1
|
Kang T, Chen J, Wan W, Pang J, Wen R, Bai X, Li L, Pan Y, He Y, Yang H. Overexpression of PLK1 Molecule Following Incomplete Thermal Ablation Promotes the Proliferation and Invasion of Residual Hepatocellular Carcinoma. Mol Biotechnol 2025; 67:2046-2059. [PMID: 38782874 DOI: 10.1007/s12033-024-01181-y] [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: 01/20/2024] [Accepted: 04/15/2024] [Indexed: 05/25/2024]
Abstract
TAT, a widely used treatment for HCC, can exacerbate the progression of residual HCC. The present study investigated the mechanism of action of PLK1 following ITA of HCC. The PLK1 levels in HCC were determined using qRT-PCR from clinical patient samples, IHC from tissue microarray, and data from globally high-throughput data and microarrays. The PLK1 levels and their effect on the biological phenotype of heat-stress HCC cells were evaluated through in vitro experiments. We detected PLK1 abnormal expression in HCC models of nude mice subjected to ITA. We detected the effects of different PLK1 expression levels on EMT pathway proteins. PLK1 exhibited an overexpression in HCC tissues with an SMD of 1.19 (3414 HCC and 3036 non-HCC tissues were included), distinguishing HCC from non-HCC effectively (AUC = 0.9). The qRT-PCR data from clinical HCC patient samples and IHC from HCC tissue microarray results also indicated an overexpressed level. In the incomplete ablation models, an increased PLK1 expression was found in both heat-stress cells and subcutaneous tumors. The upregulation of PLK1 following ITA was found to enhance the malignancy of HCC and exacerbate the proliferation, migration, and invasion of residual HCC cells, whereas PLK1 knockdown suppressed the biological malignancy of HCC cells. Meanwhile, PLK1 has different regulatory effects on various EMT pathway proteins. PLK1 promotes the progression of residual HCC by activating EMT pathway after ITA, which might provide a novel idea for the treatment and prognosis of residual HCC.
Collapse
Affiliation(s)
- Tong Kang
- Department of Medical Ultrasound, First Affiliated Hospital of Guangxi Medical University, 6 Shuangyong Road, Guangxi Zhuang Autonomous Region, Nanning, China
- Guangxi Key Laboratory of Early Prevention and Treatment for Regional High Frequency Tumor/Key Laboratory of Early Prevention and Treatment for Regional High Frequency Tumor, Guangxi Medical University, Nanning, China
| | - Jiamin Chen
- Department of Medical Ultrasound, First Affiliated Hospital of Guangxi Medical University, 6 Shuangyong Road, Guangxi Zhuang Autonomous Region, Nanning, China
| | - Weijun Wan
- Department of Medical Ultrasound, First Affiliated Hospital of Guangxi Medical University, 6 Shuangyong Road, Guangxi Zhuang Autonomous Region, Nanning, China
| | - Jinshu Pang
- Department of Medical Ultrasound, First Affiliated Hospital of Guangxi Medical University, 6 Shuangyong Road, Guangxi Zhuang Autonomous Region, Nanning, China
| | - Rong Wen
- Department of Medical Ultrasound, First Affiliated Hospital of Guangxi Medical University, 6 Shuangyong Road, Guangxi Zhuang Autonomous Region, Nanning, China
| | - Xiumei Bai
- Department of Medical Ultrasound, First Affiliated Hospital of Guangxi Medical University, 6 Shuangyong Road, Guangxi Zhuang Autonomous Region, Nanning, China
| | - Lipeng Li
- Department of Medical Ultrasound, First Affiliated Hospital of Guangxi Medical University, 6 Shuangyong Road, Guangxi Zhuang Autonomous Region, Nanning, China
| | - Yunjing Pan
- Department of Medical Ultrasound, First Affiliated Hospital of Guangxi Medical University, 6 Shuangyong Road, Guangxi Zhuang Autonomous Region, Nanning, China
| | - Yun He
- Department of Medical Ultrasound, First Affiliated Hospital of Guangxi Medical University, 6 Shuangyong Road, Guangxi Zhuang Autonomous Region, Nanning, China.
- Guangxi Key Laboratory of Early Prevention and Treatment for Regional High Frequency Tumor/Key Laboratory of Early Prevention and Treatment for Regional High Frequency Tumor, Guangxi Medical University, Nanning, China.
| | - Hong Yang
- Department of Medical Ultrasound, First Affiliated Hospital of Guangxi Medical University, 6 Shuangyong Road, Guangxi Zhuang Autonomous Region, Nanning, China.
- Collaborative Innovation Centre of Regenerative Medicine and Medical BioResource Development and Application Co-constructed by the Province and Ministry, Guangxi Medical University, Nanning, China.
- Guangxi Key Laboratory of Early Prevention and Treatment for Regional High Frequency Tumor/Key Laboratory of Early Prevention and Treatment for Regional High Frequency Tumor, Guangxi Medical University, Nanning, China.
| |
Collapse
|
2
|
Ding J, Yang A, Zhou L, Zhang F, Zhou H, Zhang Y, Wang Y, Liu Y, Liang D, Liu Y, Wu Y. PLK1 Downregulation Attenuates ET-1-Induced Cardiomyocyte Hypertrophy by Suppressing the ERK1/2 Pathway. J Cardiovasc Transl Res 2025:10.1007/s12265-025-10604-3. [PMID: 40095199 DOI: 10.1007/s12265-025-10604-3] [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: 01/14/2025] [Accepted: 03/05/2025] [Indexed: 03/19/2025]
Abstract
Cardiomyocyte hypertrophy is a key remodeling response to cardiac stress and an independent risk factor for heart failure. However, the molecular mechanism of cardiomyocyte hypertrophy is not yet fully understood. We here found Polo-like kinase 1 (PLK1) was crucial in regulating endothelin-1 (ET-1)-induced cardiomyocyte hypertrophy. Notably, PLK1 expression was significantly elevated in ET-1-induced hypertrophic cardiomyocytes and pressure overload-induced hypertrophic cardiac tissue. Knocking down Plk1 reduced the cell size of hypertrophic cardiomyocytes and suppressed the expression of hypertrophic markers, including ANP, BNP and β-MHC. The PLK1 inhibitor BI2536 had similar effects on hypertrophic cardiomyocytes. Mechanistically, the ERK1/2 pathway was identified as the key downstream pathway mediating the effects of PLK1 on ET-1-induced cardiomyocyte hypertrophy. Finally, the deficiency of PLK1 attenuated the hypertrophy of hiPSC-CMs. In summary, our study revealed that PLK1 regulates ET-1-induced cardiomyocyte hypertrophy through the ERK1/2 pathway, providing insights into the pathogenesis and potential therapies for pathological cardiac hypertrophy.
Collapse
Affiliation(s)
- Jie Ding
- Department of Cardiology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200120, China
| | - Anqi Yang
- Department of Cardiology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200120, China
| | - Liping Zhou
- Department of Cardiology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200120, China
| | - Fulei Zhang
- Department of Cardiology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200120, China
| | - Huixing Zhou
- Department of Cardiology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200120, China
| | - Yuemei Zhang
- Department of Cardiology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200120, China
| | - Yan Wang
- Jinzhou Medical University, Jinzhou, 121000, China
| | - Yi Liu
- Department of Cardiology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200120, China
| | - Dandan Liang
- Department of Cardiology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200120, China
| | - Yuanyuan Liu
- Jinzhou Medical University, Jinzhou, 121000, China.
| | - Yahan Wu
- Department of Cardiology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200120, China.
| |
Collapse
|
3
|
Oskomić M, Tomić A, Barbarić L, Matić A, Kindl DC, Matovina M. KEAP1-NRF2 Interaction in Cancer: Competitive Interactors and Their Role in Carcinogenesis. Cancers (Basel) 2025; 17:447. [PMID: 39941813 PMCID: PMC11816071 DOI: 10.3390/cancers17030447] [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: 12/23/2024] [Revised: 01/21/2025] [Accepted: 01/24/2025] [Indexed: 02/16/2025] Open
Abstract
An American Cancer Society report estimates the emergence of around 2 million new cancer cases in the US in 2024 [...].
Collapse
Affiliation(s)
| | | | | | | | | | - Mihaela Matovina
- Division of Organic Chemistry and Biochemistry, Ruđer Bošković Institute, 10000 Zagreb, Croatia; (M.O.); (A.T.); (L.B.); (A.M.); (D.C.K.)
| |
Collapse
|
4
|
Wang M, Huang Z, Li X, He P, Sun H, Peng Y, Fan Q. Apabetalone, a BET protein inhibitor, inhibits kidney damage in diabetes by preventing pyroptosis via modulating the P300/H3K27ac/PLK1 axis. Pharmacol Res 2024; 207:107306. [PMID: 39002871 DOI: 10.1016/j.phrs.2024.107306] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/24/2024] [Revised: 06/11/2024] [Accepted: 07/10/2024] [Indexed: 07/15/2024]
Abstract
Many inflammatory disorders, including diabetic kidney disease (DKD), are associated with pyroptosis, a type of inflammation-regulated cell death. The purpose of this work was to ascertain the effects of apabetalone, which targets BRD4, a specific inhibitor of the bromodomain (BRD) and extra-terminal (BET) proteins that target bromodomain 2, on kidney injury in DKD. This study utilized pharmacological and genetic approaches to investigate the effects of apabetalone on pyroptosis in db/db mice and human tubular epithelial cells (HK-2). BRD4 levels were elevated in HK-2 cells exposed to high glucose and in db/db mice. Modulating BRD4 levels led to changes in the generation of inflammatory cytokines and cell pyroptosis linked to NLRP3 inflammasome in HK-2 cells and db/db mice. Likewise, these cellular processes were mitigated by apabetalone through inhibition BRD4. Apabetalone or BRD4 siRNA suppressed PLK1 expression in HK-2 cells under high glucose by P300-dependent H3K27 acetylation on the PLK1 gene promoter, as demonstrated through chromatin immunoprecipitation and immunoprecipitation assays. To summarize, apabetalone relieves renal proptosis and fibrosis in DKD. BRD4 regulates the P300/H3K27ac/PLK1 axis, leading to the activation of the NLRP3 inflammasome and subsequent cell pyroptosis, inflammation, and fibrosis. These results may provide new perspectives on DKD treatment.
Collapse
Affiliation(s)
- Min Wang
- Department of Nephrology, First Hospital of China Medical University, Shenyang, Liaoning, China
| | - Zhaohui Huang
- Department of Nephrology, First Hospital of China Medical University, Shenyang, Liaoning, China
| | - Xin Li
- Department of Nephrology, Fourth Hospital of China Medical University, Shenyang, Liaoning, China
| | - Ping He
- Department of Nephrology, First Hospital of China Medical University, Shenyang, Liaoning, China
| | - He Sun
- Department of Endocrinology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Yali Peng
- Department of Nephrology, First Hospital of China Medical University, Shenyang, Liaoning, China
| | - QiuLing Fan
- Department of Nephrology, First Hospital of China Medical University, Shenyang, Liaoning, China; Department of Nephrology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| |
Collapse
|
5
|
Kulkarni H, Dagar N, Gaikwad AB. Targeting polo-like kinase 1 to treat kidney diseases. Cell Biochem Funct 2024; 42:e4099. [PMID: 39016459 DOI: 10.1002/cbf.4099] [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: 05/27/2024] [Revised: 06/20/2024] [Accepted: 07/10/2024] [Indexed: 07/18/2024]
Abstract
Globally, ∼850 million individuals suffer from some form of kidney disease. This staggering figure underscores the importance of continued research and innovation in the field of nephrology to develop effective treatments and improve overall global kidney health. In current research, the polo-like kinase (Plk) family has emerged as a group of highly conserved enzyme kinases vital for proper cell cycle regulation. Plks are defined by their N-terminal kinase domain and C-terminal polo-box domain, which regulate their catalytic activity, subcellular localization, and substrate recognition. Among the Plk family members, Plk1 has garnered significant attention due to its pivotal role in regulating multiple mitotic processes, particularly in the kidneys. It is a crucial serine-threonine (Ser-Thr) kinase involved in cell division and genomic stability. In this review, we delve into the types and functions of Plks, focusing on Plk1's significance in processes such as cell proliferation, spindle assembly, and DNA damage repair. The review also underscores Plk1's vital contributions to maintaining kidney homeostasis, elucidating its involvement in nuclear envelope breakdown, anaphase-promoting complex/cyclosome activation, and the regulation of mRNA translation machinery. Furthermore, the review discusses how Plk1 contributes to the development and progression of kidney diseases, emphasizing its overexpression in conditions such as acute kidney injury, chronic kidney disease, and so forth. It also highlights the importance of exploring Plk1 modulators as targeted therapies for kidney diseases in future. This review will help in understanding the role of Plk1 in kidney disease development, paving the way for the discovery and development of novel therapeutic approaches to manage kidney diseases effectively.
Collapse
Affiliation(s)
- Hrushikesh Kulkarni
- Department of Pharmacy, Birla Institute of Technology and Science, Pilani, Rajasthan, India
| | - Neha Dagar
- Department of Pharmacy, Birla Institute of Technology and Science, Pilani, Rajasthan, India
| | - Anil Bhanudas Gaikwad
- Department of Pharmacy, Birla Institute of Technology and Science, Pilani, Rajasthan, India
| |
Collapse
|
6
|
Huang QR, Jiang Q, Tan JY, Nong RB, Yan J, Yang XW, Mo LG, Ling GY, Deng T, Gong YZ. The prognostic and immunological role of MCM3 in pan-cancer and validation of prognosis in a clinical lower-grade glioma cohort. Front Pharmacol 2024; 15:1390615. [PMID: 38698811 PMCID: PMC11063780 DOI: 10.3389/fphar.2024.1390615] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2024] [Accepted: 04/05/2024] [Indexed: 05/05/2024] Open
Abstract
Background: Previous studies have shown that MCM3 plays a key role in initiating DNA replication. However, the mechanism of MCM3 function in most cancers is still unknown. The aim of our study was to explore the expression, prognostic role, and immunological characteristics of MCM3 across cancers. Methods: We explored the expression pattern of MCM3 across cancers. We subsequently explored the prognostic value of MCM3 expression by using univariate Cox regression analysis. Spearman correlation analysis was performed to determine the correlations between MCM3 and immune-related characteristics, mismatching repair (MMR) signatures, RNA modulator genes, cancer stemness, programmed cell death (PCD) gene expression, tumour mutation burden (TMB), microsatellite instability (MSI), and neoantigen levels. The role of MCM3 in predicting the response to immune checkpoint blockade (ICB) therapy was further evaluated in four immunotherapy cohorts. Single-cell data from CancerSEA were analysed to assess the biological functions associated with MCM3 in 14 cancers. The clinical correlation and independent prognostic significance of MCM3 were further analysed in the TCGA and CGGA lower-grade glioma (LGG) cohorts, and a prognostic nomogram was constructed. Immunohistochemistry in a clinical cohort was utilized to validate the prognostic utility of MCM3 expression in LGG. Results: MCM3 expression was upregulated in most tumours and strongly associated with patient outcomes in many cancers. Correlation analyses demonstrated that MCM3 expression was closely linked to immune cell infiltration, immune checkpoints, MMR genes, RNA modulator genes, cancer stemness, PCD genes and the TMB in most tumours. There was an obvious difference in outcomes between patients with high MCM3 expression and those with low MCM3 expression in the 4 ICB treatment cohorts. Single-cell analysis indicated that MCM3 was mainly linked to the cell cycle, DNA damage and DNA repair. The expression of MCM3 was associated with the clinical features of LGG patients and was an independent prognostic indicator. Finally, the prognostic significance of MCM3 in LGG was validated in a clinical cohort. Conclusion: Our study suggested that MCM3 can be used as a potential prognostic marker for cancers and may be associated with tumour immunity. In addition, MCM3 is a promising predictor of immunotherapy responses.
Collapse
Affiliation(s)
- Qian-Rong Huang
- Department of Neurosurgery, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Qian Jiang
- Department of Neurosurgery, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Ju-Yuan Tan
- Department of Neurosurgery, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Ren-Bao Nong
- Department of Neurosurgery, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Jun Yan
- Department of Neurosurgery, Guangxi Medical University Cancer Hospital, Nanning, China
| | | | - Li-Gen Mo
- Department of Neurosurgery, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Guo-Yuan Ling
- Department of Neurosurgery, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Teng Deng
- Department of Neurosurgery, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Yi-Zhen Gong
- Department of Clinical Research, Guangxi Medical University Cancer Hospital, Nanning, China
| |
Collapse
|
7
|
Shan Y, Zheng L, Zhang S, Qian B. Abnormal expression of FOXM1 in carcinogenesis of renal cell carcinoma: From experimental findings to clinical applications. Biochem Biophys Res Commun 2024; 692:149251. [PMID: 38056162 DOI: 10.1016/j.bbrc.2023.149251] [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: 09/12/2023] [Revised: 11/07/2023] [Accepted: 11/13/2023] [Indexed: 12/08/2023]
Abstract
Renal cell carcinoma (RCC) is a prevalent malignancy within the genitourinary system. At present, patients with high-grade or advanced RCC continue to have a bleak prognosis. Mounting research have emphasized the significant involvement of Forkhead box M1 (FOXM1) in RCC development and progression. Therefore, it is imperative to consolidate the existing evidence regarding the contributions of FOXM1 to RCC tumorigenesis through a comprehensive review. This study elucidated the essential functions of FOXM1 in promoting RCC growth, invasion, and metastasis by regulating cell cycle progression, DNA repair, angiogenesis, and epithelial-mesenchymal transition (EMT). Also, FOXM1 might serve as a novel diagnostic and prognostic biomarker as well as a therapeutic target for RCC. Clinical findings demonstrated that the expression of FOXM1 was markedly upregulated in RCC samples, while a high level of FOXM1 was found to be associated with a poor overall survival rate of RCC. Furthermore, it is worth noting that FOXM1 may have a significant impact on the resistance of renal cell carcinoma (RCC) to radiotherapy. This observation suggests that inhibiting FOXM1 could be a promising strategy to impede the progression of RCC and enhance its sensitivity to radiotherapy. The present review highlighted the pivotal role of FOXM1 in RCC development. FOXM1 has the capacity to emerge as not only a valuable diagnostic and prognostic tool but also a viable therapeutic option for unresectable RCC.
Collapse
Affiliation(s)
- Yanmei Shan
- Department of Nephrology, Taizhou Central Hospital (Taizhou University Hospital), Taizhou, 318000, China
| | - Liying Zheng
- Postgraduate Department, First Affiliated Hospital of Gannan Medical College, Ganzhou, China
| | - Shilong Zhang
- Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Biao Qian
- Department of Urology, First Affiliated Hospital of Gannan Medical University, Ganzhou, 341000, Jiangxi, China; Key Laboratory of Urology and Andrology of Ganzhou, Ganzhou, 341000, Jiangxi, China
| |
Collapse
|
8
|
Gong W, Liu X, Lv X, Zhang Y, Niu Y, Jin K, Li B, Zuo Q. Ubiquitination plays an important role during the formation of chicken primordial germ cells. J Anim Sci 2024; 102:skae251. [PMID: 39187982 PMCID: PMC11452721 DOI: 10.1093/jas/skae251] [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: 06/24/2024] [Accepted: 08/24/2024] [Indexed: 08/28/2024] Open
Abstract
As an important posttranslational modification, ubiquitination plays an important role in regulating protein homeostasis in eukaryotic cells. In our previous studies, both the transcriptome and proteome suggested that ubiquitination is involved in the formation of chicken primordial germ cells (PGCs). Here, affinity enrichment combined with liquid chromatography-tandem mass spectrometry (MS/MS) was used to analyze the ubiquitome during the differentiation from embryonic stem cells to PGCs, and we identify that 724 lysine ubiquitinated sites were up-regulated in 558 proteins and 138 lysine ubiquitinated sites were down-regulated in 109 proteins. Furthermore, GO and KEGG enrichment analysis showed that ubiquitination regulates key proteins to participate in the progression of key events related to PGC formation and the transduction of key signals such as Wnt, MAPK, and insulin signals, followed by the detailed explanation of the specific regulatory mechanism of ubiquitination through the combined proteome and ubiquitome analysis. Moreover, both the activation and inhibition of neddylation were detrimental to the maintenance of the biological characteristics of PGCs, which also verified the importance of ubiquitination. In conclusion, this study provides a global view of the ubiquitome during the formation of PGCs by label-free quantitative ubiquitomics, which lays a theoretical foundation for the formation mechanism and specific application of chicken PGCs.
Collapse
Affiliation(s)
- Wei Gong
- Joint International Research Laboratory of Agriculture and Agri-Product Safety of Ministry of Education of China, Yangzhou University, Yangzhou, Jiangsu, P.R. China
- Key Laboratory of Animal Breeding Reproduction and Molecular Design for Jiangsu Province, College of Animal Science and Technology, Yangzhou University, Yangzhou, Jiangsu, P.R. China
| | - Xin Liu
- Joint International Research Laboratory of Agriculture and Agri-Product Safety of Ministry of Education of China, Yangzhou University, Yangzhou, Jiangsu, P.R. China
- Key Laboratory of Animal Breeding Reproduction and Molecular Design for Jiangsu Province, College of Animal Science and Technology, Yangzhou University, Yangzhou, Jiangsu, P.R. China
| | - Xiaoqian Lv
- Joint International Research Laboratory of Agriculture and Agri-Product Safety of Ministry of Education of China, Yangzhou University, Yangzhou, Jiangsu, P.R. China
- Key Laboratory of Animal Breeding Reproduction and Molecular Design for Jiangsu Province, College of Animal Science and Technology, Yangzhou University, Yangzhou, Jiangsu, P.R. China
| | - Yani Zhang
- Joint International Research Laboratory of Agriculture and Agri-Product Safety of Ministry of Education of China, Yangzhou University, Yangzhou, Jiangsu, P.R. China
- Key Laboratory of Animal Breeding Reproduction and Molecular Design for Jiangsu Province, College of Animal Science and Technology, Yangzhou University, Yangzhou, Jiangsu, P.R. China
| | - Yingjie Niu
- Joint International Research Laboratory of Agriculture and Agri-Product Safety of Ministry of Education of China, Yangzhou University, Yangzhou, Jiangsu, P.R. China
- Key Laboratory of Animal Breeding Reproduction and Molecular Design for Jiangsu Province, College of Animal Science and Technology, Yangzhou University, Yangzhou, Jiangsu, P.R. China
| | - Kai Jin
- Joint International Research Laboratory of Agriculture and Agri-Product Safety of Ministry of Education of China, Yangzhou University, Yangzhou, Jiangsu, P.R. China
- Key Laboratory of Animal Breeding Reproduction and Molecular Design for Jiangsu Province, College of Animal Science and Technology, Yangzhou University, Yangzhou, Jiangsu, P.R. China
| | - Bichun Li
- Joint International Research Laboratory of Agriculture and Agri-Product Safety of Ministry of Education of China, Yangzhou University, Yangzhou, Jiangsu, P.R. China
- Key Laboratory of Animal Breeding Reproduction and Molecular Design for Jiangsu Province, College of Animal Science and Technology, Yangzhou University, Yangzhou, Jiangsu, P.R. China
| | - Qisheng Zuo
- Joint International Research Laboratory of Agriculture and Agri-Product Safety of Ministry of Education of China, Yangzhou University, Yangzhou, Jiangsu, P.R. China
- Key Laboratory of Animal Breeding Reproduction and Molecular Design for Jiangsu Province, College of Animal Science and Technology, Yangzhou University, Yangzhou, Jiangsu, P.R. China
| |
Collapse
|
9
|
Anselmi C, Caicci F, Bocci T, Guidetti M, Priori A, Giusti V, Levy T, Raveh T, Voskoboynik A, Weissman IL, Manni L. Multiple Forms of Neural Cell Death in the Cyclical Brain Degeneration of A Colonial Chordate. Cells 2023; 12:1041. [PMID: 37048113 PMCID: PMC10093557 DOI: 10.3390/cells12071041] [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: 02/24/2023] [Revised: 03/20/2023] [Accepted: 03/24/2023] [Indexed: 03/31/2023] Open
Abstract
Human neuronal loss occurs through different cellular mechanisms, mainly studied in vitro. Here, we characterized neuronal death in B. schlosseri, a marine colonial tunicate that shares substantial genomic homology with mammals and has a life history in which controlled neurodegeneration happens simultaneously in the brains of adult zooids during a cyclical phase named takeover. Using an ultrastructural and transcriptomic approach, we described neuronal death forms in adult zooids before and during the takeover phase while comparing adult zooids in takeover with their buds where brains are refining their structure. At takeover, we found in neurons clear morphologic signs of apoptosis (i.e., chromatin condensation, lobed nuclei), necrosis (swollen cytoplasm) and autophagy (autophagosomes, autolysosomes and degradative multilamellar bodies). These results were confirmed by transcriptomic analyses that highlighted the specific genes involved in these cell death pathways. Moreover, the presence of tubulovesicular structures in the brain medulla alongside the over-expression of prion disease genes in late cycle suggested a cell-to-cell, prion-like propagation recalling the conformational disorders typical of some human neurodegenerative diseases. We suggest that improved understanding of how neuronal alterations are regulated in the repeated degeneration-regeneration program of B. schlosseri may yield mechanistic insights relevant to the study of human neurodegenerative diseases.
Collapse
Affiliation(s)
- Chiara Anselmi
- Hopkins Marine Station, Institute for Stem Cell Biology and Regenerative Medicine, Stanford University, Pacific Grove, CA 93950, USA
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
- Wu Tsai Neurosciences Institute, Stanford University, Stanford, CA 94305, USA
| | - Federico Caicci
- Dipartimento di Biologia, Università degli Studi di Padova, 35131 Padova, Italy
| | - Tommaso Bocci
- “Aldo Ravelli” Center for Neurotechnology and Experimental Brain Therapeutics, Department of Health Sciences, University of Milan, 20142 Milan, Italy
| | - Matteo Guidetti
- “Aldo Ravelli” Center for Neurotechnology and Experimental Brain Therapeutics, Department of Health Sciences, University of Milan, 20142 Milan, Italy
- Department of Electronics, Information and Bioengineering, Politecnico di Milano, 20133 Milan, Italy
| | - Alberto Priori
- “Aldo Ravelli” Center for Neurotechnology and Experimental Brain Therapeutics, Department of Health Sciences, University of Milan, 20142 Milan, Italy
| | | | - Tom Levy
- Hopkins Marine Station, Institute for Stem Cell Biology and Regenerative Medicine, Stanford University, Pacific Grove, CA 93950, USA
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Tal Raveh
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Ayelet Voskoboynik
- Hopkins Marine Station, Institute for Stem Cell Biology and Regenerative Medicine, Stanford University, Pacific Grove, CA 93950, USA
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
- Chan Zuckerberg Biohub, San Francisco, CA 94158, USA
| | - Irving L. Weissman
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
- Department of Pathology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Lucia Manni
- Wu Tsai Neurosciences Institute, Stanford University, Stanford, CA 94305, USA
| |
Collapse
|
10
|
Boi D, Rubini E, Breccia S, Guarguaglini G, Paiardini A. When Just One Phosphate Is One Too Many: The Multifaceted Interplay between Myc and Kinases. Int J Mol Sci 2023; 24:4746. [PMID: 36902175 PMCID: PMC10003727 DOI: 10.3390/ijms24054746] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2023] [Revised: 02/19/2023] [Accepted: 02/21/2023] [Indexed: 03/06/2023] Open
Abstract
Myc transcription factors are key regulators of many cellular processes, with Myc target genes crucially implicated in the management of cell proliferation and stem pluripotency, energy metabolism, protein synthesis, angiogenesis, DNA damage response, and apoptosis. Given the wide involvement of Myc in cellular dynamics, it is not surprising that its overexpression is frequently associated with cancer. Noteworthy, in cancer cells where high Myc levels are maintained, the overexpression of Myc-associated kinases is often observed and required to foster tumour cells' proliferation. A mutual interplay exists between Myc and kinases: the latter, which are Myc transcriptional targets, phosphorylate Myc, allowing its transcriptional activity, highlighting a clear regulatory loop. At the protein level, Myc activity and turnover is also tightly regulated by kinases, with a finely tuned balance between translation and rapid protein degradation. In this perspective, we focus on the cross-regulation of Myc and its associated protein kinases underlying similar and redundant mechanisms of regulation at different levels, from transcriptional to post-translational events. Furthermore, a review of the indirect effects of known kinase inhibitors on Myc provides an opportunity to identify alternative and combined therapeutic approaches for cancer treatment.
Collapse
Affiliation(s)
- Dalila Boi
- Department of Biochemical Sciences, Sapienza University of Rome, 00185 Rome, Italy
| | - Elisabetta Rubini
- Department of Biochemical Sciences, Sapienza University of Rome, 00185 Rome, Italy
| | - Sara Breccia
- Department of Biochemical Sciences, Sapienza University of Rome, 00185 Rome, Italy
| | - Giulia Guarguaglini
- Institute of Molecular Biology and Pathology, National Research Council of Italy, Sapienza University of Rome, 00185 Rome, Italy
| | - Alessandro Paiardini
- Department of Biochemical Sciences, Sapienza University of Rome, 00185 Rome, Italy
| |
Collapse
|
11
|
Xia Y, An J, Li J, Gu W, Zhang Y, Zhao S, Zhao C, Xu Y, Li B, Zhong Z, Meng F. Transferrin-guided intelligent nanovesicles augment the targetability and potency of clinical PLK1 inhibitor to acute myeloid leukemia. Bioact Mater 2023; 21:499-510. [PMID: 36185744 PMCID: PMC9494038 DOI: 10.1016/j.bioactmat.2022.08.032] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Revised: 08/24/2022] [Accepted: 08/28/2022] [Indexed: 11/28/2022] Open
Abstract
Acute myeloid leukemia (AML) remains a most lethal hematological malignancy, partly because of its slow development of targeted therapies compared with other cancers. PLK1 inhibitor, volasertib (Vol), is among the few molecular targeted drugs granted breakthrough therapy status for AML; however, its fast clearance and dose-limiting toxicity greatly restrain its clinical benefits. Here, we report that transferrin-guided polymersomes (TPs) markedly augment the targetability, potency and safety of Vol to AML. Vol-loaded TPs (TPVol) with 4% transferrin exhibited best cellular uptake, effective down-regulation of p-PLK1, p-PTEN and p-AKT and superior apoptotic activity to free Vol in MV-4-11 leukemic cells. Intravenous injection of TPVol gave 6-fold higher AUC than free Vol and notable accumulation in AML-residing bone marrow. The efficacy studies in orthotopic MV-4-11 leukemic model demonstrated that TPVol significantly reduced leukemic cell proportions in periphery blood, bone marrow, liver and spleen, effectively enhanced mouse survival rate, and impeded bone loss. This transferrin-guided nano-delivery of molecular targeted drugs appears to be an interesting strategy towards the development of novel treatments for AML.
Collapse
Affiliation(s)
- Yifeng Xia
- Biomedical Polymers Laboratory, College of Chemistry, Chemical Engineering and Materials Science, and State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou, 215123, PR China
- College of Pharmaceutical Sciences, Soochow University, Suzhou, 215123, PR China
| | - Jingnan An
- Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Institute of Blood and Marrow Transplantation of Soochow University, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, 215123, PR China
| | - Jiaying Li
- Orthopedic Institute, Soochow University, Suzhou, 215007, PR China
| | - Wenxing Gu
- Biomedical Polymers Laboratory, College of Chemistry, Chemical Engineering and Materials Science, and State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou, 215123, PR China
| | - Yifan Zhang
- Biomedical Polymers Laboratory, College of Chemistry, Chemical Engineering and Materials Science, and State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou, 215123, PR China
| | - Songsong Zhao
- Biomedical Polymers Laboratory, College of Chemistry, Chemical Engineering and Materials Science, and State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou, 215123, PR China
| | - Cenzhu Zhao
- Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Institute of Blood and Marrow Transplantation of Soochow University, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, 215123, PR China
| | - Yang Xu
- Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Institute of Blood and Marrow Transplantation of Soochow University, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, 215123, PR China
| | - Bin Li
- Orthopedic Institute, Soochow University, Suzhou, 215007, PR China
| | - Zhiyuan Zhong
- Biomedical Polymers Laboratory, College of Chemistry, Chemical Engineering and Materials Science, and State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou, 215123, PR China
- College of Pharmaceutical Sciences, Soochow University, Suzhou, 215123, PR China
| | - Fenghua Meng
- Biomedical Polymers Laboratory, College of Chemistry, Chemical Engineering and Materials Science, and State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou, 215123, PR China
- College of Pharmaceutical Sciences, Soochow University, Suzhou, 215123, PR China
| |
Collapse
|
12
|
Zheng Z, Li X, Nie K, Wang X, Liang W, Yang F, Zheng K, Zheng Y. Identification of berberine as a potential therapeutic strategy for kidney clear cell carcinoma and COVID-19 based on analysis of large-scale datasets. Front Immunol 2023; 14:1038651. [PMID: 37033923 PMCID: PMC10076552 DOI: 10.3389/fimmu.2023.1038651] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2022] [Accepted: 03/14/2023] [Indexed: 04/11/2023] Open
Abstract
Background Regarding the global coronavirus disease 2019 (COVID)-19 pandemic, kidney clear cell carcinoma (KIRC) has acquired a higher infection probability and may induce fatal complications and death following COVID-19 infection. However, effective treatment strategies remain unavailable. Berberine exhibits significant antiviral and antitumour effects. Thus, this study aimed to provide a promising and reliable therapeutic strategy for clinical decision-making by exploring the therapeutic mechanism of berberine against KIRC/COVID-19. Methods Based on large-scale data analysis, the target genes, clinical risk, and immune and pharmacological mechanisms of berberine against KIRC/COVID-19 were systematically investigated. Results In total, 1,038 and 12,992 differentially expressed genes (DEGs) of COVID-19 and KIRC, respectively, were verified from Gene Expression Omnibus and The Cancer Genome Atlas databases, respectively, and 489 berberine target genes were obtained from official websites. After intersecting, 26 genes were considered potential berberine therapeutic targets for KIRC/COVID-19. Berberine mechanism of action against KIRC/COVID-19 was revealed by protein-protein interaction, gene ontology, and Kyoto Encyclopedia of Genes and Genomes with terms including protein interaction, cell proliferation, viral carcinogenesis, and the PI3K/Akt signalling pathway. In COVID-19 patients, ACOX1, LRRK2, MMP8, SLC1A3, CPT1A, H2AC11, H4C8, and SLC1A3 were closely related to disease severity, and the general survival of KIRC patients was closely related to ACOX1, APP, CPT1A, PLK1, and TYMS. Additionally, the risk signature accurately and sensitively depicted the overall survival and patient survival status for KIRC. Numerous neutrophils were enriched in the immune system of COVID-19 patients, and the lives of KIRC patients were endangered due to significant immune cell infiltration. Molecular docking studies indicated that berberine binds strongly to target proteins. Conclusion This study demonstrated berberine as a potential treatment option in pharmacological, immunological, and clinical practice. Moreover, its therapeutic effects may provide potential and reliable treatment options for patients with KIRC/COVID-19.
Collapse
Affiliation(s)
- Zhihua Zheng
- Department of Nephrology, Shenzhen Traditional Chinese Medicine Hospital, The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen, Guangdong, China
| | - Xiushen Li
- Shenzhen Key Laboratory, Shenzhen University General Hospital, Shenzhen, Guangdong, China
| | - Kechao Nie
- Department of Gastroenterology, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Xiaoyu Wang
- Department of Nephrology, Health College of Guangdong Pharmaceutical University, Guangzhou, Guangdong, China
| | - Wencong Liang
- Department of Nephrology, Shenzhen Traditional Chinese Medicine Hospital, The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen, Guangdong, China
| | - Fuxia Yang
- Department of Nephrology, Shenzhen Traditional Chinese Medicine Hospital, The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen, Guangdong, China
| | - Kairi Zheng
- Traditional Chinese Medicine Department, Peking University Shenzhen Hospital, Shenzhen, Guangdong, China
- *Correspondence: Kairi Zheng, ; Yihou Zheng,
| | - Yihou Zheng
- Department of Nephrology, Shenzhen Traditional Chinese Medicine Hospital, The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen, Guangdong, China
- *Correspondence: Kairi Zheng, ; Yihou Zheng,
| |
Collapse
|
13
|
Xu Y, Wu Q, Tang Z, Tan Z, Pu D, Tan W, Zhang W, Liu S. Comprehensive Analysis of Necroptosis-Related Genes as Prognostic Factors and Immunological Biomarkers in Breast Cancer. J Pers Med 2022; 13:44. [PMID: 36675706 PMCID: PMC9863352 DOI: 10.3390/jpm13010044] [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: 11/07/2022] [Revised: 12/15/2022] [Accepted: 12/20/2022] [Indexed: 12/28/2022] Open
Abstract
Breast cancer (BC) is a lethal malignancy with a poor prognosis. Necroptosis is critical in the progression of cancer. However, the expression of genes involved in necroptosis in BC and their association with prognosis remain unclear. We investigated the predictive potential of necroptosis-related genes in BC samples from the TCGA dataset. We used LASSO regression to build a risk model consisting of twelve necroptosis-related genes in BC. Using the necroptosis-related risk model, we were able to successfully classify BC patients into high- and low-risk groups with significant prognostic differences (p = 4.872 × 10 -7). Additionally, we developed a matched nomogram predicting 5, 7, and 10-year overall survival in BC patients based on this necroptosis-related risk model. Our next step was to perform multiple GSEA analyses to explore the biological pathways through which these necroptosis-related risk genes influence cancer progression. For these twelve risk model genes, we analyzed CNV, SNV, OS, methylation, immune cell infiltration, and drug sensitivity in pan-cancer. In addition, immunohistochemical data from the THPA database were used to validate the protein expression of these risk model genes in BC. Taken together, we believe that necroptosis-related genes are considered potential therapeutic targets in BC and should be further investigated.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Shengchun Liu
- Department of Breast and Thyroid Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400042, China
| |
Collapse
|
14
|
Xu Y, Wu Q, Tang Z, Tan Z, Pu D, Tan W, Zhang W, Liu S. Comprehensive Analysis of Necroptosis-Related Genes as Prognostic Factors and Immunological Biomarkers in Breast Cancer. J Pers Med 2022; 13:44. [PMID: 36675706 PMCID: PMC9863352 DOI: 10.3390/jpm13010044;] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Revised: 12/15/2022] [Accepted: 12/20/2022] [Indexed: 10/11/2024] Open
Abstract
Breast cancer (BC) is a lethal malignancy with a poor prognosis. Necroptosis is critical in the progression of cancer. However, the expression of genes involved in necroptosis in BC and their association with prognosis remain unclear. We investigated the predictive potential of necroptosis-related genes in BC samples from the TCGA dataset. We used LASSO regression to build a risk model consisting of twelve necroptosis-related genes in BC. Using the necroptosis-related risk model, we were able to successfully classify BC patients into high- and low-risk groups with significant prognostic differences (p = 4.872 × 10 −7). Additionally, we developed a matched nomogram predicting 5, 7, and 10-year overall survival in BC patients based on this necroptosis-related risk model. Our next step was to perform multiple GSEA analyses to explore the biological pathways through which these necroptosis-related risk genes influence cancer progression. For these twelve risk model genes, we analyzed CNV, SNV, OS, methylation, immune cell infiltration, and drug sensitivity in pan-cancer. In addition, immunohistochemical data from the THPA database were used to validate the protein expression of these risk model genes in BC. Taken together, we believe that necroptosis-related genes are considered potential therapeutic targets in BC and should be further investigated.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Shengchun Liu
- Department of Breast and Thyroid Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400042, China
| |
Collapse
|
15
|
Hu HJ, Wang XH, Zhang TQ, Liu Y, Chen ZR, Zhang ZZ, Huang H, Tang HF, Jiang ZS. PLK1 promotes cholesterol efflux and alleviates atherosclerosis by up-regulating ABCA1 and ABCG1 expression via the AMPK/PPARγ/LXRα pathway. Biochim Biophys Acta Mol Cell Biol Lipids 2022; 1867:159221. [PMID: 35981705 DOI: 10.1016/j.bbalip.2022.159221] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Revised: 07/31/2022] [Accepted: 08/10/2022] [Indexed: 10/15/2022]
Abstract
Polo-like kinase 1 (PLK1) is a serine/threonine kinase involving lipid metabolism and cardiovascular disease. However, its role in atherogenesis has yet to be determined. The aim of this study was to observe the impact of PLK1 on macrophage lipid accumulation and atherosclerosis development and to explore the underlying mechanisms. We found a significant reduction of PLK1 expression in lipid-loaded macrophages and atherosclerosis model mice. Lentivirus-mediated overexpression of PLK1 promoted cholesterol efflux and inhibited lipid accumulation in THP-1 macrophage-derived foam cells. Mechanistic analysis revealed that PLK1 stimulated the phosphorylation of AMP-activated protein kinase (AMPK), leading to activation of the peroxisome proliferator-activated receptor γ (PPARγ)/liver X receptor α (LXRα) pathway and up-regulation of ATP binding cassette transporter A1 (ABCA1) and ABCG1 expression. Injection of lentiviral vector expressing PLK1 increased reverse cholesterol transport, improved plasma lipid profiles and decreased atherosclerotic lesion area in apoE-deficient mice fed a Western diet. PLK1 overexpression also facilitated AMPK and HSL phosphorylation and enhanced the expression of PPARγ, LXRα, ABCA1, ABCG1 and LPL in the aorta. In summary, these data suggest that PLK1 inhibits macrophage lipid accumulation and mitigates atherosclerosis by promoting ABCA1- and ABCG1-dependent cholesterol efflux via the AMPK/PPARγ/LXRα pathway.
Collapse
Affiliation(s)
- Heng-Jing Hu
- The First Affiliated Hospital, Department of Cardiovascular Medicine, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China; Department of Cardiovascular Disease and Key Lab for Atherosclerosis of Hunan Province, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
| | - Xiu-Heng Wang
- The First Affiliated Hospital, Department of Medical-record, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
| | - Tian-Qing Zhang
- The First Affiliated Hospital, Department of Cardiovascular Medicine, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
| | - Yao Liu
- The First Affiliated Hospital, Department of Cardiovascular Medicine, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
| | - Zheng-Rong Chen
- The First Affiliated Hospital, Department of Cardiovascular Medicine, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
| | - Zhi-Zhu Zhang
- The First Affiliated Hospital, Department of Cardiovascular Medicine, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
| | - Hong Huang
- The First Affiliated Hospital, Department of Cardiovascular Medicine, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
| | - Hui-Fang Tang
- The First Affiliated Hospital, Department of Cardiovascular Medicine, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
| | - Zhi-Sheng Jiang
- The First Affiliated Hospital, Department of Cardiovascular Medicine, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China; Department of Cardiovascular Disease and Key Lab for Atherosclerosis of Hunan Province, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China.
| |
Collapse
|
16
|
Qiu Q, Li Y, Zhang Y, Hou Y, Hu J, Wang L, Chen Z, Lei Y, Du Y, Liu X. A prognosis model for clear cell renal cell carcinoma based on four necroptosis-related genes. Front Med (Lausanne) 2022; 9:942991. [PMID: 36016998 PMCID: PMC9395686 DOI: 10.3389/fmed.2022.942991] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Accepted: 07/19/2022] [Indexed: 11/24/2022] Open
Abstract
Necroptosis is a type of caspase-independent cell death, and it plays a critical role in regulating the development of cancer. To date, little is known about the role of necroptosis-related genes (NRGs) in clear cell renal cell carcinoma (ccRCC). In this study, we downloaded data regarding the expression of NRGs and overall survival (OS) from The Cancer Genome Atlas (TCGA) database and constructed a risk model to determine the prognostic features of necroptosis using COX regression analysis. Patients with ccRCC were divided into low-risk and high-risk groups based on their risk scores. Thereafter, Kaplan–Meier curves were used to evaluate OS, and receiver operating characteristic (ROC) curves were used to determine the accuracy of prediction. Stratified analyses were performed according to different clinical variables. Furthermore, we assessed the correlation between clinical variables and risk scores; the NRGs with differential expression were mainly enriched in positive regulation of intracellular transport and platinum resistance pathways. We constructed prognostic signatures for OS based on four NRGs and showed that the survival time was significantly longer in the low-risk groups than in the high-risk groups (p < 0.001). The area of the ROC curve for OS was 0.717, indicating excellent predictive accuracy of the established model. Therefore, a predictive model based on NRGs was constructed, which can predict the prognosis of patients and provides insights into the biological mechanisms underlying necroptosis in patients with ccRCC.
Collapse
Affiliation(s)
- Qiangmin Qiu
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Yanze Li
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Ye Zhang
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Yanguang Hou
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Juncheng Hu
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, China
- Wuhan University Institute of Urologic Disease, Renmin Hospital of Wuhan University, Wuhan, China
| | - Lei Wang
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Zhiyuan Chen
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Yourong Lei
- Department of Infection Prevention and Control, Renmin Hospital of Wuhan University, Wuhan, China
- *Correspondence: Yourong Lei,
| | - Yang Du
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, China
- Yang Du,
| | - Xiuheng Liu
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, China
- Xiuheng Liu,
| |
Collapse
|
17
|
Wang L, Gao M, Sun D, Wu H, Lv S, Li Y, Li L. PLK1 Is a Potential Prognostic Factor Associated with the Tumor Microenvironment in Lung Adenocarcinoma. BIOMED RESEARCH INTERNATIONAL 2022; 2022:7848771. [PMID: 35941968 PMCID: PMC9356880 DOI: 10.1155/2022/7848771] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Revised: 06/02/2022] [Accepted: 06/16/2022] [Indexed: 12/02/2022]
Abstract
More than 40% of lung cancers are lung adenocarcinoma (LUAD) worldwide. However, the prognosis of LUAD is poor for the lack of effective treatment methods. Our study identified PLK1 as a novel prognosis biomarker and treatment target for LUAD. Based on the Cancer Genome Atlas (TCGA) database, differentially expressed genes (DEGs) from 551 LUAD cases were analyzed for the Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analysis. To explore the biological pathways and the tumor-infiltrating immune cells (TICs) using gene set variation analysis (GSVA) and the CIBERSORT, as well as to analyze DEGs, a protein-protein interaction (PPI) network and Cox regression analysis were performed. Validation of DEGs was achieved through quantitative real-time PCR (qPCR) and immunoblotting. DEGs associated with the cell cycle were sorted out. Cell cycle scores were positively correlated with age, clinical stages, and metastasis and negatively correlated with overall survival of LUAD patients. PPI and Cox analyses showed that PLK1 could be a prognostic factor for LUAD patients. CIBERSORT analysis revealed a positive correlation between the transcription level of PLK1 and the function of CD8+ and activated memory CD4+ T cells, as well as a negative correlation with activated natural killer cells. Furthermore, PLK1 overexpression increased immune cytotoxicity, as measured by the cytolytic activity score, IFN- score, and IFN- level. There is a strong correlation between PLK1 and key features of TICs, indicating its potential as a promising prognostic biomarker for LUAD.
Collapse
Affiliation(s)
- Lina Wang
- The Key Laboratory of Pathobiology, Ministry of Education, Department of Pathology, College of Basic Medical Sciences, Jilin University, Changchun, Jilin, China
| | - Man Gao
- Pediatric Department of Respiration II, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Dongjie Sun
- The Key Laboratory of Pathobiology, Ministry of Education, Department of Pathology, College of Basic Medical Sciences, Jilin University, Changchun, Jilin, China
| | - Haitao Wu
- Cancer Center of the First Hospital of Jilin University, Changchun, Jilin, China
| | - Shuang Lv
- The Key Laboratory of Pathobiology, Ministry of Education, Department of Pathology, College of Basic Medical Sciences, Jilin University, Changchun, Jilin, China
| | - Yulin Li
- The Key Laboratory of Pathobiology, Ministry of Education, Department of Pathology, College of Basic Medical Sciences, Jilin University, Changchun, Jilin, China
| | - Lisha Li
- The Key Laboratory of Pathobiology, Ministry of Education, Department of Pathology, College of Basic Medical Sciences, Jilin University, Changchun, Jilin, China
| |
Collapse
|
18
|
Sun EC, Dong SS, Li ZJ, Li CX. Clinicopathological Significance of AKT1 and PLK1 Expression in Oral Squamous Cell Carcinoma. DISEASE MARKERS 2022; 2022:7300593. [PMID: 35756492 PMCID: PMC9232379 DOI: 10.1155/2022/7300593] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/27/2022] [Revised: 05/16/2022] [Accepted: 05/26/2022] [Indexed: 12/24/2022]
Abstract
Purpose Oral squamous cell carcinoma (OSCC) is the sixth leading cause of cancer-related death worldwide and is characterized by metastasis and recurrence. We aimed to evaluate the expression of AKT1 and PLK1 in OSCC and identify their correlation with the clinical and histological features and prognosis of patients with OSCC. Methods Tissue samples were collected from 70 patients with OSCC and 50 patients with normal oral mucosa. The expression levels of AKT1 and PLK1 in OSCC tissues and normal oral mucosa were detected by immunohistochemistry. The chi-square test was used to identify correlations between the expression levels of AKT1 and PLK1 with patients' clinicopathologic characteristics. Survival analysis was assessed by the Kaplan-Meier method. Spearman's rank correlation test was used to determine the relationships between AKT1 and PLK1 expressions. The bioinformatics database GEPIA was used to verify the experimental results. Results The chi-square test and Fisher's exact test showed that the positive expression rate of AKT1 and PLK1 in OSCC tissue was significantly higher than that in the normal oral mucosa (P < 0.05). PLK1 expression levels were significantly correlated with tumor stage and size (P < 0.05). Kaplan-Meier analysis showed that the survival time of AKT1 and PLK1 with high expression was significantly shorter than that of patients with low expression (P < 0.05). Spearman's rank correlation test showed a strong correlation between AKT1 and PLK1 expression in OSCC tissue (R = 0.53; P < 0.05). GEPIA bioinformatics database analysis results show that the expression and overall survival of AKT1 and PLK1 analysis and the correlation analysis of AKT1 and PLK1 were consistent with experimental results. Conclusion AKT1 and PLK1 expressions are associated with the occurrence and progression of OSCC and may be used as diagnostic and prognostic indicators of OSCC. There may be a correlation between AKT1 and PLK1 in OSCC tissue.
Collapse
Affiliation(s)
- Er-Can Sun
- Department of Stomatology, Shihezi University School of Medicine & the First Affiliated Hospital to Shihezi University School of Medicine, Shihezi, 832002 Xinjiang, China
| | - Shuang-Shuang Dong
- Department of Pathology, Northern Jiangsu People's Hospital Affiliated to Yangzhou University/Clinical Medical College, Yangzhou University, Yangzhou, Jiangsu 225000, China
| | - Zhi-Jun Li
- Department of Stomatology, Shihezi University School of Medicine & the First Affiliated Hospital to Shihezi University School of Medicine, Shihezi, 832002 Xinjiang, China
| | - Chang-Xue Li
- Department of Stomatology, Shihezi University School of Medicine & the First Affiliated Hospital to Shihezi University School of Medicine, Shihezi, 832002 Xinjiang, China
| |
Collapse
|
19
|
Chiappa M, Petrella S, Damia G, Broggini M, Guffanti F, Ricci F. Present and Future Perspective on PLK1 Inhibition in Cancer Treatment. Front Oncol 2022; 12:903016. [PMID: 35719948 PMCID: PMC9201472 DOI: 10.3389/fonc.2022.903016] [Citation(s) in RCA: 44] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Accepted: 05/09/2022] [Indexed: 12/13/2022] Open
Abstract
Polo-like kinase 1 (PLK1) is the principle member of the well conserved serine/threonine kinase family. PLK1 has a key role in the progression of mitosis and recent evidence suggest its important involvement in regulating the G2/M checkpoint, in DNA damage and replication stress response, and in cell death pathways. PLK1 expression is tightly spatially and temporally regulated to ensure its nuclear activation at the late S-phase, until the peak of expression at the G2/M-phase. Recently, new roles of PLK1 have been reported in literature on its implication in the regulation of inflammation and immunological responses. All these biological processes are altered in tumors and, considering that PLK1 is often found overexpressed in several tumor types, its targeting has emerged as a promising anti-cancer therapeutic strategy. In this review, we will summarize the evidence suggesting the role of PLK1 in response to DNA damage, including DNA repair, cell cycle progression, epithelial to mesenchymal transition, cell death pathways and cancer-related immunity. An update of PLK1 inhibitors currently investigated in preclinical and clinical studies, in monotherapy and in combination with existing chemotherapeutic drugs and targeted therapies will be discussed.
Collapse
Affiliation(s)
- Michela Chiappa
- Laboratory of Experimental Oncology, Department of Oncology, Istituto di Ricerche Farmacologiche Mario Negri-IRCCS, Milan, Italy
| | - Serena Petrella
- Laboratory of Experimental Oncology, Department of Oncology, Istituto di Ricerche Farmacologiche Mario Negri-IRCCS, Milan, Italy
| | - Giovanna Damia
- Laboratory of Experimental Oncology, Department of Oncology, Istituto di Ricerche Farmacologiche Mario Negri-IRCCS, Milan, Italy
| | - Massimo Broggini
- Laboratory of Molecular Pharmacology, Department of Oncology, Istituto di Ricerche Farmacologiche Mario Negri-IRCCS, Milan, Italy
| | - Federica Guffanti
- Laboratory of Experimental Oncology, Department of Oncology, Istituto di Ricerche Farmacologiche Mario Negri-IRCCS, Milan, Italy
| | - Francesca Ricci
- Laboratory of Experimental Oncology, Department of Oncology, Istituto di Ricerche Farmacologiche Mario Negri-IRCCS, Milan, Italy
| |
Collapse
|
20
|
Luo Y, Zhang G. Identification of a Necroptosis-Related Prognostic Index and Associated Regulatory Axis in Kidney Renal Clear Cell Carcinoma. Int J Gen Med 2022; 15:5407-5423. [PMID: 35685693 PMCID: PMC9173730 DOI: 10.2147/ijgm.s367173] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2022] [Accepted: 05/20/2022] [Indexed: 01/10/2023] Open
Affiliation(s)
- Yong Luo
- Department of Urology, the Second People’s Hospital of Foshan, Affiliated Foshan Hospital of Southern Medical University, Foshan, 528000, People’s Republic of China
- Correspondence: Yong Luo, Department of Urology, the Second People’s Hospital of Foshan, Affiliated Foshan Hospital of Southern Medical University, 78 Weiguo Road, Foshan, 528000, People’s Republic of China, Tel +86-15625093895, Fax +86-0757-88032009, Email
| | - Guian Zhang
- School of Medicine, South China University of Technology, Guangzhou, 510006, People’s Republic of China
- Guian Zhang, School of Medicine, South China University of Technology, Guangzhou, 510006, People’s Republic of China, Tel +86-13246808932, Email
| |
Collapse
|
21
|
Chen H, Lin R, Lin W, Chen Q, Ye D, Li J, Feng J, Cheng W, Zhang M, Qi Y. An immune gene signature to predict prognosis and immunotherapeutic response in lung adenocarcinoma. Sci Rep 2022; 12:8230. [PMID: 35581376 PMCID: PMC9114138 DOI: 10.1038/s41598-022-12301-6] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2022] [Accepted: 05/09/2022] [Indexed: 11/10/2022] Open
Abstract
Lung adenocarcinoma is one of the most common malignant tumors worldwide. The purpose of this study was to construct a stable immune gene signature for prediction of prognosis (IGSPP) and response to immune checkpoint inhibitors (ICIs) therapy in LUAD patients. Five genes were screened by weighted gene coexpression network analysis, Cox regression and LASSO regression analyses and were used to construct the IGSPP. The survival rate of the IGSPP low-risk group was higher than that of the IGSPP high-risk group. Multivariate Cox regression analysis showed that IGSPP could be used as an independent prognostic factor for the overall survival of LUAD patients. IGSPP genes were enriched in cell cycle pathways. IGSPP gene mutation rates were higher in the high-risk group. CD4 memory-activated T cells, M0 and M1 macrophages had higher infiltration abundance in the high-risk group, which was associated with poor overall survival. In contrast, the abundance of resting CD4 memory T cells, monocytes, resting dendritic cells and resting mast cells associated with a better prognosis was higher in the low-risk group. TIDE scores and the expressions of different immune checkpoints showed that patients in the high-risk IGSPP group benefited more from ICIs treatment. In short, an IGSPP of LUAD was constructed and characterized. It could be used to predict the prognosis and benefits of ICIs treatment in LUAD patients.
Collapse
Affiliation(s)
- Hongquan Chen
- School of Basic Medical Sciences, Fujian Medical University, Fuzhou, 350122, Fujian, China
| | - Renxi Lin
- School of Basic Medical Sciences, Fujian Medical University, Fuzhou, 350122, Fujian, China
| | - Weibin Lin
- School of Basic Medical Sciences, Fujian Medical University, Fuzhou, 350122, Fujian, China
| | - Qing Chen
- School of Basic Medical Sciences, Fujian Medical University, Fuzhou, 350122, Fujian, China
| | - Dongjie Ye
- School of Basic Medical Sciences, Fujian Medical University, Fuzhou, 350122, Fujian, China
| | - Jing Li
- School of Basic Medical Sciences, Fujian Medical University, Fuzhou, 350122, Fujian, China.,Department of Pathology, Fujian Provincial Maternity Hospital, Fuzhou, 350012, Fujian, China
| | - Jinan Feng
- School of Basic Medical Sciences, Fujian Medical University, Fuzhou, 350122, Fujian, China.,Luoyang Central Hospital Affiliated to Zhengzhou University, Luoyang, 471099, Henan, China
| | - Wenxiu Cheng
- School of Basic Medical Sciences, Fujian Medical University, Fuzhou, 350122, Fujian, China
| | - Mingfang Zhang
- School of Basic Medical Sciences, Fujian Medical University, Fuzhou, 350122, Fujian, China.
| | - Yuanlin Qi
- School of Basic Medical Sciences, Fujian Medical University, Fuzhou, 350122, Fujian, China.
| |
Collapse
|
22
|
Xin S, Mao J, Duan C, Wang J, Lu Y, Yang J, Hu J, Liu X, Guan W, Wang T, Wang S, Liu J, Song W, Song X. Identification and Quantification of Necroptosis Landscape on Therapy and Prognosis in Kidney Renal Clear Cell Carcinoma. Front Genet 2022; 13:832046. [PMID: 35237304 PMCID: PMC8882778 DOI: 10.3389/fgene.2022.832046] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2021] [Accepted: 01/19/2022] [Indexed: 01/11/2023] Open
Abstract
Kidney renal clear cell carcinoma (KIRC) has high morbidity and gradually increased in recent years, and the rate of progression once relapsed is high. At present, owing to lack of effective prognosis predicted markers and post-recurrence drug selection guidelines, the prognosis of KIRC patients is greatly affected. Necroptosis is a regulated form of cell necrosis in a way that is independent of caspase. Induced necroptosis is considered an effective strategy in chemotherapy and targeted drugs, and it can also be used to improve the efficacy of immunotherapy. Herein, we quantified the necroptosis landscape of KIRC patients from The Cancer Genome Atlas (TCGA) database and divided them into two distinct necroptosis-related patterns (C1 and C2) through the non-negative matrix factorization (NMF) algorithm. Multi-analysis revealed the differences in clinicopathological characteristics and tumor immune microenvironment (TIME). Then, we constructed the NRG prognosis signature (NRGscore), which contained 10 NRGs (PLK1, APP, TNFRSF21, CXCL8, MYCN, TNFRSF1A, TRAF2, HSP90AA1, STUB1, and FLT3). We confirmed that NRGscore could be used as an independent prognostic marker for KIRC patients and performed excellent stability and accuracy. A nomogram model was also established to provide a more beneficial prognostic indicator for the clinic. We found that NRGscore was significantly correlated with clinicopathological characteristics, TIME, and tumor mutation burden (TMB) of KIRC patients. Moreover, NRGscore had effective guiding significance for immunotherapy, chemotherapy, and targeted drugs.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | - Wen Song
- *Correspondence: Wen Song, ; Xiaodong Song,
| | | |
Collapse
|
23
|
Qian Y, Li Y, Chen K, Liu N, Hong X, Wu D, Xu Z, Zhou L, Xu L, Jia R, Ge YZ. Pan-Cancer Transcriptomic Analysis Identifies PLK1 Crucial for the Tumorigenesis of Clear Cell Renal Cell Carcinoma. J Inflamm Res 2022; 15:1099-1116. [PMID: 35210814 PMCID: PMC8859474 DOI: 10.2147/jir.s347732] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Accepted: 02/02/2022] [Indexed: 11/23/2022] Open
Affiliation(s)
- Yiguan Qian
- Department of Urology, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu, People’s Republic of China
| | - Yang Li
- Department of Urology, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu, People’s Republic of China
| | - Ke Chen
- Department of Urology, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu, People’s Republic of China
| | - Ning Liu
- Department of Urology, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu, People’s Republic of China
| | - Xi Hong
- Department of Urology, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu, People’s Republic of China
| | - Di Wu
- Department of Urology, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu, People’s Republic of China
| | - Zheng Xu
- Department of Urology, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu, People’s Republic of China
| | - Liuhua Zhou
- Department of Urology, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu, People’s Republic of China
| | - Luwei Xu
- Department of Urology, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu, People’s Republic of China
| | - Ruipeng Jia
- Department of Urology, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu, People’s Republic of China
- Correspondence: Ruipeng Jia; Yu-Zheng Ge, Department of Urology, Nanjing First Hospital, Nanjing Medical UniversityNanjing, Jiangsu, People’s Republic of China, Tel +86-15850675660, Email ;
| | - Yu-Zheng Ge
- Department of Urology, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu, People’s Republic of China
| |
Collapse
|
24
|
Zou X, Liu Y, Di J, Wei W, Watanabe N, Li J, Li X. ZMIZ2 promotes the development of triple-receptor negative breast cancer. Cancer Cell Int 2022; 22:52. [PMID: 35101047 PMCID: PMC8802436 DOI: 10.1186/s12935-021-02393-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Accepted: 12/04/2021] [Indexed: 12/25/2023] Open
Abstract
Background Triple-receptor negative breast cancer (TNBC) is an aggressive breast tumor subtype that generally has a poor prognosis. This study aimed to investigate the role and regulatory mechanisms of Zinc finger MIZ-type containing 2 (ZMIZ2) in relation to TNBC. Methods Based on data from The Cancer Genome Atlas (TCGA), the expression of ZMIZ2 in different subtypes and its correlation with androgen receptor (AR) were analyzed, and a regulatory mechanism network was constructed. The expression and prognostic value of ZMIZ2 in clinical TNBC tissue samples were also investigated. Furthermore, in vitro studies were conducted to investigate the effects of ZMIZ2 knockdown on the malignant behaviors of TNBC cells and target gene expression. Results Based on TCGA data, ZMIZ2 was found to be significantly upregulated in TNBC tissues and its expression was negatively correlated with AR expression. Key relationships, such as the ZMIZ2-CCL5, ZMIZ2/AR-MCM3, ZMIZ2/AR-E2F4, and the ZMIZ2/AR-DHX38 were identified, which were enriched in NOD-like receptor signaling pathway/toll-like receptor signaling pathway, DNA replication, cell cycle, and spliceosome, respectively. Moreover, ZMIZ2 was upregulated in clinical breast cancer tissues and its high expression was correlated with the poor prognosis of TNBC patients. Furthermore, ZMIZ2 expression was increased in breast cancer cells, and a knockdown of ZMIZ2 inhibited MDA-MB-231 cell proliferation, migration, and invasion, induced cell cycle arrest in the G1 phase, and promoted cell apoptosis. Furthermore, ZMIZ2 knockdown inhibited the mRNA and protein expression of CCL5, MCM3, E2F4, and DHX38. Conclusion Our findings reveal that ZMIZ2 is upregulated in TNBC tissues and is associated with its poor prognosis. ZMIZ2 may promote TNBC progression by promoting the expression of its target genes and affecting the corresponding pathways. Consequently, ZMIZ2 may serve as a promising target for future TNBC treatments. Supplementary Information The online version contains supplementary material available at 10.1186/s12935-021-02393-x.
Collapse
Affiliation(s)
- Xiaopan Zou
- The Key Laboratory of Molecular Epigenetic, Institute of Genetics and Cytology, Northeast Normal University, No.5268 Renmin Street, Nanguan District, Changchun, 130024, Jilin, China.,Breast and Thyroid Surgery, Jilin Province People's Hospital, Changchun, 130021, Jilin, China
| | - Yan Liu
- The Key Laboratory of Molecular Epigenetic, Institute of Genetics and Cytology, Northeast Normal University, No.5268 Renmin Street, Nanguan District, Changchun, 130024, Jilin, China
| | - Jun Di
- Pathological Diagnostic Center, Jilin Province People's Hospital, Changchun, 130021, Jilin, China
| | - Wei Wei
- The Key Laboratory of Molecular Epigenetic, Institute of Genetics and Cytology, Northeast Normal University, No.5268 Renmin Street, Nanguan District, Changchun, 130024, Jilin, China
| | - Nobumoto Watanabe
- Bio-Active Compounds Discovery Research Unit, RIKEN Center for Sustainable Resource Science, Wako, Saitama, Japan
| | - Jiang Li
- Affiliated Stomatology Hospital of Guangzhou Medical University, Guangzhou, 510180, Guangdong, China.
| | - Xiaomeng Li
- The Key Laboratory of Molecular Epigenetic, Institute of Genetics and Cytology, Northeast Normal University, No.5268 Renmin Street, Nanguan District, Changchun, 130024, Jilin, China.
| |
Collapse
|
25
|
Zhang J, Zhang H, Wang Y, Wang Q. MCM2-7 in Clear Cell Renal Cell Carcinoma: MCM7 Promotes Tumor Cell Proliferation. Front Oncol 2021; 11:782755. [PMID: 34993142 PMCID: PMC8724441 DOI: 10.3389/fonc.2021.782755] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Accepted: 12/02/2021] [Indexed: 11/23/2022] Open
Abstract
Background Clear cell renal cell carcinoma (ccRCC) accounts for 60-70% of renal cell carcinoma (RCC) cases. Finding more therapeutic targets for advanced ccRCC is an urgent mission. The minichromosome maintenance proteins 2-7 (MCM2-7) protein forms a stable heterohexamer and plays an important role in DNA replication in eukaryotic cells. In the study, we provide a comprehensive study of MCM2-7 genes expression and their potential roles in ccRCC. Methods The expression and prognosis of the MCM2-7 genes in ccRCC were analyzed using data from TCGA, GEO and ArrayExpress. MCM2-7 related genes were identified by weighted co-expression network analysis (WGCNA) and Metascape. CancerSEA and GSEA were used to analyze the function of MCM2–7 genes in ccRCC. The gene effect scores (CERES) of MCM2-7, which reflects carcinogenic or tumor suppressor, were obtained from DepMap. We used clinical and expression data of MCM2-7 from the TCGA dataset and the LASSO Cox regression analysis to develop a risk score to predict survival of patients with ccRCC. The correlations between risk score and other clinical indicators such as gender, age and stage were also analyzed. Further validation of this risk score was engaged in another cohort, E-MTAB-1980 from the ArrayExpress dataset. Results The mRNA and protein expression of MCM2-7 were increased in ccRCC compared with normal tissues. High MCM2, MCM4, MCM6 and MCM7 expression were associated with a poor prognosis of ccRCC patients. Functional enrichment analysis revealed that MCM2-7 might influence the progress of ccRCC by regulating the cell cycle. Knockdown of MCM7 can inhibit the proliferation of ccRCC cells. A two-gene risk score including MCM4 and MCM6 can predict overall survival (OS) of ccRCC patients. The risk score was successfully verified by further using Arrayexpress cohort. Conclusion We analyze MCM2-7 mRNA and protein levels in ccRCC. MCM7 is determined to promote tumor proliferation. Meanwhile, our study has determined a risk score model composed of MCM2-7 can predict the prognosis of ccRCC patients, which may help future treatment strategies.
Collapse
Affiliation(s)
- Junneng Zhang
- Laboratory Medicine Department, The Fifth Hospital of Xiamen, Xiamen, China
- *Correspondence: Junneng Zhang, ; Qingshui Wang,
| | - Huanzong Zhang
- Laboratory Medicine Department, The Fifth Hospital of Xiamen, Xiamen, China
| | - Yinghui Wang
- Laboratory Medicine Department, The Fifth Hospital of Xiamen, Xiamen, China
| | - Qingshui Wang
- Key Laboratory of Optoelectronic Science and Technology for Medicine of Ministry of Education, College of Life Sciences, Fujian Normal University, Fuzhou, China
- *Correspondence: Junneng Zhang, ; Qingshui Wang,
| |
Collapse
|
26
|
Liang XH, Feng ZP, Liu FQ, Yan R, Yin LY, Shen H, Lu HL. Identification of potential biomarkers for diagnosis of hepatocellular carcinoma. Exp Ther Med 2021; 23:51. [PMID: 34917180 PMCID: PMC8630445 DOI: 10.3892/etm.2021.10973] [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: 08/23/2019] [Accepted: 07/07/2020] [Indexed: 01/06/2023] Open
Abstract
Hepatocellular carcinoma (HCC) has a high mortality rate owing to its complexity. Identification of abnormally expressed genes in HCC tissues compared to those in normal liver tissues is a viable strategy for investigating the mechanisms of HCC tumorigenesis and progression as a means of developing novel treatments. A significant advantage of the Gene Expression Omnibus (GEO) and The Cancer Genome Atlas (TCGA) is that the data therein were collected from different independent researchers and may be integrated, allowing for a more robust data analysis. Accordingly, in the present study, the gene expression profiles for HCC and control samples were downloaded from the GEO and TCGA. Functional enrichment analysis was performed using a Metascape dataset, and a protein-protein interaction (PPI) network was constructed using the Search Tool for the Retrieval of Interacting Genes/proteins (STRING) online database. The prognostic value of mRNA for HCC was assessed using the Kaplan-Meier Plotter, a public online tool. A gene mRNA heatmap and DNA amplification numbers were obtained from cBioPortal. A total of 2,553 upregulated genes were identified. Functional enrichment analysis revealed that these differentially expressed genes (DEGs) were mainly accumulated in metabolism of RNA and the cell cycle. Considering the complexity and heterogeneity of the molecular alterations in HCC, multiple genes for the prognostication of patients with HCC are more reliable than a single gene. Thus, the PPI network and univariate Cox regression analysis were applied to screen candidate genes (small nuclear ribonucleoprotein polypeptide B and B1, nucleoporin 37, Rac GTPase activating protein 1, kinesin family member 20A, minichromosome maintenance 10 replication initiation factor, ubiquitin conjugating enzyme E2 C and hyaluronan mediated motility receptor) that are associated with the overall survival and progression-free survival of patients with HCC. In conclusion, the present study identified a set of genes that are associated with overall survival and progression-free survival of patients with HCC, providing valuable information for the prognosis of HCC.
Collapse
Affiliation(s)
- Xing-Hua Liang
- Department of Gastroenterology, The Fourth Affiliated Hospital of Guangzhou Medical University (Zengcheng District People's Hospital of Guangzhou), Guangzhou, Guangdong 511300, P.R. China
| | - Zheng-Ping Feng
- Department of Gastroenterology, The Fourth Affiliated Hospital of Guangzhou Medical University (Zengcheng District People's Hospital of Guangzhou), Guangzhou, Guangdong 511300, P.R. China
| | - Fo-Qiu Liu
- Department of Gastroenterology, The Fourth Affiliated Hospital of Guangzhou Medical University (Zengcheng District People's Hospital of Guangzhou), Guangzhou, Guangdong 511300, P.R. China
| | - Rong Yan
- Department of Gastroenterology, The Fourth Affiliated Hospital of Guangzhou Medical University (Zengcheng District People's Hospital of Guangzhou), Guangzhou, Guangdong 511300, P.R. China
| | - Liang-Yu Yin
- Department of Gastroenterology, The Fourth Affiliated Hospital of Guangzhou Medical University (Zengcheng District People's Hospital of Guangzhou), Guangzhou, Guangdong 511300, P.R. China
| | - Hao Shen
- Department of Gastroenterology, The Fourth Affiliated Hospital of Guangzhou Medical University (Zengcheng District People's Hospital of Guangzhou), Guangzhou, Guangdong 511300, P.R. China
| | - Hai-Lin Lu
- Department of Gastroenterology, The Fourth Affiliated Hospital of Guangzhou Medical University (Zengcheng District People's Hospital of Guangzhou), Guangzhou, Guangdong 511300, P.R. China
| |
Collapse
|
27
|
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: 27] [Impact Index Per Article: 6.8] [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
|
28
|
Mao S, Tian S, Luo X, Zhou M, Cao Z, Li J. Overexpression of PLK1 relieved the myocardial ischemia-reperfusion injury of rats through inducing the mitophagy and regulating the p-AMPK/FUNDC1 axis. Bioengineered 2021; 12:2676-2687. [PMID: 34115550 PMCID: PMC8806532 DOI: 10.1080/21655979.2021.1938500] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Revised: 05/31/2021] [Accepted: 05/31/2021] [Indexed: 12/23/2022] Open
Abstract
Myocardial cell injury caused by myocardial ischemia and reperfusion is one of the main causes of the occurrence and development of heart disease. Recent study has shown that inducing mitophagy of cardiomyocytes is a crucial method to alleviate ischemia-reperfusion injury. While, Polo-like kinase 1 (PLK1) can induce the mitophagy of breast cancer cells. Moreover, PLK1 was able to promote the expression of p-AMPK and FUNDC1, which are the protective factors for myocardium. Therefore, the mouse model of ischemia/reperfusion was established and the effect of PLK1 on ischemia reperfusion induced myocardial damage was investigated. The PLK1 was overexpressed in H9c2 cells and rat model of ischemia/reperfusion. Ischemia reperfusion inhibited the expression of PLK1. While overexpression of PLK1 relieved the myocardial infarction and myocardium apoptosis through inducing mitophagy in rats model of ischemia reperfusion. In vitro, the H9c2 cells overexpressing the PLK1 were treated with the hypoxia and reoxygenation and the apoptosis, survival rate and expression of mitophagy-related proteins of H9c2 cells were detected using the flow cytometry, CCK-8 assay and western blotting. The results reveled that overexpression of PLK1 alleviated the hypoxia and reoxygenation induced apoptosis of H9c2 cells and promoted the expression of mitophagy-related proteins. In addition, enhanced PLK1 expression promoted the expression of p-AMPK and FUNDC1 in H9c2 cells. However, the inhibition of FUNDC1 abolished the positive effect of PLK1 on H9c2 cells mentioned above. In conclusion, PLK1 alleviated the ischemia reperfusion induced myocardial damage by inducing the mitophagy in a p-AMPK/FUNDC1 signaling dependent pathway.
Collapse
Affiliation(s)
- Shan Mao
- Department of Cardiology, Taihe Hospital, Hubei University of Medicine, Shiyan City, Hubei Province, China
| | - Shuning Tian
- Department of Anesthesiology, Jiangmen Central Hospital Affiliated Jiangmen Hospital of Sun Yat-sen University, Jiangmen City, Guangdong Province, China
| | - Xianghong Luo
- Department of Anesthesiology, Taihe Hospital, Hubei University of Medicine, Shiyan City, Hubei Province, China
| | - Ming Zhou
- Department of Cardiology, Taihe Hospital, Hubei University of Medicine, Shiyan City, Hubei Province, China
| | - Zheng Cao
- Department of Cardiology, Taihe Hospital, Hubei University of Medicine, Shiyan City, Hubei Province, China
| | - Ji Li
- Department of Cardiovascular Internal Medicine, The Second Affiliated Hospital of Guizhou Medical University, Kaili City, Guizhou Province, China
| |
Collapse
|
29
|
Wang PC, Chen ST, Yang ZM. Effects of Aurora kinase A on mouse decidualization via Stat3-plk1-cdk1 pathway. Reprod Biol Endocrinol 2021; 19:162. [PMID: 34715887 PMCID: PMC8557062 DOI: 10.1186/s12958-021-00847-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Accepted: 10/14/2021] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Decidualization is essential to the successful pregnancy in mice. The molecular mechanisms and effects of Aurora kinase A (Aurora A) remain poorly understood during pregnancy. This study is the first to investigate the expression and role of Aurora A during mouse decidualization. METHODS Quantitative real time polymerase chain reaction, western blotting and in situ hybridization were used to determine the expression of Aurora A in mouse uteri. Aurora A activity was inhibited by Aurora A inhibitor to explore the role of Aurora A on decidualization via regulating the Aurora A/Stat3/Plk1/Cdk1 signaling pathway. RESULTS Aurora A was strongly expressed at implantation sites compared with inter-implantation sites. Furthermore, Aurora A was also significantly increased in oil-induced deciduoma compared with control. Both Aurora A mRNA and protein were significantly increased under in vitro decidualization. Under in vitro decidualization, Prl8a2, a marker of mouse decidualization, was significantly decreased by TC-S 7010, an Aurora A inhibitor. Additionally, Prl8a2 was reduced by Stat3 inhibitor, Plk1 inhibitor and Cdk1 inhibitor, respectively. Moreover, the protein levels of p-Stat3, p-Plk1 and p-Cdk1 were suppressed by TC-S 7010. The protein levels of p-Stat3, p-Plk1 and p-Cdk1 were also suppressed by S3I-201, a Stat3 inhibitor). SBE 13 HCl (Plk1 inhibitor) could reduce the protein levels of p-Plk1 and p-Cdk1. Collectively, Aurora A could regulate Stat3/Plk1/Cdk1 signaling pathway. CONCLUSION Our study shows that Aurora A is expressed in decidual cells and should be important for mouse decidualization. Aurora A/Stat3/Plk1/Cdk1 signaling pathway may be involved in mouse decidualization.
Collapse
Affiliation(s)
- Peng-Chao Wang
- College of Veterinary Medicine, Shanxi Agricultural University, Taigu, 030801 China
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642 China
| | - Si-Ting Chen
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642 China
| | - Zeng-Ming Yang
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642 China
| |
Collapse
|
30
|
Patterson DG, Kania AK, Price MJ, Rose JR, Scharer CD, Boss JM. An IRF4-MYC-mTORC1 Integrated Pathway Controls Cell Growth and the Proliferative Capacity of Activated B Cells during B Cell Differentiation In Vivo. THE JOURNAL OF IMMUNOLOGY 2021; 207:1798-1811. [PMID: 34470852 DOI: 10.4049/jimmunol.2100440] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Accepted: 07/30/2021] [Indexed: 12/13/2022]
Abstract
Cell division is an essential component of B cell differentiation to Ab-secreting plasma cells, with critical reprogramming occurring during the initial stages of B cell activation. However, a complete understanding of the factors that coordinate early reprogramming events in vivo remain to be determined. In this study, we examined the initial reprogramming by IRF4 in activated B cells using an adoptive transfer system and mice with a B cell-specific deletion of IRF4. IRF4-deficient B cells responding to influenza, 4-hydroxy-3-nitrophenylacetyl-Ficoll, and LPS divided but stalled during the proliferative response. Gene expression profiling of IRF4-deficient B cells at discrete divisions revealed IRF4 was critical for inducing MYC target genes, oxidative phosphorylation, and glycolysis. Moreover, IRF4-deficient B cells maintained an inflammatory gene expression signature. Complementary chromatin accessibility analyses established a hierarchy of IRF4 activity and identified networks of dysregulated transcription factor families in IRF4-deficient B cells, including E-box binding bHLH family members. Indeed, B cells lacking IRF4 failed to fully induce Myc after stimulation and displayed aberrant cell cycle distribution. Furthermore, IRF4-deficient B cells showed reduced mTORC1 activity and failed to initiate the B cell activation unfolded protein response and grow in cell size. Myc overexpression in IRF4-deficient cells was sufficient to overcome the cell growth defect. Together, these data reveal an IRF4-MYC-mTORC1 relationship critical for controlling cell growth and the proliferative response during B cell differentiation.
Collapse
Affiliation(s)
- Dillon G Patterson
- Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, GA; and.,The Emory Vaccine Center, Emory University School of Medicine, Emory University, Atlanta, GA
| | - Anna K Kania
- Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, GA; and.,The Emory Vaccine Center, Emory University School of Medicine, Emory University, Atlanta, GA
| | - Madeline J Price
- Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, GA; and.,The Emory Vaccine Center, Emory University School of Medicine, Emory University, Atlanta, GA
| | - James R Rose
- Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, GA; and.,The Emory Vaccine Center, Emory University School of Medicine, Emory University, Atlanta, GA
| | - Christopher D Scharer
- Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, GA; and.,The Emory Vaccine Center, Emory University School of Medicine, Emory University, Atlanta, GA
| | - Jeremy M Boss
- Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, GA; and .,The Emory Vaccine Center, Emory University School of Medicine, Emory University, Atlanta, GA
| |
Collapse
|
31
|
Al‐Qahtani QH, Moghrabi WN, Al‐Yahya S, Al‐Haj L, Al‐Saif M, Mahmoud L, Al‐Mohanna F, Al‐Souhibani N, Alaiya A, Hitti E, Khabar KSA. Kinome inhibition reveals a role for polo-like kinase 1 in targeting post-transcriptional control in cancer. Mol Oncol 2021; 15:2120-2139. [PMID: 33411958 PMCID: PMC8334256 DOI: 10.1002/1878-0261.12897] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2020] [Revised: 12/09/2020] [Accepted: 01/05/2021] [Indexed: 12/15/2022] Open
Abstract
Dysfunctions in post-transcriptional control are observed in cancer and chronic inflammatory diseases. Here, we employed a kinome inhibitor library (n = 378) in a reporter system selective for 3'-untranslated region-AU-rich elements (ARE). Fifteen inhibitors reduced the ARE-reporter activity; among the targets is the polo-like kinase 1 (PLK1). RNA-seq experiments demonstrated that the PLK1 inhibitor, volasertib, reduces the expression of cytokine and cell growth ARE mRNAs. PLK1 inhibition caused accelerated mRNA decay in cancer cells and was associated with reduced phosphorylation and stability of the mRNA decay-promoting protein, tristetraprolin (ZFP36/TTP). Ectopic expression of PLK1 increased abundance and stability of high molecular weight of ZFP36/TTP likely of the phosphorylated form. PLK1 effect was associated with the MAPK-MK2 pathway, a major regulator of ARE-mRNA stability, as evident from MK2 inhibition, in vitro phosphorylation, and knockout experiments. Mutational analysis demonstrates that TTP serine 186 is a target for PLK1 effect. Treatment of mice with the PLK1 inhibitor reduced both ZFP36/TTP phosphorylation in xenograft tumor tissues, and the tumor size. In cancer patients' tissues, PLK1/ARE-regulated gene cluster was overexpressed in solid tumors and associated with poor survival. The data showed that PLK1-mediated post-transcriptional aberration could be a therapeutic target.
Collapse
Affiliation(s)
- Qamraa H. Al‐Qahtani
- Molecular BioMedicine ProgramFaisal Specialist Hospital and Research CentreRiyadhKingSaudi Arabia
- Present address:
Department of Pharmacology and ToxicologyCollege of PharmacyKing Saud UniversityRiyadh11495Saudi Arabia
| | - Walid N. Moghrabi
- Molecular BioMedicine ProgramFaisal Specialist Hospital and Research CentreRiyadhKingSaudi Arabia
| | - Suhad Al‐Yahya
- Molecular BioMedicine ProgramFaisal Specialist Hospital and Research CentreRiyadhKingSaudi Arabia
| | - Latifa Al‐Haj
- Molecular BioMedicine ProgramFaisal Specialist Hospital and Research CentreRiyadhKingSaudi Arabia
| | - Maher Al‐Saif
- Molecular BioMedicine ProgramFaisal Specialist Hospital and Research CentreRiyadhKingSaudi Arabia
| | - Linah Mahmoud
- Molecular BioMedicine ProgramFaisal Specialist Hospital and Research CentreRiyadhKingSaudi Arabia
| | - Falah Al‐Mohanna
- Department of Comparative MedicineKing Faisal Specialist Hospital and Research CentreRiyadhSaudi Arabia
| | - Norah Al‐Souhibani
- Molecular BioMedicine ProgramFaisal Specialist Hospital and Research CentreRiyadhKingSaudi Arabia
| | - Ayodele Alaiya
- Stem Cell and Tissue Engineering ProgramFaisal Specialist Hospital and Research CentreRiyadhKingSaudi Arabia
| | - Edward Hitti
- Molecular BioMedicine ProgramFaisal Specialist Hospital and Research CentreRiyadhKingSaudi Arabia
| | - Khalid S. A. Khabar
- Molecular BioMedicine ProgramFaisal Specialist Hospital and Research CentreRiyadhKingSaudi Arabia
| |
Collapse
|
32
|
Shi D, Zhang Z, Kong C. CARMA3 Transcriptional Regulation of STMN1 by NF-κB Promotes Renal Cell Carcinoma Proliferation and Invasion. Technol Cancer Res Treat 2021; 20:15330338211027915. [PMID: 34190011 PMCID: PMC8256254 DOI: 10.1177/15330338211027915] [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] [Indexed: 01/03/2023] Open
Abstract
CARD-containing MAGUK protein 3 (CARMA3) is associated with tumor occurrence and progression. However, the signaling pathways involved in CARMA3 function remain unclear. We aimed to analyze the association between CARMA3 and stathmin (STMN1) through the NF-κB pathway, which is associated with cell proliferation and invasion, in clear cell renal cell carcinoma (ccRCC). We evaluated the effects of CARMA3 and STMN1 expression on cell migration, proliferation, and invasion in various cell lines, and their expression in tissue samples from patients with ccRCC. CARMA3 was highly expressed in ccRCC tissues and cell lines. Moreover, CARMA3 promoted the proliferation and invasion of RCC cells by activating the NF-κB pathway to transcribe STMN1. Stathmin exhibited a consistent profile with CARMA3 in ccRCC tissue, and could be an effector for CARMA3-activated cell proliferation and invasion of ccRCC cells. In summary, CARMA3 may serve as a promising target for ccRCC treatment.
Collapse
Affiliation(s)
- Du Shi
- Department of Urology, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning, China
| | - Zhe Zhang
- Department of Urology, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning, China
| | - Chuize Kong
- Department of Urology, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning, China
| |
Collapse
|
33
|
Chen SK, Wang YC, Lin TY, Wu HJ, Huang CJ, Ku WC. G-Protein-coupled Estrogen Receptor 1 Agonist G-1 Perturbs Sunitinib Resistance-related Phosphoproteomic Signatures in Renal Cell Carcinoma. Cancer Genomics Proteomics 2021; 18:207-220. [PMID: 33893075 DOI: 10.21873/cgp.20253] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Revised: 03/10/2021] [Accepted: 03/18/2021] [Indexed: 12/29/2022] Open
Abstract
BACKGROUND Metastatic renal cell carcinoma (RCC) often develops resistance to first-line targeted therapy such as sunitinib. G-Protein-coupled estrogen receptor 1 (GPER1) agonist G-1 was recently reported to regulate RCC physiology but the role of G-1 in RCC tumorigenesis and sunitinib resistance remains largely unknown. MATERIALS AND METHODS Parental and sunitinib-resistant 786-O cells were treated with GPER1 agonist G-1, and quantitative phosphoproteomics was performed. Bioinformatic analyses and validations, including immunoblotting, cell migration, and cell cycle distribution, were performed. RESULTS G-1 repressed cell proliferation and migration in both parental and sunitinib-resistant 786-O cells. Phosphoproteomic signatures, including phosphoinositide 3-kinase and protein kinase B (PI3K-AKT) as well as other pathways, were up-regulated in sunitinib-resistant cells but application of G-1 reversed this effect. Among phosphoprotein candidates, activating transcription factor 2 (ATF2) Thr69/71 phosphorylation was antagonistically regulated by sunitinib resistance and G-1. CONCLUSION Our results open up the possibility for managing RCC and sunitinib resistance by GPER1 agonist G-1 and its regulated pathways.
Collapse
Affiliation(s)
- Shao-Kuan Chen
- School of Medicine, College of Medicine, Fu Jen Catholic University, New Taipei City, Taiwan, R.O.C.,Department of Surgery, Sijhih Cathay General Hospital, New Taipei City, Taiwan, R.O.C
| | - Yen-Chieh Wang
- School of Medicine, College of Medicine, Fu Jen Catholic University, New Taipei City, Taiwan, R.O.C.,Division of Urology, Cathay General Hospital, Taipei City, Taiwan, R.O.C
| | - Tai-Yuan Lin
- School of Medicine, College of Medicine, Fu Jen Catholic University, New Taipei City, Taiwan, R.O.C
| | - Hsin-Jou Wu
- School of Medicine, College of Medicine, Fu Jen Catholic University, New Taipei City, Taiwan, R.O.C
| | - Chi-Jung Huang
- Department of Medical Research, Cathay General Hospital, Taipei City, Taiwan, R.O.C.,Department of Biochemistry, National Defense Medical Center, Taipei City, Taiwan, R.O.C
| | - Wei-Chi Ku
- School of Medicine, College of Medicine, Fu Jen Catholic University, New Taipei City, Taiwan, R.O.C.;
| |
Collapse
|
34
|
Li L, Dong S, Li S, Xu Q, Wang S, Xiong Y, Cheng Y, Zhong M, Zhang G, Hu S. Downregulation of circular RNA circDOCK7 identified from diabetic rats after sleeve gastrectomy contributes to hepatocyte apoptosis through regulating miR-139-3p and MCM3. Biochem Biophys Res Commun 2021; 548:134-142. [PMID: 33640606 DOI: 10.1016/j.bbrc.2021.02.069] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Accepted: 02/16/2021] [Indexed: 12/27/2022]
Abstract
Sleeve gastrectomy (SG) is the most widely used bariatric procedures globally, which could improve glucose and lipid metabolism dramatically. Circular RNAs (circRNAs) are being increasingly implicated in numerous pathophysiological processes. However, for diabetes mellitus (DM), the expression and function of circRNAs remain largely undetermined, in particular, whether circRNAs mediate the amelioration of DM observed after SG. Using a diabetic rat model, we subjected liver tissue from SG and sham-operated rats to RNA sequencing. Amongst the 103 differentially regulated circRNAs identified in diabetic rats after SG, we focused on circDOCK7, a highly expressed circRNA derived from the back-splicing of the DOCK7 gene. Silencing of circDOCK7 significantly inhibited cellular proliferation and induction of apoptosis in insulin-resistant rat hepatocytes. Further analysis indicated circDOCK7 harbored binding sites for miR-139-3p and regulated the expression of minichromosome maintenance 3 (MCM3) through sequestration of miR-139-3p. Our findings therefore demonstrate a novel regulatory pathway involving circDOCK7 that regulates cellular proliferation and apoptosis through increasing the expression of MCM3. Overall, our study establishes a list of specific circRNAs expressed in diabetic rat liver after SG including circDOCK7 which serve as potential biomarkers and treatment targets for DM patients.
Collapse
Affiliation(s)
- Linchuan Li
- Department of General Surgery, Shandong Qianfoshan Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Shuohui Dong
- Department of General Surgery, Shandong Qianfoshan Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Songhan Li
- Department of General Surgery, Shandong Qianfoshan Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Qian Xu
- Department of General Surgery, Shandong Qianfoshan Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Shuo Wang
- Department of General Surgery, Shandong Qianfoshan Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Yacheng Xiong
- Department of General Surgery, Shandong Qianfoshan Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Yugang Cheng
- Department of General Surgery, The First Affiliated Hospital of Shandong First Medical University, Jinan, Shandong, China
| | - Mingwei Zhong
- Department of General Surgery, The First Affiliated Hospital of Shandong First Medical University, Jinan, Shandong, China
| | - Guangyong Zhang
- Department of General Surgery, The First Affiliated Hospital of Shandong First Medical University, Jinan, Shandong, China
| | - Sanyuan Hu
- Department of General Surgery, Shandong Qianfoshan Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China.
| |
Collapse
|
35
|
Ren Z, Li J, Zhao S, Qiao Q, Li R. Knockdown of MCM8 functions as a strategy to inhibit the development and progression of osteosarcoma through regulating CTGF. Cell Death Dis 2021; 12:376. [PMID: 33828075 PMCID: PMC8027380 DOI: 10.1038/s41419-021-03621-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Revised: 03/09/2021] [Accepted: 03/11/2021] [Indexed: 12/19/2022]
Abstract
Osteosarcoma is the most common primary malignant tumor of bone derived from osteoblasts, which is a noteworthy threat to the health of children and adolescents. In this study, we found that MCM8 has significantly higher expression level in osteosarcoma tissues in comparison with normal tissues, which was also correlated with more advanced tumor grade and pathological stage. In agreement with the role of MCM proteins as indicators of cell proliferation, knockdown/overexpression of MCM8 inhibited/promoted osteosarcoma cell proliferation in vitro and tumor growth in vivo. Also, MCM8 knockdown/overexpression was also significantly associated with the promotion/inhibition of cell apoptosis and suppression/promotion of cell migration. More importantly, mechanistic study identified CTGF as a potential downstream target of MCM8, silencing of which could enhance the regulatory effects of MCM8 knockdown and alleviate the effects of MCM8 overexpression on osteosarcoma development. In summary, MCM8/CTGF axis was revealed as critical participant in the development and progression of osteosarcoma and MCM8 may be a promising therapeutic target for osteosarcoma treatment.
Collapse
Affiliation(s)
- Zhinan Ren
- Department of Orthopedics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Jun Li
- Department of Orthopedics, The Second Affiliated Hospital of Anhui Medical University, 678 Furong, Hefei, 230601, China
| | - Shanwen Zhao
- Department of Foot and Ankle Surgery, Center for Orthopaedic Surgery, The Third Affiliated Hospital of Southern Medical University, Guangzhou, 510610, China.,Orthopaedic Hospital of Guangdong Province, Guangzhou, 510630, China.,Academy of Orthopaedics, Guangdong Province, Guangzhou, 510630, China.,Guangdong Provincial Key Laboratory of Bone and Joint Degenerative Diseases, Guangzhou, 510515, China
| | - Qi Qiao
- Department of Orthopedics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China.
| | - Runguang Li
- Department of Foot and Ankle Surgery, Center for Orthopaedic Surgery, The Third Affiliated Hospital of Southern Medical University, Guangzhou, 510610, China. .,Orthopaedic Hospital of Guangdong Province, Guangzhou, 510630, China. .,Academy of Orthopaedics, Guangdong Province, Guangzhou, 510630, China. .,Guangdong Provincial Key Laboratory of Bone and Joint Degenerative Diseases, Guangzhou, 510515, China. .,Department of Orthopedics, Linzhi People's Hospital, Linzhi, 860000, China.
| |
Collapse
|
36
|
Jin B, Jin D, Zhuo Z, Zhang B, Chen K. MiR-1224-5p Activates Autophagy, Cell Invasion and Inhibits Epithelial-to-Mesenchymal Transition in Osteosarcoma Cells by Directly Targeting PLK1 Through PI3K/AKT/mTOR Signaling Pathway. Onco Targets Ther 2020; 13:11807-11818. [PMID: 33235467 PMCID: PMC7680192 DOI: 10.2147/ott.s274451] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Accepted: 09/23/2020] [Indexed: 12/12/2022] Open
Abstract
Background Osteosarcoma (OS) is one of the most common malignant bone tumors with a poor overall prognosis. MiR-1224-5p plays an important role in cancer, but its function and mechanism in OS have not been studied. Materials and Methods The expression of miR-1224-5p and PLK1 was detected by qRT-PCR in OS cells, adjacent tissues, and cell lines. Dual-luciferase reporter gene assay was used to verify the interaction between miR-1224-5p and PLK1. The expression of miR-1224-5p and PLK1 was intervened by transfection with miR-1224-5p mimic, NC mimic, pc-NC and PLK1, respectively. MTT, colony formation assay, Transwell and flow cytometry were used to observe the cell proliferation, invasion and apoptosis. Western blot was used to detect the expression levels of PLK1, PI3K/AKT/mTOR signaling pathway-related proteins, autophagy-related proteins, and epithelial-mesenchymal transition (EMT)-related proteins in the cells. Results We found that miR-1224-5p was down-regulated and PLK1 expression was up-regulated in OS tissues and cells. On the other hand, it is further confirmed that PLK1 was a target gene of miR-1224-5p. Overexpression of miR-1224-5p inhibited the proliferation, invasion while promoted the apoptosis of OS cells, whereas overexpression of PLK1 promoted the proliferation, invasion and inhibited the apoptosis of OS cells. In the miR-1224-5p group (overexpression of miR-1224-5p), PI3K, AKT, and mTOR protein phosphorylation levels were significantly reduced, while autophagic activity was significantly activated, and the degree of EMT was significantly reduced. But the results in the PLK1 group (overexpression of PLK1) were the opposite. In addition, overexpression of miR-1224-5p reversed the effect of PLK1 upregulation on OS cells. Conclusion MiR-1224-5p targets PLK1 to inhibit PI3K/AKT/mTOR signaling pathway, thus mediating the proliferation, invasion, apoptosis, autophagy and EMT in OS cells.
Collapse
Affiliation(s)
- Bicheng Jin
- Department of Surgery, Guizhou Electric Power Staff Hospital, Guiyang, Guizhou Province, People's Republic of China
| | - Dongfang Jin
- Department of Clinical Laboratory, Jinhua People's Hospital, Jinhua, Zhejiang Province, People's Republic of China
| | - Zhaozhen Zhuo
- Prenatal Diagnosis Center, Guizhou Provincial People's Hospital, Guiyang, Guizhou Province, People's Republic of China
| | - Bo Zhang
- Department of Surgery, Guizhou Electric Power Staff Hospital, Guiyang, Guizhou Province, People's Republic of China
| | - Kun Chen
- Guizhou Provincial People's Hospital Scientific Research Center Laboratory, Guiyang, Guizhou Province, People's Republic of China
| |
Collapse
|
37
|
Chen Y, Chen X, Ji Y, Zhu S, Bu F, Du X, Meng X, Huang C, Li J. PLK1 regulates hepatic stellate cell activation and liver fibrosis through Wnt/β-catenin signalling pathway. J Cell Mol Med 2020; 24:7405-7416. [PMID: 32463161 PMCID: PMC7339205 DOI: 10.1111/jcmm.15356] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2020] [Revised: 03/18/2020] [Accepted: 04/12/2020] [Indexed: 12/14/2022] Open
Abstract
As an outcome of chronic liver disease, liver fibrosis involves the activation of hepatic stellate cells (HSCs) caused by a variety of chronic liver injuries. It is important to explore approaches to inhibit the activation and proliferation of HSCs for the treatment of liver fibrosis. PLK1 is overexpressed in many human tumour cells and has become a popular drug target in tumour therapy. Therefore, further study of the function of PLK1 in the cell cycle is valid. In the present study, we found that PLK1 expression was elevated in primary HSCs isolated from CCl4 -induced liver fibrosis mice and LX-2 cells stimulated with TGF-β1. Knockdown of PLK1 inhibited α-SMA and Col1α1 expression and reduced the activation of HSCs in CCl4 -induced liver fibrosis mice and LX-2 cells stimulated with TGF-β1. We further showed that inhibiting the expression of PLK1 reduced the proliferation of HSCs and promoted HSCs apoptosis in vivo and in vitro. Furthermore, we found that the Wnt/β-catenin signalling pathway may be essential for PLK1-mediated HSCs activation. Together, blocking PLK1 effectively suppressed liver fibrosis by inhibiting HSC activation, which may provide a new treatment strategy for liver fibrosis.
Collapse
Affiliation(s)
- Yu Chen
- School of PharmacyAnhui Key Laboratory of Major Autoimmune DiseasesAnhui Institute of Innovative DrugsAnhui Medical UniversityHefeiChina
- The Key Laboratory of Anti‐inflammatory and Immune MedicinesAnhui Medical UniversityMinistry of EducationHefeiChina
- Institute for Liver Diseases of Anhui Medical UniversityILD‐AMUAnhui Medical UniversityHefeiChina
| | - Xin Chen
- School of PharmacyAnhui Key Laboratory of Major Autoimmune DiseasesAnhui Institute of Innovative DrugsAnhui Medical UniversityHefeiChina
- The Key Laboratory of Anti‐inflammatory and Immune MedicinesAnhui Medical UniversityMinistry of EducationHefeiChina
- Institute for Liver Diseases of Anhui Medical UniversityILD‐AMUAnhui Medical UniversityHefeiChina
| | - Ya‐Ru Ji
- The Key Laboratory of Anti‐inflammatory and Immune MedicinesAnhui Medical UniversityMinistry of EducationHefeiChina
- Institute for Liver Diseases of Anhui Medical UniversityILD‐AMUAnhui Medical UniversityHefeiChina
- Anhui Province Key Laboratory of Major Autoimmune DiseasesAnhui Institute of Innovative DrugsFirst Affiliated HospitalAnhui Medical UniversityHefeiChina
| | - Sai Zhu
- School of PharmacyAnhui Key Laboratory of Major Autoimmune DiseasesAnhui Institute of Innovative DrugsAnhui Medical UniversityHefeiChina
- The Key Laboratory of Anti‐inflammatory and Immune MedicinesAnhui Medical UniversityMinistry of EducationHefeiChina
- Institute for Liver Diseases of Anhui Medical UniversityILD‐AMUAnhui Medical UniversityHefeiChina
| | - Fang‐Tian Bu
- School of PharmacyAnhui Key Laboratory of Major Autoimmune DiseasesAnhui Institute of Innovative DrugsAnhui Medical UniversityHefeiChina
- The Key Laboratory of Anti‐inflammatory and Immune MedicinesAnhui Medical UniversityMinistry of EducationHefeiChina
- Institute for Liver Diseases of Anhui Medical UniversityILD‐AMUAnhui Medical UniversityHefeiChina
| | - Xiao‐Sa Du
- School of PharmacyAnhui Key Laboratory of Major Autoimmune DiseasesAnhui Institute of Innovative DrugsAnhui Medical UniversityHefeiChina
- The Key Laboratory of Anti‐inflammatory and Immune MedicinesAnhui Medical UniversityMinistry of EducationHefeiChina
- Institute for Liver Diseases of Anhui Medical UniversityILD‐AMUAnhui Medical UniversityHefeiChina
| | - Xiao‐Ming Meng
- School of PharmacyAnhui Key Laboratory of Major Autoimmune DiseasesAnhui Institute of Innovative DrugsAnhui Medical UniversityHefeiChina
- The Key Laboratory of Anti‐inflammatory and Immune MedicinesAnhui Medical UniversityMinistry of EducationHefeiChina
- Institute for Liver Diseases of Anhui Medical UniversityILD‐AMUAnhui Medical UniversityHefeiChina
| | - Cheng Huang
- School of PharmacyAnhui Key Laboratory of Major Autoimmune DiseasesAnhui Institute of Innovative DrugsAnhui Medical UniversityHefeiChina
- The Key Laboratory of Anti‐inflammatory and Immune MedicinesAnhui Medical UniversityMinistry of EducationHefeiChina
- Institute for Liver Diseases of Anhui Medical UniversityILD‐AMUAnhui Medical UniversityHefeiChina
| | - Jun Li
- School of PharmacyAnhui Key Laboratory of Major Autoimmune DiseasesAnhui Institute of Innovative DrugsAnhui Medical UniversityHefeiChina
- The Key Laboratory of Anti‐inflammatory and Immune MedicinesAnhui Medical UniversityMinistry of EducationHefeiChina
- Institute for Liver Diseases of Anhui Medical UniversityILD‐AMUAnhui Medical UniversityHefeiChina
| |
Collapse
|
38
|
ZNF143 Suppresses Cell Apoptosis and Promotes Proliferation in Gastric Cancer via ROS/p53 Axis. DISEASE MARKERS 2020; 2020:5863178. [PMID: 32076462 PMCID: PMC7017572 DOI: 10.1155/2020/5863178] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/21/2019] [Accepted: 11/06/2019] [Indexed: 12/16/2022]
Abstract
Aim This study was aimed at identifying the role of zinc finger protein 143 (ZNF143) in gastric cancer (GC) progression. Methods The impact of ZNF143 on the proliferation ability and apoptosis of GC cells was detected. The expression of ZNF143 and related targeted genes was determined using Western blot analysis. The reactive oxygen species (ROS) level of GC cells was examined using the ROS generation assay. The role of ZNF143 in the proliferation of GC cells in vivo was examined using tumor xenograft assay. Results The ectopic overexpression of ZNF143 promoted the proliferation of GC cells, while its knockdown reduced the effect in vitro. The downregulation of ZNF143 facilitated cell apoptosis. ZNF143 decreased the ROS level in GC cells, resulting in the reduction of cell apoptosis. Transfection with p53 reversed the antiapoptotic effect of ZNF143, while pifithrin-α, a specific inhibitor of p53, reduced the apoptosis in ZNF143-knockdown GC cells. However, p53 had no influence on the ROS level in GC cells. p53 played a key role in inhibiting ROS generation in GC cells, thereby inhibiting apoptosis. The transplanted tumor weight and volume were higher in the ZNF143-overexpressed group than in the ZNF143-knockdown group in vivo was examined using tumor xenograft assay. Conclusion ZNF143, as a tumor oncogene, promoted the proliferation of GC cells both in vitro and in vivo, indicating that ZNF143 might function as a novel target for GC therapy.in vitro. The downregulation of ZNF143 facilitated cell apoptosis. ZNF143 decreased the ROS level in GC cells, resulting in the reduction of cell apoptosis. Transfection with p53 reversed the antiapoptotic effect of ZNF143, while pifithrin-in vivo was examined using tumor xenograft assay.
Collapse
|