1
|
Song Y, Zhang P, Bhushan S, Wu X, Zheng H, Yang Y. The Critical Role of Inhibitor of Differentiation 4 in Breast Cancer: From Mammary Gland Development to Tumor Progression. Cancer Med 2025; 14:e70856. [PMID: 40186425 PMCID: PMC11971571 DOI: 10.1002/cam4.70856] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2024] [Revised: 03/12/2025] [Accepted: 03/26/2025] [Indexed: 04/07/2025] Open
Abstract
Inhibitor of differentiation 4 (ID4) is a highly conserved DNA-binding inhibitory protein of mammals, and its main role is to bind basic helix-loop-helix (b-HLH) so that it loses its DNA-binding activity, which in turn regulates the transcription of key genes, regulating cell differentiation and proliferation as the physiological function. Breast tissue is a highly heterogeneous tissue organ with a strong capacity for remodeling and differentiation, and studies of breast carcinogenesis suggest that the mechanisms regulating the differentiation of breast tissue interact critically with tumorigenesis. The expression level of ID4 and its regulatory mechanism play a crucial role in the study of breast cancer, but its oncogenic or oncostatic role has not yet been unanimously identified, and its regulatory mechanism in breast cancer still needs to be further elucidated. This review summarizes and analyzes the relevant studies of ID4 and the research progress in breast cancer, integrating the development of breast tissue and tumorigenesis with the regulatory role of ID4, to provide some insights into develop new treatment strategies and diagnostic biomarkers.
Collapse
Affiliation(s)
- Yuhang Song
- Department of Breast SurgeryHubei Cancer Hospital, Tongji Medical College, Huazhong University of Science and Technology, Hubei Provincial Clinical Research Center for Breast Cancer, Wuhan Clinical Research Center for Breast Cancer; National key clinical specialty construction disciplineWuhanHubeiChina
| | - Panshi Zhang
- Department of Thyroid and Breast SurgeryTongji Hospital of Huazhong University of Science and TechnologyWuhanHubeiChina
| | - Sudhanshu Bhushan
- Department of Anatomy and Cell BiologyUnit of Reproductive Biology, Justus‐Liebig‐University GiessenGiessenGermany
| | - Xinhong Wu
- Department of Breast SurgeryHubei Cancer Hospital, Tongji Medical College, Huazhong University of Science and Technology, Hubei Provincial Clinical Research Center for Breast Cancer, Wuhan Clinical Research Center for Breast Cancer; National key clinical specialty construction disciplineWuhanHubeiChina
| | - Hongmei Zheng
- Department of Breast SurgeryHubei Cancer Hospital, Tongji Medical College, Huazhong University of Science and Technology, Hubei Provincial Clinical Research Center for Breast Cancer, Wuhan Clinical Research Center for Breast Cancer; National key clinical specialty construction disciplineWuhanHubeiChina
| | - Yalong Yang
- Department of Breast SurgeryHubei Cancer Hospital, Tongji Medical College, Huazhong University of Science and Technology, Hubei Provincial Clinical Research Center for Breast Cancer, Wuhan Clinical Research Center for Breast Cancer; National key clinical specialty construction disciplineWuhanHubeiChina
| |
Collapse
|
2
|
Wang B, Bao L, Li X, Sun G, Yang W, Xie N, Lei L, Chen W, Zhang H, Chen M, Zhao X, Wan X, Yuan R, Jiang H. Identification and validation of the important role of KIF11 in the development and progression of endometrial cancer. J Transl Med 2025; 23:48. [PMID: 39806429 PMCID: PMC11727483 DOI: 10.1186/s12967-025-06081-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2024] [Accepted: 01/02/2025] [Indexed: 01/16/2025] Open
Abstract
BACKGROUND Human kinesin family member 11 (KIF11) plays a vital role in regulating the cell cycle and is implicated in the tumorigenesis and progression of various cancers, but its role in endometrial cancer (EC) is still unclear. Our current research explored the prognostic value, biological function and targeting strategy of KIF11 in EC through approaches including bioinformatics, machine learning and experimental studies. METHODS The GSE17025 dataset from the GEO database was analyzed via the limma package to identify differentially expressed genes (DEGs) in EC. Functional enrichment analysis of the DEGs was conducted using Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analyses. DEGs were further screened for hub genes through protein-protein interaction (PPI) network analysis and machine learning. The role of the hub gene KIF11 in EC was analyzed using clinical data from the TCGA database. The expression of KIF11 in EC was subsequently validated in clinical samples. In vitro experiments were utilized to evaluate the effects of KIF11 on biological functions such as proliferation, migration, apoptosis, and the cell cycle in endometrial cancer cells. RESULTS A total of 877 DEGs, which are widely involved in important biological processes such as cell division, tubulin binding, and the cell cycle, were identified. Through PPI network analysis and machine learning, KIF11 was selected as the hub gene for subsequent analysis and experimental validation. An analysis of TCGA data revealed that KIF11 is highly expressed in EC and is associated with tumor grade, stage, and a low survival rate. The overexpression of KIF11 in tumor tissues was further confirmed in EC patient samples. KIF11 knockdown had inhibitory effects on cell proliferation, migration and invasion. Flow cytometry analysis revealed that KIF11 knockdown induced G2/M phase arrest and promoted apoptosis in EC cells. CONCLUSION Our study demonstrated that KIF11 was upregulated in EC and was strongly associated with a poor prognosis. Notably, we found that reduced KIF11 expression inhibited EC cell proliferation, migration and invasion. KIF11 knockdown caused more EC cells to arrest in the G2/M phase and undergo apoptosis. The findings of our study emphasized that KIF11 may be a promising prognostic biomarker and therapeutic target for EC patients.
Collapse
Affiliation(s)
- Biying Wang
- School of Clinical Laboratory Science, Guizhou Medical University, Guiyang, Guizhou, 550000, China
| | - Lunmin Bao
- School of Clinical Laboratory Science, Guizhou Medical University, Guiyang, Guizhou, 550000, China
- Department of Laboratory Medicine, Peoples' Hospital of Anshun City, Guizhou, 561000, Anshun, China
- School of Basic Medicine, Guizhou Medical University, Guizhou, 550000, Guiyang, China
| | - Xiaoduo Li
- Department of Laboratory Medicine, Peoples' Hospital of Anshun City, Guizhou, 561000, Anshun, China
| | - Guang Sun
- Department of Laboratory Medicine, Peoples' Hospital of Anshun City, Guizhou, 561000, Anshun, China
| | - Wu Yang
- Department of Gynaecology, Peoples' Hospital of Anshun City, Guizhou, 561000, Anshun, China
| | - Nanzi Xie
- Department of Pathology, Peoples' Hospital of Anshun City, Guizhou, 561000, Anshun, China
| | - Ling Lei
- Department of Gynaecology, Peoples' Hospital of Anshun City, Guizhou, 561000, Anshun, China
| | - Wei Chen
- Department of Pathology, Peoples' Hospital of Anshun City, Guizhou, 561000, Anshun, China
| | - Hailong Zhang
- Department of Laboratory Medicine, Peoples' Hospital of Anshun City, Guizhou, 561000, Anshun, China
| | - Man Chen
- Department of Gynaecology, Peoples' Hospital of Anshun City, Guizhou, 561000, Anshun, China
| | - Xing Zhao
- School of Clinical Laboratory Science, Guizhou Medical University, Guiyang, Guizhou, 550000, China
| | - Xiufang Wan
- School of Clinical Laboratory Science, Guizhou Medical University, Guiyang, Guizhou, 550000, China
- School of Basic Medicine, Guizhou Medical University, Guizhou, 550000, Guiyang, China
| | - Rui Yuan
- School of Clinical Laboratory Science, Guizhou Medical University, Guiyang, Guizhou, 550000, China
- School of Basic Medicine, Guizhou Medical University, Guizhou, 550000, Guiyang, China
| | - Hongmei Jiang
- School of Clinical Laboratory Science, Guizhou Medical University, Guiyang, Guizhou, 550000, China.
- School of Basic Medicine, Guizhou Medical University, Guizhou, 550000, Guiyang, China.
- Guizhou Nursing Vocational College, Guizhou, 550000, Guiyang, China.
| |
Collapse
|
3
|
Shi X, Liu C, Zheng W, Cao X, Li W, Zhang D, Zhu J, Zhang X, Chen Y. Proteomic Analysis Revealed the Potential Role of MAGE-D2 in the Therapeutic Targeting of Triple-Negative Breast Cancer. Mol Cell Proteomics 2024; 23:100703. [PMID: 38128647 PMCID: PMC10835320 DOI: 10.1016/j.mcpro.2023.100703] [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: 03/05/2023] [Revised: 12/13/2023] [Accepted: 12/15/2023] [Indexed: 12/23/2023] Open
Abstract
Among all the molecular subtypes of breast cancer, triple-negative breast cancer (TNBC) is the most aggressive one. Currently, the clinical prognosis of TNBC is poor because there is still no effective therapeutic target. Here, we carried out a combined proteomic analysis involving bioinformatic analysis of the proteome database, label-free quantitative proteomics, and immunoprecipitation (IP) coupled with mass spectrometry (MS) to explore potential therapeutic targets for TNBC. The results of bioinformatic analysis showed an overexpression of MAGE-D2 (melanoma antigen family D2) in TNBC. In vivo and in vitro experiments revealed that MAGE-D2 overexpression could promote cell proliferation and metastasis. Furthermore, label-free quantitative proteomics revealed that MAGE-D2 acted as a cancer-promoting factor by activating the PI3K-AKT pathway. Moreover, the outcomes of IP-MS and cross-linking IP-MS demonstrated that MAGE-D2 could interact with Hsp70 and prevent Hsp70 degradation, but evidence for their direct interaction is still lacking. Nevertheless, MAGE-D2 is a potential therapeutic target for TNBC, and blocking MAGE-D2 may have important therapeutic implications.
Collapse
Affiliation(s)
- Xiaoyu Shi
- School of Pharmacy, Nanjing Medical University, Nanjing, China
| | - Chunyan Liu
- School of Pharmacy, Nanjing Medical University, Nanjing, China
| | - Weimin Zheng
- School of Pharmacy, Nanjing Medical University, Nanjing, China
| | - Xiao Cao
- School of Pharmacy, Nanjing Medical University, Nanjing, China
| | - Wan Li
- School of Pharmacy, Nanjing Medical University, Nanjing, China
| | - Dongxue Zhang
- School of Pharmacy, Nanjing Medical University, Nanjing, China
| | - Jianhua Zhu
- School of Pharmacy, Nanjing Medical University, Nanjing, China
| | - Xian Zhang
- School of Pharmacy, Nanjing Medical University, Nanjing, China
| | - Yun Chen
- School of Pharmacy, Nanjing Medical University, Nanjing, China; State Key Laboratory of Reproductive Medicine and Offspring Health, Nanjing, China; Key Laboratory of Cardiovascular & Cerebrovascular Medicine, Nanjing, China.
| |
Collapse
|
4
|
Thankamony AP, Ramkomuth S, Ramesh ST, Murali R, Chakraborty P, Karthikeyan N, Varghese BA, Jaikumar VS, Jolly MK, Swarbrick A, Nair R. Phenotypic heterogeneity drives differential disease outcome in a mouse model of triple negative breast cancer. Front Oncol 2023; 13:1230647. [PMID: 37841442 PMCID: PMC10570535 DOI: 10.3389/fonc.2023.1230647] [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: 05/29/2023] [Accepted: 08/24/2023] [Indexed: 10/17/2023] Open
Abstract
The triple negative breast cancer (TNBC) subtype is one of the most aggressive forms of breast cancer that has poor clinical outcome and is an unmet clinical challenge. Accumulating evidence suggests that intratumoral heterogeneity or the presence of phenotypically distinct cell populations within a tumor play a crucial role in chemoresistance, tumor progression and metastasis. An increased understanding of the molecular regulators of intratumoral heterogeneity is crucial to the development of effective therapeutic strategies in TNBC. To this end, we used an unbiased approach to identify a molecular mediator of intratumoral heterogeneity in breast cancer by isolating two tumor cell populations (T1 and T2) from the 4T1 TNBC model. Phenotypic characterization revealed that the cells are different in terms of their morphology, proliferation and self-renewal ability in vitro as well as primary tumor formation and metastatic potential in vivo. Bioinformatic analysis followed by Kaplan Meier survival analysis in TNBC patients identified Metastasis associated colon cancer 1 (Macc1) as one of the top candidate genes mediating the aggressive phenotype in the T1 tumor cells. The role of Macc1 in regulating the proliferative phenotype was validated and taken forward in a therapeutic context with Lovastatin, a small molecule transcriptional inhibitor of Macc1 to target the T1 cell population. This study increases our understanding of the molecular underpinnings of intratumoral heterogeneity in breast cancer that is critical to improve the treatment of women currently living with the highly aggressive TNBC subtype.
Collapse
Affiliation(s)
- Archana P. Thankamony
- Rajiv Gandhi Centre for Biotechnology, Trivandrum, Kerala, India
- Manipal Academy of Higher Education (MAHE), Manipal, Karnataka, India
| | - Sonny Ramkomuth
- The Kinghorn Cancer Centre and Cancer Research Theme, Garvan Institute of Medical Research, Darlinghurst, NSW, Australia
| | - Shikha T. Ramesh
- Rajiv Gandhi Centre for Biotechnology, Trivandrum, Kerala, India
- Manipal Academy of Higher Education (MAHE), Manipal, Karnataka, India
| | - Reshma Murali
- Rajiv Gandhi Centre for Biotechnology, Trivandrum, Kerala, India
| | - Priyanka Chakraborty
- Centre for BioSystems Science and Engineering, Indian Institute of Science, Bangalore, India
| | | | | | | | - Mohit Kumar Jolly
- Centre for BioSystems Science and Engineering, Indian Institute of Science, Bangalore, India
| | - Alexander Swarbrick
- The Kinghorn Cancer Centre and Cancer Research Theme, Garvan Institute of Medical Research, Darlinghurst, NSW, Australia
- St Vincent’s Clinical School, Faculty of Medicine, UNSW Sydney, Darlinghurst, NSW, Australia
| | - Radhika Nair
- Rajiv Gandhi Centre for Biotechnology, Trivandrum, Kerala, India
- Centre for Human Genetics, Bangalore, India
| |
Collapse
|
5
|
Zhang J, Wei Z, Qi X, Jiang Y, Liu D, Liu K. Kinesin family member 11 promotes progression of hepatocellular carcinoma via the OCT4 pathway. Funct Integr Genomics 2023; 23:284. [PMID: 37648881 DOI: 10.1007/s10142-023-01209-7] [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: 05/21/2023] [Revised: 08/04/2023] [Accepted: 08/11/2023] [Indexed: 09/01/2023]
Abstract
Hepatocellular carcinoma (HCC) is the tumor with the second highest mortality rate worldwide. Recent research data show that KIF11, a member of the kinesin family (KIF), plays an important role in the progression of various tumors. However, its expression and molecular mechanism in HCC remain elusive. Here, we evaluated the potential role of KIF11 in HCC. The effect of KIF11 was evaluated using the hepatocellular carcinoma cell lines, LM3 and Huh7, after genetic or pharmacological treatment. Evaluating the role of KIF11 in the xenograft animal models using its specific inhibitor. The role of KIF11 was systematically evaluated using specimens obtained from the aforementioned animal and cell models after various in vivo and in vitro experiments. The clinicopathological analysis showed that KIF11 was expressed at high levels in patients with hepatocellular carcinoma. Cell experiments in vitro showed that KIF11 deficiency significantly slowed the proliferation of liver tumor cells. And in the experiment using liver cancer cells overexpressing OCT4, overexpression of OCT4 substantially increased the proliferation of tumor cells compared with tumor cells with KIF11 knockdown alone. Both in vitro cell experiment and in vivo xenotransplantation tumor experiment showed that monastrol, an inhibitor of KIF11, could effectively delay the proliferation and migration of tumor cells. Based on these results, KIF11 is expressed at high levels in hepatocellular carcinoma and promotes tumor proliferation in an OCT4-dependent manner. KIF11 may become a therapeutic target for hepatocellular carcinoma, and its inhibitor monastrol may become a clinical antitumor drug.
Collapse
Affiliation(s)
- Ju Zhang
- Department of General Surgery, The Second Xiangya Hospital of Central South University, No. 139 Renminzhong Road, Changsha, 410012, China
| | - Zuxing Wei
- Department of General Surgery, The Second Xiangya Hospital of Central South University, No. 139 Renminzhong Road, Changsha, 410012, China
| | - Xiaoyan Qi
- Department of General Surgery, The Second Xiangya Hospital of Central South University, No. 139 Renminzhong Road, Changsha, 410012, China
| | - Yuhong Jiang
- Department of General Surgery, The Second Xiangya Hospital of Central South University, No. 139 Renminzhong Road, Changsha, 410012, China
| | - Dekun Liu
- Department of General Surgery, The Second Xiangya Hospital of Central South University, No. 139 Renminzhong Road, Changsha, 410012, China
| | - Kuijie Liu
- Department of General Surgery, The Second Xiangya Hospital of Central South University, No. 139 Renminzhong Road, Changsha, 410012, China.
| |
Collapse
|
6
|
Qiu D, Zhang G, Yan X, Xiao X, Ma X, Lin S, Wu J, Li X, Wang W, Liu J, Ma Y, Ma M. Prospects of Immunotherapy for Triple-Negative Breast Cancer. Front Oncol 2022; 11:797092. [PMID: 35111680 PMCID: PMC8801574 DOI: 10.3389/fonc.2021.797092] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Accepted: 11/30/2021] [Indexed: 01/22/2023] Open
Abstract
In the classification and typing of breast cancer, triple-negative breast cancer (TNBC) is one type of refractory breast cancer, while chemotherapy stays in the traditional treatment methods. However, the impact of chemotherapy is short-lived and may lead to recurrence due to incomplete killing of tumor cells. The occurrence, development, and relapse of breast cancer are relevant to T cell dysfunction, multiplied expression of related immune checkpoint molecules (ICIs) such as programmed death receptor 1 (PD-1), programmed cell death 1 ligand 1 (PD-L1), and cytotoxic T-lymphocyte-associated antigen 4 (CTLA-4) produce immunosuppressive effect. Immunotherapy (namely, immune checkpoint inhibitors, adoptive cellular immunotherapy, CAR-T immunotherapy and some potential treatments) provides new hope in TNBC. This review focuses on the new immune strategies of TNBC patients.
Collapse
Affiliation(s)
- Dan Qiu
- School of Traditional Chinese Medicine of Jinan University, Jinan University, Guangzhou, China
| | - Guijuan Zhang
- School of Nursing of Jinan University, Jinan University, Guangzhou, China
| | - Xianxin Yan
- School of Traditional Chinese Medicine of Jinan University, Jinan University, Guangzhou, China
| | - Xinqin Xiao
- School of Traditional Chinese Medicine of Jinan University, Jinan University, Guangzhou, China
| | - Xinyi Ma
- School Public Health, Southern Medical University (No: 3210090112), Guangzhou, China
| | - Shujun Lin
- School of Traditional Chinese Medicine of Jinan University, Jinan University, Guangzhou, China
| | - Jieyan Wu
- School of Traditional Chinese Medicine of Jinan University, Jinan University, Guangzhou, China
| | - Xinyuan Li
- School of Medicine, Jinan University, Guangzhou, China
| | - Wandi Wang
- School of Medicine, Jinan University, Guangzhou, China
| | - Junchen Liu
- School of Medicine, Jinan University, Guangzhou, China
| | - Yi Ma
- Department of Cellular Biology, Institute of Biomedicine, National Engineering, Research Center of Genetic Medicine, Key Laboratory of Bioengineering Medicine of Guangdong Province, The National Demonstration Center for Experimental Education of Life Science and Technology, Jinan University, Guangzhou, China
| | - Min Ma
- School of Traditional Chinese Medicine of Jinan University, Jinan University, Guangzhou, China.,The First Affiliated Hospital of Jinan University, Jinan University, Guangzhou, China
| |
Collapse
|
7
|
Gangapuram M, Mazzio EA, Redda KK, Soliman KFA. Transcriptome Profile Analysis of Triple-Negative Breast Cancer Cells in Response to a Novel Cytostatic Tetrahydroisoquinoline Compared to Paclitaxel. Int J Mol Sci 2021; 22:ijms22147694. [PMID: 34299315 PMCID: PMC8306781 DOI: 10.3390/ijms22147694] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Revised: 07/09/2021] [Accepted: 07/16/2021] [Indexed: 12/13/2022] Open
Abstract
The absence of chemotherapeutic target hormone receptors in breast cancer is descriptive of the commonly known triple-negative breast cancer (TNBC) subtype. TNBC remains one of the most aggressive invasive breast cancers, with the highest mortality rates in African American women. Therefore, new drug therapies are continually being explored. Microtubule-targeting agents such as paclitaxel (Taxol) interfere with microtubules dynamics, induce mitotic arrest, and remain a first-in-class adjunct drug to treat TNBC. Recently, we synthesized a series of small molecules of substituted tetrahydroisoquinolines (THIQs). The lead compound of this series, with the most potent cytostatic effect, was identified as 4-Ethyl-N-(7-hydroxy-3,4-dihydroisoquinolin-2(1H)-yl) benzamide (GM-4-53). In our previous work, GM-4-53 was similar to paclitaxel in its capacity to completely abrogate cell cycle in MDA-MB-231 TNBC cells, with the former not impairing tubulin depolymerization. Given that GM-4-53 is a cytostatic agent, and little is known about its mechanism of action, here, we elucidate differences and similarities to paclitaxel by evaluating whole-transcriptome microarray data in MDA-MB-231 cells. The data obtained show that both drugs were cytostatic at non-toxic concentrations and caused deformed morphological cytoskeletal enlargement in 2D cultures. In 3D cultures, the data show greater core penetration, observed by GM-4-53, than paclitaxel. In concentrations where the drugs entirely blocked the cell cycle, the transcriptome profile of the 48,226 genes analyzed (selection criteria: (p-value, FDR p-value < 0.05, fold change −2< and >2)), paclitaxel evoked 153 differentially expressed genes (DEGs), GM-4-53 evoked 243 DEGs, and, of these changes, 52/153 paclitaxel DEGs were also observed by GM-4-53, constituting a 34% overlap. The 52 DEGS analysis by String database indicates that these changes involve transcripts that influence microtubule spindle formation, chromosome segregation, mitosis/cell cycle, and transforming growth factor-β (TGF-β) signaling. Of interest, both drugs effectively downregulated “inhibitor of DNA binding, dominant negative helix-loop-helix” (ID) transcripts; ID1, ID3 and ID4, and amphiregulin (AREG) and epiregulin (EREG) transcripts, which play a formidable role in cell division. Given the efficient solubility of GM-4-53, its low molecular weight (MW; 296), and capacity to penetrate a small solid tumor mass and effectively block the cell cycle, this drug may have future therapeutic value in treating TNBC or other cancers. Future studies will be required to evaluate this drug in preclinical models.
Collapse
|
8
|
Li Z, Yu B, Qi F, Li F. KIF11 Serves as an Independent Prognostic Factor and Therapeutic Target for Patients With Lung Adenocarcinoma. Front Oncol 2021; 11:670218. [PMID: 33968780 PMCID: PMC8103954 DOI: 10.3389/fonc.2021.670218] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2021] [Accepted: 03/24/2021] [Indexed: 12/31/2022] Open
Abstract
Background Lung adenocarcinoma (LUAD) is challenging in clinical practice due to the poor understanding of molecular mechanisms and limited therapeutic targets. Herein, the work aimed to use bioinformatics to identify a promising molecular target for LUAD therapy. Methods Differentially expressed genes (DEGs) from the Cancer Genome Atlas (TCGA) dataset were used for a weighted gene co-expression network analysis (WGCNA) to screen the hub gene. After a prognostic estimation with meta-analysis and COX regression analysis, we performed a function analysis on the corresponding gene. The ESTIMATE and CIBERSORT methods were adopted to analyze the association of the hub gene with the tumor microenvironment (TME). A cohort of functional assays was conducted to establish the functional roles of the hub gene in A549 and PC-9 cells. Results Our screen identified KIF11 as a prognostic factor, which indicated the poor overall survival and the worse progression-free survival in LUAD patients. Additionally, KIF11 was primarily involved in cell cycle, TME alteration and tumor-infiltrating immune cells proportions. KIF11 knockdown exerted inhibitory effects on cell proliferation, migration, and invasion. Results of the flow cytometry analysis revealed that KIF11 knockdown induced a G2/M phase arrest and improved apoptosis in LUAD cells. Conclusions KIF11 is essential for LUAD cell proliferation and metastasis, and it may serve as an independent prognostic factor as well as a promising therapeutic target for LUAD patients.
Collapse
Affiliation(s)
- Zhaodong Li
- Department of Pathogenobiology, The Key Laboratory of Zoonosis, Chinese Ministry of Education, College of Basic Medicine, Jilin University, Changchun, China
| | - Bingxin Yu
- Department of Ultrasonography, The Third Hospital of Jilin University, Changchun, China
| | - Fangyuan Qi
- Department of Pathogenobiology, The Key Laboratory of Zoonosis, Chinese Ministry of Education, College of Basic Medicine, Jilin University, Changchun, China
| | - Fan Li
- Department of Pathogenobiology, The Key Laboratory of Zoonosis, Chinese Ministry of Education, College of Basic Medicine, Jilin University, Changchun, China.,The Key Laboratory for Bionics Engineering, Ministry of Education, China, Jilin University, Changchun, China.,Engineering Research Center for Medical Biomaterials of Jilin Province, Jilin University, Changchun, China.,Key Laboratory for Biomedical Materials of Jilin Province, Jilin University, Changchun, China.,State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, Urumqi, China
| |
Collapse
|