1
|
Bai F, He Z, Zhou H, Gan W. Kinesin family member 2A links with advanced tumor stage, reduced chemosensitivity and worse prognosis in gastric cancer. J Clin Lab Anal 2022; 36:e24313. [PMID: 35313389 PMCID: PMC9102491 DOI: 10.1002/jcla.24313] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Revised: 02/08/2022] [Accepted: 02/16/2022] [Indexed: 11/19/2022] Open
Abstract
Background Kinesin family member 2A (KIF2A) induces gastric cancer (GC) growth and invasion, while its clinical relevance in GC patients is not reported. This study aimed to investigate the linkage of KIF2A with clinicopathological features, prognosis, and chemosensitivity of GC. Methods A total of 160 surgical GC patients were reviewed, with their tumor and adjacent tissues acquired for immunohistochemical (IHC) assay to measure KIF2A expression, then scored by a semi‐quantitative method (IHC score: 0–12). KIF2A siRNA or nonsense‐siRNA were transfected into HGC‐27 and NCI‐N87 cells underwent various concentrations of capecitabine or oxaliplatin treatment followed by chemosensitivity assessment. Results Kinesin family member 2A expression was elevated in the tumor tissue compared to the adjacent tissue (IHC score: 5.6 ± 3.1 vs. 2.9 ± 1.7, p < 0.001). Besides, tumor KIF2A expression was related to larger tumor size (p = 0.014), higher N stage (p = 0.004) and TNM stage (p = 0.011); however, it was not linked with other clinicopathological features (all p > 0.05). Signally, tumor KIF2A high expression predicted poor overall survival (p = 0.037). After adjustment via multivariate Cox's regression, tumor KIF2A high expression independently linked with worse disease‐free survival (p = 0.033). Finally, KIF2A knockdown improved the oxaliplatin chemosensitivity vastly but only slightly affected capecitabine chemosensitivity in HGC‐27 and NCI‐N87 cells. Conclusion Kinesin family member 2A reflects larger tumor size, advanced TNM stage, improved chemosensitivity, and predicts unfavorable survival in GC.
Collapse
Affiliation(s)
- Fei Bai
- Department of Gastroduodenal Pancreas Surgery, Hunan Cancer Hospital & The Affiliated Hospital of Xiangya School of Medicine, Central South University, Changsha, China
| | - Zhuo He
- Department of Gastroduodenal Pancreas Surgery, Hunan Cancer Hospital & The Affiliated Hospital of Xiangya School of Medicine, Central South University, Changsha, China
| | - Huijun Zhou
- Department of Gastroenterology and Urology, Hunan Cancer Hospital & The Affiliated Hospital of Xiangya School of Medicine, Central South University, Changsha, China
| | - Wei Gan
- Department of General Surgery, The Second Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
2
|
Gilet JG, Ivanova EL, Trofimova D, Rudolf G, Meziane H, Broix L, Drouot N, Courraud J, Skory V, Voulleminot P, Osipenko M, Bahi-Buisson N, Yalcin B, Birling MC, Hinckelmann MV, Kwok BH, Allingham JS, Chelly J. Conditional switching of KIF2A mutation provides new insights into cortical malformation pathogeny. Hum Mol Genet 2021; 29:766-784. [PMID: 31919497 DOI: 10.1093/hmg/ddz316] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2019] [Revised: 12/04/2019] [Accepted: 12/20/2019] [Indexed: 12/19/2022] Open
Abstract
By using the Cre-mediated genetic switch technology, we were able to successfully generate a conditional knock-in mouse, bearing the KIF2A p.His321Asp missense point variant, identified in a subject with malformations of cortical development. These mice present with neuroanatomical anomalies and microcephaly associated with behavioral deficiencies and susceptibility to epilepsy, correlating with the described human phenotype. Using the flexibility of this model, we investigated RosaCre-, NestinCre- and NexCre-driven expression of the mutation to dissect the pathophysiological mechanisms underlying neurodevelopmental cortical abnormalities. We show that the expression of the p.His321Asp pathogenic variant increases apoptosis and causes abnormal multipolar to bipolar transition in newborn neurons, providing therefore insights to better understand cortical organization and brain growth defects that characterize KIF2A-related human disorders. We further demonstrate that the observed cellular phenotypes are likely to be linked to deficiency in the microtubule depolymerizing function of KIF2A.
Collapse
Affiliation(s)
- Johan G Gilet
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, 67400 Illkirch, France.,CNRS UMR 7104, 67400 Illkirch, France.,INSERM U1258, 67400 Illkirch, France.,Université de Strasbourg, 67400 Illkirch, France
| | - Ekaterina L Ivanova
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, 67400 Illkirch, France.,CNRS UMR 7104, 67400 Illkirch, France.,INSERM U1258, 67400 Illkirch, France.,Université de Strasbourg, 67400 Illkirch, France
| | - Daria Trofimova
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, ON K7L 3N6, Canada
| | - Gabrielle Rudolf
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, 67400 Illkirch, France.,CNRS UMR 7104, 67400 Illkirch, France.,INSERM U1258, 67400 Illkirch, France.,Université de Strasbourg, 67400 Illkirch, France
| | - Hamid Meziane
- CNRS UMR 7104, 67400 Illkirch, France.,CELPHEDIA, PHENOMIN, Institut Clinique de la Souris (ICS), CNRS, INSERM, Université de Strasbourg, F-67404 Illkirch-Graffenstaden, France
| | - Loic Broix
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, 67400 Illkirch, France.,CNRS UMR 7104, 67400 Illkirch, France.,INSERM U1258, 67400 Illkirch, France.,Université de Strasbourg, 67400 Illkirch, France
| | - Nathalie Drouot
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, 67400 Illkirch, France.,CNRS UMR 7104, 67400 Illkirch, France.,INSERM U1258, 67400 Illkirch, France.,Université de Strasbourg, 67400 Illkirch, France
| | - Jeremie Courraud
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, 67400 Illkirch, France.,CNRS UMR 7104, 67400 Illkirch, France.,INSERM U1258, 67400 Illkirch, France.,Université de Strasbourg, 67400 Illkirch, France
| | - Valerie Skory
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, 67400 Illkirch, France.,CNRS UMR 7104, 67400 Illkirch, France.,INSERM U1258, 67400 Illkirch, France.,Université de Strasbourg, 67400 Illkirch, France
| | - Paul Voulleminot
- Département de Neurologie, Hôpital de Hautepierre, Hôpitaux Universitaires de Strasbourg, 67200 Strasbourg, France
| | - Maria Osipenko
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, 67400 Illkirch, France.,CNRS UMR 7104, 67400 Illkirch, France.,INSERM U1258, 67400 Illkirch, France.,Université de Strasbourg, 67400 Illkirch, France
| | - Nadia Bahi-Buisson
- Imagine Institute, Paris Descartes-Sorbonne Paris Cité University, 75015 Paris, France
| | - Binnaz Yalcin
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, 67400 Illkirch, France.,CNRS UMR 7104, 67400 Illkirch, France.,INSERM U1258, 67400 Illkirch, France.,Université de Strasbourg, 67400 Illkirch, France
| | - Marie-Christine Birling
- CNRS UMR 7104, 67400 Illkirch, France.,CELPHEDIA, PHENOMIN, Institut Clinique de la Souris (ICS), CNRS, INSERM, Université de Strasbourg, F-67404 Illkirch-Graffenstaden, France
| | - Maria-Victoria Hinckelmann
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, 67400 Illkirch, France.,CNRS UMR 7104, 67400 Illkirch, France.,INSERM U1258, 67400 Illkirch, France.,Université de Strasbourg, 67400 Illkirch, France
| | - Benjamin H Kwok
- Département de médecine, Institute for Research in Immunology and Cancer (IRIC), Université de Montréal, Montréal, QC H3C 3J7, Canada
| | - John S Allingham
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, ON K7L 3N6, Canada
| | - Jamel Chelly
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, 67400 Illkirch, France.,CNRS UMR 7104, 67400 Illkirch, France.,INSERM U1258, 67400 Illkirch, France.,Université de Strasbourg, 67400 Illkirch, France.,Department of Biomedical and Molecular Sciences, Queen's University, Kingston, ON K7L 3N6, Canada.,Service de Diagnostic Génétique, Hôpital Civil de Strasbourg, Hôpitaux Universitaires de Strasbourg, 67000 Strasbourg, France
| |
Collapse
|
3
|
Li YF, Shi LJ, Wang P, Wang JW, Shi GY, Lee SC. Binding between ROCK1 and DCTN2 triggers diabetes‑associated centrosome amplification in colon cancer cells. Oncol Rep 2021; 46:151. [PMID: 34080666 PMCID: PMC8185503 DOI: 10.3892/or.2021.8102] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2021] [Accepted: 05/05/2021] [Indexed: 11/06/2022] Open
Abstract
Type 2 diabetes increases the risk various types of cancer and is associated with a poor prognosis therein. There is also evidence that the disease is associated with cancer metastasis. Centrosome amplification can initiate tumorigenesis with metastasis in vivo and increase the invasiveness of cancer cells in vitro. Our previous study reported that type 2 diabetes promotes centrosome amplification via the upregulation and centrosomal translocation of Rho-associated protein kinase 1 (ROCK1), which suggests that centrosome amplification is a candidate biological link between type 2 diabetes and cancer development. In the present study, functional proteomics analysis was used to further investigate the molecular pathways underlying centrosome amplification by targeting ROCK1 binding partners. High glucose, insulin and palmitic acid were used to induce centrosome amplification, and immunofluorescent staining was employed to visualize centrosomal alterations. Combined with immunoprecipitation, mass spectrometry-based proteomics analysis was used to identify ROCK1 binding proteins, and protein complex disruption was achieved by siRNA-knockdown. In total, 1,148 ROCK1 binding proteins were identified, among which 106 proteins were exclusively associated with the treated samples, 193 were only associated with the control samples, and 849 were found in both the control and treated samples. Of the proteins with evidence of centrosomal localization, Dynactin subunit 2 (DCTN2) was confirmed to be localized to the centrosomes. Treating the cells with high glucose, insulin and palmitic acid increased the protein levels of ROCK1 and DCTN2, promoted their binding with each other, and triggered centrosome amplification. Disruption of the protein complex by knocking down ROCK1 or DCTN2 expression partially attenuated centrosome amplification, while simultaneous knockdown of both proteins completely inhibited centrosome amplification. These results suggested ROCK1-DCTN2 binding as a signal for the regulation of centrosome homeostasis, which is key for diabetes-associated centrosome amplification, and enriches our knowledge of centrosome biology. Therefore, the ROCK1-DCTN2 complex may serve as a target for inhibiting centrosome amplification both in research or future therapeutic development.
Collapse
Affiliation(s)
- Yuan Fei Li
- Department of Oncology, The First Hospital, Shanxi Medical University, Taiyuan, Shanxi 030001, P.R. China
| | - Lin Jie Shi
- Department of Oncology, The First Hospital, Shanxi Medical University, Taiyuan, Shanxi 030001, P.R. China
| | - Pu Wang
- Changzhi Medical University, Changzhi, Shanxi 030001, P.R. China
| | - Jia Wen Wang
- Institute of Biomedical Sciences of The School of Life Sciences, Jiangsu Normal University, Xuzhou, Jiangsu 221116, P.R. China
| | - Guang Yi Shi
- Institute of Biomedical Sciences of The School of Life Sciences, Jiangsu Normal University, Xuzhou, Jiangsu 221116, P.R. China
| | - Shao Chin Lee
- Institute of Biomedical Sciences of The School of Life Sciences, Jiangsu Normal University, Xuzhou, Jiangsu 221116, P.R. China
| |
Collapse
|
4
|
Lei G, Xin X, Hu X. Clinical significance of kinesin family member 2A as a facilitating biomarker of disease surveillance and prognostication in cervical cancer patients. Ir J Med Sci 2021; 191:665-670. [PMID: 33797694 DOI: 10.1007/s11845-021-02510-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2020] [Accepted: 01/06/2021] [Indexed: 01/03/2023]
Abstract
BACKGROUND Cervical cancer is one of the most common and fatal malignancies among females, and biomarkers are essential for assisting in its management. Kinesin family member 2A (KIF2A) has been exhibited to be a potential maker in various cancers; however, its role in cervical cancer has yet to be reported. Therefore, we aimed to assess the expression of KIF2A and its correlation with clinicopathological characteristics as well as survival profile in cervical cancer patients. METHODS A hundred and thirty-five cervical cancer patients who underwent simple trachelectomy or radical hysterectomy were retrospectively analyzed. Tumor tissues and paired adjacent tissues were acquired, in which KIF2A mRNA and protein expressions were determined by reverse transcription quantitative polymerase chain reaction and immunohistochemistry assay, respectively. Disease-free survival (DFS) and overall survival (OS) were documented with a median follow-up duration of 28.0 months. RESULTS KIF2A protein (P < 0.001) and mRNA (P < 0.001) expressions were both upregulated in tumor tissues compared to paired adjacent tissues in cervical cancer patients. In addition, tumor tissue KIF2A protein and mRNA expressions were positively associated with lymph node metastasis (P = 0.025 and P = 0.010, respectively) and FIGO stage (P = 0.022 and P = 0.015, respectively) in cervical cancer patients. Moreover, patients with tumor tissue KIF2A high expression (mRNA and protein) displayed worse DFS (P = 0.010 and P = 0.046, respectively) and OS (P = 0.042 and P = 0.030, respectively) compared to patients with tumor tissue KIF2A low expression (mRNA and protein). CONCLUSION Tumor tissue KIF2A expression could serve as a biomarker enhancing the disease surveillance and prognostication in cervical cancer management.
Collapse
Affiliation(s)
- Gang Lei
- Department of Obstetrics and Gynecology, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, 7th Floor, Renji Building, No. 26 Shengli Street, Jiang'an District, Wuhan, 430000, China
| | - Xin Xin
- Department of Obstetrics and Gynecology, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, 7th Floor, Renji Building, No. 26 Shengli Street, Jiang'an District, Wuhan, 430000, China
| | - Xiaoji Hu
- Department of Obstetrics and Gynecology, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, 7th Floor, Renji Building, No. 26 Shengli Street, Jiang'an District, Wuhan, 430000, China.
| |
Collapse
|
5
|
Ding T, Li J, Sun J, Fan X, Shi C, Zhou D, Deng R. Association of kinesin family member 2A with increased disease risk, deteriorative clinical characteristics, and shorter survival profiles in acute myeloid leukemia. ACTA ACUST UNITED AC 2020; 54:e9173. [PMID: 33331418 PMCID: PMC7747876 DOI: 10.1590/1414-431x20209173] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2020] [Accepted: 08/20/2020] [Indexed: 12/14/2022]
Abstract
This study aimed to explore the correlation of kinesin family member 2A (KIF2A) expression with disease risk, clinical characteristics, and prognosis of acute myeloid leukemia (AML), and investigate the effect of KIF2A knockdown on AML cell activities in vitro. Bone marrow samples were collected from 176 AML patients and 40 healthy donors, and KIF2A expression was measured by real-time quantitative polymerase chain reaction. Treatment response, event-free survival (EFS), and overall survival (OS) were assessed in AML patients. In vitro, KIF2A expression in AML cell lines and CD34+ cells (from healthy donors) was measured, and the effect of KIF2A knockdown on AML cell proliferation and apoptosis in HL-60 and KG-1 cells was detected. KIF2A expression was greater in AML patients compared to healthy donors, and receiver operating characteristic curve indicated that KIF2A expression predicted increased AML risk (area under curve: 0.793 (95%CI: 0.724-0.826)). In AML patients, KIF2A expression positively correlated with white blood cells, monosomal karyotype, and high risk stratification. Furthermore, no correlation of KIF2A expression with complete remission or hematopoietic stem cell transplantation was found. Kaplan-Meier curves showed that KIF2A expression was negatively correlated with EFS and OS. In vitro experiments showed that KIF2A was overexpressed in AML cell lines (KG-1, HL-60, ME-1, and HT-93) compared to CD34+ cells, moreover, cell proliferation was reduced but apoptosis was increased by KIF2A knockdown in HL-60 and KG-1 cells. In conclusion, KIF2A showed potential to be a biomarker and treatment target in AML.
Collapse
Affiliation(s)
- Tianling Ding
- Department of Hematology, Huashan Hospital, Fudan University, Shanghai, China.,Department of Hematology, Huashan Hospital North, Fudan University, Shanghai, China
| | - Jialing Li
- Shanghai Qeejen Bio-tech Institution, Shanghai, China
| | - Jianhong Sun
- Shanghai Qeejen Bio-tech Institution, Shanghai, China
| | - Xiaoman Fan
- Shanghai Qeejen Bio-tech Institution, Shanghai, China
| | - Chunli Shi
- Shanghai Qeejen Bio-tech Institution, Shanghai, China
| | - Dong Zhou
- Shanghai Qeejen Bio-tech Institution, Shanghai, China
| | - Ruoyu Deng
- Shanghai Qeejen Bio-tech Institution, Shanghai, China
| |
Collapse
|
6
|
Identification of Kinesin Family Member 2A (KIF2A) as a Promising Therapeutic Target for Osteosarcoma. BIOMED RESEARCH INTERNATIONAL 2020; 2020:7102757. [PMID: 33204709 PMCID: PMC7655250 DOI: 10.1155/2020/7102757] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/10/2019] [Revised: 11/07/2019] [Accepted: 08/12/2020] [Indexed: 12/30/2022]
Abstract
Background Osteosarcoma is known as a type of common human bone malignancy, and more therapeutic targets are still required to combat this disease. In recent years, the involvement of KIF2A in cancer progression has been widely revealed; however, its potential effect on osteosarcoma development remains unknown. This study is to assess the KIF2A expression levels in human osteosarcoma tissues and explore its potential role in osteosarcoma development. Methods Immunohistochemical (IHC) assays were conducted to evaluate the expression levels of KIF2A in a total of 74 samples of osteosarcoma tissues and adjacent nontumor tissues. According to the staining intensity in tumor tissues, patients were divided into highly expressed and low expression KIF2A groups. The possible links between the KIF2A expression and the clinical pathological features were explored and analyzed, and the effects of KIF2A on osteosarcoma cell proliferation, migration, and invasion were detected through colony formation assay, MTT assay, wound closure assay, and transwell assay, respectively. The effects of KIF2A on tumor growth and metastasis were detected by the use of animal models. Results KIF2A was highly expressed in human osteosarcoma tissues. Meanwhile, KIF2A was obviously correlated to the tumor size (P = 0.001∗) and clinical stage (P = 0.014∗) of osteosarcoma patients. Our results also revealed that the ablation of KIF2A dramatically blocked the proliferation, migration, and invasion capacity of osteosarcoma cells in vitro and blocked tumor growth and metastasis in mice. Conclusions We investigated the involvement of KIF2A in the development and metastasis of osteosarcoma and therefore thought KIF2A as a promising therapeutic target for osteosarcoma treatment.
Collapse
|
7
|
Zhao J, Su L, Jiang J. Long Non-Coding RNA Paternally Expressed Imprinted Gene 10 (PEG10) Elevates Diffuse Large B-Cell Lymphoma Progression by Regulating Kinesin Family Member 2A (KIF2A) via Targeting MiR-101-3p. Med Sci Monit 2020; 26:e922810. [PMID: 32976381 PMCID: PMC7523416 DOI: 10.12659/msm.922810] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Background Diffuse large B-cell lymphoma (DLBCL) is a common malignant tumor in the immune system with high mortality. We investigated the functional effects of long non-coding RNA paternally expressed imprinted gene 10 (PEG10) on DLBCL progression. Material/Methods Real-time quantitative polymerase chain reaction was used to measure the level of PEG10, kinesin family member 2A (KIF2A) and microRNA-101-3p (miR-101-3p) in DLBCL tissues and cell lines. The relative protein level was detected by western blot analysis. The biological behaviors including cell proliferation, apoptosis, migration, and invasion were determined by MTT assay, flow cytometry analysis, and Transwell assays, respectively. Bioinformatics analysis and dual-luciferase reporter assay were performed to evaluate the interaction among PEG10, miR-101-3p, and KIF2A. Results PEG10 and KIF2A level were significantly upregulated, while miR-101-3p was downregulated in DLBCL tissues and cells. PEG10 positively regulated KIF2A level in DLBCL. PEG10, or KIF2A deletion significantly inhibited the proliferative, migratory, and invasive abilities of DLBCL cells and elevated cell apoptosis in DLBCL cells. KIF2A upregulation partially reversed the effects of PEG10 downregulation on cell growth, metastasis, and apoptosis in DLBCL. Moreover, PEG10 negatively regulated miR-101-3p level and miR-101-3p upregulation exerted inhibition effects on the progression of DLBCL. Besides, miR-101-3p was a target of PEG10 and miR-101-3p could directly target KIF2A. PEG10 promoted KIF2A level by sponging miR-101-3p. Conclusions Our findings revealed that PEG10 played an oncogenic role in DLBCL progression, which might be a potential target for the treatment of DLBCL.
Collapse
Affiliation(s)
- Jin Zhao
- Department of Hematology, Affiliated Shanxi Tumor Hospital of Shanxi Medical University, Taiyuan, Shanxi, China (mainland)
| | - Liping Su
- Department of Hematology, Affiliated Shanxi Tumor Hospital of Shanxi Medical University, Taiyuan, Shanxi, China (mainland)
| | - Jianjun Jiang
- Department of Palliative Medicine, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, Sichuan, China (mainland)
| |
Collapse
|
8
|
Wang Z, Liu X, Liu X, Niu D. Long Non-Coding RNA BLACAT1 Promotes the Tumorigenesis of Gastric Cancer by Sponging microRNA-149-5p and Targeting KIF2A. Cancer Manag Res 2020; 12:6629-6640. [PMID: 32801897 PMCID: PMC7402705 DOI: 10.2147/cmar.s258178] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Accepted: 07/09/2020] [Indexed: 12/14/2022] Open
Abstract
Objective Gastric cancer (GC) is a gastrointestinal tumor. This study is aimed to explore the regulatory mechanism of long non-coding RNA BLACAT1 (BLACAT1)/microRNA-149-5p (miR-149-5p)/KIF2A cascade on GC. Methods The expression of BLACAT1, miR-149-5p and KIF2A in GC was detected by qRT-PCR. The proliferation, migration and invasion of GC cells in vitro were analyzed by MTT, wound-healing and transwell assay, respectively. The xenograft tumor model was constructed in nude mice to confirm the inhibition effect of BLACAT1 knockdown on GC in vivo. Then, dual-luciferase reporter assay was used to detect the interactions among BLACAT1, miR-149-5p and KIF2A. Western blot assay was performed to determine the protein expression of KIF2A. Results The expression of BLACAT1 and KIF2A was up-regulated in GC, but miR-149-5p expression was down-regulated. Silencing of BLACAT1 retarded the proliferation, migration and invasion of GC cells in vitro and the growth of tumor xenograft in vivo. Moreover, BLACAT1 acted as the molecular sponge of miR-149-5p to up-regulate KIF2A expression. At last, feedback experiments suggested that BLACAT1 accelerated the proliferation, migration and invasion of GC cells by regulating miR-149-5p/KIF2A axis. Conclusion BLACAT1 facilitated the tumorigenesis of GC through regulating miR-149-5p/KIF2A axis, which indicated BLACAT1/miR-149-5p/KIF2A cascade may be a new therapeutic target.
Collapse
Affiliation(s)
- Zhengkun Wang
- Department of Gastrointestinal Surgery, Affiliated Hospital of Qingdao University, Qingdao City, Shandong Province 266000, People's Republic of China
| | - Xichun Liu
- Department of Gastrointestinal Surgery, Affiliated Hospital of Qingdao University, Qingdao City, Shandong Province 266000, People's Republic of China
| | - Xiaolei Liu
- Department of General Surgery, Affiliated Hospital of Qingdao University, Qingdao City, Shandong Province 266000, People's Republic of China
| | - Dongguang Niu
- Department of Gastrointestinal Surgery, Affiliated Hospital of Qingdao University, Qingdao City, Shandong Province 266000, People's Republic of China
| |
Collapse
|
9
|
Li X, Shu K, Wang Z, Ding D. Prognostic significance of KIF2A and KIF20A expression in human cancer: A systematic review and meta-analysis. Medicine (Baltimore) 2019; 98:e18040. [PMID: 31725680 PMCID: PMC6867763 DOI: 10.1097/md.0000000000018040] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND The kinesin family (KIF) is reported to be aberrantly expressed and significantly correlated with survival outcomes in patients with various cancers. This meta-analysis was carried out to quantitatively evaluate the prognostic values of partial KIF members in cancer patients. METHODS Two well-known KIF members, KIF2A and KIF20A, were investigated to evaluate their potential values as novel prognostic biomarkers in human cancer. A comprehensive literature search was carried out of the PubMed, EMBASE, Cochrane Library, and Web of Science databases up to April 2019. Pooled hazard ratios (HRs) and odds ratios (ORs) with 95% confidence intervals (CIs) were calculated to assess the association of KIF2A and KIF20A expression with overall survival (OS) and clinicopathological parameters. RESULTS Twenty-five studies involving 7262 patients were finally incorporated, including nine about KIF2A and sixteen about KIF20A. Our results indicated that patients with high expression of KIF2 and KIF20A tended to have shorter OS than those with low expression (HR = 2.23, 95% CI = 1.87-2.65, P < .001; HR = 1.77, 95% CI = 1.57-1.99, P < .001, respectively). Moreover, high expression of these 2 KIF members was significantly associated with advanced clinical stage (OR = 1.98, 95% CI: 1.57-2.50, P < .001; OR = 2.63, 95% CI: 2.03-3.41, P < .001, respectively), positive lymph node metastasis (OR = 2.32, 95% CI: 1.65-3.27, P < .001; OR = 2.13, 95% CI: 1.59-2.83, P < .001, respectively), and distant metastasis (OR = 2.20, 95% CI: 1.21-3.99, P = .010; OR = 5.25, 95% CI: 2.82-9.77, P < .001, respectively); only high KIF20A expression was related to poor differentiation grade (OR = 1.82, 95% CI: 1.09-3.07, P = .023). CONCLUSIONS High expression of KIF2 and KIF20A in human cancer was significantly correlated with worse prognosis and unfavorable clinicopathological features, suggesting that these 2 KIF members can be used as prognostic biomarkers for different types of tumors. PROSPERO REGISTRATION NUMBER CRD42019134928.
Collapse
Affiliation(s)
- Xing Li
- Department of Urology, People's Hospital of Zhengzhou University
| | - Kunpeng Shu
- Department of Urology, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, School of Clinical Medicine, Henan University, Zhengzhou, Henan, China
| | - Zhifeng Wang
- Department of Urology, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, School of Clinical Medicine, Henan University, Zhengzhou, Henan, China
| | - Degang Ding
- Department of Urology, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, School of Clinical Medicine, Henan University, Zhengzhou, Henan, China
| |
Collapse
|
10
|
Zhang L, Zhu G, Wang X, Liao X, Huang R, Huang C, Huang P, Zhang J, Wang P. Genome‑wide investigation of the clinical significance and prospective molecular mechanisms of kinesin family member genes in patients with lung adenocarcinoma. Oncol Rep 2019; 42:1017-1034. [PMID: 31322267 PMCID: PMC6667890 DOI: 10.3892/or.2019.7236] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2019] [Accepted: 07/11/2019] [Indexed: 12/24/2022] Open
Abstract
The current study aimed to identify the potential clinical significance and molecular mechanisms of kinesin (KIF) family member genes in lung adenocarcinoma (LUAD) using genome-wide RNA sequencing (RNA-seq) datasets derived from The Cancer Genome Atlas (TCGA) database. Clinical parameters and RNA-seq data of patients with LUAD from the TCGA database enabled the assessment of the clinical significance of KIF genes, while the potential mechanisms of their interactions in LUAD were investigated by gene set enrichment analysis (GSEA). A gene signature with potential prognostic value was constructed via a stepwise multivariable Cox analysis. In total, 23 KIF genes were identified to be differentially expressed genes (DEGs) between the LUAD tumor and adjacent non-cancerous tissues. Of these, 8 differentially expressed KIF genes were strongly found to be strongly associated with the overall survival of patients with LUAD. Three of these genes were found to be able to be grouped as a potential prognostic gene signature. Patients with higher risk scores calculated using this gene signature were found to have a markedly higher risk of mortality (adjusted P=0.003; adjusted HR, 1.576; 95% CI, 1.166–2.129). Time-dependent receiver operating characteristic analysis indicated that this prognostic signature was able to accurately predict patient prognosis with an area under curve of 0.636, 0.643,0.665, 0.670 and 0.593 for the 1-, 2-, 3-, 4- and 5-year survival, respectively. This prognostic gene signature was identified as an independent risk factor for LUAD and was able to more accurately predict prognosis in comparison to other known clinical parameters, as shown via comprehensive survival analysis. GSEA enrichment revealed that that KIF14, KIF18B and KIF20A mediated basic cell physiology through the regulation of the cell cycle, DNA replication, and DNA repair biological processes and pathways. On the whole, the findings of this study identified 23 KIF genes that were DEGs between LUAD tumor and adjacent non-cancerous tissues. In total, 8 of these genes had the potential to function as prognostic and diagnostic biomarkers in patients with LUAD.
Collapse
Affiliation(s)
- Linbo Zhang
- Department of Health Management and Division of Physical Examination, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region 530021, P.R. China
| | - Guangzhi Zhu
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region 530021, P.R. China
| | - Xiangkun Wang
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region 530021, P.R. China
| | - Xiwen Liao
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region 530021, P.R. China
| | - Rui Huang
- Department of Hematology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region 530021, P.R. China
| | - Chunxia Huang
- Department of Health Management and Division of Physical Examination, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region 530021, P.R. China
| | - Ping Huang
- Department of Health Management and Division of Physical Examination, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region 530021, P.R. China
| | - Jianquan Zhang
- Department of Respiratory Medicine, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region 530021, P.R. China
| | - Peng Wang
- Department of Health Management and Division of Physical Examination, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region 530021, P.R. China
| |
Collapse
|