1
|
Xu Y, Li C, Yin H, Nowsheen S, Xu X, Kang W, Liu X, Chen L, Lou Z, Yi J, Deng M. STK39-mediated amplification of γ-H2A.X promotes homologous recombination and contributes to PARP inhibitor resistance. Nucleic Acids Res 2024; 52:13881-13895. [PMID: 39588777 DOI: 10.1093/nar/gkae1099] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Revised: 09/17/2024] [Accepted: 10/28/2024] [Indexed: 11/27/2024] Open
Abstract
The phosphorylation of histone H2A.X into γH2A.X is a crucial early event in the DNA damage response, marking DNA damage sites and initiating repair processes. While ATM kinase is traditionally recognized as the primary mediator of H2A.X phosphorylation, our study identifies serine/threonine kinase 39 (STK39) as a novel enhancer of this critical signaling pathway. We demonstrate that after DNA damage, STK39 undergoes phosphorylation by the ATM kinase, facilitating its interaction with the Mre11-Rad50-Nbs1 complex and subsequent recruitment to chromatin. This recruitment enables STK39 to further phosphorylate H2A.X, thus amplifying γH2A.X production and promoting homologous recombination repair. Notably, we observe a significant upregulation of STK39 in pancreatic adenocarcinoma (PAAD) tissues, correlating with heightened resistance to PARPi therapy. Furthermore, we demonstrate the synergistic efficacy of combining STK39 inhibition with PARP inhibitors in suppressing and reversing PAAD growth. This study not only provides new insights into the molecular dynamics of H2A.X phosphorylation but also highlights the therapeutic potential of targeting STK39 to enhance PARPi sensitivity in PAAD (created with BioRender).
Collapse
Affiliation(s)
- Yi Xu
- State Key Laboratory of Molecular Oncology and Department of Radiation Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Panjiayuan Nanli No17, Chaoyang District, Beijing 100021, China
| | - Changying Li
- State Key Laboratory of Molecular Oncology and Department of Radiation Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Panjiayuan Nanli No17, Chaoyang District, Beijing 100021, China
| | - Huan Yin
- State Key Laboratory of Molecular Oncology and Department of Radiation Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Panjiayuan Nanli No17, Chaoyang District, Beijing 100021, China
| | - Somaira Nowsheen
- Department of Dermatology, University of California San Diego, 9500 Gilman Drive, La Jolla, San Diego, CA 92122, USA
| | - Xin Xu
- State Key Laboratory of Molecular Oncology and Department of Radiation Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Panjiayuan Nanli No17, Chaoyang District, Beijing 100021, China
| | - Wenjuan Kang
- State Key Laboratory of Molecular Oncology and Department of Radiation Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Panjiayuan Nanli No17, Chaoyang District, Beijing 100021, China
| | - Xin Liu
- State Key Laboratory of Molecular Oncology and Department of Radiation Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Panjiayuan Nanli No17, Chaoyang District, Beijing 100021, China
| | - Lifeng Chen
- Department of Gynecology, the First Affiliated Hospital, School of Medicine, Zhejiang University, No.79 Qingchun Road, Shangcheng District, Hangzhou 310003, China
| | - Zhenkun Lou
- Department of Oncology, Mayo Clinic, 200 First St SW, Rochester, MN 55905, USA
| | - Junlin Yi
- State Key Laboratory of Molecular Oncology and Department of Radiation Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Panjiayuan Nanli No17, Chaoyang District, Beijing 100021, China
| | - Min Deng
- State Key Laboratory of Molecular Oncology and Department of Radiation Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Panjiayuan Nanli No17, Chaoyang District, Beijing 100021, China
| |
Collapse
|
2
|
Naghib SM, Ahmadi B, Mikaeeli Kangarshahi B, Mozafari MR. Chitosan-based smart stimuli-responsive nanoparticles for gene delivery and gene therapy: Recent progresses on cancer therapy. Int J Biol Macromol 2024; 278:134542. [PMID: 39137858 DOI: 10.1016/j.ijbiomac.2024.134542] [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: 04/17/2024] [Revised: 08/02/2024] [Accepted: 08/04/2024] [Indexed: 08/15/2024]
Abstract
Recent cancer therapy research has found that chitosan (Ch)-based nanoparticles show great potential for targeted gene delivery. Chitosan, a biocompatible and biodegradable polymer, has exceptional properties, making it an ideal carrier for therapeutic genes. These nanoparticles can respond to specific stimuli like pH, temperature, and enzymes, enabling precise delivery and regulated release of genes. In cancer therapy, these nanoparticles have proven effective in delivering genes to tumor cells, slowing tumor growth. Adjusting the nanoparticle's surface, encapsulating protective agents, and using targeting ligands have also improved gene delivery efficiency. Smart nanoparticles based on chitosan have shown promise in improving outcomes by selectively releasing genes in response to tumor conditions, enhancing targeted delivery, and reducing off-target effects. Additionally, targeting ligands on the nanoparticles' surface increases uptake and effectiveness. Although further investigation is needed to optimize the structure and composition of these nanoparticles and assess their long-term safety, these advancements pave the way for innovative gene-focused cancer therapies.
Collapse
Affiliation(s)
- Seyed Morteza Naghib
- Nanotechnology Department, School of Advanced Technologies, Iran University of Science and Technology (IUST), Tehran 1684613114, Iran.
| | - Bahar Ahmadi
- Biomaterials and Tissue Engineering Research Group, Interdisciplinary Technologies Department, Breast Cancer Research Center, Motamed Cancer Institute, ACECR, Tehran, Iran
| | - Babak Mikaeeli Kangarshahi
- State Key Laboratory of Structure Analysis for Industrial Equipment, Department of Engineering Mechanics, Dalian University of Technology, Dalian, China
| | - M R Mozafari
- Australasian Nanoscience and Nanotechnology Initiative (ANNI), Monash University LPO, Clayton, VIC 3168, Australia
| |
Collapse
|
3
|
Liang QJ, Long QQ, Tian FQ, Long XD. Progress in research of polo-like kinase 1 in hepatocellular carcinoma. Shijie Huaren Xiaohua Zazhi 2024; 32:652-659. [DOI: 10.11569/wcjd.v32.i9.652] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Revised: 09/08/2024] [Accepted: 09/19/2024] [Indexed: 09/28/2024] Open
Abstract
Polo-like kinase 1 (PLK1) is a protein kinase that regulates the cell cycle, and it has been found that PLK1 mediates the regulation of signaling pathways associated with hepatocellular carcinoma (HCC) development, thereby affecting the biological behaviors of hepatic tumor cells such as cell proliferation, migration, and invasion. Therefore, PLK1 may be a very promising target for the treatment of HCC. This article reviews the relevant signaling pathways of PLK1 in HCC development and PLK1 inhibitors in the treatment of HCC.
Collapse
Affiliation(s)
- Qiu-Ju Liang
- Clinicopathological Diagnosis and Research Center, The Affiliated Hospital of Youjiang Medical University for Nationalities, Baise 533000, Guangxi Zhuang Autonomous Region, China
- Graduate School of Youjiang Medical University for Nationalities, Baise 533000, Guangxi Zhuang Autonomous Region, China
| | - Qin-Qin Long
- Clinicopathological Diagnosis and Research Center, The Affiliated Hospital of Youjiang Medical University for Nationalities, Baise 533000, Guangxi Zhuang Autonomous Region, China
- The Key Laboratory of Tumor Molecular Pathology of Guangxi Higher Education Institutes, Baise 533000, Guangxi Zhuang Autonomous Region, China
| | - Feng-Qin Tian
- Clinicopathological Diagnosis and Research Center, The Affiliated Hospital of Youjiang Medical University for Nationalities, Baise 533000, Guangxi Zhuang Autonomous Region, China
- The Key Laboratory of Tumor Molecular Pathology of Guangxi Higher Education Institutes, Baise 533000, Guangxi Zhuang Autonomous Region, China
| | - Xi-Dai Long
- Clinicopathological Diagnosis and Research Center, The Affiliated Hospital of Youjiang Medical University for Nationalities, Baise 533000, Guangxi Zhuang Autonomous Region, China
| |
Collapse
|
4
|
Zhou Q, Ge Y, Ma S, Xiong Z, Wang Y, Li L, Li L, Chao Z, Zhang J, Li T, Wu Z, Gao Y, Qu G, Dong H, Wang Z, Jing W, Chen G. PDIA2 is associated with the prognosis of prostate cancer, and downregulation of PDIA2 delays the progression of prostate cancer cells. Sci Rep 2024; 14:22064. [PMID: 39333312 PMCID: PMC11436862 DOI: 10.1038/s41598-024-73361-4] [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: 06/21/2024] [Accepted: 09/17/2024] [Indexed: 09/29/2024] Open
Abstract
Protein Disulfide-Isomerase A2 (PDIA2) is a gene that encodes proteins, responsible for protein folding and modification within cells. The development and course of many disorders are intimately linked to the aberrant expression of PDIA2. Nevertheless, more research is necessary to fully understand PDIA2's biological significance in pan-cancer, notably in prostate cancer (PCa). PDIA2 expression is elevated in various tumors and closely related to patient prognosis. Patients with prostate cancer who express PDIA2 high in particular have a bad prognosis in terms of progression-free survival. In addition, the upregulation of PDIA2 expression in prostate cancer patients is accompanied by higher Gleason scores, advanced tumor staging, lymph node metastasis, and elevated PSA levels. Detailed experiments further demonstrate that PDIA2 is a carcinogenic gene affecting prostate cancer cells' response to dasatinib therapy. For patients with prostate cancer, there is a clear positive connection between the expression level of PDIA2 and a bad prognosis. The prostate cancer treatment efficacy of dasatinib is hampered by PDIA2, which is intimately linked to the growth, invasion, and metastasis of PCa cells. In summary, our research highlights the potential of PDIA2 as a biomarker for the diagnosis and management of PCa.
Collapse
Affiliation(s)
- Qiang Zhou
- Department of Urology, Qinghai University Affiliated Hospital, Qinghai University Medical College, Xining, China
| | - Yue Ge
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Sheng Ma
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zezhong Xiong
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yanan Wang
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Le Li
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ling Li
- Department of Urology, Qinghai University Affiliated Hospital, Qinghai University Medical College, Xining, China
| | - Zheng Chao
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Junbiao Zhang
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Tengfei Li
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zixi Wu
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yuan Gao
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Guanyu Qu
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Haoxiao Dong
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zhihua Wang
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Wang Jing
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Guojun Chen
- Department of Urology, Qinghai University Affiliated Hospital, Qinghai University Medical College, Xining, China.
| |
Collapse
|
5
|
Peng L, Zhang Z, Du W, Zhu J, Duan W. Proteomic and Phosphoproteomic analysis of thyroid papillary carcinoma: Identification of potential biomarkers for metastasis. J Proteomics 2024; 306:105260. [PMID: 39029786 DOI: 10.1016/j.jprot.2024.105260] [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: 11/23/2023] [Revised: 07/14/2024] [Accepted: 07/15/2024] [Indexed: 07/21/2024]
Abstract
Thyroid cancer has emerged as the most rapidly proliferating solid neoplasm. In this study, we included a cohort of patients who underwent sonographic assessment and surgical intervention at the Sir Run Run Shaw Hospital, associated with the School of Medicine at Zhejiang University, spanning from January 2019 to June 2020. Stratification of cases was based on a combination of preoperative ultrasonographic evaluations and postoperative histopathological diagnoses, resulting in three distinct groups: high-risk papillary thyroid carcinoma (PTC) labeled as C1, low-risk PTC designated as C2, and a control group (N) composed of benign thyroid tissue adjacent to the carcinoma. Proteomic and phosphoproteomic analyses were conducted on PTC specimens. The comparative assessment revealed that proteins up-regulated in the C1/N and C2/N groups were predominantly involved in functions such as amino acid binding, binding of phosphorylated compounds, and serine protease activity. Notably, proteins like NADH dehydrogenase, ATP synthase, oxidoreductases, and iron ion channels were significantly elevated in the C1 versus C2 comparative group. Through meticulous analysis of differential expression multiples, statistical significance, and involvement in metabolic pathways, this study identified eight potential biomarkers pertinent to PTC metastasis diagnostics, encompassing phosphorylated myosin 10, phosphorylated proline-directed protein kinase, leucine tRNA synthetase, 2-oxo-isovalerate dehydrogenase, succinic semialdehyde dehydrogenase, ADP/ATPtranslocase, pyruvate carboxylase, and fibrinogen. Therapeutic assays employing metformin, an AMP-activated protein kinase (AMPK) activator, alongside the phosphorylation-specific inhibitor ML-7 targeting Myosin10, demonstrated attenuated cellular proliferation, migration, and invasion capabilities in thyroid cancer cells, accompanied by a reduction in amino acid pools. Cellular colocalization and interaction studies elucidated that AMPK activation imposes an inhibitory influence on Myosin10 levels. The findings of this research corroborate the utility of proteomic and phosphoproteomic platforms in the identification of metastatic markers for PTC and suggest that modulation of AMPK activity, coupled with the inhibition of Myosin10 phosphorylation, may forge novel therapeutic avenues in the management of thyroid carcinoma. SIGNIFICANCE: The significance of our research lies in its potential to transform the current understanding and management of thyroid papillary carcinoma (PTC), particularly in its metastatic form. By integrating both proteomic and phosphoproteomic analyses, our study not only sheds light on the molecular alterations associated with PTC but also identifies eight novel biomarkers that could serve as indicators of metastatic potential.
Collapse
Affiliation(s)
- Lingyao Peng
- Hangzhou TCM Hospital Affiliated to Zhejiang Chinese Medical University, Hangzhou 310007, China
| | - Zhenxian Zhang
- Hangzhou TCM Hospital Affiliated to Zhejiang Chinese Medical University, Hangzhou 310007, China
| | - Wei Du
- Hangzhou Institute of Standardization, Hangzhou 310000, China
| | - Jiang Zhu
- Women's Hospital School of Medicine Zhejiang University, 310006 Hangzhou, China.
| | - Wenkai Duan
- Hangzhou Vocational and Technical College, Hangzhou 310018, China.
| |
Collapse
|
6
|
Li C, Xin H, Hao J, Miao Y. Decreasing of serine/threonine kinase 39 has tumour inhibiting effects on acute myeloid leukaemia by impacting the PI3K/AKT and Wnt/β-catenin signalling cascades. Toxicol Appl Pharmacol 2024; 489:116982. [PMID: 38821216 DOI: 10.1016/j.taap.2024.116982] [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: 01/18/2024] [Revised: 05/26/2024] [Accepted: 05/28/2024] [Indexed: 06/02/2024]
Abstract
Serine/threonine kinase 39 (STK39) has been identified as a key regulator of tumour progression. However, whether STK39 plays a role in acute myeloid leukaemia (AML) remains undetermined. This work explored the expression and functions of STK39 in AML. STK39 was found to be overexpressed in AML and was negatively correlated with overall survival. Functionally, silencing STK39 inhibited cell proliferation, promoted cell differentiation and induced cell cycle arrest and apoptosis. The tumour inhibiting effects of STK39 downregulation were also verified by an in vivo xenograft tumour assay. Mechanistically, STK39 was closely related to the PI3K/AKT and Wnt/β-catenin signalling cascades in AML. Silencing of STK39 had suppressive effects on the PI3K/AKT and Wnt/β-catenin signalling cascades. The suppressive effect of STK39 silencing on the Wnt/β-catenin signalling cascade was significantly reversed when PI3K/AKT was reactivated. When β-catenin was re-expressed, the tumour-inhibiting effects caused by STK39 silencing were significantly eliminated. Therefore, STK39 plays a crucial role in AML and could be targeted for potential therapeutic purposes in treating AML.
Collapse
Affiliation(s)
- Chengliang Li
- Department of General Practice, Guangzhou First People's Hospital, Guangzhou 510180, China
| | - Hong Xin
- Department of Cardiovasology, Guangzhou First People's Hospital, Guangzhou 510180, China
| | - Jiajia Hao
- Department of General Practice, Guangzhou First People's Hospital, Guangzhou 510180, China
| | - Yudi Miao
- Department of Hematology, Shaanxi Provincial People's Hospital, Xi'an 710000, China.
| |
Collapse
|
7
|
Wei Y, Ma L, Peng Q, Lu L. Establishing an oxidative stress mitochondria-related prognostic model in hepatocellular carcinoma based on multi-omics characteristics and machine learning computational framework. Discov Oncol 2024; 15:287. [PMID: 39014263 PMCID: PMC11252104 DOI: 10.1007/s12672-024-01147-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/05/2024] [Accepted: 07/05/2024] [Indexed: 07/18/2024] Open
Abstract
Hepatocellular carcinoma (HCC) has high incidence and mortality rates worldwide. Damaged mitochondria are characterized by the overproduction of reactive oxygen species (ROS), which can promote cancer development. The prognostic value of the interplay between mitochondrial function and oxidative stress in HCC requires further investigation. Gene expression data of HCC samples were collected from The Cancer Genome Atlas (TCGA), Gene Expression Omnibus (GEO) and International Cancer Genome Consortium (ICGC). We screened prognostic oxidative stress mitochondria-related (OSMT) genes at the bulk transcriptome level. Based on multiple machine learning algorithms, we constructed a consensus oxidative stress mitochondria-related signature (OSMTS), which contained 26 genes. In addition, we identified six of these genes as having a suitable prognostic value for OSMTS to reduce the difficulty of clinical application. Univariate and multivariate analyses verified the OSMTS as an independent prognostic factor for overall survival (OS) in HCC patients. The OSMTS-related nomogram demonstrated to be a powerful tool for the clinical diagnosis of HCC. We observed differences in biological function and immune cell infiltration in the tumor microenvironment between the high- and low-risk groups. The highest expression of the OSMTS was detected in hepatocytes at the single-cell transcriptome level. Hepatocytes in the high- and low-risk groups differed significantly in terms of biological function and intercellular communication. Moreover, at the spatial transcriptome level, high expression of OSMTS was mainly in regions enriched in hepatocytes and B cells. Potential drugs targeting specific risk subgroups were identified. Our study revealed that the OSMTS can serve as a promising tool for prognosis prediction and precise intervention in HCC patients.
Collapse
Affiliation(s)
- Yitian Wei
- Department of Medical Oncology, the Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou, China
| | - Lujuan Ma
- Department of Medical Oncology, the Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou, China
| | - Qian Peng
- Department of Medical Oncology, the Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou, China
| | - Lin Lu
- Department of Medical Oncology, the Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou, China.
| |
Collapse
|
8
|
Liu Z, Zhang Y, Yu L, Zhang Z, Li G. A miR-361-5p/ ORC6/ PLK1 axis regulates prostate cancer progression. Exp Cell Res 2024; 440:114130. [PMID: 38885805 DOI: 10.1016/j.yexcr.2024.114130] [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: 04/17/2024] [Revised: 06/08/2024] [Accepted: 06/09/2024] [Indexed: 06/20/2024]
Abstract
Prostate cancer (PCa) is the most prevalent malignant tumor of the genitourinary system, and metastatic disease has a significant impact on the prognosis of PCa patients. As a result, knowing the processes of PCa development can help patients achieve better outcomes. Here, we investigated the expression and function of ORC6 in PCa. Our findings indicated that ORC6 was elevated in advanced PCa tissues. Patients with PCa who exhibited high levels of ORC6 had a poor prognosis. Following that, we investigated the function of ORC6 in PCa progression using a variety of functional experiments both in vivo and in vitro, and discovered that ORC6 knockdown inhibited PCa cell proliferation, growth, and migration. Furthermore, RNA-seq was employed to examine the molecular mechanism of PCa progression. The results revealed that ORC6 might promote the expression of PLK1, a serine/threonine kinase in PCa cells. We also discovered that ORC6 as a novel miR-361-5p substrate using database analysis, and miR-361-5p was found to lower ORC6 expression. Additionally, RNA immunoprecipitation (RIP) and luciferase reporter tests revealed that the transcription factor E2F1 could regulate ORC6 expression in PCa cells. PLK1 overexpression or miR-361-5p inhibitor treatment effectively removed the inhibitory effects caused by ORC6 silencing. Notably, our data showed that therapeutically targeting the miR-361-5p/ORC6/PLK1 axis may be a viable therapy option for PCa.
Collapse
Affiliation(s)
- Zhiqi Liu
- Department of Urology, The First Affiliated Hospital of Anhui Medical University, Hefei, 230000, China; Anhui Public Health Clinical Center, Hefei, 230000, China; Department of Urology, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230000, China
| | - Ying Zhang
- Department of Urology, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230000, China; Department of Urology, Peking University Shenzhen Hospital, Shenzhen, 518000, China
| | - Lin Yu
- Department of Urology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
| | - Zhiqiang Zhang
- Department of Urology, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230000, China
| | - Guangyuan Li
- Department of Urology, The First Affiliated Hospital of Anhui Medical University, Hefei, 230000, China; Anhui Public Health Clinical Center, Hefei, 230000, China.
| |
Collapse
|
9
|
He L, Li Z, Su D, Du H, Zhang K, Zhang W, Wang S, Xie F, Qiu Y, Ma S, Shi G, Yu D, Lei X, Li W, Li M, Wang Z, Gu J, Zhang Y. Tumor Microenvironment-Responsive Nanocapsule Delivery CRISPR/Cas9 to Reprogram the Immunosuppressive Microenvironment in Hepatoma Carcinoma. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2403858. [PMID: 38704691 PMCID: PMC11234430 DOI: 10.1002/advs.202403858] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Indexed: 05/07/2024]
Abstract
Cancer immunotherapy has demonstrated significant efficacy in various tumors, but its effectiveness in treating Hepatocellular Carcinoma (HCC) remains limited. Therefore, there is an urgent need to identify a new immunotherapy target and develop corresponding intervention strategies. Bioinformatics analysis has revealed that growth differentiation factor 15 (GDF15) is highly expressed in HCC and is closely related to poor prognosis of HCC patients. The previous study revealed that GDF15 can promote immunosuppression in the tumor microenvironment. Therefore, knocking out GDF15 through gene editing could potentially reverse the suppressive tumor immune microenvironment permanently. To deliver the CRISPR/Cas9 system specifically to HCC, nanocapsules (SNC) coated with HCC targeting peptides (SP94) on their surface is utilized. These nanocapsules incorporate disulfide bonds (SNCSS) that release their contents in the tumor microenvironment characterized by high levels of glutathione (GSH). In vivo, the SNCSS target HCC cells, exert a marked inhibitory effect on HCC progression, and promote HCC immunotherapy. Mechanistically, CyTOF analysis showed favorable changes in the immune microenvironment of HCC, immunocytes with killer function increased and immunocytes with inhibitive function decreased. These findings highlight the potential of the CRISPR-Cas9 gene editing system in modulating the immune microenvironment and improving the effectiveness of existing immunotherapy approaches for HCC.
Collapse
Affiliation(s)
- Lei He
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal CancersDepartment of BiopharmaceuticsSchool of PharmacyThe Fourth Military Medical UniversityXi'an710032China
| | - Zhaozhao Li
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal CancersDepartment of BiopharmaceuticsSchool of PharmacyThe Fourth Military Medical UniversityXi'an710032China
| | - Danjie Su
- Department of Obstetrics and GynecologyTangdu HospitalThe Fourth Military Medical UniversityXi'an710038China
| | - Haichen Du
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal CancersDepartment of BiopharmaceuticsSchool of PharmacyThe Fourth Military Medical UniversityXi'an710032China
- Department of Oncology940th HospitalJoint Logistic Support ForceLanzhou730050China
| | - Kuo Zhang
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal CancersDepartment of BiopharmaceuticsSchool of PharmacyThe Fourth Military Medical UniversityXi'an710032China
| | - Wangqian Zhang
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal CancersDepartment of BiopharmaceuticsSchool of PharmacyThe Fourth Military Medical UniversityXi'an710032China
| | - Shuning Wang
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal CancersDepartment of BiopharmaceuticsSchool of PharmacyThe Fourth Military Medical UniversityXi'an710032China
| | - Fei Xie
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal CancersDepartment of BiopharmaceuticsSchool of PharmacyThe Fourth Military Medical UniversityXi'an710032China
| | - Yueyuan Qiu
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal CancersDepartment of BiopharmaceuticsSchool of PharmacyThe Fourth Military Medical UniversityXi'an710032China
| | - Shuangxin Ma
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal CancersDepartment of BiopharmaceuticsSchool of PharmacyThe Fourth Military Medical UniversityXi'an710032China
| | - Gege Shi
- College of Life SciencesNorthwest UniversityXi'an710069China
| | - Duo Yu
- Department of NeurosurgeryGeneral Hospital of Central Theater CommandWuhan430012China
| | - Xiaoying Lei
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal CancersDepartment of BiopharmaceuticsSchool of PharmacyThe Fourth Military Medical UniversityXi'an710032China
| | - Weina Li
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal CancersDepartment of BiopharmaceuticsSchool of PharmacyThe Fourth Military Medical UniversityXi'an710032China
| | - Meng Li
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal CancersDepartment of BiopharmaceuticsSchool of PharmacyThe Fourth Military Medical UniversityXi'an710032China
| | - Zhaowei Wang
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal CancersDepartment of BiopharmaceuticsSchool of PharmacyThe Fourth Military Medical UniversityXi'an710032China
| | - Jintao Gu
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal CancersDepartment of BiopharmaceuticsSchool of PharmacyThe Fourth Military Medical UniversityXi'an710032China
| | - Yingqi Zhang
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal CancersDepartment of BiopharmaceuticsSchool of PharmacyThe Fourth Military Medical UniversityXi'an710032China
| |
Collapse
|
10
|
Hou Y, Li J, Li X, Lv Y, Yuan C, Tian J, Liu X. Cross-border regulation of the STK39/MAPK14 pathway by Lycium barbarum miR166a to inhibit triple-negative breast cancer. Am J Transl Res 2024; 16:2683-2698. [PMID: 39006277 PMCID: PMC11236659 DOI: 10.62347/aqew8179] [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: 03/01/2024] [Accepted: 06/01/2024] [Indexed: 07/16/2024]
Abstract
OBJECTIVE To investigate the effects of Lycium barbarum miRNA166a (Lb-miR166a) on human gene expression regulation during the therapy for triple-negative breast cancer (TNBC). METHODS Transcriptome sequencing was used to analyze the distribution and composition of miRNA in Lycium barbarum fruit. Lb-miR166a was introduced into TNBC MB-231 cells by lentiviral transfection to study its effects on cell proliferation, apoptosis, invasion, and metastasis both in vivo and in vitro. Bioinformatic and dual-luciferase assays identified the target gene of Lb-miR166a. The role of STK39 in TNBC progression was elucidated through clinical data analysis combined with cellular studies. The influence of Lb-miR166a on the STK39/MAPK14 pathway was confirmed using a target-specific knockout MB-231 cell line. RESULTS Lb-miR166a was found to be highly expressed in Lycium barbarum. It inhibited MB-231 cell proliferation, invasion, and metastasis, and promoted apoptosis. STK39 was overexpressed in TNBC and was associated with increased invasiveness and poorer patient prognosis. Gene enrichment analysis and dual-luciferase assays demonstrated that Lb-miR166a regulates STK39 expression cross-border and inhibits MAPK14 phosphorylation, impacting the phosphorylation of downstream target genes. CONCLUSION The downregulation of STK39 and subsequent inhibition of MAPK14 phosphorylation by Lb-miR166a leads to reduced proliferation, migration, and invasion of TNBC cells. These findings suggest a novel therapeutic strategy for TNBC treatment, highlighting possible clinical applications of Lb-miR166a in managing this aggressive cancer type.
Collapse
Affiliation(s)
- Yujin Hou
- Department of Oncology, General Hospital of Ningxia Medical UniversityYinchuan, Ningxia, China
| | - Jing Li
- Department of Special Technical Diagnosis and Treatment, Ning’an HospitalYinchuan, Ningxia, China
| | - Xuan Li
- Department of Oncology, General Hospital of Ningxia Medical UniversityYinchuan, Ningxia, China
| | - Ye Lv
- Department of Oncology, General Hospital of Ningxia Medical UniversityYinchuan, Ningxia, China
| | - Chunxiu Yuan
- Department of Oncology, General Hospital of Ningxia Medical UniversityYinchuan, Ningxia, China
| | - Jia Tian
- Institute of Medical Sciences, General Hospital of Ningxia Medical University, Key Laboratory of Fertility Preservation and Maintenance of Ministry of Education, Ningxia Medical UniversityYinchuan, Ningxia, China
| | - Xinlan Liu
- Department of Medical Oncology, Ningxia Hui Autonomous Region HospitalYinchuan, Ningxia, China
| |
Collapse
|
11
|
Singha M, Pu L, Srivastava G, Ni X, Stanfield BA, Uche IK, Rider PJF, Kousoulas KG, Ramanujam J, Brylinski M. Unlocking the Potential of Kinase Targets in Cancer: Insights from CancerOmicsNet, an AI-Driven Approach to Drug Response Prediction in Cancer. Cancers (Basel) 2023; 15:4050. [PMID: 37627077 PMCID: PMC10452340 DOI: 10.3390/cancers15164050] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Revised: 07/16/2023] [Accepted: 07/26/2023] [Indexed: 08/27/2023] Open
Abstract
Deregulated protein kinases are crucial in promoting cancer cell proliferation and driving malignant cell signaling. Although these kinases are essential targets for cancer therapy due to their involvement in cell development and proliferation, only a small part of the human kinome has been targeted by drugs. A comprehensive scoring system is needed to evaluate and prioritize clinically relevant kinases. We recently developed CancerOmicsNet, an artificial intelligence model employing graph-based algorithms to predict the cancer cell response to treatment with kinase inhibitors. The performance of this approach has been evaluated in large-scale benchmarking calculations, followed by the experimental validation of selected predictions against several cancer types. To shed light on the decision-making process of CancerOmicsNet and to better understand the role of each kinase in the model, we employed a customized saliency map with adjustable channel weights. The saliency map, functioning as an explainable AI tool, allows for the analysis of input contributions to the output of a trained deep-learning model and facilitates the identification of essential kinases involved in tumor progression. The comprehensive survey of biomedical literature for essential kinases selected by CancerOmicsNet demonstrated that it could help pinpoint potential druggable targets for further investigation in diverse cancer types.
Collapse
Affiliation(s)
- Manali Singha
- Department of Biological Sciences, Louisiana State University, Baton Rouge, LA 70803, USA; (M.S.); (G.S.); (X.N.)
| | - Limeng Pu
- Center for Computation and Technology, Louisiana State University, Baton Rouge, LA 70803, USA; (L.P.); (J.R.)
| | - Gopal Srivastava
- Department of Biological Sciences, Louisiana State University, Baton Rouge, LA 70803, USA; (M.S.); (G.S.); (X.N.)
| | - Xialong Ni
- Department of Biological Sciences, Louisiana State University, Baton Rouge, LA 70803, USA; (M.S.); (G.S.); (X.N.)
| | - Brent A. Stanfield
- Department of Pathobiological Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA 70803, USA; (B.A.S.); (I.K.U.); (P.J.F.R.); (K.G.K.)
| | - Ifeanyi K. Uche
- Department of Pathobiological Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA 70803, USA; (B.A.S.); (I.K.U.); (P.J.F.R.); (K.G.K.)
- Division of Biotechnology and Molecular Medicine, Department of Pathobiological Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA 70803, USA
- School of Medicine, Louisiana State University Health Sciences Center, New Orleans, LA 70112, USA
| | - Paul J. F. Rider
- Department of Pathobiological Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA 70803, USA; (B.A.S.); (I.K.U.); (P.J.F.R.); (K.G.K.)
- Division of Biotechnology and Molecular Medicine, Department of Pathobiological Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA 70803, USA
| | - Konstantin G. Kousoulas
- Department of Pathobiological Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA 70803, USA; (B.A.S.); (I.K.U.); (P.J.F.R.); (K.G.K.)
- Division of Biotechnology and Molecular Medicine, Department of Pathobiological Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA 70803, USA
| | - J. Ramanujam
- Center for Computation and Technology, Louisiana State University, Baton Rouge, LA 70803, USA; (L.P.); (J.R.)
- Division of Electrical and Computer Engineering, Louisiana State University, Baton Rouge, LA 70803, USA
| | - Michal Brylinski
- Department of Biological Sciences, Louisiana State University, Baton Rouge, LA 70803, USA; (M.S.); (G.S.); (X.N.)
- Center for Computation and Technology, Louisiana State University, Baton Rouge, LA 70803, USA; (L.P.); (J.R.)
| |
Collapse
|
12
|
Zigo M, Kerns K, Sutovsky P. The Ubiquitin-Proteasome System Participates in Sperm Surface Subproteome Remodeling during Boar Sperm Capacitation. Biomolecules 2023; 13:996. [PMID: 37371576 PMCID: PMC10296210 DOI: 10.3390/biom13060996] [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: 05/01/2023] [Revised: 06/08/2023] [Accepted: 06/12/2023] [Indexed: 06/29/2023] Open
Abstract
Sperm capacitation is a complex process endowing biological and biochemical changes to a spermatozoon for a successful encounter with an oocyte. The present study focused on the role of the ubiquitin-proteasome system (UPS) in the remodeling of the sperm surface subproteome. The sperm surface subproteome from non-capacitated and in vitro capacitated (IVC) porcine spermatozoa, with and without proteasomal inhibition, was selectively isolated. The purified sperm surface subproteome was analyzed using high-resolution, quantitative liquid chromatography-mass spectrometry (LC-MS) in four replicates. We identified 1680 HUGO annotated proteins, out of which we found 91 to be at least 1.5× less abundant (p < 0.05) and 141 to be at least 1.5× more abundant (p < 0.05) on the surface of IVC spermatozoa. These proteins were associated with sperm capacitation, hyperactivation, metabolism, acrosomal exocytosis, and fertilization. Abundances of 14 proteins were found to be significantly different (p < 0.05), exceeding a 1.5-fold abundance between the proteasomally inhibited (100 µM MG132) and vehicle control (0.2% ethanol) groups. The proteins NIF3L1, CSE1L, NDUFB7, PGLS, PPP4C, STK39, and TPRG1L were found to be more abundant; while BPHL, GSN, GSPT1, PFDN4, STYXL1, TIMM10, and UBXN4 were found to be less abundant in proteasomally inhibited IVC spermatozoa. Despite the UPS having a narrow range of targets, it modulated sperm metabolism and binding by regulating susceptible surface proteins. Changes in CSE1L, PFDN4, and STK39 during in vitro capacitation were confirmed using immunocytochemistry, image-based flow cytometry, and Western blotting. The results confirmed the active participation of the UPS in the extensive sperm surface proteome remodeling that occurs during boar sperm capacitation. This work will help us to identify new pharmacological mechanisms to positively or negatively modulate sperm fertilizing ability in food animals and humans.
Collapse
Affiliation(s)
- Michal Zigo
- Division of Animal Science, University of Missouri, Columbia, MO 65211, USA;
| | - Karl Kerns
- Division of Animal Science, University of Missouri, Columbia, MO 65211, USA;
- Department of Animal Science, Iowa State University, Ames, IA 50011, USA;
| | - Peter Sutovsky
- Department of Animal Science, Iowa State University, Ames, IA 50011, USA;
- Department of Obstetrics, Gynecology and Women’s Health, University of Missouri, Columbia, MO 65211, USA
| |
Collapse
|
13
|
Karimi K, Mojtabavi S, Tehrany PM, Nejad MM, Rezaee A, Mohtashamian S, Hamedi E, Yousefi F, Salmani F, Zandieh MA, Nabavi N, Rabiee N, Ertas YN, Salimimoghadam S, Rashidi M, Rahmanian P, Hushmandi K, Yu W. Chitosan-based nanoscale delivery systems in hepatocellular carcinoma: Versatile bio-platform with theranostic application. Int J Biol Macromol 2023; 242:124935. [PMID: 37230442 DOI: 10.1016/j.ijbiomac.2023.124935] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Revised: 04/13/2023] [Accepted: 05/15/2023] [Indexed: 05/27/2023]
Abstract
The field of nanomedicine has provided a fresh approach to cancer treatment by addressing the limitations of current therapies and offering new perspectives on enhancing patients' prognoses and chances of survival. Chitosan (CS) is isolated from chitin that has been extensively utilized for surface modification and coating of nanocarriers to improve their biocompatibility, cytotoxicity against tumor cells, and stability. HCC is a prevalent kind of liver tumor that cannot be adequately treated with surgical resection in its advanced stages. Furthermore, the development of resistance to chemotherapy and radiotherapy has caused treatment failure. The targeted delivery of drugs and genes can be mediated by nanostructures in treatment of HCC. The current review focuses on the function of CS-based nanostructures in HCC therapy and discusses the newest advances of nanoparticle-mediated treatment of HCC. Nanostructures based on CS have the capacity to escalate the pharmacokinetic profile of both natural and synthetic drugs, thus improving the effectiveness of HCC therapy. Some experiments have displayed that CS nanoparticles can be deployed to co-deliver drugs to disrupt tumorigenesis in a synergistic way. Moreover, the cationic nature of CS makes it a favorable nanocarrier for delivery of genes and plasmids. The use of CS-based nanostructures can be harnessed for phototherapy. Additionally, the incur poration of ligands including arginylglycylaspartic acid (RGD) into CS can elevate the targeted delivery of drugs to HCC cells. Interestingly, smart CS-based nanostructures, including ROS- and pH-sensitive nanoparticles, have been designed to provide cargo release at the tumor site and enhance the potential for HCC suppression.
Collapse
Affiliation(s)
- Kimia Karimi
- Faculty of Veterinary Medicine, Islamic Azad University, Science and Research Branch, Tehran, Iran
| | - Sarah Mojtabavi
- Faculty of Veterinary Medicine, Islamic Azad University, Science and Research Branch, Tehran, Iran
| | | | - Melina Maghsodlou Nejad
- Faculty of Veterinary Medicine, Islamic Azad University, Science and Research Branch, Tehran, Iran
| | - Aryan Rezaee
- Iran University of Medical Sciences, Tehran, Iran
| | - Shahab Mohtashamian
- Department of Biomedical Engineering, Faculty of Chemical Engineering, Tarbiat Modares University, Tehran, Iran
| | - Erfan Hamedi
- Department of Aquatic Animal Health & Diseases, Department of Clinical Sciences, Faculty of Veterinary Medicine, Shiraz University, Shiraz, Iran
| | - Farnaz Yousefi
- Department of Clinical Science, Faculty of Veterinary Medicine, Islamic Azad University, Science and Research Branch, Tehran, Iran
| | - Farshid Salmani
- Faculty of Veterinary Medicine, Islamic Azad University, Science and Research Branch, Tehran, Iran
| | - Mohammad Arad Zandieh
- Department of Food Hygiene and Quality Control, Division of Epidemiology, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
| | - Noushin Nabavi
- Department of Urological Sciences and Vancouver Prostate Centre, University of British Columbia, Vancouver, BC V6H3Z6, Canada
| | - Navid Rabiee
- Centre for Molecular Medicine and Innovative Therapeutics, Murdoch University, Perth, WA 6150, Australia; School of Engineering, Macquarie University, Sydney, New South Wales 2109, Australia
| | - Yavuz Nuri Ertas
- Department of Biomedical Engineering, Erciyes University, Kayseri, Turkey; ERNAM-Nanotechnology Research and Application Center, Erciyes University, Kayseri, Türkiye
| | - Shokooh Salimimoghadam
- Department of Biochemistry and Molecular Biology, Faculty of Veterinary Medicine, Shahid Chamran University of Ahvaz, Ahvaz, Iran
| | - Mohsen Rashidi
- Department Pharmacology, Faculty of Medicine, Mazandaran University of Medical Sciences, Sari, Iran; The Health of Plant and Livestock Products Research Center, Mazandaran University of Medical Sciences, Sari, Iran.
| | - Parham Rahmanian
- Faculty of Veterinary Medicine, Islamic Azad University, Science and Research Branch, Tehran, Iran.
| | - Kiavash Hushmandi
- Department of Food Hygiene and Quality Control, Division of Epidemiology, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran.
| | - Wei Yu
- School of Pharmacy, Xianning Medical College, Hubei University of Science and Technology, Xianning, 437100, China.
| |
Collapse
|
14
|
Qiu H, Su N, Wang J, Yan S, Li J. Quantitative proteomics analysis in small cell carcinoma of cervix reveals novel therapeutic targets. Clin Proteomics 2023; 20:18. [PMID: 37031178 PMCID: PMC10082492 DOI: 10.1186/s12014-023-09408-x] [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: 04/25/2022] [Accepted: 04/02/2023] [Indexed: 04/10/2023] Open
Abstract
BACKGROUND As a rare pathologic subtype, small cell carcinoma of the cervix (SCCC) is characterized by extensive aggressiveness and resistance to current therapies. To date, our knowledge of SCCC origin and progression is limited and sometimes even controversial. Herein, we explored the whole-protein expression profiles in a panel of SCCC cases, aiming to provide more evidence for the precise diagnosis and targeting therapy. METHODS Eighteen SCCC samples and six matched normal cervix tissues were collected from January 2013 to December 2017. Data independent acquisition mass spectrometry (DIA) was performed to discriminate the different proteins (DEPs) associated with SCCC. The expression of CDN2A and SYP in corresponding SCCC tissues was verified using immunohistochemistry. GO and KEGG enrichment analyses were used to identify the key DEPs related to SCCC development and tumor recurrence. RESULTS As a result, 1311 DEPs were identified in SCCC tissues (780 up-regulated and 531 down-regulated). In up-regulated DEPs, both GO analysis and KEGG analysis showed the most enriched were related to DNA replication (including nuclear DNA replication, DNA-dependent DNA replication, and cell cycle DNA replication), indicating the prosperous proliferation in SCCC. As for the down-regulated DEPs, GO analysis showed that the most enriched functions were associated with extracellular matrix collagen-containing extracellular matrix. KEGG analysis revealed that the DEPs were enriched in Complement and coagulation cascades, proteoglycans in cancer, and focal adhesion-related pathways. Down-regulation of these proteins could enhance the mobility of cancer cells and establish a favorable microenvironment for tumor metastasis, which might be accounted for the frequent local and distant metastasis in SCCC. Surprisingly, the blood vessels and circulatory system exhibit a down-regulation in SCCC, which might be partly responsible for its resistance to anti-angiogenic regimens. In the stratification analysis of early-stage tumors, a group of enzymes involved in the cancer metabolism was discriminated in these recurrence cases. CONCLUSIONS Using quantitative proteomics analysis, we first reported the whole-protein expression profiles in SCCC. Significant alterations were found in proteins associated with the enhancement of DNA replication and cellular motility. Besides the association with mitosis, a unique metabolic feature was detected in cases with tumor recurrence. These findings provided novel targets for disease surveillance and treatments, which warranted further validation in the future.
Collapse
Affiliation(s)
- Haifeng Qiu
- Department of Gynecology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Provincial Medical Key Laboratory for Gynecologic Malignancies Prevention and Treatment, Zhengzhou, Henan, China
- Zhengzhou Key Laboratory for Gynecologic Malignancies Prevention and Treatment, Zhengzhou, Henan, China
| | - Ning Su
- Department of Gynecologic Oncology, The Affiliated Cancer Hospital of Zhengzhou University and Henan Cancer Hospital, Zhengzhou, Henan, China
| | - Jing Wang
- Department of Obstetrics and Gynecology, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, Shandong, China
| | - Shuping Yan
- Department of Pathology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Jing Li
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, No.1, East Jianshe Road, Erqi District, Zhengzhou, 450000, Henan, China.
| |
Collapse
|
15
|
Kong L, Liang C, Li P, Zhang Y, Feng S, Zhang D, Yao R, Yang L, Hao Z, Zhang H, Tian X, Guo C, Du B, Dong J, Zhang Y. Myotubularin‐Related Protein14 Prevents Neointima Formation and Vascular Smooth Muscle Cell Proliferation by Inhibiting Polo‐Like Kinase1. J Am Heart Assoc 2022; 11:e026174. [DOI: 10.1161/jaha.122.026174] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Background
Restenosis is one of the main bottlenecks in restricting the further development of cardiovascular interventional therapy. New signaling molecules involved in the progress have continuously been discovered; however, the specific molecular mechanisms remain unclear. MTMR14 (myotubularin‐related protein 14) is a novel phosphoinositide phosphatase that has a variety of biological functions and is involved in diverse biological processes. However, the role of MTMR14 in vascular biology remains unclear. Herein, we addressed the role of MTMR14 in neointima formation and vascular smooth muscle cell (VSMC) proliferation after vessel injury.
Methods and Results
Vessel injury models were established using SMC‐specific conditional MTMR14‐knockout and ‐transgenic mice. Neointima formation was assessed by histopathological methods, and VSMC proliferation and migration were assessed using fluorescence ubiquitination‐based cell cycle indicator, transwell, and scratch wound assay. Neointima formation and the expression of MTMR14 was increased after injury. MTMR14 deficiency accelerated neointima formation and promoted VSMC proliferation after injury, whereas MTMR14 overexpression remarkably attenuated this process. Mechanistically, we demonstrated that MTMR14 suppressed the activation of PLK1 (polo‐like kinase 1) by interacting with it, which further leads to the inhibition of the activation of MEK/ERK/AKT (mitogen‐activated protein kinase kinase/extracellular‐signal‐regulated kinase/protein kinase B), thereby inhibiting the proliferation of VSMC from the medial to the intima and thus preventing neointima formation.
Conclusions
MTMR14 prevents neointima formation and VSMC proliferation by inhibiting PLK1. Our findings reveal that MTMR14 serves as an inhibitor of VSMC proliferation and establish a link between MTMR14 and PLK1 in regulating VSMC proliferation. MTMR14 may become a novel potential therapeutic target in the treatment of restenosis.
Collapse
Affiliation(s)
- Ling‐Yao Kong
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University Zhengzhou University Zhengzhou China
| | - Cui Liang
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University Zhengzhou University Zhengzhou China
| | - Peng‐Cheng Li
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University Zhengzhou University Zhengzhou China
| | - Yi‐Wei Zhang
- The Second School of Clinical Medicine Southern Medical University Guangzhou China
| | - Sheng‐Dong Feng
- Department of Cardiology The 7th People’s Hospital of Zhengzhou Zhengzhou China
| | - Dian‐Hong Zhang
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University Zhengzhou University Zhengzhou China
| | - Rui Yao
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University Zhengzhou University Zhengzhou China
| | - Lu‐Lu Yang
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University Zhengzhou University Zhengzhou China
| | - Zheng‐Yang Hao
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University Zhengzhou University Zhengzhou China
| | - Hao Zhang
- Department of Thoracic Surgery Union Hospital Wuhan China
- Department of Cardiovascular Surgery Union Hospital Wuhan China
| | - Xiao‐Xu Tian
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University Zhengzhou University Zhengzhou China
| | - Chen‐Ran Guo
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University Zhengzhou University Zhengzhou China
| | - Bin‐Bin Du
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University Zhengzhou University Zhengzhou China
| | - Jian‐Zeng Dong
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University Zhengzhou University Zhengzhou China
- Department of Cardiology Beijing Anzhen Hospital Capital Medical University National Clinical Research Centre for Cardiovascular Diseases Beijing China
| | - Yan‐Zhou Zhang
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University Zhengzhou University Zhengzhou China
| |
Collapse
|
16
|
Xiu M, Li L, Li Y, Gao Y. An update regarding the role of WNK kinases in cancer. Cell Death Dis 2022; 13:795. [PMID: 36123332 PMCID: PMC9485243 DOI: 10.1038/s41419-022-05249-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2022] [Revised: 09/07/2022] [Accepted: 09/08/2022] [Indexed: 01/23/2023]
Abstract
Mammalian WNK kinases (WNKs) are serine/threonine kinases that contain four members, WNK1-4. They function to maintain ion homeostasis and regulate blood pressure in mammals. Recent studies have revealed that the dysregulation of WNKs contributes to tumor growth, metastasis, and angiogenesis through complex mechanisms, especially through phosphorylating kinase substrates SPS1-related proline/alanine-rich kinase (SPAK) and oxidative stress-responsive kinase 1 (OSR1). Here, we review and discuss the relationships between WNKs and several key factors/biological processes in cancer, including ion channels, cation chloride cotransporters, sodium bicarbonate cotransporters, signaling pathways, angiogenesis, autophagy, and non-coding RNAs. In addition, the potential drugs for targeting WNK-SPAK/OSR1 signaling have also been discussed. This review summarizes and discusses knowledge of the roles of WNKs in cancer, which provides a comprehensive reference for future studies.
Collapse
Affiliation(s)
- Mengxi Xiu
- grid.24516.340000000123704535Department of Oncology, Shanghai East Hospital, Tongji University School of Medicine, 200120 Shanghai, China
| | - Li Li
- grid.24516.340000000123704535Department of Oncology, Shanghai East Hospital, Tongji University School of Medicine, 200120 Shanghai, China
| | - Yandong Li
- grid.24516.340000000123704535Department of Oncology, Shanghai East Hospital, Tongji University School of Medicine, 200120 Shanghai, China
| | - Yong Gao
- grid.24516.340000000123704535Department of Oncology, Shanghai East Hospital, Tongji University School of Medicine, 200120 Shanghai, China
| |
Collapse
|
17
|
Yang D, Li H, Chen Y, Li C, Ren W, Huang Y. A Pan-Cancer Analysis of the Oncogenic Role of BCL7B: A Potential Biomarker for Prognosis and Immunotherapy. Front Genet 2022; 13:906174. [PMID: 35910232 PMCID: PMC9334570 DOI: 10.3389/fgene.2022.906174] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Accepted: 06/09/2022] [Indexed: 12/03/2022] Open
Abstract
Background: Previous studies have partly explored the role of B-cell CLL/lymphoma 7 protein family member B (BCL7B) in tumorigenesis and development. However, the prognosis and immunoregulatory value of BCL7B in pan-cancer patients remains unclear. Methods: Through The Cancer Genome Atlas (TCGA) and Genotype-Tissue Expression (GTEx) databases, the distinct expression of BCL7B gene in 33 tumors and adjacent normal tissues was analyzed. The Kaplan–Meier method (univariate Cox regression analysis and Kaplan–Meier curve) was used to identify the cancer types whose BCL7B gene expression was related to prognosis. The receiver operating characteristic (ROC) curve was used to elucidate the diagnosis value of BCL7B gene. Spearman’s rank correlation coefficient was used to explore the relationship between BCL7B gene expression and immune cell infiltration, immune checkpoints, DNA methylation, DNA repair genes, immune-activating genes, immune-suppressing genes, immune subtypes, tumor mutation burden (TMB), and microsatellite instability (MSI). The Wilcoxon rank sum test and Kruskal–Wallis test were used to compare the expression of BCL7B gene in tumor tissues with different clinicopathological features. Gene set enrichment analysis (GSEA) was conducted to identify the tumor-related pathways in pan-cancer. The Human Protein Atlas (HPA) database was used to verify the BCL7B gene expression at the protein level. Results: High expression of BCL7B was associated with an inferior prognosis in glioblastoma multiforme (GBM), glioma (GBMLGG), kidney chromophobe (KICH), brain lower grade glioma (LGG), oral squamous cell carcinoma (OSCC), rectum adenocarcinoma (READ), and uveal melanoma (UVM). Low expression of BCL7B was associated with a poor prognosis in kidney renal clear cell carcinoma (KIRC), kidney renal papillary cell carcinoma (KIRP), skin cutaneous melanoma (SKCM), thyroid carcinoma (THCA), and sarcoma (SARC). The BCL7B gene expression had varying degrees of correlation with 24 immune cell subsets in 37 tumor environments such as adrenocortical carcinoma (ACC) and bladder urothelial carcinoma (BCLA). Spearman’s rank correlation coefficient showed that BCL7B gene expression had different degrees of correlation with 47 immune checkpoints, 46 immune-activating genes, 24 immune-suppressing genes, 5 DNA repair genes, and DNA methylation, TMB, and MSI in 39 tumors. GSEA suggested that BCL7B was notably associated with cancer-related and immune-related pathways. Conclusion: In summary, BCL7B gene has a high diagnostic and prognostic value in pan-cancer and is related to the infiltration of 24 immune cell subsets in pan-cancer.
Collapse
Affiliation(s)
- Dinglong Yang
- Second Clinical Medical College, Shanxi Medical University, Taiyuan, China
| | - Hetong Li
- Second Clinical Medical College, Shanxi Medical University, Taiyuan, China
| | - Yujing Chen
- School of Public Health, Xi’an Jiaotong University, Xian, China
| | - Chunjiang Li
- Department of Orthopedics, The Second Hospital of Shanxi Medical University, Taiyuan, China
| | - Weiping Ren
- Department of Orthopedics, The Second Hospital of Shanxi Medical University, Taiyuan, China
| | - Yongbo Huang
- Department of Orthopedics, The Second Hospital of Shanxi Medical University, Taiyuan, China
- *Correspondence: Yongbo Huang,
| |
Collapse
|
18
|
Xu H, Ma Z, Mo X, Chen X, Xu F, Wu F, Chen H, Zhou G, Xia H, Zhang C. Inducing Synergistic DNA Damage by TRIP13 and PARP1 Inhibitors Provides a Potential Treatment for Hepatocellular Carcinoma. J Cancer 2022; 13:2226-2237. [PMID: 35517402 PMCID: PMC9066198 DOI: 10.7150/jca.66020] [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: 08/12/2021] [Accepted: 12/20/2021] [Indexed: 12/26/2022] Open
Abstract
Thyroid hormone receptor interactor 13 (TRIP13), an AAA-ATPase, participates in the development of many cancers. This study explores the function of TRIP13 and synergistic effects of TRIP13 and PARP1 inhibitors in hepatocellular carcinoma (HCC). The dose-dependent effects of TRIP13 and PARP1 inhibitors on HCC cells proliferation or migration were investigated by the CCK-8 and Transwell assays. Using siRNA or lentivirus to knock down TRIP13, we tested HCC cell and tumor growth in vitro and in vivo. The DNA damage caused by TRIP13 and PARP1 inhibitors was measured by the phosphorylation of H2AX, one of the DNA damage biomarkers. The phosphorylation of H2AX was increased after treatment with DCZ0415 or TRIP13 knockdown. Combining DCZ0415 with PARP1 inhibitor, Olaparib induced synergistic anti-HCC activity. We also found that the overexpression of TRIP13 is significantly associated with early recurrent HCC and poor survival. Up-regulation of TRIP13 in HCC was regulated by transcription factor SP1. In conclusion, our study demonstrated that DCZ0415 targeting TRIP13 impaired non-homologous end-joining repair to inhibit HCC progression and had a synergistic effect with PARP1 inhibitor Olaparib in HCC, suggesting a potential treatment of HCC.
Collapse
Affiliation(s)
- Haojun Xu
- School of Basic Medical Sciences &Key Laboratory of Antibody Technique of National Health Commission & Jiangsu Antibody Drug Engineering Research Center, Nanjing Medical University, Nanjing 211166, China
| | - Zhijie Ma
- School of Basic Medical Sciences &Key Laboratory of Antibody Technique of National Health Commission & Jiangsu Antibody Drug Engineering Research Center, Nanjing Medical University, Nanjing 211166, China
| | - Xiao Mo
- Department of Pathology, The first people's hospital of Foshan, Foshan 528041, China.,School of Basic Medical Sciences &Key Laboratory of Antibody Technique of National Health Commission & Jiangsu Antibody Drug Engineering Research Center, Nanjing Medical University, Nanjing 211166, China
| | - Xiaoli Chen
- Sir Run Run Hospital, Nanjing Medical University, Nanjing 211166, China
| | - Fanggui Xu
- Sir Run Run Hospital, Nanjing Medical University, Nanjing 211166, China
| | - Fubing Wu
- Sir Run Run Hospital, Nanjing Medical University, Nanjing 211166, China
| | - Hongjin Chen
- School of Basic Medical Sciences &Key Laboratory of Antibody Technique of National Health Commission & Jiangsu Antibody Drug Engineering Research Center, Nanjing Medical University, Nanjing 211166, China
| | - Guoren Zhou
- Jiangsu Cancer Hospital & The Affiliated Cancer Hospital of Nanjing Medical University& Jiangsu Institute of Cancer Research, Nanjing 2100092, China
| | - Hongping Xia
- School of Basic Medical Sciences &Key Laboratory of Antibody Technique of National Health Commission & Jiangsu Antibody Drug Engineering Research Center, Nanjing Medical University, Nanjing 211166, China.,Sir Run Run Hospital, Nanjing Medical University, Nanjing 211166, China
| | - Chengfei Zhang
- Sir Run Run Hospital, Nanjing Medical University, Nanjing 211166, China
| |
Collapse
|
19
|
Pellegrino R, Castoldi M, Ticconi F, Skawran B, Budczies J, Rose F, Schwab C, Breuhahn K, Neumann UP, Gaisa NT, Loosen SH, Luedde T, Costa IG, Longerich T. LINC00152 Drives a Competing Endogenous RNA Network in Human Hepatocellular Carcinoma. Cells 2022; 11:cells11091528. [PMID: 35563834 PMCID: PMC9103153 DOI: 10.3390/cells11091528] [Citation(s) in RCA: 7] [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: 04/12/2022] [Revised: 04/29/2022] [Accepted: 05/01/2022] [Indexed: 11/16/2022] Open
Abstract
Genomic and epigenomic studies revealed dysregulation of long non-coding RNAs in many cancer entities, including liver cancer. We identified an epigenetic mechanism leading to upregulation of the long intergenic non-coding RNA 152 (LINC00152) expression in human hepatocellular carcinoma (HCC). Here, we aimed to characterize a potential competing endogenous RNA (ceRNA) network, in which LINC00152 exerts oncogenic functions by sponging miRNAs, thereby affecting their target gene expression. Database and gene expression data of human HCC were integrated to develop a potential LINC00152-driven ceRNA in silico. RNA immunoprecipitation and luciferase assay were used to identify miRNA binding to LINC00152 in human HCC cells. Functionally active players in the ceRNA network were analyzed using gene editing, siRNA or miRNA mimic transfection, and expression vectors in vitro. RNA expression in human HCC in vivo was validated by RNA in situ hybridization. Let-7c-5p, miR-23a-3p, miR-125a-5p, miR-125b-5p, miR-143a-3p, miR-193-3p, and miR-195-5p were detected as new components of the potential LINC00152 ceRNA network in human HCC. LINC00152 was confirmed to sponge miR143a-3p in human HCC cell lines, thereby limiting its binding to their respective target genes, like KLC2. KLC2 was identified as a central mediator promoting pro-tumorigenic effects of LINC00152 overexpression in HCC cells. Furthermore, co-expression of LINC00152 and KLC2 was observed in human HCC cohorts and high KLC2 expression was associated with shorter patient survival. Functional assays demonstrated that KLC2 promoted cell proliferation, clonogenicity and migration in vitro. The LINC00152-miR-143a-3p-KLC2 axis may represent a therapeutic target in human HCC.
Collapse
Affiliation(s)
- Rossella Pellegrino
- Institute of Pathology, Heidelberg University Hospital, 69120 Heidelberg, Germany; (J.B.); (F.R.); (C.S.); (K.B.); (T.L.)
- Correspondence: ; Tel.: +49-(0)6221-56-34094
| | - Mirco Castoldi
- Clinic for Gastroenterology, Hepatology and Infectious Diseases, University Hospital Düsseldorf, Medical Faculty of Heinrich Heine University Düsseldorf, 40225 Düsseldorf, Germany; (M.C.); (S.H.L.); (T.L.)
| | - Fabio Ticconi
- Institute for Computational Genomics, Joint Research Center for Computational Biomedicine, University Hospital RWTH Aachen, 52074 Aachen, Germany; (F.T.); (I.G.C.)
| | - Britta Skawran
- Institute of Human Genetics, Hannover Medical School, 30625 Hannover, Germany;
| | - Jan Budczies
- Institute of Pathology, Heidelberg University Hospital, 69120 Heidelberg, Germany; (J.B.); (F.R.); (C.S.); (K.B.); (T.L.)
| | - Fabian Rose
- Institute of Pathology, Heidelberg University Hospital, 69120 Heidelberg, Germany; (J.B.); (F.R.); (C.S.); (K.B.); (T.L.)
| | - Constantin Schwab
- Institute of Pathology, Heidelberg University Hospital, 69120 Heidelberg, Germany; (J.B.); (F.R.); (C.S.); (K.B.); (T.L.)
| | - Kai Breuhahn
- Institute of Pathology, Heidelberg University Hospital, 69120 Heidelberg, Germany; (J.B.); (F.R.); (C.S.); (K.B.); (T.L.)
| | - Ulf P. Neumann
- Department of General, Visceral and Transplant Surgery, University Hospital RWTH Aachen, 52074 Aachen, Germany;
- Department of Surgery, Maastricht University Medical Centre, 6229 HX Maastricht, The Netherlands
| | - Nadine T. Gaisa
- Institute of Pathology, University Hospital RWTH Aachen, 52074 Aachen, Germany;
| | - Sven H. Loosen
- Clinic for Gastroenterology, Hepatology and Infectious Diseases, University Hospital Düsseldorf, Medical Faculty of Heinrich Heine University Düsseldorf, 40225 Düsseldorf, Germany; (M.C.); (S.H.L.); (T.L.)
| | - Tom Luedde
- Clinic for Gastroenterology, Hepatology and Infectious Diseases, University Hospital Düsseldorf, Medical Faculty of Heinrich Heine University Düsseldorf, 40225 Düsseldorf, Germany; (M.C.); (S.H.L.); (T.L.)
| | - Ivan G. Costa
- Institute for Computational Genomics, Joint Research Center for Computational Biomedicine, University Hospital RWTH Aachen, 52074 Aachen, Germany; (F.T.); (I.G.C.)
| | - Thomas Longerich
- Institute of Pathology, Heidelberg University Hospital, 69120 Heidelberg, Germany; (J.B.); (F.R.); (C.S.); (K.B.); (T.L.)
| |
Collapse
|
20
|
He Q, Fan B, Du P, Jin Y. Construction and Validation of Two Hepatocellular Carcinoma-Progression Prognostic Scores Based on Gene Set Variation Analysis. Front Cell Dev Biol 2022; 10:806989. [PMID: 35356272 PMCID: PMC8959467 DOI: 10.3389/fcell.2022.806989] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Accepted: 02/01/2022] [Indexed: 11/24/2022] Open
Abstract
Background: Liver hepatocellular carcinoma (LIHC) remains a global health challenge with a low early diagnosis rate and high mortality. Therefore, finding new biomarkers for diagnosis and prognosis is still one of the current research priorities. Methods: Based on the variation of gene expression patterns in different stages, the LIHC-development genes (LDGs) were identified by differential expression analysis. Then, prognosis-related LDGs were screened out to construct the LIHC-unfavorable gene set (LUGs) and LIHC-favorable gene set (LFGs). Gene set variation analysis (GSVA) was conducted to build prognostic scoring models based on the LUGs and LFGs. ROC curve analysis and univariate and multivariate Cox regression analysis were carried out to verify the diagnostic and prognostic utility of the two GSVA scores in two independent datasets. Additionally, the key LCGs were identified by the intersection analysis of the PPI network and univariate Cox regression and further evaluated their performance in expression level and prognosis prediction. Single-sample GSEA (ssGSEA) was performed to understand the correlation between the two GSVA enrichment scores and immune activity. Result: With the development of LIHC, 83 LDGs were gradually upregulated and 247 LDGs were gradually downregulated. Combining with LIHC survival analysis, 31 LUGs and 32 LFGs were identified and used to establish the LIHC-unfavorable GSVA score (LUG score) and LIHC-favorable GSVA score (LFG score). ROC curve analysis and univariate/multivariate Cox regression analysis suggested the LUG score and LFG score could be great indicators for the early diagnosis and prognosis prediction. Four genes (ESR1, EHHADH, CYP3A4, and ACADL) were considered as the key LCGs and closely related to good prognosis. The frequency of TP53 mutation and copy number variation (CNV) were high in some LCGs. Low-LFG score patients have active metabolic activity and a more robust immune response. The high-LFG score patients characterized immune activation with the higher infiltration abundance of type I T helper cells, DC, eosinophils, and neutrophils, while the high-LUG score patients characterized immunosuppression with the higher infiltration abundance of type II T helper cells, TRegs, and iDC. The high- and low-LFG score groups differed significantly in immunotherapy response scores, immune checkpoints expression, and IC50 values of common drugs. Conclusion: Overall, the LIHC-progression characteristic genes can be great diagnostic and prognostic signatures and the two GSVA score systems may become promising indices for guiding the tumor treatment of LIHC patients.
Collapse
|
21
|
Li Z, Zhang Y, Zhou Y, Wang F, Yin C, Ding L, Zhang S. Tanshinone IIA suppresses the progression of lung adenocarcinoma through regulating CCNA2-CDK2 complex and AURKA/PLK1 pathway. Sci Rep 2021; 11:23681. [PMID: 34880385 PMCID: PMC8654884 DOI: 10.1038/s41598-021-03166-2] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Accepted: 11/29/2021] [Indexed: 12/26/2022] Open
Abstract
Lung adenocarcinoma (LUAD) belongs to a subgroup of non-small cell lung cancer (NSCLC) with an increasing incidence all over the world. Tanshinone IIA (TSA), an active compound of Salvia miltiorrhiza Bunge., has been found to have anti-tumor effects on many tumors, but its anti-LUAD effect and its mechanism have not been reported yet. In this study, bio-information analysis was applied to characterize the potential mechanism of TSA on LUA, biological experiments were used to verify the mechanisms involved. TCGA, Pubchem, SwissTargetPrediction, Venny2.1.0, STRING, DAVID, Cytoscape 3.7.2, Omicshare, GEPIA, RSCBPDB, Chem Draw, AutoDockTools, and PyMOL were utilized for analysis in the bio-information analysis and network pharmacology. Our experiments in vitro focused on the anti-LUAD effects and mechanisms of TSA on LUAD cells (A549 and NCI-H1975 cells) via MTT, plate cloning, Annexin V-FITC and PI dual staining, flow cytometry, and western blot assays. A total of 64 differentially expressed genes (DEGs) of TSA for treatment of LUAD were screened out. Gene ontology and pathway analysis revealed characteristic of the DEGs network. After GEPIA-based DEGs confirmation, 46 genes were considered having significant differences. Further, 10 key DEGs (BTK, HSD11B1, ADAM33, TNNC1, THRA, CCNA2, AURKA, MIF, PLK1, and SORD) were identified as the most likely relevant genes from overall survival analysis. Molecular Docking results showed that CCNA2, CDK2 and PLK1 had the lowest docking energy. MTT and plate cloning assays results showed that TSA inhibited the proliferation of LUAD cells in a concentration-dependent manner. Annexin V-FITC and PI dual staining and flow cytometry assays results told that TSA promoted the apoptosis of the two LUAD cells in different degrees, and induced cycle arrest in the G1/S phase. Western blot results showed that TSA significantly down-regulated the expression of CCNA2, CDK2, AURKA, PLK1, and p-ERK. In summary, TSA could suppress the progression of LUAD by inducing cell apoptosis and arresting cell cycle, and these were done by regulating CCNA2-CDK2 complex and AURKA/PLK1 pathway. These findings are the first to demonstrate the molecular mechanism of TSA in treatment of LUAD combination of network bio-information analysis and biological experiments in vitro.
Collapse
Affiliation(s)
- Ziheng Li
- Faculty of Pharmacy, Hubei University of Chinese Medicine, Wuhan, 430065, China
| | - Ying Zhang
- Faculty of Pharmacy, Hubei University of Chinese Medicine, Wuhan, 430065, China
| | - Yuan Zhou
- Faculty of Pharmacy, Hubei University of Chinese Medicine, Wuhan, 430065, China
| | - Fuqian Wang
- Faculty of Pharmacy, Hubei University of Chinese Medicine, Wuhan, 430065, China
| | - Chao Yin
- Faculty of Pharmacy, Hubei University of Chinese Medicine, Wuhan, 430065, China
| | - Li Ding
- Faculty of Pharmacy, Hubei University of Chinese Medicine, Wuhan, 430065, China.
| | - Shunbo Zhang
- Faculty of Pharmacy, Hubei University of Chinese Medicine, Wuhan, 430065, China.
| |
Collapse
|
22
|
Wang J, Fan Z, Li J, Yang J, Liu X, Cheng J. Transcription factor specificity protein 1-mediated Serine/threonine kinase 39 upregulation promotes the proliferation, migration, invasion and epithelial-mesenchymal transition of hepatocellular carcinoma cells by activating the transforming growth factor-β1 /Smad2/3 pathway. Bioengineered 2021; 12:3566-3577. [PMID: 34281492 PMCID: PMC8806741 DOI: 10.1080/21655979.2021.1947939] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 06/21/2021] [Accepted: 06/22/2021] [Indexed: 02/08/2023] Open
Abstract
Bioinformatics analysis showed that Serine/threonine kinase 39 (STK39), which was testified to play an important role in human cancers, may be a hub gene in diagnosing hepatocellular carcinoma (HCC). This study aimed to explore whether STK39 could be regulated by specificity protein 1 (SP1) to affect HCC cells malignant processes. Firstly, STK39 expression in tissues of HCC patients and several cell lines was analyzed. After STK39 silencing, cell proliferation was evaluated by methyl thiazolyl tetrazolium and colony formation assay. Tunel staining was used to detect cell apoptosis. Then, the abilities of cell migration and invasion were determined with wound healing and transwell assays. The expression of epithelial-mesenchymal transition (EMT)-related proteins and transforming growth factor-β1 (TGF-β1)/Smad2/3 pathway proteins was tested by western blot analysis. Thereafter, cells were overexpressed with SP1 under the circumstance of STK39 knockdown, and then the above cellular processes were under observation. Results revealed that the increased expression of STK39, which was found in both HHC patients and HCC cell lines, exhibited poor HCC prognosis. STK39 silencing inhibited Hep3b cell proliferation, migration, invasion, EMT and TGF-β1/Smad2/3 expression but promoted cell apoptosis. Additionally, SP1 could bind to the STK39 promoter and facilitate STK39 expression. Further studies revealed that the effects of STK39 silencing on Hep3b cells were blocked by SP1 overexpression. In conclusion, SP1-mediated STK39 up-regulation leads to the increased proliferation, migration, invasion and EMT of HCC cells via activating TGF-β1/Smad2/3 pathway. Therapies that target SP1 to knockdown STK39 expression may contribute to the inhibition of HCC progression.
Collapse
Affiliation(s)
- Jing Wang
- Department of Hepatology, Tianjin Institute of Hepatology, Tianjin Second People’s Hospital, Tianjin, China
| | - Zhenyu Fan
- Department of Gastroenterology and Hepatology, Shanghai Public Health Clinical Center, Fudan University, Shanghai, China
| | - Jia Li
- Department of Hepatology, Tianjin Institute of Hepatology, Tianjin Second People’s Hospital, Tianjin, China
| | - Jingmao Yang
- Department of Gastroenterology and Hepatology, Shanghai Public Health Clinical Center, Fudan University, Shanghai, China
| | - Xiaofei Liu
- Department of Gastroenterology and Hepatology, Shanghai Public Health Clinical Center, Fudan University, Shanghai, China
| | - Jilin Cheng
- Department of Gastroenterology and Hepatology, Shanghai Public Health Clinical Center, Fudan University, Shanghai, China
| |
Collapse
|
23
|
Qin S, Ning M, Liu Q, Ding X, Wang Y, Liu Q. Knockdown of long non-coding RNA CDKN2B-AS1 suppresses the progression of breast cancer by miR-122-5p/STK39 axis. Bioengineered 2021; 12:5125-5137. [PMID: 34374638 PMCID: PMC8806778 DOI: 10.1080/21655979.2021.1962685] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Revised: 07/26/2021] [Accepted: 07/27/2021] [Indexed: 02/07/2023] Open
Abstract
The lncRNAs have been made certain to take part in the development of most cancers in multiple ways. Here, our purpose is to making observation of the biological role and function of lncRNA CDKN2B-AS1 in human breast cancer. Twenty-eight pairs of breast cancer tissue and adjacent normal tissue from breast cancer patients were used to investigate the expression of CDKN2B-AS1 by qRT-PCR. And a lentivirus-shRNA guided CDKN2B-AS1 were to reduce its expression. The function of CDKN2B-AS1 was analyzed using a series of in vitro assays. Meanwhile, the xenograft model was used to further explicate the role of CDKN2B-AS1 in breast cancer. As for the results, there is a relative rich expression of CDKN2B-AS1 in breast cancer tissues compared with the corresponding adjacent normal tissues. Compared with the human breast epithelial cell line, the abundant expression of CDKN2B-AS1 in breast cancer cells were revealed as well. Then, knockdown CDKN2B-AS1 inhibited the malignant biological behaviors of MCF7 and T47D cells. In mechanism, CDKN2B-AS1 sponged the miR-122-5p to regulate STK39 expression. Furthermore, the inhibition effect with sh-CDKN2B-AS1 on breast cancer cells was alleviated by miR-122-5p inhibitor. Last, an in vivo model also confirmed that knockdown CDKN2B-AS1 retarded the growth of breast cancer. Our data concluded that knockdown of CDKN2B-AS1 suppresses the progression of breast cancer by miR-122-5p/STK39 axis.
Collapse
Affiliation(s)
- Shaojie Qin
- The Third Departments of Tumor Surgery, General Hospital of Ningxia Medical University, Yinchuan City, Ningxia, China
| | - Mingliang Ning
- The Third Departments of Tumor Surgery, General Hospital of Ningxia Medical University, Yinchuan City, Ningxia, China
| | - Qingyuan Liu
- The Third Departments of Tumor Surgery, General Hospital of Ningxia Medical University, Yinchuan City, Ningxia, China
| | - Xiaoyun Ding
- The Third Departments of Tumor Surgery, General Hospital of Ningxia Medical University, Yinchuan City, Ningxia, China
| | - Yanbai Wang
- Cerebrospinal Fluid Laboratory; General Hospital of Ningxia Medical University, Yinchuan City, Ningxia, China
| | - Qilun Liu
- The Third Departments of Tumor Surgery, General Hospital of Ningxia Medical University, Yinchuan City, Ningxia, China
| |
Collapse
|
24
|
STK39 enhances the progression of Cholangiocarcinoma via PI3K/AKT pathway. iScience 2021; 24:103223. [PMID: 34746696 PMCID: PMC8551078 DOI: 10.1016/j.isci.2021.103223] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Revised: 09/10/2021] [Accepted: 09/30/2021] [Indexed: 02/07/2023] Open
Abstract
Serine/threonine kinase 39 (STK39) is overexpressed in various tumor tissues and plays an essential role in tumor progression. In this study, we investigated the clinical value, as well as the potential functions and mechanisms of STK39 in cholangiocarcinoma (CCA). The results showed that STK39 was overexpressed in CCA and negatively associated with the prognosis of patients with CCA. Functionally, STK39 knockdown suppressed cell proliferation, migration, and invasion, while STK39 overexpression facilitated tumor aggressiveness. The tumor-promoting effects of STK39 in CCA were also validated by in vivo experiments. Mechanistically, RNA-seq analysis identified that STK39 enhanced the progression of CCA by activating PI3K/AKT signaling pathway. Furthermore, overexpression of STK39 could induce gemcitabine resistance in CCA cells. Moreover, the increased expression of STK39 may be mediated by the dysregulation of miR-26a-5p. In summary, STK39 could be served as a valuable prognostic candidate and a potential therapeutic target of CCA. STK39 was overexpressed in CCA, negatively associated with the prognosis of patients with CCA STK39 knockdown suppressed cell proliferation and invasion. STK39 overexpression facilitated tumor aggressiveness STK39 mediates oncogenic effects on CCA cells by activating the PI3K/AKT signaling pathway STK39 reduces CCA sensitivity to gemcitabine. Increased expression of STK39 may be mediated by dysregulation of miR-26a-5p
Collapse
|
25
|
Huang Z, Wang S, Zhang HJ, Zhou YL, Shi JH. SMOX expression predicts the prognosis of non-small cell lung cancer. ANNALS OF TRANSLATIONAL MEDICINE 2021; 9:1048. [PMID: 34422960 PMCID: PMC8339854 DOI: 10.21037/atm-21-998] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Accepted: 05/21/2021] [Indexed: 12/16/2022]
Abstract
Background The development of non-small cell lung cancer (NSCLC) is very rapid, and the effect of its treatment is often closely related to the diagnosis time of the disease. Therefore, simple and convenient tumor biomarkers are helpful for the timely diagnosis and prevention of NSCLC. Methods Through univariate and multivariate Cox regression analyses, SMOX was determined as an independent prognostic factor of GSE42127, GSE41271, GSE68465, and TCGA datasets. Furthermore, western blot, reverse transcription-polymerase chain reaction (RT-PCR), and immunohistochemical analysis were performed to confirm the predictive efficiency of SMOX expression in NSCLC. Results Patients were divided into high and low expression groups according to the median value of SMOX expression, and Kaplan-Meier curves of multiple datasets indicated that patients with low SMOX expression had a better survival rate. According to the analysis of immune infiltration, the immune microenvironment, and immune checkpoints, SMOX expression of the high and low groups showed differences in immunity in NSCLC. By comparing cancer and adjacent tissues using western blot analysis, RT-PCR and immunohistochemical analysis, we found that SMOX was highly expressed in tumor tissues and had low expression in adjacent tissues. Simultaneously, the Kaplan-Meier curve suggested that among the 155 NSCLC patients, those with low SMOX expression had better survival. Conclusions SMOX can be used as an effective predictive target for NSCLC.
Collapse
Affiliation(s)
- Zhanghao Huang
- Nantong Key Laboratory of Translational Medicine in Cardiothoracic Diseases, and Research Institution of Translational Medicine in Cardiothoracic Diseases, Affiliated Hospital of Nantong University, Nantong, China.,Department of Thoracic Surgery, Affiliated Hospital of Nantong University, Nantong, China.,Medical College of Nantong University, Nantong, China
| | - Shuo Wang
- Nantong Key Laboratory of Translational Medicine in Cardiothoracic Diseases, and Research Institution of Translational Medicine in Cardiothoracic Diseases, Affiliated Hospital of Nantong University, Nantong, China.,Department of Thoracic Surgery, Affiliated Hospital of Nantong University, Nantong, China.,Medical College of Nantong University, Nantong, China
| | - Hai-Jian Zhang
- Research Center of Clinical Medicine, Affiliated Hospital of Nantong University, Nantong, China
| | - You Lang Zhou
- Research Center of Clinical Medicine, Affiliated Hospital of Nantong University, Nantong, China
| | - Jia-Hai Shi
- Nantong Key Laboratory of Translational Medicine in Cardiothoracic Diseases, and Research Institution of Translational Medicine in Cardiothoracic Diseases, Affiliated Hospital of Nantong University, Nantong, China.,Department of Thoracic Surgery, Affiliated Hospital of Nantong University, Nantong, China
| |
Collapse
|