1
|
Ko YG, Jo JH, Song SY, Lee HS. The crucial role of CEMIP in cancer metastasis: Mechanistic insights and clinical implications. FASEB J 2025; 39:e70284. [PMID: 39758005 PMCID: PMC11701794 DOI: 10.1096/fj.202402522r] [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: 10/22/2024] [Revised: 12/04/2024] [Accepted: 12/20/2024] [Indexed: 01/07/2025]
Abstract
Cancer metastasis is the leading cause of cancer-related deaths, making early detection and the prevention of metastatic progression critical research priorities. Recent studies have expanded our understanding of CEMIP (KIAA1199, HYBID), revealing its involvement in cancer metastasis and its potential role in slowing cancer progression. CEMIP plays critical roles in several stages of cancer metastasis: First, CEMIP promotes cancer cell proliferation to maintain cell heterogeneity before the metastasis process. Second, it facilitates cancer cell detachment by promoting the epithelial-mesenchymal transition (EMT) through alterations in signaling pathways. Third, CEMIP contributes to cancer cell adherence and attachment by enabling cells to withstand cell death (anoikis and ferroptosis) and hypoxia. Fourth, during the invasion process, CEMIP induces hyaluronan depolymerization and further modulates signaling to promote EMT. Lastly, in the pre-metastatic niche, CEMIP influences the tumor microenvironment through hypoxia, angiogenesis, signaling pathway changes, and hyaluronan degradation. Recent studies have focused on leveraging CEMIP as a diagnostic tool or a predictor of metastasis and/or targeting CEMIP to overcome cancer resistance and progression. This review aims to explore the role of CEMIP at each stage of cancer metastasis and highlight recent advances in targeting CEMIP to inhibit cancer progression.
Collapse
Affiliation(s)
- Yeo Gyeong Ko
- Division of Gastroenterology, Department of Internal MedicineYonsei University College of MedicineSeoulRepublic of Korea
| | - Jung Hyun Jo
- Division of Gastroenterology, Department of Internal MedicineYonsei University College of MedicineSeoulRepublic of Korea
- Institute of GastroenterologyYonsei University College of MedicineSeoulRepublic of Korea
| | - Si Young Song
- Division of Gastroenterology, Department of Internal MedicineYonsei University College of MedicineSeoulRepublic of Korea
- Institute of GastroenterologyYonsei University College of MedicineSeoulRepublic of Korea
| | - Hee Seung Lee
- Division of Gastroenterology, Department of Internal MedicineYonsei University College of MedicineSeoulRepublic of Korea
- Institute of GastroenterologyYonsei University College of MedicineSeoulRepublic of Korea
| |
Collapse
|
2
|
Zha Y, Luo X, Ge Z, Zhang J, Li Y, Zhang S. KIAA1199/CEMIP knockdown attenuates cardiac remodeling post myocardial infarction by activating TSP4 pathway in mice. Biochim Biophys Acta Mol Basis Dis 2024; 1870:167473. [PMID: 39173890 DOI: 10.1016/j.bbadis.2024.167473] [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: 03/07/2024] [Revised: 08/04/2024] [Accepted: 08/14/2024] [Indexed: 08/24/2024]
Abstract
BACKGROUND Excessive activation of cardiac fibroblasts (CFs) significantly contributes to adverse cardiac remodeling post-myocardial infarction (MI). CEMIP, initially recognized as an enzyme involved in hyaluronic acid (HA) degradation, has also been implicated in the activation of pulmonary fibroblasts. Nevertheless, the role and mechanism of CEMIP in adverse cardiac remodeling following MI remain largely unexplored. MATERIALS AND METHODS RNA sequencing (RNA-seq) was performed on cardiac tissue harvested from the infarct/peri-infarct region of mice 28 days post-MI. RNA-seq was conducted on primary cardiac fibroblasts (CFs) transfected with adenovirus overexpressing CEMIP. Adeno-associated virus serotype 9 (AAV9) was engineered for in vivo CEMIP knockdown to elucidate its impact on cardiac remodeling. Immunoprecipitation coupled with mass spectrometry (IP-MS) and co-immunoprecipitation (co-IP) were employed to elucidate the mechanism by which CEMIP affected cardiac remodeling. KEY FINDINGS RNA-seq of fibrotic heart tissue at day 28 post-MI revealed a significant upregulation of CEMIP. In vitro, CEMIP facilitated the activation of cardiac fibroblasts. In vivo, knockdown of CEMIP markedly reduced cardiac fibrosis and improved cardiac function post-MI. IP-MS and co-immunoprecipitation (co-IP) confirmed that CEMIP interacted with TSP4 through the G8 domain. Further experiments confirmed that CEMIP promoted TSP4 degradation in lysosomes in an ACTN4-dependent manner, thereby activating the FAK signaling pathway. SIGNIFICANCE Our findings suggest that CEMIP significantly contributes to cardiac remodeling post-MI, which might be a novel approach for treating cardiac fibrosis following MI.
Collapse
Affiliation(s)
- Yafang Zha
- Department of Cardiology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, 160 Pujian Road, Shanghai, China
| | - Xueyang Luo
- Institute for Developmental and Regenerative Cardiovascular Medicine, Xinhua Hospital, Shanghai Jiao Tong University, School of Medicine, Shanghai, China
| | - Zhuowang Ge
- Department of Cardiology, Xinhua School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Jiayan Zhang
- Department of Cardiology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, 160 Pujian Road, Shanghai, China
| | - Yanyan Li
- Department of Cardiology, Xinhua School of Medicine, Shanghai Jiao Tong University, Shanghai, China.
| | - Song Zhang
- Department of Cardiology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, 160 Pujian Road, Shanghai, China.
| |
Collapse
|
3
|
Peters A, Banine F, Yasuhara K, Hoffman A, Basappa, Metri PK, Gunning L, Huffman A, VanCampen J, Shock CC, Back SA, Sherman LS. Distinct chemical structures inhibit the CEMIP hyaluronidase and promote oligodendrocyte progenitor cell maturation. J Biol Chem 2024; 300:107916. [PMID: 39454959 PMCID: PMC11742310 DOI: 10.1016/j.jbc.2024.107916] [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: 02/28/2024] [Revised: 09/05/2024] [Accepted: 10/01/2024] [Indexed: 10/28/2024] Open
Abstract
Growing evidence supports pathogenic roles for chronically elevated hyaluronidase activity in numerous conditions. Elevated expression of one such hyaluronidase, the Cell Migration Inducing and hyaluronan binding Protein (CEMIP), has been implicated in the pathogenesis and progression of several cancers as well as demyelinating diseases in the central nervous system (CNS). Developing effective and selective CEMIP inhibitors could therefore have efficacy in treating a variety of conditions where CEMIP is chronically elevated. Using two distinct screens for novel hyaluronidase inhibitors, we identified two synthetic thiocarbamates and one plant-derived flavonoid, sulfuretin, that effectively blocked CEMIP activity in live cells, including a tumorigenic cell line and primary cultures of oligodendrocyte progenitor cells (OPCs). None of these agents influenced cell proliferation, but they had differential dose-dependent and cell type-specific effects on cell survival. Furthermore, we found that each of these agents could promote oligodendrocyte maturation by OPCs in the presence of high molecular weight (>2 Mda) hyaluronan, the accumulation of which is linked to the inhibition of OPC maturation and remyelination failure in demyelinating diseases. These findings indicate that CEMIP can be inhibited through distinct chemical interactions and that CEMIP inhibitors have potential efficacy for treating demyelinating diseases or other conditions where CEMIP is elevated.
Collapse
Affiliation(s)
- Alec Peters
- Division of Neuroscience, Oregon National Primate Research Center, Beaverton, Oregon, USA; Department of Cell, Developmental and Cancer Biology, Oregon Health & Science University, Portland, Oregon, USA
| | - Fatima Banine
- Division of Neuroscience, Oregon National Primate Research Center, Beaverton, Oregon, USA
| | - Kanon Yasuhara
- Division of Neuroscience, Oregon National Primate Research Center, Beaverton, Oregon, USA
| | - Angela Hoffman
- Department of Chemistry, University of Portland, Portland, Oregon, USA
| | - Basappa
- Department of Studies in Organic Chemistry, University of Mysore, Manasagangotri, India
| | - Prashant K Metri
- Department of Studies in Organic Chemistry, University of Mysore, Manasagangotri, India
| | - Lily Gunning
- Department of Chemistry, University of Portland, Portland, Oregon, USA
| | - Ava Huffman
- Department of Chemistry, University of Portland, Portland, Oregon, USA
| | - Jake VanCampen
- Department of Chemistry, University of Portland, Portland, Oregon, USA
| | - Clinton C Shock
- College of Agricultural Sciences, Oregon State University, Corvallis, Oregon, USA
| | - Stephen A Back
- Departments of Pediatrics and Neurology, Oregon Health & Science University, Portland, Oregon, USA
| | - Larry S Sherman
- Division of Neuroscience, Oregon National Primate Research Center, Beaverton, Oregon, USA; Department of Cell, Developmental and Cancer Biology, Oregon Health & Science University, Portland, Oregon, USA.
| |
Collapse
|
4
|
Wang Y, Ding G, Chu C, Cheng XD, Qin JJ. Genomic biology and therapeutic strategies of liver metastasis from gastric cancer. Crit Rev Oncol Hematol 2024; 202:104470. [PMID: 39111457 DOI: 10.1016/j.critrevonc.2024.104470] [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: 12/15/2023] [Revised: 07/30/2024] [Accepted: 08/02/2024] [Indexed: 08/12/2024] Open
Abstract
The liver is a frequent site of metastasis in advanced gastric cancer (GC). Despite significant advancements in diagnostic and therapeutic techniques, the overall survival rate for patients afflicted with gastric cancer liver metastasis (GCLM) remains dismally low. Precision oncology has made significant progress in identifying therapeutic targets and enhancing our understanding of metastasis mechanisms through genome sequencing and molecular characterization. Therefore, it is crucial to have a comprehensive understanding of the various molecular processes involved in GCLM and the fundamental principles of systemic therapy to develop new treatment approaches. This paper aims to review recent findings on the diagnosis, potential biomarkers, and therapies targeting the multiple molecular processes of GCLM, with the goal of improving treatment strategies for patients with GCLM.
Collapse
Affiliation(s)
- Yichao Wang
- College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou 313200, China; Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou 310022, China
| | - Guangyu Ding
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou 310022, China
| | - Chu Chu
- College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou 313200, China
| | - Xiang-Dong Cheng
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou 310022, China; Key Laboratory of Prevention, Diagnosis and Therapy of Upper Gastrointestinal Cancer of Zhejiang Province, Hangzhou 310022, China.
| | - Jiang-Jiang Qin
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou 310022, China; Key Laboratory of Prevention, Diagnosis and Therapy of Upper Gastrointestinal Cancer of Zhejiang Province, Hangzhou 310022, China; Key Laboratory for Molecular Medicine and Chinese Medicine Preparations, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou 310022, China.
| |
Collapse
|
5
|
Liu Y, Jin T, Chen R, Miao R, Zhou Y, Shao S. High expression of ABL2 promotes gastric cancer cells migration, invasion and proliferation via the TGF-β and YAP signaling pathways. J Cancer 2024; 15:5719-5728. [PMID: 39308677 PMCID: PMC11414612 DOI: 10.7150/jca.99307] [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: 06/05/2024] [Accepted: 08/27/2024] [Indexed: 09/25/2024] Open
Abstract
Background: The Abelson-Related Gene (ABL2) is expressed in various malignancies. However, its role in gastric cancer (GC) regarding tumor proliferation, metastasis, and invasion remains unclear. Methods: ABL2 expression in clinical specimens was assessed using quantitative real-time fluorescence PCR (qRT-PCR). Western blotting and immunofluorescence assay determined protein levels. Additionally, Transwell migration and invasion, cell counting kit-8 (CCK-8) and colony-formation assays analyzed the effect of ABL2 on GC cells. Protein levels related to GC cells were assessed through Western blotting. The effects of si-ABL2 combined with GA-017 that activated YAP on cell migration, invasion and proliferation were investigated. Results: ABL2 expression was upregulated in human GC tissues compared to paracancer tissues, and it was positively related to tumor node metastasis classification (TNM) stage. Furthermore, high ABL2 levels promoted the proliferation, metastasis, and invasion capacity in GC cells. Elevated ABL2 expression enhanced the expression of MMP2, MMP9, and PCNA while decreasing TIMP1 and TIMP2 expression. It also increased the p-SMAD2/3 expression and YAP expression, decreased the expression of p-YAP in GC cells. Furthermore, GA-017 increased ABL2 expression in MGC-803 cells and counteracted the effects of si-ABL2 on cell migration, invasion and proliferation. Conclusion: These findings indicated that heightened ABL2 expression could activate TGF-β/SMAD2/3 and YAP signaling pathway, promoting epithelial mesenchymal transformation (EMT), and enhancing multiplication, metastasis, and invasion in GC cells.
Collapse
Affiliation(s)
- Yun Liu
- Department of Gastroenterology, Institute of Digestive Disease, The Affiliated People's Hospital, Jiangsu University, Zhenjiang, Jiangsu, China
- School of Medicine, Jiangsu University, Zhenjiang, Jiangsu, China
| | - Tao Jin
- Department of Gastroenterology, Yixing people's hospital, Yixing, Jiangsu, China
| | - Ruiyun Chen
- Department of gastrointestinal surgery, Qingdao Central Hospital, University of Health and Rehabilitation Sciences, Qingdao, Shandong, China
| | - Renjie Miao
- Department of Clinical laboratory, Affiliated Third Hospital of Zhenjiang to Jiangsu University, Zhenjiang, Jiangsu, China
| | - Yong Zhou
- Department of Gastroenterology, Institute of Digestive Disease, The Affiliated People's Hospital, Jiangsu University, Zhenjiang, Jiangsu, China
| | - Shihe Shao
- School of Medicine, Jiangsu University, Zhenjiang, Jiangsu, China
- Department of Gastroenterology, Yixing people's hospital, Yixing, Jiangsu, China
| |
Collapse
|
6
|
Michaelsen GL, da Silva LDRE, de Lima DS, Jaeger MDC, Brunetto AT, Dalmolin RJS, Sinigaglia M. A Prognostic Methylation-Driven Two-Gene Signature in Medulloblastoma. J Mol Neurosci 2024; 74:47. [PMID: 38662144 DOI: 10.1007/s12031-024-02203-9] [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: 09/29/2023] [Accepted: 02/21/2024] [Indexed: 04/26/2024]
Abstract
Medulloblastoma (MB) is one of the most common pediatric brain tumors and it is estimated that one-third of patients will not achieve long-term survival. Conventional prognostic parameters have limited and unreliable correlations with MB outcome, presenting a major challenge for patients' clinical improvement. Acknowledging this issue, our aim was to build a gene signature and evaluate its potential as a new prognostic model for patients with the disease. In this study, we used six datasets totaling 1679 samples including RNA gene expression and DNA methylation data from primary MB as well as control samples from healthy cerebellum. We identified methylation-driven genes (MDGs) in MB, genes whose expression is correlated with their methylation. We employed LASSO regression, incorporating the MDGs as a parameter to develop the prognostic model. Through this approach, we derived a two-gene signature (GS-2) of candidate prognostic biomarkers for MB (CEMIP and NCBP3). Using a risk score model, we confirmed the GS-2 impact on overall survival (OS) with Kaplan-Meier analysis. We evaluated its robustness and accuracy with receiver operating characteristic curves predicting OS at 1, 3, and 5 years in multiple independent datasets. The GS-2 showed highly significant results as an independent prognostic biomarker compared to traditional MB markers. The methylation-regulated GS-2 risk score model can effectively classify patients with MB into high and low-risk, reinforcing the importance of this epigenetic modification in the disease. Such genes stand out as promising prognostic biomarkers with potential application for MB treatment.
Collapse
Affiliation(s)
- Gustavo Lovatto Michaelsen
- Children's Cancer Institute, Porto Alegre, 90620-110, RS, Brazil
- Bioinformatics Multidisciplinary Environment-BioME, Digital Metropole Institute, Federal University of Rio Grande do Norte, Natal, 59076-550, RN, Brazil
- National Science and Technology Institute for Children's Cancer Biology and Pediatric Oncology - INCT BioOncoPed, Porto Alegre, 90035-003, RS, Brazil
| | - Lívia Dos Reis Edinger da Silva
- Children's Cancer Institute, Porto Alegre, 90620-110, RS, Brazil
- Federal University of Health Sciences of Porto Alegre, Porto Alegre, 90050-170, RS, Brazil
| | - Douglas Silva de Lima
- Children's Cancer Institute, Porto Alegre, 90620-110, RS, Brazil
- Institute of Basic Health Sciences, Federal University of Rio Grande do Sul, Porto Alegre, 90035-003, RS, Brazil
| | - Mariane da Cunha Jaeger
- Children's Cancer Institute, Porto Alegre, 90620-110, RS, Brazil
- National Science and Technology Institute for Children's Cancer Biology and Pediatric Oncology - INCT BioOncoPed, Porto Alegre, 90035-003, RS, Brazil
| | - André Tesainer Brunetto
- Children's Cancer Institute, Porto Alegre, 90620-110, RS, Brazil
- National Science and Technology Institute for Children's Cancer Biology and Pediatric Oncology - INCT BioOncoPed, Porto Alegre, 90035-003, RS, Brazil
| | - Rodrigo Juliani Siqueira Dalmolin
- Bioinformatics Multidisciplinary Environment-BioME, Digital Metropole Institute, Federal University of Rio Grande do Norte, Natal, 59076-550, RN, Brazil
- Department of Biochemistry, Federal University of Rio Grande do Norte, Natal, 59064-741, RN, Brazil
| | - Marialva Sinigaglia
- Children's Cancer Institute, Porto Alegre, 90620-110, RS, Brazil.
- Bioinformatics Multidisciplinary Environment-BioME, Digital Metropole Institute, Federal University of Rio Grande do Norte, Natal, 59076-550, RN, Brazil.
- National Science and Technology Institute for Children's Cancer Biology and Pediatric Oncology - INCT BioOncoPed, Porto Alegre, 90035-003, RS, Brazil.
| |
Collapse
|
7
|
Guo S, Guo Y, Chen Y, Cui S, Zhang C, Chen D. The role of CEMIP in cancers and its transcriptional and post-transcriptional regulation. PeerJ 2024; 12:e16930. [PMID: 38390387 PMCID: PMC10883155 DOI: 10.7717/peerj.16930] [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: 11/15/2023] [Accepted: 01/22/2024] [Indexed: 02/24/2024] Open
Abstract
CEMIP is a protein known for inducing cell migration and binding to hyaluronic acid. Functioning as a hyaluronidase, CEMIP primarily facilitates the breakdown of the extracellular matrix component, hyaluronic acid, thereby regulating various signaling pathways. Recent evidence has highlighted the significant role of CEMIP in different cancers, associating it with diverse pathological states. While identified as a biomarker for several diseases, CEMIP's mechanism in cancer seems distinct. Accumulating data suggests that CEMIP expression is triggered by chemical modifications to itself and other influencing factors. Transcriptionally, chemical alterations to the CEMIP promoter and involvement of transcription factors such as AP-1, HIF, and NF-κB regulate CEMIP levels. Similarly, specific miRNAs have been found to post-transcriptionally regulate CEMIP. This review provides a comprehensive summary of CEMIP's role in various cancers and explores how both transcriptional and post-transcriptional mechanisms control its expression.
Collapse
Affiliation(s)
- Song Guo
- Shandong University of Technology, School of Life Sciences and Medicine, Zibo, Shandong, China
| | - Yunfei Guo
- Shandong University of Technology, School of Life Sciences and Medicine, Zibo, Shandong, China
| | - Yuanyuan Chen
- Shandong University of Technology, School of Life Sciences and Medicine, Zibo, Shandong, China
| | - Shuaishuai Cui
- Shandong University of Technology, School of Life Sciences and Medicine, Zibo, Shandong, China
| | - Chunmei Zhang
- Shandong University of Technology, School of Life Sciences and Medicine, Zibo, Shandong, China
| | - Dahu Chen
- Shandong University of Technology, School of Life Sciences and Medicine, Zibo, Shandong, China
| |
Collapse
|
8
|
Liu Y, Hu G, Li Y, Kong X, Yang K, Li Z, Lao W, Li J, Zhong J, Zhang S, Leng Y, Bi C, Zhai A. Research on the biological mechanism and potential application of CEMIP. Front Immunol 2023; 14:1222425. [PMID: 37662915 PMCID: PMC10471826 DOI: 10.3389/fimmu.2023.1222425] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2023] [Accepted: 07/26/2023] [Indexed: 09/05/2023] Open
Abstract
Cell migration-inducing protein (CEMIP), also known as KIAA1199 and hyaluronan-binding protein involved in hyaluronan depolymerization, is a new member of the hyaluronidase family that degrades hyaluronic acid (HA) and remodels the extracellular matrix. In recent years, some studies have reported that CEMIP can promote the proliferation, invasion, and adhesion of various tumor cells and can play an important role in bacterial infection and arthritis. This review focuses on the pathological mechanism of CEMIP in a variety of diseases and expounds the function of CEMIP from the aspects of inhibiting cell apoptosis, promoting HA degradation, inducing inflammatory responses and related phosphorylation, adjusting cellular microenvironment, and regulating tissue fibrosis. The diagnosis and treatment strategies targeting CEMIP are also summarized. The various functions of CEMIP show its great potential application value.
Collapse
Affiliation(s)
- Yang Liu
- Department of Laboratory Medicine, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| | - Gang Hu
- The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| | - Yuetong Li
- Department of Endocrinology, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| | - Xinyi Kong
- Department of Laboratory Medicine, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| | - Kaming Yang
- Department of Endocrinology, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| | - Zhenlin Li
- Department of Endocrinology, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| | - Wanwen Lao
- Department of Endocrinology, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| | - Jiaxin Li
- Department of Endocrinology, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| | - Jianhua Zhong
- Department of Endocrinology, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| | - Shitong Zhang
- Department of General Practice, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| | - Yuxin Leng
- Department of Critical Care Medicine, Peking University Third Hospital, Beijing, China
| | - Changlong Bi
- Department of Endocrinology, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| | - Aixia Zhai
- Department of Laboratory Medicine, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| |
Collapse
|
9
|
Spataro S, Guerra C, Cavalli A, Sgrignani J, Sleeman J, Poulain L, Boland A, Scapozza L, Moll S, Prunotto M. CEMIP (HYBID, KIAA1199): structure, function and expression in health and disease. FEBS J 2023; 290:3946-3962. [PMID: 35997767 DOI: 10.1111/febs.16600] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Revised: 07/28/2022] [Accepted: 08/17/2022] [Indexed: 12/01/2022]
Abstract
CEMIP (cell migration-inducing protein), also known as KIAA1199 or HYBID, is a protein involved in the depolymerisation of hyaluronic acid (HA), a major glycosaminoglycan component of the extracellular matrix. CEMIP was originally described in patients affected by nonsyndromic hearing loss and has subsequently been shown to play a key role in tumour initiation and progression, as well as arthritis, atherosclerosis and idiopathic pulmonary fibrosis. Despite the vast literature associating CEMIP with these diseases, its biology remains elusive. The present review article summarises all the major scientific evidence regarding its structure, function, role and expression, and attempts to cast light on a protein that modulates EMT, fibrosis and tissue inflammation, an unmet key aspect in several inflammatory disease conditions.
Collapse
Affiliation(s)
- Sofia Spataro
- Institute of Pharmaceutical Sciences of Western Switzerland, University of Geneva, Switzerland
| | - Concetta Guerra
- Institute for Research in Biomedicine, Università della Svizzera Italiana, Bellinzona, Switzerland
| | - Andrea Cavalli
- Institute for Research in Biomedicine, Università della Svizzera Italiana, Bellinzona, Switzerland
- Swiss Institute of Bioinformatics, Lausanne, Switzerland
| | - Jacopo Sgrignani
- Institute for Research in Biomedicine, Università della Svizzera Italiana, Bellinzona, Switzerland
| | - Jonathan Sleeman
- European Center for Angioscience (ECAS), Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
- Institute for Biological and Chemical Systems - Biological Information Processing (IBCS - BIP), Karlsruhe Institute for Technology (KIT), Germany
| | - Lina Poulain
- Department of Molecular Biology, University of Geneva, Switzerland
| | - Andreas Boland
- Department of Molecular Biology, University of Geneva, Switzerland
| | - Leonardo Scapozza
- Institute of Pharmaceutical Sciences of Western Switzerland, University of Geneva, Switzerland
| | - Solange Moll
- Department of Pathology, University Hospital of Geneva, Switzerland
| | - Marco Prunotto
- Institute of Pharmaceutical Sciences of Western Switzerland, University of Geneva, Switzerland
| |
Collapse
|
10
|
Mi C, Zhao Y, Ren L, Zhang D. HIF1α/CCL7/KIAA1199 axis mediates hypoxia-induced gastric cancer aggravation and glycolysis alteration. J Clin Biochem Nutr 2023; 72:225-233. [PMID: 37251956 PMCID: PMC10209595 DOI: 10.3164/jcbn.22-48] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Accepted: 09/03/2022] [Indexed: 09/04/2024] Open
Abstract
Gastric cancer is a common digestion tumor with high malignant severity and prevalence. Emerging studies reported C-C motif chemokine ligand 7 (CCL7) as a regulator of various tumor diseases. Our research explored the function and underlying mechanism of CCL7 during gastric cancer development. RT-qPCR, Western blot and other datasets were employed to evaluate CCL7 expression in tissues and cells. Kaplan-Meier and Cox regression analyses were recruited to evaluate the correlations between CCL7 expression and patients' survival or clinical features. A loss-of-function assay was performed to evaluate the function of CCL7 in gastric cancer. 1% O2 was utilized to mimic hypoxic condition. KIAA1199 and HIF1α were included in the regulatory mechanism. The results showed that CCL7 was up-regulated and its high expression was correlated with poor survival of gastric cancer patients. Depressing CCL7 attenuated proliferation, migration, invasion, and induced apoptosis of gastric cancer cells. Meanwhile, CCL7 inhibition weakened hypoxia-induced gastric cancer aggravation. Besides, KIAA1199 and HIF1α were involved in the mechanism of CCL7-mediated gastric cancer aggravation under hypoxia. Our research identified CCL7 as a novel tumor-activator in gastric cancer pathogenesis and hypoxia-induced tumor aggravation was regulated by HIF1α/CCL7/KIAA1199 axis. The evidence may provide a novel target for gastric cancer treatment.
Collapse
Affiliation(s)
- Chen Mi
- Department of Gastroenterology, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi Province 710061, China
| | - Yan Zhao
- Department of Gastroenterology, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi Province 710061, China
| | - Li Ren
- Department of Gastroenterology, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi Province 710061, China
| | - Dan Zhang
- Department of Gastroenterology, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi Province 710061, China
| |
Collapse
|
11
|
Chen L, Shi K, Ditzel N, Qiu W, Figeac F, Nielsen LHD, Tencerova M, Kowal JM, Ding M, Andreasen CM, Andersen TL, Kassem M. KIAA1199 deficiency enhances skeletal stem cell differentiation to osteoblasts and promotes bone regeneration. Nat Commun 2023; 14:2016. [PMID: 37037828 PMCID: PMC10086002 DOI: 10.1038/s41467-023-37651-1] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Accepted: 03/14/2023] [Indexed: 04/12/2023] Open
Abstract
Upon transplantation, skeletal stem cells (also known as bone marrow stromal or mesenchymal stem cells) can regulate bone regeneration by producing secreted factors. Here, we identify KIAA1199 as a bone marrow stromal cell-secreted factor in vitro and in vivo. KIAA1199 plasma levels of patients positively correlate with osteoporotic fracture risk and expression levels of KIAA1199 in patient bone marrow stromal cells negatively correlates with their osteogenic differentiation potential. KIAA1199-deficient bone marrow stromal cells exhibit enhanced osteoblast differentiation in vitro and ectopic bone formation in vivo. Consistently, KIAA1199 knockout mice display increased bone mass and biomechanical strength, as well as an increased bone formation rate. They also exhibit accelerated healing of surgically generated bone defects and are protected from ovariectomy-induced bone loss. Mechanistically, KIAA1199 regulates osteogenesis by inhibiting the production of osteopontin by osteoblasts, via integrin-mediated AKT and ERK-MAPK intracellular signaling. Thus, KIAA1199 is a regulator of osteoblast differentiation and bone regeneration and could be targeted for the treatment or management of low bone mass conditions.
Collapse
Affiliation(s)
- Li Chen
- Department of Endocrinology and Metabolism, Endocrine Research Laboratory (KMEB), Odense University Hospital & University of Southern Denmark, Odense, Denmark.
- Dept. of Pathology and Physiopathology, Guangxi Key Laboratory of Tumor Immunology and Microenvironmental Regulation, Guilin Medical University, Guilin, Guangxi, China.
| | - Kaikai Shi
- Department of Endocrinology and Metabolism, Endocrine Research Laboratory (KMEB), Odense University Hospital & University of Southern Denmark, Odense, Denmark
| | - Nicholas Ditzel
- Department of Endocrinology and Metabolism, Endocrine Research Laboratory (KMEB), Odense University Hospital & University of Southern Denmark, Odense, Denmark
| | - Weimin Qiu
- Department of Endocrinology and Metabolism, Endocrine Research Laboratory (KMEB), Odense University Hospital & University of Southern Denmark, Odense, Denmark
| | - Florence Figeac
- Department of Endocrinology and Metabolism, Endocrine Research Laboratory (KMEB), Odense University Hospital & University of Southern Denmark, Odense, Denmark
| | - Louise Himmelstrup Dreyer Nielsen
- Department of Endocrinology and Metabolism, Endocrine Research Laboratory (KMEB), Odense University Hospital & University of Southern Denmark, Odense, Denmark
| | - Michaela Tencerova
- Department of Endocrinology and Metabolism, Endocrine Research Laboratory (KMEB), Odense University Hospital & University of Southern Denmark, Odense, Denmark
| | - Justyna Magdalena Kowal
- Department of Endocrinology and Metabolism, Endocrine Research Laboratory (KMEB), Odense University Hospital & University of Southern Denmark, Odense, Denmark
| | - Ming Ding
- Department of Orthopaedic Surgery and Traumatology, Odense University Hospital & University of Southern Denmark, Odense, Denmark
| | | | | | - Moustapha Kassem
- Department of Endocrinology and Metabolism, Endocrine Research Laboratory (KMEB), Odense University Hospital & University of Southern Denmark, Odense, Denmark.
- Department of Cellular and Molecular Medicine (ICMM), Faculty of Health Sciences, University of Copenhagen, Copenhagen, Denmark.
| |
Collapse
|
12
|
Ribeiro E, Costa B, Vasques-Nóvoa F, Vale N. In Vitro Drug Repurposing: Focus on Vasodilators. Cells 2023; 12:671. [PMID: 36831338 PMCID: PMC9954697 DOI: 10.3390/cells12040671] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2023] [Revised: 02/10/2023] [Accepted: 02/16/2023] [Indexed: 02/22/2023] Open
Abstract
Drug repurposing aims to identify new therapeutic uses for drugs that have already been approved for other conditions. This approach can save time and resources compared to traditional drug development, as the safety and efficacy of the repurposed drug have already been established. In the context of cancer, drug repurposing can lead to the discovery of new treatments that can target specific cancer cell lines and improve patient outcomes. Vasodilators are a class of drugs that have been shown to have the potential to influence various types of cancer. These medications work by relaxing the smooth muscle of blood vessels, increasing blood flow to tumors, and improving the delivery of chemotherapy drugs. Additionally, vasodilators have been found to have antiproliferative and proapoptotic effects on cancer cells, making them a promising target for drug repurposing. Research on vasodilators for cancer treatment has already shown promising results in preclinical and clinical studies. However, additionally research is needed to fully understand the mechanisms of action of vasodilators in cancer and determine the optimal dosing and combination therapy for patients. In this review, we aim to explore the molecular mechanisms of action of vasodilators in cancer cell lines and the current state of research on their repurposing as a treatment option. With the goal of minimizing the effort and resources required for traditional drug development, we hope to shed light on the potential of vasodilators as a viable therapeutic strategy for cancer patients.
Collapse
Affiliation(s)
- Eduarda Ribeiro
- OncoPharma Research Group, Center for Health Technology and Services Research (CINTESIS), Rua Doutor Plácido da Costa, 4200-450 Porto, Portugal
- CINTESIS@RISE, Faculty of Medicine, University of Porto, Alameda Professor Hernâni Monteiro, 4200-319 Porto, Portugal
- Institute of Biomedical Sciences Abel Salazar (ICBAS), University of Porto, Rua de Jorge Viterbo Ferreira 228, 4050-313 Porto, Portugal
| | - Bárbara Costa
- OncoPharma Research Group, Center for Health Technology and Services Research (CINTESIS), Rua Doutor Plácido da Costa, 4200-450 Porto, Portugal
- CINTESIS@RISE, Faculty of Medicine, University of Porto, Alameda Professor Hernâni Monteiro, 4200-319 Porto, Portugal
| | - Francisco Vasques-Nóvoa
- Cardiovascular R&D Center, Faculty of Medicine, University of Porto, Rua Doutor Plácido da Costa, 4200-450 Porto, Portugal
- Department of Surgery and Physiology, Faculty of Medicine, University of Porto, Rua Doutor Plácido da Costa, 4200-450 Porto, Portugal
| | - Nuno Vale
- OncoPharma Research Group, Center for Health Technology and Services Research (CINTESIS), Rua Doutor Plácido da Costa, 4200-450 Porto, Portugal
- CINTESIS@RISE, Faculty of Medicine, University of Porto, Alameda Professor Hernâni Monteiro, 4200-319 Porto, Portugal
- Department of Community Medicine, Information and Health Decision Sciences (MEDCIDS), Faculty of Medicine, University of Porto, Rua Doutor Plácido da Costa, 4200-450 Porto, Portugal
| |
Collapse
|
13
|
Liu J, Xia S, Han P, Zhang M, Wu J, Liao J. Downregulation of KIAA1199 alleviated the activation, proliferation, and migration of hepatic stellate cells by the inhibition of epithelial-mesenchymal transition. Open Med (Wars) 2023; 18:20230689. [PMID: 37034499 PMCID: PMC10080707 DOI: 10.1515/med-2023-0689] [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: 09/05/2022] [Revised: 03/04/2023] [Accepted: 03/17/2023] [Indexed: 04/11/2023] Open
Abstract
KIAA1199, a major glycosaminoglycan component of the extracellular matrix, was reported to induce a fibrosis-like process. However, the relationship between KIAA1199 and liver fibrosis remains unclear. The liver fibrosis mouse model was established with carbon tetrachloride (CCl4). Here, we found that KIAA1199 was upregulated in CCl4-induced liver fibrosis. The expression of KIAA1199 was also increased in TGF-β-stimulated LX-2 cells. To clarify the impact of KIAA1199 in hepatic stellate cells (HSCs), we downregulated the expression of KIAA1199 in LX-2 cells by RNA interference. Cell proliferation, apoptosis, and migration were determined by CCK-8, flow cytometry, and transwell assay. We found that KIAA1199 knockdown reduced the expression of fibrosis markers α-SMA and COL1A1. Depletion of KIAA1199 inhibited cell proliferation by downregulating cyclin B1 and cyclin D1 and promoted cell apoptosis by upregulating Bax and downregulating Bcl-2. Moreover, KIAA1199 knockdown decreased matrix metalloproteinase-2 (MMP-2) and MMP-9 expression to inhibit the migration ability of LX-2 cells. Silencing KIAA1199 also suppressed the epithelial-mesenchymal transition phenomenon. Collectively, our study revealed that KIAA1199 knockdown alleviated the activation, proliferation, and migration of HSCs, while promoting apoptosis of HSCs, which suggests that KIAA1199 may be a potential regulator of liver fibrosis.
Collapse
Affiliation(s)
- Jingmei Liu
- Department of Gastroenterology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Suhong Xia
- Department of Gastroenterology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Ping Han
- Department of Gastroenterology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Mingyu Zhang
- Department of Gastroenterology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Jingwen Wu
- Department of Gastroenterology, Hainan Hospital of PLA General Hospital, Sanya, Hainan, China
| | - Jiazhi Liao
- Department of Gastroenterology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| |
Collapse
|
14
|
Chen H, Wang Q, Liu J, Chen Y, Zhang Q, Chai L, Wang Y, Li D, Qiu Y, Li M. CEMIP as a prognostic biomarker for cancers: a meta- and bioinformatic analysis. Expert Rev Mol Diagn 2022; 22:1107-1115. [PMID: 36631437 DOI: 10.1080/14737159.2022.2168191] [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: 10/19/2022] [Accepted: 01/10/2023] [Indexed: 01/13/2023]
Abstract
OBJECTIVE Cell migration-inducing and hyaluronan-binding protein (CEMIP) is overexpressed in several cancers and is related to prognosis in cancer patients. Here, we conducted a meta-analysis to explore the prognostic effects of CEMIP in cancer patients. METHODS Relevant published studies were systematically searched in four databases. The role of CEMIP was evaluated using pooled hazard ratios (HRs), odd ratios (ORs), and 95% confidence intervals (95% CIs). The Cancer Genome Atlas (TCGA) was used to investigate the prognostic value of CEMIP in various cancers. RESULTS 11 literatures with 1355 patients were included in this meta-analysis. The results showed that overexpression of CEMIP was significantly associated with poor OS (HR = 3.03; 95% CI: 2.00-4.59; p < 0.001), DFS (HR = 3.38; 95% CI: 2.41-4.74; p < 0.001). Elevated CEMIP expression is associated with advanced clinical stage, lymph node metastasis, and poor histological grade. In addition, TCGA datasets were used to verify that CEMIP was found highly expressed in multiple cancers and was associated with poorer survival. CONCLUSION The results demonstrated that CEMIP could be a novel prognostic biomarker for cancer patients. However, because the included studies mainly focused on Asian populations, further research is needed to verify its applicability.
Collapse
Affiliation(s)
- Huan Chen
- Department of Respiratory & Critical Care Medicine, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Qingting Wang
- Department of Respiratory & Critical Care Medicine, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Jin Liu
- Department of Respiratory & Critical Care Medicine, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Yuqian Chen
- Department of Respiratory & Critical Care Medicine, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Qianqian Zhang
- Department of Respiratory & Critical Care Medicine, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Limin Chai
- Department of Respiratory & Critical Care Medicine, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Yan Wang
- Department of Respiratory & Critical Care Medicine, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Danyang Li
- Department of Respiratory & Critical Care Medicine, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Yuanjie Qiu
- Department of Respiratory & Critical Care Medicine, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Manxiang Li
- Department of Respiratory & Critical Care Medicine, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| |
Collapse
|
15
|
Miao X, Wang Y, Miao Z, Pan H. A comprehensive review of the progress of cell migration inducing hyaluronidase 1. Medicine (Baltimore) 2022; 101:e31610. [PMID: 36451490 PMCID: PMC9704909 DOI: 10.1097/md.0000000000031610] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/05/2022] Open
Abstract
The gene cell migration inducing hyaluronidase 1 (CEMIP) is on chromosome 15q25 and codes for a 150-kDa protein with an N-terminal secretion signal, a G8 domain, 2 GG domains, and several repeats. It was first described as a specific protein in the inner ear relating to nonsyndromic hearing loss. Recently, increasing research detected its association in various cancers, determining the progression, metastasis, and prognosis by influencing the proliferation and invasion of the cells. This relation is accomplished through various interacting pathways, such as the Wnt/β-catenin signaling pathway and the epidermal growth factor receptor signaling pathway. Thus, CEMIP could be a novel and potential focus for tumor diagnosis and treatment, but further studies on the regulatory role of CEMIP in vivo and in vitro are still needed. Herein, we summarize the process in recent studies of CEMIP, especially in cancer research.
Collapse
Affiliation(s)
- Xiangguang Miao
- Neurological Institute of Jiangxi Province and Department of Neurology, Jiangxi Provincial People’s Hospital, The First Affiliated Hospital of Nanchang Medical College, Nanchang, Jiangxi, China
| | - Yukai Wang
- Neurological Institute of Jiangxi Province and Department of Neurology, Jiangxi Provincial People’s Hospital, The First Affiliated Hospital of Nanchang Medical College, Nanchang, Jiangxi, China
| | - Zhiguo Miao
- Zhengzhou Traditional Chinese Medicine Hospital, Zhengzhou, China
| | - Haili Pan
- Neurological Institute of Jiangxi Province and Department of Neurology, Jiangxi Provincial People’s Hospital, The First Affiliated Hospital of Nanchang Medical College, Nanchang, Jiangxi, China
- School of Chemistry and Chemical Engineering, Nanchang University, Nanchang, China
- Queen Mary School, Nanchang University, Nanchang, China
- * Correspondence: Haili Pan, Nanchang University and Jiangxi Provincial People’s Hospital, Nanchang 330031, China. (e-mail: )
| |
Collapse
|
16
|
Domanegg K, Sleeman JP, Schmaus A. CEMIP, a Promising Biomarker That Promotes the Progression and Metastasis of Colorectal and Other Types of Cancer. Cancers (Basel) 2022; 14:cancers14205093. [PMID: 36291875 PMCID: PMC9600181 DOI: 10.3390/cancers14205093] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2022] [Revised: 10/05/2022] [Accepted: 10/12/2022] [Indexed: 12/04/2022] Open
Abstract
Simple Summary CEMIP (cell migration-inducing and hyaluronan-binding protein) has been implicated in the pathogenesis of numerous diseases, including colorectal and other forms of cancer. The molecular functions of CEMIP are currently under investigation and include the degradation of the extracellular matrix component hyaluronic acid (HA), as well as the regulation of a number of signaling pathways. In this review, we survey our current understanding of how CEMIP contributes to tumor growth and metastasis, focusing particularly on colorectal cancer, for which it serves as a promising biomarker. Abstract Originally discovered as a hypothetical protein with unknown function, CEMIP (cell migration-inducing and hyaluronan-binding protein) has been implicated in the pathogenesis of numerous diseases, including deafness, arthritis, atherosclerosis, idiopathic pulmonary fibrosis, and cancer. Although a comprehensive definition of its molecular functions is still in progress, major functions ascribed to CEMIP include the depolymerization of the extracellular matrix component hyaluronic acid (HA) and the regulation of a number of signaling pathways. CEMIP is a promising biomarker for colorectal cancer. Its expression is associated with poor prognosis for patients suffering from colorectal and other types of cancer and functionally contributes to tumor progression and metastasis. Here, we review our current understanding of how CEMIP is able to foster the process of tumor growth and metastasis, focusing particularly on colorectal cancer. Studies in cancer cells suggest that CEMIP exerts its pro-tumorigenic and pro-metastatic activities through stimulating migration and invasion, suppressing cell death and promoting survival, degrading HA, regulating pro-metastatic signaling pathways, inducing the epithelial–mesenchymal transition (EMT) program, and contributing to the metabolic reprogramming and pre-metastatic conditioning of future metastatic microenvironments. There is also increasing evidence indicating that CEMIP may be expressed in cells within the tumor microenvironment that promote tumorigenesis and metastasis formation, although this remains in an early stage of investigation. CEMIP expression and activity can be therapeutically targeted at a number of levels, and preliminary findings in animal models show encouraging results in terms of reduced tumor growth and metastasis, as well as combating therapy resistance. Taken together, CEMIP represents an exciting new player in the progression of colorectal and other types of cancer that holds promise as a therapeutic target and biomarker.
Collapse
Affiliation(s)
- Kevin Domanegg
- European Center for Angioscience (ECAS), Medical Faculty Mannheim, University of Heidelberg, 68167 Mannheim, Germany
| | - Jonathan P. Sleeman
- European Center for Angioscience (ECAS), Medical Faculty Mannheim, University of Heidelberg, 68167 Mannheim, Germany
- Institute of Biological and Chemical Systems-Biological Information Processing, Karlsruhe Institute of Technology (KIT) Campus Nord, 76344 Eggenstein-Leopoldshafen, Germany
- Correspondence:
| | - Anja Schmaus
- European Center for Angioscience (ECAS), Medical Faculty Mannheim, University of Heidelberg, 68167 Mannheim, Germany
- Institute of Biological and Chemical Systems-Biological Information Processing, Karlsruhe Institute of Technology (KIT) Campus Nord, 76344 Eggenstein-Leopoldshafen, Germany
| |
Collapse
|
17
|
Zeng X, Xiao J, Bai X, Liu Y, Zhang M, Liu J, Lin Z, Zhang Z. Research progress on the circRNA/lncRNA-miRNA-mRNA axis in gastric cancer. Pathol Res Pract 2022; 238:154030. [PMID: 36116329 DOI: 10.1016/j.prp.2022.154030] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Revised: 07/14/2022] [Accepted: 07/16/2022] [Indexed: 01/19/2023]
Abstract
Gastric cancer is one of the most common malignant tumours worldwide. Genetic and epigenetic alterations are key factors in gastric carcinogenesis and drug resistance to chemotherapy. Competing endogenous RNA (ceRNA) regulation models have defined circRNA/lncRNA as miRNA sponges that indirectly regulate miRNA downstream target genes. The ceRNA regulatory network is related to the malignant biological behaviour of gastric cancer. The circRNA/lncRNA-miRNA-mRNA axis may be a marker for the early diagnosis and prognosis of gastric cancer and a potential therapeutic target for gastric cancer. Exosomal ncRNAs play an important role in gastric cancer and are expected to be ideal biomarkers for the diagnosis, prognosis, and treatment of gastric cancer. This review summarizes the specific ceRNA regulatory network (circRNA/lncRNA-miRNA-mRNA) discovered in gastric cancer in recent years, which may provide new ideas or strategies for early clinical diagnosis, further development, and application.
Collapse
Affiliation(s)
- Xuemei Zeng
- Cancer Research Institute of Hengyang Medical School, University of South China; Key Laboratory of Cancer Cellular and Molecular Pathology in Hunan Province, Hengyang, Hunan 421001, China
| | - Juan Xiao
- Department of Otorhinolaryngology, The Second Affiliated Hospital, Hengyang Medical School,University of South China, Hengyang 421001, China
| | - Xue Bai
- Cancer Research Institute of Hengyang Medical School, University of South China; Key Laboratory of Cancer Cellular and Molecular Pathology in Hunan Province, Hengyang, Hunan 421001, China
| | - Yiwen Liu
- Cancer Research Institute of Hengyang Medical School, University of South China; Key Laboratory of Cancer Cellular and Molecular Pathology in Hunan Province, Hengyang, Hunan 421001, China
| | - Meilan Zhang
- Cancer Research Institute of Hengyang Medical School, University of South China; Key Laboratory of Cancer Cellular and Molecular Pathology in Hunan Province, Hengyang, Hunan 421001, China
| | - Jiangrong Liu
- Cancer Research Institute of Hengyang Medical School, University of South China; Key Laboratory of Cancer Cellular and Molecular Pathology in Hunan Province, Hengyang, Hunan 421001, China
| | - Zixuan Lin
- Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
| | - Zhiwei Zhang
- Cancer Research Institute of Hengyang Medical School, University of South China; Key Laboratory of Cancer Cellular and Molecular Pathology in Hunan Province, Hengyang, Hunan 421001, China.
| |
Collapse
|
18
|
Zhou M, Hua W, Sun Y. Cell migration inducing hyaluronidase 1 promotes growth and metastasis of papillary thyroid carcinoma. Bioengineered 2022; 13:11822-11831. [PMID: 35543351 PMCID: PMC9276010 DOI: 10.1080/21655979.2022.2074110] [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] [Indexed: 11/23/2022] Open
Abstract
Cell migration inducing hyaluronidase 1 (CEMIP) mediates catabolism of hyaluronan, and participates in the cell metastasis, invasion, and motility. Dysregulated CEMIP expression was associated with progression and prognosis of tumors. The role of CEMIP in papillary thyroid carcinoma (PTC) remains unknown. Our study showed that CEMIP was upregulated in both tissues and cells of PTC. Silencing of CEMIP reduced cell proliferation and suppressed migration and invasion of PTC. Protein expression of phosphorylated STAT3 (Signal Transducer And Activator Of Transcription 3) (p-STAT3), AKT (p-AKT) and p65 (p-p65) were decreased by CEMIP silencing in PTC cells. Pyruvate dehydrogenase kinase 4 (PDK4) over-expression attenuated CEMIP silencing-induced decrease in p-STAT3, p-AKT and p-p65. Silencing of CEMIP-induced decrease in cell proliferation and metastasis in PTC were restored by over-expression of STAT3. CEMIP functioned as an oncogenic gene in PTC through PDK4-mediated activation of STAT3/AKT/NF-κB pathway.
Collapse
Affiliation(s)
- Min Zhou
- Department of Pathology, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Wei Hua
- Department of Pathology, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Yulan Sun
- Department of Pathology, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| |
Collapse
|
19
|
Schmaus A, Rothley M, Schreiber C, Möller S, Roßwag S, Franz S, Garvalov BK, Thiele W, Spataro S, Herskind C, Prunotto M, Anderegg U, Schnabelrauch M, Sleeman J. Sulfated hyaluronic acid inhibits the hyaluronidase CEMIP and regulates the HA metabolism, proliferation and differentiation of fibroblasts. Matrix Biol 2022; 109:173-191. [DOI: 10.1016/j.matbio.2022.04.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Revised: 03/12/2022] [Accepted: 04/04/2022] [Indexed: 12/23/2022]
|
20
|
Kokoretsis D, Maniaki EK, Kyriakopoulou K, Koutsakis C, Piperigkou Z, Karamanos NK. Hyaluronan as "Agent Smith" in cancer extracellular matrix pathobiology: Regulatory roles in immune response, cancer progression and targeting. IUBMB Life 2022; 74:943-954. [PMID: 35261139 DOI: 10.1002/iub.2608] [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: 12/30/2021] [Revised: 02/08/2022] [Accepted: 02/14/2022] [Indexed: 12/16/2022]
Abstract
Extracellular matrix (ECM) critically regulates cancer cell behavior by governing cell signaling and properties. Hyaluronan (HA) acts as a structural and functional ECM component that mediates critical properties of cancer cells in a molecular size-dependent manner. HA fragments secreted by cancer-associated fibroblasts (CAFs) reveal the correlation of HA to CAF-mediated matrix remodeling, a key step for the initiation of metastasis. The main goal of this article is to highlight the vital functions of HA in cancer cell initiation and progression as well as HA-mediated paracrine interactions among cancer and stromal cells. Furthermore, the HA implication in mediating immune responses to cancer progression is also discussed. Novel data on the role of HA in the formation of pre-metastatic niche may contribute towards the improvement of current theranostic approaches that benefit cancer management.
Collapse
Affiliation(s)
- Dimitris Kokoretsis
- Biochemistry, Biochemical Analysis & Matrix Pathobiology Research Group, Laboratory of Biochemistry, Department of Chemistry, University of Patras, Patras, Greece
| | - Evangelia-Konstantina Maniaki
- Biochemistry, Biochemical Analysis & Matrix Pathobiology Research Group, Laboratory of Biochemistry, Department of Chemistry, University of Patras, Patras, Greece
| | - Konstantina Kyriakopoulou
- Biochemistry, Biochemical Analysis & Matrix Pathobiology Research Group, Laboratory of Biochemistry, Department of Chemistry, University of Patras, Patras, Greece
| | - Christos Koutsakis
- Biochemistry, Biochemical Analysis & Matrix Pathobiology Research Group, Laboratory of Biochemistry, Department of Chemistry, University of Patras, Patras, Greece
| | - Zoi Piperigkou
- Biochemistry, Biochemical Analysis & Matrix Pathobiology Research Group, Laboratory of Biochemistry, Department of Chemistry, University of Patras, Patras, Greece.,Foundation for Research and Technology-Hellas (FORTH)/Institute of Chemical Engineering Sciences (ICE-HT), Patras, Greece
| | - Nikos K Karamanos
- Biochemistry, Biochemical Analysis & Matrix Pathobiology Research Group, Laboratory of Biochemistry, Department of Chemistry, University of Patras, Patras, Greece.,Foundation for Research and Technology-Hellas (FORTH)/Institute of Chemical Engineering Sciences (ICE-HT), Patras, Greece
| |
Collapse
|
21
|
Cloud AS, Vargheese AM, Gunewardena S, Shimak RM, Ganeshkumar S, Kumaraswamy E, Jensen RA, Chennathukuzhi VM. Loss of REST in breast cancer promotes tumor progression through estrogen sensitization, MMP24 and CEMIP overexpression. BMC Cancer 2022; 22:180. [PMID: 35177031 PMCID: PMC8851790 DOI: 10.1186/s12885-022-09280-2] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Accepted: 02/08/2022] [Indexed: 02/01/2023] Open
Abstract
BACKGROUND Breast cancer is the most common malignancy in women, and is both pathologically and genetically heterogeneous, making early detection and treatment difficult. A subset of breast cancers express normal levels of REST (repressor element 1 silencing transcription factor) mRNA but lack functional REST protein. Loss of REST function is seen in ~ 20% of breast cancers and is associated with a more aggressive phenotype and poor prognosis. Despite the frequent loss of REST, little is known about the role of REST in the molecular pathogenesis of breast cancer. METHODS TCGA data was analyzed for the expression of REST target genes in breast cancer patient samples. We then utilized gene knockdown in MCF-7 cells in the presence or absence of steroid hormones estrogen and/ progesterone followed by RNA sequencing, as well as chromatin immunoprecipitation and PCR in an attempt to understand the tumor suppressor role of REST in breast cancer. RESULTS We show that REST directly regulates CEMIP (cell migration-inducing and hyaluronan-binding protein, KIAA1199) and MMP24 (matrix metallopeptidase 24), genes known to have roles in invasion and metastasis. REST knockdown in breast cancer cells leads to significant upregulation of CEMIP and MMP24. In addition, we found REST binds to RE-1 sites (repressor element-1) within the genes and influences their transcription. Furthermore, we found that the estrogen receptor (ESR1) signaling pathway is activated in the absence of REST, regardless of hormone treatment. CONCLUSIONS We demonstrate a critical role for the loss of REST in aggressive breast cancer pathogenesis and provide evidence for REST as an important diagnostic marker for personalized treatment plans.
Collapse
Affiliation(s)
- Ashley S. Cloud
- grid.412016.00000 0001 2177 6375Department of Molecular and Integrative Physiology, University of Kansas Medical Center, Kansas City, KS USA
| | - Aditya M. Vargheese
- grid.412016.00000 0001 2177 6375Department of Molecular and Integrative Physiology, University of Kansas Medical Center, Kansas City, KS USA ,grid.468219.00000 0004 0408 2680The University of Kansas Cancer Center, Kansas City, KS USA ,grid.266515.30000 0001 2106 0692University of Kansas, Lawrence, KS USA
| | - Sumedha Gunewardena
- grid.412016.00000 0001 2177 6375Department of Molecular and Integrative Physiology, University of Kansas Medical Center, Kansas City, KS USA ,grid.412016.00000 0001 2177 6375Department of Biostatistics, University of Kansas Medical Center, Kansas City, KS USA
| | - Raeann M. Shimak
- grid.468219.00000 0004 0408 2680The University of Kansas Cancer Center, Kansas City, KS USA ,grid.412016.00000 0001 2177 6375Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS USA
| | - Sornakala Ganeshkumar
- grid.412016.00000 0001 2177 6375Department of Molecular and Integrative Physiology, University of Kansas Medical Center, Kansas City, KS USA
| | - Easwari Kumaraswamy
- grid.468219.00000 0004 0408 2680The University of Kansas Cancer Center, Kansas City, KS USA ,grid.412016.00000 0001 2177 6375Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS USA
| | - Roy A. Jensen
- grid.468219.00000 0004 0408 2680The University of Kansas Cancer Center, Kansas City, KS USA ,grid.266515.30000 0001 2106 0692University of Kansas, Lawrence, KS USA ,grid.412016.00000 0001 2177 6375Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS USA ,grid.412016.00000 0001 2177 6375Department of Cancer Biology, University of Kansas Medical Center, Kansas City, KS USA ,grid.412016.00000 0001 2177 6375Department of Anatomy and Cell Biology, University of Kansas Medical Center, Kansas City, KS USA
| | - Vargheese M. Chennathukuzhi
- grid.412016.00000 0001 2177 6375Department of Molecular and Integrative Physiology, University of Kansas Medical Center, Kansas City, KS USA ,grid.468219.00000 0004 0408 2680The University of Kansas Cancer Center, Kansas City, KS USA
| |
Collapse
|
22
|
Xue J, Zhu X, Qiao X, Wang Y, Bu J, Zhang X, Ma Q, Liang L, Sun L, Liu C. CEMIP as a potential biomarker and therapeutic target for breast cancer patients. Int J Med Sci 2022; 19:434-445. [PMID: 35370456 PMCID: PMC8964326 DOI: 10.7150/ijms.58067] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Accepted: 01/07/2022] [Indexed: 11/25/2022] Open
Abstract
Purpose: We aimed to evaluate whether CEMIP plays any role in the survival outcome of breast cancer (BC) patients, as well as to explore the regulatory mechanism of CEMIP in BC. Methods: We evaluated the expression and prognostic effect of CEMIP in BC patients using the Oncomine, GEPIA, UALCAN, and Kaplan-Meier plotter databases. Additionally, we detected CEMIP mRNA and protein levels in BC and normal tissues via PCR and western blotting analyses. Through immunochemistry analysis, we quantified CEMIP expression in 233 samples from BC patients. We then analyzed the link between the survival outcomes and CEMIP expression based on these clinical samples. Furthermore, we explored the immune-related molecules regulated by CEMIP and its coexpressed genes using the STRING database. Results: CEMIP expression was higher in BC tissues than in normal tissues. Patients with high CEMIP mRNA levels had a worse survival outcome. Similarly, patients expressing CEMIP had significantly shorter overall survival and disease-free survival than those not expressing the protein (P < 0.01). Some lymphocytes, immune inhibitors, immune stimulators, MHC molecules, chemokines, and chemokine receptors can be regulated by CEMIP, and CEMIP and its coexpressed genes can participate in the hyaluronan biosynthetic process, hyaluronan catabolic process, and other related biological processes in the progression of BC. Conclusion: Compared to normal tissues, BC tissues had higher number of CEMIP transcripts. CEMIP expression was associated with an adverse prognosis. CEMIP and its coexpressed genes can participate in the progression of BC. Therefore, CEMIP may be a potential biomarker for the treatment of BC patients.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | - Caigang Liu
- Department of Oncology, Shengjing Hospital of China Medical University, Shenyang, Liaoning Province, 110004, China
| |
Collapse
|
23
|
Singh N, Sharma R, Bose S. Meta-analysis of transcriptomics data identifies potential biomarkers and their associated regulatory networks in gallbladder cancer. GASTROENTEROLOGY AND HEPATOLOGY FROM BED TO BENCH 2022; 15:311-325. [PMID: 36762219 PMCID: PMC9876761 DOI: 10.22037/ghfbb.v15i4.2292] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/24/2022] [Accepted: 09/21/2022] [Indexed: 02/11/2023]
Abstract
Aim This study aimed to identify key genes, non-coding RNAs, and their possible regulatory interactions during gallbladder cancer (GBC). Background The early detection of GBC, i.e. before metastasis, is restricted by our limited knowledge of molecular markers and mechanism(s) involved during carcinogenesis. Therefore, identifying important disease-associated transcriptome-level alterations can be of clinical importance. Methods In this study, six NCBI-GEO microarray dataseries of GBC and control tissue samples were analyzed to identify differentially expressed genes (DEGs) and non-coding RNAs {microRNAs (DEmiRNAs) and long non-coding RNAs (DElncRNAs)} with a computational meta-analysis approach. A series of bioinformatic methods were applied to enrich functional pathways, create protein-protein interaction networks, identify hub genes, and screen potential targets of DEmiRNAs and DElncRNAs. Expression and interaction data were consolidated to reveal putative DElncRNAs:DEmiRNAs:DEGs interactions. Results In total, 351 DEGs (185 downregulated, 166 upregulated), 787 DEmiRNAs (299 downregulated, 488 upregulated), and 7436 DElncRNAs (3127 downregulated, 4309 upregulated) were identified. Eight genes (FGF, CDK1, RPN2, SEC61A1, SOX2, CALR, NGFR, and NCAM) were identified as hub genes. Genes associated with ubiquitin ligase activity, N-linked glycosylation, and blood coagulation were upregulated, while those for cell-cell adhesion, cell differentiation, and surface receptor-linked signaling were downregulated. DEGs-DEmiRNAs-DElncRNAs interaction network identified 46 DElncRNAs to be associated with 28 DEmiRNAs, consecutively regulating 27 DEGs. DEmiRNAs-hsa-miR-26b-5p and hsa-miR-335-5p; and DElnRNAs-LINC00657 and CTB-89H12.4 regulated the highest number of DEGs and DEmiRNAs, respectively. Conclusion The current study has identified meaningful transcriptome-level changes and gene-miRNA-lncRNA interactions during GBC and laid a platform for future studies on novel prognostic and diagnostic markers in GBC.
Collapse
Affiliation(s)
- Nidhi Singh
- Department of Biotechnology, Gauhati University, Guwahati, Assam, India
| | - Rinku Sharma
- Department of Life Sciences, Shiv Nadar University, Noida, Uttar Pradesh, India
| | - Sujoy Bose
- Department of Biotechnology, Gauhati University, Guwahati, Assam, India
| |
Collapse
|
24
|
Periyasamy L, Muruganantham B, Park WY, Muthusami S. Phyto-targeting the CEMIP Expression as a Strategy to Prevent Pancreatic Cancer Metastasis. Curr Pharm Des 2022; 28:922-946. [PMID: 35236267 DOI: 10.2174/1381612828666220302153201] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2021] [Accepted: 12/16/2021] [Indexed: 11/22/2022]
Abstract
INTRODUCTION Metastasis of primary pancreatic cancer (PC) to adjacent or distant organs is responsible for the poor survival rate of affected individuals. Chemotherapy, radiotherapy, and immunotherapy are currently being prescribed to treat PC in addition to surgical resection. Surgical resection is the preferred treatment for PC that leads to 20% of 5-year survival, but only less than 20% of patients are eligible for surgical resection because of the poor prognosis. To improve the prognosis and clinical outcome, early diagnostic markers need to be identified, and targeting them would be of immense benefit to increase the efficiency of the treatment. Cell migration-inducing hyaluronan-binding protein (CEMIP) is identified as an important risk factor for the metastasis of various cancers, including PC. Emerging studies have pointed out the crucial role of CEMIP in the regulation of various signaling mechanisms, leading to enhanced migration and metastasis of PC. METHODS The published findings on PC metastasis, phytoconstituents, and CEMIP were retrieved from Pubmed, ScienceDirect, and Cochrane Library. Computational tools, such as gene expression profiling interactive analysis (GEPIA) and Kaplan-Meier (KM) plotter, were used to study the relationship between CEMIP expression and survival of PC individuals. RESULTS Gene expression analysis using the GEPIA database identified a stupendous increase in the CEMIP transcript in PC compared to adjacent normal tissues. KM plotter analysis revealed the impact of CEMIP on the overall survival (OS) and disease-free survival (DFS) among PC patients. Subsequently, several risk factors associated with PC development were screened, and their ability to regulate CEMIP gene expression was analyzed using computational tools. CONCLUSION The current review is focused on gathering information regarding the regulatory role of phytocomponents in PC migration and exploring their possible impact on the CEMIP expression.
Collapse
Affiliation(s)
- Loganayaki Periyasamy
- Department of Biochemistry, Karpagam Academy of Higher Education, Coimbatore, Tamil Nadu, 641 021, India
| | - Bharathi Muruganantham
- Karpagam Cancer Research Centre, Karpagam Academy of Higher Education, Coimbatore, Tamil Nadu, 641 021, India
| | - Woo-Yoon Park
- Department of Radiation Oncology, Chungbuk National University College of Medicine, Cheongju 28644, Republic of Korea
| | - Sridhar Muthusami
- Department of Biochemistry, Karpagam Academy of Higher Education, Coimbatore, Tamil Nadu, 641 021, India
- Karpagam Cancer Research Centre, Karpagam Academy of Higher Education, Coimbatore, Tamil Nadu, 641 021, India
| |
Collapse
|
25
|
Chen Y, Zhou H, Jiang WJ, Wang JF, Tian Y, Jiang Y, Xia BR. The role of CEMIP in tumors: An update based on cellular and molecular insights. Biomed Pharmacother 2021; 146:112504. [PMID: 34922110 DOI: 10.1016/j.biopha.2021.112504] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Revised: 12/01/2021] [Accepted: 12/02/2021] [Indexed: 01/17/2023] Open
Abstract
CEMIP was initially identified as an inner-ear specific protein in which three-point mutations cause folding changes in protein structure associated with non-syndromic hearing loss. CEMIP was also involved in other cellular activities, such as hyaluronan depolymerization independent of CD44 and other hyaluronidases. Growing evidence has demonstrated that CEMIP is involved in the progression of various tumors. However, whether the oncogenic effects of CEMIP relies on its enzymatic activity remain elusive. CEMIP is significantly related to metastasis and poor prognosis in patients with various tumors, suggesting that CEMIP is a potential, highly specific diagnostic tumor marker. Most preclinical experiments have shown that the overexpression of CEMIP in tumors mainly affects the adhesion, metastasis, and invasion of tumor cells and EMT. Other studies have also demonstrated that CEMIP can promote a variety of tumor processes by affecting tumor proliferation, dedifferentiation, and the tumor microenvironment. In terms of molecular mechanisms, existing research has shown that CEMIP mainly affects the WNT and EGFR signaling pathways. In addition, a variety of miRNAs have been shown to inhibit CEMIP in tumors. This paper elaborates on the clinical characteristics and regulatory dysfunction of CEMIP in different cancers. CEMIP provides a new potential target for therapy of multiple tumors, which is worthy of further study.
Collapse
Affiliation(s)
- Yu Chen
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of USTC, Division of Life, Sciences and Medicine, University of Science and Technology of China, Hefei 230031, China
| | - Hu Zhou
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of USTC, Division of Life, Sciences and Medicine, University of Science and Technology of China, Hefei 230031, China
| | - Wen-Jing Jiang
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of USTC, Division of Life, Sciences and Medicine, University of Science and Technology of China, Hefei 230031, China
| | - Jia-Fei Wang
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of USTC, Division of Life, Sciences and Medicine, University of Science and Technology of China, Hefei 230031, China
| | - Yuan Tian
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of USTC, Division of Life, Sciences and Medicine, University of Science and Technology of China, Hefei 230031, China
| | - Yan Jiang
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of USTC, Division of Life, Sciences and Medicine, University of Science and Technology of China, Hefei 230031, China
| | - Bai-Rong Xia
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of USTC, Division of Life, Sciences and Medicine, University of Science and Technology of China, Hefei 230031, China.
| |
Collapse
|
26
|
Interferon-induced transmembrane protein 2 promotes epithelial-mesenchymal transition by activating transforming growth factor-β1/small mother against decapentaplegic 2 signaling in gastric cancer. Mol Biol Rep 2021; 49:997-1006. [PMID: 34855108 DOI: 10.1007/s11033-021-06919-4] [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: 08/19/2021] [Accepted: 10/30/2021] [Indexed: 02/06/2023]
Abstract
BACKGROUND Gastric cancer (GC) is one of the most prevalent malignancy around the world. Primary tumor cells are enabled to invade and migrate into adjacent normal tissues to form secondary tumors. Epithelial-mesenchymal transitions (EMT) plays a pivotal role in facilitating tumor progression. Abundant evidence suggested that the transforming growth factor-β1 (TGF-β1) triggered the process of EMT. Nonetheless, the precise molecular mechanisms underlying EMT requires further elucidation, and there still lacks effective specific therapeutic target. In our recent research, we demonstrated that the interferon (IFN)-induced transmembrane protein 2 (IFITM2) promoted the growth and metastasis of GC. However, it remains unclear whether IFITM2 involves in TGF-β1 mediated EMT in GC. METHODS AND RESULTS In the present research, we investigated the functional role of IFITM2 in EMT process and TGF-β1 signaling pathway in two GC cell lines. We noticed that silencing IFITM2 can effectively inhibit TGF-β1 signaling mediated EMT by regulating down stream small mother against decapentaplegic (SMAD) 2/3 and transcription factors.This finding was further determined in both tumor tissues from GC patients and normal tissues adjacent to cancer. Our data demonstrated the key role of IFITM2 in TGF-β1 signaling and EMT in GC. CONCLUSION The findings enriched our understanding of the underlying mechanism in EMT during the progression of GC. In addition, IFITM2 would be a potential target for treating GC and other malignant tumors.
Collapse
|
27
|
Mi C, Zhang D, Li Y, Ren M, Ma W, Lu G, He S. miR-4677-3p participates proliferation and metastases of gastric cancer cell via CEMIP-PI3K/AKT signaling pathway. Cell Cycle 2021; 20:1978-1987. [PMID: 34437815 DOI: 10.1080/15384101.2021.1971375] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
Gastric cancer is one of the top three leading causes of cancer-related death in the world. Evidence indicated that miR-4677-3p was dysregulated and involved in modulating invasion and migration in multiple types of cancer cells. The aim of this research is to explore the function and mechanism of miR-4677-3p in the development of gastric cancer. In this study, we discovered that miR-4677-3p was down-regulated in gastric cancer tissues and cells. Over-expression of miR-4677-3p suppressed the proliferation, migration and invasion of gastric cancer cells. Furthermore, miR-4677-3p directly bond to CEMIP 3'UTR region and inhibited CEMIP expression. CEMIP promoted cell proliferation, migration and invasion of gastric cancer cells via accelerating PI3K/AKT signaling pathway. siCEMIP or PI3K/AKT signaling inhibitor (Akti-1/2 and LY294002) partly reversed the effects of miR-4677-3p on the cellular growth and metastasis of gastric cancer. In general, miR-4677-3p regulated the development of gastric cancer through CEMIP-PI3K/AKT signaling pathway axis. This study verified the function and molecular mechanism of miR-4677-3p in gastric cancer cells, and may provide a potential diagnosis/prognosis target for patients with gastric cancer.
Collapse
Affiliation(s)
- Chen Mi
- Department of Gastroenterology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi 'An City, Shaanxi Province, China
| | - Dan Zhang
- Department of Gastroenterology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi 'An City, Shaanxi Province, China
| | - Yarui Li
- Department of Gastroenterology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi 'An City, Shaanxi Province, China
| | - Mudan Ren
- Department of Gastroenterology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi 'An City, Shaanxi Province, China
| | - Wenhui Ma
- Department of Gastroenterology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi 'An City, Shaanxi Province, China
| | - Guifang Lu
- Department of Gastroenterology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi 'An City, Shaanxi Province, China
| | - Shuixiang He
- Department of Gastroenterology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi 'An City, Shaanxi Province, China
| |
Collapse
|
28
|
Guo H, Yang J, Liu S, Qin T, Zhao Q, Hou X, Ren L. Prognostic marker identification based on weighted gene co-expression network analysis and associated in vitro confirmation in gastric cancer. Bioengineered 2021; 12:4666-4680. [PMID: 34338150 PMCID: PMC8806585 DOI: 10.1080/21655979.2021.1957645] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
The aim of this study was to explore the potential molecular mechanisms of Gastric cancer (GC) and identify new prognostic markers for GC. RNA sequencing data were downloaded from the Gene Expression Omnibus database, and 418 differentially expressed genes (DEGs) were screened. Weighted correlation network analysis (WGCNA) was performed to identify six hub modules related to the clinical features of GC. Cytoscape software was used to identify five hub genes in the co-expression network, including CST1, CEMIP, COL8A1, PMEPA1, and MSLN. The TCGA database was used to verify hub gene expression in GC. The overall survival in the high CEMIP expression group was significantly lower than that of patients in the low CEMIP expression group. CEMIP expression was also found to be negatively correlated with B cell and CD4 + T cell infiltration. Further, associated in vitro experiments confirmed that CEMIP downregulation suppressed the proliferation and migration of GC cells and impaired the chemoresistance of GC cells to 5-fluorouracil. Our study effectively identified and validated prognostic biomarkers for GC, laying a new foundation for the therapeutic target, occurrence, and development of gastric cancer.
Collapse
Affiliation(s)
- Haonan Guo
- Department of Clinical Laboratory, The Affiliated Hospital of Guilin Medical University, Guangxi, Guilin, China
| | - Jun Yang
- Department of Clinical Laboratory, The Affiliated Hospital of Guilin Medical University, Guangxi, Guilin, China
| | - Shanshan Liu
- Department of Clinical Laboratory, The Affiliated Hospital of Guilin Medical University, Guangxi, Guilin, China
| | - Tao Qin
- Department of Clinical Laboratory, The Affiliated Hospital of Guilin Medical University, Guangxi, Guilin, China
| | - Qianwen Zhao
- Department of Clinical Laboratory, The Affiliated Hospital of Guilin Medical University, Guangxi, Guilin, China
| | - Xianliang Hou
- Central Laboratory, Guangxi Health Commission Key Laboratory of Glucose and Lipid Metabolism Disorders, The Second Affiliated Hospital of Guilin Medical University, Guilin, Guangxi, China
| | - Lei Ren
- Department of Clinical Laboratory, The Affiliated Hospital of Guilin Medical University, Guangxi, Guilin, China
| |
Collapse
|
29
|
Huo M, Wang H, Li L, Tong Y, Hu C, Gu Y, Liu J, Yin T. Redox-sensitive hyaluronic acid-cholesterol nanovehicles potentiate efficient transmembrane internalization and controlled release for penetrated "full-line" inhibition of pre-metastatic initiation. J Control Release 2021; 336:89-104. [PMID: 34119559 DOI: 10.1016/j.jconrel.2021.06.013] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Revised: 05/14/2021] [Accepted: 06/09/2021] [Indexed: 12/11/2022]
Abstract
Metastatic breast cancer is a major cause of cancer-related mortality worldwide. The tumor-specific penetration and triggered drug release for "full-line" inhibition of pre-metastatic initiation are of essential importance in improving mortality rates. Here, a crosslinked, redox-sensitive amphiphilic conjugate (cHLC) was constructed with a combination of features, including hyaluronic acid (HA)-mediated tumor active targeting, lipoic acid (LA) core-crosslinking based bio-stability and reducibility, and lipid raft anchoring-promoted HA-mediated endocytosis through cholesterol (CHO) modification for the penetrated co-delivery of paclitaxel (PTX) and the multi-targeted anti-metastatic agent, silibinin (SB). Resultantly, the nanodrug (cHLC/(PTX + SB)) demonstrated enhanced tumor cytoplasm-selective rapid drug delivery in a 4T1 model both in vitro and in vivo. The released SB efficiently sensitized cells to PTX treatment and inhibited the whole process of pre-metastatic initiation including epithelial-to-mesenchymal transition (EMT), local and blood vessel invasion. The exquisite design of this delivery system provides a deep insight into enhancing focus accessibility of multi-targeted drugs for an efficient inhibition of tumor metastasis.
Collapse
Affiliation(s)
- Meirong Huo
- Department of Pharmaceutics, China Pharmaceutical University, 639 Longmian Avenue, Nanjing 211198, China
| | - Honglan Wang
- Department of Pharmaceutics, China Pharmaceutical University, 639 Longmian Avenue, Nanjing 211198, China
| | - Lingchao Li
- Department of Pharmaceutics, China Pharmaceutical University, 639 Longmian Avenue, Nanjing 211198, China
| | - Yuqing Tong
- Department of Pharmaceutics, China Pharmaceutical University, 639 Longmian Avenue, Nanjing 211198, China
| | - Chengxia Hu
- Department of Pharmaceutics, China Pharmaceutical University, 639 Longmian Avenue, Nanjing 211198, China
| | - Yongwei Gu
- Department of Pharmacy, Fudan University Shanghai Cancer Center, Shanghai 200032, China
| | - Jiyong Liu
- Department of Pharmacy, Fudan University Shanghai Cancer Center, Shanghai 200032, China.
| | - Tingjie Yin
- Department of Pharmaceutics, China Pharmaceutical University, 639 Longmian Avenue, Nanjing 211198, China.
| |
Collapse
|
30
|
Tabatabai E, Khazaei M, Asgharzadeh F, Nazari SE, Shakour N, Fiuji H, Ziaeemehr A, Mostafapour A, Parizadeh MR, Nouri M, Hassanian SM, Hadizadeh F, Ferns GA, Rahmati M, Rahmani F, Avan A. Inhibition of angiotensin II type 1 receptor by candesartan reduces tumor growth and ameliorates fibrosis in colorectal cancer. EXCLI JOURNAL 2021; 20:863-878. [PMID: 34121975 PMCID: PMC8192880 DOI: 10.17179/excli2021-3421] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/19/2021] [Accepted: 04/28/2021] [Indexed: 12/23/2022]
Abstract
Colorectal cancer (CRC) is an important cause of cancer-related mortality. Aberrant activation of the renin-angiotensin system (RAS) is reported to be associated with poor clinical outcomes in patients with CRC. This study was designed to explore the anti-tumor effects of the angiotensin receptor blocker Candesartan either alone or in combination with 5-FU in in vitro and in vivo models of CRC. The cytotoxic effects of Candesartan were assessed using the MTT assay in two colorectal cancer cell lines (CT-26 and SW-480). To investigate the potential regulatory role of Candesartan on tumor growth, apoptosis, and migration, the expression levels of Cyclin D1, Survivin, MMP3, MMP9, and E-cadherin mRNAs were evaluated. The oxidant/antioxidant balance was also examined by determining the levels of MDA, thiols, SOD, and CAT. We used a xenograft model of colon cancer to investigate the effects of Candesartan alone, or in combination with 5-FU, on tumor growth following histological staining (Hematoxylin & Eosin and Masson trichrome staining) and biochemical studies as well as gene expression analyses by RT-PCR and western blotting. Candesartan suppressed tumor cell proliferation and migration by modulating Cyclin D1, MMP3/9, and E-cadherin. Treatment with Candesartan either alone, or in combination with 5-FU decreased tumor size in the mouse model, and also increased the level of oxidative markers MDA and reduced CAT, SOD, and thiols. Histological evaluation showed that Candesartan increased tumor necrosis, reduced tumor density and attenuated collagen deposition reducing tumor fibrosis in tumor xenograft. Candesartan, an inhibitor of the RAS, when used in combination with 5-FU reduced tumor growth by inhibiting fibrosis and inducing ROS production, supporting further clinical studies on this therapeutic approach for treatment of CRC.
Collapse
Affiliation(s)
- Ehsan Tabatabai
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Clinical Biochemistry, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Majid Khazaei
- Department of Physiology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.,Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Fereshteh Asgharzadeh
- Department of Physiology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Seyedeh Elnaz Nazari
- Department of Physiology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Neda Shakour
- Department of Medicinal Chemistry, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran.,Student Research Committee, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Hamid Fiuji
- Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Aghigh Ziaeemehr
- Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Asma Mostafapour
- Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | | | - Mohammad Nouri
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Clinical Biochemistry, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Seyed Mahdi Hassanian
- Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.,Department of Medical Biochemistry, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Farzin Hadizadeh
- Department of Medicinal Chemistry, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran.,Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Gordon A Ferns
- Brighton & Sussex Medical School, Division of Medical Education, Falmer, Brighton, Sussex BN1 9PH, UK
| | - Mohammad Rahmati
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Clinical Biochemistry, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Farzad Rahmani
- Iranshahr University of Medical Sciences, Iranshahr, Iran
| | - Amir Avan
- Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.,Medical Genetics Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
31
|
Tsuji S, Nakamura S, Yamada T, de Vega S, Okada Y, Inoue S, Shimazawa M, Hara H. HYBID derived from tumor cells and tumor-associated macrophages contribute to the glioblastoma growth. Brain Res 2021; 1764:147490. [PMID: 33887254 DOI: 10.1016/j.brainres.2021.147490] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Revised: 04/07/2021] [Accepted: 04/14/2021] [Indexed: 12/14/2022]
Abstract
Glioblastoma is the most malignant tumor of the brain associated with poor prognosis and outcome, and hence there is an urgent need to develop novel treatments for glioblastoma. In this study, we focused on hyaluronan binding protein (HYBID, as known as CEMIP/KIAA1199), a protein involved in hyaluronan depolymerization in chondrocytes and synoviocytes. We previously reported that Hybid-deficient (KO) mice show accumulation of hyaluronan in the brain, and memory impairment. To elucidate the role of HYBID in glioblastoma pathogenesis, we knocked down HYBID in human glioblastoma cells using siRNAs and developed a murine orthotopic xenograft model in the Hybid KO mice. Downregulation of HYBID in glioblastoma cells resulted in inhibition of cell proliferation and migration, and increased cell death. The growth of glioblastoma cells implanted in the mouse brain was suppressed in Hybid KO mice compared to that in the wild-type mice. Interestingly, infiltration of macrophages in the glioblastoma tissue was decreased in Hybid KO mice. Using intraperitoneal macrophages derived from Hybid KO mice and glioma cell supernatants, we examined the role of HYBID in macrophages in the tumor environment. We showed that HYBID contributes to macrophage migration and the release of pro-tumor factors. Moreover, we revealed that HYBID can be a poor prognostic factor in glioma patients by bioinformatics approaches. Our study provides data to support that HYBID expressed by both glioblastoma cells and tumor-associated macrophages may contribute to glioblastoma progression and suggests that HYBID may be a potential target for therapy that focuses on the tumor microenvironment of glioblastoma.
Collapse
Affiliation(s)
- Shohei Tsuji
- Molecular Pharmacology, Department of Biofunctional Evaluation, Gifu Pharmaceutical University, Gifu, Japan
| | - Shinsuke Nakamura
- Molecular Pharmacology, Department of Biofunctional Evaluation, Gifu Pharmaceutical University, Gifu, Japan
| | - Tetsuya Yamada
- Molecular Pharmacology, Department of Biofunctional Evaluation, Gifu Pharmaceutical University, Gifu, Japan; Department of Neurosurgery, Gifu University School of Medicine, Gifu, Japan
| | - Susana de Vega
- Department of Pathophysiology for Locomotive and Neoplastic Diseases, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Yasunori Okada
- Department of Pathophysiology for Locomotive and Neoplastic Diseases, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Shintaro Inoue
- Cosmetic Health Science, Gifu Pharmaceutical University, Gifu, Japan
| | - Masamitsu Shimazawa
- Molecular Pharmacology, Department of Biofunctional Evaluation, Gifu Pharmaceutical University, Gifu, Japan
| | - Hideaki Hara
- Molecular Pharmacology, Department of Biofunctional Evaluation, Gifu Pharmaceutical University, Gifu, Japan.
| |
Collapse
|
32
|
Wu C, Huang H, Choi HY, Ma Y, Zhou T, Peng Y, Pang K, Shu G, Yang X. Anti-esophageal Cancer Effect of Corilagin Extracted from Phmllanthi Fructus via the Mitochondrial and Endoplasmic Reticulum Stress Pathways. JOURNAL OF ETHNOPHARMACOLOGY 2021; 269:113700. [PMID: 33346026 DOI: 10.1016/j.jep.2020.113700] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Revised: 12/07/2020] [Accepted: 12/13/2020] [Indexed: 06/12/2023]
Abstract
HEADINGS ETHNOPHARMACOLOGICAL RELEVANCE Corilagin (β-1-O-galloyl-3,6-(R)-hexahydroxydiphenoyl-d-glucose) is a tannin isolated from the traditional ethnopharmacological plant Phmllanthi Fructus, which is widely used in not only traditional Chinese medicine but also tropical and subtropical medicine to ameliorate various diseases. AIM OF THE STUDY This study was designed to isolate the potential anti-esophageal cancer (EC) component corilagin from Phmllanthi Fructus and explain its anti-EC mechanism. MATERIALS AND METHODS Corilagin was isolated from Phmllanthi Fructus by extraction and chromatographic procedures, and its anti-esophageal cancer effect was evaluated by in vitro and in vivo experiments. In vitro experiments included MTT analysis, flow cytometry, and the Transwell assay and were used to observe corilagin-mediated inhibition of EC cell growth. Western blotting was used to analyze the apoptotic pathway of EC cells. In vivo experiments used tumor-bearing nude mice to evaluate the antitumor effect of corilagin, and its potential mechanism was explored by Western blotting. RESULTS Corilagin showed significant anti-EC activity in vitro and in vivo. Corilagin was significantly cytotoxic to EC cells and induced apoptosis in EC cells. Corilagin induced G0/G1 phase arrest by altering key G0/G1 cell cycle regulatory markers and significantly reducing the migration of EC cells and the number of cells in a time- and dose-dependent manner. Additionally, corilagin inhibited the growth of transplanted tumors in nude mice without significant toxicity. Regarding the anticancer mechanism of corilagin, the results showed that corilagin inhibited esophageal cancer progression by activating mitochondrial and endoplasmic reticulum stress signaling pathways. CONCLUSIONS Corilagin shows significant anti-EC activity in vitro and in vivo. The mechanism of the anti-EC activity of corilagin may be due to activating mitochondrial and endoplasmic reticulum stress signaling pathways.
Collapse
Affiliation(s)
- Chaoqun Wu
- School of Pharmaceutical Sciences, South-Central University for Nationalities, Wuhan, 430074, China
| | - Huiqi Huang
- School of Pharmaceutical Sciences, South-Central University for Nationalities, Wuhan, 430074, China
| | - Ho-Young Choi
- College of Korean Medicine, Kyung Hee University, Seoul, 02447, Republic of Korea
| | - Yuanren Ma
- School of Pharmaceutical Sciences, South-Central University for Nationalities, Wuhan, 430074, China
| | - Tongxi Zhou
- School of Pharmaceutical Sciences, South-Central University for Nationalities, Wuhan, 430074, China
| | - Yu Peng
- School of Pharmaceutical Sciences, South-Central University for Nationalities, Wuhan, 430074, China
| | - Kejian Pang
- Hotian Uygur Pharmaceutical Co., Ltd, Hotian, 848200, China
| | - Guangwen Shu
- School of Pharmaceutical Sciences, South-Central University for Nationalities, Wuhan, 430074, China.
| | - Xinzhou Yang
- School of Pharmaceutical Sciences, South-Central University for Nationalities, Wuhan, 430074, China.
| |
Collapse
|
33
|
Liu J, Yan W, Han P, Tian D. The emerging role of KIAA1199 in cancer development and therapy. Biomed Pharmacother 2021; 138:111507. [PMID: 33773462 DOI: 10.1016/j.biopha.2021.111507] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2020] [Revised: 03/09/2021] [Accepted: 03/11/2021] [Indexed: 12/16/2022] Open
Abstract
KIAA1199, also known as CEMIP or HYBID, is an important member of the Human Unidentified Gene-Encoded (HUGE) database. Accumulated evidence has revealed that KIAA1199 is associated with tumor progression and metastasis in numerous malignancies, including colorectal, liver, gastric, pancreatic, breast, lung, prostate, ovarian and papillary thyroid cancers. As an oncogene, it plays crucial role in the proliferation, apoptosis, invasion and migration of various tumor cells. In addition, KIAA1199 is also involved in the regulation of multiple signal pathways such as epithelial-mesenchymal transition (EMT), Wnt/ β-catenin, MEK/ERK and PI3K/Akt. In this review, we summarized up to date advancement on the role of KIAA1199 in human cancer development, progression, and metastasis. We also addressed KIAA1199 as a potential therapeutic target for cancer therapy.
Collapse
Affiliation(s)
- Jingmei Liu
- Department of Gastroenterology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Wei Yan
- Department of Gastroenterology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Ping Han
- Department of Gastroenterology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China.
| | - Dean Tian
- Department of Gastroenterology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China.
| |
Collapse
|
34
|
Piperigkou Z, Kyriakopoulou K, Koutsakis C, Mastronikolis S, Karamanos NK. Key Matrix Remodeling Enzymes: Functions and Targeting in Cancer. Cancers (Basel) 2021; 13:1441. [PMID: 33809973 PMCID: PMC8005147 DOI: 10.3390/cancers13061441] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2021] [Revised: 03/05/2021] [Accepted: 03/17/2021] [Indexed: 12/13/2022] Open
Abstract
Tissue functionality and integrity demand continuous changes in distribution of major components in the extracellular matrices (ECMs) under normal conditions aiming tissue homeostasis. Major matrix degrading proteolytic enzymes are matrix metalloproteinases (MMPs), plasminogen activators, atypical proteases such as intracellular cathepsins and glycolytic enzymes including heparanase and hyaluronidases. Matrix proteases evoke epithelial-to-mesenchymal transition (EMT) and regulate ECM turnover under normal procedures as well as cancer cell phenotype, motility, invasion, autophagy, angiogenesis and exosome formation through vital signaling cascades. ECM remodeling is also achieved by glycolytic enzymes that are essential for cancer cell survival, proliferation and tumor progression. In this article, the types of major matrix remodeling enzymes, their effects in cancer initiation, propagation and progression as well as their pharmacological targeting and ongoing clinical trials are presented and critically discussed.
Collapse
Affiliation(s)
- Zoi Piperigkou
- Biochemistry, Biochemical Analysis and Matrix Pathobiology Research Group, Laboratory of Biochemistry, Department of Chemistry, University of Patras, 265 04 Patras, Greece; (K.K.); (C.K.)
- Foundation for Research and Technology-Hellas (FORTH)/Institute of Chemical Engineering Sciences (ICE-HT), 265 04 Patras, Greece
| | - Konstantina Kyriakopoulou
- Biochemistry, Biochemical Analysis and Matrix Pathobiology Research Group, Laboratory of Biochemistry, Department of Chemistry, University of Patras, 265 04 Patras, Greece; (K.K.); (C.K.)
| | - Christos Koutsakis
- Biochemistry, Biochemical Analysis and Matrix Pathobiology Research Group, Laboratory of Biochemistry, Department of Chemistry, University of Patras, 265 04 Patras, Greece; (K.K.); (C.K.)
| | | | - Nikos K. Karamanos
- Biochemistry, Biochemical Analysis and Matrix Pathobiology Research Group, Laboratory of Biochemistry, Department of Chemistry, University of Patras, 265 04 Patras, Greece; (K.K.); (C.K.)
- Foundation for Research and Technology-Hellas (FORTH)/Institute of Chemical Engineering Sciences (ICE-HT), 265 04 Patras, Greece
| |
Collapse
|
35
|
Yu L, Shang D, Lang L, Liu Y, Yu M, Song D, Jia S, Han S, Li C, Liu J, Xu Y, Zhang X. TMF, a natural dihydroflavonoid isolated from Scutellaria javanica Jungh, stimulates anticancer activity of s180 cancer-bearing mice, induces apoptosis, inhibits invasion and migration on HepG-2 cells. JOURNAL OF ETHNOPHARMACOLOGY 2020; 263:113072. [PMID: 32738393 DOI: 10.1016/j.jep.2020.113072] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/26/2019] [Revised: 05/07/2020] [Accepted: 05/31/2020] [Indexed: 06/11/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE 5-hydroxy-7,8,2',6'-tetramethoxy flavanone (TMF) is a dihydroflavonoid extracted from Scutellaria javanica Jungh. It is a species of genus Scutellaria, and a representative southern herb and Li nationality medicine. The plant has been used as an ethnic medicine in treating cancer and the main components are dihydroflavonoids. However, the underlying mechanisms are yet to be elucidated. AIM OF THE STUDY The present study aimed at investigating the efficacy of TMF in cancer and the underlying mechanisms. MATERIALS AND METHODS The s180 cancer-bearing mice experiment in vivo was designed to study the tumor growth inhibition of TMF. Also, we investigated the latent mechanism of TMF induced apoptosis and the inhibitory action of TMF on the metastasis and proliferation in HepG-2 cells. The in vitro experimental groups were treated with TMF or hydroxycamptothecine (HCPT) for 24 h. Apoptosis was detected by flow cytometry. Caspase-3 activity was detected by ELISA. The expressions of PCNA, Bcl-2, Bax, p53, E-Cadherin, MMP-9, MMP-2, STAT3, p-STAT3, JAK2, p-JAK2, AKT, p-AKT, ERK1/2 and p-ERK1/2 were examined by Western blot. RESULTS After oral administration of TMF in s180 cancer-bearing mice, tumor growth in vivo was suppressed significantly. The MTT assay result and the reduction of PCAN proved that TMF could inhibit HepG-2 cells proliferation. TMF also caused dose-dependent apoptosis on HepG-2 cells. The experimental results showed that the expression of Bcl-2 was reduced, and the expressions of caspase-3, Bax and p53 were increased. Therefore, we speculated that TMF-induced apoptosis might be achieved by regulating the p53-Bcl-2/Bax-caspase-3 pathways. Transwell cell migration and invasion assay showed that treatment with TMF inhibited the invasion and migration in HepG-2 cells. The expressions of MMP-9 and MMP-2 were decreased while that of E-cadherin was enhanced significantly by TMF. Additionally, the expressions of p-JAK2, p-STAT3, p-AKT and p-ERK1/2 were decreased, but those of JAK2, STAT3, AKT and ERK1/2 remained unchanged. Thus, it is indicated that TMF induced apoptosis and inhibited proliferation and metastasis on HepG-2 cells via JAK2/STAT3, MAPK/ERK and PI3K/AKT pathways. CONCLUSION The present results demonstrated that TMF could stimulate anticancer activity of s180 cancer-bearing mice, induce apoptosis, and inhibit invasion and migration on HepG-2 cells. Our findings displayed a systematic insight into the mechanisms underlying anticancer action of TMF, and provided a better understanding of its use for cancer.
Collapse
Affiliation(s)
- Lei Yu
- College of Pharmacy, Engineering Research Center for Medicine, Harbin University of Commerce, Harbin, 150076, PR China.
| | - Dongyu Shang
- College of Pharmacy, Engineering Research Center for Medicine, Harbin University of Commerce, Harbin, 150076, PR China; Engineering Research Center of Natural Anticancer Drugs, Ministry of Education, Harbin, 150076, PR China.
| | - Lang Lang
- College of Pharmacy, Engineering Research Center for Medicine, Harbin University of Commerce, Harbin, 150076, PR China.
| | - Yingjie Liu
- College of Pharmacy, Engineering Research Center for Medicine, Harbin University of Commerce, Harbin, 150076, PR China.
| | - Miao Yu
- College of Pharmacy, Engineering Research Center for Medicine, Harbin University of Commerce, Harbin, 150076, PR China.
| | - Dongxue Song
- College of Pharmacy, Engineering Research Center for Medicine, Harbin University of Commerce, Harbin, 150076, PR China.
| | - Shaohua Jia
- College of Pharmacy, Engineering Research Center for Medicine, Harbin University of Commerce, Harbin, 150076, PR China.
| | - Shuang Han
- College of Pharmacy, Engineering Research Center for Medicine, Harbin University of Commerce, Harbin, 150076, PR China; Engineering Research Center of Natural Anticancer Drugs, Ministry of Education, Harbin, 150076, PR China.
| | - Chuandi Li
- College of Pharmacy, Engineering Research Center for Medicine, Harbin University of Commerce, Harbin, 150076, PR China; Engineering Research Center of Natural Anticancer Drugs, Ministry of Education, Harbin, 150076, PR China.
| | - Jun Liu
- College of Pharmacy, Engineering Research Center for Medicine, Harbin University of Commerce, Harbin, 150076, PR China.
| | - Ying Xu
- College of Pharmacy, Engineering Research Center for Medicine, Harbin University of Commerce, Harbin, 150076, PR China.
| | - Xiaopo Zhang
- Key Laboratory of Tropical Translational Medicine of Ministry of Education, Hainan Key Laboratory for Research and Development of Tropical Herbs, School of Pharmacy, Hainan Medical University, Haikou, 57199, PR China.
| |
Collapse
|
36
|
Kobayashi T, Chanmee T, Itano N. Hyaluronan: Metabolism and Function. Biomolecules 2020; 10:E1525. [PMID: 33171800 PMCID: PMC7695009 DOI: 10.3390/biom10111525] [Citation(s) in RCA: 179] [Impact Index Per Article: 35.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2020] [Revised: 11/04/2020] [Accepted: 11/05/2020] [Indexed: 12/21/2022] Open
Abstract
As a major polysaccharide component of the extracellular matrix, hyaluronan plays essential roles in the organization of tissue architecture and the regulation of cellular functions, such as cell proliferation and migration, through interactions with cell-surface receptors and binding molecules. Metabolic pathways for biosynthesis and degradation tightly control the turnover rate, concentration, and molecular size of hyaluronan in tissues. Despite the relatively simple chemical composition of this polysaccharide, its wide range of molecular weights mediate diverse functions that depend on molecular size and tissue concentration. Genetic engineering and pharmacological approaches have demonstrated close associations between hyaluronan metabolism and functions in many physiological and pathological events, including morphogenesis, wound healing, and inflammation. Moreover, emerging evidence has suggested that the accumulation of hyaluronan extracellular matrix and fragments due to the altered expression of hyaluronan synthases and hyaluronidases potentiates cancer development and progression by remodeling the tumor microenvironment. In addition to the well-known functions exerted by extracellular hyaluronan, recent metabolomic approaches have also revealed that its synthesis can regulate cellular functions via the reprogramming of cellular metabolism. This review highlights the current advances in knowledge on the biosynthesis and catabolism of hyaluronan and describes the diverse functions associated with hyaluronan metabolism.
Collapse
Affiliation(s)
- Takashi Kobayashi
- Institute for Molecular Science of Medicine, Aichi Medical University, Nagakute, Aichi 480-1195, Japan;
| | - Theerawut Chanmee
- Department of Clinical Chemistry, Faculty of Medical Technology, Mahidol University, Phutthamonthon, Nakhon Pathom 73170, Thailand;
| | - Naoki Itano
- Department of Molecular Biosciences, Faculty of Life Sciences, Kyoto Sangyo University, Kita-ku, Kyoto 603-8555, Japan
| |
Collapse
|
37
|
Wang A, Zhu J, Li J, Du W, Zhang Y, Cai T, Liu T, Fu Y, Zeng Y, Liu Z, Huang JA. Downregulation of KIAA1199 by miR-486-5p suppresses tumorigenesis in lung cancer. Cancer Med 2020; 9:5570-5586. [PMID: 32519472 PMCID: PMC7402811 DOI: 10.1002/cam4.3210] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2020] [Revised: 05/13/2020] [Accepted: 05/22/2020] [Indexed: 12/11/2022] Open
Abstract
Lung cancer is the primary cause of death among cancer patients in China, among which nonsmall cell lung cancer (NSCLC) makes up the great majority. Hence, it is imperative to identify the biomarkers and mechanisms involved in NSCLC oncogenesis. Our present research found that KIAA1199 expression was significantly increased in NSCLC and closely related to cell proliferation, motility, and poor prognosis. We demonstrated that knockdown of KIAA1199 reduced NSCLC cell growth and motility in vitro whereas overexpression of KIAA1199 had the opposite effect. Inhibition of KIAA1199 significantly suppressed tumor growth in mouse NSCLC xenograft models. Mechanistically, as an epidermal growth factor receptor (EGFR)‐binding protein, KIAA1199 promotes EGFR signaling and regulates EGFR‐dependent Src, Erk, and Akt phosphorylation, as well as downstream kinases in the EGF‐mediated EMT pathway. We demonstrated that KIAA1199 can function as a direct binding target for miR‐486‐5p and that miR‐486‐5p overexpression can attenuate proliferation and migration of NSCLC cells via regulating the EGFR signaling pathways. To conclude, our results defined KIAA1199 as an oncogenic protein that promotes cancer cell proliferation and migration by regulating EGF‐mediated signaling pathways. This study provided new insight into NSCLC oncogenesis, which could lead to the development of innovative therapeutic plans for NSCLC.
Collapse
Affiliation(s)
- Anqi Wang
- Department of Respiratory Medicine, the First Affiliated Hospital of Soochow University, Suzhou, China.,Suzhou Key Laboratory for Respiratory Diseases, Suzhou, China
| | - Jianjie Zhu
- Department of Respiratory Medicine, the First Affiliated Hospital of Soochow University, Suzhou, China.,Suzhou Key Laboratory for Respiratory Diseases, Suzhou, China.,Institute of Respiratory Diseases, Soochow University, Suzhou, China
| | - Juan Li
- Department of Respiratory Medicine, the First Affiliated Hospital of Soochow University, Suzhou, China.,Suzhou Key Laboratory for Respiratory Diseases, Suzhou, China
| | - Wenwen Du
- Department of Respiratory Medicine, the First Affiliated Hospital of Soochow University, Suzhou, China.,Suzhou Key Laboratory for Respiratory Diseases, Suzhou, China
| | - Yang Zhang
- Department of Respiratory Medicine, the First Affiliated Hospital of Soochow University, Suzhou, China.,Suzhou Key Laboratory for Respiratory Diseases, Suzhou, China
| | - Tingting Cai
- Department of Respiratory Medicine, the First Affiliated Hospital of Soochow University, Suzhou, China.,Suzhou Key Laboratory for Respiratory Diseases, Suzhou, China
| | - Ting Liu
- Department of Respiratory Medicine, the First Affiliated Hospital of Soochow University, Suzhou, China.,Suzhou Key Laboratory for Respiratory Diseases, Suzhou, China
| | - Yulong Fu
- Department of Respiratory Medicine, the First Affiliated Hospital of Soochow University, Suzhou, China.,Suzhou Key Laboratory for Respiratory Diseases, Suzhou, China
| | - Yuanyuan Zeng
- Department of Respiratory Medicine, the First Affiliated Hospital of Soochow University, Suzhou, China.,Suzhou Key Laboratory for Respiratory Diseases, Suzhou, China.,Institute of Respiratory Diseases, Soochow University, Suzhou, China
| | - Zeyi Liu
- Department of Respiratory Medicine, the First Affiliated Hospital of Soochow University, Suzhou, China.,Suzhou Key Laboratory for Respiratory Diseases, Suzhou, China.,Institute of Respiratory Diseases, Soochow University, Suzhou, China
| | - Jian-An Huang
- Department of Respiratory Medicine, the First Affiliated Hospital of Soochow University, Suzhou, China.,Suzhou Key Laboratory for Respiratory Diseases, Suzhou, China.,Institute of Respiratory Diseases, Soochow University, Suzhou, China
| |
Collapse
|
38
|
Induction of CEMIP in Chondrocytes by Inflammatory Cytokines: Underlying Mechanisms and Potential Involvement in Osteoarthritis. Int J Mol Sci 2020; 21:ijms21093140. [PMID: 32365591 PMCID: PMC7247684 DOI: 10.3390/ijms21093140] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Revised: 04/24/2020] [Accepted: 04/27/2020] [Indexed: 12/23/2022] Open
Abstract
In patients with osteoarthritis (OA), there is a decrease in both the concentration and molecular size of hyaluronan (HA) in the synovial fluid and cartilage. Cell migration-inducing hyaluronidase 1 (CEMIP), also known as hyaluronan (HA)-binding protein involved in HA depolymerization (HYBID), was recently reported as an HA depolymerization-related molecule expressed in the cartilage of patients with OA. However, the underlying mechanism of CEMIP regulation is not well understood. We found that CEMIP expression was transiently increased by interleukine-1β (IL-1β) stimulation in chondrocytic cells. We also observed that ERK activation and NF-κB nuclear translocation were involved in the induction of CEMIP by IL-1β. In addition, both administration of HA and mechanical strain attenuated the CEMIP induction in IL-1β-stimulated chondrocytes. In conclusion, we clarified the regulatory mechanism of CEMIP in chondrocytes by inflammatory cytokines and suggested the potential involvement in osteoarthritis development.
Collapse
|
39
|
Rosa N, Sneyers F, Parys JB, Bultynck G. Type 3 IP 3 receptors: The chameleon in cancer. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2020; 351:101-148. [PMID: 32247578 DOI: 10.1016/bs.ircmb.2020.02.003] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Inositol 1,4,5-trisphosphate (IP3) receptors (IP3Rs), intracellular calcium (Ca2+) release channels, fulfill key functions in cell death and survival processes, whose dysregulation contributes to oncogenesis. This is essentially due to the presence of IP3Rs in microdomains of the endoplasmic reticulum (ER) in close proximity to the mitochondria. As such, IP3Rs enable efficient Ca2+ transfers from the ER to the mitochondria, thus regulating metabolism and cell fate. This review focuses on one of the three IP3R isoforms, the type 3 IP3R (IP3R3), which is linked to proapoptotic ER-mitochondrial Ca2+ transfers. Alterations in IP3R3 expression have been highlighted in numerous cancer types, leading to dysregulations of Ca2+ signaling and cellular functions. However, the outcome of IP3R3-mediated Ca2+ transfers for mitochondrial function is complex with opposing effects on oncogenesis. IP3R3 can either suppress cancer by promoting cell death and cellular senescence or support cancer by driving metabolism, anabolic processes, cell cycle progression, proliferation and invasion. The aim of this review is to provide an overview of IP3R3 dysregulations in cancer and describe how such dysregulations alter critical cellular processes such as proliferation or cell death and survival. Here, we pose that the IP3R3 isoform is not only linked to proapoptotic ER-mitochondrial Ca2+ transfers but might also be involved in prosurvival signaling.
Collapse
Affiliation(s)
- Nicolas Rosa
- KU Leuven, Laboratory of Molecular and Cellular Signaling, Department of Cellular and Molecular Medicine and Leuven Kanker Instituut (LKI), Leuven, Belgium
| | - Flore Sneyers
- KU Leuven, Laboratory of Molecular and Cellular Signaling, Department of Cellular and Molecular Medicine and Leuven Kanker Instituut (LKI), Leuven, Belgium
| | - Jan B Parys
- KU Leuven, Laboratory of Molecular and Cellular Signaling, Department of Cellular and Molecular Medicine and Leuven Kanker Instituut (LKI), Leuven, Belgium
| | - Geert Bultynck
- KU Leuven, Laboratory of Molecular and Cellular Signaling, Department of Cellular and Molecular Medicine and Leuven Kanker Instituut (LKI), Leuven, Belgium.
| |
Collapse
|
40
|
Wang Q, Liu F, Wang L, Xie C, Wu P, Du S, Zhou S, Sun Z, Liu Q, Yu L, Liu B, Li R. Enhanced and Prolonged Antitumor Effect of Salinomycin-Loaded Gelatinase-Responsive Nanoparticles via Targeted Drug Delivery and Inhibition of Cervical Cancer Stem Cells. Int J Nanomedicine 2020; 15:1283-1295. [PMID: 32161458 PMCID: PMC7049776 DOI: 10.2147/ijn.s234679] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2019] [Accepted: 01/26/2020] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Cervical cancer stem cells (CCSCs) represent a subpopulation of tumor cells that possess self-renewal capacity and numerous intrinsic mechanisms of resistance to conventional chemotherapy and radiotherapy. These cells play a crucial role in relapse and metastasis of cervical cancer. Therefore, eradication of CCSCs is the primary objective in cervical cancer therapy. Salinomycin (Sal) is an agent used for the elimination of cancer stem cells (CSCs); however, the occurrence of several side effects hinders its application. Nanoscale drug-delivery systems offer great promise for the diagnosis and treatment of tumors. These systems can be used to reduce the side effects of Sal and improve clinical benefit. METHODS Sal-loaded polyethylene glycol-peptide-polycaprolactone nanoparticles (Sal NPs) were fabricated under mild and non-toxic conditions. The real-time biodistribution of Sal NPs was investigated through non-invasive near-infrared fluorescent imaging. The efficacy of tumor growth inhibition by Sal NPs was evaluated using tumor xenografts in nude mice. Flow cytometry, immunohistochemistry, and Western blotting were used to detect the apoptosis of CSCs after treatment with Sal NPs. Immunohistochemistry and Western blotting were used to examine epithelial-mesenchymal transition (epithelial interstitial transformation) signal-related molecules. RESULTS Sal NPs exhibited antitumor efficacy against cervical cancers by inducing apoptosis of CCSCs and inhibiting the epithelial-mesenchymal transition pathway. Besides, tumor pieces resected from Sal NP-treated mice showed decreased reseeding ability and growth speed, further demonstrating the significant inhibitory ability of Sal NPs against CSCs. Moreover, owing to targeted delivery based on the gelatinase-responsive strategy, Sal NPs was more effective and tolerable than free Sal. CONCLUSION To the best of our knowledge, this is the first study to show that CCSC-targeted Sal NPs provide a potential approach to selectively target and efficiently eradicate CCSCs. This renders them a promising strategy to improve the therapeutic effect against cervical cancer.
Collapse
Affiliation(s)
- Qin Wang
- The Comprehensive Cancer Centre of Drum Tower Hospital, Medical School of Nanjing University, Clinical Cancer Institute of Nanjing University, Nanjing210008, People’s Republic of China
| | - Fangcen Liu
- The Comprehensive Cancer Centre, Nanjing Drum Tower Hospital, Clinical College of Nanjing Medical University, Nanjing, People’s Republic of China
| | - Lifeng Wang
- The Comprehensive Cancer Centre of Drum Tower Hospital, Medical School of Nanjing University, Clinical Cancer Institute of Nanjing University, Nanjing210008, People’s Republic of China
| | - Chen Xie
- Key Laboratory for Organic Electronics and Information Displays, Institute of Advanced Materials (IAM), Jiangsu National Synergetic Innovation Center for Advanced Materials (SICAM), Nanjing University of Posts & Telecommunications, Nanjing210023, People’s Republic of China
| | - Puyuan Wu
- The Comprehensive Cancer Centre of Drum Tower Hospital, Medical School of Nanjing University, Clinical Cancer Institute of Nanjing University, Nanjing210008, People’s Republic of China
| | - Shiyao Du
- The Comprehensive Cancer Centre of Drum Tower Hospital, Medical School of Nanjing University, Clinical Cancer Institute of Nanjing University, Nanjing210008, People’s Republic of China
| | - Shujuan Zhou
- The Comprehensive Cancer Centre of Drum Tower Hospital, Medical School of Nanjing University, Clinical Cancer Institute of Nanjing University, Nanjing210008, People’s Republic of China
| | - Zhichen Sun
- The Comprehensive Cancer Centre of Drum Tower Hospital, Medical School of Nanjing University, Clinical Cancer Institute of Nanjing University, Nanjing210008, People’s Republic of China
| | - Qin Liu
- The Comprehensive Cancer Centre of Drum Tower Hospital, Medical School of Nanjing University, Clinical Cancer Institute of Nanjing University, Nanjing210008, People’s Republic of China
| | - Lixia Yu
- The Comprehensive Cancer Centre of Drum Tower Hospital, Medical School of Nanjing University, Clinical Cancer Institute of Nanjing University, Nanjing210008, People’s Republic of China
| | - Baorui Liu
- The Comprehensive Cancer Centre of Drum Tower Hospital, Medical School of Nanjing University, Clinical Cancer Institute of Nanjing University, Nanjing210008, People’s Republic of China
| | - Rutian Li
- The Comprehensive Cancer Centre of Drum Tower Hospital, Medical School of Nanjing University, Clinical Cancer Institute of Nanjing University, Nanjing210008, People’s Republic of China
| |
Collapse
|
41
|
Su J, Su B, Xia H, Liu F, Zhao X, Li J, Zhang J, Shi Y, Zeng Y, Zeng X, Ling H, Wu Y, Su Q. RORα Suppresses Epithelial-to-Mesenchymal Transition and Invasion in Human Gastric Cancer Cells via the Wnt/β-Catenin Pathway. Front Oncol 2019; 9:1344. [PMID: 31867273 PMCID: PMC6909819 DOI: 10.3389/fonc.2019.01344] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2019] [Accepted: 11/15/2019] [Indexed: 12/24/2022] Open
Abstract
Retinoid-related orphan receptor alpha (RORα) is involved in tumor development. However, the mechanisms underlying RORα inhibiting epithelial-to-mesenchymal transition (EMT) and invasion are poorly understood in gastric cancer (GC). This study revealed that the decreased expression of RORα is associated with GC development, progression, and prognosis. RORα suppressed cell proliferation, EMT, and invasion in GC cells through inhibition of the Wnt/β-catenin pathway. RORα overexpression resulted in the decreased Wnt1 expression and the increased RORα interaction with β-catenin, which could lead to the decreased intranuclear β-catenin and p-β-catenin levels, concomitant with downregulated T-cell factor-4 (TCF-4) expression and the promoter activity of c-Myc. The inhibition of Wnt/β-catenin pathway was coupled with the reduced expression of Axin, c-Myc, and c-Jun. RORα downregulated vimentin and Snail and upregulated E-cadherin protein levels in vitro and in vivo. Inversely, knockdown of RORα attenuated its inhibitory effects on Wnt/β-catenin pathway and its downstream gene expression, facilitating cell proliferation, EMT, migration, and invasion in GC cells. Therefore, RORα could play a crucial role in repressing GC cell proliferation, EMT, and invasion via downregulating Wnt/β-catenin pathway.
Collapse
Affiliation(s)
- Jian Su
- Hunan Province Key Laboratory of Cancer Cellular and Molecular Pathology, Cancer Research Institute, University of South China, Hengyang, China.,Department of Pathology, Second Affiliated Hospital, University of South China, Hengyang, China
| | - Bo Su
- Hunan Province Key Laboratory of Cancer Cellular and Molecular Pathology, Cancer Research Institute, University of South China, Hengyang, China.,Key Laboratory for Pharmacoproteomics of Hunan Provincial University, Institute of Pharmacy and Pharmacology, University of South China, Hengyang, China
| | - Hong Xia
- Hunan Province Key Laboratory of Cancer Cellular and Molecular Pathology, Cancer Research Institute, University of South China, Hengyang, China.,Center for Gastric Cancer Research of Hunan Province, First Affiliated Hospital, University of South China, Hengyang, China
| | - Fang Liu
- Hunan Province Key Laboratory of Cancer Cellular and Molecular Pathology, Cancer Research Institute, University of South China, Hengyang, China.,Center for Gastric Cancer Research of Hunan Province, First Affiliated Hospital, University of South China, Hengyang, China
| | - XiaoHong Zhao
- Hunan Province Key Laboratory of Cancer Cellular and Molecular Pathology, Cancer Research Institute, University of South China, Hengyang, China.,Department of Gynaecology, Hainan Maternal and Child Health Hospital, Haikou, China
| | - Juan Li
- Hunan Province Key Laboratory of Cancer Cellular and Molecular Pathology, Cancer Research Institute, University of South China, Hengyang, China.,Department of Gastroenterology, Loudi Center Hospital, Loudi, China
| | - JiZhen Zhang
- Hunan Province Key Laboratory of Cancer Cellular and Molecular Pathology, Cancer Research Institute, University of South China, Hengyang, China.,Department of Pathology, Affiliated Hospital, Jinggangshan University, Ji'an, China
| | - Ying Shi
- Hunan Province Key Laboratory of Cancer Cellular and Molecular Pathology, Cancer Research Institute, University of South China, Hengyang, China.,Department of Pathology and Pathophysiology, Xiamen Medical College, Xiamen, China
| | - Ying Zeng
- Hunan Province Key Laboratory of Cancer Cellular and Molecular Pathology, Cancer Research Institute, University of South China, Hengyang, China.,Center for Gastric Cancer Research of Hunan Province, First Affiliated Hospital, University of South China, Hengyang, China
| | - Xi Zeng
- Hunan Province Key Laboratory of Cancer Cellular and Molecular Pathology, Cancer Research Institute, University of South China, Hengyang, China.,Center for Gastric Cancer Research of Hunan Province, First Affiliated Hospital, University of South China, Hengyang, China
| | - Hui Ling
- Hunan Province Key Laboratory of Cancer Cellular and Molecular Pathology, Cancer Research Institute, University of South China, Hengyang, China.,Center for Gastric Cancer Research of Hunan Province, First Affiliated Hospital, University of South China, Hengyang, China
| | - YouHua Wu
- Center for Gastric Cancer Research of Hunan Province, First Affiliated Hospital, University of South China, Hengyang, China
| | - Qi Su
- Hunan Province Key Laboratory of Cancer Cellular and Molecular Pathology, Cancer Research Institute, University of South China, Hengyang, China.,Center for Gastric Cancer Research of Hunan Province, First Affiliated Hospital, University of South China, Hengyang, China
| |
Collapse
|
42
|
Wei Y, Zhang F, Zhang T, Zhang Y, Chen H, Wang F, Li Y. LDLRAD2 overexpression predicts poor prognosis and promotes metastasis by activating Wnt/β-catenin/EMT signaling cascade in gastric cancer. Aging (Albany NY) 2019; 11:8951-8968. [PMID: 31649207 PMCID: PMC6834412 DOI: 10.18632/aging.102359] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2019] [Accepted: 10/05/2019] [Indexed: 12/15/2022]
Abstract
The therapeutic strategies for advanced gastric cancer (GC) remain unsatisfying and limited. Therefore, it is still imperative to fully elucidate the mechanisms underlying GC aggressive progression. The prognostic value and biological functions of low density lipoprotein receptor class A domain containing protein 2 (LDLRAD2) in GC have never been studied yet. We found that LDLRAD2 expression was significantly upregulated in GC and closely correlated with poor prognosis in GC patients. Functionally, LDLRAD2 promoted epithelial-mesenchymal transition, migration and invasion, and metastasis of GC cells. Mechanistically, LDLRAD2 interacted with and inhibited Axin1 from binding to cytoplasmic β-catenin, which facilitated the nuclear translocation of β-catenin, thereby activating Wnt/β-catenin pathway. Inhibition of β-catenin activity markedly abolished LDLRAD2-induced migration, invasion and metastasis. Together, these results suggested that LDLRAD2 contributed to invasion and metastasis of GC through activating Wnt/β-catenin pathway. LDLRAD2/ Wnt/β-catenin axis may be a potential therapeutic target for GC treatment.
Collapse
Affiliation(s)
- Yucai Wei
- Department of Surgical Oncology, Lanzhou University Second Hospital, Lanzhou 730000, China.,Key Laboratory of Digestive System Tumors of Gansu Province, Lanzhou University Second Hospital, Lanzhou 730000, China
| | - Fan Zhang
- Key Laboratory of Digestive System Tumors of Gansu Province, Lanzhou University Second Hospital, Lanzhou 730000, China
| | - Tong Zhang
- Department of Hepatic Surgery, Liver Transplant Center, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou 510630, China
| | - Yating Zhang
- Key Laboratory of Digestive System Tumors of Gansu Province, Lanzhou University Second Hospital, Lanzhou 730000, China
| | - Hao Chen
- Department of Surgical Oncology, Lanzhou University Second Hospital, Lanzhou 730000, China.,Key Laboratory of Digestive System Tumors of Gansu Province, Lanzhou University Second Hospital, Lanzhou 730000, China
| | - Furong Wang
- Key Laboratory of Digestive System Tumors of Gansu Province, Lanzhou University Second Hospital, Lanzhou 730000, China.,Department of Pathology, Lanzhou University Second Hospital, Lanzhou 730000, China
| | - Yumin Li
- Department of Surgical Oncology, Lanzhou University Second Hospital, Lanzhou 730000, China.,Key Laboratory of Digestive System Tumors of Gansu Province, Lanzhou University Second Hospital, Lanzhou 730000, China
| |
Collapse
|
43
|
Jiao X, Ye J, Wang X, Yin X, Zhang G, Cheng X. KIAA1199, a Target of MicoRNA-486-5p, Promotes Papillary Thyroid Cancer Invasion by Influencing Epithelial-Mesenchymal Transition (EMT). Med Sci Monit 2019; 25:6788-6796. [PMID: 31501407 PMCID: PMC6752488 DOI: 10.12659/msm.918682] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Background KIAA1199 has been reported to be associated with malignant progression and poor clinical outcomes in various human malignancies. However, its clinical role and molecular function remain unknown in papillary thyroid cancer (PTC). Material/Methods The Cancer Genome Atlas (TCGA) was used to investigate the expression profiles of KIAA1199 and miR-486-5p in PTC. Immunohistochemistry was used to validate the protein expression of KIAA1199 in PTC. The Weighted Gene Co-expression Network Analysis (WGCNA) and Gene Set Enrichment Analysis (GSEA) were used to explore the potential pathway underling KIAA1199 in PTC. In vitro and in vivo experiments were performed to investigate the biological role of KIAA1199 in PTC progression. Luciferase reporter assays and Western blot analysis were performed to determine whether KIAA1199 is a downstream target of miR-486-5p. Results We found that KIAA1199 was aberrantly elevated in PTC tissues compared with normal tissues, and upregulation of KIAA1199 was positively correlated with more advanced clinical variables. Additionally, bioinformatic analysis indicated that KIAA1199 was involved in cell migration and invasion. KIAA1199 silencing inhibited the invasive ability of PTC cells by affecting epithelial-mesenchymal transition (EMT) in vitro and in vivo. Furthermore, miR-486-5p was identified as an upstream microRNA that directly targets the 3′-UTR region of KIAA1199. Conclusions The miR-486-5p/KIAA1199/EMT axis might play a critical role in PTC invasion and metastasis and offers a potential therapeutic strategy for PTC.
Collapse
Affiliation(s)
- Xuehua Jiao
- Department of Endocrinology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China (mainland).,Department of Endocrinology, The First People's Hospital of Wujiang District, Suzhou, Jiangsu, China (mainland)
| | - Jiandong Ye
- Department of General Surgery, The First People's Hospital of Wujiang District, Suzhou, Jiangsu, China (mainland)
| | - Xiujie Wang
- Department of Endocrinology, The First People's Hospital of Wujiang District, Suzhou, Jiangsu, China (mainland)
| | - Xueyan Yin
- Department of Endocrinology, The First People's Hospital of Wujiang District, Suzhou, Jiangsu, China (mainland)
| | - Guodong Zhang
- Department of Endocrinology, Suzhou Yongding Hospital, Suzhou, Jiangsu, China (mainland)
| | - Xingbo Cheng
- Department of Endocrinology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China (mainland)
| |
Collapse
|
44
|
Li Q, Zhou X, Fang Z, Zhou H. Knockdown of KLK12 inhibits viability and induces apoptosis in human colorectal cancer HT-29 cell line. Int J Mol Med 2019; 44:1667-1676. [PMID: 31485623 PMCID: PMC6777684 DOI: 10.3892/ijmm.2019.4327] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2019] [Accepted: 07/26/2019] [Indexed: 01/10/2023] Open
Abstract
Kallikrein-related peptidase 12 (KLK12) is overexpressed in cancer tissues including gastric, breast and prostate cancer. However, the role of KLK12 in colorectal cancer is not fully understood. In the present study, the level of KLK12 was determined by performing reverse transcription-polymerase chain reaction (RT-qPCR) in colorectal cancer tissues and cell lines. Lipofectamine® 2000 was used to transfect HT-29 cells to overexpress and knockdown KLK12. Cell viability, migration, invasion and apoptosis were detected by MTT, wound healing, Transwell and flow cytometry assays, respectively. The mRNA and protein expression levels of EMT-associated proteins, apoptosis-associated proteins, phosphorylated adenosine monophosphate-activated protein kinase (p-AMPK) and phosphorylated mammalian target of rapamycin (p-mTOR) were determined by RT-qPCR and western blot analysis. It was identified that the KLK12 mRNA levels were increased significantly in colorectal cancer tissues and cell lines. KLK12 small interfering RNA inhibited cell viability, migration and invasion. Furthermore, epithelial-mesenchymal transition (EMT)-associated proteins were altered by siKLK12. Cell apoptosis was induced by KLK12 downregulation, which was demonstrated by the changes in apoptosis-associated proteins; however, KLK12 overexpression produced the opposite effect. SiKLK12 enhanced the expression of p-AMPK and suppressed the expression of p-mTOR, while KLK12 overexpression had the opposite effect. Promotion of KLK12 overexpression-induced cell viability was reversed by 5-aminoimidazole-4-carboxamide ribonucleotide, an activator of the AMPK signaling pathway, and rapamycin, a specific inhibitor of the mTOR signaling pathway. Taken together, the results of the present study indicated that KLK12 was overexpressed in colorectal cancer and may regulate cell behavior, potentially via the AMPK and mTOR pathways.
Collapse
Affiliation(s)
- Qianyuan Li
- Department of Anorectal Surgery, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310006, P.R. China
| | - Xiukou Zhou
- Department of Anorectal Surgery, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310006, P.R. China
| | - Zhengyu Fang
- Department of Anorectal Surgery, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310006, P.R. China
| | - Huamiao Zhou
- Department of Oncology, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310006, P.R. China
| |
Collapse
|
45
|
Li Q, Zhou X, Fang Z, Pan Z. Effect of STC2 gene silencing on colorectal cancer cells. Mol Med Rep 2019; 20:977-984. [PMID: 31173256 PMCID: PMC6625197 DOI: 10.3892/mmr.2019.10332] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2018] [Accepted: 03/27/2019] [Indexed: 02/06/2023] Open
Abstract
Stanniocalcin 2 (STC2), a secretory glycoprotein hormone, regulates many biological processes including cell proliferation, apoptosis, tumorigenesis and atherosclerosis. However, the effect of STC2 on proliferation, migration and epithelial‑mesenchymal transition (EMT) progression in human colorectal cancer (CRC) cells remains poorly understood. The expression level of STC2 was determined by quantitative real‑time polymerase chain reaction (qPCR) and western blot analysis. Cell Counting Kit‑8 (CCK‑8) was used to detect the viability of SW480 cells. The invasion and migration of cells were identified by wound healing and Transwell assays. The mRNA and protein expression levels of β‑catenin, matrix metalloproteinase (MMP)‑2, MMP‑9, E‑cadherin and vimentin were assessed by qPCR and western blot analysis. In the present study, it was demonstrated that STC2 was highly expressed in the CRC cell lines. After silencing of STC2, the cell viability, migration and invasion were significantly reduced. Silencing of STC2 in the CRC Sw480 cells increased the expression of E‑cadherin and decreased the expression of vimentin, MMP‑2 and MMP‑9, compared to those in the normal and empty vector group. Furthermore, the expression of β‑catenin in the STC2 gene silenced group was suppressed, and the expression of β‑catenin was reversed by Wnt activator, SB216763. These results demonstrated that STC2 participates in the development and progression of CRC by promoting CRC cell proliferation, survival and migration and activating the Wnt/β‑catenin signaling pathway.
Collapse
Affiliation(s)
- Qianyuan Li
- Department of Anorectal Surgery, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310006, P.R. China
| | - Xiukou Zhou
- Department of Anorectal Surgery, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310006, P.R. China
| | - Zhengyu Fang
- Department of Anorectal Surgery, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310006, P.R. China
| | - Zhiyun Pan
- Department of Anorectal Surgery, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310006, P.R. China
| |
Collapse
|
46
|
Ding QH, Qi YY, Li XM, Chen WP, Wang XH, Ji XW. Knockdown of KIAA1199 suppresses IL-1β-induced cartilage degradation and inflammatory responses in human chondrocytes through the Wnt/β-catenin signalling pathway. Int Immunopharmacol 2019; 73:203-211. [PMID: 31103876 DOI: 10.1016/j.intimp.2019.05.018] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2019] [Revised: 04/27/2019] [Accepted: 05/10/2019] [Indexed: 12/29/2022]
Abstract
The overproduction of proteolytic enzymes and dysregulation of extracellular matrix (ECM) metabolism have been shown to accelerate the degradation process of articular cartilage. The purpose of this study was to investigate the role of KIAA1199 and its association with the pathophysiology of osteoarthritis (OA). We found that the expression of KIAA1199 was significantly upregulated in OA cartilage compared with normal tissues. Serum levels of KIAA1199 were higher in OA patients than in non-OA patients. Furthermore, knockdown of KIAA1199 inhibited interleukin-1 beta (IL-1β)-induced ECM metabolic imbalance by regulating the expression of A disintegrin-like and metalloprotease (reprolysin type) with thrombospondin type 1 motif, 5; matrix metallopeptidase-13; aggrecan; and COL2A1. In addition, silencing of KIAA1199 significantly decreased the expression of inflammatory mediators such as prostaglandin E2, IL-6, and TNF-α. Mechanistic analyses further revealed that IL-1β-induced activation of the Wnt/β-catenin pathway was suppressed during KIAA1199 knockdown. Moreover, KIAA1199 expression was also upregulated in an in vivo rat OA model. Together, these results increase our understanding of the emerging role of KIAA1199 in the process of OA degeneration, and may lead to a novel molecular target to prevent cartilage degradation.
Collapse
Affiliation(s)
- Qian-Hai Ding
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Jie Fang Road 88#, 310009 Hangzhou, People's Republic of China
| | - Yi-Ying Qi
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Jie Fang Road 88#, 310009 Hangzhou, People's Republic of China
| | - Xiu-Mao Li
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Jie Fang Road 88#, 310009 Hangzhou, People's Republic of China
| | - Wei-Ping Chen
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Jie Fang Road 88#, 310009 Hangzhou, People's Republic of China
| | - Xiang-Hua Wang
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Jie Fang Road 88#, 310009 Hangzhou, People's Republic of China
| | - Xiao-Wei Ji
- Department of Gastroenterology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Jie Fang Road 88#, 310009 Hangzhou, People's Republic of China.
| |
Collapse
|
47
|
Banach A, Jiang YP, Roth E, Kuscu C, Cao J, Lin RZ. CEMIP upregulates BiP to promote breast cancer cell survival in hypoxia. Oncotarget 2019; 10:4307-4320. [PMID: 31303964 PMCID: PMC6611512 DOI: 10.18632/oncotarget.27036] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
Cell migration-inducing protein (CEMIP) and binding immunoglobulin protein (BiP) are upregulated in human cancers, where they drive cancer progression and metastasis. It has been shown that CEMIP resides in the endoplasmic reticulum (ER) where it interacts with BiP to induce cell migration, but the relationship between the two proteins was previously unknown. Here we show that CEMIP mediates activation of the BiP promoter and upregulates BiP transcript and protein levels in breast cancer cell lines. Moreover, CEMIP overexpression confers protective adaptations to cancer cells under hypoxic conditions, by decreasing apoptosis, activating autophagy, and increasing glucose uptake, to facilitate tumor growth. We demonstrate that BiP signals downstream of CEMIP, modulating cellular resistance to hypoxia. Reducing BiP in CEMIP-expressing cells sensitized cells to hypoxia treatment, decreased glucose uptake, and resulted in tumor regression in vivo. Our study provides insights into the link between CEMIP and BiP expression and the pro-survival role they play in hypoxia. Better understanding of the mechanisms behind cancer cell adaptations to harsh tumor environments could lead to development of improved cancer treatments.
Collapse
Affiliation(s)
- Anna Banach
- Molecular and Cellular Biology Program, Stony Brook University, Stony Brook, NY, USA
| | - Ya-Ping Jiang
- Department of Physiology and Biophysics, Institute of Molecular Cardiology, Stony Brook University, Stony Brook, NY, USA
| | - Eric Roth
- Medical Scientist Training Program, Stony Brook University, Stony Brook, NY, USA
| | - Cem Kuscu
- Transplant Research Institute, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Jian Cao
- Division of Cancer Prevention, Department of Medicine, Stony Brook University, Stony Brook, NY, USA
| | - Richard Z Lin
- Department of Physiology and Biophysics, Institute of Molecular Cardiology, Stony Brook University, Stony Brook, NY, USA.,Medical Service, Northport Veterans Affairs Medical Center, Northport, NY, USA
| |
Collapse
|
48
|
Abdeyrim A, Cheng X, Lian M, Tan Y. miR‑490‑5p regulates the proliferation, migration, invasion and epithelial‑mesenchymal transition of pharyngolaryngeal cancer cells by targeting mitogen‑activated protein kinase kinasekinase 9. Int J Mol Med 2019; 44:240-252. [PMID: 31115491 PMCID: PMC6559303 DOI: 10.3892/ijmm.2019.4196] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2018] [Accepted: 05/08/2019] [Indexed: 11/05/2022] Open
Abstract
MicroRNA (miRNA/miR) has been identified to be a promising tool in treating pharyngolaryngeal cancer. The present study aimed to investigate the role of miR‑490‑5p in the regulation of proliferation, migration, invasion and epithelial‑mesenchymal transition (EMT) of pharyngolaryngeal cancer cells. The data of miR‑490‑5p expression levels of 45 cases were obtained from the People's Hospital of Xinjiang Uygur Autonomous Region, and the prediction of the target of miR‑490‑5p was conducted by bioinformatics and verified using a luciferase assay. Cell viability was determined by cell counting kit‑8. Migration and invasion rates were measured by wound healing test and Transwell apparatus, respectively. Colony formation rate was measured by plate colony formation assay. mRNA and protein levels were determined by quantitative polymerase chain reaction and western blotting, respectively. miR‑490‑5p expression was significantly depressed in primary pharyngolaryngeal cancer tissues and cell lines, leading to an unfavorable prognosis. Evidently, miR‑490‑5p overexpression decreased the cell viabilities of BICR 18 and FaDu cells. Mechanically, miR‑490‑5p could target mitogen‑activated protein kinase kinasekinase 9 (MAP3K9). The overexpression of MAP3K9 could promote cell viability, migration and invasion rates, EMT process and ability of cloning, miR‑490‑5p could target MAP3K9 and further modulate the proliferation, migration, invasion and EMT of pharyngolaryngeal cancer cells. The results of the present study provide a novel entry point to the treatment of pharyngolaryngeal cancer.
Collapse
Affiliation(s)
- Arikin Abdeyrim
- Department of Otorhinolaryngology Head and Neck Surgery, The People's Hospital of Xinjiang Uygur Autonomous Region, Ürümqi, Xinjiang 830001, P.R. China
| | - Xiuqin Cheng
- Department of Otorhinolaryngology Head and Neck Surgery, The People's Hospital of Xinjiang Uygur Autonomous Region, Ürümqi, Xinjiang 830001, P.R. China
| | - Meng Lian
- Department of Otorhinolaryngology Head and Neck Surgery, Beijing Tongren Hospital, Capital Medical University, Beijing 100005, P.R. China
| | - Yuanyouan Tan
- Department of Otorhinolaryngology Head and Neck Surgery, The People's Hospital of Xinjiang Uygur Autonomous Region, Ürümqi, Xinjiang 830001, P.R. China
| |
Collapse
|
49
|
Liu Y, Li Y, Zhao Y, Liu Y, Fan L, Jia N, Zhao Q. ILF3 promotes gastric cancer proliferation and may be used as a prognostic marker. Mol Med Rep 2019; 20:125-134. [PMID: 31115508 PMCID: PMC6579973 DOI: 10.3892/mmr.2019.10229] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2018] [Accepted: 04/25/2019] [Indexed: 12/22/2022] Open
Abstract
Interleukin enhancer-binding factor 3 (ILF3) may function as a transcriptional coactivator and has been reported to be involved in tumor proliferation and metastasis; however, its role and clinical value in gastric cancer (GC) remains unclear. To understand the value of ILF3 in GC, a total of 80 matched samples selected from GC tissues and the adjacent mucosa were used to evaluate the expression of ILF3 and its association with clinical characteristics. Furthermore, its biological functions and mechanisms were investigated using SGC-7901 and BGC823 cell lines. Immunohistochemistry demonstrated that the positive expression rates of ILF3 in GC tissue were higher compared with those in adjacent mucosa (P<0.05). Significantly overexpressed ILF3 was detected in BGC823 and SGC7901 cells, and the MTT results demonstrated decreased cell activity after ILF3 expression was inhibited. The proportions of cells in the G0/G1 phase increased, while the number of cells in the G2/M phase decreased, and the expression of the genes associated with proliferation varied following inhibition of ILF3 (P<0.05). Positive expression of ILF3 was associated with a poor prognosis for patients with GC, and was an independent risk factor for GC (P<0.05). In conclusion, ILF3 is involved in the deterioration of GC by promoting proliferation of GC cells, and ILF3 protein detection may assist in the prediction of the prognosis of patients with GC.
Collapse
Affiliation(s)
- Yü Liu
- Department of General Surgery, Hebei Medical University Fourth Affiliated Hospital and Hebei Provincial Tumor Hospital, Shijiazhuang, Hebei 050011, P.R. China
| | - Yong Li
- Department of General Surgery, Hebei Medical University Fourth Affiliated Hospital and Hebei Provincial Tumor Hospital, Shijiazhuang, Hebei 050011, P.R. China
| | - Yijie Zhao
- Department of General Surgery, Hebei Medical University Fourth Affiliated Hospital and Hebei Provincial Tumor Hospital, Shijiazhuang, Hebei 050011, P.R. China
| | - Yang Liu
- Department of General Surgery, Hebei Medical University Fourth Affiliated Hospital and Hebei Provincial Tumor Hospital, Shijiazhuang, Hebei 050011, P.R. China
| | - Liqiao Fan
- Department of General Surgery, Hebei Medical University Fourth Affiliated Hospital and Hebei Provincial Tumor Hospital, Shijiazhuang, Hebei 050011, P.R. China
| | - Nan Jia
- Department of General Surgery, Hebei Medical University Fourth Affiliated Hospital and Hebei Provincial Tumor Hospital, Shijiazhuang, Hebei 050011, P.R. China
| | - Qun Zhao
- Department of General Surgery, Hebei Medical University Fourth Affiliated Hospital and Hebei Provincial Tumor Hospital, Shijiazhuang, Hebei 050011, P.R. China
| |
Collapse
|
50
|
Tammi MI, Oikari S, Pasonen-Seppänen S, Rilla K, Auvinen P, Tammi RH. Activated hyaluronan metabolism in the tumor matrix — Causes and consequences. Matrix Biol 2019; 78-79:147-164. [PMID: 29709595 DOI: 10.1016/j.matbio.2018.04.012] [Citation(s) in RCA: 71] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2017] [Revised: 03/13/2018] [Accepted: 04/25/2018] [Indexed: 02/08/2023]
|