1
|
Deng B, Zhang D, Dai Y, Lin S, Li Y, Wen C. A biodegradable Fe-0.6Se alloy with superior strength and effective antibacterial and antitumor capabilities for orthopedic applications. Acta Biomater 2024; 189:633-650. [PMID: 39395702 DOI: 10.1016/j.actbio.2024.10.012] [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] [Received: 08/01/2024] [Revised: 10/04/2024] [Accepted: 10/08/2024] [Indexed: 10/14/2024]
Abstract
Iron-selenium (Fe-Se) alloys have potential as attractive biodegradable bone-implant materials, given the antitumor properties of Se in cancer prevention and therapy. However, the fabrication of Fe-Se alloys is challenging due to the volatility of elemental Se and the significantly different melting points of Se and Fe. In this study, we successfully fabricated Fe-xSe (x = 0.2, 0.4, 0.6, 0.8, and 1 wt.%) alloys using suction casting, with FeSe compounds as the Se source. The microstructures, tensile properties, corrosion behavior, biocompatibility, antibacterial ability, and antitumor properties of the Fe-Se alloys were evaluated. The microstructures of the Fe-Se alloys were composed of α-Fe and FeSe phases. Among the Fe-Se alloys, Fe-0.6Se showed the best combination of tensile properties, with a yield strength of 1096.5 ± 7.2 MPa, an ultimate tensile strength of 1271.6 ± 6.3 MPa, and a fracture strain of 15.6 ± 3.3 %, and a degradation rate of 56.9 ± 0.4 μm/year. Moreover, the Fe-0.6Se alloy showed superb antibacterial ability against S. aureus, antitumor activity against 143B osteosarcoma cells, and osteogenicity and biocompatibility toward pre-osteoblast MC3T3-E1 cells. In summary, adding 0.2-1.0 wt.% Se to Fe does not affect the growth of healthy cells but effectively inhibits the growth and reproduction of tumor cells, and the Fe-0.6Se alloy is promising for orthopedic applications owing to its unique combination of mechanical and biofunctional properties. STATEMENT OF SIGNIFICANCE: This work reports on Fe-xSe (x = 0.2, 0.4, 0.6, 0.8, and 1 wt.%) alloys fabricated using suction casting. The microstructures of the Fe-Se alloys were composed of α-Fe and FeSe phases. Among the Fe-Se alloys, the Fe-0.6Se showed the best combination of tensile properties, with a yield strength of 1058.6 ± 3.9 MPa, an ultimate tensile strength of 1134.1 ± 2.9 MPa, and a fracture strain of 16.8 ± 1.5 %, and a degradation rate of 56.9 ± 0.4 μm/year. Moreover, the Fe-0.6Se alloy showed superb antibacterial ability against S. aureus, antitumor activity against 143B osteosarcoma cells, and significant osteogenic ability and biocompatibility toward pre-osteoblast MC3T3-E1 cells. In summary, the Fe-0.6Se alloy is promising for orthopedic applications owing to its unique combination of mechanical and biofunctional properties.
Collapse
Affiliation(s)
- Bo Deng
- School of Materials Science and Engineering, Xiangtan University, Xiangtan 411105, China
| | - Dechuang Zhang
- School of Materials Science and Engineering, Xiangtan University, Xiangtan 411105, China.
| | - Yilong Dai
- School of Materials Science and Engineering, Xiangtan University, Xiangtan 411105, China
| | - Sihan Lin
- Department of Prosthodontics, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University, Shanghai, 200011 China.
| | - Yuncang Li
- Centre for Additive Manufacturing, School of Engineering, RMIT University, Melbourne, Victoria 3001, Australia
| | - Cuie Wen
- Centre for Additive Manufacturing, School of Engineering, RMIT University, Melbourne, Victoria 3001, Australia.
| |
Collapse
|
2
|
Hu X, Ke C, Zhong J, Chen Y, Dong J, Hao M, Chen Q, Ni J, Peng Z. Nano selenium-doped TiO 2 nanotube arrays on orthopedic implants for suppressing osteosarcoma growth. Front Bioeng Biotechnol 2023; 11:1252816. [PMID: 37731757 PMCID: PMC10508061 DOI: 10.3389/fbioe.2023.1252816] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Accepted: 08/18/2023] [Indexed: 09/22/2023] Open
Abstract
Osteosarcoma, the most common primary malignant bone tumor, is characterized by malignant cells producing osteoid or immature bone tissue. Most osteosarcoma patients require reconstructive surgery to restore the functional and structural integrity of the injured bone. Metal orthopedic implants are commonly used to restore the limb integrity in postoperative patients. However, conventional metal implants with a bioinert surface cannot inhibit the growth of any remaining cancer cells, resulting in a higher risk of cancer recurrence. Herein, we fabricate a selenium-doped TiO2 nanotube array (Se-doped TNA) film to modify the surface of medical pure titanium substrate, and evaluate the anti-tumor effect and biocompatibility of Se-doped TNA film. Moreover, we further explore the anti-tumor potential mechanism of Se-doped TNA film by studying the behaviors of human osteosarcoma cells in vitro. We provide a new pathway for achieving the anti-tumor function of orthopedic implants while keeping the biocompatibility, aiming to suppress the recurrence of osteosarcoma.
Collapse
Affiliation(s)
- Xiaodong Hu
- Affiliated Li Huili Hospital, Ningbo University, Ningbo, China
| | - Chunhai Ke
- Affiliated Li Huili Hospital, Ningbo University, Ningbo, China
| | - Jiaqi Zhong
- Affiliated Li Huili Hospital, Ningbo University, Ningbo, China
| | - Yujiong Chen
- Affiliated Li Huili Hospital, Ningbo University, Ningbo, China
| | - Jieyang Dong
- Affiliated Li Huili Hospital, Ningbo University, Ningbo, China
| | - Mingming Hao
- Affiliated Li Huili Hospital, Ningbo University, Ningbo, China
- Ningbo Institute of Innovation for Combined Medicine and Engineering (NIIME), Ningbo Medical Centre Lihuili Hospital, Ningbo University, Ningbo, China
| | - Qi Chen
- Ningbo Regen Biotech, Co, Ltd, Ningbo, Zhejiang, China
| | - Jiahua Ni
- Ningbo Regen Biotech, Co, Ltd, Ningbo, Zhejiang, China
| | - Zhaoxiang Peng
- Affiliated Li Huili Hospital, Ningbo University, Ningbo, China
| |
Collapse
|
3
|
Withdrawal Notice. Cancer Med 2023; 12:19353. [PMID: 36372952 PMCID: PMC10557851 DOI: 10.1002/cam4.5306] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Indexed: 11/16/2022] Open
Abstract
Withdrawal Notice: Zhu, Y, Pu, Q, Zhang, Q, et al. Selenium-binding protein 1 inhibits malignant progression and induces apoptosis via distinct mechanisms in non-small-cell lung cancer. Cancer Med. 2022; 00: 1-22. doi: 10.1002/cam4.5306. The above article, published online on 13th November 2022 in Wiley Online Library (https://onlinelibrary.wiley.com/doi/10.1002/cam4.5306), has been withdrawn by agreement between the journal Editor in Chief, Dr Stephen Tait, the Authors, and John Wiley & Sons, Ltd. The withdrawal has been agreed due to an editorial office error that led to the publication of the article without peer review. The revised article, which has undergone peer review may be read here: https://onlinelibrary.wiley.com/doi/10.1002/cam4.6309.
Collapse
|
4
|
Zhu Y, Pu Q, Zhang Q, Liu Y, Ma Y, Yuan Y, Liu L, Zhu W. Selenium-binding protein 1 inhibits malignant progression and induces apoptosis via distinct mechanisms in non-small cell lung cancer. Cancer Med 2023; 12:17149-17170. [PMID: 37606338 PMCID: PMC10501285 DOI: 10.1002/cam4.6309] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Revised: 05/30/2023] [Accepted: 06/23/2023] [Indexed: 08/23/2023] Open
Abstract
BACKGROUND Selenium is an essential trace element in the human body. In epidemiological and clinical studies, Se supplementation significantly reduced the incidence of lung cancer in individuals with low baseline Se levels. The significant action of selenium is based on the selenium-containing protein as a mediator. Of note, the previous studies reported that the expression of selenium-binding protein 1 (SELENBP1) was obviously decreased in many human cancer tissues including non-small cell lung cancer (NSCLC). However, its roles in the origin and development of NSCLC are still unclear. METHODS The expression of SELENBP1 was measured by qRT-PCR, Western blotting and IHC in our collected clinical NSCLC tissues and cell lines. Next, the CCK-8, colony formation, wound-haeling, Millicell, Transwell, FCM assay, and in vivo xenograft model were performed to explore the function of SELENBP1 in NSCLC. The molecular mechanisms of SELENBP1 were investigated by Western blotting or IF assay. RESULTS We further identified that the expression of SELENBP1 was significantly decreased in NSCLC tissues in TCGA database and 45 out of 59 collected clinical NSCLC tissues compared with adjacent nontumor tissues, as well as in four NSCLC cell lines compared with normal lung cells. Particularly, we unexpectedly discovered that SELENBP1 was obviously expressed in alveolar type 2 (AT-II) cells for the first time. Then, a series of in vitro experiments uncovered that overexpression of SELENBP1 inhibited the proliferation, migration, and invasion of NSCLC cells, and induced cell apoptosis. Moreover, overexpression of SELENBP1 also inhibited growth and induced apoptosis of NSCLC cells in vivo. Mechanistically, we demonstrated that overexpression of SELENBP1 inhibited the malignant characteristics of NSCLC cells in part via inactivating the PI3K/AKT/mTOR signal pathway. Meanwhile, we found that overexpression of SELENBP1 inducing the apoptosis of NSCLC cells was associated with the activation of caspase-3 signaling pathway under nonhigh level of oxidative stress, but overexpression of SELENBP1 facilitating the cell apoptosis might be related to its combining with GPX1 and colocalizing in the nucleus under high level of oxidative stress. CONCLUSIONS Our findings highlighted that SELENBP1 was an important tumor suppressor during the origin and development of NSCLC. It may help to discover novel biomarkers or drug therapy targets for NSCLC.
Collapse
Affiliation(s)
- Ying Zhu
- State Key Laboratory of Biotherapy and Cancer CenterWest China Hospital, Sichuan UniversityChengduSichuanChina
| | - Qiang Pu
- Department of Thoracic SurgeryInstitute of Thoracic Oncology, West China Hospital, Sichuan UniversityChengduSichuanChina
| | - Qiongyin Zhang
- State Key Laboratory of Biotherapy and Cancer CenterWest China Hospital, Sichuan UniversityChengduSichuanChina
| | - Yang Liu
- State Key Laboratory of Biotherapy and Cancer CenterWest China Hospital, Sichuan UniversityChengduSichuanChina
| | - Yongfang Ma
- State Key Laboratory of Biotherapy and Cancer CenterWest China Hospital, Sichuan UniversityChengduSichuanChina
| | - Yue Yuan
- State Key Laboratory of Biotherapy and Cancer CenterWest China Hospital, Sichuan UniversityChengduSichuanChina
| | - Lunxu Liu
- Department of Thoracic SurgeryInstitute of Thoracic Oncology, West China Hospital, Sichuan UniversityChengduSichuanChina
| | - Wen Zhu
- State Key Laboratory of Biotherapy and Cancer CenterWest China Hospital, Sichuan UniversityChengduSichuanChina
| |
Collapse
|
5
|
Zhang Y, He Q. The role of SELENBP1 and its epigenetic regulation in carcinogenic progression. Front Genet 2022; 13:1027726. [PMID: 36386843 PMCID: PMC9663989 DOI: 10.3389/fgene.2022.1027726] [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: 08/25/2022] [Accepted: 10/10/2022] [Indexed: 01/24/2023] Open
Abstract
The initiation and progression of cancer is modulated through diverse genetic and epigenetic modifications. The epigenetic machinery regulates gene expression through intertwined DNA methylation, histone modifications, and miRNAs without affecting their genome sequences. SELENBP1 belongs to selenium-binding proteins and functions as a tumor suppressor. Its expression is significantly downregulated and correlates with carcinogenic progression and poor survival in various cancers. The role of SELENBP1 in carcinogenesis has not been fully elucidated, and its epigenetic regulation remains poorly understood. In this review, we summarize recent findings on the function and regulatory mechanisms of SELENBP1 during carcinogenic progression, with an emphasis on epigenetic mechanisms. We also discuss the potential cancer treatment targeting epigenetic modification of SELENBP1, either alone or in combination with selenium-containing compounds or dietary selenium.
Collapse
|
6
|
Hou Y, Wang W, Bartolo P. A concise review on the role of selenium for bone cancer applications. Bone 2021; 149:115974. [PMID: 33901723 DOI: 10.1016/j.bone.2021.115974] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 04/19/2021] [Accepted: 04/19/2021] [Indexed: 01/20/2023]
Abstract
Cancer is one of the most challenging health problems in the world. Several clinical treatments have been developed, but all presenting several limitations. Among different types of cancer, bone cancer is less common, and limited new clinical treatment strategies have been proposed. Recently, a range of advanced materials has been investigated and applied for bone cancer treatment applications. However, due to the unique physiological properties of the bone tissue (a load-bearing tissue), the selection of the right type of material or the combination of suitable functional materials and base materials are critical. Selenium has been reported to present specific targeting inhibition effects on bone cancer without affecting the surrounding healthy tissue, revealing a huge potential for the development of new bone cancer treatment strategies. This paper presents a concise review on the use of selenium for bone cancer applications, discussing main synthesis methods, biocompatibility, and cytotoxicity aspects and the combination of selenium with a wide range of ceramics, metals, and polymers. Future perspectives and the novel concept of a dual-functional scaffold for both cancer treatment and new bone regeneration are also discussed.
Collapse
Affiliation(s)
- Yanhao Hou
- Department of Mechanical, Aerospace and Civil Engineering, School of Engineering, Faculty of Science and Engineering, The University of Manchester, Manchester M13 9PL, UK
| | - Weiguang Wang
- Department of Mechanical, Aerospace and Civil Engineering, School of Engineering, Faculty of Science and Engineering, The University of Manchester, Manchester M13 9PL, UK
| | - Paulo Bartolo
- Department of Mechanical, Aerospace and Civil Engineering, School of Engineering, Faculty of Science and Engineering, The University of Manchester, Manchester M13 9PL, UK.
| |
Collapse
|
7
|
Gui SJ, Ding RL, Wan YP, Zhou L, Chen XJ, Zeng GQ, He CZ. Knockdown of annexin VII enhances nasopharyngeal carcinoma cell radiosensitivity in vivo and in vitro. Cancer Biomark 2021; 28:129-139. [PMID: 31958076 DOI: 10.3233/cbm-190739] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
BACKGROUND Radioresistance leads to treatment failure in patients with nasopharyngeal carcinoma (NPC). Thus, enhancing the radiosensitivity of NPC cells would likely increase the effectiveness of radiotherapy. Annexin VII (Annexin A7, ANXA7) might be a tumor promoter in NPC but its functions in radiosensitivity remain unclear. METHODS NPC cell lines CNE2-shANXA7 and CNE2-pLKO.1 were generated and CNE2-shANXA7 nude mice xenograft tumor models were established. The main effects and molecular mechanisms of ANXA7 knockdown in NPC radiosensitivity were studied in vitro and in vivo by analyzing cell viability, clonogenicity, apoptosis, cell cycle distribution, tumor radioresponse and immunohistochemistry assay. RESULTS ANXA7 knockdown revealed potentially enhanced NPC cell radiosensitivity via apoptosis and increased the cell number at the G2/M phase. In the xenograft model, NPC cells with ANXA7 knockdown were dramatically sensitive to irradiation and tumor growth was significantly suppressed. Compared to CNE2-pLKO.1 xenografts, CNE2-shANXA7 showed more γ-H2AX foci and less phospho-DNA PKcs. CONCLUSIONS ANXA7 knockdown increased the radiosensitivity of NPC by enhancing apoptosis, modulating the cell cycle distribution into more radiosensitive phases, promoting DNA damage, and inhibiting repair. We showed that decreased ANXA7 levels enhanced radiosensitivity and provided insights into the therapeutic targets for NPC radiotherapy.
Collapse
Affiliation(s)
- Si-Jie Gui
- Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Ru-Lei Ding
- School of Nursing, University of South China, Hengyang, Hunan, China
| | - Yan-Ping Wan
- School of Nursing, University of South China, Hengyang, Hunan, China
| | - Li Zhou
- School of Nursing, University of South China, Hengyang, Hunan, China
| | - Xu-Jun Chen
- Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Gu-Qing Zeng
- School of Nursing, University of South China, Hengyang, Hunan, China
| | - Chao-Zhu He
- Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| |
Collapse
|
8
|
Li X, Wang Y, Chen Y, Zhou P, Wei K, Wang H, Wang J, Fang H, Zhang S. Hierarchically constructed selenium-doped bone-mimetic nanoparticles promote ROS-mediated autophagy and apoptosis for bone tumor inhibition. Biomaterials 2020; 257:120253. [DOI: 10.1016/j.biomaterials.2020.120253] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2019] [Revised: 07/15/2020] [Accepted: 07/19/2020] [Indexed: 02/08/2023]
|
9
|
Kim JB, Yang EY, Woo J, Kwon H, Lim W, Moon BI. Sodium Selenite Enhanced the Anti-proliferative Effect of MEK-ERK Inhibitor in Thyroid Cancer Cells. In Vivo 2020; 34:185-190. [PMID: 31882478 DOI: 10.21873/invivo.11760] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2019] [Revised: 10/10/2019] [Accepted: 10/11/2019] [Indexed: 01/02/2023]
Abstract
BACKGROUND/AIM MEK-ERK pathway plays major roles in the progression of thyroid cancer, while the use of MEK-ERK inhibitors has been limited by its toxicity. We investigated the effect of sodium selenite as an adjunct for MEK-ERK inhibitors to avoid the toxicity of ERK inhibitors. MATERIALS AND METHODS TPC1, 8505C and HTori-3 cells were treated with U0126 (MEK-ERK inhibitor) and cell viability was counted in the Neubauer chamber. The synergistic effects of sodium selenite and U0126 were also measured. The expression of ERK, p-ERK, and p90RSK was determined by western blot. RESULTS Treatment with U0126 inhibited proliferation of TPC1 and 8505C cells in a dose-dependent manner. When 5 μM sodium selenite was added to 1 μM U0126, relative cell survival further decreased. Decreased expression of p90RSK indicated that sodium selenite down-regulated ERK signaling in thyroid cancer cells. CONCLUSION The combination of U0126 and sodium selenite inhibited proliferation of thyroid cancer cells through ERK inhibition.
Collapse
Affiliation(s)
- Jong Bin Kim
- Department of Surgery, Ewha Womans University School of Medicine, Ewha Womans University Mokdong Hospital, Ewha Womans University, Seoul, Republic of Korea
| | - Eun Yeol Yang
- Department of Surgery, Ewha Womans University School of Medicine, Ewha Womans University Mokdong Hospital, Ewha Womans University, Seoul, Republic of Korea
| | - Joohyun Woo
- Department of Surgery, Ewha Womans University School of Medicine, Ewha Womans University Mokdong Hospital, Ewha Womans University, Seoul, Republic of Korea
| | - Hyungju Kwon
- Department of Surgery, Ewha Womans University School of Medicine, Ewha Womans University Mokdong Hospital, Ewha Womans University, Seoul, Republic of Korea
| | - Woosung Lim
- Department of Surgery, Ewha Womans University School of Medicine, Ewha Womans University Mokdong Hospital, Ewha Womans University, Seoul, Republic of Korea
| | - Byung-In Moon
- Department of Surgery, Ewha Womans University School of Medicine, Ewha Womans University Mokdong Hospital, Ewha Womans University, Seoul, Republic of Korea
| |
Collapse
|
10
|
Xu J, Ma J, Shi Y, Yin D, Zhang Y, Dai P, Zhao W, Zhang T. Differential Protein Expression between Cystic and Solid Vestibular Schwannoma Using Tandem Mass Tag-Based Quantitative Proteomic Analysis. Proteomics Clin Appl 2020; 14:e1900112. [PMID: 32157794 DOI: 10.1002/prca.201900112] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2019] [Revised: 02/04/2020] [Indexed: 01/02/2023]
Abstract
PURPOSE Cystic vestibular schwannoma (CVS) and solid vestibular schwannoma (SVS) are subgroups of vestibular schwannoma (VS). The tumorigenesis of CVS and SVS have not been fully elucidated, and this study is designed to identify differentially expressed proteins involved in the tumorigenesis of CVS and SVS. EXPERIMENTAL DESIGN Tandem mass tag-based proteomics is used to determine the protein expression profiles from CVS and SVS tissues. RESULTS A total of 30 differentially expressed proteins are identified between CVS and SVS, with 6 being upregulated and 24 being downregulated. Bioinformatics analyses are performed according to Gene Ontology and Kyoto Encyclopedia of Genes and Genomes analyses. These results indicate that two selected proteins (COL1A1 and COL1A2) are potential biomarkers for distinguishing CVS and SVS. CONCLUSIONS AND CLINICAL RELEVANCE Differentially expressed proteins linked to CVS and SVS are identified, and these proteins might provide potential biomarkers for human VS diagnosis. Furthermore, the present study supports the notion that decreased collagen might be the reason for bleeding associated with CVS.
Collapse
Affiliation(s)
- Jianhui Xu
- ENT Institute and Otorhinolaryngology Department of Affiliated Eye and ENT Hospital, State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai, 200031, China.,Key Laboratory of Hearing Medicine of NHFPC, Shanghai, 200031, China
| | - Jing Ma
- ENT Institute and Otorhinolaryngology Department of Affiliated Eye and ENT Hospital, State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai, 200031, China.,Key Laboratory of Hearing Medicine of NHFPC, Shanghai, 200031, China
| | - Yuxuan Shi
- ENT Institute and Otorhinolaryngology Department of Affiliated Eye and ENT Hospital, State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai, 200031, China.,Key Laboratory of Hearing Medicine of NHFPC, Shanghai, 200031, China
| | - Dongming Yin
- ENT Institute and Otorhinolaryngology Department of Affiliated Eye and ENT Hospital, State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai, 200031, China.,Key Laboratory of Hearing Medicine of NHFPC, Shanghai, 200031, China
| | - Yang Zhang
- ENT Institute and Otorhinolaryngology Department of Affiliated Eye and ENT Hospital, State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai, 200031, China.,Key Laboratory of Hearing Medicine of NHFPC, Shanghai, 200031, China
| | - Peidong Dai
- ENT Institute and Otorhinolaryngology Department of Affiliated Eye and ENT Hospital, State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai, 200031, China.,Key Laboratory of Hearing Medicine of NHFPC, Shanghai, 200031, China
| | - Weidong Zhao
- ENT Institute and Otorhinolaryngology Department of Affiliated Eye and ENT Hospital, State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai, 200031, China.,Department of Otology and Skull Base Surgery, Eye and Ear, Nose, Throat Hospital of Fudan University, Shanghai, 200031, China
| | - Tianyu Zhang
- ENT Institute and Otorhinolaryngology Department of Affiliated Eye and ENT Hospital, State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai, 200031, China.,Key Laboratory of Hearing Medicine of NHFPC, Shanghai, 200031, China
| |
Collapse
|
11
|
Ren J, Du Y, Li S, Ma S, Jiang Y, Wu C. Robust network-based regularization and variable selection for high-dimensional genomic data in cancer prognosis. Genet Epidemiol 2019; 43:276-291. [PMID: 30746793 PMCID: PMC6446588 DOI: 10.1002/gepi.22194] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2018] [Revised: 11/19/2018] [Accepted: 11/29/2018] [Indexed: 12/21/2022]
Abstract
In cancer genomic studies, an important objective is to identify prognostic markers associated with patients' survival. Network-based regularization has achieved success in variable selections for high-dimensional cancer genomic data, because of its ability to incorporate the correlations among genomic features. However, as survival time data usually follow skewed distributions, and are contaminated by outliers, network-constrained regularization that does not take the robustness into account leads to false identifications of network structure and biased estimation of patients' survival. In this study, we develop a novel robust network-based variable selection method under the accelerated failure time model. Extensive simulation studies show the advantage of the proposed method over the alternative methods. Two case studies of lung cancer datasets with high-dimensional gene expression measurements demonstrate that the proposed approach has identified markers with important implications.
Collapse
Affiliation(s)
- Jie Ren
- Department of Statistics, Kansas State University, Manhattan, KS
| | - Yinhao Du
- Department of Statistics, Kansas State University, Manhattan, KS
| | - Shaoyu Li
- Department of Mathematics and Statistics, University of North Carolina at Charlotte, Charlotte, NC
| | - Shuangge Ma
- Department of Biostatistics, Yale University, New Haven, CT
| | - Yu Jiang
- Division of Epidemiology, Biostatistics and Environmental Health, School of Public Health, University of Memphis, Memphis, TN
| | - Cen Wu
- Department of Statistics, Kansas State University, Manhattan, KS
| |
Collapse
|
12
|
Liao L, Yan WJ, Tian CM, Li MY, Tian YQ, Zeng GQ. Knockdown of Annexin A1 Enhances Radioresistance and Inhibits Apoptosis in Nasopharyngeal Carcinoma. Technol Cancer Res Treat 2019; 17:1533034617750309. [PMID: 29357787 PMCID: PMC5784564 DOI: 10.1177/1533034617750309] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
Radiotherapy is the primary treatment for nasopharyngeal carcinoma while radioresistance can hinder efficient treatment. To explore the role of annexin A1 and its potential mechanisms in radioresistance of nasopharyngeal carcinoma, human nasopharyngeal carcinoma cell line CNE2-sh annexin A1 (knockdown of annexin A1) and the control cell line CNE2-pLKO.1 were constituted and CNE2-sh annexin A1 xenograft mouse model was generated. The effect of annexin A1 knockdown on the growth of xenograft tumor after irradiation and radiation-induced DNA damage and repair was analyzed. The results of immunohistochemistry assays and Western blotting showed that the level of annexin A1 was significantly downregulated in the radioresistant nasopharyngeal carcinoma tissues or cell line compared to the radiosensitive nasopharyngeal carcinoma tissues or cell line. Knockdown of annexin A1 significantly promoted CNE2-sh annexin A1 xenograft tumor growth compared to the control groups after irradiation. Moreover, the terminal deoxynucleotidyl transferase-mediated dUTP nick end labeling assays revealed that knockdown of annexin A1 significantly inhibited apoptosis in vivo compared to the control groups. We assessed the intracellular reactive oxygen species levels and the extent of radiation-induced DNA damage and repair using reactive oxygen species assay, comet assays, and immunohistochemistry assay. The results showed that knockdown of annexin A1 remarkedly reduced the intracellular reactive oxygen species levels, level of DNA double-strand breaks, and the phosphorylation level of H2AX and increased the accumulation of DNA-dependent protein kinase in nasopharyngeal carcinoma cells after irradiation. The findings suggest that knockdown of annexin A1 inhibits DNA damage via decreasing the generation of intracellular reactive oxygen species and the formation of γ-H2AX and promotes DNA repair via increasing DNA-dependent protein kinase activity and therefore improves the radioresistance in nasopharyngeal carcinoma cells. Together, our findings suggest that knockdown of annexin A1 promotes radioresistance in nasopharyngeal carcinoma and provides insights into therapeutic targets for nasopharyngeal carcinoma radiotherapy.
Collapse
Affiliation(s)
- Li Liao
- 1 School of Public Health, Central South University, Changsha, Hunan, China.,2 School of Nursing, University of South China, Hengyang, Hunan, China
| | - Wen-Jing Yan
- 2 School of Nursing, University of South China, Hengyang, Hunan, China
| | - Chun-Mei Tian
- 2 School of Nursing, University of South China, Hengyang, Hunan, China
| | - Mao-Yu Li
- 3 Key Laboratory of Cancer Proteomics of Chinese Ministry of Health, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Yong-Quan Tian
- 1 School of Public Health, Central South University, Changsha, Hunan, China
| | - Gu-Qing Zeng
- 2 School of Nursing, University of South China, Hengyang, Hunan, China
| |
Collapse
|
13
|
Selenium-Binding Protein 1 in Human Health and Disease. Int J Mol Sci 2018; 19:ijms19113437. [PMID: 30400135 PMCID: PMC6274749 DOI: 10.3390/ijms19113437] [Citation(s) in RCA: 62] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2018] [Revised: 10/05/2018] [Accepted: 10/31/2018] [Indexed: 12/19/2022] Open
Abstract
Selenium-binding protein 1 (SBP1) is a highly conserved protein that covalently binds selenium. SBP1 may play important roles in several fundamental physiological functions, including protein degradation, intra-Golgi transport, cell differentiation, cellular motility, redox modulation, and the metabolism of sulfur-containing molecules. SBP1 expression is often reduced in many cancer types compared to the corresponding normal tissues and low levels of SBP1 are frequently associated with poor clinical outcome. In this review, the transcriptional regulation of SBP1, the different physiological roles reported for SBP1, as well as the implications of SBP1 function in cancer and other diseases are presented.
Collapse
|
14
|
Dong Y, Huo X, Sun R, Liu Z, Huang M, Yang S. lncRNA Gm15290 promotes cell proliferation and invasion in lung cancer through directly interacting with and suppressing the tumor suppressor miR-615-5p. Biosci Rep 2018; 38:BSR20181150. [PMID: 30287504 PMCID: PMC6209606 DOI: 10.1042/bsr20181150] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2018] [Revised: 09/06/2018] [Accepted: 09/13/2018] [Indexed: 01/05/2023] Open
Abstract
Long non-coding RNAs (lncRNAs) have been involved in occurrence and progression of multiple cancers. In the present study, we investigated the role of lncRNA Gm15290 in the proliferation and invasion of non-small cell lung cancer (NSCLC) cells. First, we found that lncRNA Gm15290 was markedly up-regulated in tumor tissues from NSCLC patients and NSCLC cell lines, compared with adjacent normal tissues and normal lung cell line HBE respectively. Then, different concentrations of pcDNA-Gm15290 expression vector and Gm15290 siRNA were respectively transfected into A549 NSCLC cells. Our results showed that overexpression of Gm15290 significantly increased the proliferation and invasion of A549 cells and suppressed cell apoptosis. Knockdown of Gm15290 suppressed A549 cell proliferation and invasion and promoted cell apoptosis. Subsequently, we explored the underlying mechanism through which Gm15290 promoted cell proliferation and invasion. The output of RNA hybrid bioinformatic tool revealed that Gm15290 potentially interacted with tumor suppressor miR-615-5p which displayed an opposite expression pattern in the cell lines and a strong negative correlation with the levels of Gm15290 in NSCLC patients (r2 = 0.9677, P<0.0001). The results of RNA pull-down assays confirmed that Gm15290 directly bound with miR-615-5p Gm15290 negatively regulated the expression of miR-615-5p and increased the protein levels of miR-615-5p target genes, including IGF2, AKT2, and SHMT2 Moreover, miR-615-5p mimic could antagonize the promoting effect of Gm15290 on cell proliferation and invasion.
Collapse
MESH Headings
- Adult
- Antagomirs/genetics
- Antagomirs/metabolism
- Apoptosis
- Base Sequence
- Carcinoma, Non-Small-Cell Lung/genetics
- Carcinoma, Non-Small-Cell Lung/metabolism
- Carcinoma, Non-Small-Cell Lung/pathology
- Carcinoma, Non-Small-Cell Lung/surgery
- Cell Line, Tumor
- Cell Proliferation
- Female
- Gene Expression Regulation, Neoplastic
- Glycine Hydroxymethyltransferase/genetics
- Glycine Hydroxymethyltransferase/metabolism
- Humans
- Insulin-Like Growth Factor II/genetics
- Insulin-Like Growth Factor II/metabolism
- Lung Neoplasms/genetics
- Lung Neoplasms/metabolism
- Lung Neoplasms/pathology
- Lung Neoplasms/surgery
- Male
- MicroRNAs/agonists
- MicroRNAs/antagonists & inhibitors
- MicroRNAs/genetics
- MicroRNAs/metabolism
- Middle Aged
- Neoplasm Invasiveness
- Oligoribonucleotides/genetics
- Oligoribonucleotides/metabolism
- Proto-Oncogene Proteins c-akt/genetics
- Proto-Oncogene Proteins c-akt/metabolism
- RNA, Long Noncoding/genetics
- RNA, Long Noncoding/metabolism
- RNA, Small Interfering/genetics
- RNA, Small Interfering/metabolism
- Signal Transduction
Collapse
Affiliation(s)
- Yu Dong
- Department of Respiratory Medicine, Xi'an Central Hospital, Xi'an, China
- Department of Respiration Medicine, the Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Xiaoying Huo
- Department of Respiratory Medicine, Fourth Hospital of Xi'an, Xi'an, China
| | - Ruiying Sun
- Department of Respiration Medicine, the Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Zhiyan Liu
- Department of Respiratory Medicine, Xi'an Central Hospital, Xi'an, China
| | - Miaoyi Huang
- Department of Respiratory Medicine, Xi'an Central Hospital, Xi'an, China
| | - Shuanying Yang
- Department of Respiration Medicine, the Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| |
Collapse
|
15
|
Xiao J, Lu X, Chen X, Zou Y, Liu A, Li W, He B, He S, Chen Q. Eight potential biomarkers for distinguishing between lung adenocarcinoma and squamous cell carcinoma. Oncotarget 2017; 8:71759-71771. [PMID: 29069744 PMCID: PMC5641087 DOI: 10.18632/oncotarget.17606] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2017] [Accepted: 03/29/2017] [Indexed: 11/25/2022] Open
Abstract
Lung adenocarcinoma (LADC) and squamous cell carcinoma (LSCC) are the most common non-small cell lung cancer histological phenotypes. Accurate diagnosis distinguishing between these two lung cancer types has clinical significance. For this study, we analyzed four Gene Expression Omnibus (GEO) datasets (GSE28571, GSE37745, GSE43580, and GSE50081). We then imported the datasets into the Gene-Cloud of Biotechnology Information online platform to identify genes differentially expressed in LADC and LSCC. We identified DSG3 (desmoglein 3), KRT5 (keratin 5), KRT6A (keratin 6A), KRT6B (keratin 6B), NKX2-1 (NK2 homeobox 1), SFTA2 (surfactant associated 2), SFTA3 (surfactant associated 3), and TMC5 (transmembrane channel-like 5) as potential biomarkers for distinguishing between LADC and LSCC. Receiver operating characteristic curve analysis suggested that KRT5 had the highest diagnostic value for discriminating between these two cancer types. Using the PrognoScan online survival analysis tool and the Kaplan-Meier Plotter, we found that high KRT6A or KRT6B levels, or low NKX2-1, SFTA3, or TMC5 levels correlated with unfavorable prognoses in LADC patients. Further studies will be needed to verify our findings in additional patient samples, and to elucidate the mechanisms of action of these potential biomarkers in non-small cell lung cancer.
Collapse
Affiliation(s)
- Jian Xiao
- Department of Geriatrics, Respiratory Medicine, Xiangya Hospital of Central South University, Changsha 410008, China
| | - Xiaoxiao Lu
- Department of Geriatrics, Respiratory Medicine, Xiangya Hospital of Central South University, Changsha 410008, China
| | - Xi Chen
- Department of Respiratory Medicine, Xiangya Hospital of Central South University, Changsha 410008, China
| | - Yong Zou
- Department of Geriatrics, Respiratory Medicine, Xiangya Hospital of Central South University, Changsha 410008, China
| | - Aibin Liu
- Department of Geriatrics, Xiangya Hospital of Central South University, Changsha 410008, China
| | - Wei Li
- Department of Geriatrics, Clinical Laboratory, Xiangya Hospital of Central South University, Changsha 410008, China
| | - Bixiu He
- Department of Geriatrics, Respiratory Medicine, Xiangya Hospital of Central South University, Changsha 410008, China
| | - Shuya He
- Department of Biochemistry & Biology, University of South China, Hengyang 421001, China
| | - Qiong Chen
- Department of Geriatrics, Respiratory Medicine, Xiangya Hospital of Central South University, Changsha 410008, China
| |
Collapse
|
16
|
Nan Y, Chang R, Jiang H, Yang S, Jin F, Xie Y. Downregulation of P38 phosphorylation correlates with low-grade differentiation and proliferation of lung squamous cell carcinoma. Am J Transl Res 2017; 9:1922-1933. [PMID: 28469797 PMCID: PMC5411940] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2016] [Accepted: 03/18/2017] [Indexed: 06/07/2023]
Abstract
BACKGROUND P38MAPK has been investigated as a tumor-related signaling molecule because of its apparent association with tumorigenesis. This study aimed to investigate P38MAPK expression and its role in lung squamous carcinoma (LSCC). METHODS The expression of P38MAPK and phosphorylated P38 (P-P38) in LSCC tissues and cells was examined by Western blot, real-time PCR, and immunohistochemistry. The influence of P38MAPK inhibitor SB203580 on the proliferation of LSCC cells was detected by MTT and flow cytometry. RESULTS The expression of P-P38 in LSCC tissues and cells was lower than that in cancer-adjacent normal tissues and normal bronchial epithelial cells (P<0.05). In addition, the expression of P-P38 was downregulated in LSCC tissues of poor differentiation, stages III and IV, and with lymph node metastasis compared with the LSCC tissues of well differentiation, stages I and II, and without lymph node metastasis (P<0.05). Moreover, the cell proliferation of LSCC SK-MES-1 cells treated by P38MAPK inhibitor SB203580 significantly increased in a concentration-dependent manner compared with that of SK-MES-1 cells without SB203580 (P<0.05). The inhibition of P38MAPK promoted the transition of the S phase to the G2 phase. CONCLUSIONS P-P38 was poorly expressed in LSCC tissues and cells. Its low expression was correlated with low-grade differentiation, lymph node metastasis, and advanced stage of LSCC. Inhibition of P38MAPK expression could significantly increase the proliferation of LSCC cells by promoting the transition of the S phase to the G2 phase.
Collapse
Affiliation(s)
- Yandong Nan
- Department of Respiration, Tangdu Hospital, Fourth Military Medical UniversityXi’an, China
| | - Ruijing Chang
- Department of Respiration, Tangdu Hospital, Fourth Military Medical UniversityXi’an, China
| | - Hua Jiang
- Department of Respiration, Tangdu Hospital, Fourth Military Medical UniversityXi’an, China
| | - Shuanying Yang
- Department of Respiratory Medicine, Second Affiliated Hospital, Xi’an Jiaotong UniversityXi’an, China
| | - Faguang Jin
- Department of Respiration, Tangdu Hospital, Fourth Military Medical UniversityXi’an, China
| | - Yonghong Xie
- Department of Respiration, Tangdu Hospital, Fourth Military Medical UniversityXi’an, China
| |
Collapse
|