1
|
Wang Z, Qiao X, Chen Y, Peng N, Niu C, Wang Y, Li C, Hu Z, Zhang C, Cheng C. SVIP reduces IGFBP-2 expression and inhibits glioblastoma progression via stabilizing PTEN. Cell Death Discov 2024; 10:362. [PMID: 39138166 PMCID: PMC11322382 DOI: 10.1038/s41420-024-02130-z] [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: 04/20/2024] [Revised: 07/28/2024] [Accepted: 07/31/2024] [Indexed: 08/15/2024] Open
Abstract
Glioblastoma (GBM) presents significant challenges due to its invasive nature and genetic heterogeneity. In this study, we investigated the impact of Small VCP/P97-Interacting Protein (SVIP) on GBM progression. Our results revealed elevated expression of Insulin-like Growth Factor Binding Protein 2 (IGFBP-2) and STIP1 homology and U-box containing protein 1 (STUB1), coupled with reduced SVIP levels in GBM samples. Notably, high IGFBP-2 expression correlated with poor prognosis. Mechanistically, SVIP competitively inhibited STUB1, selectively binding to VCP/p97, thereby reducing PTEN degradation. This SVIP-mediated regulation exerted influence on the PTEN/PI3K/AKT/mTOR pathway, leading to the suppression of GBM progression. Co-localization experiments demonstrated that SVIP hindered PTEN ubiquitination and degradation by outcompeting STUB1 for VCP/p97 binding. Moreover, SVIP overexpression resulted in reduced activation of AKT/mTOR signaling and facilitated autophagy. In vivo experiments using a GBM xenograft model substantiated the tumor-suppressive effects of SVIP, evident by suppressed tumor growth, decreased IGFBP-2 expression, and improved survival rates. Collectively, our findings underscore the functional significance of SVIP in GBM progression. By inhibiting STUB1 and stabilizing PTEN, SVIP modulates the expression of IGFBP-2 and attenuates the activation of the PI3K/AKT/mTOR pathway, thereby emerging as a promising therapeutic target for GBM treatment.
Collapse
Affiliation(s)
- Zixuan Wang
- Department of Neurosurgery, Centre for Leading Medicine and Advanced Technologies of IHM, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230001, China
- Dalian Medical University, Dalian, Liaoning, 116000, China
| | - Xiaolong Qiao
- Anhui University of Science and Technology, Huainan, Anhui, 232001, China
| | - Yinan Chen
- Department of Neurosurgery, Centre for Leading Medicine and Advanced Technologies of IHM, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230001, China
| | - Nan Peng
- Department of Neurosurgery, Centre for Leading Medicine and Advanced Technologies of IHM, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230001, China
| | - Chaoshi Niu
- Department of Neurosurgery, Centre for Leading Medicine and Advanced Technologies of IHM, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230001, China
| | - Yang Wang
- Dalian Medical University, Dalian, Liaoning, 116000, China
| | - Cong Li
- Dalian Medical University, Dalian, Liaoning, 116000, China.
| | - Zengchun Hu
- Department of Neurosurgery, 2nd Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, 116023, China.
| | - Caihua Zhang
- Dalian Medical University, Dalian, Liaoning, 116000, China.
| | - Chuandong Cheng
- Department of Neurosurgery, Centre for Leading Medicine and Advanced Technologies of IHM, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230001, China.
| |
Collapse
|
2
|
Zheng Y, Li W, Huang Y, Cheng H. Exosomal IGFBP2 derived from highly metastatic promotes hepatocellular carcinoma metastasis by inducing epithelial mesenchymal transition. Gene 2024; 913:148374. [PMID: 38490505 DOI: 10.1016/j.gene.2024.148374] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2023] [Revised: 02/20/2024] [Accepted: 03/12/2024] [Indexed: 03/17/2024]
Abstract
Liver cancer metastasis is the main cause of death in liver cancer patients. Exosomes, which are small vesicles released by cancer cells, play a crucial role in the metastasis of cancer. The aim of this study was to investigate the effect of exosomes derived from high metastatic potential liver cancer cells acting as cell to cell communication on liver cancer metastasis. Bioinformatics analysis was used to obtain the differential expression of exosomal mRNAs from the plasma of both liver cancer patients and healthy volunteers. Transmission electron microscopy (TEM), nanoparticle tracking analysis (NTA), and protein blot were employed to characterize the exosomes. The molecular mechanisms and were explored by conducting CCK8, Transwell, Tunel, RTqPCR, western blot, and immunofluorescence staining. We examined IGFBP2 special expression in the plasma exosomes of both liver cancer patients and healthy volunteers, and its presence was associated with a poor prognosis in liver cancer patients. Furthermore, we observed that exosomes from highly metastatic liver cancer cells (MHCC97H) contained high levels of IGFBP2 and could enhance the metastatic potential of less aggressive liver cancer cells (Hep3B). Additionally, we discovered that IGFBP2 in MHCC97H-derived exosomes activated ERK signaling pathway, which triggered epithelial-mesenchymal transition (EMT) in Hep3B cells. Our study underscores the significance of exosomal IGFBP2 from highly metastatic liver cancer cells as a driver of metastasis in less invasive liver cancer cells. This suggests that targeting IGFBP2 in exosomes could be a promising strategy for the treatment and prognosis of liver cancer patients.
Collapse
Affiliation(s)
- Yongyuan Zheng
- Department of Hepatology and Infectious Diseases, the Second Affiliated Hospital, Shantou University Medical College, Shantou, Guangdong, PR China
| | - Weibing Li
- Department of Hepatology and Infectious Diseases, the Second Affiliated Hospital, Shantou University Medical College, Shantou, Guangdong, PR China
| | - Yansong Huang
- Department of Hepatology and Infectious Diseases, the Second Affiliated Hospital, Shantou University Medical College, Shantou, Guangdong, PR China
| | - Hongqiu Cheng
- Department of Hepatology and Infectious Diseases, the Second Affiliated Hospital, Shantou University Medical College, Shantou, Guangdong, PR China.
| |
Collapse
|
3
|
Yan L, Sun J, Wang Y, Liu X, Hu J, Sun M, Suo X, Duan R, Yuan C. Lin28 affects the proliferation and osteogenic differentiation of human dental pulp stem cells by directly inhibiting let-7b maturation. BDJ Open 2024; 10:17. [PMID: 38443392 PMCID: PMC10914815 DOI: 10.1038/s41405-024-00194-8] [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: 11/08/2023] [Revised: 01/23/2024] [Accepted: 01/27/2024] [Indexed: 03/07/2024] Open
Abstract
OBJECTIVE Activation of Lin28 gene under certain conditions promotes tissue damage repair. However, it remains unknown whether conditional expression of Lin28 facilitates the recovery of damaged pulp tissue. In the study, we focus on exploring the effects and possible regulatory mechanisms of Lin28 on the proliferation and differentiation of human dental pulp stem cells (hDPSCs). MATERIALS AND METHODS We adopted techniques such as the ethynyl-2'-deoxyuridine (EdU) incorporation assay, RNA-protein immunoprecipitation (RIP) analysis, and luciferase assays to study the regulation of hDPSCs by Lin28. Furthermore, gain-of-function and loss-of-function analyses were also used in explored factors regulating hDPSCs activation. RESULTS The results show that Lin28 inhibited osteogenic differentiation by directly targets pre-let-7b. Through bioinformatics sequencing and dual luciferase experiments we learned that let-7b directly targets the IGF2BP2 3'UTR. Silencing of IGF2BP2 showed a similar biological effect as overexpression of let-7b. Overexpression of IGF2BP2 counteracted the differentiation-promoting effects produced by let-7b overexpression. DISCUSSION/CONCLUSIONS In conclusion, the RNA-binding protein Lin28 regulates osteogenic differentiation of hDPSCs by inhibiting let-7 miRNA maturation. And mature let-7b directly regulated the expression of IGF2BP2 by targeting the 3'UTR region of IGF2BP2 mRNA thus further inhibiting the differentiation of hDPSCs.
Collapse
Affiliation(s)
- Liu Yan
- School of Stomatology, Xuzhou Medical University, No.209 Tongshan Road, Xuzhou, 221004, Jiangsu, China
- Affiliated Stomatological Hospital of Xuzhou Medical University, No.130 Huaihai West Road, Xuzhou, 221000, Jiangsu, China
| | - Jing Sun
- School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University, Jinan, 250012, Shandong, China
| | - Yushan Wang
- School of Stomatology, Xuzhou Medical University, No.209 Tongshan Road, Xuzhou, 221004, Jiangsu, China
| | - Xinxin Liu
- School of Stomatology, Xuzhou Medical University, No.209 Tongshan Road, Xuzhou, 221004, Jiangsu, China
| | - Jiayi Hu
- School of Stomatology, Xuzhou Medical University, No.209 Tongshan Road, Xuzhou, 221004, Jiangsu, China
| | - Mengxin Sun
- School of Stomatology, Xuzhou Medical University, No.209 Tongshan Road, Xuzhou, 221004, Jiangsu, China
| | - Xi Suo
- School of Stomatology, Xuzhou Medical University, No.209 Tongshan Road, Xuzhou, 221004, Jiangsu, China
| | - Rongquan Duan
- School of Stomatology, Xuzhou Medical University, No.209 Tongshan Road, Xuzhou, 221004, Jiangsu, China.
| | - Changyong Yuan
- School of Stomatology, Xuzhou Medical University, No.209 Tongshan Road, Xuzhou, 221004, Jiangsu, China.
- Affiliated Stomatological Hospital of Xuzhou Medical University, No.130 Huaihai West Road, Xuzhou, 221000, Jiangsu, China.
| |
Collapse
|
4
|
Jiang C, Zhang C, Dai M, Wang F, Xu S, Han D, Wang Y, Cao Y, Liang Y, Zhang Z, Yan L, Shen Y, He K, Shen Y, Liu J. Interplay between SUMO1-related SUMOylation and phosphorylation of p65 promotes hepatocellular carcinoma progression. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2024; 1871:119595. [PMID: 37730133 DOI: 10.1016/j.bbamcr.2023.119595] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/26/2023] [Revised: 09/10/2023] [Accepted: 09/14/2023] [Indexed: 09/22/2023]
Abstract
The nuclear factor kappaB (NF-κB) subunit p65, plays an important role in the progression of hepatocellular carcinoma (HCC). Phosphorylation of p65 is considered as an important mechanism for the positive regulation of NF-κB activity. According to our previous data, p65 can be SUMOylated by small ubiquitin-related modifier 1 (SUMO1) protein, and SUMO1 promotes p65 nuclear import and HCC progression. However, the effect of SUMO1-related p65 SUMOylation on NF-κB transcriptional activity and the relationship between phosphorylation and SUMOylation of p65 remain obscure. Here, we found that phosphorylated p65 level was increased in cancer tissues of HCC patients, and similar phenomenon was found for SUMO1 expression but not for SUMO2/3. Further clinical data showed a positive correlation between SUMO1 and phosphorylated p65. We also verified that overexpression of SUMO1 upregulated phosphorylated p65 levels. Next, we verified SUMO1-related p65 SUMOylation with in vitro SUMOylation assay, constructed mutants of p65 SUMOylation and phosphorylation, and found that SUMO1-related p65 SUMOylation promoted p65 nuclear import and increased NF-κB activity. Both SUMO1-related p65 SUMOylation and p65 phosphorylation (especially at S276 site) increased the viability and invasion of hepatoma cells, and decreased the apoptosis of hepatoma cells. At last, we found that the phosphorylation of p65 promoted the level of SUMO1-related p65 SUMOylation, and SUMO1-related p65 SUMOylation upregulated phosphorylated p65 (at S276 site). Our study contributes to the exploration of the oncogenic mechanism of p65, which is the important protein in HCC.
Collapse
Affiliation(s)
- Can Jiang
- School of Basic Medical Sciences, Anhui Medical University, Hefei, China; Biopharmaceutical Institute, Anhui Medical University, Hefei, China
| | - Chunyang Zhang
- School of Basic Medical Sciences, Anhui Medical University, Hefei, China; Biopharmaceutical Institute, Anhui Medical University, Hefei, China
| | - Min Dai
- School of Basic Medical Sciences, Anhui Medical University, Hefei, China; Biopharmaceutical Institute, Anhui Medical University, Hefei, China
| | - Fuyan Wang
- School of Basic Medical Sciences, Anhui Medical University, Hefei, China; Biopharmaceutical Institute, Anhui Medical University, Hefei, China
| | - Sa Xu
- School of Basic Medical Sciences, Anhui Medical University, Hefei, China; Biopharmaceutical Institute, Anhui Medical University, Hefei, China
| | - Dan Han
- School of Basic Medical Sciences, Anhui Medical University, Hefei, China; Biopharmaceutical Institute, Anhui Medical University, Hefei, China; Clinical college, Anhui Medical University, Hefei, China
| | - Yanyan Wang
- School of Basic Medical Sciences, Anhui Medical University, Hefei, China; Biopharmaceutical Institute, Anhui Medical University, Hefei, China
| | - Yajie Cao
- School of Basic Medical Sciences, Anhui Medical University, Hefei, China; Biopharmaceutical Institute, Anhui Medical University, Hefei, China
| | - Yanyan Liang
- School of Basic Medical Sciences, Anhui Medical University, Hefei, China; Biopharmaceutical Institute, Anhui Medical University, Hefei, China
| | - Ziyu Zhang
- The First Clinical College, Anhui Medical University, Hefei, China
| | - Lina Yan
- School of Basic Medical Sciences, Anhui Medical University, Hefei, China; Biopharmaceutical Institute, Anhui Medical University, Hefei, China
| | - Yujun Shen
- School of Basic Medical Sciences, Anhui Medical University, Hefei, China; Biopharmaceutical Institute, Anhui Medical University, Hefei, China
| | - Kewu He
- The Third Affiliated Hospital, Anhui Medical University, Hefei, China
| | - Yuxian Shen
- School of Basic Medical Sciences, Anhui Medical University, Hefei, China; Biopharmaceutical Institute, Anhui Medical University, Hefei, China.
| | - Jun Liu
- School of Basic Medical Sciences, Anhui Medical University, Hefei, China; Biopharmaceutical Institute, Anhui Medical University, Hefei, China.
| |
Collapse
|
5
|
Chen X, Zhang Y, Zhang P, Wei M, Tian T, Guan Y, Han C, Wei W, Ma Y. IGFBP2 drives epithelial-mesenchymal transition in hepatocellular carcinoma via activating the Wnt/β-catenin pathway. Infect Agent Cancer 2023; 18:73. [PMID: 37957694 PMCID: PMC10644524 DOI: 10.1186/s13027-023-00543-6] [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: 08/06/2023] [Accepted: 10/04/2023] [Indexed: 11/15/2023] Open
Abstract
Metastasis has emerged as a major impediment to achieve successful therapeutic outcomes in hepatocellular carcinoma (HCC). Nonetheless, the intricate molecular mechanisms governing the progression of HCC remain elusive. Herein, we present evidence highlighting the influence exerted by insulin-like growth factor-binding protein 2 (IGFBP2) as a potent oncogene driving the malignant phenotype. Our investigation reveals a marked elevation of IGFBP2 expression in primary tumors, concomitant with the presence of mesenchymal biomarkers in HCC. Through in vitro and in vivo experimentation, we demonstrate that the overexpression of IGFBP2 expedites the progression of epithelial-mesenchymal transition (EMT) and facilitates the metastatic potential of HCC cells, chiefly mediated by the Wnt/β-catenin signaling pathway. Notably, knockdown of IGFBP2 significantly decreased the expression of total and nuclear β-catenin, N-cadherin and vimentin in the treatment of the specific activator of Wnt/β-catenin CHIR-99021. Collectively, our findings identify IGFBP2 as a pivotal regulator within the HCC EMT axis, whereby its overexpression confers the distinctly aggressive clinical features characteristic of the disease.
Collapse
Grants
- 82104187 National Natural Science Foundation of China
- 82104187 National Natural Science Foundation of China
- 82104187 National Natural Science Foundation of China
- 1308085QH130 the Natural Science Foundation of Anhui Province
- 1308085QH130 the Natural Science Foundation of Anhui Province
- JKZD20212 the Open Project Program of MOE Key Laboratory of Population Health Across Life Cycle
- JKZD20212 the Open Project Program of MOE Key Laboratory of Population Health Across Life Cycle
- KFJJ-2020-12 the Open Fund of Key Laboratory of Anti Inflammatory and Immune Medicine, Ministry of Education
- KFJJ-2020-12 the Open Fund of Key Laboratory of Anti Inflammatory and Immune Medicine, Ministry of Education
- KFJJ-2021-9 the Open Fund of Key Laboratory of Anti Inflammatory and Immune Medicine, Ministry of Education, China
- KFJJ-2021-9 the Open Fund of Key Laboratory of Anti Inflammatory and Immune Medicine, Ministry of Education, China
- AYPYS2021-2 the Key Laboratory of Dermatology, Anhui Medical University, Ministry of Education, China
- AYPYS2021-2 the Key Laboratory of Dermatology, Anhui Medical University, Ministry of Education, China
Collapse
Affiliation(s)
- Xiu Chen
- Institute of Clinical Pharmacology, Key Laboratory of Anti-Inflammatory and Immune Medicine, Ministry of Education, Anhui Collaborative Innovation Center of Anti-Inflammatory and Immune Medicine, Center of Rheumatoid Arthritis of Anhui Medical University, Anhui Medical University, Hefei, 230032, China
| | - Yu Zhang
- Institute of Clinical Pharmacology, Key Laboratory of Anti-Inflammatory and Immune Medicine, Ministry of Education, Anhui Collaborative Innovation Center of Anti-Inflammatory and Immune Medicine, Center of Rheumatoid Arthritis of Anhui Medical University, Anhui Medical University, Hefei, 230032, China
| | - Pingping Zhang
- Institute of Clinical Pharmacology, Key Laboratory of Anti-Inflammatory and Immune Medicine, Ministry of Education, Anhui Collaborative Innovation Center of Anti-Inflammatory and Immune Medicine, Center of Rheumatoid Arthritis of Anhui Medical University, Anhui Medical University, Hefei, 230032, China
| | - Mengzhu Wei
- Institute of Clinical Pharmacology, Key Laboratory of Anti-Inflammatory and Immune Medicine, Ministry of Education, Anhui Collaborative Innovation Center of Anti-Inflammatory and Immune Medicine, Center of Rheumatoid Arthritis of Anhui Medical University, Anhui Medical University, Hefei, 230032, China
| | - Tian Tian
- Institute of Clinical Pharmacology, Key Laboratory of Anti-Inflammatory and Immune Medicine, Ministry of Education, Anhui Collaborative Innovation Center of Anti-Inflammatory and Immune Medicine, Center of Rheumatoid Arthritis of Anhui Medical University, Anhui Medical University, Hefei, 230032, China
| | - Yanling Guan
- Institute of Clinical Pharmacology, Key Laboratory of Anti-Inflammatory and Immune Medicine, Ministry of Education, Anhui Collaborative Innovation Center of Anti-Inflammatory and Immune Medicine, Center of Rheumatoid Arthritis of Anhui Medical University, Anhui Medical University, Hefei, 230032, China
| | - Chenchen Han
- Institute of Clinical Pharmacology, Key Laboratory of Anti-Inflammatory and Immune Medicine, Ministry of Education, Anhui Collaborative Innovation Center of Anti-Inflammatory and Immune Medicine, Center of Rheumatoid Arthritis of Anhui Medical University, Anhui Medical University, Hefei, 230032, China
| | - Wei Wei
- Institute of Clinical Pharmacology, Key Laboratory of Anti-Inflammatory and Immune Medicine, Ministry of Education, Anhui Collaborative Innovation Center of Anti-Inflammatory and Immune Medicine, Center of Rheumatoid Arthritis of Anhui Medical University, Anhui Medical University, Hefei, 230032, China.
| | - Yang Ma
- Institute of Clinical Pharmacology, Key Laboratory of Anti-Inflammatory and Immune Medicine, Ministry of Education, Anhui Collaborative Innovation Center of Anti-Inflammatory and Immune Medicine, Center of Rheumatoid Arthritis of Anhui Medical University, Anhui Medical University, Hefei, 230032, China.
| |
Collapse
|
6
|
Huber K, Szerenos E, Lewandowski D, Toczylowski K, Sulik A. The Role of Adipokines in the Pathologies of the Central Nervous System. Int J Mol Sci 2023; 24:14684. [PMID: 37834128 PMCID: PMC10572192 DOI: 10.3390/ijms241914684] [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: 08/31/2023] [Revised: 09/24/2023] [Accepted: 09/27/2023] [Indexed: 10/15/2023] Open
Abstract
Adipokines are protein hormones secreted by adipose tissue in response to disruptions in physiological homeostasis within the body's systems. The regulatory functions of adipokines within the central nervous system (CNS) are multifaceted and intricate, and they have been identified in a number of pathologies. Therefore, specific adipokines have the potential to be used as biomarkers for screening purposes in neurological dysfunctions. The systematic review presented herein focuses on the analysis of the functions of various adipokines in the pathogenesis of CNS diseases. Thirteen proteins were selected for analysis through scientific databases. It was found that these proteins can be identified within the cerebrospinal fluid either by their ability to modify their molecular complex and cross the blood-brain barrier or by being endogenously produced within the CNS itself. As a result, this can correlate with their measurability during pathological processes, including Alzheimer's disease, amyotrophic lateral sclerosis, multiple sclerosis, depression, or brain tumors.
Collapse
Affiliation(s)
| | | | | | - Kacper Toczylowski
- Department of Pediatric Infectious Diseases, Medical University of Bialystok, Waszyngtona 17, 15-274 Bialystok, Poland
| | | |
Collapse
|
7
|
Gupta R, Kadhim MM, Turki Jalil A, Obayes AM, Aminov Z, Alsaikhan F, Ramírez-Coronel AA, Ramaiah P, Tayyib NA, Luo X. Multifaceted role of NF-κB in hepatocellular carcinoma therapy: Molecular landscape, therapeutic compounds and nanomaterial approaches. ENVIRONMENTAL RESEARCH 2023; 228:115767. [PMID: 36966991 DOI: 10.1016/j.envres.2023.115767] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/04/2023] [Revised: 03/20/2023] [Accepted: 03/22/2023] [Indexed: 05/16/2023]
Abstract
The predominant kind of liver cancer is hepatocellular carcinoma (HCC) that its treatment have been troublesome difficulties for physicians due to aggressive behavior of tumor cells in proliferation and metastasis. Moreover, stemness of HCC cells can result in tumor recurrence and angiogenesis occurs. Another problem is development of resistance to chemotherapy and radiotherapy in HCC cells. Genomic mutations participate in malignant behavior of HCC and nuclear factor-kappaB (NF-κB) has been one of the oncogenic factors in different human cancers that after nuclear translocation, it binds to promoter of genes in regulating their expression. Overexpression of NF-κB has been well-documented in increasing proliferation and invasion of tumor cells and notably, when its expression enhances, it induces chemoresistance and radio-resistance. Highlighting function of NF-κB in HCC can shed some light on the pathways regulating progression of tumor cells. The first aspect is proliferation acceleration and apoptosis inhibition in HCC cells mediated by enhancement in expression level of NF-κB. Moreover, NF-κB is able to enhance invasion of HCC cells via upregulation of MMPs and EMT, and it triggers angiogenesis as another step for increasing spread of tumor cells in tissues and organs. When NF-κB expression enhances, it stimulates chemoresistance and radio-resistance in HCC cells and by increasing stemness and population of cancer-stem cells, it can provide the way for recurrence of tumor. Overexpression of NF-κB mediates therapy resistance in HCC cells and it can be regulated by non-coding RNAs in HCC. Moreover, inhibition of NF-κB by anti-cancer and epigenetic drugs suppresses HCC tumorigenesis. More importantly, nanoparticles are considered for suppressing NF-κB axis in cancer and their prospectives and results can also be utilized for treatment of HCC. Nanomaterials are promising factors in treatment of HCC and by delivery of genes and drugs, they suppress HCC progression. Furthermore, nanomaterials provide phototherapy in HCC ablation.
Collapse
Affiliation(s)
- Reena Gupta
- Institute of Pharmaceutical Research, GLA University, District-Mathura, U. P., India
| | - Mustafa M Kadhim
- Department of Dentistry, Kut University College, Kut, Wasit, 52001, Iraq; Medical Laboratory Techniques Department, Al-Farahidi University, Baghdad, 10022, Iraq
| | - Abduladheem Turki Jalil
- Medical Laboratories Techniques Department, Al-Mustaqbal University College, Babylon, Hilla, 51001, Iraq.
| | | | - Zafar Aminov
- Department of Public Health and Healthcare Management, Samarkand State Medical University, 18 Amir Temur Street, Samarkand, Uzbekistan; Department of Scientific Affairs, Tashkent State Dental Institute, 103 Makhtumkuli Str., Tashkent, Uzbekistan
| | - Fahad Alsaikhan
- College of Pharmacy, Prince Sattam Bin Abdulaziz University, Alkharj, Saudi Arabia.
| | - 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
| | - Xuanming Luo
- Department of General Surgery, Shanghai Xuhui Central Hospital, Fudan University, Shanghai, 200031, China.
| |
Collapse
|
8
|
Li Q, Tan G, Wu F. The functions and roles of C2H2 zinc finger proteins in hepatocellular carcinoma. Front Physiol 2023; 14:1129889. [PMID: 37457025 PMCID: PMC10339807 DOI: 10.3389/fphys.2023.1129889] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Accepted: 06/20/2023] [Indexed: 07/18/2023] Open
Abstract
C2H2 zinc finger (C2H2-ZF) proteins are the majority group of human transcription factors and they have many different molecular functions through different combinations of zinc finger domains. Hepatocellular carcinoma (HCC) is one of the most prevalent malignant tumors and the main reason for cancer-related deaths worldwide. More and more findings support the abnormal expression of C2H2-ZF protein in the onset and progression of HCC. The C2H2-ZF proteins are involved in various biological functions in HCC, such as EMT, stemness maintenance, metabolic reprogramming, cell proliferation and growth, apoptosis, and genomic integrity. The study of anti-tumor drug resistance also highlights the pivotal roles of C2H2-ZF proteins at the intersection of biological functions (EMT, stemness maintenance, autophagy)and chemoresistance in HCC. The involvement of C2H2-ZF protein found recently in regulating different molecules, signal pathways and pathophysiological activities indicate these proteins as the possible therapeutic targets, and diagnostic or prognostic biomarkers for HCC.
Collapse
|
9
|
IGFBP2 derived from PO-MSCs promote epithelial barrier destruction by activating FAK signaling in nasal polyps. iScience 2023; 26:106151. [PMID: 36866245 PMCID: PMC9972572 DOI: 10.1016/j.isci.2023.106151] [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: 06/08/2022] [Revised: 11/19/2022] [Accepted: 02/02/2023] [Indexed: 02/09/2023] Open
Abstract
The nasal polyps (NPs) microenvironment comprises multiple cell types, including mesenchymal stromal cells (MSCs). Insulin-like growth factor binding protein 2 (IGFBP2) plays crucial roles in cell proliferation, differentiation and more. However, the role of NPs-derived MSCs (PO-MSCs) and IGFBP2 in NPs pathogenesis remains poorly defined. Herein, primary human nasal epithelial cells (pHNECs) and MSCs were extracted and cultured. Extracellular vesicles (EVs) and soluble proteins were isolated to investigate the role of PO-MSCs on epithelial-mesenchymal transition (EMT) and epithelial barrier function in NPs. Our data showed that IGFBP2, but not EVs from PO-MSCs (PO-MSCs-EVs), exhibited a crucial role in EMT and barrier destruction. Moreover, focal adhesion kinase (FAK) signaling pathway is necessary for IGFBP2 to exert its functions in human and mice nasal epithelial mucosa. Altogether, these findings may improve the current understanding of the role of PO-MSCs in NPs microenvironment and ultimately contribute to the prevention and treatment of NPs.
Collapse
|
10
|
Rapp E, Lu Z, Sun L, Serna SN, Almestica-Roberts M, Burrell KL, Nguyen ND, Deering-Rice CE, Reilly CA. Mechanisms and Consequences of Variable TRPA1 Expression by Airway Epithelial Cells: Effects of TRPV1 Genotype and Environmental Agonists on Cellular Responses to Pollutants in Vitro and Asthma. ENVIRONMENTAL HEALTH PERSPECTIVES 2023; 131:27009. [PMID: 36847817 PMCID: PMC9969990 DOI: 10.1289/ehp11076] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Revised: 01/20/2023] [Accepted: 01/20/2023] [Indexed: 06/18/2023]
Abstract
BACKGROUND Transient receptor potential ankyrin-1 [transient receptor potential cation channel subfamily A member 1 (TRPA1)] and vanilloid-1 [transient receptor potential cation channel subfamily V member 1 (TRPV1)] detect inhaled irritants, including air pollutants and have roles in the development and exacerbation of asthma. OBJECTIVES This study tested the hypothesis that increased expression of TRPA1, stemming from expression of the loss-of-function TRPV1 (I585V; rs8065080) polymorphic variant by airway epithelial cells may explain prior observations of worse asthma symptom control among children with the TRPV1 I585I/V genotype, by virtue of sensitizing epithelial cells to particulate materials and other TRPA1 agonists. METHODS TRP agonists, antagonists, small interfering RNA (siRNA), a nuclear factor kappa light chain enhancer of activated B cells (NF-κB) pathway inhibitor, and kinase activators and inhibitors were used to modulate TRPA1 and TRPV1 expression and function. Treatment of genotyped airway epithelial cells with particulate materials and analysis of asthma control data were used to assess consequences of TRPV1 genotype and variable TRPA1 expression on cellular responses in vitro and asthma symptom control among children as a function of voluntarily reported tobacco smoke exposure. RESULTS A relationship between higher TRPA1 expression and function and lower TRPV1 expression and function was revealed. Findings of this study pointed to a mechanism whereby NF-κB promoted TRPA1 expression, whereas NF-κB-regulated nucleotide-binding oligomerization domain, leucine rich repeat and pyrin domain containing 2 (NLRP2) limited expression. Roles for protein kinase C and p38 mitogen activated protein kinase were also demonstrated. Finally, the TRPV1 I585I/V genotype was associated with increased TRPA1 expression by primary airway epithelial cells and amplified responses to selected air pollution particles in vitro. However, the TRPV1 I585I/V genotype was not associated with worse asthma symptom control among children exposed to tobacco smoke, whereas other TRPA1 and TRPV1 variants were. DISCUSSION This study provides insights on how airway epithelial cells regulate TRPA1 expression, how TRPV1 genetics can affect TRPA1 expression, and that TRPA1 and TRPV1 polymorphisms differentially affect asthma symptom control. https://doi.org/10.1289/EHP11076.
Collapse
Affiliation(s)
- Emmanuel Rapp
- Department of Pharmacology and Toxicology, Center for Human Toxicology, University of Utah, Salt Lake City, Utah, USA
| | - Zhenyu Lu
- Department of Pharmacology and Toxicology, Center for Human Toxicology, University of Utah, Salt Lake City, Utah, USA
| | - Lili Sun
- Department of Pharmacology and Toxicology, Center for Human Toxicology, University of Utah, Salt Lake City, Utah, USA
| | - Samantha N. Serna
- Department of Pharmacology and Toxicology, Center for Human Toxicology, University of Utah, Salt Lake City, Utah, USA
| | - Marysol Almestica-Roberts
- Department of Pharmacology and Toxicology, Center for Human Toxicology, University of Utah, Salt Lake City, Utah, USA
| | - Katherine L. Burrell
- Department of Pharmacology and Toxicology, Center for Human Toxicology, University of Utah, Salt Lake City, Utah, USA
| | - Nam D. Nguyen
- Department of Pharmacology and Toxicology, Center for Human Toxicology, University of Utah, Salt Lake City, Utah, USA
| | - Cassandra E. Deering-Rice
- Department of Pharmacology and Toxicology, Center for Human Toxicology, University of Utah, Salt Lake City, Utah, USA
| | - Christopher A. Reilly
- Department of Pharmacology and Toxicology, Center for Human Toxicology, University of Utah, Salt Lake City, Utah, USA
| |
Collapse
|
11
|
Zhang B, Hong C, Luo Y, Wei L, Luo Y, Peng Y, Xu Y. Prognostic value of IGFBP2 in various cancers: a systematic review and meta-analysis. Cancer Med 2022; 11:3035-3047. [PMID: 35546443 PMCID: PMC9385590 DOI: 10.1002/cam4.4680] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Revised: 02/21/2022] [Accepted: 03/04/2022] [Indexed: 02/05/2023] Open
Abstract
BACKGROUND The prognostic significance of insulin-like growth factor binding protein 2 (IGFBP2) expression has been explored in plenty of studies in human cancers. Because of the controversial results, the meta-analysis was carried out to evaluate the relevance of IGFBP2 expression with the prognosis in various tumors. METHODS The data searched from four databases (Pubmed, Embase, Cochrane library, and Web of science) was used to calculate pooled hazard ratios (HRs) in this meta-analysis. Subgroup analyses were stratified by ethnicity, cancer type, publication year, Newcastle-Ottawa Scale score, treatments, and populations. RESULTS Twenty-one studies containing 5560 patients finally met inclusion criteria. IGFBP2 expression was associated with lower overall survival (HR = 1.57, 95% CI = 1.31-1.88) and progression-free survival (HR = 1.18, 95% CI = 1.04-1.34) in cancer patients, but not with disease-free survival (HR = 1.50, 95% CI = 0.91-2.46) or recurrence-free survival (HR = 1.50, 95% CI = 0.93-2.40). The subgroup analyses indicated IGFBP2 overexpression was significantly correlated with overall survival in Asian patients (HR = 1.42, 95% CI = 1.18-1.72), Caucasian patients (HR = 2.20, 95% CI = 1.31-3.70), glioma (HR = 1.36, 95% CI = 1.03-1.79), and colorectal cancer (HR = 2.52, 95% CI = 1.43-4.44) and surgery subgroups (HR = 1.97, 95% CI = 1.50-2.58). CONCLUSION The meta-analysis showed that IGFBP2 expression was associated with worse prognosis in several tumors, and may serve as a potential prognostic biomarker in cancer patients.
Collapse
Affiliation(s)
- Biao Zhang
- Department of Clinical Laboratory MedicineCancer Hospital of Shantou University Medical CollegeShantouChina
- Precision Medicine Research Center, Shantou University Medical CollegeShantouChina
- Guangdong Esophageal Cancer Institutethe Cancer Hospital of Shantou University Medical CollegeShantouChina
| | - Chao‐Qun Hong
- Provincial Key Laboratory of Guangdong Breast Cancer Diagnosis and TreatmentCancer Hospital of Shantou University Medical CollegeShantouChina
| | - Yu‐Hao Luo
- Department of Clinical Laboratory MedicineCancer Hospital of Shantou University Medical CollegeShantouChina
- Precision Medicine Research Center, Shantou University Medical CollegeShantouChina
| | - Lai‐Feng Wei
- Department of Clinical Laboratory MedicineCancer Hospital of Shantou University Medical CollegeShantouChina
- Precision Medicine Research Center, Shantou University Medical CollegeShantouChina
- Guangdong Esophageal Cancer Institutethe Cancer Hospital of Shantou University Medical CollegeShantouChina
| | - Yun Luo
- Department of Clinical Laboratory MedicineCancer Hospital of Shantou University Medical CollegeShantouChina
- Precision Medicine Research Center, Shantou University Medical CollegeShantouChina
- Guangdong Esophageal Cancer Institutethe Cancer Hospital of Shantou University Medical CollegeShantouChina
| | - Yu‐Hui Peng
- Department of Clinical Laboratory MedicineCancer Hospital of Shantou University Medical CollegeShantouChina
- Precision Medicine Research Center, Shantou University Medical CollegeShantouChina
- Guangdong Esophageal Cancer Institutethe Cancer Hospital of Shantou University Medical CollegeShantouChina
| | - Yi‐Wei Xu
- Department of Clinical Laboratory MedicineCancer Hospital of Shantou University Medical CollegeShantouChina
- Precision Medicine Research Center, Shantou University Medical CollegeShantouChina
- Guangdong Esophageal Cancer Institutethe Cancer Hospital of Shantou University Medical CollegeShantouChina
| |
Collapse
|
12
|
Li LY, Yang JF, Rong F, Luo ZP, Hu S, Fang H, Wu Y, Yao R, Kong WH, Feng XW, Chen BJ, Li J, Xu T. ZEB1 serves an oncogenic role in the tumourigenesis of HCC by promoting cell proliferation, migration, and inhibiting apoptosis via Wnt/β-catenin signaling pathway. Acta Pharmacol Sin 2021; 42:1676-1689. [PMID: 33514855 PMCID: PMC8463676 DOI: 10.1038/s41401-020-00575-3] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Accepted: 11/05/2020] [Indexed: 02/06/2023] Open
Abstract
Zinc finger E-box-binding homeobox 1 (ZEB1), a functional protein of zinc finger family, was aberrant expressed in many kinds of liver disease including hepatic fibrosis and Hepatitis C virus. Bioinformatics results showed that ZEB1 was abnormally expressed in HCC tissues. However, to date, the potential regulatory role and molecular mechanisms of ZEB1 are still unclear in the occurrence and development of HCC. This study demonstrated that the expression level of ZEB1 was significantly elevated both in liver tissues of HCC patients and cell lines (HepG2 and SMMC-7721 cells). Moreover, ZEB1 could promote the proliferation, migration, and invasion of HCC cells. On the downstream regulation mechanism, ZEB1 could activate the Wnt/β-catenin signaling pathway by upregulating the protein expression levels of β-catenin, c-Myc, and cyclin D1. Novel studies showed that miR-708 particularly targeted ZEB1 3'-UTR regions and inhibited the HCC cell proliferation, migration, and invasion. Furthermore, results of nude mice experiments of HCC model indicated that miR-708 could inhibit tumor growth and xenograft metastasis model was established to validate that miR-708 could inhibit HCC cell metastasis through tail-vein injection in vivo. Together, the study suggested that ZEB1 modulated by miR-708 might be a potential therapeutic target for HCC therapy.
Collapse
Affiliation(s)
- Liang-Yun Li
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, 230032, China
- Institute for Liver Diseases of Anhui Medical University, Hefei, 230032, China
| | - Jun-Fa Yang
- Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Institute of Clinical Pharmacology, Anhui Medical University, Hefei, 230032, China
| | - Fan Rong
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, 230032, China
- Institute for Liver Diseases of Anhui Medical University, Hefei, 230032, China
- Lujiang County People's Hospital of Anhui Province, Hefei, 231500, China
| | - Zhi-Pan Luo
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, 230032, China
- Institute for Liver Diseases of Anhui Medical University, Hefei, 230032, China
- The First Affiliated Hospital of Anhui Medical University, Hefei, 230032, China
| | - Shuang Hu
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, 230032, China
- Institute for Liver Diseases of Anhui Medical University, Hefei, 230032, China
| | - Hui Fang
- Department of Pharmocology, The Affiliated Hospital of Hangzhou Normal University, Hangzhou, 310015, China
| | - Ying Wu
- The First Affiliated Hospital of Anhui Medical University, Hefei, 230032, China
| | - Rui Yao
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, 230032, China
- Institute for Liver Diseases of Anhui Medical University, Hefei, 230032, China
- Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Institute of Clinical Pharmacology, Anhui Medical University, Hefei, 230032, China
| | - Wei-Hao Kong
- The First Affiliated Hospital of Anhui Medical University, Hefei, 230032, China
| | - Xiao-Wen Feng
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, 230032, China
- Institute for Liver Diseases of Anhui Medical University, Hefei, 230032, China
| | - Bang-Jie Chen
- First Clinical Medical College of Anhui Medical University, Hefei, 230032, China
| | - Jun Li
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, 230032, China
- Institute for Liver Diseases of Anhui Medical University, Hefei, 230032, China
| | - Tao Xu
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, 230032, China.
- Institute for Liver Diseases of Anhui Medical University, Hefei, 230032, China.
| |
Collapse
|
13
|
Vizoso M, van Rheenen J. Diverse transcriptional regulation and functional effects revealed by CRISPR/Cas9-directed epigenetic editing. Oncotarget 2021; 12:1651-1662. [PMID: 34434494 PMCID: PMC8378768 DOI: 10.18632/oncotarget.28037] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2020] [Accepted: 07/27/2021] [Indexed: 01/04/2023] Open
Abstract
DNA methylation is an epigenetic process that controls DNA accessibility and serves as a transcriptomic switch when deposited at regulatory regions. The adequate functioning of this process is indispensable for tissue homeostasis and cell fate determination. Conversely, altered DNA methylation patterns result in abnormal gene transcription profiles that contribute to tumor initiation and progression. However, whether the consequence of DNA methylation on gene expression and cell fate is uniform regardless of the cell type or state could so far not been tested due to the lack of technologies to target DNA methylation in-situ. Here, we have taken advantage of CRISPR/dCas9 technology adapted for epigenetic editing through site-specific targeting of DNA methylation to characterize the transcriptional changes of the candidate gene and the functional effects on cell fate in different tumor settings. As a proof-of-concept, we were able to induce de-novo site-specific methylation of the gene promoter of IGFBP2 up to 90% with long-term and bona-fide inheritance by daughter cells. Strikingly, this modification led to opposing expression profiles of the target gene in different cancer cell models and affected the expression of mesenchymal genes CDH1, VIM1, TGFB1 and apoptotic marker BCL2. Moreover, methylation-induced changes in expression profiles was also accompanied by a phenotypic switch in cell migration and cell morphology. We conclude that in different cell types the consequence of DNA methylation on gene expression and cell fate can be completely different.
Collapse
Affiliation(s)
- Miguel Vizoso
- Division of Molecular Pathology, Oncode Institute, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Jacco van Rheenen
- Division of Molecular Pathology, Oncode Institute, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| |
Collapse
|
14
|
Ngo MHT, Jeng HY, Kuo YC, Nanda JD, Brahmadhi A, Ling TY, Chang TS, Huang YH. The Role of IGF/IGF-1R Signaling in Hepatocellular Carcinomas: Stemness-Related Properties and Drug Resistance. Int J Mol Sci 2021; 22:ijms22041931. [PMID: 33669204 PMCID: PMC7919800 DOI: 10.3390/ijms22041931] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Revised: 02/10/2021] [Accepted: 02/10/2021] [Indexed: 12/12/2022] Open
Abstract
Insulin-like Growth Factor (IGF)/IGF-1 Receptor (IGF-1R) signaling is known to regulate stem cell pluripotency and differentiation to trigger cell proliferation, organ development, and tissue regeneration during embryonic development. Unbalanced IGF/IGF-1R signaling can promote cancer cell proliferation and activate cancer reprogramming in tumor tissues, especially in the liver. Hepatocellular carcinoma (HCC) is one of the leading causes of cancer-related death, with a high incidence and mortality rate in Asia. Most patients with advanced HCC develop tyrosine kinase inhibitor (TKI)-refractoriness after receiving TKI treatment. Dysregulation of IGF/IGF-1R signaling in HCC may activate expression of cancer stemness that leads to TKI refractoriness and tumor recurrence. In this review, we summarize the evidence for dysregulated IGF/IGF-1R signaling especially in hepatitis B virus (HBV)-associated HCC. The regulation of cancer stemness expression and drug resistance will be highlighted. Current clinical treatments and potential therapies targeting IGF/IGF-1R signaling for the treatment of HCC will be discussed.
Collapse
Affiliation(s)
- Mai-Huong Thi Ngo
- International PhD Program for Cell Therapy and Regeneration Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan; (M.-H.T.N.); (J.D.N.); (A.B.)
- Department of Biochemistry and Molecular Cell Biology, School of Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan
| | - Han-Yin Jeng
- Research Center of Cell Therapy and Regeneration Medicine, Taipei Medical University, Taipei 11031, Taiwan; (H.-Y.J.); (Y.-C.K.)
| | - Yung-Che Kuo
- Research Center of Cell Therapy and Regeneration Medicine, Taipei Medical University, Taipei 11031, Taiwan; (H.-Y.J.); (Y.-C.K.)
| | - Josephine Diony Nanda
- International PhD Program for Cell Therapy and Regeneration Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan; (M.-H.T.N.); (J.D.N.); (A.B.)
| | - Ageng Brahmadhi
- International PhD Program for Cell Therapy and Regeneration Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan; (M.-H.T.N.); (J.D.N.); (A.B.)
| | - Thai-Yen Ling
- Department and Graduate Institute of Pharmacology, National Taiwan University, Taipei 11031, Taiwan
- Correspondence: (T.-Y.L.); (T.-S.C.); (Y.-H.H.); Tel.: +886-2-2312-3456 (ext. 8-8322) (T.-Y.L.); +886-5-3621-000 (ext. 2242) (T.-S.C.); +886-2-2736-1661 (ext. 3150) (Y.-H.H.)
| | - Te-Sheng Chang
- School of Traditional Chinese Medicine, College of Medicine, Chang Gung University, Taoyuan 33382, Taiwan
- Division of Internal Medicine, Department of Gastroenterology and Hepatology, Chang Gung Memorial Hospital, Chiayi 61363, Taiwan
- Correspondence: (T.-Y.L.); (T.-S.C.); (Y.-H.H.); Tel.: +886-2-2312-3456 (ext. 8-8322) (T.-Y.L.); +886-5-3621-000 (ext. 2242) (T.-S.C.); +886-2-2736-1661 (ext. 3150) (Y.-H.H.)
| | - Yen-Hua Huang
- International PhD Program for Cell Therapy and Regeneration Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan; (M.-H.T.N.); (J.D.N.); (A.B.)
- Department of Biochemistry and Molecular Cell Biology, School of Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan
- Research Center of Cell Therapy and Regeneration Medicine, Taipei Medical University, Taipei 11031, Taiwan; (H.-Y.J.); (Y.-C.K.)
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan
- Center for Reproductive Medicine, Taipei Medical University Hospital, Taipei 11031, Taiwan
- Comprehensive Cancer Center, Taipei Medical University, Taipei 11031, Taiwan
- Research Center of Cancer Translational Medicine, Taipei Medical University, Taipei 11031, Taiwan
- PhD Program for Translational Medicine, College of Medical Science and Technology, Taipei Medical University, Taipei 11031, Taiwan
- Correspondence: (T.-Y.L.); (T.-S.C.); (Y.-H.H.); Tel.: +886-2-2312-3456 (ext. 8-8322) (T.-Y.L.); +886-5-3621-000 (ext. 2242) (T.-S.C.); +886-2-2736-1661 (ext. 3150) (Y.-H.H.)
| |
Collapse
|
15
|
Feder S, Bruckmann A, McMullen N, Sinal CJ, Buechler C. Chemerin Isoform-Specific Effects on Hepatocyte Migration and Immune Cell Inflammation. Int J Mol Sci 2020; 21:ijms21197205. [PMID: 33003572 PMCID: PMC7582997 DOI: 10.3390/ijms21197205] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Revised: 09/25/2020] [Accepted: 09/28/2020] [Indexed: 02/07/2023] Open
Abstract
Murine chemerin is C-terminally processed to the bioactive isoforms, muChem-156 and muChem-155, among which the longer variant protects from hepatocellular carcinoma (HCC). However, the role of muChem-155 is mostly unknown. Here, we aimed to compare the effects of these isoforms on the proliferation, migration and the secretome of the human hepatocyte cell lines HepG2 and Huh7 and the murine Hepa1-6 cell line. Therefore, huChem-157 and -156 were overexpressed in the human cells, and the respective murine variants, muChem-156 and -155, in the murine hepatocytes. Both chemerin isoforms produced by HepG2 and Hepa1-6 cells activated the chemerin receptors chemokine-like receptor 1 (CMKLR1) and G protein-coupled receptor 1 (GPR1). HuChem-157 was the active isoform in the Huh7 cell culture medium. The potencies of muChem-155 and muChem-156 to activate human GPR1 and mouse CMKLR1 were equivalent. Human CMKLR1 was most responsive to muChem-156. Chemerin variants showed no effect on cell viability and proliferation. Activation of the mitogen-activated protein kinases Erk1/2 and p38, and protein levels of the epithelial–mesenchymal transition marker, E-cadherin, were not regulated by the chemerin variants. Migration was reduced in HepG2 and Hepa1-6 cells by the longer isoform. Protective effects of chemerin in HCC include the modulation of cytokines but huChem-156 and huChem-157 overexpression did not change IL-8, CCL20 or osteopontin in the hepatocytes. The conditioned medium of the transfected hepatocytes failed to alter these soluble factors in the cell culture medium of peripheral blood mononuclear cells (PBMCs). Interestingly, the cell culture medium of Huh7 cells producing the inactive variant huChem-155 reduced CCL2 and IL-8 in PBMCs. To sum up, huChem-157 and muChem-156 inhibited hepatocyte migration and may protect from HCC metastasis. HuChem-155 was the only human isoform exerting anti-inflammatory effects on immune cells.
Collapse
Affiliation(s)
- Susanne Feder
- Department of Internal Medicine I, Regensburg University Hospital, 93053 Regensburg, Germany;
| | - Astrid Bruckmann
- Biochemistry Center Regensburg (BZR), Laboratory for RNA Biology, University of Regensburg, 93042 Regensburg, Germany;
| | - Nichole McMullen
- Department of Pharmacology, Dalhousie University, Halifax, NS B3H 4R2, Canada; (N.M.); (C.J.S.)
| | - Christopher J. Sinal
- Department of Pharmacology, Dalhousie University, Halifax, NS B3H 4R2, Canada; (N.M.); (C.J.S.)
| | - Christa Buechler
- Department of Internal Medicine I, Regensburg University Hospital, 93053 Regensburg, Germany;
- Correspondence: ; Tel.: +49-941-944-7009
| |
Collapse
|
16
|
Du Y, Gao Y. Development and validation of a novel pseudogene pair-based prognostic signature for prediction of overall survival in patients with hepatocellular carcinoma. BMC Cancer 2020; 20:887. [PMID: 32938429 PMCID: PMC7493157 DOI: 10.1186/s12885-020-07391-2] [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: 05/27/2020] [Accepted: 09/08/2020] [Indexed: 12/27/2022] Open
Abstract
BACKGROUND There is growing evidence that pseudogenes may serve as prognostic biomarkers in several cancers. The present study was designed to develop and validate an accurate and robust pseudogene pairs-based signature for the prognosis of hepatocellular carcinoma (HCC). METHODS RNA-sequencing data from 374 HCC patients with clinical follow-up information were obtained from the Cancer Genome Atlas (TCGA) database and used in this study. Survival-related pseudogene pairs were identified, and a signature model was constructed by Cox regression analysis (univariate and least absolute shrinkage and selection operator). All individuals were classified into high- and low-risk groups based on the optimal cutoff. Subgroups analysis of the novel signature was conducted and validated in an independent cohort. Pearson correlation analyses were carried out between the included pseudogenes and the protein-coding genes based on their expression levels. Enrichment analysis was performed to predict the possible role of the pseudogenes identified in the signature. RESULTS A 19-pseudogene pair signature, which included 21 pseudogenes, was established. Patients in high-risk group demonstrated an increased the risk of adverse prognosis in the TCGA cohort and the external cohort (all P < 0.001). The novel pseudogene signature was independent of other conventional clinical variables used for survival prediction in HCC patients in the two cohorts revealed by the multivariate Cox regression analysis (all P < 0.001). Subgroup analysis further demonstrated the diagnostic value of the signature across different stages, grades, sexes, and age groups. The C-index of the prognostic signature was 0.761, which was not only higher than that of several previous risk models but was also much higher than that of a single age, sex, grade, and stage risk model. Furthermore, functional analysis revealed that the potential biological mechanisms mediated by these pseudogenes are primarily involved in cytokine receptor activity, T cell receptor signaling, chemokine signaling, NF-κB signaling, PD-L1 expression, and the PD-1 checkpoint pathway in cancer. CONCLUSION The novel proposed and validated pseudogene pair-based signature may serve as a valuable independent prognostic predictor for predicting survival of patients with HCC.
Collapse
Affiliation(s)
- Yajuan Du
- Department of structural heart disease, the First Affiliated Hospital of Xi'an Jiaotong University, No.277, Yanta West Road, Xi'an, 710061, Shaanxi, People's Republic of China.
| | - Ying Gao
- Department of Radiotherapy Oncology, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, China
| |
Collapse
|