improved oncotropism to reduce toxicity and maintain a sufficient portion of the wild-type virus virulence. Finally, these technologies can also pave the way towards a more rational design of armed OVs where a transgene of interest can be delivered to TME to develop an intratumoral gene therapy to enhance specific immune stimuli.
Collapse
Affiliation(s)
- Lorella Tripodi
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università degli Studi di Napoli Federico II, 80138 Naples, Italy
- CEINGE Biotecnologie Avanzate Franco Salvatore, 80131 Naples, Italy
| | - Emanuele Sasso
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università degli Studi di Napoli Federico II, 80138 Naples, Italy
- CEINGE Biotecnologie Avanzate Franco Salvatore, 80131 Naples, Italy
| | - Sara Feola
- Laboratory of Immunovirotherapy, Drug Research Program, Faculty of Pharmacy, University of Helsinki, 00100 Helsinki, Finland
- Translational Immunology Research Program (TRIMM), University of Helsinki, 00100 Helsinki, Finland
- iCAN Digital Precision Cancer Medicine Flagship, University of Helsinki, 00100 Helsinki, Finland
| | - Ludovica Coluccino
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università degli Studi di Napoli Federico II, 80138 Naples, Italy
- CEINGE Biotecnologie Avanzate Franco Salvatore, 80131 Naples, Italy
| | - Maria Vitale
- CEINGE Biotecnologie Avanzate Franco Salvatore, 80131 Naples, Italy
| | - Guido Leoni
- Nouscom Srl, via Castel Romano 100, 00128 Rome, Italy
| | - Barbara Szomolay
- Systems Immunity Research Institute, Cardiff University School of Medicine, Cardiff CF14 4YS, UK
| | - Lucio Pastore
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università degli Studi di Napoli Federico II, 80138 Naples, Italy
- CEINGE Biotecnologie Avanzate Franco Salvatore, 80131 Naples, Italy
| | - Vincenzo Cerullo
- Laboratory of Immunovirotherapy, Drug Research Program, Faculty of Pharmacy, University of Helsinki, 00100 Helsinki, Finland
- Translational Immunology Research Program (TRIMM), University of Helsinki, 00100 Helsinki, Finland
- iCAN Digital Precision Cancer Medicine Flagship, University of Helsinki, 00100 Helsinki, Finland
| |
Collapse
|
3
|
Minaiyan G, Shafiee F, Akbari V. Survivin Promoter-Driven DFF40 Gene Expression Sensitizes Melanoma Cancer Cells to Chemotherapy. Int J Toxicol 2021; 40:380-387. [PMID: 33960240 DOI: 10.1177/10915818211014170] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Downregulation of the apoptotic protein DNA fragmentation factor 40 (DFF40) is correlated with poor overall survival in some malignancies, including melanoma. In this study, DFF40 gene expression driven by survivin promoter, a tumor-specific promoter, was used to selectively induce cytotoxicity in melanoma cells. The activity and strength of survivin promoter were examined in B16F10 murine melanoma, and L929 murine normal fibroblast cell lines using enhanced green fluorescent protein reporter assay and reverse transcription polymerase chain reaction. The effect of expression of DFF40 under the control of cytomegalovirus (CMV) or survivin promoter on viability of cancerous and normal cells was determined by MTT [3-(4,5-Dimethylthiazol-2-yl)-2,5-Diphenyltetrazolium Bromide] assay. Apoptosis induction by expression of DFF40 was evaluated using Annexin-V/propidium iodide staining. Our findings showed high activity of survivin promoter comparable to the control promoter (ie, CMV) in melanoma cells, while survivin activity in normal cells was negligible. Survivin promoter-derived DFF40 gene expression led to selective inhibition of cell viability and induction of apoptosis in cancerous cells. Low and sublethal concentrations of a chemotherapeutic drug, dacarbazine, significantly enhanced the growth inhibitory effect of DFF40 gene therapy. Combination of survivin-driven gene therapy and chemotherapy could be considered as a potential therapeutic treatment for melanoma and possibly other malignancies with similar features.
Collapse
Affiliation(s)
- Ghazale Minaiyan
- Student Research Committee, School of Pharmacy and Pharmaceutical Sciences, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Fatemeh Shafiee
- Department of Pharmaceutical Biotechnology and Isfahan Pharmaceutical Research Center, Faculty of Pharmacy, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Vajihe Akbari
- Department of Pharmaceutical Biotechnology and Isfahan Pharmaceutical Research Center, Faculty of Pharmacy, Isfahan University of Medical Sciences, Isfahan, Iran
| |
Collapse
|
4
|
Wang B, Guo H, Yu H, Chen Y, Xu H, Zhao G. The Role of the Transcription Factor EGR1 in Cancer. Front Oncol 2021; 11:642547. [PMID: 33842351 PMCID: PMC8024650 DOI: 10.3389/fonc.2021.642547] [Citation(s) in RCA: 106] [Impact Index Per Article: 26.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Accepted: 02/25/2021] [Indexed: 12/12/2022] Open
Abstract
Early growth response factor 1 (EGR1) is a transcription factor that is mainly involved in the processes of tissue injury, immune responses, and fibrosis. Recent studies have shown that EGR1 is closely related to the initiation and progression of cancer and may participate in tumor cell proliferation, invasion, and metastasis and in tumor angiogenesis. Nonetheless, the specific mechanism whereby EGR1 modulates these processes remains to be elucidated. This review article summarizes possible mechanisms of action of EGR1 in tumorigenesis and tumor progression and may serve as a reference for clinical efficacy predictions and for the discovery of new therapeutic targets.
Collapse
Affiliation(s)
- Bin Wang
- Department of Neurosurgery, The First Hospital of Jilin University, Changchun, China
| | - Hanfei Guo
- Cancer Center, The First Hospital of Jilin University, Changchun, China
| | - Hongquan Yu
- Department of Neurosurgery, The First Hospital of Jilin University, Changchun, China
| | - Yong Chen
- Department of Neurosurgery, The First Hospital of Jilin University, Changchun, China
| | - Haiyang Xu
- Department of Neurosurgery, The First Hospital of Jilin University, Changchun, China
| | - Gang Zhao
- Department of Neurosurgery, The First Hospital of Jilin University, Changchun, China
| |
Collapse
|
5
|
Monie DD, Bhandarkar AR, Parney IF, Correia C, Sarkaria JN, Vile RG, Li H. Synthetic and systems biology principles in the design of programmable oncolytic virus immunotherapies for glioblastoma. Neurosurg Focus 2021; 50:E10. [PMID: 33524942 DOI: 10.3171/2020.12.focus20855] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2020] [Accepted: 12/04/2020] [Indexed: 12/11/2022]
Abstract
Oncolytic viruses (OVs) are a class of immunotherapeutic agents with promising preclinical results for the treatment of glioblastoma (GBM) but have shown limited success in recent clinical trials. Advanced bioengineering principles from disciplines such as synthetic and systems biology are needed to overcome the current challenges faced in developing effective OV-based immunotherapies for GBMs, including off-target effects and poor clinical responses. Synthetic biology is an emerging field that focuses on the development of synthetic DNA constructs that encode networks of genes and proteins (synthetic genetic circuits) to perform novel functions, whereas systems biology is an analytical framework that enables the study of complex interactions between host pathways and these synthetic genetic circuits. In this review, the authors summarize synthetic and systems biology concepts for developing programmable, logic-based OVs to treat GBMs. Programmable OVs can increase selectivity for tumor cells and enhance the local immunological response using synthetic genetic circuits. The authors discuss key principles for developing programmable OV-based immunotherapies, including how to 1) select an appropriate chassis, a vector that carries a synthetic genetic circuit, and 2) design a synthetic genetic circuit that can be programmed to sense key signals in the GBM microenvironment and trigger release of a therapeutic payload. To illustrate these principles, some original laboratory data are included, highlighting the need for systems biology studies, as well as some preliminary network analyses in preparation for synthetic biology applications. Examples from the literature of state-of-the-art synthetic genetic circuits that can be packaged into leading candidate OV chassis are also surveyed and discussed.
Collapse
Affiliation(s)
- Dileep D Monie
- Departments of1Immunology.,6Mayo Clinic Alix School of Medicine.,7Mayo Clinic Graduate School of Biomedical Sciences; and Mayo Clinic College of Medicine and Science, Rochester, Minnesota
| | | | | | - Cristina Correia
- 5Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic
| | | | | | - Hu Li
- 5Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic
| |
Collapse
|
6
|
Montaño-Samaniego M, Bravo-Estupiñan DM, Méndez-Guerrero O, Alarcón-Hernández E, Ibáñez-Hernández M. Strategies for Targeting Gene Therapy in Cancer Cells With Tumor-Specific Promoters. Front Oncol 2020; 10:605380. [PMID: 33381459 PMCID: PMC7768042 DOI: 10.3389/fonc.2020.605380] [Citation(s) in RCA: 57] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2020] [Accepted: 10/30/2020] [Indexed: 12/16/2022] Open
Abstract
Cancer is the second cause of death worldwide, surpassed only by cardiovascular diseases, due to the lack of early diagnosis, and high relapse rate after conventional therapies. Chemotherapy inhibits the rapid growth of cancer cells, but it also affects normal cells with fast proliferation rate. Therefore, it is imperative to develop other safe and more effective treatment strategies, such as gene therapy, in order to significantly improve the survival rate and life expectancy of patients with cancer. The aim of gene therapy is to transfect a therapeutic gene into the host cells to express itself and cause a beneficial biological effect. However, the efficacy of the proposed strategies has been insufficient for delivering the full potential of gene therapy in the clinic. The type of delivery vehicle (viral or non viral) chosen depends on the desired specificity of the gene therapy. The first gene therapy trials were performed with therapeutic genes driven by viral promoters such as the CMV promoter, which induces non-specific toxicity in normal cells and tissues, in addition to cancer cells. The use of tumor-specific promoters over-expressed in the tumor, induces specific expression of therapeutic genes in a given tumor, increasing their localized activity. Several cancer- and/or tumor-specific promoters systems have been developed to target cancer cells. This review aims to provide up-to-date information concerning targeting gene therapy with cancer- and/or tumor-specific promoters including cancer suppressor genes, suicide genes, anti-tumor angiogenesis, gene silencing, and gene-editing technology, as well as the type of delivery vehicle employed. Gene therapy can be used to complement traditional therapies to provide more effective treatments.
Collapse
Affiliation(s)
- Mariela Montaño-Samaniego
- Laboratorio de Terapia Génica, Departamento de Bioquímica, Escuela Nacional de Ciencias Biológicas del Instituto Politécnico Nacional, Ciudad de México, México
| | - Diana M. Bravo-Estupiñan
- Laboratorio de Terapia Génica, Departamento de Bioquímica, Escuela Nacional de Ciencias Biológicas del Instituto Politécnico Nacional, Ciudad de México, México
| | - Oscar Méndez-Guerrero
- Laboratorio de Terapia Génica, Departamento de Bioquímica, Escuela Nacional de Ciencias Biológicas del Instituto Politécnico Nacional, Ciudad de México, México
| | - Ernesto Alarcón-Hernández
- Laboratorio de Genética Molecular, Departamento de Bioquímica, Escuela Nacional de Ciencias Biológicas del Instituto Politécnico Nacional, Ciudad de México, México
| | - Miguel Ibáñez-Hernández
- Laboratorio de Terapia Génica, Departamento de Bioquímica, Escuela Nacional de Ciencias Biológicas del Instituto Politécnico Nacional, Ciudad de México, México
| |
Collapse
|
7
|
Wang N, Li Y, Zhou RM, Cui SJ, Cao SR, Huang X, Huo XR, Shan BE. The effect of polymorphisms in the promoter of the BIRC5 gene on the risk of oesophageal squamous cell carcinoma and patient’s outcomes. Mutagenesis 2019; 34:307-313. [PMID: 31165868 DOI: 10.1093/mutage/gez012] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2019] [Accepted: 05/17/2019] [Indexed: 12/24/2022] Open
Abstract
Abstract
Baculoviral inhibitor of apoptosis repeat-containing 5 (BIRC5) is an inhibitor of apoptosis proteins and plays a key role in apoptosis or programmed cell death. In the present study, we evaluated the effect of BIRC5 gene polymorphisms on the risk of developing oesophageal squamous cell carcinoma (ESCC) and patients’ outcomes in a high-incidence population from northern China. A population-based case-control study was performed in 597 ESCC patients and 597 control subjects.Survival data were available for 211 patients who received platinum-based chemotherapy after surgery. Five polymorphisms (-31 C>G, -241 C>T, -625 G>C, -644 T>C and -1547 A>G) in the promoter of the BIRC5 gene were genotyped by the polymerase chain reaction-ligase detection reaction (PCR-LDR) method. Compared with the -31 CC genotype, the -31 CG/GG genotype of -31 C>G single nucleotide polymorphism (SNP) was associated with a significant elevated risk of ESCC [adjusted odds ratio (OR) = 1.40, 95% confidence interval (CI) = 1.07–1.84]. Interestingly, this association was stronger among females, younger patients and non-smokers in stratified analyses (adjusted OR = 1.72, 95% CI = 1.07–2.75; adjusted OR = 1.61, 95% CI = 1.10–2.36; adjusted OR = 1.80, 95% CI = 1.26–2.58, respectively]. Survival analyses showed that the T allele of -241 C>T SNP was associated with poor prognosis [hazard ratio (HR) = 2.99, 95% CI = 1.09–8.19) and that the C allele of -625 G>C SNP was associated with good prognosis (HR = 0.62, 95% CI = 0.38–0.99) in ESCC patients. The -31 C>G polymorphism may be involved in the development of ESCC, and the -241 C>T and -625 G>C polymorphisms may be useful prognostic markers for ESCC.
Collapse
Affiliation(s)
- Na Wang
- Cancer Institute, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, PR China
| | - Yan Li
- Cancer Institute, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, PR China
| | - Rong-Miao Zhou
- Cancer Institute, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, PR China
| | - Sai-Jin Cui
- Cancer Institute, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, PR China
| | - Shi-Ru Cao
- Cancer Institute, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, PR China
| | - Xi Huang
- Cancer Institute, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, PR China
| | - Xiang-Ran Huo
- Cancer Institute, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, PR China
| | - Bao-En Shan
- Cancer Institute, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, PR China
| |
Collapse
|
8
|
Zhao Z, Huang R, Cai H, Liu B, Zeng Y, Kuang A. Improved radioiodine-131 imaging of prostatic carcinoma using the sodium iodide symporter gene under control of the survivin promoter. INTERNATIONAL JOURNAL OF CLINICAL AND EXPERIMENTAL PATHOLOGY 2018; 11:4067-4072. [PMID: 31949797 PMCID: PMC6962784] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 05/21/2018] [Accepted: 06/29/2018] [Indexed: 06/10/2023]
Abstract
Improvement of radioiodine accumulation in non-thyroidal tumors by transfecting the sodium iodide symporter (NIS) gene has been successfully investigated in many studies. However, regarding the uncertain iodine influx and efflux efficiencies in different cells, the optimal imaging time by radioiodine following NIS gene transport remains unclear. This study aimed to investigate the serial expression of NIS under control of survivin promoter in prostate cancer PC-3 cells and xenografts by adenoviral vector (Ad-Sur-NIS), and determine the optimal imaging time for radioiodine application. In vitro, the 125I accumulation in Ad-Sur-NIS-infected PC-3 cells was 44 times higher than that in control cells (P<0.05). Moreover, the expression efficiency of NIS reached a peak at 48 h post transfection, at which a 1.9-fold or 1.4-fold increase of 125I accumulation was found compared with 24 h or 72 h. In the clonogenic assay, the cell inhibition rates induced by 131I were 93.4 ± 11.2% in Ad-Sur-NIS and 71.8 ± 10.1% in Ad-NIS infected cells, both of which were significantly higher than that in Ad-Sur-GFP infected cells (10.9 ± 1.9%, P<0.05). In in vivo studies, the 131I uptake of tumor-to-muscle ratios were more prominent on day 2 (15.23 ± 4.55) and day-9 (9.78 ± 2.34) compared to the day 16 (1.29 ± 0.49), which showed a gradual reduction (P<0.05). Therefore, the Ad-Sur-NIS transfection allowed PC-3 tumor imaging by iodine-131 with an optimal time no later than 9 days post-transfection.
Collapse
Affiliation(s)
- Zhen Zhao
- Department of Nuclear Medicine, West China Hospital of Sichuan University Chengdu, Sichuan, China
| | - Rui Huang
- Department of Nuclear Medicine, West China Hospital of Sichuan University Chengdu, Sichuan, China
| | - Huawei Cai
- Department of Nuclear Medicine, West China Hospital of Sichuan University Chengdu, Sichuan, China
| | - Bin Liu
- Department of Nuclear Medicine, West China Hospital of Sichuan University Chengdu, Sichuan, China
| | - Yu Zeng
- Department of Nuclear Medicine, West China Hospital of Sichuan University Chengdu, Sichuan, China
| | - Anren Kuang
- Department of Nuclear Medicine, West China Hospital of Sichuan University Chengdu, Sichuan, China
| |
Collapse
|
9
|
Li J, Han Y, Zhou D, Zhou Y, Ye M, Wang H, Du Z. Downregulation of Survivin Gene Expression Affects Ionizing Radiation Resistance of Human T98 Glioma Cells. Cell Mol Neurobiol 2018; 38:861-868. [PMID: 29098505 DOI: 10.1007/s10571-017-0560-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2017] [Accepted: 10/16/2017] [Indexed: 12/15/2022]
Abstract
Survivin is a tumor-associated gene, which has been detected in a wide variety of human tumors. Previous research has shown that Survivin can affect hepatoma carcinoma cell radiosensitivity. However, little is known about the role of Survivin in ionizing radiation resistance in glioma cells. In this study, we aimed to identify the effects of Survivin on ionizing radiation resistance in glioma cell line T98. Our results showed that downregulation of Survivin gene expression and ionizing irradiation could both inhibit T98 cell proliferation by assays in vitro including CCK-8 and immunohistochemistry. The inhibitory effect of downregulation of Survivin combined with irradiation was the most significant compared with other groups. Results of Western blotting and flow cytometric analysis also showed that downregulation of Survivin combined with the irradiation group achieved the highest apoptosis rate. Experimental results in vivo by intracranial implanting into nude mice were consistent with those in vitro. These findings indicated that ionizing radiation resistance of human T98 glioma cells can be inhibited effectively after Survivin gene silencing.
Collapse
Affiliation(s)
- Jicheng Li
- Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, 215006, Jiangsu, People's Republic of China
| | - Yong Han
- Department of Neurosurgery, Children's Hospital of Soochow University, Suzhou, 215000, Jiangsu, People's Republic of China
| | - Dai Zhou
- Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, 215006, Jiangsu, People's Republic of China
| | - Youxin Zhou
- Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, 215006, Jiangsu, People's Republic of China
| | - Ming Ye
- Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, 215006, Jiangsu, People's Republic of China.
| | - Hangzhou Wang
- Department of Neurosurgery, Children's Hospital of Soochow University, Suzhou, 215000, Jiangsu, People's Republic of China.
| | - Ziwei Du
- Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, 215006, Jiangsu, People's Republic of China
| |
Collapse
|
10
|
Arafat W, El Ashry MS, Abd Alrazek MAA, Matta CA, El Aleem EA, Kamel EM, Hussein M, Ezzat G, Samir M. The Relation between Survivin Gene Expression and Urinary Bladder Cancer Disease. HOSPICE AND PALLIATIVE MEDICINE INTERNATIONAL JOURNAL 2017; 1. [DOI: 10.15406/hpmij.2017.01.00007] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/01/2023]
|