1151
|
Liu J, Hao Y, Wang C, Jin Y, Yang Y, Gu J, Chen X. An Optimized Isotopic Photocleavable Tagging Strategy for Site-Specific and Quantitative Profiling of Protein O-GlcNAcylation in Colorectal Cancer Metastasis. ACS Chem Biol 2022; 17:513-520. [PMID: 35254053 DOI: 10.1021/acschembio.1c00981] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
O-linked-β-N-acetylglucosamine (O-GlcNAc) glycosylation is a ubiquitous protein post-translational modification of the emerging importance in metazoans. Of the thousands of O-GlcNAcylated proteins identified, many carry multiple modification sites with varied stoichiometry. To better match the scale of O-GlcNAc sites and their dynamic nature, we herein report an optimized strategy, termed isotopic photocleavable tagging for O-GlcNAc profiling (isoPTOP), which enables quantitative and site-specific profiling of O-GlcNAcylation with excellent specificity and sensitivity. In HeLa cells, ∼1500 O-GlcNAcylation sites were identified with the optimized procedures, which led to quantification of ∼1000 O-GlcNAcylation sites with isoPTOP. Furthermore, we apply isoPTOP to probe the O-GlcNAcylation dynamics in a pair of colorectal cancer (CRC) cell lines, SW480 and SW620 cells, which represent primary carcinoma and metastatic cells, representatively. The stoichiometric differences of 625 O-GlcNAcylation sites are quantified. Of these quantified sites, many occur on important regulators involved in tumor progression and metastasis. Our results provide a valuable database for understanding the functional role of O-GlcNAc in CRC. IsoPTOP should be applicable for investigating O-GlcNAcylation dynamics in various pathophysiological processes.
Collapse
Affiliation(s)
- Jialin Liu
- College of Chemistry and Molecular Engineering, Peking University, Beijing, 100871, China
- Beijing National Laboratory for Molecular Sciences, Peking University, Beijing, 100871, China
| | - Yi Hao
- College of Chemistry and Molecular Engineering, Peking University, Beijing, 100871, China
- Beijing National Laboratory for Molecular Sciences, Peking University, Beijing, 100871, China
| | - Chunting Wang
- College of Chemistry and Molecular Engineering, Peking University, Beijing, 100871, China
- Beijing National Laboratory for Molecular Sciences, Peking University, Beijing, 100871, China
| | - Yangya’nan Jin
- College of Chemistry and Molecular Engineering, Peking University, Beijing, 100871, China
- Beijing National Laboratory for Molecular Sciences, Peking University, Beijing, 100871, China
| | - Yong Yang
- Department of Gastrointestinal Surgery, Peking University Shougang Hospital, Beijing, 100144, China
- Gastrointestinal Cancer Center, Peking University Cancer Hospital, Beijing, 100142, China
| | - Jin Gu
- Department of Gastrointestinal Surgery, Peking University Shougang Hospital, Beijing, 100144, China
- Gastrointestinal Cancer Center, Peking University Cancer Hospital, Beijing, 100142, China
- Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, 100871, China
| | - Xing Chen
- College of Chemistry and Molecular Engineering, Peking University, Beijing, 100871, China
- Beijing National Laboratory for Molecular Sciences, Peking University, Beijing, 100871, China
- Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, 100871, China
- Synthetic and Functional Biomolecules Center, Peking University, Beijing, 100871, China
- Key Laboratory of Bioorganic Chemistry and Molecular Engineering of Ministry of Education, Peking University, Beijing, 100871, China
| |
Collapse
|
1152
|
Du J, Tao Q, Liu Y, Huang Z, Jin H, Lin W, Huang X, Zeng J, Zhao Y, Liu L, Xu Q, Han X, Chen L, Chen XL, Wen Y. Assessment of the targeted effect of Sijunzi decoction on the colorectal cancer microenvironment via the ESTIMATE algorithm. PLoS One 2022; 17:e0264720. [PMID: 35303006 PMCID: PMC8932555 DOI: 10.1371/journal.pone.0264720] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2021] [Accepted: 02/15/2022] [Indexed: 11/18/2022] Open
Abstract
Objective Sijunzi decoction (SJZD) was used to treat patients with colorectal cancer (CRC) as an adjuvant method. The aim of the study was to investigate the therapeutic targets and pathways of SJZD towards the tumor microenvironment of CRC via network pharmacology and the ESTIMATE algorithm. Methods The ESTIMATE algorithm was used to calculate immune and stromal scores to predict the level of infiltrating immune and stromal cells. The active targets of SJZD were searched in the Traditional Chinese Medicine Systems Pharmacology Database and Analysis Platform (TCMSP) and UniProt database. The core targets were obtained by matching the differentially expressed genes in CRC tissues and the targets of SJZD. Then, GO, KEGG and validation in TCGA were carried out. Results According to the ESTIMATE algorithm and survival analysis, the median survival time of the low stromal score group was significantly higher than that of the high stromal score group (P = 0.018), while the patients showed no significant difference of OS between different immune groups (P = 0.19). A total of 929 genes were upregulated and 115 genes were downregulated between the stromal score groups (|logFC| > 2, adjusted P < 0.05); 357 genes were upregulated and 472 genes were downregulated between the immune score groups. The component-target network included 139 active components and 52 related targets. The core targets were HSPB1, SPP1, IGFBP3, and TGFB1, which were significantly associated with poor prognosis in TCGA validation. GO terms included the response to hypoxia, the extracellular space, protein binding and the TNF signaling pathway. Immunoreaction was the main enriched pathway identified by KEGG analysis. Conclusion The core genes (HSPB1, SPP1, IGFBP3 and TGFB1) affected CRC development and prognosis by regulating hypoxia, protein binding and epithelial-mesenchymal transition in the extracellular matrix.
Collapse
Affiliation(s)
- Jiaxin Du
- School of Basic Medical Science, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Quyuan Tao
- School of Basic Medical Science, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Ying Liu
- School of Basic Medical Science, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Zhanming Huang
- The First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - He Jin
- School of Basic Medical Science, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Wenjia Lin
- School of Basic Medical Science, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Xinying Huang
- School of Basic Medical Science, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Jingyan Zeng
- Shenzhen Clinical College, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yongchang Zhao
- The First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Lingyu Liu
- School of Basic Medical Science, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Qian Xu
- School of Basic Medical Science, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Xue Han
- School of Basic Medical Science, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Lixia Chen
- School of Basic Medical Science, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Xin-lin Chen
- School of Basic Medical Science, Guangzhou University of Chinese Medicine, Guangzhou, China
- * E-mail: (XC); (YW)
| | - Yi Wen
- The First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, China
- * E-mail: (XC); (YW)
| |
Collapse
|
1153
|
Zhang S, He Y, Xuan Q, Ling X, Men K, Zhao X, Xue D, Li L, Zhang Y. TMEM139 prevents NSCLC metastasis by inhibiting lysosomal degradation of E-cadherin. Cancer Sci 2022; 113:1999-2007. [PMID: 35302694 PMCID: PMC9207374 DOI: 10.1111/cas.15341] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Revised: 03/10/2022] [Accepted: 03/12/2022] [Indexed: 11/29/2022] Open
Abstract
Non‐small‐cell lung cancer (NSCLC) accounts for approximately 85% of all lung cancer cases and has the highest mortality rate among all solid tumors. It is characterized by early metastasis, and investigations of the molecular mechanisms underlying the progression and metastasis of NSCLC are urgently needed for the development of therapeutic targets. Here, we report that the transmembrane protein TMEM139 is significantly downregulated in NSCLC and that reduced expression of TMEM139 is correlated with a poor prognosis in NSCLC patients. Mechanistically, we found that TMEM139 directly interacts with E‐cadherin at the plasma membrane and at focal adhesion sites. Moreover, TMEM139 can prevent the lysosomal degradation of E‐cadherin, which inhibits epithelial‐mesenchymal transition, migration and invasion of NSCLC cells both in vitro and in vivo. Our study not only expands our understanding of NSCLC metastasis but also provides a foundation to develop novel therapeutic strategies.
Collapse
Affiliation(s)
- Shuai Zhang
- Department of Medical Oncology, Harbin Medical University Cancer Hospital, Harbin, China
| | - Yunlong He
- Department of radiation oncology, Harbin Medical University Cancer Hospital, Harbin, China
| | - Qijia Xuan
- Department of Medical Oncology, The Fourth Affiliated Hospital Zhejiang University School of Medicine, Yiwu, Zhejiang Province, China
| | - Xiaodong Ling
- Department of Thoracic Surgery, Harbin Medical University Cancer Hospital, Harbin, China
| | - Kaiya Men
- Department of Medical Oncology, Harbin Medical University Cancer Hospital, Harbin, China
| | - Xu Zhao
- Department of Medical Oncology, Harbin Medical University Cancer Hospital, Harbin, China
| | - Dinglong Xue
- Department of Medical Oncology, Harbin Medical University Cancer Hospital, Harbin, China
| | - Ling Li
- Department of Medical Oncology, Harbin Medical University Cancer Hospital, Harbin, China
| | - Yingyin Zhang
- Department of Medical Oncology, Hu Lun Bei Er Ren Min Hospital, Inner Mongolia Province, China
| |
Collapse
|
1154
|
Wang Y, Huang B, Jin T, Ocansey DKW, Jiang J, Mao F. Intestinal Fibrosis in Inflammatory Bowel Disease and the Prospects of Mesenchymal Stem Cell Therapy. Front Immunol 2022; 13:835005. [PMID: 35370998 PMCID: PMC8971815 DOI: 10.3389/fimmu.2022.835005] [Citation(s) in RCA: 52] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Accepted: 01/26/2022] [Indexed: 12/12/2022] Open
Abstract
Intestinal fibrosis is an important complication of inflammatory bowel disease (IBD). In the course of the development of fibrosis, certain parts of the intestine become narrowed, significantly destroying the structure and function of the intestine and affecting the quality of life of patients. Chronic inflammation is an important initiating factor of fibrosis. Unfortunately, the existing anti-inflammatory drugs cannot effectively prevent and alleviate fibrosis, and there is no effective anti-fibrotic drug, which makes surgical treatment the mainstream treatment for intestinal fibrosis and stenosis. Mesenchymal stem cells (MSCs) are capable of tissue regeneration and repair through their self-differentiation, secretion of cytokines, and secretion of extracellular vesicles. MSCs have been shown to play an important therapeutic role in the fibrosis of many organs. However, the role of MSC in intestinal fibrosis largely remained unexplored. This review summarizes the mechanism of intestinal fibrosis, including the role of immune cells, TGF-β, and the gut microbiome and metabolites. Available treatment options for fibrosis, particularly, MSCs are also discussed.
Collapse
Affiliation(s)
- Yifei Wang
- Aoyang Institute of Cancer, Affiliated Aoyang Hospital of Jiangsu University, Suzhou, China
- Key Laboratory of Medical Science and Laboratory Medicine of Jiangsu Province, School of Medicine, Jiangsu University, Zhenjiang, China
| | - Bin Huang
- Aoyang Institute of Cancer, Affiliated Aoyang Hospital of Jiangsu University, Suzhou, China
- General Surgery Department, Affiliated Aoyang Hospital of Jiangsu University, Suzhou, China
| | - Tao Jin
- Department of Gastrointestinal and Endoscopy, The Affiliated Yixing Hospital of Jiangsu University, Yixing, China
| | - Dickson Kofi Wiredu Ocansey
- Key Laboratory of Medical Science and Laboratory Medicine of Jiangsu Province, School of Medicine, Jiangsu University, Zhenjiang, China
- Directorate of University Health Services, University of Cape Coast, Cape Coast, Ghana
| | - Jiajia Jiang
- Aoyang Institute of Cancer, Affiliated Aoyang Hospital of Jiangsu University, Suzhou, China
- Key Laboratory of Medical Science and Laboratory Medicine of Jiangsu Province, School of Medicine, Jiangsu University, Zhenjiang, China
- *Correspondence: Jiajia Jiang, ; Fei Mao,
| | - Fei Mao
- Aoyang Institute of Cancer, Affiliated Aoyang Hospital of Jiangsu University, Suzhou, China
- Key Laboratory of Medical Science and Laboratory Medicine of Jiangsu Province, School of Medicine, Jiangsu University, Zhenjiang, China
- *Correspondence: Jiajia Jiang, ; Fei Mao,
| |
Collapse
|
1155
|
Chen S, Li D, Yu D, Li M, Ye L, Jiang Y, Tang S, Zhang R, Xu C, Jiang S, Wang Z, Aschner M, Zheng Y, Chen L, Chen W. Determination of tipping point in course of PM 2.5 organic extracts-induced malignant transformation by dynamic network biomarkers. JOURNAL OF HAZARDOUS MATERIALS 2022; 426:128089. [PMID: 34933256 DOI: 10.1016/j.jhazmat.2021.128089] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/06/2021] [Revised: 12/11/2021] [Accepted: 12/13/2021] [Indexed: 06/14/2023]
Abstract
The dynamic network biomarkers (DNBs) are designed to identify the tipping point and specific molecules in initiation of PM2.5-induced lung cancers. To discover early-warning signals, we analyzed time-series gene expression datasets over a course of PM2.5 organic extraction-induced human bronchial epithelial (HBE) cell transformation (0th~16th week). A composition index of DNB (CIDNB) was calculated to determine correlations and fluctuations in molecule clusters at each timepoint. We identified a group of genes with the highest CIDNB at the 10th week, implicating a tipping point and corresponding DNBs. Functional experiments revealed that manipulating respective DNB genes at the tipping point led to remarkable changes in malignant phenotypes, including four promoters (GAB2, NCF1, MMP25, LAPTM5) and three suppressors (BATF2, DOK3, DAP3). Notably, co-altered expression of seven core DNB genes resulted in an enhanced activity of malignant transformation compared to effects of single-gene manipulation. Perturbation of pathways (EMT, HMGB1, STAT3, NF-κB, PTEN) appeared in HBE cells at the tipping point. The core DNB genes were involved in regulating lung cancer cell growth and associated with poor survival, indicating their synergistic effects in initiation and development of lung cancers. These findings provided novel insights into the mechanism of dynamic networks attributable to PM2.5-induced cell transformation.
Collapse
Affiliation(s)
- Shen Chen
- Department of Toxicology, School of Public Health, Sun Yat-sen University, 74 Zhongshan Road 2, Guangzhou 510080, China
| | - Daochuan Li
- Department of Toxicology, School of Public Health, Sun Yat-sen University, 74 Zhongshan Road 2, Guangzhou 510080, China
| | - Dianke Yu
- Department of Toxicology, School of Public Health, Qingdao University, Qingdao 266021, China
| | - Miao Li
- Department of Toxicology, School of Public Health, Sun Yat-sen University, 74 Zhongshan Road 2, Guangzhou 510080, China
| | - Lizhu Ye
- Department of Toxicology, School of Public Health, Sun Yat-sen University, 74 Zhongshan Road 2, Guangzhou 510080, China
| | - Yue Jiang
- Department of Toxicology, School of Public Health, Sun Yat-sen University, 74 Zhongshan Road 2, Guangzhou 510080, China
| | - Shijie Tang
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai 200031, China
| | - Rui Zhang
- Department of Toxicology, School of Public Health, Sun Yat-sen University, 74 Zhongshan Road 2, Guangzhou 510080, China
| | - Chi Xu
- Department of Toxicology, School of Public Health, Sun Yat-sen University, 74 Zhongshan Road 2, Guangzhou 510080, China
| | - Shuyun Jiang
- Department of Toxicology, School of Public Health, Sun Yat-sen University, 74 Zhongshan Road 2, Guangzhou 510080, China
| | - Ziwei Wang
- Department of Toxicology, School of Public Health, Sun Yat-sen University, 74 Zhongshan Road 2, Guangzhou 510080, China
| | - Michael Aschner
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Forchheimer 209, 1300 Morris Park Avenue, Bronx, NY, 10461, USA
| | - Yuxin Zheng
- Department of Toxicology, School of Public Health, Qingdao University, Qingdao 266021, China
| | - Liping Chen
- Department of Toxicology, School of Public Health, Sun Yat-sen University, 74 Zhongshan Road 2, Guangzhou 510080, China.
| | - Wen Chen
- Department of Toxicology, School of Public Health, Sun Yat-sen University, 74 Zhongshan Road 2, Guangzhou 510080, China.
| |
Collapse
|
1156
|
Abstract
Natural killer (NK) cells are innate immune cells that are critical to the body's antitumor and antimetastatic defense. As such, novel therapies are being developed to utilize NK cells as part of a next generation of immunotherapies to treat patients with metastatic disease. Therefore, it is essential for us to examine how metastatic cancer cells and NK cells interact with each other throughout the metastatic cascade. In this Review, we highlight the recent body of work that has begun to answer these questions. We explore how the unique biology of cancer cells at each stage of metastasis alters fundamental NK cell biology, including how cancer cells can evade immunosurveillance and co-opt NK cells into cells that promote metastasis. We also discuss the translational potential of this knowledge.
Collapse
Affiliation(s)
- Isaac S. Chan
- Department of Internal Medicine, Division of Hematology and Oncology, and
- Harold C. Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Andrew J. Ewald
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, and
- Department of Cell Biology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
1157
|
Le F, Li HM, Lv QL, Chen JJ, Lin QX, Ji YL, Yi B. lncRNA ZNF674-AS1 inhibits the migration, invasion and epithelial-mesenchymal transition of thyroid cancer cells by modulating the miR-181a/SOCS4 axis. Mol Cell Endocrinol 2022; 544:111551. [PMID: 34990740 DOI: 10.1016/j.mce.2021.111551] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Revised: 12/28/2021] [Accepted: 12/31/2021] [Indexed: 01/11/2023]
Abstract
Thyroid cancer (TC) is a very common endocrine cancer worldwide. Further understanding and revealing the molecular mechanism underlying thyroid cancer are indispensable for the development of effective diagnosis and treatments. Long non-coding RNAs (lncRNAs), a series of non-coding RNAs with a length of >200 nts, have been regarded as crucial regulators of many cancers playing a tumor suppressive or oncogenic role, depending on circumstances. lncRNA ZNF674-AS1 was reported to be abnormally expressed in TC, but the exact mechanism remains unclear. This study aims to probe the mechanism and roles of ZNF674-AS1 in TC. The expression patterns of RNAs and proteins were determined via qRT-PCR and western blotting, respectively. Cell proliferation, migration and invasion were detected using MTT and Transwell assays. ZNF674-AS1 and SOCS4 expression were remarkably reduced while miR-181a was upregulated in TC tissues and cells. Enforced expression of ZNF674-AS1 inhibited proliferation, migration, invasion and epithelial-mesenchymal transition (EMT) in vitro and reduced tumour growth in vivo. Mechanistic assays verified that ZNF674-AS1 directly interacted with miR-181a to increase SOCS4 expression. In addition, miR-181a overexpression aggravated proliferation, metastasis and EMT by inhibiting SOCS4. Interestingly, inhibition of miR-181a diminished the promoting effects of ZNF674-AS1 silencing on the malignant behaviours of TC cells. These data illustrated that ZNF674-AS1 alleviated TC progression by modulating the miR-181a/SOCS4 axis (graphical abstract), further suggesting that ZNF674-AS1 might be used as a therapheutic target in TC treatment.
Collapse
Affiliation(s)
- Fei Le
- Department of head and neck Surgery, Jiangxi Cancer hospital, Nanchang, 330029, Jiangxi Province, PR China
| | - Hong-Mi Li
- Department of Comprehensive Radiation Oncology, Jiangxi Cancer hospital, Nanchang, 330029, Jiangxi Province, PR China
| | - Qiao-Li Lv
- Department of Science and Education, Jiangxi Cancer hospital, Nanchang, 330029, Jiangxi Province, PR China
| | - Jun-Jun Chen
- Department of Science and Education, Jiangxi Cancer hospital, Nanchang, 330029, Jiangxi Province, PR China
| | - Qian-Xia Lin
- Department of Science and Education, Jiangxi Cancer hospital, Nanchang, 330029, Jiangxi Province, PR China
| | - Yu-Long Ji
- Department of Science and Education, Jiangxi Cancer hospital, Nanchang, 330029, Jiangxi Province, PR China
| | - Bo Yi
- Department of Abdominal Surgery, Jiangxi Cancer hospital, Nanchang, 330029, Jiangxi Province, PR China.
| |
Collapse
|
1158
|
Wu L, Yu K, Chen K, Zhu X, Yang Z, Wang Q, Gao J, Wang Y, Cao T, Xu H, Pan X, Wang L, Xia J, Li Y, Wang ZP, Ma J. Fbxo45 facilitates pancreatic carcinoma progression by targeting USP49 for ubiquitination and degradation. Cell Death Dis 2022; 13:231. [PMID: 35279684 PMCID: PMC8918322 DOI: 10.1038/s41419-022-04675-2] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Revised: 02/12/2022] [Accepted: 02/18/2022] [Indexed: 12/27/2022]
Abstract
Fbxo45, a conserved F-box protein, comprises of an atypical SKP1, CUL1, F-box protein (SCF) ubiquitin ligase complex that promotes tumorigenesis and development. However, the biological function and molecular mechanisms of Fbxo45 involved in pancreatic carcinogenesis are ambiguous. We conducted several approaches, including transfection, coIP, real-time polymerase chain reaction (RT-PCR), Western blotting, ubiquitin assays, and animal studies, to explore the role of Fbxo45 in pancreatic cancer. Here, we report that USP49 stability is governed by Fbxo45-mediated ubiquitination and is enhanced by the absence of Fbxo45. Moreover, Fbxo45 binds to a short consensus sequence of USP49 through its SPRY domain. Furthermore, Fbxo45-mediated USP49 ubiquitination and degradation are enhanced by NEK6 kinase. Functionally, Fbxo45 increases cell viability and motility capacity by targeting USP49 in pancreatic cancer cells. Xenograft mouse experiments demonstrated that ectopic expression of Fbxo45 enhanced tumor growth in mice and that USP49 overexpression inhibited tumor growth in vivo. Notably, Fbxo45 expression was negatively associated with USP49 expression in pancreatic cancer tissues. Fbxo45 serves as an oncoprotein to facilitate pancreatic oncogenesis by regulating the stability of the tumor suppressor USP49 in pancreatic cancer.
Collapse
Affiliation(s)
- Linhui Wu
- Bengbu Medical College Key Laboratory of Cancer Research and Clinical Laboratory Diagnosis, Bengbu Medical College, Bengbu, Anhui, 233030, China
| | - Ke Yu
- Bengbu Medical College Key Laboratory of Cancer Research and Clinical Laboratory Diagnosis, Bengbu Medical College, Bengbu, Anhui, 233030, China
| | - Kai Chen
- Bengbu Medical College Key Laboratory of Cancer Research and Clinical Laboratory Diagnosis, Bengbu Medical College, Bengbu, Anhui, 233030, China
| | - Xuelian Zhu
- Department of Laboratory Medicine, School of Laboratory Medicine, Bengbu Medical College, Bengbu, Anhui, 233030, China
| | - Zheng Yang
- Department of Laboratory Medicine, School of Laboratory Medicine, Bengbu Medical College, Bengbu, Anhui, 233030, China
| | - Qi Wang
- Department of Pathology, Bengbu Medical College, Bengbu, Anhui, 233030, China
| | - Junjie Gao
- Bengbu Medical College Key Laboratory of Cancer Research and Clinical Laboratory Diagnosis, Bengbu Medical College, Bengbu, Anhui, 233030, China
| | - Yingying Wang
- Bengbu Medical College Key Laboratory of Cancer Research and Clinical Laboratory Diagnosis, Bengbu Medical College, Bengbu, Anhui, 233030, China
| | - Tong Cao
- Department of Clinical Laboratory, The First Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui, 233004, China
| | - Hui Xu
- Department of Clinical Laboratory Diagnostics, School of Laboratory Medicine, Bengbu Medical College, Bengbu, Anhui, 233030, China
| | - Xueshan Pan
- Department of Biochemistry and Molecular Biology, School of Laboratory Medicine, Bengbu Medical College, Bengbu, Anhui, 233030, China
| | - Lixia Wang
- Department of Urology, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, 215006, China
| | - Jun Xia
- Department of Biochemistry and Molecular Biology, School of Laboratory Medicine, Bengbu Medical College, Bengbu, Anhui, 233030, China
| | - Yuyun Li
- Department of Clinical Laboratory Diagnostics, School of Laboratory Medicine, Bengbu Medical College, Bengbu, Anhui, 233030, China
| | - Zhiwei Peter Wang
- Department of Biochemistry and Molecular Biology, School of Laboratory Medicine, Bengbu Medical College, Bengbu, Anhui, 233030, China.
- Center of Scientific Research, the Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325027, China.
| | - Jia Ma
- Department of Biochemistry and Molecular Biology, School of Laboratory Medicine, Bengbu Medical College, Bengbu, Anhui, 233030, China.
| |
Collapse
|
1159
|
Transcriptional and post-transcriptional control of epithelial-mesenchymal plasticity: why so many regulators? Cell Mol Life Sci 2022; 79:182. [PMID: 35278142 PMCID: PMC8918127 DOI: 10.1007/s00018-022-04199-0] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2021] [Revised: 01/18/2022] [Accepted: 02/07/2022] [Indexed: 12/12/2022]
Abstract
The dynamic transition between epithelial-like and mesenchymal-like cell states has been a focus for extensive investigation for decades, reflective of the importance of Epithelial-Mesenchymal Transition (EMT) through development, in the adult, and the contributing role EMT has to pathologies including metastasis and fibrosis. Not surprisingly, regulation of the complex genetic networks that underlie EMT have been attributed to multiple transcription factors and microRNAs. What is surprising, however, are the sheer number of different regulators (hundreds of transcription factors and microRNAs) for which critical roles have been described. This review seeks not to collate these studies, but to provide a perspective on the fundamental question of whether it is really feasible that so many regulators play important roles and if so, what does this tell us about EMT and more generally, the genetic machinery that controls complex biological processes.
Collapse
|
1160
|
Circulating tumour cells in the -omics era: how far are we from achieving the 'singularity'? Br J Cancer 2022; 127:173-184. [PMID: 35273384 PMCID: PMC9296521 DOI: 10.1038/s41416-022-01768-9] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Revised: 01/27/2022] [Accepted: 02/17/2022] [Indexed: 12/22/2022] Open
Abstract
Over the past decade, cancer diagnosis has expanded to include liquid biopsies in addition to tissue biopsies. Liquid biopsies can result in earlier and more accurate diagnosis and more effective monitoring of disease progression than tissue biopsies as samples can be collected frequently. Because of these advantages, liquid biopsies are now used extensively in clinical care. Liquid biopsy samples are analysed for circulating tumour cells (CTCs), cell-free DNA, RNA, proteins and exosomes. CTCs originate from the tumour, play crucial roles in metastasis and carry information on tumour heterogeneity. Multiple single-cell omics approaches allow the characterisation of the molecular makeup of CTCs. It has become evident that CTCs are robust biomarkers for predicting therapy response, clinical development of metastasis and disease progression. This review describes CTC biology, molecular heterogeneity within CTCs and the involvement of EMT in CTC dynamics. In addition, we describe the single-cell multi-omics technologies that have provided insights into the molecular features within therapy-resistant and metastasis-prone CTC populations. Functional studies coupled with integrated multi-omics analyses have the potential to identify therapies that can intervene the functions of CTCs.
Collapse
|
1161
|
Yanagihara K, Iino Y, Yokozaki H, Kubo T, Oda T, Kubo T, Komatsu M, Sasaki H, Ichikawa H, Kuwata T, Seyama T, Ochiai A. A Comparative Study of Patient-Derived Tumor Models of Pancreatic Ductal Adenocarcinoma Involving Orthotopic Implantation. Pathobiology 2022; 89:222-232. [PMID: 35272288 DOI: 10.1159/000521714] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Accepted: 12/23/2021] [Indexed: 02/05/2023] Open
Abstract
BACKGROUND Pancreatic ductal adenocarcinoma (PDA) is associated with very poor prognoses. Therefore, new therapies and preclinical models are urgently needed. In the present study, we sought to develop more realistic experimental models for use in PDA research. METHODS We developed patient-derived xenografts (PDXs), established PDX-derived cell lines (PDCLs), and generated cell line-derived xenografts (CDXs), which we integrated to create 13 matched "trios" - i.e., patient-derived tumor models of PDA. We then compared and contrasted histological and molecular alterations between these three model systems. RESULTS Orthotopic implantation (OI) of the PDCLs resulted in tumorigenesis and metastases to the liver and peritoneum. Morphological comparisons of OI-CDXs and OI-PDXs with passaged tumors revealed that the histopathological features of the original tumor were maintained in both models. Molecular alterations in PDX tumors (including those to KRAS, TP53, SMAD4, and CDKN2A) were similar to those in the respective PDCLs and CDX tumors. When gene expression levels in the PDCLs, ectopic tumors, and OI tumors were compared, the distant metastasis-promoting gene CXCR4 was specifically upregulated in OI tumors, whose immunohistochemical profiles suggested epithelial-mesenchymal transition and adeno-squamous trans-differentiation. CONCLUSION These patient-derived tumor models provide useful tools for monitoring responses to antineoplastic agents and for studying PDA biology.
Collapse
Affiliation(s)
- Kazuyoshi Yanagihara
- Division of Biomarker Discovery, Exploratory Oncology and Clinical Trial Center, National Cancer Center, Chiba, Japan
- Division of Pathology, Department of Pathology, Kobe University Graduate School of Medicine, Kobe, Japan
- Department of Life Sciences, Yasuda Women's University Faculty of Pharmacy, Hiroshima, Japan
| | - Yuki Iino
- Division of Biomarker Discovery, Exploratory Oncology and Clinical Trial Center, National Cancer Center, Chiba, Japan
| | - Hiroshi Yokozaki
- Division of Pathology, Department of Pathology, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Takanori Kubo
- Department of Life Sciences, Yasuda Women's University Faculty of Pharmacy, Hiroshima, Japan
| | - Tatsuya Oda
- Department of Gastrointestinal and Hepato-Biliary-Pancreatic Surgery, Faculty of Medicine, University of Tsukuba, Tsukuba, Japan
| | - Takashi Kubo
- Department of Pathology and Clinical Laboratories, National Cancer Center Hospital, Tokyo, Japan
- Department of Clinical Genomics, National Cancer Center Research Institute, Tokyo, Japan
| | - Masayuki Komatsu
- Department of Translational Oncology, Fundamental Innovative Oncology Core Center, National Cancer Center Research Institute, Tokyo, Japan
| | - Hiroki Sasaki
- Department of Translational Oncology, Fundamental Innovative Oncology Core Center, National Cancer Center Research Institute, Tokyo, Japan
| | - Hitoshi Ichikawa
- Department of Clinical Genomics, National Cancer Center Research Institute, Tokyo, Japan
| | - Takeshi Kuwata
- Department of Pathology and Clinical Laboratories, National Cancer Center Hospital East, Chiba, Japan
| | - Toshio Seyama
- Department of Life Sciences, Yasuda Women's University Faculty of Pharmacy, Hiroshima, Japan
| | - Atsushi Ochiai
- Division of Biomarker Discovery, Exploratory Oncology and Clinical Trial Center, National Cancer Center, Chiba, Japan
| |
Collapse
|
1162
|
Zhao R, Trainor PA. Epithelial to mesenchymal transition during mammalian neural crest cell delamination. Semin Cell Dev Biol 2022; 138:54-67. [PMID: 35277330 DOI: 10.1016/j.semcdb.2022.02.018] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Revised: 02/08/2022] [Accepted: 02/21/2022] [Indexed: 11/18/2022]
Abstract
Epithelial to mesenchymal transition (EMT) is a well-defined cellular process that was discovered in chicken embryos and described as "epithelial to mesenchymal transformation" [1]. During EMT, epithelial cells lose their epithelial features and acquire mesenchymal character with migratory potential. EMT has subsequently been shown to be essential for both developmental and pathological processes including embryo morphogenesis, wound healing, tissue fibrosis and cancer [2]. During the past 5 years, interest and study of EMT especially in cancer biology have increased exponentially due to the implied role of EMT in multiple aspects of malignancy such as cell invasion, survival, stemness, metastasis, therapeutic resistance and tumor heterogeneity [3]. Since the process of EMT in embryogenesis and cancer progression shares similar phenotypic changes, core transcription factors and molecular mechanisms, it has been proposed that the initiation and development of carcinoma could be attributed to abnormal activation of EMT factors usually required for normal embryo development. Therefore, developmental EMT mechanisms, whose timing, location, and tissue origin are strictly regulated, could prove useful for uncovering new insights into the phenotypic changes and corresponding gene regulatory control of EMT under pathological conditions. In this review, we initially provide an overview of the phenotypic and molecular mechanisms involved in EMT and discuss the newly emerging concept of epithelial to mesenchymal plasticity (EMP). Then we focus on our current knowledge of a classic developmental EMT event, neural crest cell (NCC) delamination, highlighting key differences in our understanding of NCC EMT between mammalian and non-mammalian species. Lastly, we highlight available tools and future directions to advance our understanding of mammalian NCC EMT.
Collapse
Affiliation(s)
- Ruonan Zhao
- Stowers Institute for Medical Research, Kansas City, MO, USA; Department of Anatomy and Cell Biology, University of Kansas Medical Center, Kansas City, KS, USA
| | - Paul A Trainor
- Stowers Institute for Medical Research, Kansas City, MO, USA; Department of Anatomy and Cell Biology, University of Kansas Medical Center, Kansas City, KS, USA.
| |
Collapse
|
1163
|
Ge H, Xu C, Chen H, Liu L, Zhang L, Wu C, Lu Y, Yao Q. Traditional Chinese Medicines as Effective Reversals of Epithelial-Mesenchymal Transition Induced-Metastasis of Colorectal Cancer: Molecular Targets and Mechanisms. Front Pharmacol 2022; 13:842295. [PMID: 35308223 PMCID: PMC8931761 DOI: 10.3389/fphar.2022.842295] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Accepted: 01/28/2022] [Indexed: 12/24/2022] Open
Abstract
Colorectal cancer (CRC) is the third most common type of cancer worldwide. Distant metastasis is the major cause of cancer-related mortality in patients with CRC. Epithelial-mesenchymal transition (EMT) is a critical process triggered during tumor metastasis, which is also the main impetus and the essential access within this duration. Therefore, targeting EMT-related molecular pathways has been considered a novel strategy to explore effective therapeutic agents against metastatic CRC. Traditional Chinese medicines (TCMs) with unique properties multi-target and multi-link that exert their therapeutic efficacies holistically, which could inhibit the invasion and metastasis ability of CRC cells via inhibiting the EMT process by down-regulating transforming growth factor-β (TGF-β)/Smads, PI3K/Akt, NF-κB, Wnt/β-catenin, and Notch signaling pathways. The objective of this review is to summarize and assess the anti-metastatic effect of TCM-originated bioactive compounds and Chinese medicine formulas by mediating EMT-associated signaling pathways in CRC therapy, providing a foundation for further research on the exact mechanisms of action through which TCMs affect EMT transform in CRC.
Collapse
Affiliation(s)
- Hongzhang Ge
- Department of Integrated Traditional Chinese and Western Medicine, The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, China
- Key Laboratory of Integration of Chinese and Western Medicine Oncology, Zhejiang Cancer Hospital, Hangzhou, China
- Key Laboratory of Head and Neck Cancer Translational Research of Zhejiang Province, Zhejiang Cancer Hospital, Hangzhou, China
| | - Chao Xu
- Department of Integrated Traditional Chinese and Western Medicine, The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, China
- Key Laboratory of Integration of Chinese and Western Medicine Oncology, Zhejiang Cancer Hospital, Hangzhou, China
- Key Laboratory of Head and Neck Cancer Translational Research of Zhejiang Province, Zhejiang Cancer Hospital, Hangzhou, China
| | - Haitao Chen
- Department of Integrated Traditional Chinese and Western Medicine, The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, China
- Key Laboratory of Integration of Chinese and Western Medicine Oncology, Zhejiang Cancer Hospital, Hangzhou, China
- Key Laboratory of Head and Neck Cancer Translational Research of Zhejiang Province, Zhejiang Cancer Hospital, Hangzhou, China
| | - Ling Liu
- Department of Integrated Traditional Chinese and Western Medicine, The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, China
- Key Laboratory of Integration of Chinese and Western Medicine Oncology, Zhejiang Cancer Hospital, Hangzhou, China
- Key Laboratory of Head and Neck Cancer Translational Research of Zhejiang Province, Zhejiang Cancer Hospital, Hangzhou, China
| | - Lei Zhang
- Department of Integrated Traditional Chinese and Western Medicine, The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, China
- Key Laboratory of Integration of Chinese and Western Medicine Oncology, Zhejiang Cancer Hospital, Hangzhou, China
- Key Laboratory of Head and Neck Cancer Translational Research of Zhejiang Province, Zhejiang Cancer Hospital, Hangzhou, China
| | - Changhong Wu
- Second Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, China
| | - Yi Lu
- Department of Clinical Nutrition, The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, China
| | - Qinghua Yao
- Department of Integrated Traditional Chinese and Western Medicine, The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, China
- Key Laboratory of Integration of Chinese and Western Medicine Oncology, Zhejiang Cancer Hospital, Hangzhou, China
- Key Laboratory of Head and Neck Cancer Translational Research of Zhejiang Province, Zhejiang Cancer Hospital, Hangzhou, China
- Second Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, China
- Department of Clinical Nutrition, The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, China
- *Correspondence: Qinghua Yao,
| |
Collapse
|
1164
|
Yang G, Xiog Y, Wang G, Li W, Tang T, Sun J, Li J. miR‑374c‑5p regulates PTTG1 and inhibits cell growth and metastasis in hepatocellular carcinoma by regulating epithelial‑mesenchymal transition. Mol Med Rep 2022; 25:148. [PMID: 35234260 PMCID: PMC8915393 DOI: 10.3892/mmr.2022.12664] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Accepted: 02/15/2022] [Indexed: 11/25/2022] Open
Abstract
A large number of studies have reported that microRNA (miR)-374c-5p plays an important role in the occurrence and development of malignant tumors, but there is no research on the role of miR-374c-5p in hepatocellular carcinoma (HCC). The aim of the present study was to investigate the role of miR-374c-5p in HCC and the underlying molecular mechanism. The expression of miR-374c-5p in HCC tissues and HCC cell lines was analyzed via reverse transcription-quantitative PCR. The association between miR-374c-5p and clinical pathology was also analyzed in patients with HCC. Kaplan-Meier analysis and Cox multivariate analysis were used to evaluate the prognostic significance of miR-374c-5p in HCC. The biological functions of miR-374c-5p, including cell proliferation, migration and invasion and its potential molecular mechanism were analyzed in vivo and in vitro. In addition, the molecular mechanism of miR-374c-5p in HCC was further explored. The results demonstrated that miR-374c-5p expression was lower in HCC than in matched adjacent tissue samples. Patients with low expression of miR-374c-5p had poor prognosis and short survival time. Overexpression of miR-374c-5p inhibited HCC cell proliferation, migration and invasion in vitro. In vivo, it was found that overexpression of miR-374c-5p significantly inhibited the growth and proliferation of HCC cells. Dual-luciferase reporter assays verified that miR-374c-5p directly targets the 3′-untranslated region of pituitary tumor-transforming 1 (PTTG1) and regulates PTTG1 expression. In general, it was revealed that miR-374c-5p regulates the malignant biological behavior of HCC through PTTG1, thereby affecting epithelial-mesenchymal transition. Thus, miR-374c-5p is a potential biological indicator to predict poor prognosis in patients with HCC.
Collapse
Affiliation(s)
- Gang Yang
- First Clinical Medical College, Jinan University, Tianhe, Guangzhou, Guangdong 510632, P.R. China
| | - Yongfu Xiog
- Department of Hepatobiliary Surgery, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan 637000, P.R. China
| | - Guan Wang
- Physical Examination Center, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan 637000, P.R. China
| | - Weinan Li
- Department of Hepatobiliary Surgery, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan 637000, P.R. China
| | - Tao Tang
- Department of Hepatobiliary Surgery, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan 637000, P.R. China
| | - Ji Sun
- Department of Hepatobiliary Surgery, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan 637000, P.R. China
| | - Jingdong Li
- First Clinical Medical College, Jinan University, Tianhe, Guangzhou, Guangdong 510632, P.R. China
| |
Collapse
|
1165
|
Design, synthesis and biological evaluation of antitumor platinum(II) agents conjugated with non-steroidal anti-inflammatory drug species. Bioorg Chem 2022; 120:105633. [DOI: 10.1016/j.bioorg.2022.105633] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2021] [Revised: 01/06/2022] [Accepted: 01/16/2022] [Indexed: 12/12/2022]
|
1166
|
Liang W, Chen S, Yang G, Feng J, Ling Q, Wu B, Yan H, Cheng J. Overexpression of zinc-finger protein 677 inhibits proliferation and invasion by and induces apoptosis in clear cell renal cell carcinoma. Bioengineered 2022; 13:5292-5304. [PMID: 35164660 PMCID: PMC8973725 DOI: 10.1080/21655979.2022.2038891] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Revised: 01/29/2022] [Accepted: 02/02/2022] [Indexed: 11/28/2022] Open
Abstract
Recent studies have demonstrated that zinc-finger protein 677 (ZNF677) acts as a tumor suppressor gene in cancer. However, the expression and function of ZNF677 in clear cell renal cell carcinoma (ccRCC) are still unclear. In this study, we used bioinformatics analysis and in vitro experiments to investigate the expression of ZNF677 in ccRCC tissues and the malignant biological behavior of ZNF677 in 786-0 cells. We demonstrated that ZNF677 is hypermethylated in ccRCC and is associated with clinicopathological features. The results of the functional assays indicate that ZNF677 inhibits tumor cell proliferation and invasion and induces apoptosis. Further prognostic analysis indicated that low expression of ZNF677 is associated with shorter overall survival. Additionally, ZNF677 overexpression suppressed the invasion and epithelial-mesenchymal transition of 786-0 cells by inactivating the PI3K/AKT signaling pathway. This is the first report to evaluate the influence of ZNF677 on ccRCC cells malignant biological behavior. The results indicate that high expression of ZNF677 could be considered as a favorable prognostic indicator for ccRCC.
Collapse
Affiliation(s)
- W Liang
- Department of Urology, The First Affiliated Hospital of Guangxi Medical University, Nanning, People’s Republic of China
| | - Sh Chen
- Department of Urology, The First Affiliated Hospital of Guangxi Medical University, Nanning, People’s Republic of China
| | - Gl Yang
- Department of Urology, The First Affiliated Hospital of Guangxi Medical University, Nanning, People’s Republic of China
| | - Jy Feng
- Department of Urology, The First Affiliated Hospital of Guangxi Medical University, Nanning, People’s Republic of China
| | - Q Ling
- Department of Urology, The First Affiliated Hospital of Guangxi Medical University, Nanning, People’s Republic of China
| | - B Wu
- Department of Urology, The First Affiliated Hospital of Guangxi Medical University, Nanning, People’s Republic of China
| | - Hb Yan
- Department of Urology, The First Affiliated Hospital of Guangxi Medical University, Nanning, People’s Republic of China
| | - Jw Cheng
- Department of Urology, The First Affiliated Hospital of Guangxi Medical University, Nanning, People’s Republic of China
| |
Collapse
|
1167
|
Bai L, Yang ZX, Ma PF, Liu JS, Wang DS, Yu HC. Overexpression of SLC25A51 promotes hepatocellular carcinoma progression by driving aerobic glycolysis through activation of SIRT5. Free Radic Biol Med 2022; 182:11-22. [PMID: 35182732 DOI: 10.1016/j.freeradbiomed.2022.02.014] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/04/2021] [Revised: 02/13/2022] [Accepted: 02/14/2022] [Indexed: 11/21/2022]
Abstract
Solute carrier family 25 member 20 (SLC25A51) is a newly identified mammalian mitochondrial NAD+ transporter. However, the clinicopathological and biological significance of SLC25A51 in human cancers, including hepatocellular carcinoma (HCC), remains unclear. The aim of this study was to define the role of SLC25A51 in HCC progression. Here we demonstrate that SLC25A51 is significantly overexpressed in human HCC specimens and cell lines, caused by, at least in partial, the decrease of miR-212-3p. SLC25A51 overexpression is positively correlated with the clinicopathological characteristics of vascular invasion and tumor diameter, as well as poor survival in patients with HCC. Knockdown of SLC25A51 attenuated, while overexpression of SLC25A51 enhanced the growth and metastasis of HCC cells both in vitro and in vivo. Mechanistically, glucose metabolism reprogramming from oxidative phosphorylation to glycolysis by activation of mitochondrial sirtuin 5 (SIRT5) was found to contribute to the promotion of growth and metastasis by SLC25A51 in HCC cells. Together, these findings reveal important roles of SLC25A51 in HCC tumorigenesis and suggest SLC25A51 as a promising prognostic marker and therapeutic target for treating HCC.
Collapse
Affiliation(s)
- Lu Bai
- Department of Clinical Laboratory, Xijing Hospital, Air Force Medical University, Xi'an, China
| | - Zhao-Xu Yang
- Department of Hepatobiliary Surgery, Xijing Hospital, Air Force Medical University, Xi'an, China
| | - Peng-Fei Ma
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Jian-Shan Liu
- Department of Hepatobiliary Surgery, Xijing Hospital, Air Force Medical University, Xi'an, China
| | - De-Sheng Wang
- Department of Hepatobiliary Surgery, Xijing Hospital, Air Force Medical University, Xi'an, China.
| | - Heng-Chao Yu
- Department of Hepatobiliary Surgery, Xijing Hospital, Air Force Medical University, Xi'an, China.
| |
Collapse
|
1168
|
Shah PP, Saurabh K, Kurlawala Z, Vega AA, Siskind LJ, Beverly LJ. Towards a molecular understanding of the overlapping and distinct roles of UBQLN1 and UBQLN2 in lung cancer progression and metastasis. Neoplasia 2022; 25:1-8. [PMID: 35063704 PMCID: PMC8864381 DOI: 10.1016/j.neo.2021.11.010] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2021] [Revised: 10/29/2021] [Accepted: 11/17/2021] [Indexed: 11/30/2022]
Abstract
The Ubiquilin family of proteins (UBQLN) consists of five related proteins (UBQLN1-4 and UBQLNL). Herein, we showed that loss of UBQLN1 and/or UBQLN2 induces cellular processes involved in tumor progression and metastasis, including proliferation, clonogenic potential and migration in lung adenocarcinoma cells. Molecular, biochemical and RNAseq analyses in multiple cellular systems, identified overlapping and distinct gene sets and pathways that were altered following loss of UBQLN1 and/or UBQLN2. The present study, provide evidence that UBQLN1 and UBQLN2 perform similar, but distinct molecular functions in a variety of cell types.
The Ubiquilin family of proteins (UBQLN) consists of five related proteins (UBQLN1-4 and UBQLNL) that all contain ubiquitin-like (UBL) and ubiquitin-associated (UBA) domains. UBQLN1 and UBQLN2 are the most closely related and have been the most well-studied, however their biochemical, biological and cellular functions are still not well understood. Previous studies from our lab reported that loss of UBQLN1 or UBQLN2 induces epithelial mesenchymal transition (EMT) in lung adenocarcinoma cells. Herein, we showed that loss of UBQLN1 and/or UBQLN2 induces cellular processes involved in tumor progression and metastasis, including proliferation, clonogenic potential and migration in lung adenocarcinoma cells. In fact, following simultaneous loss of both UBQLN1 and UBQLN2 many of these processes were further enhanced. To understand the molecular mechanisms by which UBQLN1 and UBQLN2 loss could be additive, we performed molecular, biochemical and RNAseq analyses in multiple cellular systems. We identified overlapping and distinct gene sets and pathways that were altered following loss of UBQLN1 and/or UBQLN2. We have also begun to define cell type specific gene regulation of UBQLN1 and UBQLN2, as well as understand how loss of either gene can alter differentiation of normal cells. The data presented here demonstrate that UBQLN1 and UBQLN2 perform similar, but distinct molecular functions in a variety of cell types.
Collapse
Affiliation(s)
- Parag P Shah
- James Graham Brown Cancer Center, University of Louisville, 505 S. Hancock Street, CTRB rm 204, Louisville, KY 40202, USA
| | - Kumar Saurabh
- James Graham Brown Cancer Center, University of Louisville, 505 S. Hancock Street, CTRB rm 204, Louisville, KY 40202, USA
| | - Zimple Kurlawala
- James Graham Brown Cancer Center, University of Louisville, 505 S. Hancock Street, CTRB rm 204, Louisville, KY 40202, USA
| | - Alexis A Vega
- Department of Biochemistry and Molecular Genetics, University of Louisville, Louisville, KY 40202, USA
| | - Leah J Siskind
- James Graham Brown Cancer Center, University of Louisville, 505 S. Hancock Street, CTRB rm 204, Louisville, KY 40202, USA; Department of Pharmacology and Toxicology, University of Louisville School of Medicine, Louisville, KY 40202, USA
| | - Levi J Beverly
- James Graham Brown Cancer Center, University of Louisville, 505 S. Hancock Street, CTRB rm 204, Louisville, KY 40202, USA; Department of Biochemistry and Molecular Genetics, University of Louisville, Louisville, KY 40202, USA; Department of Medicine, Division of Hematology and Oncology, University of Louisville School of Medicine, Louisville, KY 40202, USA; Department of Pharmacology and Toxicology, University of Louisville School of Medicine, Louisville, KY 40202, USA.
| |
Collapse
|
1169
|
Juárez-Cruz JC, Okoniewski M, Ramírez M, Ortuño-Pineda C, Navarro-Tito N, Castañeda-Saucedo E. Chronic Leptin Treatment Induces Epithelial-Mesenchymal Transition in MCF10A Mammary Epithelial Cells. J Mammary Gland Biol Neoplasia 2022; 27:19-36. [PMID: 35195812 DOI: 10.1007/s10911-022-09515-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/04/2021] [Accepted: 02/08/2022] [Indexed: 01/06/2023] Open
Abstract
Leptin is a cytokine-like hormone that functions as a link between obesity and breast cancer (BC). Leptin treatment induces Epithelial to Mesenchymal Transition (EMT) in BC cell lines. In non-tumoral breast epithelial MCF10A cells, acute leptin treatment induces partial EMT. However, the effect of chronic leptin treatment on EMT in non-tumorigenic breast cells has not been fully explored. This study aimed to evaluate the effect of chronic leptin treatment on the induction of EMT in MCF10A cells. We found that chronic leptin treatment induces a switch from an epithelial to a mesenchymal morphology, partial loss of E-cadherin and gain of vimentin expression. Immunolocalization experiments showed a partial loss of E-cadherin at cell junctions and increased cytoplasmic localization of vimentin in leptin-treated cells. Moreover, chronic leptin treatment increased collective cell migration and invasion. Furthermore, when cultured in non-adherent conditions leptin treated cells exhibited reduced cell aggregation, increased survival, and decreased apoptosis, which correlates with increased FAK and AKT phosphorylation. Finally, bioinformatic analysis in two publicly available RNAseq datasets from normal breast tissue shows that high levels of leptin mRNA correlate positively with the expression of mesenchymal markers, and negatively with epithelial markers. Thus, our results demonstrate that chronic leptin treatment induces EMT in non-tumorigenic MCF10A cells and suggest that high leptin expression in normal breast tissue may induce EMT and contribute to increased risk of breast cancer.
Collapse
Affiliation(s)
- Juan Carlos Juárez-Cruz
- Laboratorio de Biología Celular del Cáncer. Facultad de Ciencias Químico-Biológicas, Universidad Autónoma de Guerrero. Av. Lázaro Cárdenas S/N Ciudad Universitaria. C.P, 39087, Chilpancingo de los Bravo, Guerrero, México
| | | | - Mónica Ramírez
- CONACYT, Universidad Autónoma de Guerrero, Chilpancingo de los Bravo, Guerrero, México
| | - Carlos Ortuño-Pineda
- Laboratorio de Ácidos Nucleicos y Proteínas. Facultad de Ciencias Químico-Biológicas, Universidad Autónoma de Guerrero, Chilpancingo de los Bravo, Guerrero, México
| | - Napoleón Navarro-Tito
- Laboratorio de Biología Celular del Cáncer. Facultad de Ciencias Químico-Biológicas, Universidad Autónoma de Guerrero. Av. Lázaro Cárdenas S/N Ciudad Universitaria. C.P, 39087, Chilpancingo de los Bravo, Guerrero, México
| | - Eduardo Castañeda-Saucedo
- Laboratorio de Biología Celular del Cáncer. Facultad de Ciencias Químico-Biológicas, Universidad Autónoma de Guerrero. Av. Lázaro Cárdenas S/N Ciudad Universitaria. C.P, 39087, Chilpancingo de los Bravo, Guerrero, México.
| |
Collapse
|
1170
|
Ye G, Huang M, Li Y, Ouyang J, Chen M, Wen Q, Li X, Zeng H, Long P, Fan Z, Yin J, Ye W, Zhang D. The FAP α -activated prodrug Z-GP-DAVLBH inhibits the growth and pulmonary metastasis of osteosarcoma cells by suppressing the AXL pathway. Acta Pharm Sin B 2022; 12:1288-1304. [PMID: 35530139 PMCID: PMC9072247 DOI: 10.1016/j.apsb.2021.08.015] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Revised: 07/27/2021] [Accepted: 07/30/2021] [Indexed: 01/02/2023] Open
Abstract
Osteosarcoma is a kind of bone tumor with highly proliferative and invasive properties, a high incidence of pulmonary metastasis and a poor prognosis. Chemotherapy is the mainstay of treatment for osteosarcoma. Currently, there are no molecular targeted drugs approved for osteosarcoma treatment, particularly effective drugs for osteosarcoma with pulmonary metastases. It has been reported that fibroblast activation protein alpha (FAPα) is upregulated in osteosarcoma and critically associated with osteosarcoma progression and metastasis, demonstrating that FAPα-targeted agents might be a promising therapeutic strategy for osteosarcoma. In the present study, we reported that the FAPα-activated vinblastine prodrug Z-GP-DAVLBH exhibited potent antitumor activities against FAPα-positive osteosarcoma cells in vitro and in vivo. Z-GP-DAVLBH inhibited the growth and induced the apoptosis of osteosarcoma cells. Importantly, it also decreased the migration and invasion capacities and reversed epithelial–mesenchymal transition (EMT) of osteosarcoma cells in vitro and suppressed pulmonary metastasis of osteosarcoma xenografts in vivo. Mechanistically, Z-GP-DAVLBH suppressed the AXL/AKT/GSK-3β/β-catenin pathway, leading to inhibition of the growth and metastatic spread of osteosarcoma cells. These findings demonstrate that Z-GP-DAVLBH is a promising agent for the treatment of FAPα-positive osteosarcoma, particularly osteosarcoma with pulmonary metastases.
Collapse
|
1171
|
Hirway SU, Weinberg SH. A review of computational modeling, machine learning and image analysis in cancer metastasis dynamics. COMPUTATIONAL AND SYSTEMS ONCOLOGY 2022. [DOI: 10.1002/cso2.1044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Affiliation(s)
- Shreyas U. Hirway
- Department of Biomedical Engineering The Ohio State University Columbus Ohio USA
| | - Seth H. Weinberg
- Department of Biomedical Engineering The Ohio State University Columbus Ohio USA
| |
Collapse
|
1172
|
Sun Q, Zhang T, Xiao Q, Mei B, Zhang X. Procyanidin B2 inhibits angiogenesis and cell growth in oral squamous cell carcinoma cells through the vascular endothelial growth factor (VEGF)/VEGF receptor 2 (VEGFR2) pathway. Bioengineered 2022; 13:6500-6508. [PMID: 35220896 PMCID: PMC8973926 DOI: 10.1080/21655979.2022.2033013] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
This study aimed to explore the therapy role of procyanidin B2 (PB2) in inhibiting angiogenesis and cell growth in oral squamous cell carcinoma. After oral mucosa epithelial cell (OMEC) and human oral squamous cell carcinoma (OSCC) cell line (SCC-25) were treated with PB2 or SCC-25 were treated with PB2 and rhVEGF, cell counting kit-8 (CCK-8) assay was used to determine the cell viability. The apoptosis, migration, invasion and angiogenesis of SCC-25 after indicated treatment were detected by Tunel, wound healing, transwell and tube formation assays. The protein expression related to apoptosis, metastasis and epithelial-mesenchymal transition (EMT) and changed expression of vascular endothelial growth factor (VEGF)/VEGF receptor 2 (VEGFR2) signaling was analyzed by Western blot. As a result, PB2 inhibited viability, invasion, migration and EMT and promoted apoptosis of SCC-25 cells. In addition, PB2 inhibited VEGF/VEGFR2 signaling and tumor itangiogenesis in OSCC. As expected, activation of VEGF/VEGFR2 signaling suppressed the effect of PB2 on growth and metastasis of OSCC cells. In conclusion, PB2 inhibited the VEGF/VEGFR2 pathway to suppress the angiogenesis and cell growth of SCC-25 cells.
Collapse
Affiliation(s)
- Qiurong Sun
- Department of Stomatology, The First Affiliated Hosital of Gannan Medical University, Ganzhou, Jiangxi, China
| | - Taiyang Zhang
- Department of Stomatology, The First Affiliated Hosital of Gannan Medical University, Ganzhou, Jiangxi, China
| | - Qingchun Xiao
- Department of Stomatology, The First Affiliated Hosital of Gannan Medical University, Ganzhou, Jiangxi, China
| | - Bingxin Mei
- Department of Stomatology, The First Affiliated Hosital of Gannan Medical University, Ganzhou, Jiangxi, China
| | - Xingwang Zhang
- Department of Oral Surgery, Daqing Longnan Hospital, Daqing, Heilongjiang, China
| |
Collapse
|
1173
|
Liu J, Cui G, Shen S, Gao F, Zhu H, Xu Y. Establishing a Prognostic Signature Based on Epithelial-Mesenchymal Transition-Related Genes for Endometrial Cancer Patients. Front Immunol 2022; 12:805883. [PMID: 35095892 PMCID: PMC8795518 DOI: 10.3389/fimmu.2021.805883] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2021] [Accepted: 12/21/2021] [Indexed: 12/24/2022] Open
Abstract
Backgrounds Epithelial-mesenchymal transition (EMT) is a sequential process where tumor cells develop from the epithelial state to the mesenchymal state. EMT contributes to various tumor functions including initiation, propagating potential, and resistance to therapy, thus affecting the survival time of patients. The aim of this research is to set up an EMT-related prognostic signature for endometrial cancer (EC). Methods EMT-related gene (ERG) expression and clinical data were acquired from The Cancer Genome Atlas (TCGA). The entire set was randomly divided into two sets, one for contributing the risk model (risk score) and the other for validating. Univariate and multivariate Cox proportional hazards regression analyses were applied to the training set to select the prognostic ERGs. The expression of 10 ERGs was confirmed by qRT-PCR in clinical samples. Then, we developed a nomogram predicting 1-/3-/5-year survival possibility combining the risk score and clinical factors. The entire set was stratified into the high- and low-risk groups, which was used to analyze the immune infiltrating, tumorigenesis pathways, and response to drugs. Results A total of 220 genes were screened out from 1,316 ERGs for their differential expression in tumor versus normal. Next, 10 genes were found to be associated with overall survival (OS) in EC, and the expression was validated by qRT-PCR using clinical samples, so we constructed a 10-ERG-based risk score to distinguish high-/low-risk patients and a nomogram to predict survival rate. The calibration plots proved the predictive value of our model. Gene Set Enrichment Analysis (GSEA) discovered that in the low-risk group, immune-related pathways were enriched; in the high-risk group, tumorigenesis pathways were enriched. The low-risk group showed more immune activities, higher tumor mutational burden (TMB), and higher CTAL4/PD1 expression, which was in line with a better response to immune checkpoint inhibitors. Nevertheless, response to chemotherapeutic drugs turned out better in the high-risk group. The high-risk group had higher N 6-methyladenosine (m6A) RNA expression, microsatellite instability level, and stemness indices. Conclusion We constructed the ERG-related signature model to predict the prognosis of EC patients. What is more, it might offer a reference for predicting individualized response to immune checkpoint inhibitors and chemotherapeutic drugs.
Collapse
Affiliation(s)
- Jinhui Liu
- Department of Gynecology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Guoliang Cui
- Department of Gastroenterology, The Second Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
| | - Shuning Shen
- Department of Gynecology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Feng Gao
- Department of Orthopedic Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Hongjun Zhu
- Department of Oncology, Nantong Third People's Hospital Affiliated to Nantong University, Nantong, China
| | - Yinghua Xu
- Department of Radiation Oncology, Nantong Third People's Hospital Affiliated to Nantong University, Nantong, China
| |
Collapse
|
1174
|
Wu Z, Lu Z, Li L, Ma M, Long F, Wu R, Huang L, Chou J, Yang K, Zhang Y, Li X, Hu G, Zhang Y, Lin C. Identification and Validation of Ferroptosis-Related LncRNA Signatures as a Novel Prognostic Model for Colon Cancer. Front Immunol 2022; 12:783362. [PMID: 35154072 PMCID: PMC8826443 DOI: 10.3389/fimmu.2021.783362] [Citation(s) in RCA: 82] [Impact Index Per Article: 27.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2021] [Accepted: 12/28/2021] [Indexed: 12/13/2022] Open
Abstract
Background Ferroptosis is a newly defined form of programmed cell death that plays an important role in many cancers. However, ferroptosis-related lncRNAs (FRLs) involved in the regulation of colon cancer are not thoroughly understood. This study aimed to identify a prognostic FRL signature in colon cancer and explore its potential molecular function. Methods RNA-seq data and relevant clinical information were obtained from The Cancer Genome Atlas (TCGA) database, and a list of ferroptosis-related genes was extracted from the FerrDb website. Analysis of differentially expressed FRLs was performed using the 'limma' package in R software. By implementing coexpression analysis and univariate Cox analysis, we then identified prognostic FRLs. Using Cox regression analysis with the least absolute shrinkage and selection operator (LASSO) algorithm, we constructed a prognostic model based on 4 FRLs. We evaluated the prognostic power of this model using Kaplan-Meier (K-M) survival curve analysis and receiver operating characteristic (ROC) curve analysis. Moreover, the relationships between the signature and immune landscape, somatic mutation and drug sensitivity were explored. Finally, in vitro experiments were conducted to validate the functions of AP003555.1 and AC000584.1. Results A 4-FRL signature was constructed. Two risk groups were classified based on the risk score calculated by this signature. The signature-based risk score exhibited a more powerful capacity for survival prediction than traditional clinicopathological features in colon patients. Additionally, we observed a significant difference in immune cells, such as CD4+ and CD8+ T cells and macrophages, between the two groups. Moreover, the high-risk group exhibited lower IC50 values for certain chemotherapy drugs, such as cisplatin, docetaxel, bleomycin or axitinib. Finally, the in vitro experiments showed that ferroptosis processes were suppressed after AP003555.1 and AC000584.1 knockdown. Conclusion The proposed 4-FRL signature is a promising biomarker to predict clinical outcomes and therapeutic responses in colon cancer patients.
Collapse
Affiliation(s)
- Zhiwei Wu
- Department of Gastrointestinal Surgery, The Third XiangYa Hospital of Central South University, Changsha, China
| | - Zhixing Lu
- Department of Gastrointestinal Surgery, The Third XiangYa Hospital of Central South University, Changsha, China
| | - Liang Li
- Department of Gastrointestinal Surgery, The Third XiangYa Hospital of Central South University, Changsha, China
| | - Min Ma
- Department of Gastrointestinal Surgery, The Third XiangYa Hospital of Central South University, Changsha, China
| | - Fei Long
- Department of Gastrointestinal Surgery, The Third XiangYa Hospital of Central South University, Changsha, China
| | - Runliu Wu
- Department of Gastrointestinal Surgery, The Third XiangYa Hospital of Central South University, Changsha, China
| | - Lihua Huang
- School of Life Sciences, Central South University, Changsha, China
| | - Jing Chou
- Department of Gastrointestinal Surgery, The Third XiangYa Hospital of Central South University, Changsha, China
| | - Kaiyan Yang
- Department of Gastrointestinal Surgery, The Third XiangYa Hospital of Central South University, Changsha, China
| | - Yi Zhang
- Department of General Surgery, Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Xiaorong Li
- Department of Gastrointestinal Surgery, The Third XiangYa Hospital of Central South University, Changsha, China
| | - Gui Hu
- Department of Gastrointestinal Surgery, The Third XiangYa Hospital of Central South University, Changsha, China
| | - Yi Zhang
- Department of Gastrointestinal Surgery, The Third XiangYa Hospital of Central South University, Changsha, China
| | - Changwei Lin
- Department of Gastrointestinal Surgery, The Third XiangYa Hospital of Central South University, Changsha, China
| |
Collapse
|
1175
|
Wang C, Che J, Jiang Y, Chen P, Bao G, Li C. CDT1 facilitates metastasis in prostate cancer and correlates with cell cycle regulation. Cancer Biomark 2022; 34:459-469. [PMID: 35253732 DOI: 10.3233/cbm-210389] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
BACKGROUND: CDT1 is the essential regulator of the initiation of DNA replication. Overexpressed CDT1 can cause DNA damage through re-replication. However, the function of CDT1 in prostate cancer (PCa) development has not been established. METHODS: Through bioinformatics, expression levels of CDT1 were found to be higher in metastatic PCa when compared to primary PCa. Then, immunohistochemical staining confirmed that the expression of CDT1 was significantly correlated with the occurrence of distant metastasis. For PCa cells, we established a stable clones knockdown CDT1. MTT was used in analyzing the proliferation ability of cells. Migration as well as invasion assays were performed. Effects of CDT1 knockdown on the cell cycle were evaluated by flow cytometry. Expression levels of EMT-associated markers in PCa cells were determined by Western blotting. And PI3K/AKT/GSK3β, a signaling molecule recognized in PCa that can regulate EMT, was detected in protein level. RESULTS: Over expression of CDT1 in PCa cells enhanced cell migration, invasion, tumor metastasis and was correlated with cell cycle regulation. Our results showed that knockdown of CDT1 inhibited G1 to S phase transition and induced the G1 phase cell cycle arrest in PCa cells. Moreover, it upregulated the expressions of epithelial markers (E-cadherin) and down-regulated mesenchymal markers (including Slug, N-cadherin, MMP2, vimentin, Snail, and MMP9) via regulating the phosphorylation level of PI3K, AKT and GSK3β. CONCLUSIONS: CDT1 promotes PCa cell metastasis by promoting cell cycle and PI3K/AKT/GSK3β mediated epithelial-mesenchymal transition (EMT) progression and may be a therapeutic target for metastatic PCa.
Collapse
Affiliation(s)
- Chunhui Wang
- Departments of Urology, Affiliated Hospital of Chifeng University, Chifeng, Inner Mongolia, China
- Urology Research Center, Chifeng University, Chifeng, Inner Mongolia, China
- Departments of Urology, Affiliated Hospital of Chifeng University, Chifeng, Inner Mongolia, China
| | - Jizhong Che
- Departments of Urology, Yan Tai Affiliated Hospital of Bin Zhou Medical University, Binzhou, Shandong, China
- Departments of Urology, Affiliated Hospital of Chifeng University, Chifeng, Inner Mongolia, China
| | - Ying Jiang
- Medical Reproductive Center, Affiliated Hospital of Chifeng University, Chifeng, Inner Mongolia, China
- Departments of Urology, Affiliated Hospital of Chifeng University, Chifeng, Inner Mongolia, China
| | - Ping Chen
- Physical Examination Center, Affiliated Hospital of Chifeng University, Chifeng, Inner Mongolia, China
| | - Guochang Bao
- Departments of Urology, Affiliated Hospital of Chifeng University, Chifeng, Inner Mongolia, China
- Urology Research Center, Chifeng University, Chifeng, Inner Mongolia, China
| | - Chunsheng Li
- Departments of Urology, Affiliated Hospital of Chifeng University, Chifeng, Inner Mongolia, China
- Urology Research Center, Chifeng University, Chifeng, Inner Mongolia, China
| |
Collapse
|
1176
|
Song D, Li S, Ning L, Zhang S, Cai Y. Smurf2 suppresses the metastasis of hepatocellular carcinoma via ubiquitin degradation of Smad2. Open Med (Wars) 2022; 17:384-396. [PMID: 35509688 PMCID: PMC8874264 DOI: 10.1515/med-2022-0437] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2021] [Revised: 12/27/2021] [Accepted: 01/17/2022] [Indexed: 11/15/2022] Open
Abstract
Purpose Smurf2, one of C2-WW-HECT domain E3 ubiquitin ligases, is closely related to the development and progression in different cancer types, including hepatocellular carcinoma (HCC). This study aims to illustrate the expression and molecular mechanism of Smurf2 in regulating the progression of HCC. Methods The expression of Smurf2 in human HCC and adjacent non-tumor liver specimens was detected using tissue microarray studies from 220 HCC patients who underwent curative resection. The relationships of Smurf2 and HCC progression and survival were analyzed using the chi-square test, Kaplan–Meier analysis, and Cox proportional hazards model. For Smurf2 was low expression in HCC cell lines, Smurf2 overexpression cell lines were established. The effect of Smurf2 on cell proliferation and migration was detected by Cell Counting Kit-8 and colony formation assay, and the epithelial–mesenchymal transition (EMT) markers and its transcription factors were tested by immunoblotting. The interaction and ubiquitination of Smad2 by Smurf2 were detected by co-immunoprecipitation and immunoprecipitation assay. Finally, the effect of Smurf2 on HCC was verified using the mouse lung metastasis model. Results Smurf2 was downregulated in HCC tissues compared to that of corresponding non-tumor liver specimens. The low expression of Smurf2 in HCC was significantly associated with macrovascular or microvascular tumor thrombus and the impairment of overall survival and disease-free survival. In vitro and in vivo analysis showed that Smurf2 overexpression decreased the EMT potential of HCC cells by promoting the ubiquitination of Smad2 via the proteasome-dependent degradation pathway. Conclusion The expression of Smurf2 was downregulated in HCC specimens and affected the survival of patients. Smurf2 inhibited the EMT of HCC by enhancing Smad2 ubiquitin-dependent proteasome degradation.
Collapse
Affiliation(s)
- Dongqiang Song
- Liver Cancer Institute, Zhongshan Hospital of Fudan University , Xuhui District , Shanghai , P. R. China
| | - Shuyu Li
- Department of Gastroenterology and Hepatology, Zhongshan Hospital of Fudan University , Xuhui District , Shanghai , P. R. China
| | - Liuxin Ning
- Department of Gastroenterology and Hepatology, Zhongshan Hospital of Fudan University , Xuhui District , Shanghai , P. R. China
| | - Shuncai Zhang
- Department of Gastroenterology and Hepatology, Zhongshan Hospital of Fudan University , Xuhui District , Shanghai , P. R. China
| | - Yu Cai
- Department of Gastroenterology and Hepatology, Zhongshan Hospital of Fudan University , Xuhui District , Shanghai , P. R. China
| |
Collapse
|
1177
|
Brown MS, Muller KE, Pattabiraman DR. Quantifying the Epithelial-to-Mesenchymal Transition (EMT) from Bench to Bedside. Cancers (Basel) 2022; 14:1138. [PMID: 35267444 PMCID: PMC8909103 DOI: 10.3390/cancers14051138] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Revised: 02/04/2022] [Accepted: 02/17/2022] [Indexed: 02/07/2023] Open
Abstract
The epithelial-to-mesenchymal transition (EMT) and its reversal, the mesenchymal-to-epithelial transition (MET) are critical components of the metastatic cascade in breast cancer and many other solid tumor types. Recent work has uncovered the presence of a variety of states encompassed within the EMT spectrum, each of which may play unique roles or work collectively to impact tumor progression. However, defining EMT status is not routinely carried out to determine patient prognosis or dictate therapeutic decision-making in the clinic. Identifying and quantifying the presence of various EMT states within a tumor is a critical first step to scoring patient tumors to aid in determining prognosis. Here, we review the major strides taken towards translating our understanding of EMT biology from bench to bedside. We review previously used approaches including basic immunofluorescence staining, flow cytometry, single-cell sequencing, and multiplexed tumor mapping. Future studies will benefit from the consideration of multiple methods and combinations of markers in designing a diagnostic tool for detecting and measuring EMT in patient tumors.
Collapse
Affiliation(s)
- Meredith S. Brown
- Department of Molecular and Systems Biology, Geisel School of Medicine at Dartmouth, Hanover, NH 03755, USA;
| | - Kristen E. Muller
- Department of Pathology, Dartmouth-Hitchcock Medical Center, Lebanon, NH 03756, USA;
| | - Diwakar R. Pattabiraman
- Department of Molecular and Systems Biology, Geisel School of Medicine at Dartmouth, Hanover, NH 03755, USA;
- Norris Cotton Cancer Center, Dartmouth-Hitchcock Medical Center, Lebanon, NH 03756, USA
| |
Collapse
|
1178
|
Jin Y, Kang Y, Wang M, Wu B, Su B, Yin H, Tang Y, Li Q, Wei W, Mei Q, Hu G, Lukacs-Kornek V, Li J, Wu K, Yuan X, Wang W. Targeting polarized phenotype of microglia via IL6/JAK2/STAT3 signaling to reduce NSCLC brain metastasis. Signal Transduct Target Ther 2022; 7:52. [PMID: 35194016 PMCID: PMC8864012 DOI: 10.1038/s41392-022-00872-9] [Citation(s) in RCA: 89] [Impact Index Per Article: 29.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2021] [Revised: 12/29/2021] [Accepted: 12/31/2021] [Indexed: 12/27/2022] Open
Abstract
Tumor-associated macrophages have emerged as crucial factors for metastases. Microglia are indispensable components of the brain microenvironment and play vital roles in brain metastasis (BM). However, the underlying mechanism of how activated microglia promote brain metastasis of non-small cell lung cancer (NSCLC) remains elusive. Here, we purified cell lines with brain-metastatic tropism and employed a co-culture system to reveal their communication with microglia. By single-cell RNA-sequencing and transcriptome difference analysis, we identified IL6 as the key regulator in brain-metastatic cells (A549-F3) to induce anti-inflammatory microglia via JAK2/STAT3 signaling, which in turn promoted the colonization process in metastatic A549-F3 cells. In our clinical samples, patients with higher levels of IL6 in serum showed higher propensity for brain metastasis. Additionally, the TCGA (The Cancer Genome Atlas) data revealed that NSCLC patients with a lower level of IL6 had a longer overall survival time compared to those with a higher level of IL6. Overall, our data indicate that the targeting of IL6/JAK2/STAT3 signaling in activated microglia may be a promising new approach for inhibiting brain metastasis in NSCLC patients.
Collapse
Affiliation(s)
- Yu Jin
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei Province, China
| | - Yalin Kang
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei Province, China
| | - Minghuan Wang
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei Province, China
| | - Bili Wu
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei Province, China
| | - Beibei Su
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei Province, China
| | - Han Yin
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei Province, China
| | - Yang Tang
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei Province, China
| | - Qianxia Li
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei Province, China
| | - Wenjie Wei
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei Province, China
| | - Qi Mei
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei Province, China
| | - Guangyuan Hu
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei Province, China
| | - Veronika Lukacs-Kornek
- Institute of Experimental Immunology, University Clinic of Rheinische Friedrich-Wilhelms-University, Bonn, Germany
| | - Jian Li
- Institute of Experimental Immunology, University Clinic of Rheinische Friedrich-Wilhelms-University, Bonn, Germany
| | - Kongming Wu
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei Province, China
| | - Xianglin Yuan
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei Province, China.
| | - Wei Wang
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei Province, China.
| |
Collapse
|
1179
|
Smit MM, Feller KJ, You L, Storteboom J, Begce Y, Beerens C, Chien MP. Spatially Annotated Single Cell Sequencing for Unraveling Intratumor Heterogeneity. Front Bioeng Biotechnol 2022; 10:829509. [PMID: 35273957 PMCID: PMC8902076 DOI: 10.3389/fbioe.2022.829509] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2021] [Accepted: 01/11/2022] [Indexed: 11/27/2022] Open
Abstract
Intratumor heterogeneity is a major obstacle to effective cancer treatment. Current methods to study intratumor heterogeneity using single-cell RNA sequencing (scRNA-seq) lack information on the spatial organization of cells. While state-of-the art spatial transcriptomics methods capture the spatial distribution, they either lack single cell resolution or have relatively low transcript counts. Here, we introduce spatially annotated single cell sequencing, based on the previously developed functional single cell sequencing (FUNseq) technique, to spatially profile tumor cells with deep scRNA-seq and single cell resolution. Using our approach, we profiled cells located at different distances from the center of a 2D epithelial cell mass. By profiling the cell patch in concentric bands of varying width, we showed that cells at the outermost edge of the patch responded strongest to their local microenvironment, behaved most invasively, and activated the process of epithelial-to-mesenchymal transition (EMT) to migrate to low-confluence areas. We inferred cell-cell communication networks and demonstrated that cells in the outermost ∼10 cell wide band, which we termed the invasive edge, induced similar phenotypic plasticity in neighboring regions. Applying FUNseq to spatially annotate and profile tumor cells enables deep characterization of tumor subpopulations, thereby unraveling the mechanistic basis for intratumor heterogeneity.
Collapse
Affiliation(s)
- Myrthe M. Smit
- Department of Molecular Genetics, Erasmus University Medical Center, Rotterdam, Netherlands
- Erasmus MC Cancer Institute, Rotterdam, Netherlands
| | - Kate J. Feller
- Department of Molecular Genetics, Erasmus University Medical Center, Rotterdam, Netherlands
- Erasmus MC Cancer Institute, Rotterdam, Netherlands
| | - Li You
- Department of Molecular Genetics, Erasmus University Medical Center, Rotterdam, Netherlands
- Erasmus MC Cancer Institute, Rotterdam, Netherlands
| | - Jelle Storteboom
- Department of Molecular Genetics, Erasmus University Medical Center, Rotterdam, Netherlands
- Erasmus MC Cancer Institute, Rotterdam, Netherlands
| | - Yasin Begce
- Department of Molecular Genetics, Erasmus University Medical Center, Rotterdam, Netherlands
- Erasmus MC Cancer Institute, Rotterdam, Netherlands
| | - Cecile Beerens
- Department of Molecular Genetics, Erasmus University Medical Center, Rotterdam, Netherlands
- Erasmus MC Cancer Institute, Rotterdam, Netherlands
| | - Miao-Ping Chien
- Department of Molecular Genetics, Erasmus University Medical Center, Rotterdam, Netherlands
- Erasmus MC Cancer Institute, Rotterdam, Netherlands
- Oncode Institute, Utrecht, Netherlands
- *Correspondence: Miao-Ping Chien,
| |
Collapse
|
1180
|
Golgi phosphoprotein 3 induces autophagy and epithelial-mesenchymal transition to promote metastasis in colon cancer. Cell Death Dis 2022; 8:76. [PMID: 35190555 PMCID: PMC8861175 DOI: 10.1038/s41420-022-00864-2] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Revised: 01/15/2022] [Accepted: 02/01/2022] [Indexed: 12/19/2022]
Abstract
In this study, we aimed to investigate whether and how Golgi phosphoprotein 3 (GOLPH3) facilitates colon cancer metastasis via the regulation of autophagy and epithelial-mesenchymal transition (EMT). The role GOLPH3 plays in colon cancer metastasis was analyzed using western blotting, immunohistochemistry, transwell, wound-healing, and zebrafish assays. Autophagy and EMT were assessed via RNA-sequencing (RNA-seq) analysis, mRFP-GFP-LC3 reporter assays, and their related markers. Significant associations were found between colon cancer clinical and pathological stages and poor prognosis. GOLPH3 facilitates colon cancer metastasis, both in vitro and in vivo. RNA-seq analysis of GOLPH3-overexpressing and control cell models revealed that GOLPH3 enhances EMT and autophagy. Moreover, examination of autophagic, epithelial, and mesenchymal markers in GOLPH3-overexpressing, -silenced, and control cell lines revealed that GOLPH3 promotes EMT and autophagy. When autophagy was inhibited, GOLPH3-promoted metastasis and EMT were counteracted in vitro and in vivo. Using RNA-seq, PI3K/Akt signaling was identified as the key downstream pathway on which GOLPH3 acts. Mechanistically, we demonstrated that GOLPH3 stimulates autophagy and induces EMT via the suppression of the phosphorylation of protein kinase B (Akt) at Ser473. In summary, GOLPH3 induces autophagy and EMT, promoting metastasis in colon cancer. Beyond this, and in contrast to conventional perspectives, we discovered that GOLPH3 represses the phosphorylation of Akt at Ser473.
Collapse
|
1181
|
Hsueh CY, Huang Q, Gong H, Shen Y, Sun J, Lau HC, Zhang D, Tang D, Wu C, Guo Y, Huang H, Cao P, Tao L, Zhang M, Zhou L. A positive feed-forward loop between Fusobacterium nucleatum and ethanol metabolism reprogramming drives laryngeal cancer progression and metastasis. iScience 2022; 25:103829. [PMID: 35198889 PMCID: PMC8851092 DOI: 10.1016/j.isci.2022.103829] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Revised: 12/10/2021] [Accepted: 01/21/2022] [Indexed: 12/21/2022] Open
Abstract
Alcohol consumption, which affects the structure and composition of the laryngeal microbiota, is one of the most important risk factors for laryngeal squamous cell cancer (LSCC). Our results demonstrated that high enrichment of Fusobacterium nucleatum (F. nucleatum) in LSCC was associated with poor prognosis. F. nucleatum increased miR-155-5p and miR-205-5p expression to suppress alcohol dehydrogenase 1B (ADH1B) and transforming growth factor β receptor 2 (TGFBR2) expression by activating innate immune signaling, resulting in ethanol metabolism reprogramming to allow F. nucleatum accumulation and PI3K/AKT signaling pathway activation to promote epithelial-mesenchymal transition, further exacerbating the uncontrolled progression and metastasis of LSCC. Therefore, the positive feed-forward loop between F. nucleatum and ethanol metabolism reprogramming promotes cell proliferation, migration, and invasion to affect LSCC patient prognosis. The amount of F. nucleatum is a potential prognostic biomarker, which yields valuable insight into clinical management that may improve the oncologic outcome of patients with LSCC.
Collapse
Affiliation(s)
- Chi-Yao Hsueh
- Department of Otorhinolaryngology, Eye & ENT Hospital, Fudan University, 83 Fen Yang Road, Shanghai 200031, China
| | - Qiang Huang
- Department of Otorhinolaryngology, Eye & ENT Hospital, Fudan University, 83 Fen Yang Road, Shanghai 200031, China
| | - Hongli Gong
- Department of Otorhinolaryngology, Eye & ENT Hospital, Fudan University, 83 Fen Yang Road, Shanghai 200031, China
| | - Yujie Shen
- Department of Otorhinolaryngology, Eye & ENT Hospital, Fudan University, 83 Fen Yang Road, Shanghai 200031, China
| | - Ji Sun
- Department of Pathology, Eye & ENT Hospital, Fudan University, 83 Fen Yang Road, Shanghai 200031, China
| | - Hui-Ching Lau
- Department of Otorhinolaryngology, Eye & ENT Hospital, Fudan University, 83 Fen Yang Road, Shanghai 200031, China
| | - Duo Zhang
- Department of Otorhinolaryngology, Eye & ENT Hospital, Fudan University, 83 Fen Yang Road, Shanghai 200031, China
| | - Di Tang
- Department of Otorhinolaryngology, Eye & ENT Hospital, Fudan University, 83 Fen Yang Road, Shanghai 200031, China
| | - Chunping Wu
- Department of Otorhinolaryngology, Eye & ENT Hospital, Fudan University, 83 Fen Yang Road, Shanghai 200031, China
| | - Yang Guo
- Department of Otorhinolaryngology, Eye & ENT Hospital, Fudan University, 83 Fen Yang Road, Shanghai 200031, China
| | - Huiying Huang
- Department of Otorhinolaryngology, Eye & ENT Hospital, Fudan University, 83 Fen Yang Road, Shanghai 200031, China
| | - Pengyu Cao
- Department of Otorhinolaryngology, Eye & ENT Hospital, Fudan University, 83 Fen Yang Road, Shanghai 200031, China
| | - Lei Tao
- Department of Otorhinolaryngology, Eye & ENT Hospital, Fudan University, 83 Fen Yang Road, Shanghai 200031, China
| | - Ming Zhang
- Department of Otorhinolaryngology, Eye & ENT Hospital, Fudan University, 83 Fen Yang Road, Shanghai 200031, China
| | - Liang Zhou
- Department of Otorhinolaryngology, Eye & ENT Hospital, Fudan University, 83 Fen Yang Road, Shanghai 200031, China
| |
Collapse
|
1182
|
The Role of SMAD4 Inactivation in Epithelial-Mesenchymal Plasticity of Pancreatic Ductal Adenocarcinoma: The Missing Link? Cancers (Basel) 2022; 14:cancers14040973. [PMID: 35205719 PMCID: PMC8870198 DOI: 10.3390/cancers14040973] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Revised: 02/11/2022] [Accepted: 02/11/2022] [Indexed: 01/27/2023] Open
Abstract
Simple Summary Pancreatic ductal adenocarcinoma (PDAC) is currently one of the deadliest cancers. Despite the progress that has been made in the research of patient care and the understanding of pancreatic cancer, the survival rate remains mediocre. SMAD4, a tumor-suppressor gene, is specifically inactivated in 50–55% of pancreatic cancers. The role of SMAD4 protein loss in PDAC remains controversial, but seems to be associated with worse overall survival and metastasis. Here, we review the function of SMAD4 inactivation in the context of a specific biological process called epithelial–mesenchymal transition, as it has been increasingly associated with tumor formation, metastasis and resistance to therapy. By improving our understanding of these molecular mechanisms, we hope to find new targets for therapy and improve the care of patients with PDAC. Abstract Pancreatic ductal adenocarcinoma (PDAC) presents a five-year survival rate of 10% and its incidence increases over the years. It is, therefore, essential to improve our understanding of the molecular mechanisms that promote metastasis and chemoresistance in PDAC, which are the main causes of death in these patients. SMAD4 is inactivated in 50% of PDACs and its loss has been associated with worse overall survival and metastasis, although some controversy still exists. SMAD4 is the central signal transducer of the transforming growth factor-beta (TGF-beta) pathway, which is notably known to play a role in epithelial–mesenchymal transition (EMT). EMT is a biological process where epithelial cells lose their characteristics to acquire a spindle-cell phenotype and increased motility. EMT has been increasingly studied due to its potential implication in metastasis and therapy resistance. Recently, it has been suggested that cells undergo EMT transition through intermediary states, which is referred to as epithelial–mesenchymal plasticity (EMP). The intermediary states are characterized by enhanced aggressiveness and more efficient metastasis. Therefore, this review aims to summarize and analyze the current knowledge on SMAD4 loss in patients with PDAC and to investigate its potential role in EMP in order to better understand its function in PDAC carcinogenesis.
Collapse
|
1183
|
Privat-Maldonado A, Verloy R, Cardenas Delahoz E, Lin A, Vanlanduit S, Smits E, Bogaerts A. Cold Atmospheric Plasma Does Not Affect Stellate Cells Phenotype in Pancreatic Cancer Tissue in Ovo. Int J Mol Sci 2022; 23:ijms23041954. [PMID: 35216069 PMCID: PMC8878510 DOI: 10.3390/ijms23041954] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Revised: 02/04/2022] [Accepted: 02/08/2022] [Indexed: 02/06/2023] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a challenging neoplastic disease, mainly due to the development of resistance to radio- and chemotherapy. Cold atmospheric plasma (CAP) is an alternative technology that can eliminate cancer cells through oxidative damage, as shown in vitro, in ovo, and in vivo. However, how CAP affects the pancreatic stellate cells (PSCs), key players in the invasion and metastasis of PDAC, is poorly understood. This study aims to determine the effect of an anti-PDAC CAP treatment on PSCs tissue developed in ovo using mono- and co-cultures of RLT-PSC (PSCs) and Mia PaCa-2 cells (PDAC). We measured tissue reduction upon CAP treatment and mRNA expression of PSC activation markers and extracellular matrix (ECM) remodelling factors via qRT-PCR. Protein expression of selected markers was confirmed via immunohistochemistry. CAP inhibited growth in Mia PaCa-2 and co-cultured tissue, but its effectiveness was reduced in the latter, which correlates with reduced ki67 levels. CAP did not alter the mRNA expression of PSC activation and ECM remodelling markers. No changes in MMP2 and MMP9 expression were observed in RLT-PSCs, but small changes were observed in Mia PaCa-2 cells. Our findings support the ability of CAP to eliminate PDAC cells, without altering the PSCs.
Collapse
Affiliation(s)
- Angela Privat-Maldonado
- PLASMANT, Chemistry Department, Faculty of Sciences, University of Antwerp, 2610 Antwerp, Belgium; (R.V.); (A.L.); (A.B.)
- Solid Tumor Immunology Group, Center for Oncological Research, Integrated Personalized and Precision Oncology Network, Department of Molecular Imaging, Pathology, Radiotherapy and Oncology, University of Antwerp, 2610 Antwerp, Belgium;
- Correspondence: ; Tel.: +32-3265-25-76
| | - Ruben Verloy
- PLASMANT, Chemistry Department, Faculty of Sciences, University of Antwerp, 2610 Antwerp, Belgium; (R.V.); (A.L.); (A.B.)
- Solid Tumor Immunology Group, Center for Oncological Research, Integrated Personalized and Precision Oncology Network, Department of Molecular Imaging, Pathology, Radiotherapy and Oncology, University of Antwerp, 2610 Antwerp, Belgium;
| | - Edgar Cardenas Delahoz
- Industrial Vision Lab InViLab, Faculty of Applied Engineering, University of Antwerp, 2610 Antwerp, Belgium; (E.C.D.); (S.V.)
| | - Abraham Lin
- PLASMANT, Chemistry Department, Faculty of Sciences, University of Antwerp, 2610 Antwerp, Belgium; (R.V.); (A.L.); (A.B.)
- Solid Tumor Immunology Group, Center for Oncological Research, Integrated Personalized and Precision Oncology Network, Department of Molecular Imaging, Pathology, Radiotherapy and Oncology, University of Antwerp, 2610 Antwerp, Belgium;
| | - Steve Vanlanduit
- Industrial Vision Lab InViLab, Faculty of Applied Engineering, University of Antwerp, 2610 Antwerp, Belgium; (E.C.D.); (S.V.)
| | - Evelien Smits
- Solid Tumor Immunology Group, Center for Oncological Research, Integrated Personalized and Precision Oncology Network, Department of Molecular Imaging, Pathology, Radiotherapy and Oncology, University of Antwerp, 2610 Antwerp, Belgium;
| | - Annemie Bogaerts
- PLASMANT, Chemistry Department, Faculty of Sciences, University of Antwerp, 2610 Antwerp, Belgium; (R.V.); (A.L.); (A.B.)
| |
Collapse
|
1184
|
Li J, Lai Z, Li H, Niu W, Du Z, Han Y, Chen L, Zhang C, Tang Y, Zou H, Gao X, Gao L. On-Cell Catalytic Detection of Epithelial-to-Mesenchymal Transition by a Clusterzyme Bioprobe. Anal Chem 2022; 94:3023-3028. [PMID: 35133805 DOI: 10.1021/acs.analchem.1c05556] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
We construct a peptide-conjugated metal cluster as an enzyme-like catalytic bioprobe to enhance quantitative analysis of a membrane protein biomarker and detect epithelial-to-mesenchymal transition of tumor cells. This bioprobe with atomically precise formula, termed clusterzyme, possesses selective recognition and intrinsic enzyme-like activity. These favorable features facilitate sensitive quantitative analysis of the membrane protein in situ through on-cell catalytic signal amplification. This clusterzyme-based analytical method exhibits excellent compatibility with a traditional enzyme-linked immunosorbent assay and improved detection sensitivity with accuracy and robustness. Further, the expression level of the membrane protein reflects the ability of migration and invasion of model tumor cells, revealing epithelial-to-mesenchymal transition process. This work offers a facile and sensitive approach to monitor tumor cell type evolution at the molecular level, demonstrating a potential application of early cancer diagnosis and therapy assessment.
Collapse
Affiliation(s)
- Jiaojiao Li
- Department of Chemistry and Biology, Faculty of Environment and Life Science, Center of Excellence for Environmental Safety and Biological Effects, Beijing University of Technology, Beijing 100124, China
| | - Zijing Lai
- Department of Chemistry and Biology, Faculty of Environment and Life Science, Center of Excellence for Environmental Safety and Biological Effects, Beijing University of Technology, Beijing 100124, China
| | - Han Li
- Department of Chemistry and Biology, Faculty of Environment and Life Science, Center of Excellence for Environmental Safety and Biological Effects, Beijing University of Technology, Beijing 100124, China
| | - Wenchao Niu
- Department of Chemistry and Biology, Faculty of Environment and Life Science, Center of Excellence for Environmental Safety and Biological Effects, Beijing University of Technology, Beijing 100124, China
| | - Zhongying Du
- Department of Chemistry and Biology, Faculty of Environment and Life Science, Center of Excellence for Environmental Safety and Biological Effects, Beijing University of Technology, Beijing 100124, China
| | - Ying Han
- Department of Chemistry and Biology, Faculty of Environment and Life Science, Center of Excellence for Environmental Safety and Biological Effects, Beijing University of Technology, Beijing 100124, China
| | - Lu Chen
- Department of Environmental Protection and Biopharmaceuticals, Beijing Industrial Technician College, Beijing 100023, China
| | - Chunyu Zhang
- Institute of Materia Medica, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan 250000, Shandong China
| | - Yuhua Tang
- Department of Chemistry and Biology, Faculty of Environment and Life Science, Center of Excellence for Environmental Safety and Biological Effects, Beijing University of Technology, Beijing 100124, China
| | - Haolin Zou
- Department of Chemistry and Biology, Faculty of Environment and Life Science, Center of Excellence for Environmental Safety and Biological Effects, Beijing University of Technology, Beijing 100124, China
| | - Xueyun Gao
- Department of Chemistry and Biology, Faculty of Environment and Life Science, Center of Excellence for Environmental Safety and Biological Effects, Beijing University of Technology, Beijing 100124, China
| | - Liang Gao
- Department of Chemistry and Biology, Faculty of Environment and Life Science, Center of Excellence for Environmental Safety and Biological Effects, Beijing University of Technology, Beijing 100124, China
| |
Collapse
|
1185
|
Wang HY, Zhang XP, Wang W. Regulation of epithelial-to-mesenchymal transition in hypoxia by the HIF-1α network. FEBS Lett 2022; 596:338-349. [PMID: 34905218 DOI: 10.1002/1873-3468.14258] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2021] [Revised: 11/30/2021] [Accepted: 12/08/2021] [Indexed: 12/17/2022]
Abstract
Epithelial-to-mesenchymal transition (EMT) plays a significant role in cancer metastasis. A series of models have focused on EMT regulation by TGF-β network. However, how EMT is regulated under hypoxia is less understood. We developed a model of HIF-1α network to explore the potential link between EMT and the network topology. Our results revealed that three positive feedback loops, composed of HIF-1α and its three targets SNAIL, TWIST, and miR-210, should be sequentially activated to induce EMT under aggravating hypoxia. We suggested that the number of the positive feedback loops is critical for determining the number of stable states in EMT. Our work may advance the understanding of the significance of network topology in the regulation of EMT.
Collapse
Affiliation(s)
- Hang-Yu Wang
- Kuang Yaming Honors School, Nanjing University, China
| | - Xiao-Peng Zhang
- Kuang Yaming Honors School, Nanjing University, China
- Institute for Brain Sciences, Nanjing University, China
| | - Wei Wang
- Institute for Brain Sciences, Nanjing University, China
- National Laboratory of Solid State Microstructure and Department of Physics, Nanjing University, China
| |
Collapse
|
1186
|
Wang S, Xie J, Zou X, Pan T, Yu Q, Zhuang Z, Zhong Y, Zhao X, Wang Z, Li R, Lei Y, Yin J, Yuan Y, Wei X, Liu L, Liu S, Yang H, Wu L. Establish an assessment model to characterized metastasis ability Single-cell multiomics reveals heterogeneous cell states linked to metastatic potential in liver cancer cell lines. iScience 2022; 25:103857. [PMID: 35198910 PMCID: PMC8850337 DOI: 10.1016/j.isci.2022.103857] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Revised: 01/01/2022] [Accepted: 01/28/2022] [Indexed: 11/16/2022] Open
Abstract
Hepatocellular carcinoma (HCC) is the most common liver cancer with a high rate of metastasis. However, the molecular mechanisms that drive metastasis remain unclear. We combined single-cell transcriptomic, proteomic, and chromatin accessibility data to investigate how heterogeneous phenotypes contribute to metastatic potential in five HCC cell lines. We confirmed that the prevalence of a mesenchymal state and levels of cell proliferation are linked to the metastatic potential. We also identified a rare hypoxic subtype that has a higher capacity for glycolysis and exhibits dormant, invasive, and malignant characteristics. This subtype has increased metastatic potential. We further identified a robust 14-gene panel representing this hypoxia signature and this hypoxia signature could serve as a prognostic index. Our data provide a valuable data resource, facilitate a deeper understanding of metastatic mechanisms, and may help diagnosis of metastatic potential in individual patients, thus supporting personalized medicine. Provide a high-resolution single-cell triple-omics data of five liver cancer cell lines Identify a robust 14-gene set representing hypoxia signature The hypoxia signature is associated with prognosis Establish an assessment model to characterized metastasis ability
Collapse
|
1187
|
Cook DP, Wrana JL. A specialist-generalist framework for epithelial-mesenchymal plasticity in cancer. Trends Cancer 2022; 8:358-368. [DOI: 10.1016/j.trecan.2022.01.014] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Revised: 01/17/2022] [Accepted: 01/24/2022] [Indexed: 12/12/2022]
|
1188
|
Wu F, Zhang Y, Chen X, Wang Y, Peng H, Zhang Z, Yang Y, Wang Q. Bioinformatics analysis of key genes and potential mechanism in cadmium-induced breast cancer progression. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2022; 29:11883-11892. [PMID: 34558042 DOI: 10.1007/s11356-021-16542-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Accepted: 09/10/2021] [Indexed: 06/13/2023]
Abstract
Cadmium (Cd) may be associated with breast cancer progression, but the detailed molecular mechanism has not been fully elucidated. In this study, one public dataset (GSE136595) was used to identify differentially expressed genes (DEGs) in Cd-treated MCF-7 breast cancer cells. We determined a total of 2077 DEGs, and Ingenuity Pathway Analysis (IPA) software showed that 246 of them were related to tumor progression. Pathway analysis of these DEGs indicated that the HIF1α signaling and the epithelial-mesenchymal transition (EMT) pathway regulated by growth factors might be activated. Moreover, twist family bHLH transcription factor 1 (TWIST1), lysine demethylase 3A (KDM3A), Kruppel-like factor 4 (KLF4), nuclear protein 1 (NUPR1), neurogenin 3 (NEUROG3), and HNF1 homeobox B (HNF1B) might be the key transcription factors. And the result of protein-protein interaction (PPI) analysis showed that the hub genes in these 246 DEGs were tumor protein p53 (TP53), polo-like kinase 1 (PLK1), sirtuin 1 (SIRT1), protein tyrosine phosphatase non-receptor type 11 (PTPN11), caspase 8 (CASP8), cyclin-dependent kinase 6 (CDK6), calmodulin 3 (CALM3), KRAS proto-oncogene (KRAS), extra spindle pole bodies like 1 (ESPL1), and marker of proliferation Ki-67 (MKI67). Further analysis indicated that TWIST1, NUPR1, KRAS, and PTPN11 were related to the prognostic of breast cancer based on the Cancer Genome Atlas (TCGA) and they were validated to be upregulated in the Cd-treated MCF-7 cells. Our results suggested that the HIF1α signaling and the EMT pathway regulated by growth factors might be participant in the Cd-induced breast cancer progression and TWIST1, NUPR1, KRAS, and PTPN11 might be potential key genes.
Collapse
Affiliation(s)
- Fei Wu
- Department of Toxicology, School of Public Health, Sun Yat-sen University, Guangzhou, China
| | - Yangchun Zhang
- Department of Toxicology, School of Public Health, Sun Yat-sen University, Guangzhou, China
| | - Xu Chen
- Department of Toxicology, School of Public Health, Sun Yat-sen University, Guangzhou, China
| | - Yuqing Wang
- Department of Toxicology, School of Public Health, Sun Yat-sen University, Guangzhou, China
| | - Honghao Peng
- Department of Toxicology, School of Public Health, Sun Yat-sen University, Guangzhou, China
| | - Zhengbao Zhang
- Department of Quality Management, Guangdong Provincial Center for Disease Prevention and Control, Guangzhou, China
| | - Ying Yang
- Department of Quality Management, Guangdong Provincial Center for Disease Prevention and Control, Guangzhou, China.
| | - Qing Wang
- Department of Toxicology, School of Public Health, Sun Yat-sen University, Guangzhou, China.
| |
Collapse
|
1189
|
Kisoda S, Mouri Y, Kitamura N, Yamamoto T, Miyoshi K, Kudo Y. The role of partial-EMT in the progression of head and neck squamous cell carcinoma. J Oral Biosci 2022; 64:176-182. [DOI: 10.1016/j.job.2022.02.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Revised: 02/07/2022] [Accepted: 02/09/2022] [Indexed: 10/19/2022]
|
1190
|
Feng X, Jiang J, Sun L, Zhou Q. CDK5RAP3 acts as a putative tumor inhibitor in papillary thyroid carcinoma via modulation of Akt/GSK-3β/Wnt/β-catenin signaling. Toxicol Appl Pharmacol 2022; 440:115940. [DOI: 10.1016/j.taap.2022.115940] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Revised: 02/14/2022] [Accepted: 02/21/2022] [Indexed: 02/07/2023]
|
1191
|
Huang Y, Wang Y, Wang Y, Wang N, Duan Q, Wang S, Liu M, Bilal MA, Zheng Y. LPCAT1 Promotes Cutaneous Squamous Cell Carcinoma via EGFR-Mediated Protein Kinase B/p38MAPK Signaling Pathways. J Invest Dermatol 2022; 142:303-313.e9. [PMID: 34358528 DOI: 10.1016/j.jid.2021.07.163] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2021] [Revised: 07/10/2021] [Accepted: 07/12/2021] [Indexed: 02/07/2023]
Abstract
Cutaneous squamous cell carcinoma (cSCC) is the second most common form of skin cancer. LPCAT1, a lysophosphatidylcholine acyltransferase, takes a center stage in membrane lipid remodeling. LPCAT1 is elevated in several cancers and contributes to cancer development. However, its role and molecular mechanisms in cSCC remain to be elucidated. In this study, we found that LPCAT1 was upregulated in cSCC tissues and in cell lines. In vitro, loss-of-function and gain-of-function experiments demonstrated that LPCAT1 facilitated cSCC cell proliferation, protected cells against apoptosis, accelerated epithelial‒mesenchymal transition, and enhanced cell metastasis. Mechanistically, LPCAT1 regulated EGFR signaling. The oncogenic effect of LPCAT1 was mediated by EGFR/protein kinase B and EGFR/p38MAPK pathways in cSCC. Using the xenograft mouse model, we consolidated the results mentioned earlier. In conclusion, LPCAT1 contributed to cSCC progression through EGFR-mediated protein kinase B and p38MAPK signaling pathways. LPCAT1 may serve as a target for therapeutic intervention in cSCC.
Collapse
Affiliation(s)
- Yingjian Huang
- Department of Dermatology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Yuqian Wang
- Department of Dermatology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Yan Wang
- Center for Mitochondrial Biology and Medicine, The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, China
| | - Ning Wang
- Department of Dermatology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Qiqi Duan
- Department of Dermatology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Shengbang Wang
- Department of Dermatology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Meng Liu
- Department of Dermatology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Muhammad Ahsan Bilal
- Department of Dermatology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Yan Zheng
- Department of Dermatology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China.
| |
Collapse
|
1192
|
Zhou H, Zou J, Shao C, Zhou A, Yu J, Chen S, Xu C. Prolyl 4-hydroxylase subunit alpha 3 facilitates human colon cancer growth and metastasis through the TGF-β/Smad signaling pathway. Pathol Res Pract 2022; 230:153749. [PMID: 34959098 DOI: 10.1016/j.prp.2021.153749] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Revised: 12/21/2021] [Accepted: 12/21/2021] [Indexed: 02/07/2023]
Abstract
Prolyl 4-hydroxylase subunit alpha 3 (P4HA3) has been known to be associated with a variety of human cancers. However, the role of P4HA3 on colon cancer growth and metastasis is unclear. In this study, we investigated the effect of P4HA3 on the growth and metastasis of colon cancer and its possible molecular mechanism. First of all, we demonstrated that P4HA3 expression was greatly higher in cells and tissues of colon cancer than that in non-tumor tissues and cells, and the prognosis of patients who had higher P4HA3 was distinctively poorer than patients who had lower level of P4HA3. Second, it was shown that P4HA3 knockdown strongly inhibited the migration, proliferation and invasion ability of colon cancer cells. However, P4HA3 over-expression accelerated the abilities. Meanwhile, P4HA3 could promote subcutaneous tumorigenesis in nude mice in vivo. In addition, P4HA3 knockdown significantly decreased mesenchymal markers Vimentin, N-cadherin and Snail expression and increased epithelial marker E-cadherin expression. And conversely, over-expression of P4HA3 produced the opposite effects. In the current study, there was further evidence that down-regulating P4HA3 significantly reduced both TGF-β and its following molecules including p-Smad2 as well as p-Smad3. However, overexpression of P4HA3 showed the opposite effect. In conclusion, this study shows that P4HA3 promotes the human colon cancer growth and metastasis by affecting TGF-β/Smad signaling pathway. P4HA3 may become a new target for early diagnosis, treatment and prognosis assessment of colon cancer.
Collapse
Affiliation(s)
- Hailang Zhou
- Department of Gastroenterology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, PR China; Department of Gastroenterology, Lianshui People's Hospital Affiliated to Kangda College of Nanjing Medical University, Huaian, Jiangsu 223400, PR China
| | - Junwei Zou
- Department of General Surgery, The Second Affiliated Hospital of Wannan Medical College, Wuhu, Anhui 241000, PR China
| | - Changjiang Shao
- Department of Gastroenterology, The Second People's Hospital of Lianyungang, Lianyungang, Jiangsu 222006, PR China
| | - Aijun Zhou
- Department of Gastroenterology, Lianshui People's Hospital Affiliated to Kangda College of Nanjing Medical University, Huaian, Jiangsu 223400, PR China
| | - Jiufeng Yu
- Department of Traditional Chinese Medicine, Lianshui People's Hospital Affiliated to Kangda College of Nanjing Medical University, Huaian, Jiangsu 223400, PR China
| | - Song Chen
- The Institute of Life Sciences, Jiangsu College of Nursing,Huaian, Jiangsu 223300, PR China
| | - Chunfang Xu
- Department of Gastroenterology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, PR China.
| |
Collapse
|
1193
|
Long Non-coding RNA ZFPM2-AS1: A Novel Biomarker in the Pathogenesis of Human Cancers. Mol Biotechnol 2022; 64:725-742. [DOI: 10.1007/s12033-021-00443-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Accepted: 12/22/2021] [Indexed: 10/19/2022]
|
1194
|
Role of glycosyltransferases in carcinogenesis; growth factor signaling and EMT/MET programs. Glycoconj J 2022; 39:167-176. [PMID: 35089466 PMCID: PMC8795723 DOI: 10.1007/s10719-022-10041-3] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Revised: 01/05/2022] [Accepted: 01/11/2022] [Indexed: 02/06/2023]
Abstract
The glycosylation of cell surface receptors has been shown to regulate each step of signal transduction, including receptor trafficking to the cell surface, ligand binding, dimerization, phosphorylation, and endocytosis. In this review we focus on the role of glycosyltransferases that are involved in the modification of N-glycans, such as the effect of branching and elongation in signaling by various cell surface receptors. In addition, the role of those enzymes in the EMT/MET programs, as related to differentiation and cancer development, progress and therapy resistance is discussed.
Collapse
|
1195
|
Xu Y, Shi H, Wang M, Huang P, Xu M, Han S, Li H, Wang Y. LACTB suppresses carcinogenesis in lung cancer and regulates the EMT pathway. Exp Ther Med 2022; 23:247. [PMID: 35222724 PMCID: PMC8815028 DOI: 10.3892/etm.2022.11172] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Accepted: 12/31/2021] [Indexed: 11/24/2022] Open
Abstract
Lung cancer causes thousands of deaths worldwide every year, and present therapeutics show little benefit for advanced-stage patients. Researchers do not know why and how lung cancer begins. Lactamase β (LACTB) is a tumor-suppressor in some cancers. However, its role in lung cancer is unknown. By analyzing the TCGA database and Kaplan-Meier Plotter database, LACTB was found to be downregulated in lung cancer tissues but the methylation level was increased. Patients with high LACTB expression exhibited improved survival. Then, in vitro assays demonstrated that LACTB overexpression inhibited cell migration and invasion, and induced apoptosis in H1299 and H1975 cells. Knockdown of LACTB caused the reverse effects. Moreover, a much higher apoptotic rate and more potent inhibitory effects on H1299 and H1975 cells were obtained when LACTB was combined with docetaxel. In addition, members of the epithelial-mesenchymal transition (EMT) signaling pathway were assessed using western blot analysis. The expression of E-cadherin was decreased while levels of N-cadherin and vimentin were increased after knockdown of LACTB in lung cancer cells. By contrast, overexpression of LACTB increased the level of E-cadherin but decreased N-cadherin and vimentin. Therefore, LACTB is a tumor suppressor in lung cancer that inhibits cell migration and invasion and induces cell apoptosis. Meanwhile, LACTB was found to strengthen the anticancer role of docetaxel and to suppress the EMT pathway in lung cancer.
Collapse
Affiliation(s)
- Yihui Xu
- Medical Research and Laboratory Diagnostic Center, Central Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250013, P.R. China
| | - Hubo Shi
- Department of Thoracic Surgery, Shangdong Public Health Clinical Center, Jinan, Shandong 250102, P.R. China
| | - Min Wang
- Medical Research and Laboratory Diagnostic Center, Central Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250013, P.R. China
| | - Ping Huang
- Medical Research and Laboratory Diagnostic Center, Central Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250013, P.R. China
| | - Mingjie Xu
- Medical Research and Laboratory Diagnostic Center, Central Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250013, P.R. China
| | - Shuyi Han
- Medical Research and Laboratory Diagnostic Center, Central Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250013, P.R. China
| | - Huanjie Li
- School of Medicine, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, P.R. China
| | - Yunshan Wang
- Medical Research and Laboratory Diagnostic Center, Central Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250013, P.R. China
| |
Collapse
|
1196
|
Zu F, Chen H, Liu Q, Zang H, Li Z, Tan X. Syntenin Regulated by miR-216b Promotes Cancer Progression in Pancreatic Cancer. Front Oncol 2022; 12:790788. [PMID: 35155233 PMCID: PMC8831246 DOI: 10.3389/fonc.2022.790788] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Accepted: 01/05/2022] [Indexed: 12/24/2022] Open
Abstract
Outcomes for patients with pancreatic cancer (PC) are poor; therefore, there is an urgent need to identify novel therapeutic targets involved in the progression of PC. We previously identified 161 differentially expressed proteins (DEPs) in PC. Syntenin (SDCBP) was identified as a survival-related protein through integrated, survival, and Cox analyses. High expression of SDCBP was associated with a poor prognosis in PC tissue and promoted the proliferation, migration, and invasion of PC cells, and induced epithelial–mesenchymal transition (EMT) via the PI3K/AKT pathway. Additionally, we elucidated the regulatory mechanism underlying these roles of SDCBP at the post-transcriptional level. microRNAs (miRNAs) of SDCBP were predicted using bioinformatics. Low levels of miR-216b expression were confirmed in PC tissues and were negatively correlated with SDCBP expression. miR-216b was found to directly regulate SDCBP expression through luciferase reporter assays. Furthermore, agomiR-216b restrained PC proliferation, migration, invasion, and EMT via the PI3K/AKT pathway, whereas antagomiR-216b facilitated this process. Notably, the knockout of SDCBP counteracted the effect of antagomiR-216b in PC, which suggested that miR-216b and SDCBP represent molecular targets underlying PC progression and EMT. Finally, the results were validated in in vivo studies. These findings indicated that low expression of miR-216b and the oncogene SDCBP contributes to PC migration, invasion, and EMT, and that they have potential as future therapeutic targets for patients with PC.
Collapse
Affiliation(s)
- Fuqiang Zu
- Department of Pancreatic and Thyroid Surgery, General Surgery, Shengjing Hospital of China Medical University, Shenyang, China
| | - Hui Chen
- Department of Pancreatic and Thyroid Surgery, General Surgery, Shengjing Hospital of China Medical University, Shenyang, China
| | - Qingfeng Liu
- Department of General Surgery, The People’s Hospital of China Medical University, Shenyang, China
| | - Hui Zang
- Department of General Surgery, The People’s Hospital of China Medical University, Shenyang, China
| | - Zeyu Li
- Department of Pancreatic and Thyroid Surgery, General Surgery, Shengjing Hospital of China Medical University, Shenyang, China
| | - Xiaodong Tan
- Department of Pancreatic and Thyroid Surgery, General Surgery, Shengjing Hospital of China Medical University, Shenyang, China
- *Correspondence: Xiaodong Tan,
| |
Collapse
|
1197
|
Sobolev VV, Khashukoeva AZ, Evina OE, Geppe NA, Chebysheva SN, Korsunskaya IM, Tchepourina E, Mezentsev A. Role of the Transcription Factor FOSL1 in Organ Development and Tumorigenesis. Int J Mol Sci 2022; 23:1521. [PMID: 35163444 PMCID: PMC8835756 DOI: 10.3390/ijms23031521] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2021] [Revised: 01/26/2022] [Accepted: 01/27/2022] [Indexed: 12/25/2022] Open
Abstract
The transcription factor FOSL1 plays an important role in cell differentiation and tumorigenesis. Primarily, FOSL1 is crucial for the differentiation of several cell lineages, namely adipocytes, chondrocytes, and osteoblasts. In solid tumors, FOSL1 controls the progression of tumor cells through the epithelial-mesenchymal transformation. In this review, we summarize the available data on FOSL1 expression, stabilization, and degradation in the cell. We discuss how FOSL1 is integrated into the intracellular signaling mechanisms and provide a comprehensive analysis of FOSL1 influence on gene expression. We also analyze the pathological changes caused by altered Fosl1 expression in genetically modified mice. In addition, we dedicated a separate section of the review to the role of FOSL1 in human cancer. Primarily, we focus on the FOSL1 expression pattern in solid tumors, FOSL1 importance as a prognostic factor, and FOSL1 perspectives as a molecular target for anticancer therapy.
Collapse
Affiliation(s)
- Vladimir V. Sobolev
- Center for Theoretical Problems in Physico-Chemical Pharmacology, Russian Academy of Sciences, 109029 Moscow, Russia; (I.M.K.); (E.T.)
| | - Asiat Z. Khashukoeva
- Federal State Autonomous Educational Institution of Higher Education, N.I. Pirogov Russian National Research Medical University of the Ministry of Health of the Russian Federation, 117997 Moscow, Russia;
| | - Olga E. Evina
- “JSC DK Medsi”, Medical and Diagnostics Center, 125284 Moscow, Russia;
| | - Natalia A. Geppe
- NF Filatov Clinical Institute of Children’s Health, I.M. Sechenov First MSMU, 119435 Moscow, Russia; (N.A.G.); (S.N.C.)
| | - Svetlana N. Chebysheva
- NF Filatov Clinical Institute of Children’s Health, I.M. Sechenov First MSMU, 119435 Moscow, Russia; (N.A.G.); (S.N.C.)
| | - Irina M. Korsunskaya
- Center for Theoretical Problems in Physico-Chemical Pharmacology, Russian Academy of Sciences, 109029 Moscow, Russia; (I.M.K.); (E.T.)
| | - Ekaterina Tchepourina
- Center for Theoretical Problems in Physico-Chemical Pharmacology, Russian Academy of Sciences, 109029 Moscow, Russia; (I.M.K.); (E.T.)
| | - Alexandre Mezentsev
- Center for Theoretical Problems in Physico-Chemical Pharmacology, Russian Academy of Sciences, 109029 Moscow, Russia; (I.M.K.); (E.T.)
| |
Collapse
|
1198
|
Shi C, Xie Y, Li X, Li G, Liu W, Pei W, Liu J, Yu X, Liu T. Identification of Ferroptosis-Related Genes Signature Predicting the Efficiency of Invasion and Metastasis Ability in Colon Adenocarcinoma. Front Cell Dev Biol 2022; 9:815104. [PMID: 35155451 PMCID: PMC8826729 DOI: 10.3389/fcell.2021.815104] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Accepted: 12/03/2021] [Indexed: 01/14/2023] Open
Abstract
Background: Colon adenocarcinoma (COAD) is one of the most prevalent cancers worldwide and has become a leading cause of cancer death. Although many potential biomarkers of COAD have been screened with the bioinformatics method, it is necessary to explore novel markers for the diagnosis and appropriate individual treatments for COAD patients due to the high heterogeneity of this disease. Epithelial-to-mesenchymal transition (EMT)-mediated tumor metastasis suggests poor prognosis of cancers. Ferroptosis is involved in tumor development. EMT signaling can increase the cellular sensitivity to ferroptosis in tumors. The aim of our study is finding novel prognostic biomarkers to determine COAD patients for predicting efficiency of metastasis status and targeting precise ferroptosis-related therapy. Methods: A novel gene signature related to metastasis and ferroptosis was identified combing with risk model and WGCNA analysis with R software. The biological functions and predictive ability of the signature in COAD were explored through bioinformatics analysis. Results: We established a four-gene prognostic signature (MMP7, YAP1, PCOLCE, and HOXC11) based on EMT and ferroptosis related genes and validated the reliability and effectiveness of this model in COAD. This four-gene prognostic signature was closely connected with metastasis and ferroptosis sensitivity of COAD. Moreover, WGCNA analysis further confirmed the correlation between PCOLCE, HOXC11, and liver and lymphatic invasion of COAD. Conclusion: The four genes may become potential prognostic biomarkers to identify COAD patients with metastasis. Moreover, this four-gene signature may be able to determine the COAD suitable with ferroptosis induction therapy. Finally, PCOLCE2 and HOXC11 were selected individually because of their novelties and precise prediction ability. Overall, this signature provided novel possibilities for better prognostic evaluation of COAD patients and may be of great guiding significance for individualized treatment and clinical decision.
Collapse
Affiliation(s)
- Chunlei Shi
- Department of General Surgery, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin General Surgery Institute, Tianjin, China
| | - Yongjie Xie
- Key Laboratory of Cancer Prevention, Department of Pancreatic Cancer, National Clinical Research Center for Cancer, Tianjin’s Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Xueyang Li
- Key Laboratory of Cancer Prevention, Department of Pancreatic Cancer, National Clinical Research Center for Cancer, Tianjin’s Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
- Department of Breast Oncoplastic Surgery, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education, Tianjin, China
| | - Guangming Li
- Department of General Surgery, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin General Surgery Institute, Tianjin, China
| | - Weishuai Liu
- Key Laboratory of Cancer Prevention, Department of Pancreatic Cancer, National Clinical Research Center for Cancer, Tianjin’s Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
- Department of Pain Relief, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, China
| | - Wenju Pei
- Department of General Surgery, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin General Surgery Institute, Tianjin, China
| | - Jing Liu
- Key Laboratory of Cancer Prevention, Department of Pancreatic Cancer, National Clinical Research Center for Cancer, Tianjin’s Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
- Department of Breast Oncoplastic Surgery, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education, Tianjin, China
- *Correspondence: Jing Liu, ; Xiaozhou Yu, ; Tong Liu,
| | - Xiaozhou Yu
- Key Laboratory of Cancer Prevention, Department of Pancreatic Cancer, National Clinical Research Center for Cancer, Tianjin’s Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
- Department of Molecular Imaging and Nuclear Medicine, National Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
- *Correspondence: Jing Liu, ; Xiaozhou Yu, ; Tong Liu,
| | - Tong Liu
- Department of General Surgery, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin General Surgery Institute, Tianjin, China
- *Correspondence: Jing Liu, ; Xiaozhou Yu, ; Tong Liu,
| |
Collapse
|
1199
|
Chang LC, Chiu HM, Wu MS, Shen TL. The Role of Small Extracellular Vesicles in the Progression of Colorectal Cancer and Its Clinical Applications. Int J Mol Sci 2022; 23:1379. [PMID: 35163305 PMCID: PMC8835972 DOI: 10.3390/ijms23031379] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2022] [Revised: 01/17/2022] [Accepted: 01/24/2022] [Indexed: 02/01/2023] Open
Abstract
Colorectal cancer (CRC) is one of the most common cancers worldwide and a longstanding critical challenge for public health. Screening has been suggested to effectively reduce both the incidence and mortality of CRC. However, the drawback of the current screening modalities, both stool-based tests and colonoscopies, is limited screening adherence, which reduces the effectiveness of CRC screening. Blood tests are more acceptable than stool tests or colonoscopy as a first-line screening approach. Therefore, identifying blood biomarkers for detecting CRC and its precancerous neoplasms is urgently needed to fulfill the unmet clinical need. Currently, many kinds of blood contents, such as circulating tumor cells, circulating tumor nucleic acids, and extracellular vesicles, have been investigated as biomarkers for CRC detection. Among these, small extracellular vesicles (sEVs) have been demonstrated to detect CRC effectively in recent reports. sEVs enable intercellular shuttling-for instance, trafficking between recipient cancer cells and stromal cells-which can affect tumor initiation, proliferation, angiogenesis, immune regulation; metastasis, the cancer-specific molecules, such as proteins, microRNAs, long noncoding RNAs, and circular RNAs, loaded into cancer-derived sEVs may serve as biomarkers for the detection of cancers, including CRC. Indeed, accumulating evidence has shown that nucleic acids and proteins contained in CRC-derived sEVs are effective as blood biomarkers for CRC detection. However, investigations of the performance of sEVs for diagnosing CRC in clinical trials remains limited. Thus, the effectiveness of sEV biomarkers for diagnosing CRC needs further validation in clinical trials.
Collapse
Affiliation(s)
- Li-Chun Chang
- Department of Internal Medicine, National Taiwan University Hospital, Taipei 100, Taiwan; (L.-C.C.); (H.-M.C.); (M.-S.W.)
- Health Management Center, National Taiwan University Hospital, Taipei 100, Taiwan
| | - Han-Mo Chiu
- Department of Internal Medicine, National Taiwan University Hospital, Taipei 100, Taiwan; (L.-C.C.); (H.-M.C.); (M.-S.W.)
- Health Management Center, National Taiwan University Hospital, Taipei 100, Taiwan
| | - Ming-Shiang Wu
- Department of Internal Medicine, National Taiwan University Hospital, Taipei 100, Taiwan; (L.-C.C.); (H.-M.C.); (M.-S.W.)
| | - Tang-Long Shen
- Department of Plant Pathology and Microbiology, National Taiwan University, Taipei 100, Taiwan
- Center for Biotechnology, National Taiwan University, Taipei 100, Taiwan
- Genome and Systems Biology Degree Program, National Taiwan University, Taipei 100, Taiwan
| |
Collapse
|
1200
|
Yu K, Tan Z, Xin Y. Systematic evaluation of the anti-tumor effect of Phellinus linteus polysaccharide in thyroid carcinoma in vitro. Mol Biol Rep 2022; 49:2785-2793. [DOI: 10.1007/s11033-021-07090-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Accepted: 12/15/2021] [Indexed: 10/19/2022]
|