1
|
Zhong M, Zhang P, Yao S. LTBP2 silence suppresses glioblastoma proliferation and tumor growth of xenograft tumor mice through modulating JAK2/STAT2 signaling pathway. Tissue Cell 2025; 93:102697. [PMID: 39752891 DOI: 10.1016/j.tice.2024.102697] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2024] [Revised: 11/29/2024] [Accepted: 12/18/2024] [Indexed: 03/05/2025]
Abstract
Glioblastoma is considered the most malignant central nervous system tumor. This study aimed to investigate effects of latent transforming growth factor-β binding protein-2 (LTBP2) on glioblastoma growth and associated mechanisms. LTBP2 gene transcription in glioblastoma was determined using RT-PCR. LTBP2 gene silencing lentiviral vectors were synthesized, and the highest efficient vector was selected to package sh-LTBP2 lentivirus. Xenograft tumor model was constructed by injecting sh-LTBP2-infected U87MG cells into mice, and tumor growth was evaluated. Proliferation, colony formation and migration of U87MG were verified with CCK-8, colony-formation assay and migration assay, and cell cycle was examined. Western blot and immunohistochemistry were performed to examine Janus kinase 2 (JAK2) and signal transducers and activators of transcription 2 (STAT2) phosphorylation. LTBP2 gene transcription in SVGp12 cells was significantly lower compared to U87MG, U251 and T98G cells (P < 0.001). LTBP2 gene silence suppressed U87MG cell proliferation, migration and colony formation compared to U87MG group and sh-NC group (P < 0.05). LTBP2 gene silence regulated phases of cell cycle in U87MG cells. JAK2/STAT2 participated in LTBP2 silence-induced decreased U87MG proliferation. LTBP2 gene silence suppressed tumor growth by modulating JAK2/STAT2 pathway in xenograft tumor mice. In conclusion, LTBP2 silence inhibited proliferation and migration of U87MG cells and tumor growth of xenograft tumor mice through modulating JAK2/STAT2 signaling pathway.
Collapse
Affiliation(s)
- Musheng Zhong
- Zunyi Medical University, Zhuhai, China; Pingsha Community Health Service center, Zhuhai, China
| | - Peng Zhang
- Department of Neurosurgery, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Shengtao Yao
- Department of Neurosurgery, Affiliated Hospital of Zunyi Medical University, Zunyi, China.
| |
Collapse
|
2
|
Kim OH, Jeon TJ, Kang H, Chang ES, Hong SA, Kim MK, Lee HJ. hsa-mir-483-3p modulates delayed breast cancer recurrence. Sci Rep 2025; 15:693. [PMID: 39753688 PMCID: PMC11698896 DOI: 10.1038/s41598-024-84437-6] [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: 08/07/2024] [Accepted: 12/23/2024] [Indexed: 01/06/2025] Open
Abstract
Patients with estrogen receptor-positive breast cancer undergoing continuous adjuvant hormone therapy often experience delayed recurrence with tamoxifen use, potentially causing adverse effects. However, the lack of biomarkers hampers patient selection for extended endocrine therapy. This study aimed to elucidate the molecular mechanisms underlying delayed recurrence and identify biomarkers. When miRNA expression was assessed in luminal breast cancer tissues with and without delayed recurrence using NanoString, a significant increase in the expression of miR483-3p was observed in samples from patients with delayed recurrence compared with those without. miR483-3p expression was elevated in tamoxifen resistant (TAMR) EFM19 cells than in non-resistant EFM19 cells. Notably, genes associated with cancer metastasis (AMOTL2, ANKRD1, CTGF, and VEGF) were upregulated in TAMR EFM19 cells, although cell motility and proliferation were reduced. Transfection of miR483-3p mimics into both non-resistant EFM19 and MCF7 cells resulted in increased expression of cancer metastasis-related genes, but decreased proliferation and migration. Given that miR483-3p can bind to the 3'UTR region of O-GlcNAc transferase (OGT) and potentially affect its protein expression, we examined OGT protein levels and found that transfection with miR483-3p mimics selectively reduced OGT expression. Overall, breast cancer cells subjected to long-term hormone therapy displayed elevated miR483-3p expression, reducing motility and dormancy induction via decreased OGT expression. These findings suggest that miR483-3p is a potential biomarker for long-term endocrine therapy.
Collapse
Affiliation(s)
- Ok-Hyeon Kim
- Department of Anatomy and Cell Biology, College of Medicine, Chung-Ang University, Seoul, 06974, South Korea
| | - Tae Jin Jeon
- Department of Global Innovative Drugs, Graduate School of Chung-Ang University, Seoul, 06974, South Korea
| | - Hana Kang
- Department of Global Innovative Drugs, Graduate School of Chung-Ang University, Seoul, 06974, South Korea
| | - Eun Seo Chang
- Department of Global Innovative Drugs, Graduate School of Chung-Ang University, Seoul, 06974, South Korea
| | - Soon Auck Hong
- Department of Pathology, College of Medicine, Chung-Ang University, Seoul, 06974, South Korea
| | - Min Kyoon Kim
- Department of Surgery, College of Medicine, Chung-Ang University, Seoul, 06974, South Korea.
| | - Hyun Jung Lee
- Department of Anatomy and Cell Biology, College of Medicine, Chung-Ang University, Seoul, 06974, South Korea.
- Department of Global Innovative Drugs, Graduate School of Chung-Ang University, Seoul, 06974, South Korea.
| |
Collapse
|
3
|
Xia C, Zhao J, Huang Y, Miao H, Zhao F. Angiogenesis in nasopharyngeal carcinoma: insights, imaging, and therapeutic strategies. Front Oncol 2024; 14:1331064. [PMID: 38863627 PMCID: PMC11165036 DOI: 10.3389/fonc.2024.1331064] [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: 10/31/2023] [Accepted: 05/09/2024] [Indexed: 06/13/2024] Open
Abstract
Nasopharyngeal carcinoma (NPC) is a highly prevalent head and neck malignancy in southern China frequently diagnosed at advanced stages owing to subtle early symptoms and associated metastasis. Angiogenesis emerges as a pivotal factor in NPC progression, with numerous angiogenesis-related factors showing aberrant expression and contributing to increased neovascularization within NPC tumors. These abnormal vessels not only nourish tumor growth but also facilitate metastasis, culminating in unfavorable patient outcomes. Multiple studies have demonstrated the applicability of various imaging techniques for assessing angiogenesis in NPC tumors, thus serving as a foundation for personalized treatment strategies and prognostic assessments. Anti-angiogenic therapies have exhibited significant potential for inhibiting NPC angiogenesis and exerting anti-tumor effects. To enhance efficacy, anti-angiogenic drugs are frequently combined with other treatment modalities to synergistically enhance anti-tumor effects while mitigating the side effects associated with single-agent therapies, consequently improving patient prognosis. Identifying the potential mechanisms and key targets underlying NPC angiogenesis and exploring more effective detection and treatment approaches holds promise for shaping the future of NPC diagnosis, treatment, and prognosis, thereby offering new avenues and perspectives for research and clinical practice.
Collapse
Affiliation(s)
- Chenxi Xia
- Department of Otolaryngology-Head and Neck Surgery, the Affiliated Hospital of Southwest Medical University, Southwest Medical University, Luzhou, Sichuan, China
| | - Jia Zhao
- Department of Otolaryngology-Head and Neck Surgery, the Affiliated Hospital of Southwest Medical University, Southwest Medical University, Luzhou, Sichuan, China
| | - Yu Huang
- Department of Otolaryngology-Head and Neck Surgery, Chengdu Second People’s Hospital, Chengdu, Sichuan, China
| | - Hongbin Miao
- Department of Otolaryngology-Head and Neck Surgery, Bishan hospital of Chongqing Medical University, Bishan Hospital of Chongqing, Bishan, Chongqing, China
| | - Feipeng Zhao
- Department of Otolaryngology-Head and Neck Surgery, the Affiliated Hospital of Southwest Medical University, Southwest Medical University, Luzhou, Sichuan, China
- Department of Otolaryngology-Head and Neck Surgery, Chengdu Second People’s Hospital, Chengdu, Sichuan, China
| |
Collapse
|
4
|
LTBP2 Knockdown Promotes Ferroptosis in Gastric Cancer Cells through p62-Keap1-Nrf2 Pathway. BIOMED RESEARCH INTERNATIONAL 2022; 2022:6532253. [PMID: 35968244 PMCID: PMC9371865 DOI: 10.1155/2022/6532253] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Accepted: 07/06/2022] [Indexed: 11/18/2022]
Abstract
Gastric cancer (GC) is one of the most common gastrointestinal malignancies. Ferroptosis is a new type of peroxidation-driven and iron-dependent cell death. However, the biological functions and exact regulatory mechanisms of ferroptosis in GC remain elusive. Here, we performed RNAi and gene transfection, cell viability assay, lipid peroxidation assay, reactive oxygen species (ROS) assay, glutathione assay, qRT-PCR, Western blotting, and transmission electron microscopy (TEM) to study ferroptosis in gastric cancer. The results revealed that silencing latent transforming growth factor β binding proteins (LTBP2) can significantly inhibit GC cell proliferation and decrease cellular GSH levels, reduce GPX4 activity, and increase ROS generation and malondialdehyde (MDA) levels, leading to ferroptosis in GC cells. In addition, we demonstrate that suppression of LTBP2 could regulate the p62-Keap1-Nrf2 pathway, thereby downregulating the GPX4 and xCT expression and upregulating the PTGS2 and 4HNE expression. Our findings described a new role of LTBP2 in regulating ferroptosis, which heralds the prospect of ferroptosis-mediated cancer therapy.
Collapse
|
5
|
Zhang X, Tian C, Tian C, Cheng J, Mao W, Li M, Chen M. LTBP2 inhibits prostate cancer progression and metastasis via the PI3K/AKT signaling pathway. Exp Ther Med 2022; 24:563. [PMID: 36034756 PMCID: PMC9400130 DOI: 10.3892/etm.2022.11500] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Accepted: 05/11/2022] [Indexed: 12/02/2022] Open
Abstract
Biochemical recurrence (BCR) is a cause of concern in advanced prostate cancer (PCa). Thus, novel diagnostic biomarkers are required to improve clinical care. However, research on PCa immunotherapy is also scarce. Hence, the present study aimed to explore promising BCR-related diagnostic biomarkers, and their expression pattern, prognostic value, immune response effects, biological functions, and possible molecular mechanisms were evaluated. GEO datasets (GSE46602, GSE70768, and GSE116918) were downloaded and merged as the training cohort, and differential expression analysis was performed. Lasso regression and SVM-RFE algorithm, as well as PPI analysis and MCODE algorithm, were then applied to filter BCR-related biomarker genes. The CIBERSORT and estimation of stromal and immune cells in malignant tumor tissues using expression data (ESTIMATE) methods were used to calculate the fractions of tumor-infiltrating immune cells. GO/DO enrichment analyses were used to identify the biological functions. The expression of latent transforming growth factor β-binding protein 2 (LTBP2) was determined by RT-qPCR and western blotting. The role of LTBP2 in PCa was determined by CCK-8, Transwell, and the potential mechanism was investigated by KEGG and GSEA and confirmed by western blotting. In total, 44 BCR-related differentially expressed genes (DEGs) in the training cohort were screened. LTBP2 was found to be a diagnostic biomarker of BCR in PCa and was associated with CD4+ T-cell infiltration and response to anti-PD-1/PD-L1 immunotherapy. Subsequently, using the ESTIMATE algorithm, it was identified that LTBP2 was associated with the tumor microenvironment and could be a predictor of the clinical benefit of immune checkpoint blockade. Finally, the expression and biological function of LTBP2 were evaluated via cellular experiments. The results showed that LTBP2 was downregulated in PCa cells and inhibited PCa proliferation and metastasis via the PI3K/AKT signaling pathway in vitro. In conclusion, LTBP2 was a promising diagnostic biomarker of BCR of PCa and had an important role in CD4+ T-cell recruitment. Moreover, it was associated with immunotherapy in patients with PCa who developed BCR, and it inhibited PCa proliferation and metastasis via the PI3K/AKT signaling pathway in vitro.
Collapse
Affiliation(s)
- Xiaowen Zhang
- Department of Urology, Affiliated Zhongda Hospital of South‑East University, Nanjing, Jiangsu 210009, P.R. China
| | - Chuanjie Tian
- Department of Urology, Langxi County People's Hospital, Xuancheng, Anhui 242100, P.R. China
| | - Chuanjie Tian
- Department of Urology, Langxi County People's Hospital, Xuancheng, Anhui 242100, P.R. China
| | - Jianbin Cheng
- Department of Urology Surgery, Heqiao Hospital, Yixing, Jiangsu 214200, P.R. China
| | - Weipu Mao
- Department of Urology, Affiliated Zhongda Hospital of South‑East University, Nanjing, Jiangsu 210009, P.R. China
| | - Menglan Li
- NHC Contraceptives Adverse Reaction Surveillance Center, Jiangsu Health Development Research Center, Nanjing, Jiangsu 210036, P.R. China
| | - Ming Chen
- Department of Urology, Affiliated Zhongda Hospital of South‑East University, Nanjing, Jiangsu 210009, P.R. China
| |
Collapse
|
6
|
The Role of Latent Transforming Growth Factor β Binding Protein 2 (LTBP2) in the Diagnosis and Stage Discrimination of Gastric Cancer. Indian J Surg 2021. [DOI: 10.1007/s12262-021-03133-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
|
7
|
Akkoc Y, Peker N, Akcay A, Gozuacik D. Autophagy and Cancer Dormancy. Front Oncol 2021; 11:627023. [PMID: 33816262 PMCID: PMC8017298 DOI: 10.3389/fonc.2021.627023] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2020] [Accepted: 01/22/2021] [Indexed: 12/13/2022] Open
Abstract
Metastasis and relapse account for the great majority of cancer-related deaths. Most metastatic lesions are micro metastases that have the capacity to remain in a non-dividing state called “dormancy” for months or even years. Commonly used anticancer drugs generally target actively dividing cancer cells. Therefore, cancer cells that remain in a dormant state evade conventional therapies and contribute to cancer recurrence. Cellular and molecular mechanisms of cancer dormancy are not fully understood. Recent studies indicate that a major cellular stress response mechanism, autophagy, plays an important role in the adaptation, survival and reactivation of dormant cells. In this review article, we will summarize accumulating knowledge about cellular and molecular mechanisms of cancer dormancy, and discuss the role and importance of autophagy in this context.
Collapse
Affiliation(s)
- Yunus Akkoc
- Koç University Research Center for Translational Medicine (KUTTAM), Istanbul, Turkey
| | - Nesibe Peker
- Koç University Research Center for Translational Medicine (KUTTAM), Istanbul, Turkey
| | - Arzu Akcay
- Yeni Yüzyıl University, School of Medicine, Private Gaziosmanpaşa Hospital, Department of Pathology, Istanbul, Turkey
| | - Devrim Gozuacik
- Koç University Research Center for Translational Medicine (KUTTAM), Istanbul, Turkey.,Koç University School of Medicine, Istanbul, Turkey.,Sabancı University Nanotechnology Research and Application Center (SUNUM), Istanbul, Turkey
| |
Collapse
|
8
|
Liang X, Zhang L, Ji Q, Wang B, Wei D, Cheng D. miR-421 promotes apoptosis and suppresses metastasis of osteosarcoma cells via targeting LTBP2. J Cell Biochem 2019; 120:10978-10987. [PMID: 30924175 DOI: 10.1002/jcb.28144] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2018] [Accepted: 10/31/2018] [Indexed: 01/24/2023]
Abstract
Increasing evidence has confirmed that microRNAs (miRs) are involved in tumor development and progression. A previous study reported that miR-421 could serve as a diagnostic marker in patients with osteosarcoma (OS). The present study explored the potential roles of miR-421 in the regulation of cell proliferation, apoptosis, migration, invasion, and epithelial-mesenchymal transition of OS cells. Our results showed that miR-421 was upregulated in OS tissues and cell lines (MG63, U2OS, HOS, and Saos-2) compared with the corresponding adjacent tissues or human osteoblast cells hFOB1.19, while the latent transforming growth factor β-binding protein 2 (LTBP2) expression was reduced. In MG63 and U2OS cells, CCK8 assay displayed that cell proliferation was repressed by the miR-421 inhibitor, conversely increased by miR-421 mimics. Inhibition of miR-421 promoted cell apoptosis rate, caspase 3 activity, cleaved-caspase 3 (c-caspase 3) expression, and Bax/Bcl-2 ratio, restoration of miR-421 showed the opposite functions. Suppression of miR-421 blocked migration and invasion, whereas miR-421 overexpression promoted the migration and invasion of MG63 and U2OS cells. In addition, real-time polymerase chain reaction and Western blot analysis revealed that miR-421 negatively regulated E-cadherin expression, and positively regulated the expression of N-cadherin and vimentin. The luciferase reporter assay determined that miR-421 could target LTBP2-3'-UTR, and LTBP2 expression was regulated negatively by miR-421 both in mRNA and protein levels. Depletion of LTBP2 partly abolished the biological functions of miR-421 inhibitor in OS. In conclusion, miR-421 plays an oncogenic role in OS via targeting LTBP2, suggesting that miR-421 may be a potential therapeutic target against OS.
Collapse
Affiliation(s)
- Xiaoju Liang
- Department of Pediatric Orthopedics, Honghui Hospital, Xi'an Jiaotong University, Xi'an, China
| | - Lijun Zhang
- Department of Hand Surgery, Honghui Hospital, Xi'an Jiaotong University, Xi'an, China
| | - Qiang Ji
- Department of Pediatric Orthopedics, Honghui Hospital, Xi'an Jiaotong University, Xi'an, China
| | - Bing Wang
- Department of Pediatric Orthopedics, Honghui Hospital, Xi'an Jiaotong University, Xi'an, China
| | - Dengke Wei
- Department of Hand Surgery, Honghui Hospital, Xi'an Jiaotong University, Xi'an, China
| | - Deliang Cheng
- Department of Hand Surgery, Honghui Hospital, Xi'an Jiaotong University, Xi'an, China
| |
Collapse
|
9
|
High Expression of LTBP2 Contributes to Poor Prognosis in Colorectal Cancer Patients and Correlates with the Mesenchymal Colorectal Cancer Subtype. DISEASE MARKERS 2019; 2019:5231269. [PMID: 30956730 PMCID: PMC6431450 DOI: 10.1155/2019/5231269] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/10/2018] [Accepted: 02/07/2019] [Indexed: 01/12/2023]
Abstract
Colorectal cancer (CRC) is a complex and heterogeneous disease with four consensus molecular subtypes (CMS1-4). LTBP2 is a member of the fibrillin/LTBP super family and plays a critical role in tumorigenesis by activating TGF-β in the CMS4 CRC subtype. So far, the expression and prognostic significance of LTBP2 in CRC remains obscure. In this study, we aimed to analyze the mRNA and protein expression levels of LTBP2 in CRC tissues and then estimate their values as a potential prognostic biomarker. We detected the mRNA expression of LTBP2 in 28 cases of fresh CRC tissues and 4 CRC cell lines and the protein expression of LTBP2 in 483 samples of CRC tissues, matched tumor-adjacent tissues, and benign colorectal diseases. LTBP2 protein expression was then correlated to patients' clinical features and overall survival. Both LTBP2 mRNA and protein expression levels in CRC tissues were remarkably superior to those in adjacent normal colorectal tissues (P = 0.0071 and P < 0.001, respectively), according to TCGA dataset of CRC. High LTBP2 protein expression was correlated with TNM stage (P < 0.001), T stage (P < 0.001), N stage (P < 0.001), and M stage (P < 0.001). High LTBP2 protein expression was related to poor overall survival in CRC patients and was an independent prognostic factor for CRC. LTBP2 mRNA expression was especially higher in the CMS4 subtype (P < 0.001), which was confirmed in CRC cell lines. Our data suggested that LTBP2 may act as an oncogene in the development of colorectal cancer and have important significance in predicting CRC prognosis. LTBP2 could be a novel biomarker and potential therapeutic target for mesenchymal colorectal cancer and can improve the outcome of high-risk CRC.
Collapse
|
10
|
Wang J, Liang WJ, Min GT, Wang HP, Chen W, Yao N. LTBP2 promotes the migration and invasion of gastric cancer cells and predicts poor outcome of patients with gastric cancer. Int J Oncol 2018; 52:1886-1898. [PMID: 29620158 PMCID: PMC5919710 DOI: 10.3892/ijo.2018.4356] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2017] [Accepted: 03/08/2018] [Indexed: 12/20/2022] Open
Abstract
Latent transforming growth factor-β-binding protein (LTBP)2 is a member of the fibrillin/LTBP superfamily of extracellular matrix proteins, and has been demonstrated to exhibit tumor-promoting and tumor-suppressive functions in different types of cancer. However, the function of LTBP2 in gastric cancer (GC) remains unknown. The aim of the present study was to investigate the expression and molecular function of LTBP2 in GC, and to evaluate its prognostic value for patients with GC. The results revealed that the expression of LTBP2 was upregulated in GC tissues and cell lines. Increased LTBP2 expression was associated with poor overall survival in patients with early-stage [tumor-node-metastasis (TNM) I/II] and late-stage (TNM III/IV) GC. Furthermore, silencing of LTBP2 effectively suppressed the proliferation, migration, invasion and epithelial-mesenchymal transition in GC cells. These results suggested that LTBP2 may be considered as a potential therapeutic target and a promising prognostic biomarker for human GC.
Collapse
Affiliation(s)
- Jun Wang
- Fourth Department of General Surgery, The First Hospital of Lanzhou University, Lanzhou, Gansu 730000, P.R. China
| | - Wen-Jia Liang
- Department of Ultrasound, Gansu Provincial Hospital, Lanzhou, Gansu 730000, P.R. China
| | - Guang-Tao Min
- Fourth Department of General Surgery, The First Hospital of Lanzhou University, Lanzhou, Gansu 730000, P.R. China
| | - Hong-Peng Wang
- Fourth Department of General Surgery, The First Hospital of Lanzhou University, Lanzhou, Gansu 730000, P.R. China
| | - Wei Chen
- Fourth Department of General Surgery, The First Hospital of Lanzhou University, Lanzhou, Gansu 730000, P.R. China
| | - Nan Yao
- Fourth Department of General Surgery, The First Hospital of Lanzhou University, Lanzhou, Gansu 730000, P.R. China
| |
Collapse
|
11
|
Chai AWY, Cheung AKL, Dai W, Ko JMY, Ip JCY, Chan KW, Kwong DLW, Ng WT, Lee AWM, Ngan RKC, Yau CC, Tung SY, Lee VHF, Lam AKY, Pillai S, Law S, Lung ML. Metastasis-suppressing NID2, an epigenetically-silenced gene, in the pathogenesis of nasopharyngeal carcinoma and esophageal squamous cell carcinoma. Oncotarget 2018; 7:78859-78871. [PMID: 27793011 PMCID: PMC5346683 DOI: 10.18632/oncotarget.12889] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2016] [Accepted: 10/19/2016] [Indexed: 12/13/2022] Open
Abstract
Nidogen-2 (NID2) is a key component of the basement membrane that stabilizes the extracellular matrix (ECM) network. The aim of the study is to analyze the functional roles of NID2 in the pathogenesis of nasopharyngeal carcinoma (NPC) and esophageal squamous cell carcinoma (ESCC). We performed genome-wide methylation profiling of NPC and ESCC and validated our findings using the methylation-sensitive high-resolution melting (MS-HRM) assay. Results showed that promoter methylation of NID2 was significantly higher in NPC and ESCC samples than in their adjacent non-cancer counterparts. Consistently, down-regulation of NID2 was observed in the clinical samples and cell lines of both NPC and ESCC. Re-expression of NID2 suppresses clonogenic survival and migration abilities of transduced NPC and ESCC cells. We showed that NID2 significantly inhibits liver metastasis. Mechanistic studies of signaling pathways also confirm that NID2 suppresses the EGFR/Akt and integrin/FAK/PLCγ metastasis-related pathways. This study provides novel insights into the crucial tumor metastasis suppression roles of NID2 in cancers.
Collapse
Affiliation(s)
- Annie Wai Yeeng Chai
- Department of Clinical Oncology, The University of Hong Kong, Hong Kong (SAR), People's Republic of China
| | - Arthur Kwok Leung Cheung
- Department of Clinical Oncology, The University of Hong Kong, Hong Kong (SAR), People's Republic of China
| | - Wei Dai
- Department of Clinical Oncology, The University of Hong Kong, Hong Kong (SAR), People's Republic of China
| | - Josephine Mun Yee Ko
- Department of Clinical Oncology, The University of Hong Kong, Hong Kong (SAR), People's Republic of China
| | - Joseph Chok Yan Ip
- Department of Clinical Oncology, The University of Hong Kong, Hong Kong (SAR), People's Republic of China
| | - Kwok Wah Chan
- Center for Cancer Research, The University of Hong Kong, Hong Kong (SAR), People's Republic of China.,Department of Pathology, The University of Hong Kong, Hong Kong (SAR), People's Republic of China
| | - Dora Lai-Wan Kwong
- Department of Clinical Oncology, The University of Hong Kong, Hong Kong (SAR), People's Republic of China.,Center for Nasopharyngeal Carcinoma Research, The University of Hong Kong, Hong Kong (SAR), People's Republic of China
| | - Wai Tong Ng
- Center for Nasopharyngeal Carcinoma Research, The University of Hong Kong, Hong Kong (SAR), People's Republic of China.,Department of Clinical Oncology, Pamela Youde Nethersole Eastern Hospital, Hong Kong (SAR), People's Republic of China
| | - Anne Wing Mui Lee
- Department of Clinical Oncology, The University of Hong Kong, Hong Kong (SAR), People's Republic of China.,Center for Nasopharyngeal Carcinoma Research, The University of Hong Kong, Hong Kong (SAR), People's Republic of China
| | - Roger Kai Cheong Ngan
- Center for Nasopharyngeal Carcinoma Research, The University of Hong Kong, Hong Kong (SAR), People's Republic of China.,Department of Clinical Oncology, Queen Elizabeth Hospital, Hong Kong (SAR), People's Republic of China
| | - Chun Chung Yau
- Center for Nasopharyngeal Carcinoma Research, The University of Hong Kong, Hong Kong (SAR), People's Republic of China.,Department of Oncology, Princess Margaret Hospital, Hong Kong (SAR), People's Republic of China
| | - Stewart Yuk Tung
- Center for Nasopharyngeal Carcinoma Research, The University of Hong Kong, Hong Kong (SAR), People's Republic of China.,Department of Clinical Oncology, Tuen Mun Hospital, Hong Kong (SAR), People's Republic of China
| | - Victor Ho Fun Lee
- Department of Clinical Oncology, The University of Hong Kong, Hong Kong (SAR), People's Republic of China.,Center for Nasopharyngeal Carcinoma Research, The University of Hong Kong, Hong Kong (SAR), People's Republic of China
| | - Alfred King-Yin Lam
- Department of Cancer Molecular Pathology, Griffith Medical School and Menzies Health Institute Queensland, Griffith University, Gold Coast, Australia
| | - Suja Pillai
- Department of Cancer Molecular Pathology, Griffith Medical School and Menzies Health Institute Queensland, Griffith University, Gold Coast, Australia
| | - Simon Law
- Center for Cancer Research, The University of Hong Kong, Hong Kong (SAR), People's Republic of China.,Department of Surgery, The University of Hong Kong, Hong Kong (SAR), People's Republic of China
| | - Maria Li Lung
- Department of Clinical Oncology, The University of Hong Kong, Hong Kong (SAR), People's Republic of China.,Center for Cancer Research, The University of Hong Kong, Hong Kong (SAR), People's Republic of China.,Center for Nasopharyngeal Carcinoma Research, The University of Hong Kong, Hong Kong (SAR), People's Republic of China
| |
Collapse
|
12
|
Han L, Tang MM, Xu X, Jiang B, Huang J, Feng X, Qiang J. LTBP2 is a prognostic marker in head and neck squamous cell carcinoma. Oncotarget 2018; 7:45052-45059. [PMID: 27281608 PMCID: PMC5216705 DOI: 10.18632/oncotarget.8855] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2016] [Accepted: 04/02/2016] [Indexed: 12/15/2022] Open
Abstract
Latent transforming growth factor (TGF)-beta binding protein 2 (LTBP2) belongs to the fibrillin/LTBP extracellular matrix glycoprotein superfamily. It plays vital roles in tumorigenesis through regulating TGFβ activity, elastogenesis and maintenance of the extracellular matrix (ECM) structure. In this study, we determined the expression levels of LTBP2 mRNA and protein in head and neck squamous cell carcinoma (HNSCC) tissues and adjacent normal tissues by quantitative reverse transcription PCR (qRT-PCR) and tissue microarray immunohistochemistry analysis (TMA-IHC) respectively. LTBP2 protein levels in cancer tissues were correlated with HNSCC patients' clinical characteristics and overall survival. Both LTBP2 mRNA and protein levels were significantly higher in HNSCC tissues than in adjacent normal tissues. High LTBP2 protein level was associated with lymph node metastasis and higher pTNM stages. High LTBP2 protein level is an independent prognostic marker in HNSCC. Our data suggest that LTBP2 acts as an oncogene in HNSCC development and progression. Detection of LTBP2 expression could be a useful prognosis marker and targeting LTBP2 may represent a novel strategy for cancer treatment through regulating activities of TGFβ.
Collapse
Affiliation(s)
- Liang Han
- Department of Head and Neck Surgery, Affiliated Tumor Hospital of Nantong University, Nantong Tumor Hospital, Nantong, Jiangsu, China
| | - Ming Ming Tang
- Department of Head and Neck Surgery, Affiliated Tumor Hospital of Nantong University, Nantong Tumor Hospital, Nantong, Jiangsu, China
| | - Xinjiang Xu
- Department of Head and Neck Surgery, Affiliated Tumor Hospital of Nantong University, Nantong Tumor Hospital, Nantong, Jiangsu, China
| | - Bin Jiang
- Department of Head and Neck Surgery, Affiliated Tumor Hospital of Nantong University, Nantong Tumor Hospital, Nantong, Jiangsu, China
| | - Jianfei Huang
- Department of Clinical Pathology, Affiliated Hospital of Nantong University, Nantong, Jiangsu, China
| | - Xingmei Feng
- Department of Stomatology, Affiliated Hospital of Nantong University, Nantong, Jiangsu, China
| | - Jianfeng Qiang
- Department of Graduate, Medical School of Nantong University, Nantong, Jiangsu, China
| |
Collapse
|
13
|
Liu Q, Zhang H, Jiang X, Qian C, Liu Z, Luo D. Factors involved in cancer metastasis: a better understanding to "seed and soil" hypothesis. Mol Cancer 2017; 16:176. [PMID: 29197379 PMCID: PMC5712107 DOI: 10.1186/s12943-017-0742-4] [Citation(s) in RCA: 215] [Impact Index Per Article: 26.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2017] [Accepted: 11/07/2017] [Indexed: 02/07/2023] Open
Abstract
Metastasis has intrigued researchers for more than 100 years. Despite the development of technologies and therapeutic strategies, metastasis is still the major cause of cancer-related death until today. The famous "seed and soil" hypothesis is widely cited and accepted, and it still provides significant instructions in cancer research until today. To our knowledge, there are few reviews that comprehensively and correlatively focus on both the seed and soil factors involved in cancer metastasis; moreover, despite the fact that increasingly underlying mechanisms and concepts have been defined recently, previous perspectives are appealing but may be limited. Hence, we reviewed factors involved in cancer metastasis, including both seed and soil factors. By integrating new concepts with the classic hypothesis, we aim to provide a comprehensive understanding of the "seed and soil" hypothesis and to conceptualize the framework for understanding factors involved in cancer metastasis. Based on a dynamic overview of this field, we also discuss potential implications for future research and clinical therapeutic strategies.
Collapse
Affiliation(s)
- Qiang Liu
- First Clinical Medical College, School of Medicine, Nanchang University, Nanchang, People's Republic of China
| | - Hongfei Zhang
- Queen Mary School, School of Medicine, Nanchang University, Nanchang, People's Republic of China
| | - Xiaoli Jiang
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Nanchang University, Bayi Road, No.461, 330006, Nanchang, People's Republic of China
| | - Caiyun Qian
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Nanchang University, Bayi Road, No.461, 330006, Nanchang, People's Republic of China
| | - Zhuoqi Liu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Nanchang University, Bayi Road, No.461, 330006, Nanchang, People's Republic of China.
| | - Daya Luo
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Nanchang University, Bayi Road, No.461, 330006, Nanchang, People's Republic of China.
- Jiangxi Province Key Laboratory of Tumor Pathogens and Molecular Pathology, Nanchang University, Nanchang, Bayi Road, No.461, 330006, Nanchang, People's Republic of China.
| |
Collapse
|
14
|
Gao XL, Zhang M, Tang YL, Liang XH. Cancer cell dormancy: mechanisms and implications of cancer recurrence and metastasis. Onco Targets Ther 2017; 10:5219-5228. [PMID: 29138574 PMCID: PMC5667781 DOI: 10.2147/ott.s140854] [Citation(s) in RCA: 90] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
More recently, disease metastasis and relapse in many cancer patients several years (even some decades) after surgical remission are regarded as tumor dormancy. However, the knowledge of this phenomenon is cripplingly limited. Substantial quantities of reviews have summarized three main potential models that can be put forth to explain such process, including angiogenic dormancy, immunologic dormancy, and cellular dormancy. In this review, newly uncovered mechanisms governing cancer cell dormancy are discussed, with an emphasis on the cross talk between dormant cancer cells and their microenvironments. In addition, potential mechanisms of reactivation of these dormant cells in certain anatomic sites including lymph nodes and bone marrow are discussed. Molecular mechanism of cellular dormancy in head and neck cancer is also involved.
Collapse
Affiliation(s)
- Xiao-Lei Gao
- State Key Laboratory of Oral Diseases.,Department of Oral and Maxillofacial Surgery
| | - Mei Zhang
- State Key Laboratory of Oral Diseases.,Department of Oral Pathology, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, People's Republic of China
| | - Ya-Ling Tang
- State Key Laboratory of Oral Diseases.,Department of Oral Pathology, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, People's Republic of China
| | - Xin-Hua Liang
- State Key Laboratory of Oral Diseases.,Department of Oral and Maxillofacial Surgery
| |
Collapse
|
15
|
Wang C, Wang G, Zhang L, Pan J, Wei Y. Latent Transforming Growth Factor β Binding Protein 2 (LTBP2) as a Novel Biomarker for the Diagnosis and Prognosis of Pancreatic Carcinoma. Med Sci Monit 2017; 23:3232-3239. [PMID: 28669978 PMCID: PMC5507795 DOI: 10.12659/msm.905284] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2017] [Accepted: 06/05/2017] [Indexed: 01/01/2023] Open
Abstract
BACKGROUND Latent transforming growth factor b binding protein 2 (LTBP2) is proven to be associated with ECM and involved in the advancement of several kinds of cancer. The present study evaluated the diagnosis and prognosis of pancreatic carcinoma (PC) using LTBP2 as a biomarker. MATERIAL AND METHODS Protein levels of LTBP2 were evaluated in 111 pairs of pancreatic ductal adenocarcinoma (PDAC) tissues and adjacent nontumor tissues via immunohistochemistry. ELISA method was used to quantify the serum concentration of LTBP2. The subjects in this study included 141 PDAC patients, 20 patients with benign pancreatic disease, and 20 healthy volunteers. RESULTS IHC results showed that LTBP2 levels were significantly elevated in the PDAC tissues as compared with the adjacent nontumor tissues (P<0.05). Sixty-one of the 111 (54.9%) PDAC tissues showed high expression of the protein. LTBP2 overexpression was significantly correlated with poor differentiation (P=0.018) and advanced TNM stage (P=0.036). Moreover, Kaplan-Meier analysis showed that high levels of LTBP2 predicted worse overall survival (P=0.001) and disease-free survival (P=0.001). Multivariate Cox regression analysis indicated that high expression of LTBP2 was an autonomous prognostic factor for poor overall and disease-free survival (P=0.001; P=0.002). Receiver operating characteristic (ROC) curve analyses of showed that LTBP-2 had an area under the curve (AUC) of 0.846 (95% confidence intervals: 0.757-0.934) and cut-off value of 19.12. CONCLUSIONS LTBP2 is a novel biomarker for the diagnosis of PC and may be a potential target for PDAC clinical therapy.
Collapse
Affiliation(s)
- Cheng Wang
- Corresponding Author: Cheng Wang, e-mail:
| | | | | | | | | |
Collapse
|
16
|
Lu J, Liu Q, Wang L, Tu W, Chu H, Ding W, Jiang S, Ma Y, Shi X, Pu W, Zhou X, Jin L, Wang J, Wu W. Increased expression of latent TGF-β-binding protein 4 affects the fibrotic process in scleroderma by TGF-β/SMAD signaling. J Transl Med 2017; 97:591-601. [PMID: 28263294 DOI: 10.1038/labinvest.2017.20] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2016] [Revised: 01/03/2017] [Accepted: 01/13/2017] [Indexed: 01/06/2023] Open
Abstract
Scleroderma is a fibrosis-related disorder characterized by cutaneous and internal organ fibrosis, and excessive collagen deposition in extracellular matrix (ECM) is a major cause of fibrosis. Transforming growth factor-β (TGF-β)/SMAD signaling has a central role in the pathogenesis of fibrosis by inducing abnormal collagen accumulation in ECM, and latent TGF-β-binding protein 4 (LTBP-4) affects the secretion of latent TGF-β to ECM. A previous study indicated that bleomycin (BLM) treatment increased LTBP-4 expression in lung fibroblasts of Thy-1 knockout mice with lung fibrosis, and LTBP-4 further promoted TGF-β bioavailability as well as SMAD3 phosphorylation. However, the expression and function of LTBP-4 in human scleroderma remain unclear. We aimed to investigate the potential role of LTBP-4 in scleroderma through clinical, in vivo and in vitro studies. LTBP-4 and TGF-β expressions were significantly upregulated in systemic scleroderma (SSc) patients' plasma compared with normal controls (LTBP-4, 1,215±100.2 vs 542.8±41.7 ng/ml, P<0.0001; TGF-β, 1.5±0.2 vs 0.7±0.1 ng/ml, P=0.0031), while no significant difference was found between localized scleroderma (LSc) and normal controls. The plasma concentrations of LTBP-4 and TGF-β were even higher in SSc patients with lung fibrosis (LTBP-4, 1462± 137.3 vs 892.8±113.4 ng/ml, P=0.0037; TGF-β, 2.0±0.4 vs 0.9±0.2 ng/ml, P=0.0212) and esophagus involvement (1390±134.4 vs 940.7±127.0 ng/ml, P=0.0269; TGF-β, 1.9±0.3 vs 0.9±0.2 ng/ml, P=0.0426). The area under receiver operating characteristics (ROC) curve of LTBP-4 was 0.86. Immunohistochemistry measurement also demonstrated a higher LTBP-4 expression in sclerotic skin tissue of LSc and SSc compared with normal controls. More positive fibroblasts were also found in BLM-induced scleroderma mouse model than the saline-treated group. In in vitro studies, knockdown of LTBP-4 in SSc skin fibroblasts prominently reduced downstream COL1A1, COL1A2, and COL3A1 mRNA level by 84%, 82%, and 43%, respectively, and other fibrosis-related genes' expression were also decreased. Furthermore, extracellular TGF-β level and the SMAD2/3 phosphorylation were inhibited through LTBP-4 knockdown treatment, suggesting that the knockdown of LTBP-4 reduced the collagen expression through TGF-β/SMAD signaling pathway. Taken together, these data suggest that LTBP-4 affects fibrotic process in scleroderma, and the high expression of LTBP-4 in SSc plasma may serve as a clinical biomarker in diagnosing this disease. In addition, this study also lays the theoretical foundation for targeting LTBP-4 as treatment of scleroderma.
Collapse
Affiliation(s)
- Jiaying Lu
- Department of Dermatology, Huashan Hospital, Fudan University, Shanghai, China
| | - Qingmei Liu
- Department of Dermatology, Huashan Hospital, Fudan University, Shanghai, China.,State Key Laboratory of Genetic Engineering, Collaborative Innovation Center for Genetics and Development, School of Life Sciences, Fudan University, Shanghai, China
| | - Lei Wang
- Division of Rheumatology, Shanghai TCM-integrated Hospital, Shanghai, China
| | - Wenzhen Tu
- Division of Rheumatology, Shanghai TCM-integrated Hospital, Shanghai, China
| | - Haiyan Chu
- State Key Laboratory of Genetic Engineering, Collaborative Innovation Center for Genetics and Development, School of Life Sciences, Fudan University, Shanghai, China
| | - Weifeng Ding
- State Key Laboratory of Genetic Engineering, Collaborative Innovation Center for Genetics and Development, School of Life Sciences, Fudan University, Shanghai, China
| | - Shuai Jiang
- State Key Laboratory of Genetic Engineering, Collaborative Innovation Center for Genetics and Development, School of Life Sciences, Fudan University, Shanghai, China
| | - Yanyun Ma
- State Key Laboratory of Genetic Engineering, Collaborative Innovation Center for Genetics and Development, School of Life Sciences, Fudan University, Shanghai, China
| | - Xiangguang Shi
- State Key Laboratory of Genetic Engineering, Collaborative Innovation Center for Genetics and Development, School of Life Sciences, Fudan University, Shanghai, China
| | - Weilin Pu
- State Key Laboratory of Genetic Engineering, Collaborative Innovation Center for Genetics and Development, School of Life Sciences, Fudan University, Shanghai, China
| | - Xiaodong Zhou
- University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Li Jin
- State Key Laboratory of Genetic Engineering, Collaborative Innovation Center for Genetics and Development, School of Life Sciences, Fudan University, Shanghai, China
| | - Jiucun Wang
- State Key Laboratory of Genetic Engineering, Collaborative Innovation Center for Genetics and Development, School of Life Sciences, Fudan University, Shanghai, China.,Institute of Rheumatology, Immunology and Allergy, Fudan University, Shanghai, China
| | - Wenyu Wu
- Department of Dermatology, Huashan Hospital, Fudan University, Shanghai, China
| |
Collapse
|
17
|
Wan F, Peng L, Zhu C, Zhang X, Chen F, Liu T. Knockdown of Latent Transforming Growth Factor-β (TGF-β)-Binding Protein 2 (LTBP2) Inhibits Invasion and Tumorigenesis in Thyroid Carcinoma Cells. Oncol Res 2016; 25:503-510. [PMID: 27712597 PMCID: PMC7841189 DOI: 10.3727/096504016x14755368915591] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Latent transforming growth factor-β (TGF-β)-binding protein 2 (LTBP2) is one of four proteins in the LTBP family of proteins (LTBP1-4) and was shown to play a vital role in tumorigenesis. However, little is known regarding the functional role of LTBP2 in thyroid carcinoma. Therefore, the current study aimed to evaluate the effect of LTBP2 expression on the proliferation, invasion, and tumorigenesis in thyroid carcinoma cells and to explore the molecular mechanism of LTBP2 in tumor progression. Our results showed that the expression of LTBP2 is upregulated in human thyroid carcinoma and cell lines. Knockdown of LTBP2 inhibits the proliferation, invasion, and EMT phenotype in thyroid carcinoma cells. Furthermore, knockdown of LTBP2 attenuates thyroid carcinoma growth in nude mice. Finally, knockdown of LTBP2 inhibits activation of the PI3K/Akt pathway in thyroid carcinoma cells. In summary, the present study has provided further evidence that knockdown of LTBP2 inhibits invasion and tumorigenesis in thyroid carcinoma cells. Our findings may help to further elucidate the molecular mechanisms underlying thyroid carcinoma progression and provide candidate targets for the prevention and treatment of thyroid carcinoma.
Collapse
Affiliation(s)
- Fuqiang Wan
- Department of Head and Neck Surgery, Linyi Tumor Hospital, Linyi, P.R. China
| | - Li Peng
- Department of Head and Neck Surgery, Linyi Tumor Hospital, Linyi, P.R. China
| | - ChaoYu Zhu
- Department No. 2 of Abdominal Surgery, Linyi Tumor Hospital, Linyi, P.R. China
| | - XinFa Zhang
- Department of Breast Surgery, Linyi Tumor Hospital, Linyi, P.R. China
| | - FangWen Chen
- Department of Head and Neck Surgery, Linyi Tumor Hospital, Linyi, P.R. China
| | - Tao Liu
- Department of General Surgery, Linyi People's Hospital, Linyi, P.R. China
| |
Collapse
|
18
|
Mitra A, Mishra L, Li S. EMT, CTCs and CSCs in tumor relapse and drug-resistance. Oncotarget 2016; 6:10697-711. [PMID: 25986923 PMCID: PMC4484413 DOI: 10.18632/oncotarget.4037] [Citation(s) in RCA: 388] [Impact Index Per Article: 43.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2015] [Accepted: 04/20/2015] [Indexed: 12/15/2022] Open
Abstract
Tumor relapse and metastasis are the primary causes of poor survival rates in patients with advanced cancer despite successful resection or chemotherapeutic treatment. A primary cause of relapse and metastasis is the persistence of cancer stem cells (CSCs), which are highly resistant to chemotherapy. Although highly efficacious drugs suppressing several subpopulations of CSCs in various tissue-specific cancers are available, recurrence is still common in patients. To find more suitable therapy for relapse, the mechanisms underlying metastasis and drug-resistance associated with relapse-initiating CSCs need to be identified. Recent studies in circulating tumor cells (CTCs) of some cancer patients manifest phenotypes of both CSCs and epithelial-mesenchymal transition (EMT). These patients are unresponsive to standard chemotherapies and have low progression free survival, suggesting that EMT-positive CTCs are related to co-occur with or transform into relapse-initiating CSCs. Furthermore, EMT programming in cancer cells enables in the remodeling of extracellular matrix to break the dormancy of relapse-initiating CSCs. In this review, we extensively discuss the association of the EMT program with CTCs and CSCs to characterize a subpopulation of patients prone to relapses. Identifying the mechanisms by which EMT-transformed CTCs and CSCs initiate relapse could facilitate the development of new or enhanced personalized therapeutic regimens.
Collapse
Affiliation(s)
- Abhisek Mitra
- Department of Pediatrics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Lopa Mishra
- Department of Gastroenterology, Hepatology and Nutrition, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Shulin Li
- Department of Pediatrics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|
19
|
Evans EB, Lin SY. New insights into tumor dormancy: Targeting DNA repair pathways. World J Clin Oncol 2015; 6:80-88. [PMID: 26468441 PMCID: PMC4600194 DOI: 10.5306/wjco.v6.i5.80] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/29/2015] [Revised: 07/02/2015] [Accepted: 07/27/2015] [Indexed: 02/06/2023] Open
Abstract
Over the past few decades, major strides have advanced the techniques for early detection and treatment of cancer. However, metastatic tumor growth still accounts for the majority of cancer-related deaths worldwide. In fact, breast cancers are notorious for relapsing years or decades after the initial clinical treatment, and this relapse can vary according to the type of breast cancer. In estrogen receptor-positive breast cancers, late tumor relapses frequently occur whereas relapses in estrogen receptor-negative cancers or triple negative tumors arise early resulting in a higher mortality risk. One of the main causes of metastasis is tumor dormancy in which cancer cells remain concealed, asymptomatic, and untraceable over a prolonged period of time. Under certain conditions, dormant cells can re-enter into the cell cycle and resume proliferation leading to recurrence. However, the molecular and cellular regulators underlying this transition remain poorly understood. To date, three mechanisms have been identified to trigger tumor dormancy including cellular, angiogenic, and immunologic dormancies. In addition, recent studies have suggested that DNA repair mechanisms may contribute to the survival of dormant cancer cells. In this article, we summarize the recent experimental and clinical evidence governing cancer dormancy. In addition, we will discuss the role of DNA repair mechanisms in promoting the survival of dormant cells. This information provides mechanistic insight to explain why recurrence occurs, and strategies that may enhance therapeutic approaches to prevent disease recurrence.
Collapse
|
20
|
Lung ML, Cheung AKL, Ko JMY, Lung HL, Cheng Y, Dai W. The interplay of host genetic factors and Epstein-Barr virus in the development of nasopharyngeal carcinoma. CHINESE JOURNAL OF CANCER 2015; 33:556-68. [PMID: 25367335 PMCID: PMC4244319 DOI: 10.5732/cjc.014.10170] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
The interplay between host cell genetics and Epstein-Barr virus (EBV) infection contributes to the development of nasopharyngeal carcinoma (NPC). Understanding the host genetic and epigenetic alterations and the influence of EBV on cell signaling and host gene regulation will aid in understanding the molecular pathogenesis of NPC and provide useful biomarkers and targets for diagnosis and therapy. In this review, we provide an update of the oncogenes and tumor suppressor genes associated with NPC, as well as genes associated with NPC risk including those involved in carcinogen detoxification and DNA repair. We also describe the importance of host genetics that govern the human leukocyte antigen (HLA) complex and immune responses, and we describe the impact of EBV infection on host cell signaling changes and epigenetic regulation of gene expression. High-power genomic sequencing approaches are needed to elucidate the genetic basis for inherited susceptibility to NPC and to identify the genes and pathways driving its molecular pathogenesis.
Collapse
Affiliation(s)
- Maria Li Lung
- Department of Clinical Oncology and Center for Nasopharyngeal Carcinoma Research, University of Hong Kong, Hong Kong, P. R. China.
| | | | | | | | | | | |
Collapse
|
21
|
Phoon YP, Cheung AKL, Cheung FMF, Chan KF, Wong S, Wong BWY, Tung SY, Yau CC, Ng WT, Lung ML. IKBB tumor suppressive role in nasopharyngeal carcinomaviaNF-κB-mediated signalling. Int J Cancer 2015; 138:160-70. [PMID: 26227166 DOI: 10.1002/ijc.29702] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2015] [Revised: 06/29/2015] [Accepted: 07/21/2015] [Indexed: 12/30/2022]
Affiliation(s)
- Yee Peng Phoon
- Department of Clinical Oncology; University of Hong Kong; People's Republic of China
| | - Arthur Kwok Leung Cheung
- Department of Clinical Oncology; University of Hong Kong; People's Republic of China
- Center for Cancer Research, University of Hong Kong; People's Republic of China
| | - Florence Man Fung Cheung
- Center for Nasopharyngeal Carcinoma Research, University of Hong Kong; People's Republic of China
- Department of Pathology; HKU-Shenzhen Hospital; Shenzhen People's Republic of China
| | - Kui Fat Chan
- Department of Pathology; Tuen Mun Hospital; Hong Kong
| | - Shun Wong
- Department of Pathology; Princess Margaret Hospital; Hong Kong
| | - Bonnie Wing Yan Wong
- Department of Clinical Oncology; University of Hong Kong; People's Republic of China
| | - Stewart Yuk Tung
- Center for Nasopharyngeal Carcinoma Research, University of Hong Kong; People's Republic of China
- Department of Clinical Oncology; Tuen Mun Hospital; Hong Kong
| | - Chun Chung Yau
- Center for Nasopharyngeal Carcinoma Research, University of Hong Kong; People's Republic of China
- Department of Oncology; Princess Margaret Hospital; Hong Kong
| | - Wai Tong Ng
- Center for Nasopharyngeal Carcinoma Research, University of Hong Kong; People's Republic of China
- Department of Clinical Oncology; Pamela Youde Nethersole Eastern Hospital; Hong Kong
| | - Maria Li Lung
- Department of Clinical Oncology; University of Hong Kong; People's Republic of China
- Center for Cancer Research, University of Hong Kong; People's Republic of China
- Center for Nasopharyngeal Carcinoma Research, University of Hong Kong; People's Republic of China
| |
Collapse
|
22
|
Kan R, Shuen WH, Lung HL, Cheung AKL, Dai W, Kwong DLW, Ng WT, Lee AWM, Yau CC, Ngan RKC, Tung SY, Lung ML. NF-κB p65 Subunit Is Modulated by Latent Transforming Growth Factor-β Binding Protein 2 (LTBP2) in Nasopharyngeal Carcinoma HONE1 and HK1 Cells. PLoS One 2015; 10:e0127239. [PMID: 25974126 PMCID: PMC4431814 DOI: 10.1371/journal.pone.0127239] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2014] [Accepted: 04/12/2015] [Indexed: 01/22/2023] Open
Abstract
NF-κB is a well-characterized transcription factor, widely known as a key player in tumor-derived inflammation and cancer development. Herein, we present the functional and molecular relevance of the canonical NF-κB p65 subunit in nasopharyngeal carcinoma (NPC). Loss- and gain-of-function approaches were utilized to reveal the functional characteristics of p65 in propagating tumor growth, tumor-associated angiogenesis, and epithelial-to-mesenchymal transition in NPC cells. Extracellular inflammatory stimuli are critical factors that trigger the NF-κB p65 signaling; hence, we investigated the components of the tumor microenvironment that might potentially influence the p65 signaling pathway. This led to the identification of an extracellular matrix (ECM) protein that was previously reported as a candidate tumor suppressor in NPC. Our studies on the Latent Transforming Growth Factor-β Binding Protein 2 (LTBP2) protein provides substantial evidence that it can modulate the p65 transcriptional activity. Re-expression of LTBP2 elicits tumor suppressive effects that parallel the inactivation of p65 in NPC cells. LTBP2 was able to reduce phosphorylation of p65 at Serine 536, inhibit nuclear localization of active phosphorylated p65, and impair the p65 DNA-binding ability. This results in a consequential down-regulation of p65-related gene expression. Therefore, the data suggest that the overall up-regulation of p65 expression and the loss of this candidate ECM tumor suppressor are milestone events contributing to NPC development.
Collapse
Affiliation(s)
- Rebecca Kan
- Department of Clinical Oncology, Li Ka Shing Faculty of Medicine, University of Hong Kong, Hong Kong (SAR), PR China
| | - Wai Ho Shuen
- Department of Clinical Oncology, Li Ka Shing Faculty of Medicine, University of Hong Kong, Hong Kong (SAR), PR China
| | - Hong Lok Lung
- Department of Clinical Oncology, Li Ka Shing Faculty of Medicine, University of Hong Kong, Hong Kong (SAR), PR China
- Center for Cancer Research, Li Ka Shing Faculty of Medicine, University of Hong Kong, Hong Kong (SAR), PR China
| | - Arthur Kwok Leung Cheung
- Department of Clinical Oncology, Li Ka Shing Faculty of Medicine, University of Hong Kong, Hong Kong (SAR), PR China
- Center for Cancer Research, Li Ka Shing Faculty of Medicine, University of Hong Kong, Hong Kong (SAR), PR China
| | - Wei Dai
- Department of Clinical Oncology, Li Ka Shing Faculty of Medicine, University of Hong Kong, Hong Kong (SAR), PR China
| | - Dora Lai-Wan Kwong
- Department of Clinical Oncology, Li Ka Shing Faculty of Medicine, University of Hong Kong, Hong Kong (SAR), PR China
- Center for Cancer Research, Li Ka Shing Faculty of Medicine, University of Hong Kong, Hong Kong (SAR), PR China
- Center for Nasopharyngeal Carcinoma Research, Li Ka Shing Faculty of Medicine, University of Hong Kong, Hong Kong (SAR), PR China
| | - Wai Tong Ng
- Center for Nasopharyngeal Carcinoma Research, Li Ka Shing Faculty of Medicine, University of Hong Kong, Hong Kong (SAR), PR China
- Department of Clinical Oncology, Pamela Youde Nethersole Eastern Hospital, Hong Kong (SAR), PR China
| | - Anne Wing Mui Lee
- Center for Nasopharyngeal Carcinoma Research, Li Ka Shing Faculty of Medicine, University of Hong Kong, Hong Kong (SAR), PR China
- Department of Clinical Oncology, The University of Hong Kong—Shen Zhen Hospital, PR China
| | - Chun Chung Yau
- Center for Nasopharyngeal Carcinoma Research, Li Ka Shing Faculty of Medicine, University of Hong Kong, Hong Kong (SAR), PR China
- Department of Oncology, Princess Margaret Hospital, Hong Kong (SAR), PR China
| | - Roger Kai Cheong Ngan
- Center for Nasopharyngeal Carcinoma Research, Li Ka Shing Faculty of Medicine, University of Hong Kong, Hong Kong (SAR), PR China
- Department of Clinical Oncology, Queen Elizabeth Hospital, Hong Kong (SAR), PR China
| | - Stewart Yuk Tung
- Center for Nasopharyngeal Carcinoma Research, Li Ka Shing Faculty of Medicine, University of Hong Kong, Hong Kong (SAR), PR China
- Department of Clinical Oncology, Tuen Mun Hospital, Hong Kong (SAR), PR China
| | - Maria Li Lung
- Department of Clinical Oncology, Li Ka Shing Faculty of Medicine, University of Hong Kong, Hong Kong (SAR), PR China
- Center for Cancer Research, Li Ka Shing Faculty of Medicine, University of Hong Kong, Hong Kong (SAR), PR China
- Center for Nasopharyngeal Carcinoma Research, Li Ka Shing Faculty of Medicine, University of Hong Kong, Hong Kong (SAR), PR China
- * E-mail:
| |
Collapse
|
23
|
Chiang MS, Yang JR, Liao SC, Hsu CC, Hsu CW, Yuan K. Latent transforming growth factor-β binding proteins (LTBP-1 and LTBP-2) and gingiva keratinization. Oral Dis 2015; 21:762-9. [PMID: 25858550 DOI: 10.1111/odi.12344] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2014] [Revised: 03/31/2015] [Accepted: 04/01/2015] [Indexed: 11/30/2022]
Abstract
OBJECTIVE Transforming growth factor-beta (TGF-β) proteins are involved in epithelial keratinization. The major function of latent TGF-β binding proteins (LTBPs) is modulating TGF-β activity. However, whether LTBP-1 and LTBP-2 play roles in gingiva keratinization remains unclear. MATERIALS AND METHODS Human keratinized gingiva and non-keratinized alveolar mucosa were processed for LTBP-1, LTBP-2, cytokeratin-1 (K1), cytokeratin-4 (K4), and TGF-β immunohistochemical (IHC) staining. Porcine heterotopically transplanted connective tissues and newly grown epithelia were harvested for IHC staining. The expression levels of LTBP-1 and LTBP-2 were compared between differentiated and undifferentiated human normal oral keratinocytes (hNOK). The expression of LTBP-1 and LTBP-2 was knocked down in a cell line (OEC-M1) to evaluate the effects on the expression of K1, K4, and involucrin (INV). RESULTS In human and porcine specimens, LTBP-2 expression patterns distinguished keratinized and non-keratinized oral epithelia. Western blotting results showed that K1, LTBP-1, and INV proteins were upregulated in differentiated hNOK. In OEC-M1 cells, LTBP-2 knockdown resulted in upregulated the expression of K1 and INV and downregulated the expression of K4. LTBP-1 knockdown resulted in opposite effects. CONCLUSION The expression patterns of LTBP-2 differ in keratinized gingiva and non-keratinized mucosa. LTBP-1 and LTBP-2 are involved in the keratinization of oral epithelium; however, the underlying mechanism remains to be elucidated.
Collapse
Affiliation(s)
- M-S Chiang
- Department of Oral Medicine, National Cheng Kung University Hospital, Tainan, Taiwan.,Institute of Oral Medicine, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - J-R Yang
- Division of Physiology, Livestock Research Institute, Council of Agriculture, Executive Yuan, Tainan, Taiwan
| | - S-C Liao
- Department of Oral Medicine, National Cheng Kung University Hospital, Tainan, Taiwan
| | - C-C Hsu
- Department of Oral Medicine, National Cheng Kung University Hospital, Tainan, Taiwan
| | - C-W Hsu
- Institute of Oral Medicine, College of Medicine, National Cheng Kung University, Tainan, Taiwan.,Dental Department, Tainan Municipal Hospital, Tainan, Taiwan
| | - K Yuan
- Department of Oral Medicine, National Cheng Kung University Hospital, Tainan, Taiwan.,Institute of Oral Medicine, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| |
Collapse
|
24
|
Translational research in nasopharyngeal carcinoma. Oral Oncol 2014; 50:345-52. [DOI: 10.1016/j.oraloncology.2013.11.005] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2013] [Revised: 11/11/2013] [Accepted: 11/13/2013] [Indexed: 11/20/2022]
|
25
|
Szeto CYY, Lin CH, Choi SC, Yip TTC, Ngan RKC, Tsao GSW, Li Lung M. Integrated mRNA and microRNA transcriptome sequencing characterizes sequence variants and mRNA-microRNA regulatory network in nasopharyngeal carcinoma model systems. FEBS Open Bio 2014; 4:128-40. [PMID: 24490137 PMCID: PMC3907684 DOI: 10.1016/j.fob.2014.01.004] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2013] [Revised: 01/09/2014] [Accepted: 01/09/2014] [Indexed: 01/28/2023] Open
Abstract
Nasopharyngeal carcinoma (NPC) is a prevalent malignancy in Southeast Asia among the Chinese population. Aberrant regulation of transcripts has been implicated in many types of cancers including NPC. Herein, we characterized mRNA and miRNA transcriptomes by RNA sequencing (RNASeq) of NPC model systems. Matched total mRNA and small RNA of undifferentiated Epstein–Barr virus (EBV)-positive NPC xenograft X666 and its derived cell line C666, well-differentiated NPC cell line HK1, and the immortalized nasopharyngeal epithelial cell line NP460 were sequenced by Solexa technology. We found 2812 genes and 149 miRNAs (human and EBV) to be differentially expressed in NP460, HK1, C666 and X666 with RNASeq; 533 miRNA–mRNA target pairs were inversely regulated in the three NPC cell lines compared to NP460. Integrated mRNA/miRNA expression profiling and pathway analysis show extracellular matrix organization, Beta-1 integrin cell surface interactions, and the PI3K/AKT, EGFR, ErbB, and Wnt pathways were potentially deregulated in NPC. Real-time quantitative PCR was performed on selected mRNA/miRNAs in order to validate their expression. Transcript sequence variants such as short insertions and deletions (INDEL), single nucleotide variant (SNV), and isomiRs were characterized in the NPC model systems. A novel TP53 transcript variant was identified in NP460, HK1, and C666. Detection of three previously reported novel EBV-encoded BART miRNAs and their isomiRs were also observed. Meta-analysis of a model system to a clinical system aids the choice of different cell lines in NPC studies. This comprehensive characterization of mRNA and miRNA transcriptomes in NPC cell lines and the xenograft provides insights on miRNA regulation of mRNA and valuable resources on transcript variation and regulation in NPC, which are potentially useful for mechanistic and preclinical studies. Using RNASeq we characterized the mRNA and miRNA transcriptomes in NPC and NP models. 2812 Genes and 149 miRNAs (human and EBV) were differentially expressed in NPC vs NP models. 533 miRNA–mRNA target pairs were inversely regulated in HK1, C666, and X666 vs NP460. ECM, β1 integrin, PI3K/AKT, EGFR, ErbB, and Wnt pathways appeared to be deregulated in NPC. A novel TP53 mutation was identified in NP460, HK1, and C666.
Collapse
Key Words
- AIP, aryl hydrocarbon receptor interacting protein
- BAX, BCL2-asscoiated X protein
- CIITA, class II, major histocompatibility complex, transactivator
- DKK1, Dickkopf-Like protein 1
- EBV, Epstein–Barr virus
- ECM, extracellular matrix
- EGFR, epidermal growth factor receptor
- EGR1, early growth response 1
- FBLN2, fibulin 2
- GADD45, growth arrest and DNA-damage-inducible
- GNG11, guanine nucleotide binding protein (G protein), Gamma 11
- GO, gene ontology
- GSTP1, glutathione S-transferase pi 1
- IL18, interleukin 18
- INDEL, insertion and deletion
- LMP1, Epstein–Barr virus latent membrane protein 1
- LTBP2, latent transforming growth factor beta binding protein 2
- MDM2, MDM2 oncogene, E3 ubiquitin protein ligase
- MET, met proto-oncogene
- MMP19, matrix metallopeptidase 19
- NGS, next-generation sequencing
- NPC, nasopharyngeal carcinoma
- Nasopharyngeal carcinoma
- Nasopharyngeal cell lines/xenograft (NP460, HK1, C666, X666)
- PI3K, phosphoinositide 3-kinase
- PTEN, phosphatase and tensin homolog
- RNA sequencing
- RNASeq, RNA sequencing
- SNP, single nucleotide polymorphism
- TNFRSF9, tumour necrosis factor receptor superfamily, member 9
- TP53
- Transcriptome analysis
- UTR, untranslated region
- miRNA, microRNA
Collapse
Affiliation(s)
- Carol Ying-Ying Szeto
- Center for Nasopharyngeal Cancer Research, The University of Hong Kong, PR China ; Department of Clinical Oncology, The University of Hong Kong, PR China
| | - Chi Ho Lin
- Centre for Genomic Sciences, The University of Hong Kong, PR China
| | - Siu Chung Choi
- Centre for Genomic Sciences, The University of Hong Kong, PR China
| | - Timothy T C Yip
- Center for Nasopharyngeal Cancer Research, The University of Hong Kong, PR China ; Department of Clinical Oncology, Queen Elizabeth Hospital, PR China
| | - Roger Kai-Cheong Ngan
- Center for Nasopharyngeal Cancer Research, The University of Hong Kong, PR China ; Department of Clinical Oncology, Queen Elizabeth Hospital, PR China
| | - George Sai-Wah Tsao
- Center for Nasopharyngeal Cancer Research, The University of Hong Kong, PR China ; Department of Anatomy, The University of Hong Kong, PR China
| | - Maria Li Lung
- Center for Nasopharyngeal Cancer Research, The University of Hong Kong, PR China ; Department of Clinical Oncology, The University of Hong Kong, PR China
| |
Collapse
|
26
|
De Wever O, Van Bockstal M, Mareel M, Hendrix A, Bracke M. Carcinoma-associated fibroblasts provide operational flexibility in metastasis. Semin Cancer Biol 2014; 25:33-46. [PMID: 24406210 DOI: 10.1016/j.semcancer.2013.12.009] [Citation(s) in RCA: 97] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2013] [Revised: 12/20/2013] [Accepted: 12/30/2013] [Indexed: 01/11/2023]
Abstract
Malignant cancer cells do not act as lone wolves to achieve metastasis, as they exist within a complex ecosystem consisting of an extracellular matrix scaffold populated by carcinoma-associated fibroblasts (CAFs), endothelial cells and immune cells. We recognize local (primary tumor) and distant ecosystems (metastasis). CAFs, also termed myofibroblasts, may have other functions in the primary tumor versus the metastasis. Cellular origin and tumor heterogeneity lead to the expression of specific markers. The molecular characteristics of a CAF remain in evolution since CAFs show operational flexibility. CAFs respond dynamically to a cancer cell's fluctuating demands by shifting profitable signals necessary in metastasis. Local, tissue-resident fibroblasts and mesenchymal stem cells (MSCs) coming from reservoir sites such as bone marrow and adipose tissue are the main progenitor cells of CAFs. CAFs may induce awakening from metastatic dormancy, a major cause of cancer-specific death. Cancer management protocols influence CAF precursor recruitment and CAF activation. Since CAF signatures represent early changes in metastasis, including formation of pre-metastatic niches, we discuss whether liquid biopsies, including exosomes, may detect and monitor CAF reactions allowing optimized prognosis of cancer patients.
Collapse
Affiliation(s)
- Olivier De Wever
- Laboratory of Experimental Cancer Research, Department of Radiotherapy and Experimental Cancer Research, Ghent University Hospital, Ghent, Belgium.
| | | | - Marc Mareel
- Laboratory of Experimental Cancer Research, Department of Radiotherapy and Experimental Cancer Research, Ghent University Hospital, Ghent, Belgium
| | - An Hendrix
- Laboratory of Experimental Cancer Research, Department of Radiotherapy and Experimental Cancer Research, Ghent University Hospital, Ghent, Belgium
| | - Marc Bracke
- Laboratory of Experimental Cancer Research, Department of Radiotherapy and Experimental Cancer Research, Ghent University Hospital, Ghent, Belgium
| |
Collapse
|
27
|
Wang SH, Lin SY. Tumor dormancy: potential therapeutic target in tumor recurrence and metastasis prevention. Exp Hematol Oncol 2013; 2:29. [PMID: 24502434 PMCID: PMC4176492 DOI: 10.1186/2162-3619-2-29] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2013] [Accepted: 10/12/2013] [Indexed: 12/14/2022] Open
Abstract
In past decades, cancer patient survival has been improved with earlier detection and advancements in therapy. However, many patients who exhibit no clinical symptoms after frontline therapy subsequently suffer, often many years later, aggressive tumor recurrence. Cancer recurrence represents a critical clinical challenge in effectively treating malignancies and for patients’ quality of life. Tumor cell dormancy may help to explain treatment resistance and recurrence or metastatic reactivation. Understanding the dormant stage of tumor cells may help in discovering ways to maintain the dormant state or permanently eliminate dormant residual disseminated tumor cells. Over the past decade, numerous studies indicate that various mechanisms of tumor dormancy exist, including cellular dormancy (quiescence), angiogenic dormancy, and immunologic dormancy. In this short review, we summarize recent experimental and clinical evidence for these three mechanisms and other possible tumor microenvironment mechanisms that may influence tumor dormancy.
Collapse
Affiliation(s)
| | - Shiaw-Yih Lin
- Department of Systems Biology, Unit 950, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd,, Houston, TX 77054, USA.
| |
Collapse
|
28
|
Bultmann I, Conradi A, Kretschmer C, Sterner-Kock A. Latent transforming growth factor β-binding protein 4 is downregulated in esophageal cancer via promoter methylation. PLoS One 2013; 8:e65614. [PMID: 23741501 PMCID: PMC3669142 DOI: 10.1371/journal.pone.0065614] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2013] [Accepted: 04/26/2013] [Indexed: 02/07/2023] Open
Abstract
Latent transforming growth factor β-binding protein 4 (LTBP4) is an extracellular matrix molecule that is a member of important connective tissue networks and is needed for the correct folding and the secretion of TGF-β1. LTBP4 is downregulated in carcinomas of various tissues. Here we show that LTBP4 is also downregulated in adenocarcinomas and squamous cell carcinomas of the esophagus in vitro and in vivo. Re-expression of LTBP4 in esophageal cancer cell lines reduced cell migration ability, whereas cell viability and cell proliferation remained unchanged. Hypermethylation of the promoter regions of the two main human LTBP4 transcriptional forms, LTBP4L and LTBP4S, was found to be involved in LTBP4 silencing. Detailed investigations of the methylation patterns of the promoter regions of LTBP4L and LTBP4S identified GATA1, SP1, E2F4 and SMAD3 as potential transcription factors involved in LTBP4 expression. In in vitro transcription factor activity studies we discovered E2F4 as novel powerful regulator for LTBP4S expression.
Collapse
Affiliation(s)
- Insa Bultmann
- Center for Experimental Medicine, Medical Faculty, University of Cologne, Cologne, Germany
| | | | | | | |
Collapse
|