1
|
Li X, Xu H, Li C, Guan Y, Liu Y, Zhang T, Meng F, Cheng H, Song X, Jia Z, He R, Zhao J, Chen S, Guan C, Yan S, Wang J, Wei Y, Zhang J, Tang J, Peng J, Wang Y. Biological characteristics of tissue engineered-nerve grafts enhancing peripheral nerve regeneration. Stem Cell Res Ther 2024; 15:215. [PMID: 39020413 PMCID: PMC11256578 DOI: 10.1186/s13287-024-03827-9] [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: 04/15/2024] [Accepted: 07/02/2024] [Indexed: 07/19/2024] Open
Abstract
BACKGROUND A favorable regenerative microenvironment is essential for peripheral nerve regeneration. Neural tissue-specific extracellular matrix (ECM) is a natural material that helps direct cell behavior and promote axon regeneration. Both bone marrow-derived mesenchymal stem cells (BMSCs) and adipose-derived mesenchymal stem cells (ADSCs) transplantation are effective in repairing peripheral nerve injury (PNI). However, there is no study that characterizes the in vivo microenvironmental characteristics of these two MSCs for the early repair of PNI when combined with neural tissue-derived ECM materials, i.e., acellular nerve allograft (ANA). METHODS In order to investigate biological characteristics, molecular mechanisms of early stage, and effectiveness of ADSCs- or BMSCs-injected into ANA for repairing PNI in vivo, a rat 10 mm long sciatic nerve defect model was used. We isolated primary BMSCs and ADSCs from bone marrow and adipose tissue, respectively. First, to investigate the in vivo response characteristics and underlying molecular mechanisms of ANA combined with BMSCs or ADSCs, eighty-four rats were randomly divided into three groups: ANA group, ANA+BMSC group, and ANA+ADSC group. We performed flow cytometry, RT-PCR, and immunofluorescence staining up to 4 weeks postoperatively. To further elucidate the underlying molecular mechanisms, changes in long noncoding RNAs (lncRNAs), circular RNAs (circRNAs), microRNAs (miRNAs), and messenger RNAs (mRNAs) were systematically investigated using whole transcriptome sequencing. We then constructed protein-protein interaction networks to find 10 top ranked hub genes among differentially expressed mRNAs. Second, in order to explore the effectiveness of BMSCs and ADSCs on neural tissue-derived ECM materials for repairing PNI, sixty-eight rats were randomized into four groups: ANA group, ANA+BMSC group, ANA+ADSC group, and AUTO group. In the ANA+BMSC and ANA+ADSC groups, ADSCs/BMSCs were equally injected along the long axis of the 10-mm ANA. Then, we performed histological and functional assessments up to 12 weeks postoperatively. RESULTS The results of flow cytometry and RT-PCR showed that ANA combined with BMSCs exhibited more significant immunomodulatory effects, as evidenced by the up-regulation of interleukin (IL)-10, down-regulation of IL-1β and tumor necrosis factor-alpha (TNF-α) expression, promotion of M1-type macrophage polarization to M2-type, and a significant increase in the number of regulatory T cells (Tregs). ANA combined with ADSCs exhibited more pronounced features of pro-myelination and angiogenesis, as evidenced by the up-regulation of myelin-associated protein gene (MBP and MPZ) and angiogenesis-related factors (TGF-β, VEGF). Moreover, differentially expressed genes from whole transcriptome sequencing results further indicated that ANA loaded with BMSCs exhibited notable immunomodulatory effects and ANA loaded with ADSCs was more associated with angiogenesis, axonal growth, and myelin formation. Notably, ANA infused with BMSCs or ADSCs enhanced peripheral nerve regeneration and motor function recovery with no statistically significant differences. CONCLUSIONS This study revealed that both ANA combined with BMSCs and ADSCs enhance peripheral nerve regeneration and motor function recovery, but their biological characteristics (mainly including immunomodulatory effects, pro-vascular regenerative effects, and pro-myelin regenerative effects) and underlying molecular mechanisms in the process of repairing PNI in vivo are different, providing new insights into MSC therapy for peripheral nerve injury and its clinical translation.
Collapse
Affiliation(s)
- Xiangling Li
- The Fourth Medical Center of the General Hospital of People's Liberation Army, Beijing, 100853, China
- Department of Rehabilitation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Institute of Orthopedics, Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, Beijing, 100853, China
| | - Hang Xu
- Department of General Surgery, General Hospital, Tianjin Medical University, Tianjin, 300052, China
| | - Chaochao Li
- Institute of Orthopedics, Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, Beijing, 100853, China
| | - Yanjun Guan
- Institute of Orthopedics, Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, Beijing, 100853, China
| | - Yuli Liu
- Institute of Orthopedics, Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, Beijing, 100853, China
| | - Tieyuan Zhang
- Institute of Orthopedics, Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, Beijing, 100853, China
- Shandong University Center for Orthopaedics, Cheeloo College of Medicine, Shandong University, Jinan, 250012, China
| | - Fanqi Meng
- Department of Anesthesiology, Xuanwu Hospital Capital Medical University, Beijing, 100053, China
| | - Haofeng Cheng
- Institute of Orthopedics, Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, Beijing, 100853, China
- School of Medicine, Nankai University, Tianjin, 300071, China
| | - Xiangyu Song
- Institute of Orthopedics, Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, Beijing, 100853, China
- School of Medicine, Hebei North University, Zhangjiakou, 075132, China
| | - Zhibo Jia
- Institute of Orthopedics, Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, Beijing, 100853, China
- School of Medicine, Hebei North University, Zhangjiakou, 075132, China
| | - Ruichao He
- Institute of Orthopedics, Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, Beijing, 100853, China
- School of Medicine, Nankai University, Tianjin, 300071, China
| | - Jinjuan Zhao
- The Fourth Medical Center of the General Hospital of People's Liberation Army, Beijing, 100853, China
- Institute of Orthopedics, Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, Beijing, 100853, China
| | - Shengfeng Chen
- Institute of Orthopedics, Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, Beijing, 100853, China
| | - Congcong Guan
- Institute of Orthopedics, Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, Beijing, 100853, China
- School of Medicine, Nankai University, Tianjin, 300071, China
| | - Shi Yan
- Institute of Orthopedics, Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, Beijing, 100853, China
- School of Medicine, Nankai University, Tianjin, 300071, China
| | - Jinpeng Wang
- Institute of Orthopedics, Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, Beijing, 100853, China
| | - Yu Wei
- Institute of Orthopedics, Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, Beijing, 100853, China
| | - Jian Zhang
- Institute of Orthopedics, Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, Beijing, 100853, China
| | - Jinshu Tang
- The Fourth Medical Center of the General Hospital of People's Liberation Army, Beijing, 100853, China.
| | - Jiang Peng
- The Fourth Medical Center of the General Hospital of People's Liberation Army, Beijing, 100853, China.
- Institute of Orthopedics, Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, Beijing, 100853, China.
| | - Yu Wang
- The Fourth Medical Center of the General Hospital of People's Liberation Army, Beijing, 100853, China.
- Institute of Orthopedics, Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, Beijing, 100853, China.
- Co-Innovation Center of Neuroregeneration, Nantong University, Nantong, 226001, China.
| |
Collapse
|
2
|
Wang Y, Cheng S, Fleishman JS, Chen J, Tang H, Chen ZS, Chen W, Ding M. Targeting anoikis resistance as a strategy for cancer therapy. Drug Resist Updat 2024; 75:101099. [PMID: 38850692 DOI: 10.1016/j.drup.2024.101099] [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: 04/07/2024] [Revised: 05/25/2024] [Accepted: 05/27/2024] [Indexed: 06/10/2024]
Abstract
Anoikis, known as matrix detachment-induced apoptosis or detachment-induced cell death, is crucial for tissue development and homeostasis. Cancer cells develop means to evade anoikis, e.g. anoikis resistance, thereby allowing for cells to survive under anchorage-independent conditions. Uncovering the mechanisms of anoikis resistance will provide details about cancer metastasis, and potential strategies against cancer cell dissemination and metastasis. Here, we summarize the principal elements and core molecular mechanisms of anoikis and anoikis resistance. We discuss the latest progress of how anoikis and anoikis resistance are regulated in cancers. Furthermore, we summarize emerging data on selective compounds and nanomedicines, explaining how inhibiting anoikis resistance can serve as a meaningful treatment modality against cancers. Finally, we discuss the key limitations of this therapeutic paradigm and possible strategies to overcome them. In this review, we suggest that pharmacological modulation of anoikis and anoikis resistance by bioactive compounds could surmount anoikis resistance, highlighting a promising therapeutic regimen that could be used to overcome anoikis resistance in cancers.
Collapse
Affiliation(s)
- Yumin Wang
- Department of Respiratory and Critical Care Medicine, Aerospace Center Hospital, Peking University Aerospace School of Clinical Medicine, Beijing 100049, China
| | - Sihang Cheng
- Department of Radiology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing 100730, China
| | - Joshua S Fleishman
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, NY 11439, USA
| | - Jichao Chen
- Department of Respiratory and Critical Care Medicine, Aerospace Center Hospital, Peking University Aerospace School of Clinical Medicine, Beijing 100049, China
| | - Hailin Tang
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-Sen University Cancer Center, Guangzhou, China
| | - Zhe-Sheng Chen
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, NY 11439, USA.
| | - Wenkuan Chen
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-Sen University Cancer Center, Guangzhou, China.
| | - Mingchao Ding
- Department of Peripheral Vascular Intervention, Aerospace Center Hospital, Peking University Aerospace School of Clinical Medicine, Beijing 100049, China.
| |
Collapse
|
3
|
Zhao D, Guo Y, Wei H, Jia X, Zhi Y, He G, Nie W, Huang L, Wang P, Laster KV, Liu Z, Wang J, Lee MH, Dong Z, Liu K. Multi-omics characterization of esophageal squamous cell carcinoma identifies molecular subtypes and therapeutic targets. JCI Insight 2024; 9:e171916. [PMID: 38652547 PMCID: PMC11141925 DOI: 10.1172/jci.insight.171916] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Accepted: 04/12/2024] [Indexed: 04/25/2024] Open
Abstract
Esophageal squamous cell carcinoma (ESCC) is the predominant form of esophageal cancer and is characterized by an unfavorable prognosis. To elucidate the distinct molecular alterations in ESCC and investigate therapeutic targets, we performed a comprehensive analysis of transcriptomics, proteomics, and phosphoproteomics data derived from 60 paired treatment-naive ESCC and adjacent nontumor tissue samples. Additionally, we conducted a correlation analysis to describe the regulatory relationship between transcriptomic and proteomic processes, revealing alterations in key metabolic pathways. Unsupervised clustering analysis of the proteomics data stratified patients with ESCC into 3 subtypes with different molecular characteristics and clinical outcomes. Notably, subtype III exhibited the worst prognosis and enrichment in proteins associated with malignant processes, including glycolysis and DNA repair pathways. Furthermore, translocase of inner mitochondrial membrane domain containing 1 (TIMMDC1) was validated as a potential prognostic molecule for ESCC. Moreover, integrated kinase-substrate network analysis using the phosphoproteome nominated candidate kinases as potential targets. In vitro and in vivo experiments further confirmed casein kinase II subunit α (CSNK2A1) as a potential kinase target for ESCC. These underlying data represent a valuable resource for researchers that may provide better insights into the biology and treatment of ESCC.
Collapse
Affiliation(s)
- Dengyun Zhao
- Department of Pathophysiology, School of Basic Medical Sciences, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, Henan, China
- China-US (Henan) Hormel Cancer Institute, Zhengzhou, Henan, China
- Tianjian Laboratory of Advanced Biomedical Sciences, Zhengzhou, Henan, China
| | - Yaping Guo
- Department of Pathophysiology, School of Basic Medical Sciences, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, Henan, China
- The Collaborative Innovation Center of Henan Province for Cancer Chemoprevention, Zhengzhou, Henan, China
- State Key Laboratory of Esophageal Cancer Prevention and Treatment, Zhengzhou University, Zhengzhou, Henan, China
| | - Huifang Wei
- Department of Pathophysiology, School of Basic Medical Sciences, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, Henan, China
- China-US (Henan) Hormel Cancer Institute, Zhengzhou, Henan, China
| | - Xuechao Jia
- Department of Pathophysiology, School of Basic Medical Sciences, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, Henan, China
- China-US (Henan) Hormel Cancer Institute, Zhengzhou, Henan, China
| | - Yafei Zhi
- Department of Pathophysiology, School of Basic Medical Sciences, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, Henan, China
- China-US (Henan) Hormel Cancer Institute, Zhengzhou, Henan, China
| | - Guiliang He
- China-US (Henan) Hormel Cancer Institute, Zhengzhou, Henan, China
| | - Wenna Nie
- China-US (Henan) Hormel Cancer Institute, Zhengzhou, Henan, China
| | - Limeng Huang
- China-US (Henan) Hormel Cancer Institute, Zhengzhou, Henan, China
| | - Penglei Wang
- Department of Pathophysiology, School of Basic Medical Sciences, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, Henan, China
- China-US (Henan) Hormel Cancer Institute, Zhengzhou, Henan, China
| | | | - Zhicai Liu
- Linzhou Cancer Hospital, Anyang, Henan, China
| | - Jinwu Wang
- Linzhou Cancer Hospital, Anyang, Henan, China
| | - Mee-Hyun Lee
- Department of Pathophysiology, School of Basic Medical Sciences, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, Henan, China
- China-US (Henan) Hormel Cancer Institute, Zhengzhou, Henan, China
- College of Korean Medicine, Dongshin University, Naju, Jeonnam, Republic of Korea
| | - Zigang Dong
- Department of Pathophysiology, School of Basic Medical Sciences, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, Henan, China
- China-US (Henan) Hormel Cancer Institute, Zhengzhou, Henan, China
- Tianjian Laboratory of Advanced Biomedical Sciences, Zhengzhou, Henan, China
- The Collaborative Innovation Center of Henan Province for Cancer Chemoprevention, Zhengzhou, Henan, China
- State Key Laboratory of Esophageal Cancer Prevention and Treatment, Zhengzhou University, Zhengzhou, Henan, China
- Provincial Cooperative Innovation Center for Cancer Chemoprevention, Zhengzhou University, Zhengzhou, Henan, China
| | - Kangdong Liu
- Department of Pathophysiology, School of Basic Medical Sciences, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, Henan, China
- China-US (Henan) Hormel Cancer Institute, Zhengzhou, Henan, China
- Tianjian Laboratory of Advanced Biomedical Sciences, Zhengzhou, Henan, China
- The Collaborative Innovation Center of Henan Province for Cancer Chemoprevention, Zhengzhou, Henan, China
- State Key Laboratory of Esophageal Cancer Prevention and Treatment, Zhengzhou University, Zhengzhou, Henan, China
- Provincial Cooperative Innovation Center for Cancer Chemoprevention, Zhengzhou University, Zhengzhou, Henan, China
| |
Collapse
|
4
|
Chen P, Long J, Hua T, Zheng Z, Xiao Y, Chen L, Yu K, Wu W, Zhang S. Transcriptome and open chromatin analysis reveals the process of myocardial cell development and key pathogenic target proteins in Long QT syndrome type 7. J Transl Med 2024; 22:307. [PMID: 38528561 PMCID: PMC10964537 DOI: 10.1186/s12967-024-05125-7] [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: 09/23/2023] [Accepted: 03/20/2024] [Indexed: 03/27/2024] Open
Abstract
OBJECTIVE Long QT syndrome type 7 (Andersen-Tawil syndrome, ATS), which is caused by KCNJ2 gene mutation, often leads to ventricular arrhythmia, periodic paralysis and skeletal malformations. The development, differentiation and electrophysiological maturation of cardiomyocytes (CMs) changes promote the pathophysiology of Long QT syndrome type 7(LQT7). We aimed to specifically reproduce the ATS disease phenotype and study the pathogenic mechanism. METHODS AND RESULTS We established a cardiac cell model derived from human induced pluripotent stem cells (hiPSCs) to the phenotypes and electrophysiological function, and the establishment of a human myocardial cell model that specifically reproduces the symptoms of ATS provides a reliable platform for exploring the mechanism of this disease or potential drugs. The spontaneous pulsation rate of myocardial cells in the mutation group was significantly lower than that in the repair CRISPR group, the action potential duration was prolonged, and the Kir2.1 current of the inward rectifier potassium ion channel was decreased, which is consistent with the clinical symptoms of ATS patients. Only ZNF528, a chromatin-accessible TF related to pathogenicity, was continuously regulated beginning from the cardiac mesodermal precursor cell stage (day 4), and continued to be expressed at low levels, which was identified by WGCNA method and verified with ATAC-seq data in the mutation group. Subsequently, it indicated that seven pathways were downregulated (all p < 0.05) by used single sample Gene Set Enrichment Analysis to evaluate the overall regulation of potassium-related pathways enriched in the transcriptome and proteome of late mature CMs. Among them, the three pathways (GO: 0008076, GO: 1990573 and GO: 0030007) containing the mutated gene KCNJ2 is involved that are related to the whole process by which a potassium ion enters the cell via the inward rectifier potassium channel to exert its effect were inhibited. The other four pathways are related to regulation of the potassium transmembrane pathway and sodium:potassium exchange ATPase (p < 0.05). ZNF528 small interfering (si)-RNA was applied to hiPSC-derived cardiomyocytes for CRISPR group to explore changes in potassium ion currents and growth and development related target protein levels that affect disease phenotype. Three consistently downregulated proteins (KCNJ2, CTTN and ATP1B1) associated with pathogenicity were verificated through correlation and intersection analysis. CONCLUSION This study uncovers TFs and target proteins related to electrophysiology and developmental pathogenicity in ATS myocardial cells, obtaining novel targets for potential therapeutic candidate development that does not rely on gene editing.
Collapse
Affiliation(s)
- Peipei Chen
- Department of Clinical Nutrition & Health Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- Department of Cardiology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
| | - Junyu Long
- Department of Liver Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Tianrui Hua
- Department of Cardiology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
| | - Zhifa Zheng
- Department of Orthopedic Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Ying Xiao
- Department of Cardiology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
| | - Lianfeng Chen
- Department of Cardiology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
| | - Kang Yu
- Department of Clinical Nutrition & Health Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
| | - Wei Wu
- Department of Cardiology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China.
- State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
| | - Shuyang Zhang
- Department of Cardiology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China.
- State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
| |
Collapse
|
5
|
Zhao Y, Hu F, Wang Q. Cortactin contributes to the tumorigenesis of gastric cancer by activating ERK/MMP pathway. Heliyon 2023; 9:e18289. [PMID: 37539204 PMCID: PMC10395536 DOI: 10.1016/j.heliyon.2023.e18289] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2022] [Revised: 07/10/2023] [Accepted: 07/13/2023] [Indexed: 08/05/2023] Open
Abstract
Gastric cancer is a malignant tumor with high mortality and high incidence. This study aims to explore the function and molecular mechanism of Cortactin on gastric cancer progression in vitro and in vivo. A bioinformatics analysis from TCGA displayed that Cortactin was highly expressed in gastric cancer samples, and patients with a high Cortactin level had a worse survival rate. Subsequently, we investigated the specific mechanism of action of A in gastric cancer by collecting patient samples for immunohistochemistry, WB, qRT-PCR, cell transfection, cell invasion and metastasis, and constructing tumor xenografts in nude mice. Overexpression of Cortactin inhibited apoptosis and enhanced cellular proliferation and mobility in AGS cells, while those activities were reversed by the knockdown of MMP2 or MMP9. Conversely, the deletion of Cortactin induced apoptosis and suppressed cell growth and metastasis in SGC7901 cells, whereas those behaviors were inhibited by overexpression of MMP2 or MMP9. Additionally, the ERK pathway was activated by Cortactin upregulation. In vivo studies presented that overexpression of Cortactin promoted tumor growth, increased Ki67 expression, and reduced caspase 3 expression, which was reversed by ERK inhibitor treatment. In conclusion, Cortactin acted as an oncogene in gastric cancer and exerted its function by ERK/MMP2/MMP9 signaling pathway.
Collapse
Affiliation(s)
- Yi Zhao
- Department of Gastroenterology, The First Affiliated Hospital of Bengbu Medical College, Bengbu Anhui 233004, China
| | - Fang Hu
- Department of Hematology, The First Affiliated Hospital of Bengbu Medical College, Bengbu Anhui 233004, China
| | - Qizhi Wang
- Department of Gastroenterology, The First Affiliated Hospital of Bengbu Medical College, Bengbu Anhui 233004, China
| |
Collapse
|
6
|
Banushi B, Joseph SR, Lum B, Lee JJ, Simpson F. Endocytosis in cancer and cancer therapy. Nat Rev Cancer 2023:10.1038/s41568-023-00574-6. [PMID: 37217781 DOI: 10.1038/s41568-023-00574-6] [Citation(s) in RCA: 41] [Impact Index Per Article: 41.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 04/11/2023] [Indexed: 05/24/2023]
Abstract
Endocytosis is a complex process whereby cell surface proteins, lipids and fluid from the extracellular environment are packaged, sorted and internalized into cells. Endocytosis is also a mechanism of drug internalization into cells. There are multiple routes of endocytosis that determine the fate of molecules, from degradation in the lysosomes to recycling back to the plasma membrane. The overall rates of endocytosis and temporal regulation of molecules transiting through endocytic pathways are also intricately linked with signalling outcomes. This process relies on an array of factors, such as intrinsic amino acid motifs and post-translational modifications. Endocytosis is frequently disrupted in cancer. These disruptions lead to inappropriate retention of receptor tyrosine kinases on the tumour cell membrane, changes in the recycling of oncogenic molecules, defective signalling feedback loops and loss of cell polarity. In the past decade, endocytosis has emerged as a pivotal regulator of nutrient scavenging, response to and regulation of immune surveillance and tumour immune evasion, tumour metastasis and therapeutic drug delivery. This Review summarizes and integrates these advances into the understanding of endocytosis in cancer. The potential to regulate these pathways in the clinic to improve cancer therapy is also discussed.
Collapse
Affiliation(s)
- Blerida Banushi
- Frazer Institute, University of Queensland, Woolloongabba, Queensland, Australia
| | - Shannon R Joseph
- Frazer Institute, University of Queensland, Woolloongabba, Queensland, Australia
| | - Benedict Lum
- Frazer Institute, University of Queensland, Woolloongabba, Queensland, Australia
| | - Jason J Lee
- Frazer Institute, University of Queensland, Woolloongabba, Queensland, Australia
| | - Fiona Simpson
- Frazer Institute, University of Queensland, Woolloongabba, Queensland, Australia.
| |
Collapse
|
7
|
Moon SJ, Choi HJ, Kye YH, Jeong GY, Kim HY, Myung JK, Kong G. CTTN Overexpression Confers Cancer Stem Cell-like Properties and Trastuzumab Resistance via DKK-1/WNT Signaling in HER2 Positive Breast Cancer. Cancers (Basel) 2023; 15:cancers15041168. [PMID: 36831511 PMCID: PMC9954024 DOI: 10.3390/cancers15041168] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2023] [Revised: 02/03/2023] [Accepted: 02/09/2023] [Indexed: 02/17/2023] Open
Abstract
BACKGROUND Despite the therapeutic success of trastuzumab, HER2 positive (HER2+) breast cancer patients continue to face significant difficulties due to innate or acquired drug resistance. In this study we explored the potential role of CTTN in inducing trastuzumab resistance of HER2+ breast cancers. METHODS Genetic changes of CTTN and survival of HER2+ breast cancer patients were analyzed in multiple breast cancer patient cohorts (METABRIC, TCGA, Kaplan-Meier (KM) plotter, and Hanyang University cohort). The effect of CTTN on cancer stem cell activity was assessed using the tumorsphere formation, ALDEFLUOR assay, and by in vivo xenograft experiments. CTTN-induced trastuzumab resistance was assessed by the sulforhodamine B (SRB) assay, colony formation assays, and in vivo xenograft model. RNA-seq analysis was used to clarify the mechanism of trastuzumab resistance conferred by CTTN. RESULTS Survival analysis indicated that CTTN overexpression is related to a poor prognosis in HER2+ breast cancers (OS, p = 0.05 in the Hanyang University cohort; OS, p = 0.0014 in KM plotter; OS, p = 0.008 and DFS, p = 0.010 in METABRIC). CTTN overexpression-induced cancer stem cell-like characteristics in experiments of tumorsphere formation, ALDEFLUOR assays, and in vivo limiting dilution assays. CTTN overexpression resulted in trastuzumab resistance in SRB, colony formation assays, and in vivo xenograft models. Mechanistically, the mRNA and protein levels of DKK-1, a Wnt antagonist, were downregulated by CTTN. Treatment of the β-catenin/TCF inhibitor reversed CTTN-induced cancer stem cell-like properties in vitro. Combination treatment with trastuzumab and β-catenin/TCF inhibitor overcame trastuzumab resistance conferred by CTTN overexpression in in vitro colony formation assays. CONCLUSIONS CTTN activates DKK-1/Wnt/β-catenin signaling to induce trastuzumab resistance. We propose that CTTN is a novel biomarker indicating a poor prognosis and a possible therapeutic target for overcoming trastuzumab resistance.
Collapse
Affiliation(s)
- So-Jeong Moon
- Department of HY-KIST Bio-Convergence, Hanyang University, Seoul 04763, Republic of Korea
- Department of Pathology, College of Medicine, Hanyang University, Seoul 04763, Republic of Korea
| | - Hyung-Jun Choi
- Department of Pathology, College of Medicine, Hanyang University, Seoul 04763, Republic of Korea
| | - Young-Hyeon Kye
- Department of HY-KIST Bio-Convergence, Hanyang University, Seoul 04763, Republic of Korea
- Department of Pathology, College of Medicine, Hanyang University, Seoul 04763, Republic of Korea
| | - Ga-Young Jeong
- Department of Pathology, College of Medicine, Hanyang University, Seoul 04763, Republic of Korea
| | - Hyung-Yong Kim
- Department of Pathology, College of Medicine, Hanyang University, Seoul 04763, Republic of Korea
| | - Jae-Kyung Myung
- Department of Pathology, College of Medicine, Hanyang University, Seoul 04763, Republic of Korea
| | - Gu Kong
- Department of HY-KIST Bio-Convergence, Hanyang University, Seoul 04763, Republic of Korea
- Department of Pathology, College of Medicine, Hanyang University, Seoul 04763, Republic of Korea
- Correspondence: ; Tel.: +82-2-2290-8251; Fax: +82-2-2295-1091
| |
Collapse
|
8
|
Wang H, Tian RF, Liang X, Fan J, Duan ZC, Fan XY, Zhang JJ, Yao DS, Chen ZN, Li L. A four oxidative stress gene prognostic model and integrated immunity-analysis in pancreatic adenocarcinoma. Front Oncol 2023; 12:1015042. [PMID: 36713541 PMCID: PMC9880292 DOI: 10.3389/fonc.2022.1015042] [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: 08/09/2022] [Accepted: 12/28/2022] [Indexed: 01/15/2023] Open
Abstract
Background and aims Pancreatic adenocarcinoma (PAAD) is highly aggressive and characterized by a poor prognosis. Oxidative stress has great impacts on the occurrence and development of tumors. However, the predictive role of oxidative stress related genes on PAAD patients' prognosis remains unclear. In this study, we aimed to construct a prognostic model for PAAD based on oxidative stress genes and to evaluate its predictive value. Methods The Cancer Genome Atlas (TCGA) and three Gene Expression Omnibus (GEO) datasets were used to identify differentially expressed oxidative stress genes. Univariate Cox regression, Kaplan-Meier and multivariate Cox regression analysis were used to select genes and to construct a prognosis model. According to the median value of the model's risk score, patients were divided into high and low risk groups, and gene set enrichment analysis (GSEA), immune infiltration and immunotherapy effect, drug resistance and the expression of immune checkpoint related genes and synthetic driver genes of T cell proliferation were analyzed. Finally, the mRNA and protein levels of four genes in PAAD were verified by the clinical proteomic tumor analysis consortium (CPTAC) database and the immunostaining of patients' tissue. Results 55 differentially expressed oxidative stress genes were identified, and four genes including MET, FYN, CTTN and CDK1 were selected to construct a prognosis model. GESA indicated that immune related pathways, metabolic pathways and DNA repair pathways were significantly enriched in the high risk group as compared to the low risk group. The frequency of genetic mutations was also significantly higher in high risk groups than that in low risk groups. Moreover, the infiltration level of 23 immune cells as well as the expression of immune checkpoint related and synthetic driver genes of T cell proliferation were significantly altered, with the better immunotherapy effect occurring in low risk group. In patient PAAD tissues, the mRNA and protein levels of these four genes were up-regulated. Conclusion We have successfully constructed a four oxidative stress gene prognostic model that has important predictive value for PAAD patients, and this model might be a promising guidance for prognostic prediction and efficacy monitoring in clinical individualized therapy.
Collapse
Affiliation(s)
- Hao Wang
- Institutes of Biomedicine and Department of Cell Biology, Jinan University, Guangzhou, China
| | - Ruo-Fei Tian
- Department of Cell Biology, National Translational Science Center for Molecular Medicine, Fourth Military Medical University, Xi’an, China
| | - Xue Liang
- Department of Cell Biology, National Translational Science Center for Molecular Medicine, Fourth Military Medical University, Xi’an, China
| | - Jing Fan
- Department of Cell Biology, National Translational Science Center for Molecular Medicine, Fourth Military Medical University, Xi’an, China
| | - Zi-Chuan Duan
- Department of Cell Biology, National Translational Science Center for Molecular Medicine, Fourth Military Medical University, Xi’an, China
| | - Xin-Yu Fan
- Department of Cell Biology, National Translational Science Center for Molecular Medicine, Fourth Military Medical University, Xi’an, China
| | - Jia-Jia Zhang
- Department of Cell Biology, National Translational Science Center for Molecular Medicine, Fourth Military Medical University, Xi’an, China
| | - Dong-Sheng Yao
- Institutes of Biomedicine and Department of Cell Biology, Jinan University, Guangzhou, China,*Correspondence: Zhi-Nan Chen, ; Ling Li, ; Dong-Sheng Yao,
| | - Zhi-Nan Chen
- Institutes of Biomedicine and Department of Cell Biology, Jinan University, Guangzhou, China,Department of Cell Biology, National Translational Science Center for Molecular Medicine, Fourth Military Medical University, Xi’an, China,*Correspondence: Zhi-Nan Chen, ; Ling Li, ; Dong-Sheng Yao,
| | - Ling Li
- Department of Cell Biology, National Translational Science Center for Molecular Medicine, Fourth Military Medical University, Xi’an, China,*Correspondence: Zhi-Nan Chen, ; Ling Li, ; Dong-Sheng Yao,
| |
Collapse
|
9
|
Li W, Lei T, Song X, Deng C, Lu J, Zhang W, Kuang Z, He Y, Zhou Q, Luo Z, Mo F, Yang H, Hang J, Xiao B, Li L. CBLC inhibits the proliferation and metastasis of breast cancer cells via ubiquitination and degradation of CTTN. J Recept Signal Transduct Res 2022; 42:588-598. [PMID: 36043996 DOI: 10.1080/10799893.2022.2116049] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The E3 ubiquitin ligase is an important regulator of cell signaling and proteostasis and is tightly controlled in many diseases, including cancer. Our study aimed to investigate the biological role of the E3 ubiquitin ligase CBLC in breast cancer and elucidate the specific mechanistic network underlying CBLC-mediated target substrate degradation, cell proliferation and metastasis. Here, we showed that CBLC expression was higher in breast cancer tissues and cells than that in normal tissues and cells. Higher expression of CBLC predicted a better prognosis for breast cancer patients. CBLC inhibited the proliferation, migration and invasion of breast cancer cells. Co-IP and immunofluorescence co-localization assays demonstrated that CBLC interacted with CTTN in the cytoplasm. CBLC promoted the degradation of CTTN through the ubiquitin-proteasome pathway without affecting its mRNA level. The inhibitory effect of CBLC on breast cancer cell proliferation, migration and invasion could partly be reversed by CTTN. Taken together, our study clarified the biological role of CBLC as a tumor suppressor and discovered its functional substrate, providing a molecular basis for CBLC/CTTN as a potential therapeutic target in breast cancer.
Collapse
Affiliation(s)
- Weiwei Li
- Department of Laboratory Medicine, The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People's Hospital, Guangdong, China.,Department of Laboratory Medicine, Guiyang Maternity & Child Health Hospital, Guiyang, China.,Department of Basic Clinical Laboratory Medicine, School of Clinical Laboratory Science, Guizhou Medical University, Guiyang, China
| | - Ting Lei
- Department of Laboratory Medicine, The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People's Hospital, Guangdong, China.,Department of Laboratory Medicine, General Hospital of Southern Theatre Command of PLA, Guangzhou, China
| | - Xiaoyu Song
- Department of Laboratory Medicine, The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People's Hospital, Guangdong, China
| | - Chun Deng
- Department of Laboratory Medicine, The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People's Hospital, Guangdong, China.,Department of Basic Clinical Laboratory Medicine, School of Clinical Laboratory Science, Guizhou Medical University, Guiyang, China
| | - Jingrun Lu
- Department of Laboratory Medicine, The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People's Hospital, Guangdong, China.,Department of Basic Clinical Laboratory Medicine, School of Clinical Laboratory Science, Guizhou Medical University, Guiyang, China
| | - Wenwu Zhang
- Department of Laboratory Medicine, The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People's Hospital, Guangdong, China
| | - Zhenzhan Kuang
- Department of Laboratory Medicine, The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People's Hospital, Guangdong, China.,Department of Laboratory Medicine, General Hospital of Southern Theatre Command of PLA, Guangzhou, China
| | - Yongyin He
- Department of Laboratory Medicine, The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People's Hospital, Guangdong, China.,Department of Laboratory Medicine, General Hospital of Southern Theatre Command of PLA, Guangzhou, China
| | - Quan Zhou
- Department of Laboratory Medicine, General Hospital of Southern Theatre Command of PLA, Guangzhou, China
| | - Zhaoxun Luo
- School of Pediatrics, Guizhou Medical University, Guiyang, China
| | - Fei Mo
- Department of Basic Clinical Laboratory Medicine, School of Clinical Laboratory Science, Guizhou Medical University, Guiyang, China
| | - Hanlin Yang
- Department of Laboratory Medicine, The Guiyang Second People's Hospital, Guiyang, China
| | - Jianfeng Hang
- Department of Laboratory Medicine, General Hospital of Southern Theatre Command of PLA, Guangzhou, China
| | - Bin Xiao
- Department of Laboratory Medicine, The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People's Hospital, Guangdong, China
| | - Linhai Li
- Department of Laboratory Medicine, The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People's Hospital, Guangdong, China
| |
Collapse
|
10
|
Yuan X, Ma S, Fa B, Wei T, Ma Y, Wang Y, Lv W, Zhang Y, Zheng J, Chen G, Sun J, Yu Z. A high-efficiency differential expression method for cancer heterogeneity using large-scale single-cell RNA-sequencing data. Front Genet 2022; 13:1063130. [DOI: 10.3389/fgene.2022.1063130] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2022] [Accepted: 11/14/2022] [Indexed: 12/03/2022] Open
Abstract
Colorectal cancer is a highly heterogeneous disease. Tumor heterogeneity limits the efficacy of cancer treatment. Single-cell RNA-sequencing technology (scRNA-seq) is a powerful tool for studying cancer heterogeneity at cellular resolution. The sparsity, heterogeneous diversity, and fast-growing scale of scRNA-seq data pose challenges to the flexibility, accuracy, and computing efficiency of the differential expression (DE) methods. We proposed HEART (high-efficiency and robust test), a statistical combination test that can detect DE genes with various sources of differences beyond mean expression changes. To validate the performance of HEART, we compared HEART and the other six popular DE methods on various simulation datasets with different settings by two simulation data generation mechanisms. HEART had high accuracy (F1 score >0.75) and brilliant computational efficiency (less than 2 min) on multiple simulation datasets in various experimental settings. HEART performed well on DE genes detection for the PBMC68K dataset quantified by UMI counts and the human brain single-cell dataset quantified by read counts (F1 score = 0.79, 0.65). By applying HEART to the single-cell dataset of a colorectal cancer patient, we found several potential blood-based biomarkers (CTTN, S100A4, S100A6, UBA52, FAU, and VIM) associated with colorectal cancer metastasis and validated them on additional spatial transcriptomic data of other colorectal cancer patients.
Collapse
|
11
|
Xia W, Jiang H, Guo H, Liu Y, Gou X. Integrated gene co-expression network analysis reveals unique developmental processes of Aurelia aurita. Gene X 2022; 840:146733. [PMID: 35863715 DOI: 10.1016/j.gene.2022.146733] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2022] [Revised: 06/15/2022] [Accepted: 07/08/2022] [Indexed: 11/04/2022] Open
Abstract
The typical life cycle of the moon jellyfish (Aurelia aurita) includes the planula, polyp, strobila, ephyra, and medusa developmental stages. These stages exhibit huge differences in both external morphology and internal physiological functions. However, the gene co-expression network involved in these post-embryonic developmental processes has not been studied yet. Here, based on 15 RNA sequencing samples covering all five stages of the A. aurita life cycle, we systematically analyzed the gene co-expression network and obtained 35 relevant modules. Furthermore, we identified the highly correlated modules and hub genes for each stage. These hub genes are implicated to play important roles in the developmental processes of A. aurita, which should help improve our understanding of the jellyfish life cycle.
Collapse
Affiliation(s)
- Wangxiao Xia
- Shaanxi Key Laboratory of Brain Disorders,Institute of Basic Translational Medicine, Xi'an Medical University, Xi'an 710021, China
| | - Hui Jiang
- College of Life Science, Hainan Normal University, Haikou 571158, China
| | - Huifang Guo
- Shaanxi Key Laboratory of Infection and Immune Disorders, School of Basic Medical Science, Xi'an Medical University, Xi'an 710021, China
| | - Yaowen Liu
- College of Veterinary Medicine, Yunnan Agricultural University, Kunming 650231, China.
| | - Xingchun Gou
- Shaanxi Key Laboratory of Brain Disorders,Institute of Basic Translational Medicine, Xi'an Medical University, Xi'an 710021, China.
| |
Collapse
|
12
|
Zhang Y, Wu T, Li F, Cheng Y, Han Q, Lu X, Lu S, Xia W. FGF19 Is Coamplified With CCND1 to Promote Proliferation in Lung Squamous Cell Carcinoma and Their Combined Inhibition Shows Improved Efficacy. Front Oncol 2022; 12:846744. [PMID: 35463335 PMCID: PMC9021371 DOI: 10.3389/fonc.2022.846744] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Accepted: 02/28/2022] [Indexed: 12/09/2022] Open
Abstract
Lung squamous cell carcinoma (LUSC) remains as a major cause of cancer-associated mortality with few therapeutic options. Continued research on new driver genes is particularly important. FGF19, a fibroblast growth factor, is frequently observed as amplified in human LUSC, which is also associated with multiple genomic gains and losses. However, the importance of these associated changes is largely unknown. In this study, we aimed to clarify a novel mechanism that link neighboring oncogene co-amplification in the development of LUSC. We found that FGF19 was co-amplified and co-expressed with its neighboring gene CCND1 in a subset of LUSC patients and associated with poor prognosis. Moreover, FGF19 combined with CCND1 promoted the cell cycle progression of LUSC cells. Mechanistically, FGF19 also enhanced CCND1 expression by activating FGFR4-ERK1/2 signaling and strengthening CCND1-induced phosphorylation and inactivation of retinoblastoma (RB). In a murine model of lung orthotopic cancer, knockdown of CCND1 was found to prolong survival by attenuating FGF19-induced cell proliferation. Furthermore, the combination treatment of the FGFR4 inhibitor BLU9931 and the CDK4/6 inhibitor palbociclib potentiated the growth inhibition and arrested cells in G1 phase. In vivo, co-targeting FGFR4 and CDK4/6 also showed marked inhibition of tumor growth than single agent treatment. These findings further elucidate the oncogenic role of FGF19 in LUSC and provide insights into how the co-amplification of neighboring genes synergistically function to promote cancer growth, and combined inhibition against both FGF19 and CCND1 is more effective.
Collapse
Affiliation(s)
- Yanshuang Zhang
- State Key Laboratory of Oncogenes and Related Genes, Ren Ji Hospital, School of Medicine and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Tingyu Wu
- State Key Laboratory of Oncogenes and Related Genes, Ren Ji Hospital, School of Medicine and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Fan Li
- State Key Laboratory of Oncogenes and Related Genes, Ren Ji Hospital, School of Medicine and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Yirui Cheng
- State Key Laboratory of Oncogenes and Related Genes, Ren Ji Hospital, School of Medicine and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Qing Han
- State Key Laboratory of Oncogenes and Related Genes, Ren Ji Hospital, School of Medicine and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Xin Lu
- State Key Laboratory of Oncogenes and Related Genes, Ren Ji Hospital, School of Medicine and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Shun Lu
- Shanghai Lung Cancer Center, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Weiliang Xia
- State Key Laboratory of Oncogenes and Related Genes, Ren Ji Hospital, School of Medicine and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
13
|
Oshima K, Kato K, Ito Y, Daiko H, Nozaki I, Nakagawa S, Shibuya Y, Kojima T, Toh Y, Okada M, Hironaka S, Akiyama Y, Komatsu Y, Maejima K, Nakagawa H, Onuki R, Nagai M, Kato M, Kanato K, Kuchiba A, Nakamura K, Kitagawa Y. A prognostic biomarker study in patients with clinical stage I esophageal squamous cell carcinoma: JCOG0502-A1. Cancer Sci 2021; 113:1018-1027. [PMID: 34962019 PMCID: PMC8898710 DOI: 10.1111/cas.15251] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Revised: 12/11/2021] [Accepted: 12/13/2021] [Indexed: 11/03/2022] Open
Abstract
We conducted genomic analyses of Japanese patients with stage I esophageal squamous cell carcinoma (ESCC) to investigate the frequency of genomic alterations and the association with survival outcomes. Biomarker analysis was conducted for patients with clinical stage T1bN0M0 ESCC enrolled in JCOG0502 (UMIN000000551). Whole-exome sequencing (WES) was performed using DNA extracted from formalin-fixed, paraffin-embedded tissue of ESCC and normal tissue or blood sample. Single nucleotide variants (SNVs), insertions/deletions (indels), and copy number alterations (CNAs) were identified. We then evaluated the associations between each gene alteration with a frequency ≥10% and progression-free survival (PFS) using a Cox regression model. We controlled for family-wise errors at 0.05 using the Bonferroni method. Among the 379 patients who were enrolled in JCOG0502, 127 patients were successfully analyzed using WES. The median patient age was 63 years (IQR, 57-67 years), and 78.0% of the patients ultimately underwent surgery. The 3-year PFS probability was 76.3%. We detected 20 genes with SNVs, indels, or amplifications with a frequency of ≥10%. Genomic alterations in FGF19 showed the strongest association with PFS with a borderline level of statistical significance of p = 0.00252 (Bonferroni-adjusted significance level is 0.0025). Genomic alterations in FGF4, MYEOV, CTTN, and ORAOV1 showed a marginal association with PFS (p < 0.05). These genomic alterations were all CNAs at chromosome 11q13.3. We have identified new genomic alterations associated with the poor efficacy of ESCC (T1bN0M0). These findings open avenues for the development of new potential treatments for patients with ESCC.
Collapse
Affiliation(s)
- Kotoe Oshima
- Department of Gastrointestinal Medical Oncology, National Cancer Center Hospital, Tokyo
| | - Ken Kato
- Department of Head and Neck, Esophageal Medical Oncology, National Cancer Center Hospital, Tokyo
| | - Yoshinori Ito
- Department of Radiation Oncology, Showa University School of Medicine, Tokyo
| | - Hiroyuki Daiko
- Esophageal Surgery Division, National Cancer Center Hospital, Tokyo
| | - Isao Nozaki
- Department of Gastroenterological Surgery, National Hospital Organization Shikoku Cancer Center, Matsuyama
| | - Satoru Nakagawa
- Department of Gastroenterological Surgery, Niigata Cancer Center Hospital, Niigata
| | - Yuichi Shibuya
- Department of Gastroenterology Surgery, Kochi Health Sciences Center, Kochi
| | - Takashi Kojima
- Department of Gastroenterology and Gastrointestinal Oncology, National Cancer Center Hospital East, Kashiwa
| | - Yasushi Toh
- Department of Gastroenterological Surgery, National Hospital Organization Kyushu Cancer Center, Fukuoka
| | | | - Shuichi Hironaka
- Clinical Trial Promotion Department, Chiba Cancer Center, Chiba.,Department of Medical Oncology and Hematology, Oita University Faculty of Medicine, Yufu
| | - Yuji Akiyama
- Department of Surgery, Iwate Medical University, Iwate
| | - Yoshito Komatsu
- Cancer Chemotherapy, Hokkaido University Hospital Cancer Center, Sapporo
| | - Kazuhiro Maejima
- Laboratory for Cancer Genomics, RIKEN Center for integrative Medical Sciences, Yokohama
| | - Hidewaki Nakagawa
- Laboratory for Cancer Genomics, RIKEN Center for integrative Medical Sciences, Yokohama
| | - Ritsuko Onuki
- Division of Bioinformatics, Research Institute, National Cancer Center, Tokyo
| | - Momoko Nagai
- Division of Bioinformatics, Research Institute, National Cancer Center, Tokyo
| | - Mamoru Kato
- Division of Bioinformatics, Research Institute, National Cancer Center, Tokyo
| | - Keisuke Kanato
- Research Management Division, Clinical Research Support Office, National Cancer Center Hospital, Tokyo
| | - Aya Kuchiba
- Biostatistics Division, Center for Research Administration and Support, National Cancer Center, Tokyo.,Graduate School of Health Innovation, Kanagawa University of Human Services, Kanagawa
| | - Kenichi Nakamura
- Research Management Division, Clinical Research Support Office, National Cancer Center Hospital, Tokyo
| | - Yuko Kitagawa
- Department of Surgery, Keio University School of Medicine, Tokyo
| |
Collapse
|
14
|
Mazloomi SM, Foroutan-Ghaznavi M, Montazeri V, Tavoosidana G, Fakhrjou A, Nozad-Charoudeh H, Pirouzpanah S. Profiling the expression of pro-metastatic genes in association with the clinicopathological features of primary breast cancer. Cancer Cell Int 2021; 21:6. [PMID: 33407452 PMCID: PMC7789694 DOI: 10.1186/s12935-020-01708-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2020] [Accepted: 12/07/2020] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Metastasis accounts for ninety percent of breast cancer (BrCa) mortality. Cortactin, Ras homologous gene family member A (RhoA), and Rho-associated kinase (ROCK) raise cellular motility in favor of metastasis. Claudins (CLDN) belong to tight junction integrity and are dysregulated in BrCa. Thus far, epidemiologic evidence regarding the association of different pro-metastatic genes with pathological phenotypes of BrCa is largely inconsistent. This study aimed to determine the possible transcriptional models of pro-metastatic genes incorporate in holding the integrity of epithelial cell-cell junctions (CTTN, RhoA, ROCK, CLDN-1, CLDN-2, and CLDN-4), for the first time, in association with clinicopathological features of primary BrCa. METHODS In a consecutive case-series design, 206 newly diagnosed non-metastatic eligible BrCa patients with histopathological confirmation (30-65 years) were recruited in Tabriz, Iran (2015-2017). Real-time RT-PCR was used. Then fold changes in the expression of target genes were measured. RESULTS ROCK amplification was associated with the involvement of axillary lymph node metastasis (ALNM; ORadj. = 3.05, 95%CI 1.01-9.18). Consistently, inter-correlations of CTTN-ROCK (β = 0.226, P < 0.05) and RhoA-ROCK (β = 0.311, P < 0.01) were determined among patients diagnosed with ALNM+ BrCa. In addition, the overexpression of CLDN-4 was frequently observed in tumors identified by ALNM+ or grade III (P < 0.05). The overexpression of CTTN, CLDN-1, and CLDN-4 genes was correlated positively with the extent of tumor size. CTTN overexpression was associated with the increased chance of luminal-A positivity vs. non-luminal-A (ORadj. = 1.96, 95%CI 1.02-3.77). ROCK was also expressed in luminal-B BrCa tumors (P < 0.05). The estrogen receptor-dependent transcriptions were extended to the inter-correlations of RhoA-ROCK (β = 0.280, P < 0.01), ROCK-CLDN-2 (β = 0.267, P < 0.05), and CLDN-1-CLDN-4 (β = 0.451, P < 0.001). CONCLUSIONS For the first time, our findings suggested that the inter-correlations of CTTN-ROCK and RhoA-ROCK were significant transcriptional profiles determined in association with ALNM involvement; therefore the overexpression of ROCK may serve as a potential molecular marker for lymphatic metastasis. The provided binary transcriptional profiles need more approvals in different clinical features of BrCa metastasis.
Collapse
Affiliation(s)
- Seyed-Mohammad Mazloomi
- Nutrition Research Center, Department of Food Hygiene and Quality Control, Faculty of Nutrition and Food Sciences, Shiraz University of Medical Sciences, Shiraz, 7193635899 Iran
| | - Mitra Foroutan-Ghaznavi
- Students’ Research Committee, Faculty of Nutrition and Food Sciences, Shiraz University of Medical Sciences, Shiraz, 7134814336 Iran
- Molecular Medicine Research Center, Biomedicine Institute, Tabriz University of Medical Sciences, Tabriz, 5166414766 Iran
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, 5166614756 Iran
| | - Vahid Montazeri
- Department of Thoracic Surgery, Faculty of Medicine, Tabriz University of Medical Sciences, Surgery Ward, Nour-Nejat Hospital, Tabriz, 5166614766 Iran
| | - Gholamreza Tavoosidana
- Department of Molecular Medicine, Faculty of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, 1417755469 Iran
| | - Ashraf Fakhrjou
- Department of Pathology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, 5166614766 Iran
| | | | - Saeed Pirouzpanah
- Molecular Medicine Research Center, Biomedicine Institute, Tabriz University of Medical Sciences, Tabriz, 5166414766 Iran
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, 5166614756 Iran
- Department of Biochemistry and Dietetics, Faculty of Nutrition and Food Sciences, Tabriz University of Medical Sciences, Tabriz, 5166614711 Iran
| |
Collapse
|
15
|
Zeng YZ, Zhang YQ, Lin XQ, Chen JY, Zhang F, Zhu JL, Wei XL. Co-expression of VEGF-C and survivin predicts poor prognosis in esophageal squamous cell carcinoma. Transl Cancer Res 2021; 10:210-222. [PMID: 35116253 PMCID: PMC8799162 DOI: 10.21037/tcr-20-2498] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2020] [Accepted: 11/16/2020] [Indexed: 02/05/2023]
Abstract
BACKGROUND Lymphatic metastasis is one of the main factors affecting prognosis in esophageal squamous cell carcinoma (ESCC). Vascular endothelial growth factor-C (VEGF-C) is an important factor that promotes lymphangiogenesis. Survivin also plays a significant role in lymphatic invasion. However, the role and mechanism of their co-expression are still unclear in ESCC. The purpose of this study was to investigate whether the co-expression of VEGF-C and survivin could be a potential marker to predict patient prognosis and survival in ESCC. METHODS The levels of VEGF-C, vascular endothelial growth factor receptor 3 (VEGFR-3), survivin, and Ki-67 were determined by immunohistochemistry (IHC) in 97 ESCC patient tumors. The correlations of co-expression of VEGF-C and survivin with pathological features and survival results were also assessed. RESULTS High VEGF-C expression was observed in 64.9% of the patients and significantly correlated with T stage (P=0.024), node status (P=0.038), and lymph node metastasis (P=0.015). High survivin expression was significantly associated with T stage (P=0.013), N stage (P=0.016), lymph node metastasis (P=0.005), and differentiation (P=0.044) in 67.0% of the patients. Co-expression of VEGF-C and survivin (V+S+) was significantly associated with T stage (P<0.001), N stage (P=0.015), lymph node metastasis (P=0.003), differentiation (P=0.0045), and Ki-67 levels (P=0.024). High expression of VEGF-C or survivin was associated significantly with worse disease-free survival (DFS) and overall survival (OS) (P<0.05). Moreover, the V+S+ group had a worse DFS (P<0.001) and OS (P=0.001) than any other group (i.e., V-S-, V+S-, V-S+). Furthermore, multivariate DFS analyses (95% CI: 1.147-2.220, P=0.006) and multivariate OS analyses (95% CI: 1.080-2.193, P=0.017) revealed that co-expression of VEGF-C and survivin was an independent prognostic factor in ESCC patients. CONCLUSIONS Co-expression of VEGF-C and survivin was predictive of poor prognosis in ESCC. Combined detection of VEGF-C and survivin could represent a feasible and effective marker to predict the prognosis and survival of ESCC patients.
Collapse
Affiliation(s)
- Yun-Zhu Zeng
- Department of Pathology, Cancer Hospital of Shantou University Medical College, Shantou, China
| | - Yong-Qu Zhang
- Department of Breast-Thyroid-Surgery, Xiang’an Hospital of Xiamen University, Xiamen, China
| | - Xue-Qiong Lin
- Clinical Laboratory, Cancer Hospital of Shantou University Medical College, Shantou, China
| | - Jiong-Yu Chen
- Oncological Research Lab, Cancer Hospital of Shantou University Medical College, Shantou, China
| | - Fan Zhang
- Guangdong Provincial Key Laboratory for Breast Cancer Diagnosis and Treatment, Cancer Hospital of Shantou University Medical College, Shantou, China
| | - Jian-Ling Zhu
- Department of Pathology, Cancer Hospital of Shantou University Medical College, Shantou, China
| | - Xiao-Long Wei
- Department of Pathology, Cancer Hospital of Shantou University Medical College, Shantou, China
| |
Collapse
|
16
|
Zeng YZ, Zhang YQ, Chen JY, Zhang LY, Gao WL, Lin XQ, Huang SM, Zhang F, Wei XL. TRPC1 Inhibits Cell Proliferation/Invasion and Is Predictive of a Better Prognosis of Esophageal Squamous Cell Carcinoma. Front Oncol 2021; 11:627713. [PMID: 33854967 PMCID: PMC8039442 DOI: 10.3389/fonc.2021.627713] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Accepted: 03/08/2021] [Indexed: 02/05/2023] Open
Abstract
BACKGROUND AND OBJECTIVES In China, over 90% of esophageal cancer (EC) cases are esophageal squamous cell carcinoma (ESCC). ESCC is a frequently malignant tumor with poor prognosis despite the development of comprehensive therapeutic strategies, for which there is still a lack of effective prognostic factors. Previous studies found that the abnormal expression of TRPC1 is closely related to the proliferation, invasion, metastasis, and differentiation of various tumors. However, the relationship between TRPC1 and ESCC is currently unclear. The present study aimed to clarify the clinical significance of TRPC1 and to preliminarily assess the molecular mechanism by which TRPC1 regulates cell proliferation, migration, and invasion in ESCC. MATERIALS AND METHODS Immunohistochemistry (IHC) was used to determine the expression of TRPC1 and Ki-67 in 165 cases of ESCC. The correlations between TRPC1 expression and clinicopathological characteristics were determined, and both univariate and multivariate analyses were utilized to quantify the impact of TRPC1 expression on patient survival. Cell Counting Kit-8, scratch wound healing, and transwell assays were used to determine the effects of TRPC1 on proliferation, migration, and invasion in ESCC in vitro, respectively. RESULTS The positive expression rate of TRPC1 showed significantly decreased in ESCC (45.50%) compared with the levels in normal esophageal mucosa (NEM; 80.80%) and high-grade intraepithelial neoplasia (HGIEN; 63.20%) (P<0.001). Higher expression rate of TRPC1 was associated with low lymph node metastasis (P<0.001), high differentiation (rs = 0.232, P=0.003), and low Ki-67 (rs = -0.492, P<0.001). We further revealed that low expression of TRPC1 was associated with poor prognosis (Disease-free survival, DFS: 95% CI=0.545-0.845, P=0.001; Overall survival, OS: 95% CI=0.553-0.891, P=0.004). Furthermore, we showed that downregulation of TRPC1 promoted the proliferation, migration, and invasion of human esophageal squamous cell carcinoma cell line EC9706 in vitro. In contrast, overexpression of TRPC1 inhibited the proliferation, migration, and invasion of human esophageal squamous cell carcinoma cell line KYSE150 (P<0.01), in a manner at least in part mediated through the AKT/p27 pathway. CONCLUSION TRPC1 inhibited the proliferation, migration, and invasion of EC9706 and KYSE150 cells, at least, in part mediated through the AKT/p27 pathway in vitro. The downregulation of TRPC1 may be one of the most important molecular events in the malignant progression of ESCC. TRPC1 could be a new candidate tumor suppressor gene and a new prognostic factor of ESCC.
Collapse
Affiliation(s)
- Yun-Zhu Zeng
- Department of Pathology, Cancer Hospital of Shantou University Medical College, Shantou, China
| | - Yong-Qu Zhang
- Department of Breast-Thyroid-Surgery and Cancer Research Center, Xiang’an Hospital of Xiamen University, Xiamen, China
| | - Jiong-Yu Chen
- Oncological Research Laboratory, Cancer Hospital of Shantou University Medical College, Shantou, China
| | - Li-Ying Zhang
- Department of Pathology, Cancer Hospital of Shantou University Medical College, Shantou, China
| | - Wen-Liang Gao
- Department of Breast-Thyroid-Surgery and Cancer Research Center, Xiang’an Hospital of Xiamen University, Xiamen, China
| | - Xue-Qiong Lin
- Clinical Laboratory, Cancer Hospital of Shantou University Medical College, Shantou, China
| | - Shao-Min Huang
- Department of Pathology, Cancer Hospital of Shantou University Medical College, Shantou, China
| | - Fan Zhang
- Guangdong Provincial Key Laboratory for Breast Cancer Diagnosis and Treatment, Cancer Hospital of Shantou University Medical College, Shantou, China
| | - Xiao-Long Wei
- Department of Pathology, Cancer Hospital of Shantou University Medical College, Shantou, China
- *Correspondence: Xiao-Long Wei,
| |
Collapse
|
17
|
Qi T, Cao H, Sun H, Feng H, Li N, Wang C, Wang L. piR-19166 inhibits migration and metastasis through CTTN/MMPs pathway in prostate carcinoma. Aging (Albany NY) 2020; 12:18209-18220. [PMID: 32881713 PMCID: PMC7585067 DOI: 10.18632/aging.103677] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2020] [Accepted: 05/27/2020] [Indexed: 01/24/2023]
Abstract
Tumor metastasis is one of death causes for patients of prostate carcinoma. PIWI-interacting RNAs (piRNAs) are a subtype of noncoding protein RNAs that are involved in tumorigenesis, but the effect of piRNAs in prostate carcinoma (PCa) remains unclear. This article showed the identification of piRNAs was performed using a piRNA microarray screen in PCa tissues and several piRNAs were identified as dysregulated. The two up-regulated piRNAs (piR-19004 and piR-2878) and one down-regulated piR-19166 have been validated in the tissues and cell lines of PCa using quantitative reverse transcription polymerase chain reaction (qRT-PCR). Further studies showed that piR-19166 is transfected into PCa cells to suppress its migration and metastasis. Mechanistically, cortactin (CTTN) 3' untranslated region (UTR) was complementary combined with piR-19166 by bioinformatic prediction and identified as a direct target of piR-19166 through dual-luciferase reporter assay. Over-expression and knockdown of CTTN could respectively rescue and simulate the effects induced by piR-19166. Finally, piR-19166 suppresses migration and metastasis by the CTTN/matrix metalloproteinases (MMPs) pathway in PCa cells. Thus, these findings suggested that piR-19166 targets the CTTN of prostate cancer cells to inhibit migration and distant metastasis, and may represent a new marker of diagnosis and treatment for PCa patients in early stages.
Collapse
Affiliation(s)
- Tingyue Qi
- Department of Ultrasound, Medical Imaging Center, Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou 225009, China
| | - Haiyan Cao
- Department of Ultrasound, Medical Imaging Center, Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou 225009, China
| | - Hongguang Sun
- Department of Ultrasound, Medical Imaging Center, Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou 225009, China
| | - Hao Feng
- Department of Ultrasound, Medical Imaging Center, Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou 225009, China
| | - Nianfeng Li
- Department of Ultrasound, Medical Imaging Center, Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou 225009, China
| | - Chenghai Wang
- Department of Pathology, Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou 225009, China
| | - Lei Wang
- Department of Pathology, Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou 225009, China
| |
Collapse
|
18
|
Rivello F, Matuła K, Piruska A, Smits M, Mehra N, Huck WTS. Probing single-cell metabolism reveals prognostic value of highly metabolically active circulating stromal cells in prostate cancer. SCIENCE ADVANCES 2020; 6:6/40/eaaz3849. [PMID: 32998889 PMCID: PMC7527228 DOI: 10.1126/sciadv.aaz3849] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/04/2019] [Accepted: 08/06/2020] [Indexed: 05/05/2023]
Abstract
Despite their important role in metastatic disease, no general method to detect circulating stromal cells (CStCs) exists. Here, we present the Metabolic Assay-Chip (MA-Chip) as a label-free, droplet-based microfluidic approach allowing single-cell extracellular pH measurement for the detection and isolation of highly metabolically active cells (hm-cells) from the tumor microenvironment. Single-cell mRNA-sequencing analysis of the hm-cells from metastatic prostate cancer patients revealed that approximately 10% were canonical EpCAM+ hm-CTCs, 3% were EpCAM- hm-CTCs with up-regulation of prostate-related genes, and 87% were hm-CStCs with profiles characteristic for cancer-associated fibroblasts, mesenchymal stem cells, and endothelial cells. Kaplan-Meier analysis shows that metastatic prostate cancer patients with more than five hm-cells have a significantly poorer survival probability than those with zero to five hm-cells. Thus, prevalence of hm-cells is a prognosticator of poor outcome in prostate cancer, and a potentially predictive and therapy response biomarker for agents cotargeting stromal components and preventing epithelial-to-mesenchymal transition.
Collapse
Affiliation(s)
- Francesca Rivello
- Institute for Molecules and Materials, Radboud University, Heyendaalseweg 135, 6525 AJ Nijmegen, the Netherlands
| | - Kinga Matuła
- Institute for Molecules and Materials, Radboud University, Heyendaalseweg 135, 6525 AJ Nijmegen, the Netherlands
| | - Aigars Piruska
- Institute for Molecules and Materials, Radboud University, Heyendaalseweg 135, 6525 AJ Nijmegen, the Netherlands
| | - Minke Smits
- Department of Medical Oncology, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Niven Mehra
- Department of Medical Oncology, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Wilhelm T S Huck
- Institute for Molecules and Materials, Radboud University, Heyendaalseweg 135, 6525 AJ Nijmegen, the Netherlands.
| |
Collapse
|
19
|
Barczak W, Jin L, Carr SM, Munro S, Ward S, Kanapin A, Samsonova A, La Thangue NB. PRMT5 promotes cancer cell migration and invasion through the E2F pathway. Cell Death Dis 2020; 11:572. [PMID: 32709847 PMCID: PMC7382496 DOI: 10.1038/s41419-020-02771-9] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2020] [Revised: 07/04/2020] [Accepted: 07/09/2020] [Indexed: 12/31/2022]
Abstract
The pRb-E2F pathway is a critical point of regulation in the cell cycle and loss of control of the pathway is a hallmark of cancer. E2F1 is the major target through which pRb exerts its effects and arginine methylation by PRMT5 plays a key role in dictating E2F1 activity. Here we have explored the functional role of the PRMT5-E2F1 axis and highlight its influence on different aspects of cancer cell biology including viability, migration, invasion and adherence. Through a genome-wide expression analysis, we identified a distinct set of genes under the control of PRMT5 and E2F1, including some highly regulated genes, which influence cell migration, invasio and adherence through a PRMT5-dependent mechanism. Most significantly, a coincidence was apparent between the expression of PRMT5 and E2F1 in human tumours, and elevated levels of PRMT5 and E2F1 correlated with poor prognosis disease. Our results suggest a causal relationship between PRMT5 and E2F1 in driving the malignant phenotype and thereby highlight an important pathway for therapeutic intervention.
Collapse
Affiliation(s)
- Wojciech Barczak
- Laboratory of Cancer Biology Department of Oncology, University of Oxford, Old Road Campus Research Building, Oxford, OX3 7DQ, UK
| | - Li Jin
- Laboratory of Cancer Biology Department of Oncology, University of Oxford, Old Road Campus Research Building, Oxford, OX3 7DQ, UK
| | - Simon Mark Carr
- Laboratory of Cancer Biology Department of Oncology, University of Oxford, Old Road Campus Research Building, Oxford, OX3 7DQ, UK
| | - Shonagh Munro
- Argonaut Therapeutics Ltd Magdalen Centre, Oxford Science Park, Oxford, OX4 4GA, UK
| | - Samuel Ward
- Argonaut Therapeutics Ltd Magdalen Centre, Oxford Science Park, Oxford, OX4 4GA, UK
| | - Alexander Kanapin
- Centre for Genome Bioinformatics, St. Petersburg State University, St. Petersburg, 199034, Russia
| | - Anastasia Samsonova
- Centre for Genome Bioinformatics, St. Petersburg State University, St. Petersburg, 199034, Russia
| | - Nicholas B La Thangue
- Laboratory of Cancer Biology Department of Oncology, University of Oxford, Old Road Campus Research Building, Oxford, OX3 7DQ, UK.
| |
Collapse
|
20
|
Yu Y, Cao J, Wu W, Zhu Q, Tang Y, Zhu C, Dai J, Li Z, Wang J, Xue L, Zhen F, Liu J, Huang C, Zhao F, Zhou Y, Wen W, Pan X, Wei H, Zhu Y, He Y, Que J, Wang W, Luo J, Xu J, Chen L. Genome-wide copy number variation analysis identified ANO1 as a novel oncogene and prognostic biomarker in esophageal squamous cell cancer. Carcinogenesis 2020; 40:1198-1208. [PMID: 31050728 DOI: 10.1093/carcin/bgz077] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2018] [Revised: 03/27/2019] [Accepted: 04/22/2019] [Indexed: 12/11/2022] Open
Abstract
Copy number variations (CNVs) represent one of the most common genomic alterations. This study aimed to evaluate the roles of genes within highly aberrant genome regions in the prognosis of esophageal squamous cell cancer (ESCC). Exome sequencing data from 81 paired ESCC tissues were used to screen aberrant genomic regions. The associations between CNVs and gene expression were evaluated using gene expression data from the same individuals. Then, an RNA expression array profile from 119 ESCC samples was adopted for differential gene expression and prognostic analyses. Two independent ESCC cohorts with 315 subjects were further recruited to validate the prognostic value using immunohistochemistry tests. Finally, we explored the potential mechanism of our identified novel oncogene in ESCC. In total, 2003 genes with CNVs were observed, of which 76 genes showed recurrent CNVs in more than three samples. Among them, 32 genes were aberrantly expressed in ESCC tumor tissues and statistically correlated with CNVs. Strikingly, 4 (CTTN, SHANK2, INPPL1 and ANO1) of the 32 genes were significantly associated with the prognosis of ESCC patients. Patients with a positive expression of ANO1 had a poorer prognosis than ANO1 negative patients (overall survival rate: 42.91% versus 26.22% for ANO1-/+ samples, P < 0.001). Functionally, ANO1 promoted ESCC cell proliferation, migration and invasion by activating transforming growth factor-β pathway. Knockdown of ANO1 significantly inhibited tumor progression in vitro and in vivo. In conclusion, ANO1 is a novel oncogene in ESCC and may serve as a prognostic biomarker for ESCC.
Collapse
Affiliation(s)
- Yue Yu
- Department of Thoracic Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China.,Department of Thoracic Surgery, Cancer Institute and Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | - Jing Cao
- Department of Thoracic Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Weibing Wu
- Department of Thoracic Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Quan Zhu
- Department of Thoracic Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Yu Tang
- Department of Thoracic Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Chengxiang Zhu
- Department of Thoracic Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Juncheng Dai
- Department of Epidemiology and Biostatistics, Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Medicine, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Zhihua Li
- Department of Thoracic Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China.,Department of Epidemiology and Biostatistics, Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Medicine, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Jun Wang
- Department of Thoracic Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Lei Xue
- Department of Thoracic Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Fuxi Zhen
- Department of Thoracic Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Jinyuan Liu
- Department of Thoracic Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Chenjun Huang
- Department of Thoracic Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Fei Zhao
- Department of Thoracic Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Yue Zhou
- Department of Thoracic Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Wei Wen
- Department of Thoracic Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Xianglong Pan
- Department of Thoracic Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Haixing Wei
- Department of Thoracic Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Yining Zhu
- Department of Thoracic Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Yaozhou He
- Department of Thoracic Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Jun Que
- Department of Thoracic Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Wei Wang
- Department of Thoracic Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Jinhua Luo
- Department of Thoracic Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Jing Xu
- Department of Thoracic Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Liang Chen
- Department of Thoracic Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| |
Collapse
|
21
|
Zhao Y, Lei Y, He SW, Li YQ, Wang YQ, Hong XH, Liang YL, Li JY, Chen Y, Luo WJ, Zhang PP, Yang XJ, He QM, Ma J, Liu N, Tang LL. Hypermethylation of UCHL1 Promotes Metastasis of Nasopharyngeal Carcinoma by Suppressing Degradation of Cortactin (CTTN). Cells 2020; 9:E559. [PMID: 32120844 PMCID: PMC7140450 DOI: 10.3390/cells9030559] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2020] [Revised: 02/15/2020] [Accepted: 02/26/2020] [Indexed: 12/25/2022] Open
Abstract
Epigenetic regulation plays an important role in the development and progression of nasopharyngeal carcinoma (NPC), but the epigenetic mechanisms underlying NPC metastasis remain poorly understood. Here, we demonstrate that hypermethylation of the UCHL1 promoter leads to its downregulation in NPC. Restoration of UCHL1 inhibited the migration and invasion of NPC cells in vitro and in vivo, and knockdown of UCHL1 promoted NPC cell migration and invasion in vitro and in vivo. Importantly, we found that UCHL1 interacts with CTTN, and may function as a ligase promoting CTTN degradation by increasing K48-linked ubiquitination of CTTN. Additionally, restoration of CTTN in NPC cells that overexpressed UCHL1 rescued UCHL1 suppressive effects on NPC cell migration and invasion, which indicated that CTTN is a functional target of UCHL1 in NPC. Our findings revealed that UCHL1 acts as a tumor suppressor gene in NPC and thus provided a novel therapeutic target for NPC treatment.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | - Ling-Long Tang
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center of Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangzhou 510060, China; (Y.Z.); (Y.L.); (S.-W.H.); (Y.-Q.L.); (Y.-Q.W.); (X.-H.H.); (Y.-L.L.); (J.-Y.L.); (Y.C.); (W.-J.L.); (P.-P.Z.); (X.-J.Y.); (Q.-M.H.); (J.M.); (N.L.)
| |
Collapse
|
22
|
Peng L, Guo JC, Long L, Pan F, Zhao JM, Xu LY, Li EM. A Novel Clinical Six-Flavoprotein-Gene Signature Predicts Prognosis in Esophageal Squamous Cell Carcinoma. BIOMED RESEARCH INTERNATIONAL 2019; 2019:3869825. [PMID: 31815134 PMCID: PMC6878914 DOI: 10.1155/2019/3869825] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/04/2019] [Revised: 08/23/2019] [Accepted: 10/04/2019] [Indexed: 02/05/2023]
Abstract
Flavoproteins and their interacting proteins play important roles in mitochondrial electron transport, fatty acid degradation, and redox regulation. However, their clinical significance and function in esophageal squamous cell carcinoma (ESCC) are little known. Here, using survival analysis and machine learning, we mined 179 patient expression profiles with ESCC in GSE53625 from the Gene Expression Omnibus (GEO) database and constructed a signature consisting of two flavoprotein genes (GPD2 and PYROXD2) and four flavoprotein interacting protein genes (CTTN, GGH, SRC, and SYNJ2BP). Kaplan-Meier analysis revealed the signature was significantly associated with the survival of ESCC patients (mean survival time: 26.77 months in the high-risk group vs. 54.97 months in the low-risk group, P < 0.001, n = 179), and time-dependent ROC analysis demonstrated that the six-gene signature had good predictive ability for six-year survival for ESCC (AUC = 0.86, 95% CI: 0.81-0.90). We then validated its prediction performance in an independent set by RT-PCR (mean survival: 15.73 months in the high-risk group vs. 21.1 months in the low-risk group, P=0.032, n = 121). Furthermore, RNAi-mediated knockdown of genes in the flavoprotein signature led to decreased proliferation and migration of ESCC cells. Taken together, CTTN, GGH, GPD2, PYROXD2, SRC, and SYNJ2BP have an important clinical significance for prognosis of ESCC patients, suggesting they are efficient prognostic markers and potential targets for ESCC therapy.
Collapse
Affiliation(s)
- Liu Peng
- The Key Laboratory of Molecular Biology for High Cancer Incidence Coastal Chaoshan Area, Shantou University Medical College, Shantou 515041, Guangdong, China
- Department of Biochemistry and Molecular Biology, Shantou University Medical College, Shantou 515041, Guangdong, China
| | - Jin-Cheng Guo
- The Key Laboratory of Molecular Biology for High Cancer Incidence Coastal Chaoshan Area, Shantou University Medical College, Shantou 515041, Guangdong, China
- Department of Biochemistry and Molecular Biology, Shantou University Medical College, Shantou 515041, Guangdong, China
| | - Lin Long
- The Key Laboratory of Molecular Biology for High Cancer Incidence Coastal Chaoshan Area, Shantou University Medical College, Shantou 515041, Guangdong, China
- Department of Biochemistry and Molecular Biology, Shantou University Medical College, Shantou 515041, Guangdong, China
| | - Feng Pan
- The Key Laboratory of Molecular Biology for High Cancer Incidence Coastal Chaoshan Area, Shantou University Medical College, Shantou 515041, Guangdong, China
- Department of Biochemistry and Molecular Biology, Shantou University Medical College, Shantou 515041, Guangdong, China
| | - Jian-Mei Zhao
- The Key Laboratory of Molecular Biology for High Cancer Incidence Coastal Chaoshan Area, Shantou University Medical College, Shantou 515041, Guangdong, China
- Department of Biochemistry and Molecular Biology, Shantou University Medical College, Shantou 515041, Guangdong, China
| | - Li-Yan Xu
- The Key Laboratory of Molecular Biology for High Cancer Incidence Coastal Chaoshan Area, Shantou University Medical College, Shantou 515041, Guangdong, China
- Institute of Oncologic Pathology, Shantou University Medical College, Shantou 515041, Guangdong, China
| | - En-Min Li
- The Key Laboratory of Molecular Biology for High Cancer Incidence Coastal Chaoshan Area, Shantou University Medical College, Shantou 515041, Guangdong, China
- Department of Biochemistry and Molecular Biology, Shantou University Medical College, Shantou 515041, Guangdong, China
| |
Collapse
|
23
|
Cortactin as a potential predictor of second esophageal neoplasia in hypopharyngeal carcinoma. Auris Nasus Larynx 2018; 46:260-266. [PMID: 30107961 DOI: 10.1016/j.anl.2018.08.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2018] [Revised: 06/22/2018] [Accepted: 08/01/2018] [Indexed: 12/20/2022]
Abstract
OBJECTIVE Hypopharyngeal carcinoma has a very poor prognosis. The high incidence of second esophageal neoplasia is one of the major causes. To establish an efficient follow-up scheme for increasing the diagnostic yield and reducing the adverse impact of second esophageal neoplasia on survival, the purpose of this study was to explore a biomarker to predict second esophageal neoplasia. METHODS In this retrospective cohort study, consecutive tissue specimens from those patients who underwent tumor resection between September 2007 and October 2015 were collected. Gene amplification was performed by real-time PCR. The expression of cortactin was evaluated by immunohistochemistry. The predictive risk factors of developing second esophageal neoplasia and prognostic factors related to survival were analyzed. RESULTS A total of 187 patients were included with a mean follow-up of 48months (12-118months). Second esophageal tumors were found in 53 (28.3%), including 41 (21.9%) esophageal squamous cell carcinoma and 12 severe dysplasia. The results of multivariate analyses revealed that age (OR 2.81, 95% CI 1.16-6.78), cortactin overexpression (OR 2.49, 95% CI 1.17-5.33), and stage IV versus I (OR 6.49, 95% CI 1.68-25.18) were independent predictors of second esophageal neoplasia, and second esophageal neoplasia (HR 1.78, 95% CI 1.05-3.01) was an independent predictor of overall survival. CONCLUSION This is the first report to identify a potential biomarker for predicting second esophageal neoplasia in patients with hypopharyngeal carcinoma. In those patients with cortactin overexpression and younger age (≤60years old), close surveillance for second esophageal neoplasia is required. In addition, the real effect of cortactin overexpression on development of primary esophageal carcinoma is required to be validated in a large cohort study.
Collapse
|
24
|
Villaronga MÁ, Hermida-Prado F, Granda-Díaz R, Menéndez ST, Álvarez-Teijeiro S, Quer M, Vilaseca I, Allonca E, Garzón-Arango M, Sanz-Moreno V, Astudillo A, Rodrigo JP, García-Pedrero JM. Immunohistochemical Expression of Cortactin and Focal Adhesion Kinase Predicts Recurrence Risk and Laryngeal Cancer Risk Beyond Histologic Grading. Cancer Epidemiol Biomarkers Prev 2018; 27:805-813. [PMID: 29654156 DOI: 10.1158/1055-9965.epi-17-1082] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2017] [Revised: 01/02/2018] [Accepted: 04/09/2018] [Indexed: 11/16/2022] Open
Abstract
Background: Cortactin (CTTN) and the focal adhesion kinase (FAK) are two major candidate genes to, respectively, drive 11q13- and 8q24-associated aggressive behavior in various cancers. Recent evidence uncovered their clinical relevance in early stages of tumorigenesis as promising biomarkers for cancer risk assessment.Methods: Using a multicenter validation study, CTTN and FAK expression was evaluated by immunohistochemistry (IHC) in a cohort of 109 patients with laryngeal precancerous lesions, and correlated with clinicopathologic parameters and laryngeal cancer risk. The pathophysiologic role of CTTN and FAK was further investigated using functional studies in cellular models.Results: Positive CTTN and FAK expression (scores 2 and 3) was detected in 49 (41%) and 35 (32%) laryngeal dysplasias, respectively. Univariate Cox analysis showed that CTTN and FAK expression but not histologic grading was significantly associated with both recurrence risk and laryngeal cancer risk. Patients carrying strong CTTN- or FAK-expressing lesions (score 3) experienced the highest laryngeal cancer incidence (log-rank P < 0.001). In multivariate stepwise analysis, FAK expression [HR = 13.91; 95% CI, 4.82-40.15; P < 0.001] and alcohol consumption (HR = 2.22; 95% confidence interval, 1.17-4.20; P = 0.014) were significant independent predictors of laryngeal cancer development. Targeting FAK by either RNAi or pharmacologic inhibitors effectively blocked cell growth, colony formation, and invasion into 3D collagen matrices.Conclusions: CTTN and FAK emerge as powerful predictors of laryngeal cancer risk and recurrence risk beyond histologic grading.Impact: Our work supports the applicability of IHC CTTN and FAK as complementary markers for risk stratification in patients with laryngeal precancerous lesions. Cancer Epidemiol Biomarkers Prev; 27(7); 805-13. ©2018 AACR.
Collapse
Affiliation(s)
- M Ángeles Villaronga
- Department of Otolaryngology, Hospital Universitario Central de Asturias and Instituto Universitario de Oncología del Principado de Asturias, University of Oviedo, CIBERONC, Oviedo, Spain.
| | - Francisco Hermida-Prado
- Department of Otolaryngology, Hospital Universitario Central de Asturias and Instituto Universitario de Oncología del Principado de Asturias, University of Oviedo, CIBERONC, Oviedo, Spain
| | - Rocío Granda-Díaz
- Department of Otolaryngology, Hospital Universitario Central de Asturias and Instituto Universitario de Oncología del Principado de Asturias, University of Oviedo, CIBERONC, Oviedo, Spain
| | - Sofía T Menéndez
- Department of Otolaryngology, Hospital Universitario Central de Asturias and Instituto Universitario de Oncología del Principado de Asturias, University of Oviedo, CIBERONC, Oviedo, Spain
| | - Saúl Álvarez-Teijeiro
- Department of Otolaryngology, Hospital Universitario Central de Asturias and Instituto Universitario de Oncología del Principado de Asturias, University of Oviedo, CIBERONC, Oviedo, Spain
| | - Miquel Quer
- Department of Otolaryngology, Hospital Santa Creu i Sant Pau, Barcelona, Spain
| | - Isabel Vilaseca
- Department of Otolaryngology, Hospital Clínic, Barcelona, Spain
| | - Eva Allonca
- Department of Otolaryngology, Hospital Universitario Central de Asturias and Instituto Universitario de Oncología del Principado de Asturias, University of Oviedo, CIBERONC, Oviedo, Spain
| | - Marta Garzón-Arango
- Department of Otolaryngology, Hospital Universitario Central de Asturias and Instituto Universitario de Oncología del Principado de Asturias, University of Oviedo, CIBERONC, Oviedo, Spain
| | - Victoria Sanz-Moreno
- Tumour Plasticity Laboratory, Randall Division of Cell and Molecular Biophysics, Guy's Campus, King's College London, London, United Kingdom
| | - Aurora Astudillo
- Department of Pathology, Hospital Universitario Central de Asturias, Oviedo, Spain
| | - Juan P Rodrigo
- Department of Otolaryngology, Hospital Universitario Central de Asturias and Instituto Universitario de Oncología del Principado de Asturias, University of Oviedo, CIBERONC, Oviedo, Spain.
| | - Juana M García-Pedrero
- Department of Otolaryngology, Hospital Universitario Central de Asturias and Instituto Universitario de Oncología del Principado de Asturias, University of Oviedo, CIBERONC, Oviedo, Spain.
| |
Collapse
|
25
|
Ribeiro IP, Rodrigues JM, Mascarenhas A, Kosyakova N, Caramelo F, Liehr T, Melo JB, Carreira IM. Cytogenetic, genomic, and epigenetic characterization of the HSC-3 tongue cell line with lymph node metastasis. J Oral Sci 2018; 60:70-81. [PMID: 29479029 DOI: 10.2334/josnusd.16-0811] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/01/2022]
Abstract
Oral carcinoma develops from squamous epithelial cells by the acquisition of multiple (epi) genetic alterations that target different genes and molecular pathways. Herein, we performed a comprehensive genomic and epigenetic characterization of the HSC-3 cell line through karyotyping, multicolor fluorescence in situ hybridization, array comparative genomic hybridization, and methylation-specific multiplex ligation-dependent probe amplification. HSC-3 turned out to be a near-triploid cell line with a modal number of 61 chromosomes. Banding and molecular cytogenetic analyses revealed that nonrandom gains of chromosomal segments occurred more frequently than losses. Overall, gains of chromosome 1, 3q, 5p, 7p, 8q, 9q, 10, 11p, 11q13, 12, 13, 14, 17, 18p, 20, Yp, and Xq were observed. The largest region affected by copy number loss was observed at chromosome 18q. Several of the observed genomic imbalances and their mapped genes were already associated with oral carcinoma and/or adverse prognosis, invasion, and metastasis in cancer. The most common rearrangements observed were translocations in the centromeric/near-centromeric regions. RARB, ESR1, and CADM1 genes were methylated and showed copy number losses, whereas TP73 and GATA5 presented with methylation and copy number gains. Thus, the current study presents a comprehensive characterization of the HSC-3 cell line; the use of this cell line may contribute to enriching the resources available for oral cancer research, especially for the testing of therapeutic agents.
Collapse
Affiliation(s)
- Ilda P Ribeiro
- Cytogenetics and Genomics Laboratory, Faculty of Medicine, University of Coimbra.,Center of Investigation on Environment Genetics and Oncobiology, Faculty of Medicine, University of Coimbra
| | - Joana M Rodrigues
- Cytogenetics and Genomics Laboratory, Faculty of Medicine, University of Coimbra
| | | | - Nadezda Kosyakova
- Institute of Human Genetics, Jena University Hospital, Friedrich Schiller University
| | - Francisco Caramelo
- Laboratory of Biostatistics and Medical Informatics, Faculty of Medicine, University of Coimbra
| | - Thomas Liehr
- Institute of Human Genetics, Jena University Hospital, Friedrich Schiller University
| | - Joana B Melo
- Cytogenetics and Genomics Laboratory, Faculty of Medicine, University of Coimbra.,Center of Investigation on Environment Genetics and Oncobiology, Faculty of Medicine, University of Coimbra
| | - Isabel M Carreira
- Cytogenetics and Genomics Laboratory, Faculty of Medicine, University of Coimbra.,Center of Investigation on Environment Genetics and Oncobiology, Faculty of Medicine, University of Coimbra
| |
Collapse
|
26
|
Luo ML, Zhou Z, Sun L, Yu L, Sun L, Liu J, Yang Z, Ran Y, Yao Y, Hu H. An ADAM12 and FAK positive feedback loop amplifies the interaction signal of tumor cells with extracellular matrix to promote esophageal cancer metastasis. Cancer Lett 2018; 422:118-128. [PMID: 29476791 DOI: 10.1016/j.canlet.2018.02.031] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2017] [Revised: 02/16/2018] [Accepted: 02/19/2018] [Indexed: 02/06/2023]
Abstract
Esophageal squamous cell carcinomas (ESCCs) have a poor prognosis mostly due to early metastasis. To explore the early event of metastasis in ESCC, we established an in vitro selection model to mimic the interaction of tumor cells with extracellular matrix, through which a sub-line of ESCC cells with high invasive ability was generated. By comparing the gene expression profile of the highly invasive sub-line to that of the parental cells, ADAM12-L was identified as a candidate gene promoting ESCC cell invasion. Immunohistochemistry revealed that the ADAM12-L was overexpressed in human ESCC tissues, especially at cancer invasive edge, and ADAM12-L overexpression tightly correlated with increased metastasis and poor outcome of ESCC patients. Indeed, ADAM12-L knockdown reduced the invasion and metastasis of ESCC cells both in vitro and in vivo. Furthermore, we demonstrated that ADAM12-L participated in focal adhesion turnover and promoted the activation of focal adhesion kinase (FAK), which in turn increased ADAM12-L transcription through FAK/JNK/c-Jun axis. Therefore, a loop initiated from the cancer cell upon the engagement with extracellular matrix through FAK and c-Jun to enhance ADAM12-L expression is established, leading to the positive feedback of further FAK activation and prompting metastasis. Our study indicates that overexpression of ADAM12-L can serve as a precision marker to determine the activation of this loop. Targeting ADAM12-L to disrupt this positive feedback loop represents a promising strategy to treat the metastasis of esophageal cancers.
Collapse
Affiliation(s)
- Man-Li Luo
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, China; Medical Research Center, SunYat-Sen Memorial Hospital, SunYat-Sen University, Guangzhou 510120, China
| | - Zhuan Zhou
- State Key Laboratory of Molecular Oncology, Cancer Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing 100021, China
| | - Lichao Sun
- State Key Laboratory of Molecular Oncology, Cancer Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing 100021, China
| | - Long Yu
- State Key Laboratory of Molecular Oncology, Cancer Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing 100021, China
| | - Lixin Sun
- State Key Laboratory of Molecular Oncology, Cancer Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing 100021, China
| | - Jun Liu
- State Key Laboratory of Molecular Oncology, Cancer Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing 100021, China
| | - Zhihua Yang
- State Key Laboratory of Molecular Oncology, Cancer Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing 100021, China
| | - Yuliang Ran
- State Key Laboratory of Molecular Oncology, Cancer Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing 100021, China
| | - Yandan Yao
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, China; Breast Tumor Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, China.
| | - Hai Hu
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, China; Department of Oncology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, China.
| |
Collapse
|
27
|
Wu MH, Luo JD, Wang WC, Chang TH, Hwang WL, Lee KH, Liu SY, Yang JW, Chiou CT, Chang CH, Chiang WF. Risk analysis of malignant potential of oral verrucous hyperplasia: A follow-up study of 269 patients and copy number variation analysis. Head Neck 2018; 40:1046-1056. [DOI: 10.1002/hed.25076] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2017] [Revised: 09/26/2017] [Accepted: 12/06/2017] [Indexed: 12/28/2022] Open
Affiliation(s)
- Ming-Heng Wu
- Graduate Institute of Translational Medicine, College of Medical Science and Technology; Taipei Medical University; Taipei Taiwan
| | - Ji-Dung Luo
- Bioinformatics Core Laboratory, Molecular Medicine Research Center; Chang Gung University; Taoyuan Taiwan
| | - Wen-Chang Wang
- Graduate Institute of Translational Medicine, College of Medical Science and Technology; Taipei Medical University; Taipei Taiwan
| | - Tzu-Hao Chang
- Graduate Institute of Biomedical Informatics, College of Medical Science and Technology; Taipei Medical University; Taipei Taiwan
| | - Wei-Lun Hwang
- Graduate Institute of Translational Medicine, College of Medical Science and Technology; Taipei Medical University; Taipei Taiwan
| | - Kuen-Haur Lee
- Graduate Institute of Cancer Biology and Drug Discovery, College of Medical Science and Technology; Taipei Medical University; Taipei Taiwan
| | - Shyun-Yeu Liu
- Department of Oral and Maxillofacial Surgery, Chi-Mei Medical Center; Liouying Taiwan
| | - Jung-Wu Yang
- Department of Oral and Maxillofacial Surgery; Sin-Lau Hospital; Tainan Taiwan
| | - Chang-Ta Chiou
- Department of Oral and Maxillofacial Surgery; An-Nan Hospital; Tainan Taiwan
| | - Chi-Hua Chang
- Department of Oral and Maxillofacial Surgery; Chang-Chung Memorial Hospital; Kaohsiung Taiwan
| | - Wei-Fan Chiang
- Graduate Institute of Translational Medicine, College of Medical Science and Technology; Taipei Medical University; Taipei Taiwan
- Department of Oral and Maxillofacial Surgery, Chi-Mei Medical Center; Liouying Taiwan
- School of Dentistry; National Yang-Ming University; Taipei Taiwan
| |
Collapse
|
28
|
Lin DC, Wang MR, Koeffler HP. Genomic and Epigenomic Aberrations in Esophageal Squamous Cell Carcinoma and Implications for Patients. Gastroenterology 2018; 154:374-389. [PMID: 28757263 PMCID: PMC5951382 DOI: 10.1053/j.gastro.2017.06.066] [Citation(s) in RCA: 172] [Impact Index Per Article: 28.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/29/2017] [Revised: 06/05/2017] [Accepted: 06/07/2017] [Indexed: 12/28/2022]
Abstract
Esophageal squamous cell carcinoma (ESCC) is a common malignancy without effective therapy. The exomes of more than 600 ESCCs have been sequenced in the past 4 years, and numerous key aberrations have been identified. Recently, researchers reported both inter- and intratumor heterogeneity. Although these are interesting observations, their clinical implications are unclear due to the limited number of samples profiled. Epigenomic alterations, such as changes in DNA methylation, histone acetylation, and RNA editing, also have been observed in ESCCs. However, it is not clear what proportion of ESCC cells carry these epigenomic aberrations or how they contribute to tumor development. We review the genomic and epigenomic characteristics of ESCCs, with a focus on emerging themes. We discuss their clinical implications and future research directions.
Collapse
Affiliation(s)
- De-Chen Lin
- Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, California.
| | - Ming-Rong Wang
- State Key Laboratory of Molecular Oncology, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
| | - H Phillip Koeffler
- Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, California; Cancer Science Institute of Singapore, National University of Singapore, Singapore; National University Cancer Institute, National University Hospital Singapore, Singapore
| |
Collapse
|
29
|
Abstract
Actin remodeling plays an essential role in diverse cellular processes such as cell motility, vesicle trafficking or cytokinesis. The scaffold protein and actin nucleation promoting factor Cortactin is present in virtually all actin-based structures, participating in the formation of branched actin networks. It has been involved in the control of endocytosis, and vesicle trafficking, axon guidance and organization, as well as adhesion, migration and invasion. To migrate and invade through three-dimensional environments, cells have developed specialized actin-based structures called invadosomes, a generic term to designate invadopodia and podosomes. Cortactin has emerged as a critical regulator of invadosome formation, function and disassembly. Underscoring this role, Cortactin is frequently overexpressed in several types of invasive cancers. Herein we will review the roles played by Cortactin in these specific invasive structures.
Collapse
Affiliation(s)
- Pauline Jeannot
- CRCT INSERM UMR1037, Université Toulouse III Paul Sabatier , CNRS ERL5294, Toulouse, France.,Cell Signalling Group, Cancer Research UK Manchester Institute, The University of Manchester , Manchester M20 4BX, UK
| | - Arnaud Besson
- CRCT INSERM UMR1037, Université Toulouse III Paul Sabatier , CNRS ERL5294, Toulouse, France.,LBCMCP , Centre de Biologie Intégrative, Université de Toulouse , CNRS, UPS, Toulouse Cedex, France
| |
Collapse
|
30
|
Zhang Y, Zhang Y, Fan H, Shi Y, Zhan C, Wang Q. Study on the role of transient receptor potential C6 channels in esophageal squamous cell carcinoma radiosensitivity. J Thorac Dis 2017; 9:3802-3809. [PMID: 29268388 DOI: 10.21037/jtd.2017.09.108] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Background To study the effect of transient receptor potential C6 (TRPC6) channels on esophageal squamous cell carcinoma (ESCC) cell lines Eca109 cell cycle and to confirm whether TRPC6 channel is candidate radiosensitivity in vitro and in vivo. Methods We chose Eca109 cell line with a strong TRPC6 channels expression. Cell cycle was investigated after TRPC6 channel inhibitor SKF96365 treated with a 5 µM concentration. According to the results of cell cycle, radiation was performed. CCK-8 test was used to test the cell proliferation. Then we performed the same study in vivo. Total of 40 male nude mice were randomly divided into four groups as follows: SKF96365, radio, combined radio-SKF96365 and control. In SKF96365 group, 20 mg/kg 5 µM SKF96365 was injected into the abdominal cavity of the nude mice at day 5-11. In radiation group, the nude mice received radiotherapy 2 Gy per day at day 7-11. In combined radio-SKF96365 group, 20 mg/kg 5 µM SKF96365 was injected into the abdominal cavity of the nude mice at day 5-11 and 2 Gy radiotherapy was delivered to the tumor site at day 7-11. In control group, nude mice were injected saline into the abdominal cavity at day 5-11. General states of health were observed, the tumor size in volume was measured with calipers two times every week. Six weeks after seeding, mice were sacrificed by neck-break. The tumor size was measured in volume with caliper and in weigh with scale. Results Treatment with SKF96365 substantially increased the percentage of Eca109 cells in the G2/M phase and reduced that in G0/G1 phase in a time-dependent manner. Most of the cells (85.26%), 24 h after SKF96365 treatment were arrested in the G2/M phase. CCK-8 test showed that Eca109 ESCC cells received both SKF96365 and radiation showed the worst ability of cell proliferation. The same result was obtained in vivo. Nude mice received combined radio-SKF96365 showed the smallest tumor size and volume. Conclusions TRPC6 plays an important role in development of esophageal cancer, and SKF96365 may increase the sensitivity of radiotherapy. TRPC6 may become a new radiotherapy target in esophageal cancer.
Collapse
Affiliation(s)
- Yi Zhang
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Yongxing Zhang
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Hong Fan
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Yu Shi
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Cheng Zhan
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Qun Wang
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| |
Collapse
|
31
|
Yin M, Ma W, An L. Cortactin in cancer cell migration and invasion. Oncotarget 2017; 8:88232-88243. [PMID: 29152154 PMCID: PMC5675706 DOI: 10.18632/oncotarget.21088] [Citation(s) in RCA: 54] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2016] [Accepted: 08/29/2017] [Indexed: 12/20/2022] Open
Abstract
Cortactin, a substrate of sarcoma (Src) kinases, is an actin-binding protein that is involved in cytoskeletal regulation, and is frequently overexpressed in cancer cells. Binding to the actin related protein 2/3 (Arp2/3) complex stimulates cortactin activity, which promotes F-actin nucleation and assembly. Cortactin promotes cancer cell migration and invasion, and plays a pivotal role in invadopodia formation and extra cellular matrix degradation. Overexpression of cortactin, by amplification of the chromosomal band 11q13, increases tumor aggressiveness. In this review, we report on the current knowledge and potential mechanisms of action of cortactin as a critical mediator of cancer cell migration and invasion.
Collapse
Affiliation(s)
- Miao Yin
- Shandong Provincial Key Laboratory of Animal Resistance Biology, College of Life Sciences, Shandong Normal University, Jinan 250014, China
| | - Wenqing Ma
- Shandong Provincial Key Laboratory of Animal Resistance Biology, College of Life Sciences, Shandong Normal University, Jinan 250014, China
| | - Liguo An
- Shandong Provincial Key Laboratory of Animal Resistance Biology, College of Life Sciences, Shandong Normal University, Jinan 250014, China
| |
Collapse
|
32
|
Song Q, Jiang D, Wang H, Huang J, Liu Y, Xu C, Hou Y. Chromosomal and Genomic Variations in Esophageal Squamous Cell Carcinoma: A Review of Technologies, Applications, and Prospections. J Cancer 2017; 8:2492-2500. [PMID: 28900487 PMCID: PMC5595079 DOI: 10.7150/jca.19601] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2017] [Accepted: 05/30/2017] [Indexed: 12/14/2022] Open
Abstract
Esophageal squamous cell carcinoma (ESCC) is one of the most common malignant tumors with poor prognosis worldwide. The poor prognosis is due to the advanced stage at the time of diagnosis and the limited clinical staging lacking significant molecular biomarkers to effectively stratify patients for treatment options. As cancer is a disease of genome instability and a resulting of accumulation of genetic alteration, mounting chromosomal and genomic technologies were developed and progressed rapidly which could be used for characterizing patients in genomics level. In this review, we summarized applications of multiple technologies and research progress at chromosomal and genomic level in ESCC.
Collapse
Affiliation(s)
- Qi Song
- Department of Pathology, Zhongshan Hospital, Fudan University, Shanghai 200032, P. R. China
| | - Dongxian Jiang
- Department of Pathology, Zhongshan Hospital, Fudan University, Shanghai 200032, P. R. China
| | - Haixing Wang
- Department of Pathology, Zhongshan Hospital, Fudan University, Shanghai 200032, P. R. China
| | - Jie Huang
- Department of Pathology, Zhongshan Hospital, Fudan University, Shanghai 200032, P. R. China
| | - Yalan Liu
- Department of Pathology, Zhongshan Hospital, Fudan University, Shanghai 200032, P. R. China
| | - Chen Xu
- Department of Pathology, Zhongshan Hospital, Fudan University, Shanghai 200032, P. R. China
| | - Yingyong Hou
- Department of Pathology, Zhongshan Hospital, Fudan University, Shanghai 200032, P. R. China
- Department of Pathology, School of Basic Medical Sciences & Zhongshan Hospital, Fudan University, Shanghai 200032, P. R. China
| |
Collapse
|
33
|
Jiang YY, Lin DC, Mayakonda A, Hazawa M, Ding LW, Chien WW, Xu L, Chen Y, Xiao JF, Senapedis W, Baloglu E, Kanojia D, Shang L, Xu X, Yang H, Tyner JW, Wang MR, Koeffler HP. Targeting super-enhancer-associated oncogenes in oesophageal squamous cell carcinoma. Gut 2017; 66:1358-1368. [PMID: 27196599 PMCID: PMC5912916 DOI: 10.1136/gutjnl-2016-311818] [Citation(s) in RCA: 157] [Impact Index Per Article: 22.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/08/2016] [Revised: 04/11/2016] [Accepted: 04/20/2016] [Indexed: 12/12/2022]
Abstract
OBJECTIVES Oesophageal squamous cell carcinoma (OSCC) is an aggressive malignancy and the major histological subtype of oesophageal cancer. Although recent large-scale genomic analysis has improved the description of the genetic abnormalities of OSCC, few targetable genomic lesions have been identified, and no molecular therapy is available. This study aims to identify druggable candidates in this tumour. DESIGN High-throughput small-molecule inhibitor screening was performed to identify potent anti-OSCC compounds. Whole-transcriptome sequencing (RNA-Seq) and chromatin immunoprecipitation sequencing (ChIP-Seq) were conducted to decipher the mechanisms of action of CDK7 inhibition in OSCC. A variety of in vitro and in vivo cellular assays were performed to determine the effects of candidate genes on OSCC malignant phenotypes. RESULTS The unbiased high-throughput small-molecule inhibitor screening led us to discover a highly potent anti-OSCC compound, THZ1, a specific CDK7 inhibitor. RNA-Seq revealed that low-dose THZ1 treatment caused selective inhibition of a number of oncogenic transcripts. Notably, further characterisation of the genomic features of these THZ1-sensitive transcripts demonstrated that they were frequently associated with super-enhancer (SE). Moreover, SE analysis alone uncovered many OSCC lineage-specific master regulators. Finally, integrative analysis of both THZ1-sensitive and SE-associated transcripts identified a number of novel OSCC oncogenes, including PAK4, RUNX1, DNAJB1, SREBF2 and YAP1, with PAK4 being a potential druggable kinase. CONCLUSIONS Our integrative approaches led to a catalogue of SE-associated master regulators and oncogenic transcripts, which may significantly promote both the understanding of OSCC biology and the development of more innovative therapies.
Collapse
Affiliation(s)
- Yan-Yi Jiang
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, Singapore
| | - De-Chen Lin
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, Singapore
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Anand Mayakonda
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, Singapore
| | - Masaharu Hazawa
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, Singapore
| | - Ling-Wen Ding
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, Singapore
| | - Wen-Wen Chien
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, Singapore
| | - Liang Xu
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, Singapore
| | - Ye Chen
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, Singapore
| | - Jin-Fen Xiao
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, Singapore
| | - William Senapedis
- Department of Drug Discovery, Karyopharm Therapeutics Inc., Newton, Massachusetts, USA
| | - Erkan Baloglu
- Department of Drug Discovery, Karyopharm Therapeutics Inc., Newton, Massachusetts, USA
| | - Deepika Kanojia
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, Singapore
| | - Li Shang
- State Key Laboratory of Molecular Oncology, Cancer Institute (Hospital), Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | - Xin Xu
- State Key Laboratory of Molecular Oncology, Cancer Institute (Hospital), Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | - Henry Yang
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, Singapore
| | - Jeffrey W Tyner
- Department of Cell, Developmental & Cancer Biology, Knight Cancer Institute, Oregon Health & Science University, Portland, Oregon, USA
| | - Ming-Rong Wang
- State Key Laboratory of Molecular Oncology, Cancer Institute (Hospital), Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | - H Phillip Koeffler
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, Singapore
- Division of Hematology/Oncology, Cedars-Sinai Medical Center, University of California, Los Angeles School of Medicine, Los Angeles, California, USA
- National University Cancer Institute, National University Health System and National University of Singapore, Singapore, Singapore
| |
Collapse
|
34
|
Cortactin and phosphorylated cortactin tyr421 and tyr466 expression in supraglottic laryngeal carcinomas and lymph node metastases. Int J Biol Markers 2017; 33:79-86. [DOI: 10.5301/ijbm.5000297] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Background: The most important adverse prognostic factor for laryngeal squamous cell carcinoma (LSCC) is the presence of cervical lymph node metastases. The supraglottic area of the larynx is richly supplied with lymphatics, and 25%-75% of supraglottic carcinomas metastasize in neck lymph nodes. Cortactin is a multidomain protein related to actin cytoskeleton regulation, podosome and lamellipodia formation, integrin signaling, axon guidance and extracellular matrix degradation. Cortactin is involved in metastasis formation because of its role in cell mobility. The present study focused mainly on the role of cortactin and phosphorylated cortactin (residues tyr421 and tyr466) expression and subcellular localization in primary supraglottic LSCCs and their cervical lymph node metastases. Methods: The immunohistochemical expression of cortactin, p-Y466-cortactin and p-Y421-cortactin was assessed in 38 primary supraglottic LSCCs and 10 lymph node metastases. The statistical approach included bootstrapping analysis. Results: Despite a significantly higher expression of cortactin in carcinoma cells than in adjacent normal laryngeal mucosa, no associations emerged between prognosis and the expression of cortactin or its isoforms in supraglottic LSCC. Statistical analysis found cortactin expression higher in less-differentiated LSCCs (p = 0.03). A significant direct correlation was found between cortactin and p-Y466-cortactin levels (p = 0.031), and between p-Y466-cortactin and p-Y421-cortactin levels (p = 0.001). Conclusions: Cortactin expression in carcinoma cells and its known involvement in the EGFR pathway suggest a role for this protein as a target for LSCC therapy. Further prospective studies are needed to investigate the potential of cortactin, p-Y466-cortactin and p-Y421-cortactin expression as markers of response to treatment (particularly EGFR-directed agents) in LSCC.
Collapse
|
35
|
Pattle SB, Utjesanovic N, Togo A, Wells L, Conn B, Monaghan H, Junor E, Johannessen I, Cuschieri K, Talbot S. Copy number gain of 11q13.3 genes associates with pathological stage in hypopharyngeal squamous cell carcinoma. Genes Chromosomes Cancer 2016; 56:185-198. [DOI: 10.1002/gcc.22425] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2016] [Revised: 10/11/2016] [Accepted: 10/12/2016] [Indexed: 12/13/2022] Open
Affiliation(s)
- Samuel B. Pattle
- Division of Infection and Pathway Medicine; The University of Edinburgh, Little France Crescent, Scotland
| | - Natasa Utjesanovic
- Division of Infection and Pathway Medicine; The University of Edinburgh, Little France Crescent, Scotland
| | - Athena Togo
- Department of Otolaryngology, The Laurieston Building; NHS Lothian, Edinburgh
| | - Lucy Wells
- Western General Hospital; The Edinburgh Cancer Centre; NHS Lothian, Edinburgh
| | - Brendan Conn
- Department of Pathology; Royal Infirmary of Edinburgh; NHS Lothian, Edinburgh
| | - Hannah Monaghan
- Department of Pathology; Royal Infirmary of Edinburgh; NHS Lothian, Edinburgh
| | - Elizabeth Junor
- Western General Hospital; The Edinburgh Cancer Centre; NHS Lothian, Edinburgh
| | | | - Kate Cuschieri
- Scottish HPV Reference Laboratory; Royal Infirmary of Edinburgh; NHS Lothian, Edinburgh
| | - Simon Talbot
- Division of Infection and Pathway Medicine; The University of Edinburgh, Little France Crescent, Scotland
| |
Collapse
|
36
|
Spatial intratumoral heterogeneity and temporal clonal evolution in esophageal squamous cell carcinoma. Nat Genet 2016; 48:1500-1507. [PMID: 27749841 PMCID: PMC5127772 DOI: 10.1038/ng.3683] [Citation(s) in RCA: 176] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2016] [Accepted: 08/31/2016] [Indexed: 12/14/2022]
Abstract
Esophageal squamous cell carcinoma (ESCC) is among the most common malignancies, but little is known about its spatial intratumor heterogeneity (ITH) and temporal clonal evolutionary processes. To address this, we performed multiregion whole-exome sequencing on 51 tumor regions from 13 ESCCs, and multiregion global methylation profiling on three of these 13 cases. We found an average of 35.8% heterogeneous somatic mutations with strong evidence of ITH. Half of driver mutations located on the branches targeted oncogenes, including PIK3CA, NFE2L2, MTOR, etc. By contrast, the majority of truncal and clonal driver mutations occurred in tumor suppressor genes, including TP53, KMT2D, ZNF750, etc. Interestingly, the phyloepigenetic trees robustly recapitulated the topologic structures of the phylogenetic ones, indicating the possible relationship between genetic and epigenetic alterations. Our integrated investigations of the spatial ITH and clonal evolution provide an important molecular foundation for enhanced understanding of the tumorigenesis and progression of ESCC.
Collapse
|
37
|
Jing X, Wu H, Ji X, Wu H, Shi M, Zhao R. Cortactin promotes cell migration and invasion through upregulation of the dedicator of cytokinesis 1 expression in human colorectal cancer. Oncol Rep 2016; 36:1946-1952. [PMID: 27633051 DOI: 10.3892/or.2016.5058] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2016] [Accepted: 08/16/2016] [Indexed: 11/06/2022] Open
Abstract
Cortactin (CTTN), a major substrate of the Src tyrosine kinase, has been implicated in cell proliferation, motility and invasion in various types of cancer. However, the molecular mechanisms of CTTN-driven malignant behavior remain unclear. In the current study, we determined the expression of CTTN in colorectal cancer and investigated its underlying mechanism in the metastasis of colorectal cancer. We confirmed increased CTTN expression in lymph node-positive CRC specimens and highly invasive CRC cell lines. Further study has shown that overexpression of CTTN promoted CRC cell migration and invasion, whereas CTTN silencing inhibited CRC cell migratory and invasive capacities in vitro. Mechanistically, CTTN increases expression of dedicator of cytokinesis 1 (DOCK1) and gene silencing of DOCK1 partially abolishes the migration and invasion capacity by CTTN. Our findings indicate that CTTN promotes metastasis of CRC cells by increasing DOCK1 expression and this could offer a promising therapeutic target for colorectal cancer treatment.
Collapse
Affiliation(s)
- Xiaoqian Jing
- Department of Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, P.R. China
| | - Huo Wu
- Department of Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, P.R. China
| | - Xiaopin Ji
- Department of Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, P.R. China
| | - Haoxuan Wu
- Department of Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, P.R. China
| | - Minmin Shi
- Department of Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, P.R. China
| | - Ren Zhao
- Department of Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, P.R. China
| |
Collapse
|
38
|
Divergent synthesis and identification of the cellular targets of deoxyelephantopins. Nat Commun 2016; 7:12470. [PMID: 27539788 PMCID: PMC4992173 DOI: 10.1038/ncomms12470] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2016] [Accepted: 06/23/2016] [Indexed: 12/12/2022] Open
Abstract
Herbal extracts containing sesquiterpene lactones have been extensively used in traditional medicine and are known to be rich in α,β-unsaturated functionalities that can covalently engage target proteins. Here we report synthetic methodologies to access analogues of deoxyelephantopin, a sesquiterpene lactone with anticancer properties. Using alkyne-tagged cellular probes and quantitative proteomics analysis, we identified several cellular targets of deoxyelephantopin. We further demonstrate that deoxyelephantopin antagonizes PPARγ activity in situ via covalent engagement of a cysteine residue in the zinc-finger motif of this nuclear receptor. Deoxyelephantopin is a naturally occurring sesquiterpene lactone with known anticancer properties. Here, the authors synthesize deoxyelephantopins and a range of analogues including alkyne-tagged probes, using them to identify its cellular targets.
Collapse
|
39
|
Calpain Genetic Disruption and HSP90 Inhibition Combine To Attenuate Mammary Tumorigenesis. Mol Cell Biol 2016; 36:2078-88. [PMID: 27215381 DOI: 10.1128/mcb.01062-15] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2015] [Accepted: 04/26/2016] [Indexed: 12/17/2022] Open
Abstract
Calpain is an intracellular Ca(2+)-regulated protease system whose substrates include proteins involved in proliferation, survival, migration, invasion, and sensitivity to therapeutic drugs. Genetic disruption of calpain attenuated the tumorigenic potential of breast cancer cells and hypersensitized cells to 17AAG, an inhibitor of the molecular chaperone HSP90. Calpain-1 or -2 overexpression rendered cells resistant to 17AAG, whereas downregulation or inhibition of calpain-1/2 led to increased cell death in multiple breast cancer cell lines, including models of HER2(+) (SKBR3) and triple-negative basal-cell-like (MDA-MB-231) breast cancer. In an MDA-MB-231 orthotopic xenograft model, calpain knockdown or 17AAG treatment independently attenuated tumor growth and metastasis, while the combination was most effective. Calpain knockdown was associated with increased 17AAG-induced degradation of the HSP90 clients cyclin D1 and AKT and multidrug resistance protein 2, which correlated with increased expression of antimitogenic p27(KIP1) and proapoptotic BIM proteins. Like other therapeutics, 17AAG can be effluxed by specific ABC transporters. Calpain expression positively correlated with the expression of P glycoprotein in mouse embryonic fibroblasts. Importantly, we show that calpain affects ABC transporter function and efflux of clinically relevant doxorubicin. These observations provide a compelling rationale for exploring the combination of calpain inhibition with new or existing cancer therapeutics.
Collapse
|
40
|
LONG HAOCHENG, GAO XIA, LEI CHANGJIANG, ZHU BIN, LI LEI, ZENG CHENG, HUANG JIANBIN, FENG JIARUI. miR-542-3p inhibits the growth and invasion of colorectal cancer cells through targeted regulation of cortactin. Int J Mol Med 2016; 37:1112-8. [DOI: 10.3892/ijmm.2016.2505] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2015] [Accepted: 02/08/2016] [Indexed: 11/05/2022] Open
|
41
|
Microtubule-associated protein 4 is an important regulator of cell invasion/migration and a potential therapeutic target in esophageal squamous cell carcinoma. Oncogene 2016; 35:4846-56. [PMID: 26876215 DOI: 10.1038/onc.2016.17] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2015] [Revised: 12/06/2015] [Accepted: 12/11/2015] [Indexed: 01/01/2023]
Abstract
Cell invasion and migration significantly contribute to tumor metastasis. Microtubule-associated protein 4 (MAP4) protein is one member of microtubule-associate proteins family. It is responsible for stabilization of microtubules by modulation of microtubule dynamics. However, there is little information about the involvement of MAP4 in human cancer. Here we show that MAP4 serves as a regulator of invasion and migration in esophageal squamous cancer cells. By activating the ERK-c-Jun-vascular endothelial growth factor A signaling pathway, MAP4 promotes cell invasion and migration in vitro, tumor growth and metastasis in mouse models. Immunohistochemical staining of operative tissues indicated that MAP4 expression was associated with tumor stage, lymph node metastasis and shorter survival of the patients with esophageal squamous cell carcinoma (ESCC). Multivariate Cox regression analysis showed that MAP4 is an independent prognostic indicator. In the serial sections of ESCC tissues, there was a positive correlation between MAP4 and vascular endothelial growth factor A expression. Notably, an intratumoral injection of MAP4-small interfering RNA (siRNA) remarkably inhibited the growth of the tumors that formed by the MAP4-expressing ESCC cells in nude mice, and a combination of MAP4-siRNA and Bevacizumab significantly enhanced the inhibition effect. Our data suggest that MAP4 is probably a useful prognostic biomarker and a potential therapeutic target for the disease.
Collapse
|
42
|
Hu X, Moon JW, Li S, Xu W, Wang X, Liu Y, Lee JY. Amplification and overexpression of CTTN and CCND1 at chromosome 11q13 in Esophagus squamous cell carcinoma (ESCC) of North Eastern Chinese Population. Int J Med Sci 2016; 13:868-874. [PMID: 27877079 PMCID: PMC5118758 DOI: 10.7150/ijms.16845] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/14/2016] [Accepted: 09/01/2016] [Indexed: 12/13/2022] Open
Abstract
Esophageal squamous cell carcinoma (ESCC) is a genetically complex tumor type and is a major cause of cancer-related mortality. The combination of genetics, diet, behavior, and environment plays an important role in the carcinogenesis of ESCC. To characterize the genomic aberrations of this disease, we investigated the genomic imbalances in 19 primary ESCC cases using high-resolution array comparative genomic hybridization (CGH). All cases showed either loss or gain of whole chromosomes or segments of chromosome(s) with variable genomic sizes. The copy number alterations per case affected the median 34% (~ 1,034Mb/3,000Mb) of the whole genome. Recurrent gains were 1q21.3-qter, 3q13.11-qter, 5pter-p11, 7pter-p15.3, 7p12.1-p11.2, 7q11-q11.2, 8p12-qter, 11q13.2-q13.3, 12pter-p13.31, 17q24.2, 20q11.21-qter, and 22q11.21-q11.22 whereas the recurrent losses were 3pter-p11.1, 4pter-p12, 4q28.3-q31.22, 4q31.3-q32.1, 9pter-p12, 11q22.3-qter and 13q12.11-q22.1. Amplification of 11q13 resulting in overexpression of CTTN/CCND1 was the most prominent finding, which was observed in 13 of 19 ESCC cases. These unique profiles of copy number alteration should be validated by further studies and need to be taken into consideration when developing biomarkers for early detection of ESCC.
Collapse
Affiliation(s)
- Xiaoxia Hu
- Department of Pediatrics, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104 USA; Department of Clinical Medicine, College of Medicine and Health, Lishui University, Zhejiang, 323000, P.R. China
| | - Ji Wook Moon
- Department of Pathology, Korea University College of Medicine, Seoul, 02841, Republic of Korea
| | - Shibo Li
- Department of Pediatrics, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104 USA
| | - Weihong Xu
- Department of Pathology, Korea University College of Medicine, Seoul, 02841, Republic of Korea
| | - Xianfu Wang
- Department of Pathology, Korea University College of Medicine, Seoul, 02841, Republic of Korea
| | - Yuanyuan Liu
- Department of Internal Medicine, the First Hospital of Jilin University, Jilin, 130021, P.R. China
| | - Ji-Yun Lee
- Department of Pathology, Korea University College of Medicine, Seoul, 02841, Republic of Korea
| |
Collapse
|
43
|
Hwang YS, Lee J, Zhang X, Lindholm PF. Lysophosphatidic acid activates the RhoA and NF-κB through Akt/IκBα signaling and promotes prostate cancer invasion and progression by enhancing functional invadopodia formation. Tumour Biol 2015; 37:6775-85. [PMID: 26662305 DOI: 10.1007/s13277-015-4549-x] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2015] [Accepted: 11/30/2015] [Indexed: 12/31/2022] Open
Abstract
We have demonstrated previously that increased RhoA and nuclear factor (NF)-κB activities are associated with increased PC-3 prostate cancer cell invasion and that lysophosphatidic acid (LPA) significantly increases cancer invasion through RhoA and NF-κB activation. In this study, we identified the intermediate signaling molecules and specialized cell structures which are activated by LPA, resulting in enhanced cellular invasion. LPA-induced Akt and IκBα signaling pathways were necessary for RhoA and NF-κB activation, and these LPA effects were abolished by RhoA inhibition. Mice injected with PC-3 cells expressing dominant-negative RhoA N19 developed significantly less tumor growth compared with those injected with control (pcDNA 3.1). In addition, LPA treatment increased functional invadopodia formation. Activation of RhoA and NF-κB through the Akt and IκBα signaling pathway was required for LPA-stimulated gelatin degradation activity. LPA administration increased tumor growth and osteolytic lesions in a mouse xenograft model. These results indicate that LPA promotes PC-3 cell invasion by increasing functional invadopodia formation via upregulating RhoA and NF-κB signaling which contributes to prostate cancer progression. Therefore, the LPA and RhoA-NF-κB signaling axis may represent key molecular targets to inhibit prostate cancer invasion and progression.
Collapse
Affiliation(s)
- Young Sun Hwang
- Department of Dental Hygiene, College of Health Science, Eulji University, Seongnam, Republic of Korea
| | - Jongsung Lee
- Department of Genetic Engineering, Sungkyunkwan University, 2066, Seobu-Ro, Jangan Gu, Suwon City, Gyunggi Do, 164-19, Republic of Korea
| | - Xianglan Zhang
- Oral Cancer Research Institute, Yonsei University College of Dentistry, Seoul, Republic of Korea.,Department of Pathology, Yanbian University Hospital, Yanji City, Jilin Province, China
| | - Paul F Lindholm
- Department of Pathology, Northwestern University, Feinberg School of Medicine, Chicago, IL, 60611, USA.
| |
Collapse
|
44
|
Rhie A, Park WS, Choi MK, Kim JH, Ryu J, Ryu CH, Kim JI, Jung YS. Genomic Copy Number Variations Characterize the Prognosis of Both P16-Positive and P16-Negative Oropharyngeal Squamous Cell Carcinoma After Curative Resection. Medicine (Baltimore) 2015; 94:e2187. [PMID: 26683928 PMCID: PMC5058900 DOI: 10.1097/md.0000000000002187] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Recently increasing high-risk HPV+ OSCC exhibits unique clinical and molecular characteristics compared to HPV-unrelated (HPV-) counterpart. Genomic copy number variations (CNVs), unique in HPV+ OSCCs, and their role for the prognosis prediction remains poorly studied. Here, we analyzed the distinct genomic copy number variations (CNVs) in human papillomavirus-related (HPV+) oropharyngeal squamous cell carcinoma (OSCC) and their role as a prognosticator after curative resection. For 58 consecutive, Korean OSCC patients that underwent surgery-based treatment with median 10 years of follow-up, HPV-related markers, and genome-wide CNV analysis were analyzed. Clinical associations between the CNV profile and survival analyses were followed. p16 expression predicted the overall survival (OS) (hazard ratio [HR] = 0.27, confidence interval [CI]: 0.39-0.80, P = 0.0006) better than HPV L1 PCR (HR = 0.83, CI: 0.66-1.29, P = 0.64), smoking, or other variables. Although the overall number of CNVs was not significantly different, 30 loci showed unique CNV patterns between the p16+ and p16- groups. A region containing PRDM2 was amplified only in the p16+ group, whereas EGFR and 11q13.3 showed increased amplification in p16- counterpart. Loss of a locus containing FGF18 led to a worse, but gain of region including CDK10 and RAD18 led to better overall survival (OS) in all OSCC patients. Meanwhile, subgroup analysis of p16+ OSCC revealed that amplification of regions harboring HRAS and loss of locus bearing KDR led to better OS. p16+ OSCC exhibit distinct CNV patterns compared with p16- counterpart. Specific patterns of CNVs predict better survival, especially in p16+ OSCC. This might allow better insights of the outcome after curative resection for HPV+ and HPV- OSCC.
Collapse
Affiliation(s)
- Arang Rhie
- From the Department of Biomedical Science, Department of Biochemistry and Molecular Biology, Genomic Medicine Institute (GMI), Medical Research Center, Seoul National University, Seoul (AR, J-IIK); Department of Pathology, Center for Specific Organs Cancer, Hematologic Malignancy Branch, National Cancer Center, Goyang, Gyeonggi (WSP, MKC); and Department of Otolaryngology-Head and Neck Surgery, Graduate School of Cancer Science and Policy, Specific Organs Cancer Branch, National Cancer Center, Goyang, Gyeonggi, Korea (J-HK, JR, CHR, Y-SJ)
| | | | | | | | | | | | | | | |
Collapse
|
45
|
Cao W, Wu W, Yan M, Tian F, Ma C, Zhang Q, Li X, Han P, Liu Z, Gu J, Biddle FG. Multiple region whole-exome sequencing reveals dramatically evolving intratumor genomic heterogeneity in esophageal squamous cell carcinoma. Oncogenesis 2015; 4:e175. [PMID: 26619400 PMCID: PMC4670960 DOI: 10.1038/oncsis.2015.34] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2015] [Revised: 10/02/2015] [Accepted: 10/08/2015] [Indexed: 12/13/2022] Open
Abstract
Cancer is a disease of genome instability and genomic alterations; now, genomic heterogeneity is rapidly emerging as a defining feature of cancer, both within and between tumors. Motivation for our pilot study of tumor heterogeneity in esophageal squamous cell carcinoma (ESCC) is that it is not well studied, but the highest incidences of esophageal cancers are found in China and ESCC is the most common type. We profiled the mutations and changes in copy number that were identified by whole-exome sequencing and array-based comparative genomic hybridization in multiple regions within an ESCC from two patients. The average mutational heterogeneity rate was 90% in all regions of the individual tumors in each patient; most somatic point mutations were nonsynonymous substitutions, small Indels occurred in untranslated regions of genes, and copy number alterations varied among multiple regions of a tumor. Independent Sanger sequencing technology confirmed selected gene mutations with more than 88% concordance. Phylogenetic analysis of the somatic mutation frequency demonstrated that multiple, genomically heterogeneous divergent clones evolve and co-exist within a primary ESCC and metastatic subclones result from the dispersal and adaptation of an initially non-metastatic parental clone. Therefore, a single-region sampling will not reflect the evolving architecture of a genomically heterogeneous landscape of mutations in ESCC tumors and the divergent complexity of this genomic heterogeneity among patients will complicate any promise of a simple genetic or epigenetic diagnostic signature in ESCC. We conclude that any potential for informative biomarker discovery in ESCC and targeted personalized therapies will require a deeper understanding of the functional biology of the ontogeny and phylogeny of the tumor heterogeneity.
Collapse
Affiliation(s)
- W Cao
- Translational Medical Center, Zhengzhou Central Hospital Affiliated to Zhengzhou University, Zhengzhou, China
| | - W Wu
- Translational Medical Center, Zhengzhou Central Hospital Affiliated to Zhengzhou University, Zhengzhou, China.,Department of Pathology and Laboratory Medicine, Arnie Charbonneau Cancer Institute, Cumming School of Medicine, University of Calgary, Calgary Alberta, Canada
| | - M Yan
- Medical School, Zhengzhou University, Zhengzhou, China
| | - F Tian
- Translational Medical Center, Zhengzhou Central Hospital Affiliated to Zhengzhou University, Zhengzhou, China
| | - C Ma
- Translational Medical Center, Zhengzhou Central Hospital Affiliated to Zhengzhou University, Zhengzhou, China
| | - Q Zhang
- Translational Medical Center, Zhengzhou Central Hospital Affiliated to Zhengzhou University, Zhengzhou, China
| | - X Li
- Translational Medical Center, Zhengzhou Central Hospital Affiliated to Zhengzhou University, Zhengzhou, China
| | - P Han
- Translational Medical Center, Zhengzhou Central Hospital Affiliated to Zhengzhou University, Zhengzhou, China
| | - Z Liu
- Linzhou Cancer Hospital, Linzhou, China
| | - J Gu
- Science and Education Department, Health Bureau of Zhengzhou, Zhengzhou, China
| | - F G Biddle
- Departments of Medical Genetics and Biological Sciences, University of Calgary, Calgary, Canada
| |
Collapse
|
46
|
He J, Zhu G, Gao L, Chen P, Long Y, Liao S, Yi H, Yi W, Pei Z, Wu M, Li X, Xiang J, Peng S, Ma J, Zhou M, Xiong W, Zeng Z, Xiang B, Tang K, Cao L, Li G, Zhou Y. Fra-1 is upregulated in gastric cancer tissues and affects the PI3K/Akt and p53 signaling pathway in gastric cancer. Int J Oncol 2015; 47:1725-34. [PMID: 26330014 DOI: 10.3892/ijo.2015.3146] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2015] [Accepted: 07/30/2015] [Indexed: 11/06/2022] Open
Abstract
Gastric cancer is an aggressive disease that continues to have a daunting impact on global health. Fra-1 (FOSL1) plays important roles in oncogenesis in various malignancies. We investigated the expression of Fra-1 in gastric cancer (GC) tissues by qPCR, immunohistochemistry (IHC) and western blot technologies. The results showed that Fra-1 was overexpressed in gastric cancer tissues compared with the adjacent non‑cancerous tissues. To explore the possible mechanism of Fra-1 in GC, we elucidated the effect of Fra-1 in the apoptosis and cell cycle of gastric cancer cells, AGS, and found that a considerable decrease in apoptotic cells and increase of S phase rate were observed for AGS cells with Fra-1 overexpession. We identified and confirmed that Fra-1 affected the expression level of CTTN and EZR in vitro through LC-MS/MS analyses and western blot technology. Furthermore, we found that Fra-1 was correlated with dysregulation PI3K/Akt and p53 signaling pathway in gastric cancer tissues in vitro. Moreover, we found that Fra-1 overexpression affected the expression of PI3K, Akt, MDM2 and p53 in vivo. In summary, our results suggest that Fra-1 is upregulated in gastric cancer tissues and plays its function by affecting the PI3K/Akt and p53 signaling pathway in gastric cancer.
Collapse
Affiliation(s)
- Junyu He
- Hunan Provincial Tumor Hospital and the Affiliated Tumor Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan 410006, P.R. China
| | - Guangchao Zhu
- Hunan Provincial Tumor Hospital and the Affiliated Tumor Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan 410006, P.R. China
| | - Lu Gao
- Cancer Research Institute, Central South University, Changsha, Hunan 410078, P.R. China
| | - Pan Chen
- Hunan Provincial Tumor Hospital and the Affiliated Tumor Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan 410006, P.R. China
| | - Yuehua Long
- Cancer Research Institute, Central South University, Changsha, Hunan 410078, P.R. China
| | - Shan Liao
- Cancer Research Institute, Central South University, Changsha, Hunan 410078, P.R. China
| | - Hong Yi
- Research Center of Carcinogenesis and Targeted Therapy, Xiangya Hospital, Central South University, Changsha, Hunan 410008, P.R. China
| | - Wei Yi
- Cancer Research Institute, Central South University, Changsha, Hunan 410078, P.R. China
| | - Zhen Pei
- Cancer Research Institute, Central South University, Changsha, Hunan 410078, P.R. China
| | - Minghua Wu
- Hunan Provincial Tumor Hospital and the Affiliated Tumor Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan 410006, P.R. China
| | - Xiaoling Li
- Hunan Provincial Tumor Hospital and the Affiliated Tumor Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan 410006, P.R. China
| | - Juanjuan Xiang
- Hunan Provincial Tumor Hospital and the Affiliated Tumor Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan 410006, P.R. China
| | - Shuping Peng
- Hunan Provincial Tumor Hospital and the Affiliated Tumor Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan 410006, P.R. China
| | - Jian Ma
- Hunan Provincial Tumor Hospital and the Affiliated Tumor Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan 410006, P.R. China
| | - Ming Zhou
- Hunan Provincial Tumor Hospital and the Affiliated Tumor Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan 410006, P.R. China
| | - Wei Xiong
- Hunan Provincial Tumor Hospital and the Affiliated Tumor Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan 410006, P.R. China
| | - Zhaoyang Zeng
- Hunan Provincial Tumor Hospital and the Affiliated Tumor Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan 410006, P.R. China
| | - Bo Xiang
- Hunan Provincial Tumor Hospital and the Affiliated Tumor Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan 410006, P.R. China
| | - Ke Tang
- Cancer Research Institute, Central South University, Changsha, Hunan 410078, P.R. China
| | - Li Cao
- Cancer Research Institute, Central South University, Changsha, Hunan 410078, P.R. China
| | - Guiyuan Li
- Hunan Provincial Tumor Hospital and the Affiliated Tumor Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan 410006, P.R. China
| | - Yanhong Zhou
- Hunan Provincial Tumor Hospital and the Affiliated Tumor Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan 410006, P.R. China
| |
Collapse
|
47
|
Expression and clinical significance of cortactin protein in ovarian neoplasms. Clin Transl Oncol 2015; 18:220-7. [DOI: 10.1007/s12094-015-1360-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2015] [Accepted: 07/13/2015] [Indexed: 12/30/2022]
|
48
|
Regulators of Actin Dynamics in Gastrointestinal Tract Tumors. Gastroenterol Res Pract 2015; 2015:930157. [PMID: 26345720 PMCID: PMC4539459 DOI: 10.1155/2015/930157] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/15/2015] [Revised: 07/09/2015] [Accepted: 07/21/2015] [Indexed: 02/07/2023] Open
Abstract
Reorganization of the actin cytoskeleton underlies cell migration in a wide variety of physiological and pathological processes, such as embryonic development, wound healing, and tumor cell invasion. It has been shown that actin assembly and disassembly are precisely regulated by intracellular signaling cascades that respond to changes in the cell microenvironment, ligand binding to surface receptors, or oncogenic transformation of the cell. Actin-nucleating and actin-depolymerizing (ANFs/ADFs) and nucleation-promoting factors (NPFs) regulate cytoskeletal dynamics at the leading edge of migrating cells, thereby modulating cell shape; these proteins facilitate cellular movement and mediate degradation of the surrounding extracellular matrix by secretion of lytic proteases, thus eliminating barriers for tumor cell invasion. Accordingly, expression and activity of these actin-binding proteins have been linked to enhanced metastasis and poor prognosis in a variety of malignancies. In this review, we will summarize what is known about expression patterns and the functional role of actin regulators in gastrointestinal tumors and evaluate first pharmacological approaches to prevent invasion and metastatic dissemination of malignant cells.
Collapse
|
49
|
Kim DY, Lee JH, Kim KY, Kang DB, Park WC, Chae SC, Lee JK. Association between genetic polymorphisms in cortactin and susceptibility to gastric cancer. Ann Surg Treat Res 2015; 89:74-80. [PMID: 26236696 PMCID: PMC4518033 DOI: 10.4174/astr.2015.89.2.74] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2014] [Revised: 03/12/2015] [Accepted: 03/19/2015] [Indexed: 01/14/2023] Open
Abstract
PURPOSE Overexpression of cortactin (CTTN) in human tumors has been proposed to result in increased cell migration and metastatic potential. Here, we determined the frequencies of CTTN g.-9101C>T, g.-8748C>T, and g.72C>T polymorphisms in apparently healthy subjects and gastric cancer patients, respectively, and the influence of the CTTN polymorphisms on gastric cancer susceptibility. METHODS Blood samples were collected from 267 patients and 533 controls. CTTN g.-8748C>T and g.-9101C>T polymorphisms were determined using polymerase chain reaction-restriction fragment length polymorphism; the g.72C>T polymorphism was determined using the TaqMan method. RESULTS Genotype frequencies of the CTTN g.-9101C>T polymorphism were 97.5% (TT), 2.5% (TC), and 0% (CC) in the patient group, and 98.6% (TT), 1.4% (TC), and 0% (CC) in the control group. Genotype frequencies of the CTTN g.-8748C>T polymorphism were 93.3% (TT), 6.8% (TC), and 0% (CC) in the patient group, and 94.2% (TT), 5.8% (TC), and 0% (CC) in the control group. Genotype frequencies of the CTTN g.72C>T polymorphism were 82.4% (CC), 17.2% (CT), and 0.4% (TT) in the patient group, and 78.0% (CC), 20.1% (CT), and 1.9% (TT) in the control group. Genotype and allele frequencies of the CTTN g.-9101C>T polymorphism differed significantly between the advanced gastric cancer and control groups. Patients with advanced gastric cancer, possessing the TC genotype, had a significantly poorer prognosis than the group with the TT genotype. CONCLUSION The CTTN g.-9101C>T polymorphism might influence advanced gastric cancer susceptibility. However, the role of the CTTN g.-9101C>T, g.-8748C>T, and g.72C>T polymorphisms requires careful interpretation and confirmation through larger studies.
Collapse
Affiliation(s)
- Dae Yong Kim
- Department of Surgery, Institute of Medical Science, Wonkwang University School of Medicine, Iksan, Korea
| | - Joo Hyun Lee
- Department of Surgery, Institute of Medical Science, Wonkwang University School of Medicine, Iksan, Korea
| | - Keun Young Kim
- Department of Surgery, Institute of Medical Science, Wonkwang University School of Medicine, Iksan, Korea
| | - Dong Baek Kang
- Department of Surgery, Institute of Medical Science, Wonkwang University School of Medicine, Iksan, Korea
| | - Won Cheol Park
- Department of Surgery, Institute of Medical Science, Wonkwang University School of Medicine, Iksan, Korea
| | - Soo Cheon Chae
- Department of Pathology, Institute of Medical Science, Wonkwang University School of Medicine, Iksan, Korea
| | - Jeong Kyun Lee
- Department of Surgery, Institute of Medical Science, Wonkwang University School of Medicine, Iksan, Korea
| |
Collapse
|
50
|
Markwell SM, Weed SA. Tumor and stromal-based contributions to head and neck squamous cell carcinoma invasion. Cancers (Basel) 2015; 7:382-406. [PMID: 25734659 PMCID: PMC4381264 DOI: 10.3390/cancers7010382] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2015] [Revised: 02/10/2015] [Accepted: 02/15/2015] [Indexed: 12/11/2022] Open
Abstract
Head and neck squamous cell carcinoma (HNSCC) is typically diagnosed at advanced stages with evident loco-regional and/or distal metastases. The prevalence of metastatic lesions directly correlates with poor patient outcome, resulting in high patient mortality rates following metastatic development. The progression to metastatic disease requires changes not only in the carcinoma cells, but also in the surrounding stromal cells and tumor microenvironment. Within the microenvironment, acellular contributions from the surrounding extracellular matrix, along with contributions from various infiltrating immune cells, tumor associated fibroblasts, and endothelial cells facilitate the spread of tumor cells from the primary site to the rest of the body. Thus far, most attempts to limit metastatic spread through therapeutic intervention have failed to show patient benefit in clinic trails. The goal of this review is highlight the complexity of invasion-promoting interactions in the HNSCC tumor microenvironment, focusing on contributions from tumor and stromal cells in order to assist future therapeutic development and patient treatment.
Collapse
Affiliation(s)
- Steven M Markwell
- Department of Neurobiology and Anatomy, Program in Cancer Cell Biology, Mary Babb Randolph Cancer Center, West Virginia University, Morgantown, WV 26506, USA.
| | - Scott A Weed
- Department of Neurobiology and Anatomy, Program in Cancer Cell Biology, Mary Babb Randolph Cancer Center, West Virginia University, Morgantown, WV 26506, USA.
| |
Collapse
|