1
|
Din ZU, Cui B, Wang C, Zhang X, Mehmood A, Peng F, Liu Q. Crosstalk between lipid metabolism and EMT: emerging mechanisms and cancer therapy. Mol Cell Biochem 2025; 480:103-118. [PMID: 38622439 DOI: 10.1007/s11010-024-04995-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2023] [Accepted: 03/19/2024] [Indexed: 04/17/2024]
Abstract
Lipids are the key component of all membranes composed of a variety of molecules that transduce intracellular signaling and provide energy to the cells in the absence of nutrients. Alteration in lipid metabolism is a major factor for cancer heterogeneity and a newly identified cancer hallmark. Reprogramming of lipid metabolism affects the diverse cancer phenotypes, especially epithelial-mesenchymal transition (EMT). EMT activation is considered to be an essential step for tumor metastasis, which exhibits a crucial role in the biological processes including development, wound healing, and stem cell maintenance, and has been widely reported to contribute pathologically to cancer progression. Altered lipid metabolism triggers EMT and activates multiple EMT-associated oncogenic pathways. Although the role of lipid metabolism-induced EMT in tumorigenesis is an attractive field of research, there are still significant gaps in understanding the underlying mechanisms and the precise contributions of this interplay. Further study is needed to clarify the specific molecular mechanisms driving the crosstalk between lipid metabolism and EMT, as well as to determine the potential therapeutic implications. The increased dependency of tumor cells on lipid metabolism represents a novel therapeutic target, and targeting altered lipid metabolism holds promise as a strategy to suppress EMT and ultimately inhibit metastasis.
Collapse
Affiliation(s)
- Zaheer Ud Din
- Institute of Cancer Stem Cell, Dalian Medical University, 9 Western Section, Lvshun South Street, Lvshunkou District, Dalian, 116044, Liaoning, China
- Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, Institute of Aging Research, Guangdong Medical University, Dongguan, China
| | - Bai Cui
- Institute of Cancer Stem Cell, Dalian Medical University, 9 Western Section, Lvshun South Street, Lvshunkou District, Dalian, 116044, Liaoning, China
- State Key Laboratory of Oncology in South China, Cancer Center, Sun Yat-Sen University, Guangzhou, 510060, China
| | - Cenxin Wang
- Institute of Cancer Stem Cell, Dalian Medical University, 9 Western Section, Lvshun South Street, Lvshunkou District, Dalian, 116044, Liaoning, China
| | - Xiaoyu Zhang
- Institute of Cancer Stem Cell, Dalian Medical University, 9 Western Section, Lvshun South Street, Lvshunkou District, Dalian, 116044, Liaoning, China
| | - Arshad Mehmood
- Department of Neurology, The Second Hospital of Hebei Medical University, Shijiazhuang, 050000, China
| | - Fei Peng
- Institute of Cancer Stem Cell, Dalian Medical University, 9 Western Section, Lvshun South Street, Lvshunkou District, Dalian, 116044, Liaoning, China.
| | - Quentin Liu
- Institute of Cancer Stem Cell, Dalian Medical University, 9 Western Section, Lvshun South Street, Lvshunkou District, Dalian, 116044, Liaoning, China.
- State Key Laboratory of Oncology in South China, Cancer Center, Sun Yat-Sen University, Guangzhou, 510060, China.
| |
Collapse
|
2
|
Wei J, Sun J, Pan Y, Cao M, Wang Y, Yuan T, Guo A, Han R, Ding X, Yang G, Yu T, Ding R. Revealing genes related teat number traits via genetic variation in Yorkshire pigs based on whole-genome sequencing. BMC Genomics 2024; 25:1217. [PMID: 39695943 DOI: 10.1186/s12864-024-11109-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2024] [Accepted: 11/29/2024] [Indexed: 12/20/2024] Open
Abstract
BACKGROUND Teat number is one of the most important indicators to evaluate the lactation performance of sows, and increasing the teat number has become an important method to improve the economic efficiency of farms. Therefore, it is particularly important to deeply analyze the genetic mechanism of teat number traits in pigs. In this study, we detected Single Nucleotide Ploymorphism (SNP), Insertion-Deletion (InDel) and Structural variant (SV) by high-coverage whole-genome resequencing data, and selected teat number at birth and functional teat number as two types of teat number traits for genome-wide association study (GWAS) to reveal candidate genes associated with pig teat number traits. RESULTS In this study, we used whole genome resequencing data from 560 Yorkshire sows to detect SNPs, InDels and SVs, and performed GWAS for the traits of born teat number and functional teat number, and detected a total of 85 significant variants and screened 214 candidate genes, including HEG1, XYLT1, SULF1, MUC13, VRTN, RAP1A and NPVF. Among them, HEG1 and XYLT1 were the new candidate genes in this study. The co-screening and population validation of multiple traits suggested that HEG1 may have a critical effect on the born teat number. CONCLUSION Our study shows that more candidate genes associated with pig teat number traits can be identified by GWAS with different variant types. Through large population validation, we found that HEG1 may be a new key candidate gene affecting pig teat number traits. In conclusion, the results of this study provide new information for exploring the genetic mechanisms affecting pig teat number traits and genetic improvement of pigs.
Collapse
Affiliation(s)
- Jialin Wei
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, Laboratory of Animal Fat Deposition & Muscle Development, College of Animal Science and Technology, Northwest A&F University, Yangling, 712100, Shaanxi, China
| | - Jingchun Sun
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, Laboratory of Animal Fat Deposition & Muscle Development, College of Animal Science and Technology, Northwest A&F University, Yangling, 712100, Shaanxi, China
- Key Laboratory of Agroecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, 410125, Hunan, China
| | - Yi Pan
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, Laboratory of Animal Fat Deposition & Muscle Development, College of Animal Science and Technology, Northwest A&F University, Yangling, 712100, Shaanxi, China
| | - Minghao Cao
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, Laboratory of Animal Fat Deposition & Muscle Development, College of Animal Science and Technology, Northwest A&F University, Yangling, 712100, Shaanxi, China
| | - Yulong Wang
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, Laboratory of Animal Fat Deposition & Muscle Development, College of Animal Science and Technology, Northwest A&F University, Yangling, 712100, Shaanxi, China
| | - Tiantian Yuan
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, Laboratory of Animal Fat Deposition & Muscle Development, College of Animal Science and Technology, Northwest A&F University, Yangling, 712100, Shaanxi, China
| | - Ao Guo
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, Laboratory of Animal Fat Deposition & Muscle Development, College of Animal Science and Technology, Northwest A&F University, Yangling, 712100, Shaanxi, China
| | - Ruihua Han
- Tongchuan Animal Husbandry Technology Extension Station, Tongchuan, 727000, Shaanxi, China
| | - Xiangdong Ding
- Key Laboratory of Animal Genetics and Breeding of Ministry of Agriculture and Rural Affairs, National Engineering Laboratory of Animal Breeding, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China
| | - Gongshe Yang
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, Laboratory of Animal Fat Deposition & Muscle Development, College of Animal Science and Technology, Northwest A&F University, Yangling, 712100, Shaanxi, China
| | - Taiyong Yu
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, Laboratory of Animal Fat Deposition & Muscle Development, College of Animal Science and Technology, Northwest A&F University, Yangling, 712100, Shaanxi, China
| | - Rongrong Ding
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, Laboratory of Animal Fat Deposition & Muscle Development, College of Animal Science and Technology, Northwest A&F University, Yangling, 712100, Shaanxi, China.
| |
Collapse
|
3
|
Liu M, Tang B, Xiang R, Hu P, Xu C, Hu L, Li Q. Aberrant expression of MRAS and HEG1 as the biomarkers for osimertinib resistance in LUAD. Discov Oncol 2024; 15:678. [PMID: 39560891 DOI: 10.1007/s12672-024-01552-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/03/2024] [Accepted: 11/07/2024] [Indexed: 11/20/2024] Open
Abstract
Epidermal growth factor receptor (EGFR)-tyrosine kinase inhibitors (TKIs) are the most applied targeted therapy for EGFR-mutant lung adenocarcinoma (LUAD). The third-generation EGFR-TKI, osimertinib, is widely used throughout lung cancer treatment, with single or combination modes. One of the main barriers in osimertinib treatment is the acquired resistance and mechanisms are not fully understood. Gene expression other than genetic mutations might predict drug response and mediate resistance occurrence. We analyzed six datasets of osimertinib-resistant LUAD cells from the Gene Expression Omnibus (GEO) database and identified two hub genes, named MRAS and HEG1. We found that the expression mode of MRAS/HEG1 in LUAD was osimertinib-dependent and contributed to drug resistance. We also explored potential mechanisms of hub genes related osimertinib resistance and emphasized the M2 infiltration involved. Moreover, potential therapeutic agents conquering MRAS/HEG1-related resistance were also identified. In conclusion, MRAS and HEG1 might be responsible for osimertinib resistance and could be promising prognostic biomarkers for osimertinib response in LUAD, which might provide insights into therapeutic strategies.
Collapse
Affiliation(s)
- Mingxin Liu
- Department of Thoracic Surgery, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital and Institute, Sichuan Cancer Center, School of Medicine, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu, 610042, China
| | - Bo Tang
- Department of Oncology and Cancer Institute, Sichuan Academy of Medical Science and Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610072, China
| | - Run Xiang
- Department of Thoracic Surgery, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital and Institute, Sichuan Cancer Center, School of Medicine, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu, 610042, China
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, Sichuan University, Chengdu, 610042, China
| | - Peihong Hu
- Department of Thoracic Surgery, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital and Institute, Sichuan Cancer Center, School of Medicine, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu, 610042, China
| | - Chuan Xu
- Department of Oncology and Cancer Institute, Sichuan Academy of Medical Science and Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610072, China
- Yu-Yue Pathology Scientific Research Center, Chongqing, 400039, China
- Jinfeng Laboratory, Chongqing, 401329, China
| | - Lanlin Hu
- Department of Oncology and Cancer Institute, Sichuan Academy of Medical Science and Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610072, China.
- Yu-Yue Pathology Scientific Research Center, Chongqing, 400039, China.
- Jinfeng Laboratory, Chongqing, 401329, China.
| | - Qiang Li
- Department of Thoracic Surgery, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital and Institute, Sichuan Cancer Center, School of Medicine, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu, 610042, China.
| |
Collapse
|
4
|
Ma S, Meng G, Liu T, You J, He R, Zhao X, Cui Y. The Wnt signaling pathway in hepatocellular carcinoma: Regulatory mechanisms and therapeutic prospects. Biomed Pharmacother 2024; 180:117508. [PMID: 39362068 DOI: 10.1016/j.biopha.2024.117508] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2024] [Revised: 08/26/2024] [Accepted: 09/25/2024] [Indexed: 10/05/2024] Open
Abstract
Hepatocellular carcinoma (HCC) is a malignant tumor that arises from hepatocytes. Multiple signaling pathways play a regulatory role in the occurrence and development of HCC, with the Wnt signaling pathway being one of the primary regulatory pathways. In normal hepatocytes, the Wnt signaling pathway maintains cell regeneration and organ development. However, when aberrant activated, the Wnt pathway is closely associated with invasion, cancer stem cells(CSCs), drug resistance, and immune evasion in HCC. Among these factors, the development of drug resistance is one of the most important factors affecting the efficacy of HCC treatment. These mechanisms form the basis for tumor cell adaptation and evolution within the body, enabling continuous changes in tumor cells, resistance to drugs and immune system attacks, leading to metastasis and recurrence. In recent years, there have been numerous new discoveries regarding these mechanisms. An increasing number of drugs targeting the Wnt signaling pathway have been developed, with some already entering clinical trials. Therefore, this review encompasses the latest research on the role of the Wnt signaling pathway in the onset and progression of HCC, as well as advancements in its therapeutic strategies.
Collapse
Affiliation(s)
- Shihui Ma
- Department of General Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150000, China
| | - Guorui Meng
- Department of General Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150000, China
| | - Tong Liu
- Department of General Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150000, China
| | - Junqi You
- Department of General Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150000, China
| | - Risheng He
- Department of General Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150000, China
| | - Xudong Zhao
- Department of General Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150000, China
| | - Yunfu Cui
- Department of General Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150000, China.
| |
Collapse
|
5
|
Sun X, Chen B, Shan Y, Jian M, Wang Z. Lectin microarray based glycan profiling of exosomes for dynamic monitoring of colorectal cancer progression. Anal Chim Acta 2024; 1316:342819. [PMID: 38969421 DOI: 10.1016/j.aca.2024.342819] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Revised: 06/02/2024] [Accepted: 06/03/2024] [Indexed: 07/07/2024]
Abstract
BACKGROUND Exosomes, as emerging biomarkers in liquid biopsies in recent years, offer profound insights into cancer diagnostics due to their unique molecular signatures. The glycosylation profiles of exosomes have emerged as potential biomarkers, offering a novel and less invasive method for cancer diagnosis and monitoring. Colorectal cancer (CRC) represents a substantial global health challenge and burden. Thus there is a great need for the aberrant glycosylation patterns on the surface of CRC cell-derived exosomes, proposing them as potential biomarkers for tumor characterization. RESULTS The interactions of 27 lectins with exosomes from three CRC cell lines (SW480, SW620, HCT116) and one normal colon epithelial cell line (NCM460) have been analyzed by the lectin microarray. The result indicates that Ulex Europaeus Agglutinin I (UEA-I) exhibits high affinity and specificity towards exosomes derived from SW480 cells. The expression of glycosylation related genes within cells has been analyzed by high-throughput quantitative polymerase chain reaction (HT-qPCR). The experimental result of HT-qPCR is consistent with that of lectin microarray. Moreover, the limit of detection (LOD) of UEA-I microarray is calculated to be as low as 2.7 × 105 extracellular vehicles (EVs) mL-1 (three times standard deviation (3σ) of blank sample). The UEA-I microarray has been successfully utilized to dynamically monitor the progression of tumors in mice-bearing SW480 CRC subtype, applicable in tumor sizes ranging from 2 mm to 20 mm in diameter. SIGNIFICANCE The results reveal that glycan expression pattern of exosome is linked to specific CRC subtypes, and regulated by glycosyltransferase and glycosidase genes of mother cells. Our findings illuminate the potential of glycosylation molecules on the surface of exosomes as reliable biomarkers for diagnosis of tumor at early stage and monitoring of cancer progression.
Collapse
Affiliation(s)
- Xudong Sun
- State Key Laboratory of Electroanalytical Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, PR China; School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei, 230026, PR China
| | - Bowen Chen
- Department of Thyroid Surgery, General Surgery Center, The First Hospital of Jilin University, Changchun, 130021, PR China
| | - Yongjie Shan
- State Key Laboratory of Electroanalytical Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, PR China; School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei, 230026, PR China
| | - Minghong Jian
- State Key Laboratory of Electroanalytical Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, PR China
| | - Zhenxin Wang
- State Key Laboratory of Electroanalytical Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, PR China; School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei, 230026, PR China; National Analytical Research Center of Electrochemistry and Spectroscopy, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, PR China.
| |
Collapse
|
6
|
Liu F, Yan T, Cui D, Jiang J. Identification and validation of a prognostic model based on four genes related to satellite nodules in hepatocellular carcinoma. Sci Rep 2024; 14:15633. [PMID: 38972883 PMCID: PMC11228042 DOI: 10.1038/s41598-024-66610-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2024] [Accepted: 07/02/2024] [Indexed: 07/09/2024] Open
Abstract
Satellite nodules is a key clinical characteristic which has prognostic value of hepatocellular carcinoma (HCC). Currently, there is no gene-level predictive model for Satellite nodules in liver cancer. For the 377 HCC cases collected from the dataset of Cancer Genome Atlas (TCGA), their original pathological data were analyzed to extract information regarding satellite nodules status as well as other relevant pathological data. Then, this study employed statistical modeling for prognostic model establishment in TCGA, and validation in International Cancer Genome Consortium (ICGC) cohorts and GSE76427. Through rigorous statistical analyses, 253 differential satellite nodules-related genes (SNRGs) were identified, and four key genes related to satellite nodules and prognosis were selected to construct a prognostic model. The high-risk group predicted by our model exhibited an unfavorable overall survival (OS) outlook and demonstrated an association with adverse worse clinical characteristics such as larger tumor size, higher alpha-fetoprotein, microvascular invasion and advanced stage. Moreover, the validation of the model's prognostic value in the ICGC and GSE76427 cohorts mirrored that of the TCGA cohort. Besides, the high-risk group also showed higher levels of resting Dendritic cells, M0 macrophages infiltration, alongside decreased levels of CD8+ T cells and γδT cells infiltration. The prognostic model based on SNRGs can reliability predict the OS of HCC and is likely to have predictive value of immunotherapy for HCC.
Collapse
Affiliation(s)
- Feng Liu
- Department of Interventional Oncology, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Tinghua Yan
- The First Clinical Medical College of Jinan University, Guangzhou, China
| | - Dan Cui
- Department of Interventional Oncology, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Jinhua Jiang
- Department of Interventional Oncology, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China.
| |
Collapse
|
7
|
Tong M, Bai Y, Han X, Kong L, Ren L, Zhang L, Li X, Yao J, Yan B. Single-cell profiling transcriptomic reveals cellular heterogeneity and cellular crosstalk in choroidal neovascularization model. Exp Eye Res 2024; 242:109877. [PMID: 38537669 DOI: 10.1016/j.exer.2024.109877] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2023] [Revised: 03/18/2024] [Accepted: 03/21/2024] [Indexed: 04/01/2024]
Abstract
Choroidal neovascularization (CNV) is a hallmark of neovascular age-related macular degeneration (nAMD) and a major contributor to vision loss in nAMD cases. However, the identification of specific cell types associated with nAMD remains challenging. Herein, we performed single-cell sequencing to comprehensively explore the cellular diversity and understand the foundational components of the retinal pigment epithelium (RPE)/choroid complex. We unveiled 10 distinct cell types within the RPE/choroid complex. Notably, we observed significant heterogeneity within endothelial cells (ECs), fibroblasts, and macrophages, underscoring the intricate nature of the cellular composition in the RPE/choroid complex. Within the EC category, four distinct clusters were identified and EC cluster 0 was tightly associated with choroidal neovascularization. We identified five clusters of fibroblasts actively involved in the pathogenesis of nAMD, influencing fibrotic responses, angiogenic effects, and photoreceptor function. Additionally, three clusters of macrophages were identified, suggesting their potential roles in regulating the progression of nAMD through immunomodulation and inflammation regulation. Through CellChat analysis, we constructed a complex cell-cell communication network, revealing the role of EC clusters in interacting with fibroblasts and macrophages in the context of nAMD. These interactions were found to govern angiogenic effects, fibrotic responses, and inflammatory processes. In summary, this study reveals noteworthy cellular heterogeneity in the RPE/choroid complex and provides valuable insights into the pathogenesis of CNV. These findings will open up potential avenues for deep understanding and targeted therapeutic interventions in nAMD.
Collapse
Affiliation(s)
- Ming Tong
- Eye Institute and Department of Ophthalmology, Eye and ENT Hospital, Fudan University, Shanghai, 200031, China
| | - Yun Bai
- College of Information Science, Shanghai Ocean University, Shanghai, 201306, China
| | - Xiaoyan Han
- Eye Institute and Department of Ophthalmology, Eye and ENT Hospital, Fudan University, Shanghai, 200031, China
| | - Lingjie Kong
- Eye Institute and Department of Ophthalmology, Eye and ENT Hospital, Fudan University, Shanghai, 200031, China
| | - Ling Ren
- Eye Institute and Department of Ophthalmology, Eye and ENT Hospital, Fudan University, Shanghai, 200031, China
| | - Linyu Zhang
- The Fourth School of Clinical Medicine, Nanjing Medical University, Nanjing, 210000, China; The Affiliated Eye Hospital, Nanjing Medical University, Nanjing, 210000, China
| | - Xiumiao Li
- The Affiliated Eye Hospital, Nanjing Medical University, Nanjing, 210000, China
| | - Jin Yao
- The Fourth School of Clinical Medicine, Nanjing Medical University, Nanjing, 210000, China; The Affiliated Eye Hospital, Nanjing Medical University, Nanjing, 210000, China.
| | - Biao Yan
- Department of Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China.
| |
Collapse
|
8
|
Zhang J, Li R, Zhang H, Wang S, Zhao Y. ITGA2 as a prognostic factor of glioma promotes GSCs invasion and EMT by activating STAT3 phosphorylation. Carcinogenesis 2024; 45:235-246. [PMID: 38142122 DOI: 10.1093/carcin/bgad096] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2023] [Revised: 11/24/2023] [Accepted: 12/20/2023] [Indexed: 12/25/2023] Open
Abstract
Glioma is the most common malignant brain tumor in adults with a high mortality and recurrence rate. Integrin alpha 2 (ITGA2) is involved in cell adhesion, stem cell regulation, angiogenesis and immune cell function. The role of ITGA2 in glioma malignant invasion remains unknown. The function and clinical relevance of ITGA2 were analysed by bioinformatics databases. The expression of ITGA2 in parent cells and GSCs was detected by flow cytometry and immunofluorescence double staining. The role of ITGA2 on the malignant phenotype of GSCs and epithelial-mesenchymal transition (EMT) was identified by stem cell function assays and Western blot. The effect of ITGA2 on glioma progression in vivo was determined by the intracranial orthotopic xenograft model. Immunohistochemistry, Spearman correlation and Kaplan-Meier were used to analyse the relationship of ITGA2 with clinical features and glioma prognosis. Biological analysis showed that ITGA2 might be related to cell invasion and migration. ITGA2, enriched in GSCs and co-expressed with SOX2, promoted the invasion and migration of GSCs by activating STAT3 phosphorylation and enhancing EMT. ITGA2 knockout suppressed the intracranial orthotopic xenograft growth and prolonged the survival of xenograft mice. In addition, the expression level of ITGA2 was significantly correlated to the grade of malignancy, N-cadherin and Ki67. High expression of ITGA2 indicated a worse prognosis of glioma patients. As a biomarker for the prediction of prognosis, ITGA2 promotes the malignant invasion of GSCs by activating STAT3 phosphorylation and enhancing EMT, leading to tumor recurrence and poor prognosis.
Collapse
Affiliation(s)
- Jin Zhang
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- Stoke Center, Beijing Institute for Brain Disorders, Beijing, China
- Beijing Key Laboratory of Translational Medicine for Cerebrovascular Disease, Beijing, China
- China National Clinical Research Center for Neurological Diseases, Beijing, China
| | - Ruinan Li
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- Stoke Center, Beijing Institute for Brain Disorders, Beijing, China
- Beijing Key Laboratory of Translational Medicine for Cerebrovascular Disease, Beijing, China
- China National Clinical Research Center for Neurological Diseases, Beijing, China
| | - Haibin Zhang
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- Stoke Center, Beijing Institute for Brain Disorders, Beijing, China
- Beijing Key Laboratory of Translational Medicine for Cerebrovascular Disease, Beijing, China
- China National Clinical Research Center for Neurological Diseases, Beijing, China
| | - Shanshan Wang
- Department of Oncology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, China
- Tianjin's Clinical Research Center for Cancer, Tianjin, China
- Key Laboratory of Cancer Prevention and Therapy, Tianjin, China
| | - Yuanli Zhao
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- Stoke Center, Beijing Institute for Brain Disorders, Beijing, China
- Beijing Key Laboratory of Translational Medicine for Cerebrovascular Disease, Beijing, China
- China National Clinical Research Center for Neurological Diseases, Beijing, China
| |
Collapse
|
9
|
Wang KY, Wang KJ, Shen LL, Wang XH. The down-regulation of GADD45B leads to a conversion of cellular oxidative phosphorylation to glycolysis and promotes the progression of bladder cancer. Heliyon 2024; 10:e27427. [PMID: 38501008 PMCID: PMC10945183 DOI: 10.1016/j.heliyon.2024.e27427] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Revised: 02/28/2024] [Accepted: 02/28/2024] [Indexed: 03/20/2024] Open
Abstract
Background The predominant feature of cancer cells during the process of carcinogenesis is the inclination towards glycolytic metabolism rather than mitochondrial oxidative phosphorylation. Nevertheless, there is a scarcity of research investigating the correlation between bladder cancer and mitochondrial energy metabolism. Methods A qPCR array comprising 90 genes associated with mitochondrial oxidative phosphorylation was employed to discern metabolic disparities between three sets of bladder cancer tissue and adjacent normal tissue. Wound healing and transwell assays were utilized to assess cell migration and invasion capabilities, respectively. Colony formation assays were conducted to ascertain the tumorigenic potential of the cells. The proliferative capacity of the cells was examined through in vitro CCK-8 assays. Additionally, nude mouse models were established to evaluate the impact of bladder tumor cells on in vivo proliferation. The Seahorse XFe96 Analyzer was utilized to quantify mitochondrial oxidative phosphorylation, while the levels of glucose-6-phosphate and pyruvate were assessed to evaluate glycolysis. Results Examination of qPCR array data demonstrated a noteworthy inhibition of mitochondrial oxidative phosphorylation in bladder cancer tissue, as evidenced by the down-regulation of a majority of genes associated with mitochondrial energy metabolism. Notably, GADD45B may potentially exert a significant influence on bladder cancer development, warranting further investigation. The down-regulation of GADD45B in bladder cancer cells resulted in impaired mitochondrial respiration and elevated levels of glycolysis, thereby enhancing cell migration and invasion. Conversely, up-regulation of GADD45B had the opposite effect. Furthermore, over-expression of GADD45B inhibited tumor proliferation and tumorigenesis in both in vitro and in vivo settings. Conclusion These findings from our study indicate that the down-regulation of GADD45B promotes the shift of cell mitochondrial oxidative phosphorylation towards glycolysis, thereby facilitating the progression of bladder cancer.
Collapse
Affiliation(s)
- Kai-yun Wang
- Department of Urology, the Affiliated People's Hospital of Ningbo University, Ningbo, Zhejiang, China
| | - Ke-jie Wang
- Department of Urology, the Affiliated People's Hospital of Ningbo University, Ningbo, Zhejiang, China
| | - Li-liang Shen
- Department of Urology, the Affiliated People's Hospital of Ningbo University, Ningbo, Zhejiang, China
| | - Xu-hui Wang
- Department of Urology, the Affiliated People's Hospital of Ningbo University, Ningbo, Zhejiang, China
| |
Collapse
|
10
|
Li Q, Sharkey A, Sheridan M, Magistrati E, Arutyunyan A, Huhn O, Sancho-Serra C, Anderson H, McGovern N, Esposito L, Fernando R, Gardner L, Vento-Tormo R, Turco MY, Moffett A. Human uterine natural killer cells regulate differentiation of extravillous trophoblast early in pregnancy. Cell Stem Cell 2024; 31:181-195.e9. [PMID: 38237587 DOI: 10.1016/j.stem.2023.12.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2023] [Revised: 10/19/2023] [Accepted: 12/20/2023] [Indexed: 02/04/2024]
Abstract
In humans, balanced invasion of trophoblast cells into the uterine mucosa, the decidua, is critical for successful pregnancy. Evidence suggests that this process is regulated by uterine natural killer (uNK) cells, but how they influence reproductive outcomes is unclear. Here, we used our trophoblast organoids and primary tissue samples to determine how uNK cells affect placentation. By locating potential interaction axes between trophoblast and uNK cells using single-cell transcriptomics and in vitro modeling of these interactions in organoids, we identify a uNK cell-derived cytokine signal that promotes trophoblast differentiation at the late stage of the invasive pathway. Moreover, it affects transcriptional programs involved in regulating blood flow, nutrients, and inflammatory and adaptive immune responses, as well as gene signatures associated with disorders of pregnancy such as pre-eclampsia. Our findings suggest mechanisms on how optimal immunological interactions between uNK cells and trophoblast enhance reproductive success.
Collapse
Affiliation(s)
- Qian Li
- Department of Pathology, University of Cambridge, Cambridge CB2 1QP, UK; Centre for Trophoblast Research, University of Cambridge, Cambridge CB2 3EG, UK.
| | - Andrew Sharkey
- Department of Pathology, University of Cambridge, Cambridge CB2 1QP, UK; Centre for Trophoblast Research, University of Cambridge, Cambridge CB2 3EG, UK
| | - Megan Sheridan
- Department of Pathology, University of Cambridge, Cambridge CB2 1QP, UK; Centre for Trophoblast Research, University of Cambridge, Cambridge CB2 3EG, UK
| | - Elisa Magistrati
- Friedrich Miescher Institute for Biomedical Research, 4058 Basel, Switzerland
| | - Anna Arutyunyan
- Centre for Trophoblast Research, University of Cambridge, Cambridge CB2 3EG, UK; Wellcome Sanger Institute, Cambridge CB10 1SA, UK
| | - Oisin Huhn
- Department of Pathology, University of Cambridge, Cambridge CB2 1QP, UK; Centre for Trophoblast Research, University of Cambridge, Cambridge CB2 3EG, UK
| | - Carmen Sancho-Serra
- Centre for Trophoblast Research, University of Cambridge, Cambridge CB2 3EG, UK; Wellcome Sanger Institute, Cambridge CB10 1SA, UK
| | - Holly Anderson
- Department of Pathology, University of Cambridge, Cambridge CB2 1QP, UK
| | - Naomi McGovern
- Department of Pathology, University of Cambridge, Cambridge CB2 1QP, UK; Centre for Trophoblast Research, University of Cambridge, Cambridge CB2 3EG, UK
| | - Laura Esposito
- Department of Pathology, University of Cambridge, Cambridge CB2 1QP, UK; Centre for Trophoblast Research, University of Cambridge, Cambridge CB2 3EG, UK
| | - Ridma Fernando
- Friedrich Miescher Institute for Biomedical Research, 4058 Basel, Switzerland
| | - Lucy Gardner
- Department of Pathology, University of Cambridge, Cambridge CB2 1QP, UK; Centre for Trophoblast Research, University of Cambridge, Cambridge CB2 3EG, UK
| | - Roser Vento-Tormo
- Centre for Trophoblast Research, University of Cambridge, Cambridge CB2 3EG, UK; Wellcome Sanger Institute, Cambridge CB10 1SA, UK.
| | | | - Ashley Moffett
- Department of Pathology, University of Cambridge, Cambridge CB2 1QP, UK; Centre for Trophoblast Research, University of Cambridge, Cambridge CB2 3EG, UK.
| |
Collapse
|
11
|
Li M, Li T, Jin T, Chen Y, Cheng L, Liang Q, Yan S, Li T, Ran Q, Chen W. Abnormal activation of the Wnt3a/β-catenin signaling pathway promotes the expression of T-box transcription factor 3(TBX3) and the epithelial-mesenchymal transition pathway to mediate the occurrence of adenomyosis. Mol Biol Rep 2023; 50:9935-9950. [PMID: 37878207 DOI: 10.1007/s11033-023-08870-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Accepted: 09/29/2023] [Indexed: 10/26/2023]
Abstract
BACKGROUND T-box transcription factor 3(TBX3) is a transcription factor that can regulate cell proliferation, apoptosis, invasion, and migration in different tumor cells; however, its role in adenomyosis (ADM) has not been previously studied. Some of ADM's pathophysiological characteristics are similar to those of malignant tumors (e.g., abnormal proliferation, migration, and invasion). METHODS AND RESULTS We hypothesized that TBX3 might have a role in ADM. We used tamoxifen-induced Institute of Cancer research (ICR) mice to establish ADM disease model. The study procedure included western blotting and immunohistochemistry to analyze protein levels; additionally, we used intraperitoneal injection of Wnt/β-catenin pathway inhibitor XAV-939 to study the relationship between TBX3 and Wnt/β-catenin pathway as well as Anti-proliferation cell nuclear antigen( PCNA) and TUNEL to detect cell proliferation and apoptosis, respectively. TBX3 overexpression and epithelial-to-mesenchymal transition (EMT) in ADM mice was found to be associated with activation of the Wnt3a/β-catenin pathway. Treatment with XAV-939 in ADM mice led to the inhibition of both TBX3 and EMT; moreover, abnormal cell proliferation was suppressed, the depth of invasion of endometrium cells was limited. Thus, the use of XAV-939 effectively inhibited further invasion of endometrial cells. CONCLUSION These findings suggest that TBX3 may play an important role in the development of ADM. The expression of TBX3 in ADM was regulated by the Wnt3a/β-catenin pathway. The activation of the Wnt3a/β-catenin pathway in ADM promoted TBX3 expression and induced the occurrence of EMT, thus promoting cell proliferation and inhibiting apoptosis, ultimately accelerating the development of ADM. The study provides a reference for the diagnosis of ADM.
Collapse
Affiliation(s)
- Mengqi Li
- Department of Biochemistry and Molecular Biology, School of Medicine, Jinan University, 601 Huangpu Avenue West, Guangzhou, 510632, People's Republic of China
| | - Ting Li
- Department of Biochemistry and Molecular Biology, School of Medicine, Jinan University, 601 Huangpu Avenue West, Guangzhou, 510632, People's Republic of China
| | - Tingting Jin
- Department of Biochemistry and Molecular Biology, School of Medicine, Jinan University, 601 Huangpu Avenue West, Guangzhou, 510632, People's Republic of China
| | - Yi Chen
- Department of Gynecology, Guangdong Provincial Hospital of Chinese Medicine, 111 Dade Road, Guangzhou, 510120, People's Republic of China
| | - Lan Cheng
- Department of Gynecology, Guangdong Provincial Hospital of Chinese Medicine, 111 Dade Road, Guangzhou, 510120, People's Republic of China
| | - Qiheng Liang
- Department of Gynecology, Guangdong Provincial Hospital of Chinese Medicine, 111 Dade Road, Guangzhou, 510120, People's Republic of China
| | - Simiao Yan
- Department of Biochemistry and Molecular Biology, School of Medicine, Jinan University, 601 Huangpu Avenue West, Guangzhou, 510632, People's Republic of China
| | - Tingting Li
- Division of Histology and Embryology, International Joint Laboratory for Embryonic Development & Prenatal Medicine, School of Medicine, Jinan University, Guangzhou, 510632, People's Republic of China
| | - Qingzhen Ran
- Department of Gynecology, Guangdong Provincial Hospital of Chinese Medicine, 111 Dade Road, Guangzhou, 510120, People's Republic of China.
| | - Wanqun Chen
- Department of Biochemistry and Molecular Biology, School of Medicine, Jinan University, 601 Huangpu Avenue West, Guangzhou, 510632, People's Republic of China.
| |
Collapse
|
12
|
Mori K, Matsumoto K, Ikeda M, Koguchi D, Shimizu Y, Tsumura H, Ishii D, Tsuji S, Sato Y, Iwamura M. Membranous Expression of Heart Development Protein with EGF-like Domain 1 Is Associated with a Good Prognosis in Patients with Bladder Cancer. Diagnostics (Basel) 2023; 13:3067. [PMID: 37835810 PMCID: PMC10572329 DOI: 10.3390/diagnostics13193067] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Revised: 09/21/2023] [Accepted: 09/25/2023] [Indexed: 10/15/2023] Open
Abstract
OBJECTIVE To investigate the correlation between total protein expression of heart development protein with EGF-like domain 1 (HEG1) and clinicopathological characteristics in patients with bladder cancer (BC) after radical cystectomy (RC). PATIENTS AND METHODS We retrospectively analyzed data from 110 patients who underwent RC at Kitasato University Hospital. And we prepared an anti-HEG1 monoclonal antibody W10B9, which can detect total HEG1 protein. HEG1 protein expression in tumor cells was evaluated separately for membrane and cytoplasmic staining using immunohistochemistry. RESULTS Membranous HEG1 expression was associated with absent lymphovascular invasion (p < 0.01) and low pT stage (p < 0.01). Kaplan-Meier analysis revealed that the membranous HEG1-positive group had significantly long recurrence-free survival (RFS) (p < 0.01) and cancer-specific survival (p = 0.01). Expression of membranous HEG1 was identified as an independent prognostic factor for RFS (p = 0.04). There were no significant differences between cytoplasmic HEG1 expression and clinicopathologic factors including prognosis. CONCLUSION The expression of membranous HEG1 could serve as a favorable prognostic indicator in patients with BC treated with RC.
Collapse
Affiliation(s)
- Kohei Mori
- Department of Urology, Kitasato University School of Medicine, 1-15-1 Kitasato, Minami-ku, Sagamihara 252-0374, Kanagawa, Japan; (K.M.); (M.I.); (D.K.); (Y.S.); (H.T.); (D.I.); (Y.S.); (M.I.)
| | - Kazumasa Matsumoto
- Department of Urology, Kitasato University School of Medicine, 1-15-1 Kitasato, Minami-ku, Sagamihara 252-0374, Kanagawa, Japan; (K.M.); (M.I.); (D.K.); (Y.S.); (H.T.); (D.I.); (Y.S.); (M.I.)
| | - Masaomi Ikeda
- Department of Urology, Kitasato University School of Medicine, 1-15-1 Kitasato, Minami-ku, Sagamihara 252-0374, Kanagawa, Japan; (K.M.); (M.I.); (D.K.); (Y.S.); (H.T.); (D.I.); (Y.S.); (M.I.)
| | - Dai Koguchi
- Department of Urology, Kitasato University School of Medicine, 1-15-1 Kitasato, Minami-ku, Sagamihara 252-0374, Kanagawa, Japan; (K.M.); (M.I.); (D.K.); (Y.S.); (H.T.); (D.I.); (Y.S.); (M.I.)
| | - Yuriko Shimizu
- Department of Urology, Kitasato University School of Medicine, 1-15-1 Kitasato, Minami-ku, Sagamihara 252-0374, Kanagawa, Japan; (K.M.); (M.I.); (D.K.); (Y.S.); (H.T.); (D.I.); (Y.S.); (M.I.)
| | - Hideyasu Tsumura
- Department of Urology, Kitasato University School of Medicine, 1-15-1 Kitasato, Minami-ku, Sagamihara 252-0374, Kanagawa, Japan; (K.M.); (M.I.); (D.K.); (Y.S.); (H.T.); (D.I.); (Y.S.); (M.I.)
| | - Daisuke Ishii
- Department of Urology, Kitasato University School of Medicine, 1-15-1 Kitasato, Minami-ku, Sagamihara 252-0374, Kanagawa, Japan; (K.M.); (M.I.); (D.K.); (Y.S.); (H.T.); (D.I.); (Y.S.); (M.I.)
| | - Shoutaro Tsuji
- Department of Medical Technology & Clinical Engineering, Gunma University of Health and Welfare, 191-1 Kawamagari-machi, Maebashi-shi 371-0823, Gunma, Japan;
| | - Yuichi Sato
- Department of Urology, Kitasato University School of Medicine, 1-15-1 Kitasato, Minami-ku, Sagamihara 252-0374, Kanagawa, Japan; (K.M.); (M.I.); (D.K.); (Y.S.); (H.T.); (D.I.); (Y.S.); (M.I.)
- KITASATO-OTSUKA Biomedical Assay Laboratories Co., Ltd., 1-15-1 Kitasato, Minami-ku, Sagamihara 252-0329, Kanagawa, Japan
| | - Masatsugu Iwamura
- Department of Urology, Kitasato University School of Medicine, 1-15-1 Kitasato, Minami-ku, Sagamihara 252-0374, Kanagawa, Japan; (K.M.); (M.I.); (D.K.); (Y.S.); (H.T.); (D.I.); (Y.S.); (M.I.)
| |
Collapse
|
13
|
Alqurashi YE, Al-Hetty HRAK, Ramaiah P, Fazaa AH, Jalil AT, Alsaikhan F, Gupta J, Ramírez-Coronel AA, Tayyib NA, Peng H. Harnessing function of EMT in hepatocellular carcinoma: From biological view to nanotechnological standpoint. ENVIRONMENTAL RESEARCH 2023; 227:115683. [PMID: 36933639 DOI: 10.1016/j.envres.2023.115683] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/26/2023] [Revised: 03/08/2023] [Accepted: 03/11/2023] [Indexed: 05/08/2023]
Abstract
Management of cancer metastasis has been associated with remarkable reduction in progression of cancer cells and improving survival rate of patients. Since 90% of mortality are due to cancer metastasis, its suppression can improve ability in cancer fighting. The EMT has been an underlying cause in increasing cancer migration and it is followed by mesenchymal transformation of epithelial cells. HCC is the predominant kind of liver tumor threatening life of many people around the world with poor prognosis. Increasing patient prognosis can be obtained via inhibiting tumor metastasis. HCC metastasis modulation by EMT and HCC therapy by nanoparticles are discussed here. First of all, EMT happens during progression and advanced stages of HCC and therefore, its inhibition can reduce tumor malignancy. Moreover, anti-cancer compounds including all-trans retinoic acid and plumbaging, among others, have been considered as inhibitors of EMT. The EMT association with chemoresistance has been evaluated. Moreover, ZEB1/2, TGF-β, Snail and Twist are EMT modulators in HCC and enhancing cancer invasion. Therefore, EMT mechanism and related molecular mechanisms in HCC are evaluated. The treatment of HCC has not been only emphasized on targeting molecular pathways with pharmacological compounds and since drugs have low bioavailability, their targeted delivery by nanoparticles promotes HCC elimination. Moreover, nanoparticle-mediated phototherapy impairs tumorigenesis in HCC by triggering cell death. Metastasis of HCC and even EMT mechanism can be suppressed by cargo-loaded nanoparticles.
Collapse
Affiliation(s)
- Yaser E Alqurashi
- Department of Biology, College of Science Al-zulfi, Majmaah University, Al-Majmaah, 11952, Saudi Arabia
| | | | | | | | - Abduladheem Turki Jalil
- Medical Laboratories Techniques Department, Al-Mustaqbal University College, Babylon, Hilla, 51001, Iraq
| | - Fahad Alsaikhan
- College of Pharmacy, Prince Sattam Bin Abdulaziz University, Alkharj, Saudi Arabia.
| | - Jitendra Gupta
- Institute of Pharmaceutical Research, GLA University, Mathura, Pin Code 281406, U. P., India
| | - Andrés Alexis Ramírez-Coronel
- Azogues Campus Nursing Career, Health and Behavior Research Group (HBR), Psychometry and Ethology Laboratory, Catholic University of Cuenca, Ecuador; Epidemiology and Biostatistics Research Group, CES University, Colombia; Educational Statistics Research Group (GIEE), National University of Education, Ecuador
| | - Nahla A Tayyib
- Faculty of Nursing, Umm Al- Qura University, Makkah, Saudi Arabia
| | - Hu Peng
- Department of Emergency, Shanghai Tenth People's Hospital, Tongji University, Shanghai, 200072, China.
| |
Collapse
|
14
|
Ding B, Lou W, Fan W, Pan J. Exosomal miR-374c-5p derived from mesenchymal stem cells suppresses epithelial-mesenchymal transition of hepatocellular carcinoma via the LIMK1-Wnt/β-catenin axis. ENVIRONMENTAL TOXICOLOGY 2023; 38:1038-1052. [PMID: 36722453 DOI: 10.1002/tox.23746] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Revised: 01/11/2023] [Accepted: 01/16/2023] [Indexed: 06/18/2023]
Abstract
Metastasis is a leading cause to treatment failure in hepatocellular carcinoma (HCC) patients. Exosomes act as pivotal mediators in communication between different cells and exert effects on recipient cells by delivering bioactive cargoes, such as microRNAs (miRNAs). MiRNAs function in multiple steps of HCC development, including metastasis. MiR-374c-5p was previously identified as a tumor suppressor in some malignancies, while the current knowledge of its role in HCC metastasis is still limited. Herein, miR-374c-5p was found to be downregulated in HCC cell lines and clinical samples, and positively related with favorable prognosis in HCC patients. MiR-374c-5p transferred by exosomes derived from bone marrow mesenchymal stem cell (BMSC) suppressed migration, invasion and proliferation of HCC cells. LIMK1 was verified as downstream target gene of miR-374c-5p. Knockdown of LIMK1 reduced invasion, migration and proliferation of HCC cells, whereas overexpression functioned oppositely. The miR-374c-5p/LIMK1 axis suppressed epithelial-mesenchymal transition (EMT) by inactivating Wnt/β-catenin pathway. In addition, miR-374c-5p was downregulated and LIMK1 upregulated in TGF-β1 induced EMT. This EMT model could be reversed by LIMK1 silencing or miR-374c-5p overexpression. These results suggest that exo-miR-374c-5p suppresses EMT via targeting LIMK1-Wnt/β-catenin axis and the axis is involved in TGF-β1 induced metastasis of HCC, thereby identifying miR-374c-5p as a potential target for HCC treatment.
Collapse
Affiliation(s)
- Bisha Ding
- Cancer Center, Department of Medical Oncology, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Zhejiang, Hangzhou, China
| | - Weiyang Lou
- Department of Breast Surgery, The First Affiliated Hospital, College of Medicine, Zhejiang University, Zhejiang, Hangzhou, China
| | - Weimin Fan
- College of Medicine, Zhejiang University, Zhejiang, Hangzhou, China
| | - Jie Pan
- School of Basic Medical Sciences and Forensic Medicine, Hangzhou Medical College, Zhejiang, Hangzhou, China
| |
Collapse
|
15
|
Shi Y, Niu Y, Yuan Y, Li K, Zhong C, Qiu Z, Li K, Lin Z, Yang Z, Zuo D, Qiu J, He W, Wang C, Liao Y, Wang G, Yuan Y, Li B. PRMT3-mediated arginine methylation of IGF2BP1 promotes oxaliplatin resistance in liver cancer. Nat Commun 2023; 14:1932. [PMID: 37024475 PMCID: PMC10079833 DOI: 10.1038/s41467-023-37542-5] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Accepted: 03/22/2023] [Indexed: 04/08/2023] Open
Abstract
Although oxaliplatin-based chemotherapy has been effective in the treatment of hepatocellular carcinoma (HCC), primary or acquired resistance to oxaliplatin remains a major challenge in the clinic. Through functional screening using CRISPR/Cas9 activation library, transcriptomic profiling of clinical samples, and functional validation in vitro and in vivo, we identify PRMT3 as a key driver of oxaliplatin resistance. Mechanistically, PRMT3-mediated oxaliplatin-resistance is in part dependent on the methylation of IGF2BP1 at R452, which is critical for the function of IGF2BP1 in stabilizing the mRNA of HEG1, an effector of PRMT3-IGF2BP1 axis. Also, PRMT3 overexpression may serve as a biomarker for oxaliplatin resistance in HCC patients. Collectively, our study defines the PRTM3-IGF2BP1-HEG1 axis as important regulators and therapeutic targets in oxaliplatin-resistance and suggests the potential to use PRMT3 expression level in pretreatment biopsy as a biomarker for oxaliplatin-resistance in HCC patients.
Collapse
Affiliation(s)
- Yunxing Shi
- State Key Laboratory of Oncology in South China and Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
- Department of Liver Surgery, Sun Yat-Sen University Cancer Center, Guangzhou, China
| | - Yi Niu
- State Key Laboratory of Oncology in South China and Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
- Department of Liver Surgery, Sun Yat-Sen University Cancer Center, Guangzhou, China
| | - Yichuan Yuan
- State Key Laboratory of Oncology in South China and Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
- Department of Liver Surgery, Sun Yat-Sen University Cancer Center, Guangzhou, China
| | - Kai Li
- State Key Laboratory of Oncology in South China and Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
- Department of Liver Surgery, Sun Yat-Sen University Cancer Center, Guangzhou, China
| | - Chengrui Zhong
- State Key Laboratory of Oncology in South China and Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
- Department of Liver Surgery, Sun Yat-Sen University Cancer Center, Guangzhou, China
| | - Zhiyu Qiu
- State Key Laboratory of Oncology in South China and Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
- Department of Liver Surgery, Sun Yat-Sen University Cancer Center, Guangzhou, China
| | - Keren Li
- State Key Laboratory of Oncology in South China and Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
- Department of Liver Surgery, Sun Yat-Sen University Cancer Center, Guangzhou, China
| | - Zhu Lin
- State Key Laboratory of Oncology in South China and Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
- Department of Liver Surgery, Sun Yat-Sen University Cancer Center, Guangzhou, China
| | - Zhiwen Yang
- State Key Laboratory of Oncology in South China and Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Dinglan Zuo
- State Key Laboratory of Oncology in South China and Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Jiliang Qiu
- State Key Laboratory of Oncology in South China and Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
- Department of Liver Surgery, Sun Yat-Sen University Cancer Center, Guangzhou, China
| | - Wei He
- State Key Laboratory of Oncology in South China and Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
- Department of Liver Surgery, Sun Yat-Sen University Cancer Center, Guangzhou, China
| | - Chenwei Wang
- State Key Laboratory of Oncology in South China and Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
- Department of Liver Surgery, Sun Yat-Sen University Cancer Center, Guangzhou, China
| | - Yadi Liao
- State Key Laboratory of Oncology in South China and Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Guocan Wang
- Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| | - Yunfei Yuan
- State Key Laboratory of Oncology in South China and Collaborative Innovation Center for Cancer Medicine, Guangzhou, China.
- Department of Liver Surgery, Sun Yat-Sen University Cancer Center, Guangzhou, China.
| | - Binkui Li
- State Key Laboratory of Oncology in South China and Collaborative Innovation Center for Cancer Medicine, Guangzhou, China.
- Department of Liver Surgery, Sun Yat-Sen University Cancer Center, Guangzhou, China.
| |
Collapse
|
16
|
Xu H, Liu J, Zhang Y, Zhou Y, Zhang L, Kang J, Ning C, He Z, Song S. KIF23, under regulation by androgen receptor, contributes to nasopharyngeal carcinoma deterioration by activating the Wnt/β-catenin signaling pathway. Funct Integr Genomics 2023; 23:116. [PMID: 37010644 DOI: 10.1007/s10142-023-01044-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2023] [Revised: 03/22/2023] [Accepted: 03/27/2023] [Indexed: 04/04/2023]
Abstract
Our study aimed to explore the potential mechanisms of KIF23 regulating function in the progression of nasopharyngeal carcinoma and pinpoint novel therapeutic targets for the clinical treatment of nasopharyngeal carcinoma patients. Firstly, the mRNA and protein level of KIF23 in nasopharyngeal carcinoma was measured using quantitative real-time PCR and western blot. Then, the influence of KIF23 on tumor metastasis and growth in nasopharyngeal carcinoma was determined through the in vivo and in vitro experiments. Lastly, the regulatory mechanisms of KIF23 in nasopharyngeal carcinoma were illustrated in the chromatin immunoprecipitation assay. KIF23 was first found to be overexpressed in nasopharyngeal carcinoma samples, and its expression was associated with poor prognosis. Then, the nasopharyngeal carcinoma cell's proliferation, migration, and invasion potential could be improved by inducing KIF23 expression both in vivo and in vitro. Furthermore, androgen receptor (AR) was found to bind to the KIF23 promoter region directly and enhance KIF23 transcription. At last, KIF23 could accelerate nasopharyngeal carcinoma deterioration via activating the Wnt/β-catenin signaling pathway. AR/KIF23/Wnt/β-catenin pathway promotes nasopharyngeal carcinoma deterioration. Our findings could serve as a new therapeutic strategy for nasopharyngeal carcinoma in the clinical practice.
Collapse
Affiliation(s)
- Hongbo Xu
- Department of Radiation Oncology, The First Affiliated Hospital of Bengbu Medical College, No.287, Changhuai Road, Longzihu District, Bengbu, 233004, Anhui, China
- Anhui Province Key Laboratory of Translational Cancer Research Affiliated to Bengbu Medical College, Bengbu, Anhui, China
| | - Jingjing Liu
- Department of Radiation Oncology, The First Affiliated Hospital of Bengbu Medical College, No.287, Changhuai Road, Longzihu District, Bengbu, 233004, Anhui, China
| | - Yajun Zhang
- Department of Radiation Oncology, The First Affiliated Hospital of Bengbu Medical College, No.287, Changhuai Road, Longzihu District, Bengbu, 233004, Anhui, China
| | - Yan Zhou
- Department of Radiation Oncology, The First Affiliated Hospital of Bengbu Medical College, No.287, Changhuai Road, Longzihu District, Bengbu, 233004, Anhui, China
| | - Lei Zhang
- Department of Radiation Oncology, The First Affiliated Hospital of Bengbu Medical College, No.287, Changhuai Road, Longzihu District, Bengbu, 233004, Anhui, China
| | - Jia Kang
- Department of Radiation Oncology, The First Affiliated Hospital of Bengbu Medical College, No.287, Changhuai Road, Longzihu District, Bengbu, 233004, Anhui, China
| | - Can Ning
- Department of Radiation Oncology, The First Affiliated Hospital of Bengbu Medical College, No.287, Changhuai Road, Longzihu District, Bengbu, 233004, Anhui, China
| | - Zelai He
- Department of Radiation Oncology, The First Affiliated Hospital of Bengbu Medical College, No.287, Changhuai Road, Longzihu District, Bengbu, 233004, Anhui, China.
- Anhui Province Key Laboratory of Translational Cancer Research Affiliated to Bengbu Medical College, Bengbu, Anhui, China.
| | - Shilong Song
- Department of Radiation Oncology, The First Affiliated Hospital of Bengbu Medical College, No.287, Changhuai Road, Longzihu District, Bengbu, 233004, Anhui, China.
- Anhui Province Key Laboratory of Translational Cancer Research Affiliated to Bengbu Medical College, Bengbu, Anhui, China.
| |
Collapse
|
17
|
Hu P, Zong B, Chen Q, Shao R, Chen M, Yang Y, Shao G. Microtubule-associated protein 4 promotes epithelial mesenchymal transition in hepatocellular cancer cells via regulating GSK3β/β-catenin pathway. Heliyon 2023; 9:e14309. [PMID: 36938447 PMCID: PMC10020083 DOI: 10.1016/j.heliyon.2023.e14309] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Revised: 02/23/2023] [Accepted: 02/28/2023] [Indexed: 03/08/2023] Open
Abstract
Metastasis is a major obstacle in the treatment of hepatocellular carcinoma (HCC). Microtubule-associated protein 4 (MAP4) plays an important role as a coordinator between microtubules and microfilaments. However, the role of MAP4 in HCC migration and epithelial mesenchymal transition (EMT) is unclear. We compared the protein and mRNA levels of MAP4 in human HCC and adjacent normal tissues using western blotting, immunohistochemistry and RT-qPCR. The migration and invasion abilities and the levels of EMT markers (E-Cadherin, N-Cadherin, Vimentin, and Snail) were compared between MAP4-knockdown and MAP4-overexpressed HCC cells. Finally, we examined whether β-catenin and glycogen synthase kinase 3β (GSK3β) are involved in the stimulatory effects of MAP4 on HCC migration, invasion and EMT. The results revealed that MAP4 levels were higher in the HCC tissues than in the normal hepatic tissues. More importantly, MAP4 knockdown suppressed migration and invasion abilities and EMT processes in HCC cells, which were confirmed by the stimulatory effects of MAP4 overexpression on EMT processes in HCC cells. Further evidence demonstrated that the up-regulation of β-catenin activity induced by the interaction between MAP4 and GSK3β possibly accounted for the pro-migration and pro-EMT effects of MAP4 on HCC cells. Taken together, these results suggest that MAP4 promotes migration, invasion, and EMT in HCC cells by regulating the GSK3β/β-catenin pathway.
Collapse
Affiliation(s)
- Pingping Hu
- Department of Pathology, Zhenjiang Hospital of Chinese Traditional and Western Medicine, Affiliated to Jiangsu University, Zhenjiang, China
- Corresponding author.
| | - Bin Zong
- Department of Rehabilitation Medicine, Zhenjiang Hospital of Chinese Traditional and Western Medicine, Affiliated to Jiangsu University, Zhenjiang, China
| | - Qian Chen
- School of Medicine, Jiangsu University, Zhenjiang, China
| | - Rui Shao
- Department of Pathology, Zhenjiang First People’s Hospital, Affiliated to Jiangsu University, Zhenjiang, China
| | - Miao Chen
- Department of Pathology, Zhenjiang First People’s Hospital, Affiliated to Jiangsu University, Zhenjiang, China
| | - Yujie Yang
- School of Medicine, Jiangsu University, Zhenjiang, China
| | - Genbao Shao
- School of Medicine, Jiangsu University, Zhenjiang, China
| |
Collapse
|
18
|
TSUJI S, IMAI K. Medical application of the monoclonal antibody SKM9-2 against sialylated HEG1, a new precision marker for malignant mesothelioma. PROCEEDINGS OF THE JAPAN ACADEMY. SERIES B, PHYSICAL AND BIOLOGICAL SCIENCES 2023; 99:39-47. [PMID: 36775341 PMCID: PMC10020423 DOI: 10.2183/pjab.99.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/14/2022] [Accepted: 11/28/2022] [Indexed: 06/18/2023]
Abstract
Malignant pleural mesothelioma (MPM) is an aggressive tumor of the pleural cavity. Pathologically distinguishing MPM from other pleural lesions is often difficult. We searched for marker antigens to facilitate the pathological diagnosis of MPM and found useful markers for the pathological detection of malignant mesothelioma. Among them, the anti-mesothelioma monoclonal antibody SKM9-2, which was isolated as a clone binding to specimens of MPM (but not to specimens of lung adenocarcinoma) by immunohistochemical screening, showed higher specificity and sensitivity than traditional mesothelioma markers. SKM9-2 recognizes both sialylated O-glycans and peptide sequences in HEG1, and its glycan modifications are specific to mesothelioma. New effective treatments for MPM are needed because the prognosis of patients with MPM is usually poor. SKM9-2 can be used as a seed for next-generation antibody drugs with strong cytotoxic activities. In this review, we have summarized our research on antibody development for MPM diagnosis and treatment.
Collapse
Affiliation(s)
- Shoutaro TSUJI
- Department of Medical Technology & Clinical Engineering, Gunma University of Health and Welfare, Maebashi, Gunma, Japan
| | - Kohzoh IMAI
- Institute for Genetic Medicine, Hokkaido University, Sapporo, Hokkaido, Japan
| |
Collapse
|
19
|
Deng H, Chen G, Zhang J. β-Elemene regulates epithelial-mesenchymal transformation and inhibits invasion and metastasis of colorectal cancer cells. JOURNAL OF COMPLEMENTARY & INTEGRATIVE MEDICINE 2022:jcim-2022-0295. [PMID: 36480470 DOI: 10.1515/jcim-2022-0295] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Accepted: 11/29/2022] [Indexed: 05/03/2023]
Abstract
OBJECTIVES To study the inhibitory effect of β-elemene on invasion and metastasis of colorectal cancer cells and its possible mechanism. METHODS Human colon cancer HCT116 cells were treated with different concentrations of β-elemene. The proliferation inhibition rate of the cells was detected by MTT assay, cell migration rate was detected by scratched assay, and cell invasion rate was evaluated by Transwell cell invasion assay. The expressions of Vimentin, E-cadherin, N-cadherin, and β-catenin were detected by Western blotting. The mRNA expressions of Vimentin, E-cadherin, N-cadherin, and β-catenin were detected by real-time PCR. RESULTS Compared with the control group, the expressions of migration rate, invasion rate, scratch healing rate, N-cadherin, and Vimentin protein of HCT116 cells were decreased after β-elemene treatment, while the expression of E-cadherin protein was increased, and the inhibition rate of cell proliferation was increased (p<0.05). CONCLUSIONS β-Elemene may inhibit cell proliferation and invasion and metastasis by inhibiting EMT signaling pathway in human colon cancer cell line HCT116.
Collapse
Affiliation(s)
- Heng Deng
- Department of Proctology, Second Hospital Affiliated Anhui University of Traditional Chinese Medicine, Hefei, China
| | - Guo Chen
- Graduate Department, Anhui University of Chinese Medicine, Hefei, China
| | - Jun Zhang
- Chinese Medicine Teaching and Research Section, Anhui University of Chinese Medicine, Hefei, China
| |
Collapse
|
20
|
Li Z, Yang HY, Zhang XL, Zhang X, Huang YZ, Dai XY, Shi L, Zhou GR, Wei JF, Ding Q. Kinesin family member 23, regulated by FOXM1, promotes triple negative breast cancer progression via activating Wnt/β-catenin pathway. J Exp Clin Cancer Res 2022; 41:168. [PMID: 35524313 PMCID: PMC9077852 DOI: 10.1186/s13046-022-02373-7] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2022] [Accepted: 04/25/2022] [Indexed: 11/13/2022] Open
Abstract
Background Triple negative breast cancer (TNBC) is highly malignant and has a worse prognosis, compared with other subtypes of breast cancer due to the absence of therapeutic targets. KIF23 plays a crucial role in the tumorigenesis and cancer progression. However, the role of KIF23 in development of TNBC and the underlying mechanism remain unknown. The study aimed to elucidate the biological function and regulatory mechanism of KIF23 in TNBC. Methods Quantitative real-time PCR and Western blot were used to determine the KIF23 expression in breast cancer tissues and cell lines. Then, functional experiments in vitro and in vivo were performed to investigate the effects of KIF23 on tumor growth and metastasis in TNBC. Chromatin immunoprecipitation assay was conducted to illustrate the potential regulatory mechanisms of KIF23 in TNBC. Results We found that KIF23 was significantly up-regulated and associated with poor prognosis in TNBC. KIF23 could promote TNBC proliferation, migration and invasion in vitro and in vivo. KIF23 could activate Wnt/β-catenin pathway and promote EMT progression in TNBC. In addition, FOXM1, upregulated by WDR5 via H3K4me3 modification, directly bound to the promoter of KIF23 gene to promote its transcription and accelerated TNBC progression via Wnt/β-catenin pathway. Both of small inhibitor of FOXM1 and WDR5 could inhibit TNBC progression. Conclusions Our findings elucidate WDR5/FOXM1/KIF23/Wnt/β-catenin axis is associated with TNBC progression and may provide a novel and promising therapeutic target for TNBC treatment. Supplementary Information The online version contains supplementary material available at 10.1186/s13046-022-02373-7.
Collapse
|
21
|
Li R, Shang R, Li S, Ren Y, Shen L, Yang L, Chen S, Chen X, Li J, Xu M. LOXL3-promoted hepatocellular carcinoma progression via promotion of Snail1/USP4-mediated epithelial-mesenchymal transition. ENVIRONMENTAL TOXICOLOGY 2022; 37:2540-2551. [PMID: 35841383 DOI: 10.1002/tox.23617] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/17/2022] [Revised: 06/19/2022] [Accepted: 07/03/2022] [Indexed: 06/15/2023]
Abstract
Lysyl-oxidase-like 3 (LOXL3) was reported to be essential in epithelial-mesenchymal transition (EMT) of cancers. However, the role of LOXL3 in hepatocellular carcinoma (HCC) remained unclear. In this study, we explored clinical significance, biological functions, and regulatory mechanisms of LOXL3 in HCC. Our study found that LOXL3 expression was markedly associated with the tumor size and clinical stage of HCC, and it was highly expressed in tumor tissues of metastatic HCC patients. High expression of LOXL3 predicted a poor prognosis of HCC. TGF-β1 treatment elevated LOXL3 protein expression and cell invasion, and reduced cell apoptosis in HCC cell lines (SMMC-7721 and Huh-7), while downregulation of LOXL3 reversed the promotive effects of TGF-β1 treatment on LOXL3 protein expression and cell invasion, and the inhibitory effect on cell apoptosis. Mechanistically, LOXL3 interacted with snail family transcriptional repressor 1 (Snail1) through STRING database and RIP assay, and Snail1 bound to ubiquitin-specific peptidase 4 (USP4) promoter by JASPAR database, luciferase reporter gene and Co-IP assays. Overexpression of USP4 reversed the inhibitory effect of LOXL3 silence on EMT in HCC cells through deubiquitinating and stabilizing the expression of Snail1. Moreover, LOXL3-promoted HCC EMT through Wnt/β-catenin/Snail1 signaling pathway. In vivo study revealed that silence of LOXL3-inhibited HCC tumor growth. In conclusion, LOXL3 silence inhibited HCC invasion and EMT through Snail1/USP4-mediated circulation loop and Wnt/β-catenin signaling pathway.
Collapse
Affiliation(s)
- Rong Li
- Department of Anesthesiology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Runze Shang
- Department of General Surgery, Affiliated Haixia Hospital of Huaqiao University (The 910 Hospital), Quanzhou, China
| | - Shunle Li
- Department of General Surgery, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Yifan Ren
- Department of General Surgery, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Lin Shen
- Department of Gastroenterology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Longbao Yang
- Department of Gastroenterology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Shuo Chen
- Department of General Surgery, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Xi Chen
- Department of General Surgery, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Junhui Li
- Department of General Surgery, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Meng Xu
- Department of General Surgery, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| |
Collapse
|
22
|
CMaf-Inducing Protein Promotes LUAD Proliferation and Metastasis by Activating the MAPK/ERK Pathway. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2022; 2022:2501846. [PMID: 36159575 PMCID: PMC9492370 DOI: 10.1155/2022/2501846] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Revised: 08/19/2022] [Accepted: 08/25/2022] [Indexed: 11/30/2022]
Abstract
Objective Previous studies have shown that cMaf-inducing protein (CMIP) promotes tumorigenesis and progression, however, the role of CMIP in lung adenocarcinoma (LUAD) and its molecular mechanism remain unclear. Methods In this study, the Human Protein Atlas and Kaplan–Meier Plotter database were used to analyze the expression and prognostic value of CMIP in LUAD. Then, the expression levels of CMIP in LUAD tissues and cells were detected by qRT-PCR and western blot. The lentiviral vector was used to establish a stable transfected cell line, and the transfection efficiency was detected by qRT-PCR. MTT assay, colony formation assay, transwell assay, and wound healing assay were used to evaluate the function of CMIP in LUAD. In addition, the effect of CMIP on the MAPK/ERK pathway in LUAD cells was analyzed by western blot. Results The expression level of CMIP was significantly increased in LUAD cell and tissue samples, and the high expression of CMIP was associated with overall survival (OS) and progression-free survival (PFS) in LUAD patients. In vitro experiments showed that CMIP overexpression significantly promoted the proliferation, migration, and invasion of A549 cells. CMIP knockout significantly inhibited the proliferation, migration, and invasion of H1299 cells. In addition, it was observed that the expression levels of the MAPK/ERK pathway-related proteins were significantly increased in CMIP-overexpressed A549 cells, and promoted cell proliferation, migration, and invasion, while U0126 could significantly reverse the activation of the MAPK/ERK pathway by CMIP overexpression, and inhibit the proliferation, migration, and invasion of A549 cells. Conclusion Our study shows that CMIP, as an oncogene, is associated with poor patient prognosis, and may promote the proliferation and metastasis of LUAD by activating the MAPK/ERK pathway. Therefore, CMIP may be a new potential therapeutic target for LUAD.
Collapse
|
23
|
Yu H, Liu Y, Wang Y, Li Y, Sun J, Liu L. Circ_0005397 enhances hepatocellular carcinoma progression through miR-1283/HEG1. Ann Hepatol 2022; 27:100712. [PMID: 35500803 DOI: 10.1016/j.aohep.2022.100712] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Revised: 04/07/2022] [Accepted: 04/07/2022] [Indexed: 02/04/2023]
Abstract
INTRODUCTION AND OBJECTIVES Circular RNA (circRNA) has been confirmed to be an important regulator for the progression of hepatocellular carcinoma (HCC). However, the role and regulatory mechanism of circ_0005397 in HCC are not completely clear. PATIENTS AND METHODS Fifty HCC patients were included in this study. Reverse transcription-qPCR analysis was used to appraise circ_0005397, microRNA (miR)-1283, HEG homolog 1 (HEG1) mRNA expression levels, while western blot was used to identify HEG1, PCNA, Bax and Bcl-2 protein expression levels. Furthermore, cell proliferation, apoptosis, migration, invasion and angiogenesis were judged through cell counting kit-8 assay, EdU assay, Caspase3 activity test, flow cytometry, transwell assay and tube formation experiment. Dual-luciferase reporter assay and RIP assay were used to verify the targeting relationship between miR-1283 and circ_0005397 or HEG1. Finally, the effect of circ_0005397 on HCC tumor development was detected by mice experiments in vivo. RESULTS Circ_0005397 was highly expressed in HCC tissues and cells, in HCC cell lines. Circ_0005397 silencing inhibited proliferation, migration, invasion and angiogenesis, while induced apoptosis in HCC cells. Circ_0005397 could sponge miR-1283, and miR-1283 could target HEG1. MiR-1283 inhibitor incompletely counteracted the effect of si-circ_0005397 on HCC cell progression, while HEG1 overexpression partially overturned the effect of miR-1283 on HCC cell progression. Circ_0005397 regulated the expression of HEG1 through targeting miR-1283. Moreover, circ_0005397 silencing blocked the growth of HCC tumors in vivo. CONCLUSIONS Circ_0005397 regulated HEG1 by targeting miR-1283, thereby promoting HCC development.
Collapse
Affiliation(s)
- Haifeng Yu
- Department of Hepatology, Yantai Qishan Hospital, Yantai 264001, China
| | - Youde Liu
- Department of Hepatology, Yantai Qishan Hospital, Yantai 264001, China
| | - Yanna Wang
- Department of Hepatology, Yantai Qishan Hospital, Yantai 264001, China
| | - Yanfang Li
- Department of Hepatology, Yantai Qishan Hospital, Yantai 264001, China
| | - Jing Sun
- Department of Hepatology, Yantai Qishan Hospital, Yantai 264001, China
| | - Lijuan Liu
- Department of Nuclear Medicine, Yantai Yuhuangding Hospital, No. 20, Yuhuangding East Road, Zhifu District, Yantai 264000, China.
| |
Collapse
|
24
|
Zou X, Zhang Y, Wang N, Shi J, Li Q, Hao W, Zhu W, Han W. HEG1 as a novel potential biomarker for the prognosis of lung adenocarcinoma. Cancer Med 2022; 12:3288-3298. [PMID: 35950222 PMCID: PMC9939152 DOI: 10.1002/cam4.5081] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Revised: 05/15/2022] [Accepted: 07/03/2022] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Heart development protein with EGF-like domains 1 (HEG1), generally related to angiogenesis and embryonic development, was reported to participate in the occurrence and progression of some tumors recently. However, the role of HEG1 in lung adenocarcinoma (LUAD) is unclear. PATIENTS AND METHODS To explore the effect of HEG1 on LUAD, GEPIA platform and UALCAN database, as well as Kaplan-Meier plotter were adopted to analyze the association of HEG1 with clinicopathological characteristics and survival outcomes for LUAD firstly. And then the HEG1 in LUAD tissues, blood and cell lines were detected by qRT-PCR, western blot, immunofluorescence, immunohistochemistry, and ELISA. Gene set enrichment analysis (GSEA) was conducted to identify pathways that might be affected by HEG1 in LUAD. RESULTS In this study, HEG1 in lung tissues and cell lines of LUAD were significantly downregulated compared to benign pulmonary disease tissues and alveolar epithelial cells (p < 0.05). Moreover, compared with other groups, patients with advanced tumor stage had lower HEG1 mRNA expression levels (p = 0.025), which were negatively correlated with Ki67 index in tumor tissues (r = -0.427, p = 0.033). On the other hand, the LUAD patients with lower HEG1 had shorter overall survival (OS) (HR = 0.51, 95% CI: 0.40-0.65, p < 0.001) according to Kaplan-Meier plotter. In addition, HEG1 in serum of LUAD patients was negatively associated with CEA (r = -0.636, p < 0.001). GSEA showed that HEG1 was enriched in various metabolic-related pathways, including glucose metabolism, lipid metabolism, and nucleotide metabolism signaling. CONCLUSIONS HEG1 was downregulated in LUAD patients and associated with poor prognosis, which indicating HEG1 may serve as a potential biomarker for diagnosis and prognosis of LUAD.
Collapse
Affiliation(s)
- Xin Zou
- Department of Pathology, Qingdao Municipal HospitalDalian Medical UniversityQingdaoChina,Respiratory Disease Key Laboratory of QingdaoQingdao Municipal HospitalQingdaoChina
| | - Yue Zhang
- Department of RespiratoryJilin Provincial People's HospitalJilinChina
| | - Ning Wang
- Department of Pulmonary and Critical Care Medicine, Qingdao Municipal Hospital, School of MedicineQingdao UniversityQingdaoChina
| | - Jie Shi
- NMPA Key Laboratory for Quality Research and Evaluation of Traditional Marine Chinese MedicineQingdaoChina
| | - Qinghai Li
- Respiratory Disease Key Laboratory of QingdaoQingdao Municipal HospitalQingdaoChina,Department of Pulmonary and Critical Care Medicine, Qingdao Municipal Hospital, School of MedicineQingdao UniversityQingdaoChina
| | - Wanming Hao
- Department of Pulmonary and Critical Care Medicine, Qingdao Municipal Hospital, School of MedicineQingdao UniversityQingdaoChina
| | - Wenjing Zhu
- Respiratory Disease Key Laboratory of QingdaoQingdao Municipal HospitalQingdaoChina,NMPA Key Laboratory for Quality Research and Evaluation of Traditional Marine Chinese MedicineQingdaoChina,Clinical Research Center, Qingdao Municipal HospitalQingdao UniversityQingdaoChina
| | - Wei Han
- Respiratory Disease Key Laboratory of QingdaoQingdao Municipal HospitalQingdaoChina,Department of Pulmonary and Critical Care Medicine, Qingdao Municipal Hospital, School of MedicineQingdao UniversityQingdaoChina
| |
Collapse
|
25
|
Li Y, Xu C, Sun B, Zhong F, Cao M, Yang L. Sema3d Restrained Hepatocellular Carcinoma Progression Through Inactivating Pi3k/Akt Signaling via Interaction With FLNA. Front Oncol 2022; 12:913498. [PMID: 35957887 PMCID: PMC9358705 DOI: 10.3389/fonc.2022.913498] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Accepted: 06/06/2022] [Indexed: 12/24/2022] Open
Abstract
Hepatocellular carcinoma (HCC) is one of the most lethal malignant tumors worldwide due to the high incidence rate of metastasis and recurrence. Semaphorin 3d (Sema3d) has been shown to play a critical role in vascular development during early embryogenesis and several forms of cancer progression via regulating cell migration. However, the function of Sema3d in hepatocellular carcinoma (HCC) remains elusive. This study aimed to explore the function and mechanisms of Sema3d in HCC. In our study, Sema3d expression was significantly downregulated in HCC tissues and cell lines. Downregulated Sema3d was closely correlated with aggressive clinicopathological features and poor clinical outcomes in HCC patients. Moreover, overexpression of Sema3d in HCCLM3 cells was significantly inhibited and knockdown of Sema3d in PLC/PRF/5 cells promoted proliferation, migration, invasion, and epithelial–mesenchymal transition (EMT) of HCC cells in vitro and tumor growth, EMT, and metastasis in vivo. Furthermore, the RNA sequencing and gene set enrichment analysis (GSEA) indicated that these phenotypic and functional changes in Sema3d-interfered HCC cells were mediated by the Pi3k/Akt signaling pathway, and co-IP–combined mass spectrometry indicated Sema3d might interact with FLNA. Finally, we proved that Sema3d exerted its tumor-restraining effect by interacting with FLNA to inactivate the Pi3k/Akt signaling pathway and remodel the cytoskeleton. Our data showed that Sema3d restrained hepatocellular carcinoma proliferation, invasion, and metastasis through inactivating Pi3k/Akt via interaction with FLNA, which may serve as a novel prognostic predictor and a potential therapeutic target for HCC patients.
Collapse
|
26
|
Cao D, Liu H, Bai L, Tang H. EMT-Related Gene Signature: A New Method for Personalized Risk Assessment in Patients with Hepatocellular Carcinoma. Dig Dis 2022; 41:282-295. [PMID: 35636408 DOI: 10.1159/000525232] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Accepted: 05/16/2022] [Indexed: 02/02/2023]
Abstract
BACKGROUND Hepatocellular carcinoma (HCC) is the fourth leading cause of cancer-related death worldwide, accounting for more than 700,000 deaths annually. Epithelial-mesenchymal transition (EMT) is posited to contribute to HCC progression. We, therefore, aimed to elucidate the relationship between EMT-related gene (ERG) expression and prognosis in patients with HCC. METHODS Univariate and multivariate Cox regression analyses were performed to identify prognostic-related differentially expressed EMT-related genes (DE-ERGs). Time-dependent receiver operating characteristic and Kaplan-Meier curves were used to assess the prognostic performance of the EMT-related signature. pRRophetic R package was used to evaluate sorafenib sensitivity in the GSE14520 cohort. Gene expression in Huh7 and L02 cell lines was detected by real-time PCR and Western blotting. Differential expression of the three genes between HCC tissues and normal tissues was validated using immunohistochemical analyses. RESULTS Of the 76 identified DE-ERGs, 29 were associated with overall survival. Three prognosis-related ERGs (GOLM1, SOX4, and CD14) were screened out by multivariate Cox regression. A gene signature was identified based on the three prognostic-related ERGs. HCC patients with a low-risk score had a better prognosis and were more sensitive to sorafenib compared to those with a high-risk score. Moreover, we further confirmed increased expression of GOLM1 and SOX4, and decreased expression of CD14, in liver cancer cell line and HCC tissue. CONCLUSIONS The results of the present study demonstrate the utility of an ERG signature as a potential biomarker informing prognosis in patients with HCC, which may contribute to the implementation of personalized therapies.
Collapse
Affiliation(s)
- Dan Cao
- Center of Infectious Diseases, West China Hospital, Sichuan University, Chengdu, China, .,Division of Infectious Diseases, State Key Laboratory of Biotherapy, Sichuan University, Chengdu, China,
| | - Huan Liu
- Center of Infectious Diseases, West China Hospital, Sichuan University, Chengdu, China.,Division of Infectious Diseases, State Key Laboratory of Biotherapy, Sichuan University, Chengdu, China
| | - Lang Bai
- Center of Infectious Diseases, West China Hospital, Sichuan University, Chengdu, China.,Division of Infectious Diseases, State Key Laboratory of Biotherapy, Sichuan University, Chengdu, China
| | - Hong Tang
- Center of Infectious Diseases, West China Hospital, Sichuan University, Chengdu, China.,Division of Infectious Diseases, State Key Laboratory of Biotherapy, Sichuan University, Chengdu, China
| |
Collapse
|
27
|
Li YM, Xu C, Sun B, Zhong FJ, Cao M, Yang LY. Piezo1 promoted hepatocellular carcinoma progression and EMT through activating TGF-β signaling by recruiting Rab5c. Cancer Cell Int 2022; 22:162. [PMID: 35461277 PMCID: PMC9035260 DOI: 10.1186/s12935-022-02574-2] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Accepted: 04/06/2022] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Piezo1 has been revealed to play a regulatory role in vascular development and progression of variety tumors. However, whether and how the progression of hepatocellular carcinoma (HCC) regulated by Piezo1 remains elusive. This study aimed to elucidate the effect and mechanisms of Piezo1 in HCC. METHODS The mRNA and protein expression level of Piezo1 in HCC samples and cell lines was determined by qRT-PCR, western blot and immunohistochemistry analyses. Two independent study cohorts containing 280 patients were analyzed to reveal the association between Piezo1 expression and clinicopathological characteristics. Series of in vitro and in vivo experiments were used to validate the function of Piezo1 in HCC. Gene set enrichment analysis (GSEA) was performed to explore the signaling pathway of Piezo1. Immunoprecipitation, immunofluorescence and in vitro and in vivo experiments were used to explore the molecular mechanism of Piezo1 in HCC progression. RESULTS Our results demonstrated the Piezo1 expression was significantly upregulated in HCC tissues and cell lines, and upregulation of Piezo1 closely correlated with aggressive clinicopathological features and poor prognosis. Knockdown of Piezo1 in HCCLM3 and Hep3B cells significantly restrained proliferation, migration, invasion and epithelial-mesenchymal transition (EMT) of HCC cells in vitro, and tumor growth, metastasis, EMT in vivo. TGF-β signaling pathway was most significant enriched pathway in GSEA. Finally, tumor promotion effect of Piezo1 was found to exerted through recruiting and combining Rab5c to activating TGF-β signaling pathway. CONCLUSIONS Piezo1 significantly related to poor prognosis and promotes progression of hepatocellular carcinoma via activating TGF-β signaling, which suggesting that Piezo1 may serve as a novel prognostic predictor and the potential therapeutic target for HCC patients.
Collapse
Affiliation(s)
- Yi-ming Li
- Liver Cancer Laboratory, Department of Surgery, Xiangya Hospital, Central South University, Xiangya Road 87, Changsha, 410008 Hunan China
| | - Cong Xu
- Liver Cancer Laboratory, Department of Surgery, Xiangya Hospital, Central South University, Xiangya Road 87, Changsha, 410008 Hunan China
| | - Bo Sun
- Liver Cancer Laboratory, Department of Surgery, Xiangya Hospital, Central South University, Xiangya Road 87, Changsha, 410008 Hunan China
| | - Fang-jing Zhong
- Liver Cancer Laboratory, Department of Surgery, Xiangya Hospital, Central South University, Xiangya Road 87, Changsha, 410008 Hunan China
| | - Momo Cao
- Liver Cancer Laboratory, Department of Surgery, Xiangya Hospital, Central South University, Xiangya Road 87, Changsha, 410008 Hunan China
| | - Lian-yue Yang
- Liver Cancer Laboratory, Department of Surgery, Xiangya Hospital, Central South University, Xiangya Road 87, Changsha, 410008 Hunan China
| |
Collapse
|
28
|
Li W, Pei S, Zhang X, Qi D, Zhang W, Dou Y, Yang R, Yao X, Zhang Z, Xie S, Fang D, Sun H. Cinobufotalin inhibits the epithelial-mesenchymal transition of hepatocellular carcinoma cells through down-regulate β-catenin in vitro and in vivo. Eur J Pharmacol 2022; 922:174886. [DOI: 10.1016/j.ejphar.2022.174886] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2021] [Revised: 03/03/2022] [Accepted: 03/09/2022] [Indexed: 02/07/2023]
|
29
|
Zhu S, Rao X, Qian Y, Chen J, Song R, Yan H, Yang X, Hu J, Wang X, Han Z, Zhu Y, Liu R, Jong-Leong Wong J, McCaughan GW, Zheng X. Liver Endothelial Heg Regulates Vascular/Biliary Network Patterning and Metabolic Zonation Via Wnt Signaling. Cell Mol Gastroenterol Hepatol 2022; 13:1757-1783. [PMID: 35202885 PMCID: PMC9059100 DOI: 10.1016/j.jcmgh.2022.02.010] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/04/2021] [Revised: 02/11/2022] [Accepted: 02/14/2022] [Indexed: 12/18/2022]
Abstract
BACKGROUND & AIMS The liver has complex interconnecting blood vessel and biliary networks; however, how the vascular and biliary network form and regulate each other and liver function are not well-understood. We aimed to examine the role of Heg in mammalian liver development and functional maintenance. METHODS Global (Heg-/-) or liver endothelial cell (EC)-specific deletion of Heg (Lyve1-Cre;Hegfl/fl ) mice were used to study the in vivo function of Heg in the liver. Carbon-ink anterograde and retrograde injection were used to visualize the 3-dimensional patterning of liver portal and biliary networks, respectively. RNA sequencing, histology, and molecular and biochemical assays were used to assess liver gene expression, protein distribution, liver injury response, and function. RESULTS Heg deficiency in liver ECs led to a sparse liver vascular and biliary network. This network paucity does not compromise liver function under baseline conditions but did alter liver zonation. Molecular analysis revealed that endothelial Heg deficiency decreased expression of Wnt ligands/agonists including Wnt2, Wnt9b, and Rspo3 in ECs, which limits Axin2 mediated canonical Wnt signaling and the expression of cytochrome P450 enzymes in hepatocytes. Under chemical-induced stressed conditions, Heg-deficiency in liver ECs protected mice from drug-induced liver injuries. CONCLUSION Our study found that endothelial Heg is essential for the 3-D patterning of the liver vascular and indirectly regulates biliary networks and proper liver zonation via its regulation of Wnt ligand production in liver endothelial cells. The endothelial Heg-initiated changes of the liver metabolic zonation and metabolic enzyme expression in hepatocytes was functionally relevant to xenobiotic metabolism and drug induced liver toxicity.
Collapse
Affiliation(s)
- Shichao Zhu
- Department of Pharmacology and Tianjin Key Laboratory of Inflammation Biology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Xiyun Rao
- Department of Pharmacology and Tianjin Key Laboratory of Inflammation Biology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Yude Qian
- Department of Pharmacology and Tianjin Key Laboratory of Inflammation Biology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Jinbiao Chen
- Liver Injury and Cancer Program Centenary Institute and Sydney Medical School, The University of Sydney, A.W Morrow Gastroenterology and Liver Center, Royal Prince Alfred Hospital, Sydney, NSW, Australia
| | - Renhua Song
- Epigenetics and RNA Biology Program Centenary Institute and Sydney Medical School, The University of Sydney, Sydney, NSW, Australia
| | - Huili Yan
- Department of Pharmacology and Tianjin Key Laboratory of Inflammation Biology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Xi Yang
- Department of Pharmacology and Tianjin Key Laboratory of Inflammation Biology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Junhao Hu
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai, China
| | - Xiaohong Wang
- Department of Pharmacology and Tianjin Key Laboratory of Inflammation Biology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Zhiming Han
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | - Yi Zhu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Renjing Liu
- Vascular Epigenetics Laboratory, Victor Chang Cardiac Research Institute, Sydney, NSW, Australia
| | - Justin Jong-Leong Wong
- Epigenetics and RNA Biology Program Centenary Institute and Sydney Medical School, The University of Sydney, Sydney, NSW, Australia
| | - Geoffrey W. McCaughan
- Liver Injury and Cancer Program Centenary Institute and Sydney Medical School, The University of Sydney, A.W Morrow Gastroenterology and Liver Center, Royal Prince Alfred Hospital, Sydney, NSW, Australia
| | - Xiangjian Zheng
- Department of Pharmacology and Tianjin Key Laboratory of Inflammation Biology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China,Correspondence Address correspondence to: Dr Xiangjian Zheng, Pharmacology, Tianjin Medical University, No 22 Qi Xiang Tai Rd, Tianjin 300070, China. tel: 86-22-8333-6835.
| |
Collapse
|
30
|
Ruan Y, Xu H, Ji X. High expression of NPM1 via the Wnt/β-catenin signalling pathway might predict poor prognosis for patients with prostate adenocarcinoma. Clin Exp Pharmacol Physiol 2022; 49:525-535. [PMID: 35108408 DOI: 10.1111/1440-1681.13628] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2021] [Revised: 01/21/2022] [Accepted: 01/26/2022] [Indexed: 11/29/2022]
Abstract
Prostate adenocarcinoma (PRAD) occurs only in males and has a higher incidence rate than other cancers. NPM1 is a nucleocytoplasmic shuttling protein that participates in the development of multiple tumours. The aim of this research was to explore the effect of the upregulation or downregulation of the NPM1 protein on the malignancy of prostate cancer and its possible signalling pathway. Prostate adenocarcinoma cell lines were used in this study, including RWPE-1, PC3, LNCap, and 22RV1 cells. Our research revealed that NPM1 was widely expressed in the PRAD cell lines, as determined by Western blotting, and that the levels of NPM1 protein were positively correlated with the degree of malignancy of the PRAD cell lines. Through interference and overexpression experiments, we found that PC3 cells growth was inhibited after NPM1 knockdown and that this inhibition was partly reversed by CTNNB1 overexpression; in contrast, PC3 cells growth was promoted after NPM1 overexpression, and this promotion was partly reversed by CTNNB1 knockdown, suggesting that NPM1 and CTNNB1 play important roles in the progression of prostate cancer cells via the Wnt/β-catenin signalling pathway. NPM1 may serve as an important biomarker and candidate therapeutic for patients with prostate cancer.
Collapse
Affiliation(s)
- Yong Ruan
- Key Laboratory of Animal Genetics, Breeding and Reproduction in the Plateau Mountainous Region, Ministry of Education, Guiyang, 550025, China.,College of Animal Science, Guizhou University, Guiyang, 550025, China.,Medical College, Guizhou University, Guiyang, 550025, China
| | - Houqiang Xu
- Key Laboratory of Animal Genetics, Breeding and Reproduction in the Plateau Mountainous Region, Ministry of Education, Guiyang, 550025, China.,College of Animal Science, Guizhou University, Guiyang, 550025, China.,Medical College, Guizhou University, Guiyang, 550025, China
| | - Xinqin Ji
- College of Animal Science, Guizhou University, Guiyang, 550025, China.,Medical College, Guizhou University, Guiyang, 550025, China
| |
Collapse
|
31
|
Yang M, Ju L, Li C, Cheng H, Li N, Zhang Q, Sun S, Ding L, Sui X, Zhang C, Yang M, Cao J, Ding R. MiR-582-3p participates in the regulation of biological behaviors of A549 cells by ambient PM 2.5 exposure. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2022; 29:13624-13634. [PMID: 34590234 DOI: 10.1007/s11356-021-16801-2] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/25/2021] [Accepted: 09/24/2021] [Indexed: 06/13/2023]
Abstract
Ambient fine particulate matter (PM2.5) is one of the main environmental air pollutants that is closely related to the development of lung cancer, but the mechanisms are unclear. In this study, A549 cells were exposed to ambient PM2.5 to investigate the alterations of biological behaviors, and the possible role of miR-582-3p in the effects was further explored. The findings showed that PM2.5 exposure could significantly enhance the biological behaviors of A549 cells, and promote their epithelial-mesenchymal transition (EMT) transformation, especially at relatively low doses. Over-activation of Wnt/β-catenin signaling pathway and increased expression of miR-582-3p were also found in A549 cells after PM2.5 exposure. After the knockdown of miR-582-3p in A549 cells, the effects of PM2.5 on malignant biological behavior changes, EMT, and the activation of Wnt/β-catenin signaling pathway were all significantly alleviated. Furthermore, the inhibition of Wnt/β-catenin signaling pathway also inhibited the EMT process of A549 cells, which was rescued by the overexpression of miR-582-3p. Therefore, this study showed that ambient PM2.5 can upregulate the expression of miR-582-3p, consequently activate the Wnt/β-catenin signaling pathway, and thereby enhance EMT transformation and promote the malignant biological behaviors of A549 cells. These findings provide evidence for further research into the mechanisms by which exposure to PM2.5 in the environment promotes lung cancer.
Collapse
Affiliation(s)
- Mei Yang
- Department of Occupational Health and Environmental Health, School of Public Health, Anhui Medical University, 81 Meishan Road, Hefei, 230032, Anhui, China
- Department of Environmental Hygiene, Anhui Provincial Center for Disease Control and Prevention, Hefei, Anhui, China
| | - Liangliang Ju
- Department of Occupational Health and Environmental Health, School of Public Health, Anhui Medical University, 81 Meishan Road, Hefei, 230032, Anhui, China
| | - Changlian Li
- Department of Occupational Health and Environmental Health, School of Public Health, Anhui Medical University, 81 Meishan Road, Hefei, 230032, Anhui, China
| | - Han Cheng
- The First Clinical College of Anhui Medical University, Hefei, 230032, Anhui, China
| | - Na Li
- Department of Oncology, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Qi Zhang
- Department of Occupational Health and Environmental Health, School of Public Health, Anhui Medical University, 81 Meishan Road, Hefei, 230032, Anhui, China
| | - Shu Sun
- Department of Occupational Health and Environmental Health, School of Public Health, Anhui Medical University, 81 Meishan Road, Hefei, 230032, Anhui, China
| | - Liu Ding
- Department of Occupational Health and Environmental Health, School of Public Health, Anhui Medical University, 81 Meishan Road, Hefei, 230032, Anhui, China
| | - Xinmiao Sui
- Department of Occupational Health and Environmental Health, School of Public Health, Anhui Medical University, 81 Meishan Road, Hefei, 230032, Anhui, China
- Department of Environmental Hygiene, Anhui Provincial Center for Disease Control and Prevention, Hefei, Anhui, China
| | - Chao Zhang
- Department of Maternal, Child and Adolescent Health, School of Public Health, Anhui Medical University, Hefei, Anhui, China
| | - Mingwei Yang
- Department of Radiation Oncology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China.
| | - Jiyu Cao
- Department of Occupational Health and Environmental Health, School of Public Health, Anhui Medical University, 81 Meishan Road, Hefei, 230032, Anhui, China.
- Department of Teaching Center for Preventive Medicine, School of Public Health, Anhui Medical University, 81 Meishan Road, Hefei, 230032, Anhui, China.
| | - Rui Ding
- Department of Occupational Health and Environmental Health, School of Public Health, Anhui Medical University, 81 Meishan Road, Hefei, 230032, Anhui, China.
| |
Collapse
|
32
|
Proteomic Analysis of Human Serum for Patients at Different Pathological Stages of Hepatic Fibrosis. BIOMED RESEARCH INTERNATIONAL 2021; 2021:3580090. [PMID: 34877354 PMCID: PMC8645358 DOI: 10.1155/2021/3580090] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Accepted: 10/15/2021] [Indexed: 12/24/2022]
Abstract
Background Hepatic fibrosis is a severe liver disease that has threatened human health for a long time. In order to undergo timely and adequate therapy, it is important for patients to obtain an accurate diagnosis of fibrosis. Laboratory inspection methods have been efficient in distinguishing between advanced hepatic fibrosis stages (F3, F4), but the identification of early stages of fibrosis has not been achieved. The development of proteomics may provide us with a new direction to identify the stages of fibrosis. Methods We established serum proteomic maps for patients with hepatic fibrosis at different stages and identified differential expression of proteins between fibrosis stages through ultra-high-performance liquid chromatography tandem mass spectrometry proteomic analysis. Results From the proteomic profiles of the serum of patients with different stages of liver fibrosis, a total of 1,338 proteins were identified. Among three early fibrosis stages (control, F1, and F2), 55 differential proteins were identified, but no proteins simultaneously exhibited differential expression between control, F1, and F2. Differential proteins were detected in the comparison between different fibrosis stages. Significant differences were found between advanced fibrosis stages (F2-vs.-F3 and F3-vs.-F4) through a series of statistical analysis, including hierarchical clustering, Gene Ontology (GO) functional annotation, Kyoto Encyclopedia of Genes and Genomes pathway, and protein-protein interaction network analysis. The differential proteins identified by GO annotation were associated with biological processes (mainly platelet degranulation and cell adhesion), molecular functions, and cellular components. Conclusions All potential biomarkers identified between the stages of fibrosis could be key points in determining the fibrosis staging. The differences between early stages may provide a useful reference in addressing the challenge of early fibrosis staging.
Collapse
|
33
|
Resveratrol inhibits the migration, invasion and epithelial-mesenchymal transition in liver cancer cells through up- miR-186-5p expression. Zhejiang Da Xue Xue Bao Yi Xue Ban 2021; 50:582-590. [PMID: 34986537 DOI: 10.3724/zdxbyxb-2021-0197] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
To investigate the molecular mechanism of resveratrol inhibiting the metastasis of liver cancer . HepG2 and Huh7 cells were treated with different concentrations of resveratrol, and the cell viability was determined by CCK-8 assay to determine the optimal concentration of resveratrol for subsequent experiments. The expressions of miR-186-5p in liver cancer tissues and liver cancer cells were determined by quantitative real-time RT-PCR. The migration and invasion of HepG2 and Huh7 cells were detected by wound healing assay and Transwell assay, and the expression levels of epithelial-mesenchymal transition (EMT) related proteins were determined by Western blotting. Resveratrol with concentration of had no effect on the viability of HepG2 and Huh7 cells, so the concentration of resveratrol in subsequent experiments was 6.25 μmol/L. Resveratrol inhibited the wound healing and invasion of liver cancer cells; increased the expression of E-cadherin, and decreased the expression of vimentin and Twist1. The expression of miR-186-5p was significantly down-regulated in liver cancer tissues and cells compared with the adjacent tissues and normal liver cells (both <0.05). Furthermore, resveratrol induced the expression of miR-186-5p in liver cancer cells (both <0.01). Overexpression of miR-186-5p suppressed the migration, invasion and EMT of liver cancer cells. Knockdown of miR-186-5p blocked the inhibition effects of resveratrol on the migration, invasion and EMT of liver cancer cells. Resveratrol could inhibit the metastasis of liver cancer , which might be associated with up-regulating miR-186-5p.
Collapse
|
34
|
Abstract
Liver metastasis, originating either from a primary liver or other cancer types, represent a large cancer-related burden. Therefore, studies that add to better understanding of its molecular basis are needed. Herein, the role of the Wnt signaling pathway in liver metastasis is outlined. Its role in hepatocellular carcinoma (HCC) epithelial-mesenchymal transition (EMT), motility, migration, metastasis formation, and other steps of the metastatic cascade are presented. Additionally, the roles of the Wnt signaling pathway in the liver metastasis formation of colorectal, breast, gastric, lung, melanoma, pancreatic, and prostate cancer are explored. The special emphasis is given to the role of the Wnt signaling pathway in the communication between the many of the components of the primary and secondary cancer microenvironment that contribute to the metastatic outgrowth in the liver. The data presented herein are a review of the most recent publications and advances in the field that add to the idea that the Wnt pathway is among the drivers of liver metastasis and that its targeting could potentially relieve liver metastasis–related complications.
Collapse
|
35
|
Peng P, Song H, Xie C, Zheng W, Ma H, Xin D, Zhan J, Yuan X, Chen A, Tao J, Qin J. miR-146a-5p-mediated suppression on trophoblast cell progression and epithelial-mesenchymal transition in preeclampsia. Biol Res 2021; 54:30. [PMID: 34517910 PMCID: PMC8438983 DOI: 10.1186/s40659-021-00351-5] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2021] [Accepted: 08/24/2021] [Indexed: 12/29/2022] Open
Abstract
Objective This study aims to identify the effect of miR-146a-5p on trophoblast cell invasion as well as the mechanism in preeclampsia (PE). Methods Expression levels of miR-146a-5p and Wnt2 in preeclamptic and normal placentae were quantified. Trophoblast cells (HTR-8) were separately transfected with miR-146a-5p mimic, miR-146a-5p inhibitor, pcDNA3.1-Wnt2 or sh-Wnt2, and then the expression levels of miR-146a-5p, Wnt2, and epithelial-mesenchymal transition (EMT)-related proteins (Vimentin, N-cadherin and E-cadherin) were measured. Moreover, the proliferative, migratory and invasive capacities of trophoblast cells were detected, respectively. Dual luciferase reporter assay determined the binding of miR-146a-5p and Wnt2. Results Compared with normal placental tissues, the placentae from PE patients showed higher miR-146a-5p expression and lower Wnt2 expression. Transfection of miR-146a-5p inhibitor or pcDNA3.1-Wnt2 exerted pro-migratory and pro-invasive effects on HTR-8 cells and encouraged EMT in HTR-8 cells; transfection with miR-146a-5p mimic or sh-Wnt2 weakened the proliferative, migratory and invasive capacities as well as reduced EMT process of HTR-8 cells. Moreover, Wnt2 overexpression could partially counteract the suppressive effects of miR-146a-5p overexpression on the progression and EMT of HTR-8 cells. Conclusion miR-146a-5p mediates trophoblast cell proliferation and invasion through regulating Wnt2 expression.
Collapse
Affiliation(s)
- Pingping Peng
- Department of Gynecology and Obstetrics, the First People's Hospital of Yichang, Yichang, 443000, Hubei, People's Republic of China.,Department of Gynecology and Obstetrics, the People's Hospital of China Three Gorges University, Yichang, 443000, Hubei, People's Republic of China
| | - Huamei Song
- Department of Gynecology and Obstetrics, the First People's Hospital of Yichang, Yichang, 443000, Hubei, People's Republic of China.,Department of Gynecology and Obstetrics, the People's Hospital of China Three Gorges University, Yichang, 443000, Hubei, People's Republic of China
| | - Chenghong Xie
- Department of Gynecology and Obstetrics, the First People's Hospital of Yichang, Yichang, 443000, Hubei, People's Republic of China.,Department of Gynecology and Obstetrics, the People's Hospital of China Three Gorges University, Yichang, 443000, Hubei, People's Republic of China
| | - Wenfei Zheng
- Department of Gynecology and Obstetrics, the First People's Hospital of Yichang, Yichang, 443000, Hubei, People's Republic of China.,Department of Gynecology and Obstetrics, the People's Hospital of China Three Gorges University, Yichang, 443000, Hubei, People's Republic of China
| | - Huigai Ma
- Department of Gynecology and Obstetrics, the First People's Hospital of Yichang, Yichang, 443000, Hubei, People's Republic of China.,Department of Gynecology and Obstetrics, the People's Hospital of China Three Gorges University, Yichang, 443000, Hubei, People's Republic of China
| | - Dandan Xin
- Department of Gynecology and Obstetrics, the First People's Hospital of Yichang, Yichang, 443000, Hubei, People's Republic of China.,Department of Gynecology and Obstetrics, the People's Hospital of China Three Gorges University, Yichang, 443000, Hubei, People's Republic of China
| | - Jingqiong Zhan
- Department of Gynecology and Obstetrics, the First People's Hospital of Yichang, Yichang, 443000, Hubei, People's Republic of China.,Department of Gynecology and Obstetrics, the People's Hospital of China Three Gorges University, Yichang, 443000, Hubei, People's Republic of China
| | - Xiaoqing Yuan
- Department of Gynecology and Obstetrics, the First People's Hospital of Yichang, Yichang, 443000, Hubei, People's Republic of China.,Department of Gynecology and Obstetrics, the People's Hospital of China Three Gorges University, Yichang, 443000, Hubei, People's Republic of China
| | - Aihua Chen
- Department of Gynecology and Obstetrics, the First People's Hospital of Yichang, Yichang, 443000, Hubei, People's Republic of China.,Department of Gynecology and Obstetrics, the People's Hospital of China Three Gorges University, Yichang, 443000, Hubei, People's Republic of China
| | - Jing Tao
- Department of Gynecology and Obstetrics, the First People's Hospital of Yichang, Yichang, 443000, Hubei, People's Republic of China.,Department of Gynecology and Obstetrics, the People's Hospital of China Three Gorges University, Yichang, 443000, Hubei, People's Republic of China
| | - Jufang Qin
- Department of Gynecology and Obstetrics, the First People's Hospital of Yichang, Yichang, 443000, Hubei, People's Republic of China. .,Department of Gynecology and Obstetrics, the People's Hospital of China Three Gorges University, Yichang, 443000, Hubei, People's Republic of China. .,Department of Gynecology and Obstetrics, the First People's Hospital of Yichang, the People's Hospital of China Three Gorges University, No. 4, Hudi Street, Xiling District, Yichang, 443000, Hubei, People's Republic of China.
| |
Collapse
|
36
|
Zhang H, Yang X, Zhu L, Li Z, Zuo P, Wang P, Feng J, Mi Y, Zhang C, Xu Y, Jin G, Zhang J, Ye H. ASPM promotes hepatocellular carcinoma progression by activating Wnt/β-catenin signaling through antagonizing autophagy-mediated Dvl2 degradation. FEBS Open Bio 2021; 11:2784-2799. [PMID: 34428354 PMCID: PMC8487047 DOI: 10.1002/2211-5463.13278] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Revised: 07/30/2021] [Accepted: 08/23/2021] [Indexed: 12/23/2022] Open
Abstract
Hepatocellular carcinoma (HCC) is one of the most fatal cancers worldwide. In this article, we show that expression of abnormal spindle‐like microcephaly‐associated protein (ASPM) is up‐regulated in liver cancer samples, and this up‐regulation is significantly associated with tumor aggressiveness and reduced survival times of patients. Down‐regulation of ASPM expression inhibits the proliferation, invasion, migration and epithelial‐to‐mesenchymal transition of HCC cells in vitro and inhibits tumor formation in nude mice. ASPM interacts with disheveled‐2 (Dvl2) and antagonizes autophagy‐mediated Dvl2 degradation by weakening the functional interaction between Dvl2 and the lipidated form of microtubule‐associated proteins 1A/1B light chain 3A (LC3II), thereby increasing Dvl2 protein abundance and leading to Wnt/β‐catenin signaling activation in HCC cells. Thus, our results define ASPM as a novel oncoprotein in HCC and indicate that disruption of the Wnt–ASPM–Dvl2–β‐catenin signaling axis might have potential clinical value.
Collapse
Affiliation(s)
- Haifeng Zhang
- Department of Biochemistry & Molecular Biology, School of Basic Medical Sciences, Zhengzhou University, China
| | - Xiaobei Yang
- Department of Biochemistry & Molecular Biology, School of Basic Medical Sciences, Zhengzhou University, China
| | - Lili Zhu
- Department of Biochemistry & Molecular Biology, School of Basic Medical Sciences, Zhengzhou University, China
| | - Zhihui Li
- Department of Biochemistry & Molecular Biology, School of Basic Medical Sciences, Zhengzhou University, China
| | - Peipei Zuo
- Academy of Medical Sciences, Zhengzhou University, China
| | - Peng Wang
- Henan Institute of Medical and Pharmaceutical Sciences, Zhengzhou University, China
| | - Jingyu Feng
- Department of Biochemistry & Molecular Biology, School of Basic Medical Sciences, Zhengzhou University, China
| | - Yang Mi
- Department of Biochemistry & Molecular Biology, School of Basic Medical Sciences, Zhengzhou University, China
| | - Chengjuan Zhang
- Center of Repository, The Affiliated Cancer Hospital of Zhengzhou University, China
| | - Yan Xu
- Department of Biochemistry & Molecular Biology, School of Basic Medical Sciences, Zhengzhou University, China
| | - Ge Jin
- Department of Biochemistry & Molecular Biology, School of Basic Medical Sciences, Zhengzhou University, China
| | | | - Hua Ye
- College of Public Health, Zhengzhou University, China
| |
Collapse
|
37
|
Hiroshima K, Wu D, Koh E, Sekine Y, Ozaki D, Yusa T, Nakazawa T, Tsuji S, Miyagi Y, Walts AE, Marchevsky AM, Husain AN, Imai K. Membranous HEG1 expression is a useful marker in the differential diagnosis of epithelioid and biphasic malignant mesothelioma versus carcinomas. Pathol Int 2021; 71:604-613. [PMID: 34240508 PMCID: PMC8519072 DOI: 10.1111/pin.13140] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Accepted: 06/06/2021] [Indexed: 12/24/2022]
Abstract
Sialylated HEG1 has been reported as a highly specific and sensitive mesothelioma marker but a comprehensive evaluation of its expression in carcinomas in different organs, various sarcomas and reactive mesothelial proliferations has not been reported. The aim of this study was to evaluate the clinical applicability of HEG1 as a marker in the diagnosis of mesothelioma. HEG1 immunoreactivity was evaluated in whole sections of 122 mesotheliomas, 75 pulmonary carcinomas, 55 other carcinomas, 16 mesenchymal tumors, and 24 reactive mesothelial proliferations and in tissue microarrays containing 70 epithelioid (EM), 36 biphasic (BM), and 2 sarcomatoid mesotheliomas (SM). In whole sections and tissue microarrays, respectively, membranous HEG1 was expressed in 93.0% and 85.5% of EM, 81.3% and 69.4% of BM, 0% and 0% of SM. HEG1 was not expressed in pulmonary adenocarcinomas. HEG1 was expressed as cytoplasmic immunoreactivity in pulmonary squamous cell carcinomas (21.7%). Membranous HEG1 staining was seen in ovarian carcinomas (66.7%), thyroid carcinomas (100%), reactive conditions (16.7%), and mesenchymal tumors (18.8%). The sensitivity of membranous HEG1 expression to distinguish EM/BM from all carcinomas was 88.8%. The specificity for the differential diagnosis between EM/BM and all carcinomas and pulmonary carcinomas was 92.3% and 98.7%, respectively.
Collapse
Affiliation(s)
- Kenzo Hiroshima
- Department of Biochemistry and Genetics, Chiba University Graduate School of Medicine, Chiba, Japan.,Department of Pathology, Tokyo Women's Medical University, Yachiyo Medical Center, Yachiyo, Japan.,Department of Medicine, Sodegaura Satsukidai Hospital, Sodegaura, Japan
| | - Di Wu
- Department of Pathology, Tokyo Women's Medical University, Yachiyo Medical Center, Yachiyo, Japan
| | - Eitetsu Koh
- Department of Thoracic Surgery, Tokyo Women's Medical University, Yachiyo Medical Center, Yachiyo, Japan
| | - Yasuo Sekine
- Department of Thoracic Surgery, Tokyo Women's Medical University, Yachiyo Medical Center, Yachiyo, Japan
| | - Daisuke Ozaki
- Department of Pathology, Chiba Rosai Hospital, Ichihara, Japan
| | - Toshikazu Yusa
- Department of General Thoracic Surgery and Asbestos Disease Center, Chiba Rosai Hospital, Ichihara, Japan
| | - Tadao Nakazawa
- Department of Pathology, Tokyo Women's Medical University, Yachiyo Medical Center, Yachiyo, Japan
| | - Shoutaro Tsuji
- Division of Cancer Therapeutics, Kanagawa Cancer Center Research Institute, Yokohama, Japan
| | - Yohei Miyagi
- Molecular Pathology and Genetics Division, Kanagawa Cancer Center Research Institute, Yokohama, Japan
| | - Ann E Walts
- Department of Pathology and Laboratory Medicine, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Alberto M Marchevsky
- Department of Pathology and Laboratory Medicine, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Aliya N Husain
- Department of Pathology, University of Chicago, Chicago, Illinois, USA
| | - Kohzoh Imai
- Research Platform Office, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| |
Collapse
|
38
|
Kotipalli A, Banerjee R, Kasibhatla SM, Joshi R. Analysis of H3K4me3-ChIP-Seq and RNA-Seq data to understand the putative role of miRNAs and their target genes in breast cancer cell lines. Genomics Inform 2021; 19:e17. [PMID: 34261302 PMCID: PMC8261273 DOI: 10.5808/gi.21020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Accepted: 05/25/2021] [Indexed: 11/26/2022] Open
Abstract
Breast cancer is one of the leading causes of cancer in women all over the world and accounts for ~25% of newly observed cancers in women. Epigenetic modifications influence differential expression of genes through non-coding RNA and play a crucial role in cancer regulation. In the present study, epigenetic regulation of gene expression by in-silico analysis of histone modifications using chromatin immunoprecipitation sequencing (ChIP-Seq) has been carried out. Histone modification data of H3K4me3 from one normal-like and four breast cancer cell lines were used to predict miRNA expression at the promoter level. Predicted miRNA promoters (based on ChIP-Seq) were used as a probe to identify gene targets. Five triple-negative breast cancer (TNBC)‒specific miRNAs (miR153-1, miR4767, miR4487, miR6720, and miR-LET7I) were identified and corresponding 13 gene targets were predicted. Eight miRNA promoter peaks were predicted to be differentially expressed in at least three breast cancer cell lines (miR4512, miR6791, miR330, miR3180-3, miR6080, miR5787, miR6733, and miR3613). A total of 44 gene targets were identified based on the 3′-untranslated regions of downregulated mRNA genes that contain putative binding targets to these eight miRNAs. These include 17 and 15 genes in luminal-A type and TNBC respectively, that have been reported to be associated with breast cancer regulation. Of the remaining 12 genes, seven (A4GALT, C2ORF74, HRCT1, ZC4H2, ZNF512, ZNF655, and ZNF608) show similar relative expression profiles in large patient samples and other breast cancer cell lines thereby giving insight into predicted role of H3K4me3 mediated gene regulation via the miRNA-mRNA axis.
Collapse
Affiliation(s)
- Aneesh Kotipalli
- HPC-Medical and Bioinformatics Applications Group, Centre for Development of Advanced Computing, Pune 411008, India
| | - Ruma Banerjee
- HPC-Medical and Bioinformatics Applications Group, Centre for Development of Advanced Computing, Pune 411008, India
| | - Sunitha Manjari Kasibhatla
- HPC-Medical and Bioinformatics Applications Group, Centre for Development of Advanced Computing, Pune 411008, India
| | - Rajendra Joshi
- HPC-Medical and Bioinformatics Applications Group, Centre for Development of Advanced Computing, Pune 411008, India
| |
Collapse
|
39
|
Zhang J, Zhang X, Yang S, Bao Y, Xu D, Liu L. FOXH1 promotes lung cancer progression by activating the Wnt/β-catenin signaling pathway. Cancer Cell Int 2021; 21:293. [PMID: 34090445 PMCID: PMC8180118 DOI: 10.1186/s12935-021-01995-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2021] [Accepted: 05/26/2021] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND The expression of forkhead box protein H1 (FOXH1) is frequently upregulated in various cancers. However, the molecular mechanisms underlying the association between FOXH1 expression and lung cancer progression still remain poorly understood. Thus, the main objective of this study is to explore the role of FOXH1 in lung cancer. METHODS The Cancer Genome Atlas dataset was used to investigate FOXH1 expression in lung cancer tissues, and the Kaplan-Meier plotter dataset was used to determine the role of FOXH1 in patient prognosis. A549 and PC9 cells were transfected with short hairpin RNA targeting FOXH1 mRNA. The Cell Counting Kit-8, colony formation, soft agar, wound healing, transwell invasion and flow cytometry assays were performed to evaluate proliferation, migration and invasion of lung cancer cells. Tumorigenicity was examined in a BALB/c nude mice model. Western blot analysis was performed to assess the molecular mechanisms, and β-catenin activity was measured by a luciferase reporter system assay. RESULTS Higher expression level of FOXH1 was observed in tumor tissue than in normal tissue, and this was associated with poor overall survival. Knockdown of FOXH1 significantly inhibited lung cancer cell proliferation, migration, invasion, and cycle. In addition, the mouse xenograft model showed that knockdown of FOXH1 suppressed tumor growth in vivo. Further experiments revealed that FOXH1 depletion inhibited the epithelial-mesenchymal transition of lung cancer cells by downregulating the expression of mesenchymal markers (Snail, Slug, matrix metalloproteinase-2, N-cadherin, and Vimentin) and upregulating the expression of an epithelial marker (E-cadherin). Moreover, knockdown of FOXH1 significantly downregulated the activity of β-catenin and its downstream targets, p-GSK-3β and cyclin D1. CONCLUSION FOXH1 exerts oncogenic functions in lung cancer through regulation of the Wnt/β-catenin signaling pathway. FOXH1 might be a potential therapeutic target for patients with certain types of lung cancer.
Collapse
Affiliation(s)
- Jun Zhang
- Department of Morphological Experiment Center, Medical College of Yanbian University, Yanji, Jilin, 133000, China.,Department of Histology and Embryology, Jilin Medical University, Jilin, Jilin, 132013, China
| | - Xian Zhang
- Department of General Surgery, Affiliated Hospital of Yanbian University, Yanji, Jilin, 133000, China
| | - Shasha Yang
- Department of Morphological Experiment Center, Medical College of Yanbian University, Yanji, Jilin, 133000, China
| | - Yanqiu Bao
- Department of Pathology, Affiliated Hospital of Yanbian University, Yanji, Jilin, 133000, China
| | - Dongyuan Xu
- Department of Morphological Experiment Center, Medical College of Yanbian University, Yanji, Jilin, 133000, China.
| | - Lan Liu
- Department of Pathology, Affiliated Hospital of Yanbian University, Yanji, Jilin, 133000, China.
| |
Collapse
|
40
|
Cheng X, Li F, Tao Z. Tenascin-C promotes epithelial-to-mesenchymal transition and the mTOR signaling pathway in nasopharyngeal carcinoma. Oncol Lett 2021; 22:570. [PMID: 34113398 PMCID: PMC8185706 DOI: 10.3892/ol.2021.12831] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2020] [Accepted: 04/22/2021] [Indexed: 12/24/2022] Open
Abstract
Tenascin-C (TNC) is a large extracellular matrix glycoprotein that promotes cell adhesion and tissue remodeling, and is involved in the transduction of cellular signaling pathways. The present study aimed to investigate the role of TNC and determine its effect in nasopharyngeal carcinoma (NPC). TNC gene transcription and expression were analyzed using the NPC dataset and immunohistochemistry analysis of NPC tissues. Weighted gene co-expression network and gene enrichment analyses were performed to determine the potential molecular mechanisms underlying the effects of TNC in NPC. TNC expression was suppressed in NPC cells, and the effects were determined both in vitro and in vivo. The results demonstrated that TNC gene transcription and expression were high in NPC tissues compared with normal tissues. Notably, TNC knockdown inhibited NPC cell proliferation, migration and invasion. In addition, TNC knockdown inhibited tumor growth in mice. In vitro, TNC knockdown inhibited epithelial-to-mesenchymal transition (EMT) and decreased activity of the PI3K/AKT/mTOR signaling pathway in NPC cells. Taken together, these results suggest that TNC promotes cell proliferation, EMT and activity of the PI3K/AKT/mTOR signaling pathway in NPC cells, and thus functions as an oncogene.
Collapse
Affiliation(s)
- Xiang Cheng
- Department of Otolaryngology-Head and Neck Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Fen Li
- Department of Otolaryngology-Head and Neck Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Zezhang Tao
- Department of Otolaryngology-Head and Neck Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| |
Collapse
|
41
|
EB2 promotes hepatocellular carcinoma proliferation and metastasis via MAPK/ERK pathway by modulating microtubule dynamics. Clin Sci (Lond) 2021; 135:847-864. [PMID: 33755094 DOI: 10.1042/cs20201500] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2020] [Revised: 03/09/2021] [Accepted: 03/23/2021] [Indexed: 01/22/2023]
Abstract
Metastasis is the main cause of poor postoperative survival of hepatocellular carcinoma (HCC) patients. Cytoskeleton rearrangement is a key event in cancer metastasis. However, the significance of microtubule (MT), one of the core components of cytoskeleton, in this process is only beginning to be revealed. Here, we find that the MT dynamics regulator end-binding protein 2 (EB2) is highly expressed in HCC and predicts poor prognosis of HCC patients. Functional studies show that EB2 overexpression promotes HCC proliferation, invasion and metastasis in vitro and in vivo, while EB2 knockdown has opposite results. Mechanistically, EB2 mediates MTs destabilization, increases Src (Src proto-oncogene non-receptor tyrosine kinase) activity, and thus facilitates extracellular signal-regulated kinase (ERK) signaling activation, which could in turn promote EB2 expression in HCC, eventually resulting in enhanced HCC proliferation, invasion and metastasis. Furthermore, U0126, a specific ERK inhibitor, could effectively inhibit EB2-mediated HCC proliferation and metastasis in vitro and in vivo. In conclusion, EB2 coordinates MT cytoskeleton and intracellular signal transduction, forming an EB2-MT-ERK positive feedback loop, to facilitate HCC proliferation, invasion and metastasis. EB2 could serve as a promising prognostic biomarker and potential therapeutic target for HCC; HCC patients with high EB2 expression may benefit from treatment with ERK inhibitors.
Collapse
|
42
|
Chen PP, Zhang ZS, Wu JC, Zheng JF, Lin F. LncRNA SNHG12 promotes proliferation and epithelial mesenchymal transition in hepatocellular carcinoma through targeting HEG1 via miR-516a-5p. Cell Signal 2021; 84:109992. [PMID: 33774129 DOI: 10.1016/j.cellsig.2021.109992] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2021] [Revised: 02/20/2021] [Accepted: 03/22/2021] [Indexed: 12/17/2022]
Abstract
Hepatocellular carcinoma (HCC) is the most common cancer and its prognosis is poor due to metastasis and recurrence. EMT is associated with metastasis. A deep understanding of regulatory mechanism of EMT is critical. LncRNA is involved in regulation of various biological processes including EMT. This study aimed to investigate the regulatory signal axis among lncRNA SNHG12, miR-516a-5p and the target gene HEG1 during EMT. Cell cycle and apoptosis were analyzed by flow cytometry. Tumorigenesis was analyzed by clone formation assay. Wound healing assay and transwell assay was performed to detect migration and invasion, respectively. Interaction among SNHG12, miR-516a-5p and HEG1 were analyzed by dual luciferase assay and RIP assay. We also detected expression of RNA and protein by QPCR and western blotting. Finally, tumor growth was analyzed by tumorigenesis assay in vivo. Ki-67 and HEG1 level in tumor tissues was analyzed by IHC. SNHG12 and HEG1 were upregulated, miR-516a-5p was downregulated in HCC cell lines. SNHG12 could interact with and inhibit miR-516a-5p. MiR-516a-5p could interact with HEG1 and inhibit HEG1 expression. Knock down SNHG12 inhibited proliferation, migration, invasion, EMT and promoted apoptosis of HCC cells. Such effects were antagonized by inhibiting miR-516a-5p. SNHG12 overexpression lead to opposite results. Similar results were observed in mice. SNHG12 could promote EMT in HCC through targeting and inhibiting miR-516a-5p, which eventually upregulated HEG1 expression, in both cell and mice.
Collapse
Affiliation(s)
- Ping-Ping Chen
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Jinan University, Guangzhou 510630, Guangdong Province, PR China; Department of Hepatobiliary Surgery, Hainan Provincial People's Hospital, Haikou 570311, Hainan Province, PR China
| | - Zhen-Sheng Zhang
- Department of Hepatobiliary Surgery, Hainan Provincial People's Hospital, Haikou 570311, Hainan Province, PR China
| | - Jin-Cai Wu
- Department of Hepatobiliary Surgery, Hainan Provincial People's Hospital, Haikou 570311, Hainan Province, PR China
| | - Jin-Fang Zheng
- Department of Hepatobiliary Surgery, Hainan Provincial People's Hospital, Haikou 570311, Hainan Province, PR China
| | - Fan Lin
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Jinan University, Guangzhou 510630, Guangdong Province, PR China; Department of Hepatobiliary Surgery, The First Clinical Medical College of Jinan University, Guangzhou 510630, Guangdong Province, PR China.
| |
Collapse
|
43
|
Song L, Guo X, Zhao F, Wang W, Zhao Z, Jin L, Wu C, Yao J, Ma Z. TTC36 inactivation induce malignant properties via Wnt-β-catenin pathway in gastric carcinoma. J Cancer 2021; 12:2598-2609. [PMID: 33854620 PMCID: PMC8040709 DOI: 10.7150/jca.47292] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2020] [Accepted: 02/16/2021] [Indexed: 01/23/2023] Open
Abstract
Objective: Tetratricopeptide repeat (TRP)-mediated cofactor proteins are involved in a wide range of cancers. TTC36 is little studied member of TRP subfamily. This study aimed to investigate the role of TTC36 in human gastric carcinoma (GC) and explore the potential underlying mechanisms. Methods: The analysis of TTC36 differential expression in GC was conducted using data from TCGA and a human tissue microarray. And effects of TTC36 expression on the prognosis of patients with gastric carcinoma were analyzed using the data from the GEO database. Lentivirus was transfected into the cell lines of AGS and BGC823 to construct overexpression and knocked down TTC36 cell model respectively. The effect of TTC36 expression on the growth, apoptosis and cell cycle of cells was explored in vitro. Downstream molecules were detected by western blotting. GSEA predicted signal pathway and related proteins were then detected. Results: TTC36 expression in human GC tissues was found significantly lower than that in adjacent normal tissues and closely related to clinical prognosis. The overexpression of TTC36 notably inhibited tumor progression, cell cycle G1/S transfer and increased apoptosis in AGS cells. Conversely, the opposite effects were observed when TTC36 was suppressed in BGC823 cells. The expression of cleaved caspase3, Survivin, cyclin D1 and c-Myc were consistent with the phenotype in TTC36 operated GC cell lines. Intriguingly, GSEA analysis predicted Wnt-β-catenin pathway involved in TTC36 induced effects in GC cells, expression of β-catenin and downstream molecules such as TCF4, c-jun and pAKT were found negative related to TTC36 expression in GC cells. Notably, treatment with the Wnt/β-catenin inhibitor XAV939 dramatically attenuated the effects of TTC36 in GC cells. Conclusion: These results signify a critical role for TTC36 as a tumor suppressor in gastric carcinoma via regulating Wnt-β-catenin pathway.
Collapse
Affiliation(s)
- Lei Song
- Key Laboratory of Biotechnology and Bioengineering of State Ethnic Affairs Commission, Biomedical Research Center, Northwest Minzu University, Lanzhou 730030, Gansu, China.,Department of Medicine, Northwest Minzu University, Lanzhou 730030, Gansu, China
| | - Xiaonong Guo
- Key Laboratory of Biotechnology and Bioengineering of State Ethnic Affairs Commission, Biomedical Research Center, Northwest Minzu University, Lanzhou 730030, Gansu, China
| | - Fei Zhao
- Key Laboratory of Biotechnology and Bioengineering of State Ethnic Affairs Commission, Biomedical Research Center, Northwest Minzu University, Lanzhou 730030, Gansu, China.,Department of Medicine, Northwest Minzu University, Lanzhou 730030, Gansu, China
| | - Wei Wang
- Department of Urology, Institute of Urology, Gansu Nephro-Urological Clinical Center, Key Laboratory of Urological Diseases in Gansu Province, Lanzhou University Second Hospital, Lanzhou 730030, Gansu, China
| | - Zhifang Zhao
- Department of Medicine, Northwest Minzu University, Lanzhou 730030, Gansu, China
| | - Long Jin
- Department of Medicine, Northwest Minzu University, Lanzhou 730030, Gansu, China
| | - Chengli Wu
- Department of Medicine, Northwest Minzu University, Lanzhou 730030, Gansu, China
| | - Jibin Yao
- Department of Surgical Oncology, Gansu Provincial Hospital, Lanzhou 730000, Gansu, China
| | - Zhongren Ma
- Key Laboratory of Biotechnology and Bioengineering of State Ethnic Affairs Commission, Biomedical Research Center, Northwest Minzu University, Lanzhou 730030, Gansu, China
| |
Collapse
|
44
|
Xiao S, Hu J, Hu N, Sheng L, Rao H, Zheng G. Identification of a Novel Epithelial-to-Mesenchymal-related Gene Signature in Predicting Survival of Patients with Hepatocellular Carcinoma. Comb Chem High Throughput Screen 2021; 25:1254-1270. [PMID: 33655854 DOI: 10.2174/1386207324666210303093629] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2020] [Revised: 12/11/2020] [Accepted: 02/09/2021] [Indexed: 11/22/2022]
Abstract
BACKGROUND Epithelial-mesenchymal transformation (EMT) promotes cancer metastasis including hepatocellular carcinoma. Therefore, EMT-related gene signature was explored. OBJECTIVE The present study was designed to develop an EMT-related gene signature for predicting the prognosis of patients with hepatocellular carcinoma. METHODS We conducted an integrated gene expression analysis based on tumor data of the patients with hepatocellular carcinoma from The Cancer Genome Atlas (TCGA), HCCDB18 and GSE14520 dataset. An EMT-related gene signature was constructed by least absolute shrinkage and selection operator (LASSO) and COX regression analysis of univariate and multivariate survival. RESULTS A 3-EMT gene signature was developed and validated based on gene expression profiles of hepatocellular carcinoma from three microarray platforms. Patients with a high risk score had a significantly worse overall survival (OS) than those with low risk scores. The EMT-related gene signature showed a high performance in accurately predicting prognosis and in examining the clinical characteristics and immune score analysis. Univariate and multivariate Cox regression analyses confirmed that the EMT-related gene signature was an independent prognostic factor for predicting survival in hepatocellular carcinoma patients. Compared with the existing models, our EMT-related gene signature reached higher area under curve (AUC). CONCLUSION Our findings provide novel insight into understanding EMT and help identify hepatocellular carcinoma patients with poor prognosis.
Collapse
Affiliation(s)
- Simeng Xiao
- College of Pharmacy, Hubei University of Chinese Medicine, Wuhan, Hubei, 430065. China
| | - Junjie Hu
- College of Pharmacy, Hubei University of Chinese Medicine, Wuhan, Hubei, 430065. China
| | - Na Hu
- College of Pharmacy, Hubei University of Chinese Medicine, Wuhan, Hubei, 430065. China
| | - Lei Sheng
- College of Pharmacy, Hubei University of Chinese Medicine, Wuhan, Hubei, 430065. China
| | - Hui Rao
- College of Pharmacy, Hubei University of Chinese Medicine, Wuhan, Hubei, 430065. China
| | - Guohua Zheng
- Key Laboratory for Chinese Medicine Resource and Compound Prescription of Ministry of Education, Hubei University of Chinese Medicine, Wuhan, Hubei, 430065. China
| |
Collapse
|
45
|
Zha Z, Li D, Zhang P, Wang P, Fang X, Liu X, Weng C, Li B, Wu Y, Mao H, Wang L, Xu L, Dong J, Guan M, Lu L, Liu G. Neuron specific enolase promotes tumor metastasis by activating the Wnt/β-catenin pathway in small cell lung cancer. Transl Oncol 2021; 14:101039. [PMID: 33618068 PMCID: PMC7905480 DOI: 10.1016/j.tranon.2021.101039] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2020] [Revised: 02/03/2021] [Accepted: 02/05/2021] [Indexed: 01/07/2023] Open
Abstract
Neuron-specific enolase (NSE) has been used as a specific biomarker for small cell lung cancer (SCLC) patients. Nevertheless, the biological function and mechanism of NSE in SCLC are still unclear. In this study, we clarified the role of NSE in the progression of SCLC and found that NSE expression was positively correlated with distant metastasis. Functional analysis showed that overexpression of NSE promoted migration and invasion of SCLC cells. Mechanism analysis showed that NSE overexpression induced epithelial-mesenchymal transition (EMT) of SCLC cells. Moreover, overexpression of NSE increased the protein expression of β-catenin and its downstream target genes, and silencing β-catenin eliminated NSE-mediated cell migration, invasion and EMT process. Furthermore, NSE interacted with β-catenin and inhibited the degradation of β-catenin. Besides, the animal experiments also indicated that NSE could promote the EMT process and distant metastasis of SCLC cells in vivo. In summary, our results revealed that NSE could promote the EMT process of SCLC cells by activating the Wnt/β-catenin signaling pathway, thereby promoting cell migration, invasion and distant metastasis, which might serve as a potential target for the therapy of SCLC patients.
Collapse
Affiliation(s)
- Zhiqiang Zha
- Department of Medical Oncology, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, 510180, Guangdong, China
| | - Dailing Li
- Department of Medical Oncology, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, 510180, Guangdong, China
| | - Peiling Zhang
- Department of Medical Oncology, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, 510180, Guangdong, China
| | - Peipei Wang
- Department of Medical Oncology, Guangzhou First People's Hospital, Guangzhou Medical University, Guangzhou 510180, Guangdong, China
| | - Xisheng Fang
- Department of Medical Oncology, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, 510180, Guangdong, China; Department of Medical Oncology, Guangzhou First People's Hospital, Guangzhou Medical University, Guangzhou 510180, Guangdong, China
| | - Xia Liu
- Department of Medical Oncology, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, 510180, Guangdong, China; Department of Medical Oncology, Guangzhou First People's Hospital, Guangzhou Medical University, Guangzhou 510180, Guangdong, China
| | - Chengyin Weng
- Department of Medical Oncology, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, 510180, Guangdong, China; Department of Medical Oncology, Guangzhou First People's Hospital, Guangzhou Medical University, Guangzhou 510180, Guangdong, China
| | - Baoxiu Li
- Department of Medical Oncology, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, 510180, Guangdong, China; Department of Medical Oncology, Guangzhou First People's Hospital, Guangzhou Medical University, Guangzhou 510180, Guangdong, China
| | - Yong Wu
- Department of Medical Oncology, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, 510180, Guangdong, China; Department of Medical Oncology, Guangzhou First People's Hospital, Guangzhou Medical University, Guangzhou 510180, Guangdong, China
| | - Haibo Mao
- Department of Medical Oncology, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, 510180, Guangdong, China; Department of Medical Oncology, Guangzhou First People's Hospital, Guangzhou Medical University, Guangzhou 510180, Guangdong, China
| | - Lina Wang
- Department of Medical Oncology, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, 510180, Guangdong, China; Department of Medical Oncology, Guangzhou First People's Hospital, Guangzhou Medical University, Guangzhou 510180, Guangdong, China
| | - Lin Xu
- Department of Medical Oncology, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, 510180, Guangdong, China; Department of Medical Oncology, Guangzhou First People's Hospital, Guangzhou Medical University, Guangzhou 510180, Guangdong, China
| | - Jiaming Dong
- Department of Medical Oncology, Guangzhou First People's Hospital, Guangzhou Medical University, Guangzhou 510180, Guangdong, China
| | - Mingmei Guan
- Department of Medical Oncology, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, 510180, Guangdong, China; Department of Medical Oncology, Guangzhou First People's Hospital, Guangzhou Medical University, Guangzhou 510180, Guangdong, China
| | - Lin Lu
- Department of Medical Oncology, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, 510180, Guangdong, China; Department of Medical Oncology, Guangzhou First People's Hospital, Guangzhou Medical University, Guangzhou 510180, Guangdong, China.
| | - Guolong Liu
- Department of Medical Oncology, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, 510180, Guangdong, China; Department of Medical Oncology, Guangzhou First People's Hospital, Guangzhou Medical University, Guangzhou 510180, Guangdong, China.
| |
Collapse
|
46
|
Le J, Fu Y, Han Q, Ma Y, Ji H, Wei X, Chen Y, Sun Y, Gao Y, Wu H. Transcriptome Analysis of the Inhibitory Effect of Sennoside A on the Metastasis of Hepatocellular Carcinoma Cells. Front Pharmacol 2021; 11:566099. [PMID: 33708105 PMCID: PMC7942274 DOI: 10.3389/fphar.2020.566099] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Accepted: 10/30/2020] [Indexed: 12/24/2022] Open
Abstract
Sennoside A (SA) is a bioactive component of Rheum officinale Baill. with an activity of irritant laxative, which has been reported to possess therapeutic potential in various diseases or conditions including obesity, insulin resistance, liver steatosis, prostate cancer and pancreatic cancer progression. However, whether SA has therapeutic potential in hepatocellular carcinoma (HCC) treatment remains elusive. In this study, we treated two HCC cell lines, HepG2 and SMMC-7721 with SA and found that SA selectively inhibited the growth of HCC cells by proliferation assay. SA has a good inhibitory effect on proliferation of HepG2 cells in a concentration dependent manner, but there was no effect on SMMC-7721 cells. Then we conducted transwell assays and transcriptome analysis in HCC cells and examined the effects of SA on HCC in vivo. The results showed that SA significantly inhibited the migration and invasion of HCC. Comparison of RNA-seq transcriptome profiles from control groups and SA-treated groups identified 171 and 264 differentially expressed genes (DEGs) in HepG2 and SMMC-7721 cells respectively, in which includes 2 overlapping up-regulated DEGs and 12 overlapping down-regulated DEGs between HepG2 and SMMC-7721 cells. The qPCR were applied to investigate the transcriptional level of 9 overlapping down-regulated DEGs related to cancer metastasis, and the results were consistent with RNA-seq data. The dominate pathways including Wnt signaling pathway, TNF signaling pathway, VEGF signaling pathway, and NF-κB signaling pathway were strongly inhibited by SA, which are involved in regulating cancer metastasis. Finally, we confirmed that the downregulation of KRT7 and KRT81 could inhibit HCC metastasis. This study has provided new insight into the understanding of the inhibitory effects and potential targets of SA on the metastasis of HCC.
Collapse
Affiliation(s)
- Jiamei Le
- Shanghai University of Medicine & Health Sciences Affiliated Zhoupu Hospital, Shanghai, China.,Shanghai Key Laboratory of Molecular Imaging, Collaborative Innovation Center for Biomedicine, Shanghai University of Medicine & Health Sciences, Shanghai, China
| | - Yi Fu
- Shanghai University of Medicine & Health Sciences Affiliated Zhoupu Hospital, Shanghai, China.,Shanghai Key Laboratory of Molecular Imaging, Collaborative Innovation Center for Biomedicine, Shanghai University of Medicine & Health Sciences, Shanghai, China
| | - Qiuqin Han
- Shanghai University of Medicine & Health Sciences Affiliated Zhoupu Hospital, Shanghai, China.,Shanghai Key Laboratory of Molecular Imaging, Collaborative Innovation Center for Biomedicine, Shanghai University of Medicine & Health Sciences, Shanghai, China
| | - Yujie Ma
- Shanghai University of Medicine & Health Sciences Affiliated Zhoupu Hospital, Shanghai, China.,Shanghai Key Laboratory of Molecular Imaging, Collaborative Innovation Center for Biomedicine, Shanghai University of Medicine & Health Sciences, Shanghai, China
| | - Houlin Ji
- Shanghai University of Medicine & Health Sciences Affiliated Zhoupu Hospital, Shanghai, China.,Shanghai Key Laboratory of Molecular Imaging, Collaborative Innovation Center for Biomedicine, Shanghai University of Medicine & Health Sciences, Shanghai, China
| | - Xindong Wei
- Shanghai University of Medicine & Health Sciences Affiliated Zhoupu Hospital, Shanghai, China.,Shanghai Key Laboratory of Molecular Imaging, Collaborative Innovation Center for Biomedicine, Shanghai University of Medicine & Health Sciences, Shanghai, China
| | - Yifan Chen
- Shanghai University of Medicine & Health Sciences Affiliated Zhoupu Hospital, Shanghai, China.,Shanghai Key Laboratory of Molecular Imaging, Collaborative Innovation Center for Biomedicine, Shanghai University of Medicine & Health Sciences, Shanghai, China
| | - Yongning Sun
- Department of Traditional Chinese Medicine, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China.,Department of Cardiology, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yueqiu Gao
- Institute of Clinical Immunology, Department of Liver Diseases, Central Laboratory, ShuGuang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China.,Laboratory of Cellular Immunity, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Hailong Wu
- Shanghai University of Medicine & Health Sciences Affiliated Zhoupu Hospital, Shanghai, China.,Shanghai Key Laboratory of Molecular Imaging, Collaborative Innovation Center for Biomedicine, Shanghai University of Medicine & Health Sciences, Shanghai, China
| |
Collapse
|
47
|
HEG1 Is a Highly Specific and Sensitive Marker of Epithelioid Malignant Mesothelioma. Am J Surg Pathol 2020; 44:1143-1148. [PMID: 32205484 DOI: 10.1097/pas.0000000000001469] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Malignant mesothelioma can be difficult to distinguish from other malignancies, particularly non-small cell lung carcinomas (NSCLCs), without immunohistochemistry. However, conventional markers of mesothelial lineage all have variable degrees of cross-reactivity with other neoplasms, including NSCLCs, necessitating the use of multiple mesothelioma and carcinoma markers in every case for accurate diagnosis. A recently described monoclonal HEG homolog 1 (HEG1) antibody was proposed to be a specific marker for mesothelioma. Here we performed a large scale assessment of the SKM9-2 HEG1 antibody using tissue microarrays containing 69 epithelioid mesotheliomas, 32 sarcomatoid mesotheliomas, 167 NSCLCs, and 17 ovarian high-grade serous carcinomas. Strong membrane staining, usually diffuse, for HEG1 was seen in 65/69 (94%) epithelioid mesotheliomas, 0/60 pulmonary squamous cell carcinomas, 0/73 pulmonary adenocarcinomas, and 0/13 pulmonary large cell carcinomas. HEG1 showed staining in 14/32 (44%) sarcomatoid mesotheliomas compared with 0/21 sarcomatoid pulmonary carcinomas. Three of 17 (18%) high-grade serous carcinomas demonstrated membrane staining. Ten B3 thymoma whole sections were negative. On the microarrays, the conventional mesothelial markers calretinin, WT1, D2-40, and CK5/6 had sensitivities for epithelioid mesothelioma of 94%, 90%, 96%, and 91%, respectively. We conclude that HEG1 SKM9-2 antibody offers sensitivity comparable to conventional markers for epithelioid mesotheliomas, but provides considerably better specificity, such that the diagnosis of epithelioid mesothelioma versus NSCLC potentially could be confirmed with a combination of HEG1 and a suitable broad spectrum carcinoma marker such as claudin-4. HEG1 is specific but insensitive for separating sarcomatoid mesotheliomas from sarcomatoid lung carcinomas.
Collapse
|
48
|
Xiong C, Wang G, Bai D. A novel prognostic models for identifying the risk of hepatocellular carcinoma based on epithelial-mesenchymal transition-associated genes. Bioengineered 2020; 11:1034-1046. [PMID: 32951492 PMCID: PMC8291854 DOI: 10.1080/21655979.2020.1822715] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Several epithelial-mesenchymal transition (EMT)-associated genes (EAGs) have been confirmed to correlate with the prognosis of hepatocellular carcinoma (HCC) patients. Herein, we explored the value of EAGs in the prognosis of HCC relying on data from The Cancer Genome Atlas (TCGA) database. A total of 200 EMT-associated genes were downloaded from the Gene set enrichment analysis (GSEA) website. Moreover, 96 differentially expressed EAGs were identified. Using Gene Ontology (GO) enrichment analysis and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analysis, we forecasted the potential molecular mechanisms of EAGs. To identify prognostic EAGs, Cox regression was used in developing a prognostic risk model. Then, the Kaplan-Meier and receiver operating characteristic (ROC) curves were plotted to validate the prognostic significance of the model. A total of 5 prognostic correlated EAGs (P3H1, SPP1, MMP1, LGALS1, and ITGB5) were screened via Cox regression, which provided the basis for developing a novel prognostic risk model. Based on the risk model, patients were subdivided into high-risk and low-risk groups. The overall survival of the low-risk group was better compared to the high-risk group (P < 0.00001). The ROC curve of the risk model showed a higher AUC (Area under Curve) (AUC = 0.723) compared to other clinical features (AUC ≤ 0.511). A nomogram based on this model was constructed to predict the 1-year, 2-year, and 3-year overall survival rates (OS) of patients. Conclusively, we developed a novel HCC prognostic risk model based on the expression of EAGs, which help advance the prognostic management of HCC patients. Abbreviations: HCC: hepatocellular carcinoma; TCGA: The Cancer Genome Atlas; EMT: epithelial-mesenchymal transition; EAGs: EMT-associated genes; GSEA: gene set enrichment analysis; GO: Gene Ontology; KEGG: Kyoto Encyclopedia of Genes and Genomes; PPI: protein-protein interaction; TF: transcription factor; ROC: receiver operating characteristic; K-M: Kaplan-Meier; AUC: the area under the ROC curve; FDR: false discovery rate; TNM: Tumor size/lymph nodes/distance metastasis.
Collapse
Affiliation(s)
- Chen Xiong
- Dalian Medical University , Dalian, P.R. China
| | - Guifu Wang
- Dalian Medical University , Dalian, P.R. China
| | - Dousheng Bai
- Department of Hepatobiliary Surgery, Clinical Medical College, Yangzhou University , Yangzhou, P.R. China
| |
Collapse
|
49
|
Wang Z, Li Z, Wu Q, Li C, Li J, Zhang Y, Wang C, Sun S, Sun S. DNER promotes epithelial-mesenchymal transition and prevents chemosensitivity through the Wnt/β-catenin pathway in breast cancer. Cell Death Dis 2020; 11:642. [PMID: 32811806 PMCID: PMC7434780 DOI: 10.1038/s41419-020-02903-1] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2019] [Revised: 08/02/2020] [Accepted: 08/03/2020] [Indexed: 12/24/2022]
Abstract
Breast cancer (BC) is the most common malignant tumour in women worldwide, and one of the most common fatal tumours in women. Delta/Notch-like epidermal growth factor (EGF)-related receptor (DNER) is a transmembrane protein involved in the development of tumours. The role and potential mechanism of DNER in epithelial-mesenchymal transition (EMT) and apoptosis in BC are not fully understood. We find that DNER is overexpressed in BC tissue, especially triple-negative breast cancer (TNBC) tissue, and related to the survival of BC and TNBC patients. In addition, DNER regulates cell EMT to enhance the proliferation and metastasis of BC cells via the Wnt/β-catenin pathway in vitro and in vivo. Moreover, the expression levels of β-catenin and DNER in BD tissue are positively correlated. The simultaneously high expression of DNER and β-catenin contributes to poor prognosis in BC patients. Finally, DNER protects BC cells from epirubicin-induced growth inhibition and apoptosis via the Wnt/β-catenin pathway. In conclusion, these results suggest that DNER induces EMT and prevents apoptosis by the Wnt/β-catenin pathway, ultimately promoting the malignant progression of BC. In conclusion, our study demonstrates that DNER functions as an oncogene and potentially valuable therapeutic target for BC.
Collapse
Affiliation(s)
- Zhong Wang
- Department of Breast and Thyroid Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Zhiyu Li
- Department of Breast and Thyroid Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Qi Wu
- Department of Breast and Thyroid Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Chenyuan Li
- Department of Breast and Thyroid Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Juanjuan Li
- Department of Breast and Thyroid Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Yimin Zhang
- Department of Breast and Thyroid Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Changhua Wang
- Department of Pathophysiology, Wuhan University School of Basic Medical Sciences, Wuhan, Hubei, China
| | - Si Sun
- Department of Clinical Laboratory, Renmin Hospital of Wuhan University, Wuhan, Hubei, China.
| | - Shengrong Sun
- Department of Breast and Thyroid Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei, China.
| |
Collapse
|
50
|
Colorectal cancer-derived exosomal miR-106b-3p promotes metastasis by down-regulating DLC-1 expression. Clin Sci (Lond) 2020; 134:419-434. [PMID: 32065214 DOI: 10.1042/cs20191087] [Citation(s) in RCA: 71] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2019] [Revised: 02/04/2020] [Accepted: 02/17/2020] [Indexed: 12/12/2022]
Abstract
Cancer-derived exosomal miRNAs play an important role in the development of metastasis, but the effects and underlying mechanisms remain unclear. In the present study, we investigated the miRNA expression profiles of 5 paired serum exosomal samples from metastatic colorectal cancer (mCRC) and non-mCRC patients via RNA sequencing. After we evaluated the differentially expressed miRNAs in 80 CRC patients, miR-106b-3p was selected as a metastasis-associated miRNA of CRC. We showed that the expression level of serum exosomal miR-106b-3p was significantly higher in CRC patients with metastasis than those without metastasis. Additionally, high serum exosomal miR-106b-3p expression in patients was correlated with a poor prognosis. Coculture of low-metastatic CRC cells with high-metastatic CRC cell-derived exosomes promoted cell migration, invasion, and epithelial-to-mesenchymal transition (EMT), which was caused by the transport and transduction of miR-106b-3p in vitro. Moreover, exosomal miR-106b-3p promoted lung metastasis of CRC cells in vivo. In addition, we demonstrated that miR-106b-3p regulated metastasis by targeting deleted in liver cancer-1 (DLC-1). A negative correlation was also identified between miR-106b-3p and DLC-1 expression in human CRC tumour tissues and in mouse lung metastatic lesions. Collectively, our study indicated that metastasis-associated miR-106b-3p from serum exosomes could be used as a potential prognostic biomarker and therapeutic target for CRC patients.
Collapse
|