1
|
Esquer H, Zhou Q, LaBarbera DV. Targeted Inhibition of CHD1L by OTI-611 Reprograms Chemotherapy and Targeted Therapy-Induced Cell Cycle Arrest and Suppresses Proliferation to Produce Synergistic Antitumor Effects in Breast and Colorectal Cancer. Cells 2025; 14:318. [PMID: 40072047 PMCID: PMC11898988 DOI: 10.3390/cells14050318] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2025] [Revised: 02/08/2025] [Accepted: 02/11/2025] [Indexed: 03/15/2025] Open
Abstract
The second and third most frequently diagnosed cancers worldwide are breast (2.3 million new cases) and colorectal (1.9 million new cases), respectively. Although advances in cancer therapies and early detection have improved the overall survival of patients, patients still develop resistance or cancer recurrence. Thus, the development of novel therapies that can affect multiple mechanisms of drug resistance and cell survival is ideal for the treatment of advanced and metastatic cancers. CHD1L is a novel oncogenic protein involved in regulating chromatin remodeling, DNA damage repair, epithelial-mesenchymal transition (EMT), and programmed cell death via PARthanatos. Herein, we assess in real-time how the CHD1L inhibitor (CHD1Li) OTI-611 modulates cell cycle progression in Colo678, SUM149PT, and SW620 cell lines. By utilizing a cell cycle reporter, we tracked the real-time cell cycle progression of cancer cells treated with OTI-611 alone and in combination with standard-of-care (SOC) therapies. Our results indicate that OTI-611 causes G1 phase cell cycle arrest through a CHD1L-mediated mechanism that regulates Cyclin D1 expression and localization. As a result of this mechanism, OTI-611 can reprogram the cell cycle effects of other antitumor agents to modulate and arrest cells in G1 when used in combination, including agents commonly known to arrest cells in the G2/M phase. Therefore, we conclude that OTI-611-induced G1 arrest represents a critical component of its unique mechanism of action, contributing significantly to its anticancer activity.
Collapse
Affiliation(s)
- Hector Esquer
- Department of Pharmaceutical Sciences, The Skaggs School of Pharmacy and Pharmaceutical Sciences, The University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA; (H.E.); (Q.Z.)
- The Center for Drug Discovery, The University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
- The University of Colorado Cancer Center, Aurora, CO 80045, USA
| | - Qiong Zhou
- Department of Pharmaceutical Sciences, The Skaggs School of Pharmacy and Pharmaceutical Sciences, The University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA; (H.E.); (Q.Z.)
- The Center for Drug Discovery, The University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
- The University of Colorado Cancer Center, Aurora, CO 80045, USA
| | - Daniel V. LaBarbera
- Department of Pharmaceutical Sciences, The Skaggs School of Pharmacy and Pharmaceutical Sciences, The University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA; (H.E.); (Q.Z.)
- The Center for Drug Discovery, The University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
- The University of Colorado Cancer Center, Aurora, CO 80045, USA
| |
Collapse
|
2
|
Clune S, Awolade P, Zhou Q, Esquer H, Matter B, Kearns JT, Kellett T, Akintayo DC, Kompella UB, LaBarbera DV. The validation of new CHD1L inhibitors as a therapeutic strategy for cancer. Biomed Pharmacother 2024; 170:116037. [PMID: 38128184 PMCID: PMC10792906 DOI: 10.1016/j.biopha.2023.116037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Revised: 12/04/2023] [Accepted: 12/14/2023] [Indexed: 12/23/2023] Open
Abstract
Chromodomain helicase DNA-binding protein 1 like (CHD1L) is an oncogene that promotes tumor progression, metastasis, and multidrug resistance. CHD1L expression is indicative of poor outcomes and low survival in cancer patients with various cancer types. Herein, we report a set of CHD1L inhibitors (CHD1Li) discovered from high-throughput screening and evaluated using enzyme inhibition, 3D tumor organoid cytotoxicity and mechanistic assays. The structurally distinct compounds 8-11 emerged as hits with promising bioactivity by targeting CHD1L. CHD1Li were further examined for their stability in human and mouse liver microsomes, which showed compounds 9 and 11 to be the most metabolically stable. Additionally, molecular modeling studies of CHD1Li with the target protein shed light on key pharmacophore features driving CHD1L binding. Taken together, these results expand the chemical space of CHD1Li as a potential targeted therapy for colorectal cancer and other cancers.
Collapse
Affiliation(s)
- Sophia Clune
- The Skaggs School of Pharmacy and Pharmaceutical Sciences, Department of Pharmaceutical Sciences, The University of Colorado (CU) Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Paul Awolade
- The Skaggs School of Pharmacy and Pharmaceutical Sciences, Department of Pharmaceutical Sciences, The University of Colorado (CU) Anschutz Medical Campus, Aurora, CO 80045, USA; The CU Anschutz Center for Drug Discovery, USA
| | - Qiong Zhou
- The Skaggs School of Pharmacy and Pharmaceutical Sciences, Department of Pharmaceutical Sciences, The University of Colorado (CU) Anschutz Medical Campus, Aurora, CO 80045, USA; The CU Anschutz Center for Drug Discovery, USA; The CU Cancer Center, USA
| | - Hector Esquer
- The Skaggs School of Pharmacy and Pharmaceutical Sciences, Department of Pharmaceutical Sciences, The University of Colorado (CU) Anschutz Medical Campus, Aurora, CO 80045, USA; The CU Anschutz Center for Drug Discovery, USA; The CU Cancer Center, USA
| | - Brock Matter
- The Skaggs School of Pharmacy and Pharmaceutical Sciences, Department of Pharmaceutical Sciences, The University of Colorado (CU) Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Jeffrey T Kearns
- The Skaggs School of Pharmacy and Pharmaceutical Sciences, Department of Pharmaceutical Sciences, The University of Colorado (CU) Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Timothy Kellett
- The Skaggs School of Pharmacy and Pharmaceutical Sciences, Department of Pharmaceutical Sciences, The University of Colorado (CU) Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Damilola Caleb Akintayo
- The Skaggs School of Pharmacy and Pharmaceutical Sciences, Department of Pharmaceutical Sciences, The University of Colorado (CU) Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Uday B Kompella
- The Skaggs School of Pharmacy and Pharmaceutical Sciences, Department of Pharmaceutical Sciences, The University of Colorado (CU) Anschutz Medical Campus, Aurora, CO 80045, USA; The CU Anschutz Center for Drug Discovery, USA; The CU Cancer Center, USA
| | - Daniel V LaBarbera
- The Skaggs School of Pharmacy and Pharmaceutical Sciences, Department of Pharmaceutical Sciences, The University of Colorado (CU) Anschutz Medical Campus, Aurora, CO 80045, USA; The CU Anschutz Center for Drug Discovery, USA; The CU Cancer Center, USA.
| |
Collapse
|
3
|
Prigaro BJ, Esquer H, Zhou Q, Pike LA, Awolade P, Lai XH, Abraham AD, Abbott JM, Matter B, Kompella UB, Messersmith WA, Gustafson DL, LaBarbera DV. Design, Synthesis, and Biological Evaluation of the First Inhibitors of Oncogenic CHD1L. J Med Chem 2022; 65:3943-3961. [PMID: 35192363 DOI: 10.1021/acs.jmedchem.1c01778] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Chromodomain helicase DNA-binding protein 1 like (CHD1L) is an oncogene implicated in tumor progression, multidrug resistance, and metastasis in many types of cancer. In this article, we described the optimization of the first lead CHD1L inhibitors (CHD1Li) through drug design and medicinal chemistry. More than 30 CHD1Li were synthesized and evaluated using a variety of colorectal cancer (CRC) tumor organoid models and functional assays. The results led to the prioritization of six lead CHD1Li analogues with improved potency, antitumor activity, and drug-like properties including metabolic stability and in vivo pharmacokinetics. Furthermore, lead CHD1Li 6.11 proved to be an orally bioavailable antitumor agent, significantly reducing the tumor volume of CRC xenografts generated from isolated quasi mesenchymal cells (M-phenotype), which possess enhanced tumorigenic properties. In conclusion, we reported the optimization of first-in-class inhibitors of oncogenic CHD1L as a novel therapeutic strategy with potential for the treatment of cancer.
Collapse
Affiliation(s)
- Brett J Prigaro
- The Skaggs School of Pharmacy and Pharmaceutical Sciences, Department of Pharmaceutical Sciences, The CU Cancer Center, Aurora, Colorado 80045, United States
| | - Hector Esquer
- The Skaggs School of Pharmacy and Pharmaceutical Sciences, Department of Pharmaceutical Sciences, The CU Cancer Center, Aurora, Colorado 80045, United States
| | - Qiong Zhou
- The Skaggs School of Pharmacy and Pharmaceutical Sciences, Department of Pharmaceutical Sciences, The CU Cancer Center, Aurora, Colorado 80045, United States
| | - Laura A Pike
- The Skaggs School of Pharmacy and Pharmaceutical Sciences, Department of Pharmaceutical Sciences, The CU Cancer Center, Aurora, Colorado 80045, United States
| | - Paul Awolade
- The Skaggs School of Pharmacy and Pharmaceutical Sciences, Department of Pharmaceutical Sciences, The CU Cancer Center, Aurora, Colorado 80045, United States
| | - Xin-He Lai
- The Skaggs School of Pharmacy and Pharmaceutical Sciences, Department of Pharmaceutical Sciences, The CU Cancer Center, Aurora, Colorado 80045, United States
| | - Adedoyin D Abraham
- The Skaggs School of Pharmacy and Pharmaceutical Sciences, Department of Pharmaceutical Sciences, The CU Cancer Center, Aurora, Colorado 80045, United States
| | - Joshua M Abbott
- The Skaggs School of Pharmacy and Pharmaceutical Sciences, Department of Pharmaceutical Sciences, The CU Cancer Center, Aurora, Colorado 80045, United States
| | - Brock Matter
- The Skaggs School of Pharmacy and Pharmaceutical Sciences, Department of Pharmaceutical Sciences, The CU Cancer Center, Aurora, Colorado 80045, United States
| | - Uday B Kompella
- The Skaggs School of Pharmacy and Pharmaceutical Sciences, Department of Pharmaceutical Sciences, The CU Cancer Center, Aurora, Colorado 80045, United States.,The University of Colorado (CU) Anschutz Medical Campus (AMC) Center for Drug Discovery, The CU Cancer Center, Aurora, Colorado 80045, United States
| | - Wells A Messersmith
- The School of Medicine, Division of Medical Oncology, The CU Cancer Center, Aurora, Colorado 80045, United States.,The University of Colorado (CU) Anschutz Medical Campus (AMC) Center for Drug Discovery, The CU Cancer Center, Aurora, Colorado 80045, United States.,The CU Cancer Center, Aurora, Colorado 80045, United States
| | - Daniel L Gustafson
- Flint Animal Cancer Center and Department of Clinical Sciences, School of Biomedical Engineering, Colorado State University, Fort Collins, Colorado 80523, United States.,The University of Colorado (CU) Anschutz Medical Campus (AMC) Center for Drug Discovery, The CU Cancer Center, Aurora, Colorado 80045, United States.,The CU Cancer Center, Aurora, Colorado 80045, United States
| | - Daniel V LaBarbera
- The Skaggs School of Pharmacy and Pharmaceutical Sciences, Department of Pharmaceutical Sciences, The CU Cancer Center, Aurora, Colorado 80045, United States.,The University of Colorado (CU) Anschutz Medical Campus (AMC) Center for Drug Discovery, The CU Cancer Center, Aurora, Colorado 80045, United States.,The CU Cancer Center, Aurora, Colorado 80045, United States
| |
Collapse
|
4
|
Zhang X, Bai Y, Huang L, Liu S, Mo Y, Cheng W, Wang G, Cao Z, Chen X, Cui H, Qi L, Ma L, Liu M, Guan XY, Ma NF. CHD1L augments autophagy-mediated migration of hepatocellular carcinoma through targeting ZKSCAN3. Cell Death Dis 2021; 12:950. [PMID: 34654797 PMCID: PMC8520006 DOI: 10.1038/s41419-021-04254-x] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2021] [Revised: 09/25/2021] [Accepted: 10/04/2021] [Indexed: 01/18/2023]
Abstract
Autophagy is an important biological process in normal cells. However, how it affects tumor progression still remains poorly understood. Herein, we demonstrated that the oncogenic protein Chromodomain-helicase-DNA-binding-protein 1-like gene (CHD1L) might promote HCC cells migration and metastasis through autophagy. CHD1L could bind to the promotor region of Zinc finger with KRAB and SCAN domain 3 (ZKSCAN3), a pivotal autophagy suppressor, and inhibit its transcription. We established inducible CHD1L conditional knockout cell line (CHD1L-iKO cell) and found that the deletion of CHD1L significantly increased ZKSCAN3 expression both at mRNA and protein level. Deletion of CHD1L impaired the autophagic flux and migration of HCC cells, while specifically inhibiting ZKSCAN3 blocked these effects. Further exploration demonstrated that the enhanced tumor cell migration and metastasis induced by CHD1L was mediated through ZKSCAN3-induced autophagic degradation of Paxillin. In summary, we have characterized a previously unknown function of CHD1L in regulating tumor migration via ZKSCAN3-mediated autophagy in HCC. Further inhibition of CHD1L and its downstream autophagy signaling might shed new light on cancer therapeutics.
Collapse
MESH Headings
- Animals
- Autophagy
- Autophagy-Related Protein 5/metabolism
- Carcinoma, Hepatocellular/genetics
- Carcinoma, Hepatocellular/metabolism
- Carcinoma, Hepatocellular/pathology
- Carcinoma, Hepatocellular/ultrastructure
- Cell Line, Tumor
- Cell Movement
- DNA Helicases/metabolism
- DNA-Binding Proteins/metabolism
- Female
- Gene Expression Regulation, Neoplastic
- Gene Knockdown Techniques
- Humans
- Liver Neoplasms/genetics
- Liver Neoplasms/metabolism
- Liver Neoplasms/pathology
- Liver Neoplasms/ultrastructure
- Mice, Inbred BALB C
- Mice, Nude
- Microtubule-Associated Proteins/metabolism
- Neoplasm Metastasis
- Paxillin/metabolism
- RNA, Small Interfering/metabolism
- Transcription Factors/genetics
- Transcription Factors/metabolism
- Transcription, Genetic
- Mice
Collapse
Affiliation(s)
- Xiaofeng Zhang
- Affiliated Cancer Hospital and Institute of Guangzhou Medical University, Guangzhou, Guangdong, China
- Guangzhou Municipal and Guangdong ProvincialKey Laboratory of Protein Modification and Degradation, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Yinshan Bai
- Guangzhou Municipal and Guangdong ProvincialKey Laboratory of Protein Modification and Degradation, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, Guangdong, China
- School of Life Science and Engineering, Foshan University, Foshan, Guangdong, China
| | - Li Huang
- Guangzhou Municipal and Guangdong ProvincialKey Laboratory of Protein Modification and Degradation, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Shanshan Liu
- Guangzhou Municipal and Guangdong ProvincialKey Laboratory of Protein Modification and Degradation, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Yanxuan Mo
- Guangzhou Municipal and Guangdong ProvincialKey Laboratory of Protein Modification and Degradation, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Wei Cheng
- Guangzhou Municipal and Guangdong ProvincialKey Laboratory of Protein Modification and Degradation, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Guangliang Wang
- Guangzhou Municipal and Guangdong ProvincialKey Laboratory of Protein Modification and Degradation, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Zhiming Cao
- Affiliated Cancer Hospital and Institute of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Xiaogang Chen
- Guangzhou Municipal and Guangdong ProvincialKey Laboratory of Protein Modification and Degradation, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Huiqing Cui
- Guangzhou Municipal and Guangdong ProvincialKey Laboratory of Protein Modification and Degradation, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Ling Qi
- The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People's Hospital, Qingyuan, Guangdong, China
| | - Lei Ma
- Affiliated Cancer Hospital and Institute of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Ming Liu
- Guangzhou Municipal and Guangdong ProvincialKey Laboratory of Protein Modification and Degradation, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Xin-Yuan Guan
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Cancer Center, Sun Yat-sen University, Guangzhou, Guangdong, China
- Department of Clinical Oncology, Center for Cancer Research, and State Key Laboratory for Liver Research, University of Hong Kong, Pok Fu Lam, Hong Kong
| | - Ning-Fang Ma
- Affiliated Cancer Hospital and Institute of Guangzhou Medical University, Guangzhou, Guangdong, China.
- Guangzhou Municipal and Guangdong ProvincialKey Laboratory of Protein Modification and Degradation, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, Guangdong, China.
- Department of Histology and Embryology, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, Guangdong, China.
| |
Collapse
|
5
|
Zhang L, Jiang Y, Jiao P, Deng X, Xie Y. The high expression of CHD1L and its clinical significance in human solid tumors: A meta-analysis. Medicine (Baltimore) 2021; 100:e24851. [PMID: 33725840 PMCID: PMC7969280 DOI: 10.1097/md.0000000000024851] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/28/2019] [Accepted: 01/26/2021] [Indexed: 01/05/2023] Open
Abstract
BACKGROUND Chromodomain helicase DNA-binding protein 1-like (CHD1L) is an oncogene. It was cloned from 1q21 chromosome region of hepatocellular carcinoma in 1991. CHD1L is up-regulated in many kinds of cancers and is involved in the carcinogenesis and development of tumors. More and more studies have shown that over-expression of CHD1L is associated with poor prognosis of tumors. The purpose of this study was to evaluate the prognostic value of CHD1L in human solid tumors. METHODS The key words in the database of PubMed, Web of Science, Embase, Cochrane library, and TCGA were searched for systematic literature retrieval. We collected relevant articles and data about CHD1L and prognosis of cancer and screened them according to the eligible criteria to evaluate the prognostic value of CHD1L in cancer patients. Then Stata SE12.0 software is used to analyze the data. RESULTS In our meta-analysis, 2720 patients with a total of 15 articles involving multiple types of tumors showed that high expression levels of CHD1L were associated with shorter overall survival (OS) (hazard ratio = 2.21, 95% confidence interval [CI]: (1.49-3.30)] and (hazard ratio = 1.16, 95% CI: (1.01-1.32)] in the TCGA database, in addition, the pooled odds ratios (ORs) indicated high expression levels of CHD1L in tumors significantly are associated with TNM stage (OR = 1.61, 95% CI: 1.01-2.55, P < .05), tumor size (OR = 1.38, 95% CI: 1.07-1.78, P < .05), tumor differentiation (OR = 2.13, 95% CI: 1.43-3.16, P < .05), and distant metastasis (OR = 1.86, 95% CI: 1.45-2.39 P < .05). However, we did not observe a significant correlation between the high expression of CHD1L and age, gender. CONCLUSION The high expression of CHD1L is associated with poor OS as well as related to tumor differentiation, tumor size, and distant metastasis, which can be served as a prognostic marker and a potential predictor of clinical pathology in human solid tumors.
Collapse
Affiliation(s)
- Long Zhang
- Department of Hepatopancreatobiliary Surgery, Ganzhou People's Hospital of Jiangxi Province (Ganzhou Hospital Affiliated to Nanchang University), Ganzhou, Jiangxi
| | - Yufen Jiang
- Department of Gastroenterology, Kezhou People's Hospital, Atushi, Xinjiang
| | - Panpan Jiao
- Hospital Infection Management Office, Binzhou People's Hospital, Binzhou, Shandong, P.R. China
| | - Xiaohong Deng
- Department of Hepatopancreatobiliary Surgery, Ganzhou People's Hospital of Jiangxi Province (Ganzhou Hospital Affiliated to Nanchang University), Ganzhou, Jiangxi
| | - Yuancai Xie
- Department of Hepatopancreatobiliary Surgery, Ganzhou People's Hospital of Jiangxi Province (Ganzhou Hospital Affiliated to Nanchang University), Ganzhou, Jiangxi
| |
Collapse
|
6
|
Xiong X, Lai X, Li A, Liu Z, Ma N. Diversity roles of CHD1L in normal cell function and tumorigenesis. Biomark Res 2021; 9:16. [PMID: 33663617 PMCID: PMC7934534 DOI: 10.1186/s40364-021-00269-w] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Accepted: 02/16/2021] [Indexed: 12/14/2022] Open
Abstract
Chromodomain helicase/ATPase DNA binding protein 1-like gene (CHD1L) is a multifunctional protein participated in diverse cellular processes, including chromosome remodeling, cell differentiation and development. CHD1L is a regulator of chromosomal integrity maintenance, DNA repair and transcriptional regulation through its bindings to DNA. By regulating kinds of complex networks, CHD1L has been identified as a potent anti-apoptotic and pro-proliferative factor. CHD1L is also an oncoprotein since its overexpression leads to dysregulation of related downstream targets in various cancers. The latest advances in the functional molecular basis of CHD1L in normal cells will be described in this review. As the same time, we will describe the current understanding of CHD1L in terms of structure, characteristics, function and the molecular mechanisms underlying CHD1L in tumorigenesis. We inference that the role of CHD1L which involve in multiple cellular processes and oncogenesis is well worth further studying in basic biology and clinical relevance.
Collapse
Affiliation(s)
- Xifeng Xiong
- Guangzhou Institute of Traumatic Surgery, Guangzhou Red Cross Hospital, Jinan University, Guangzhou, 510220, China
| | - Xudong Lai
- Departement of infectious disease, Guangzhou Red Cross Hospital, Jinan University, Guangzhou, 510220, China
| | - Aiguo Li
- Guangzhou Institute of Traumatic Surgery, Guangzhou Red Cross Hospital, Jinan University, Guangzhou, 510220, China.
| | - Zhihe Liu
- Guangzhou Institute of Traumatic Surgery, Guangzhou Red Cross Hospital, Jinan University, Guangzhou, 510220, China.
| | - Ningfang Ma
- Affiliated Cancer Hospital and Institute of Guangzhou Medical University, Guangzhou, 510095, China. .,Department of Histology and Embryology, Guangzhou Medical University, Xinzao Town, Panyu District, Guangzhou, 511436, China.
| |
Collapse
|
7
|
Abbott JM, Zhou Q, Esquer H, Pike L, Broneske TP, Rinaldetti S, Abraham AD, Ramirez DA, Lunghofer PJ, Pitts TM, Regan DP, Tan AC, Gustafson DL, Messersmith WA, LaBarbera DV. First-in-Class Inhibitors of Oncogenic CHD1L with Preclinical Activity against Colorectal Cancer. Mol Cancer Ther 2020; 19:1598-1612. [PMID: 32499299 DOI: 10.1158/1535-7163.mct-20-0106] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2020] [Revised: 04/03/2020] [Accepted: 05/21/2020] [Indexed: 12/29/2022]
Abstract
Since the discovery of CHD1L in 2008, it has emerged as an oncogene implicated in the pathology and poor prognosis of a variety of cancers, including gastrointestinal cancers. However, a mechanistic understanding of CHD1L as a driver of colorectal cancer has been limited. Until now, there have been no reported inhibitors of CHD1L, also limiting its development as a molecular target. We sought to characterize the clinicopathologic link between CHD1L and colorectal cancer, determine the mechanism(s) by which CHD1L drives malignant colorectal cancer, and discover the first inhibitors with potential for novel treatments for colorectal cancer. The clinicopathologic characteristics associated with CHD1L expression were evaluated using microarray data from 585 patients with colorectal cancer. Further analysis of microarray data indicated that CHD1L may function through the Wnt/TCF pathway. Thus, we conducted knockdown and overexpression studies with CHD1L to determine its role in Wnt/TCF-driven epithelial-to-mesenchymal transition (EMT). We performed high-throughput screening (HTS) to identify the first CHD1L inhibitors. The mechanism of action, antitumor efficacy, and drug-like properties of lead CHD1L inhibitors were determined using biochemical assays, cell models, tumor organoids, patient-derived tumor organoids, and in vivo pharmacokinetics and pharmacodynamics. Lead CHD1L inhibitors display potent in vitro antitumor activity by reversing TCF-driven EMT. The best lead CHD1L inhibitor possesses drug-like properties in pharmacokinetic/pharmacodynamic mouse models. This work validates CHD1L as a druggable target and establishes a novel therapeutic strategy for the treatment of colorectal cancer.
Collapse
Affiliation(s)
- Joshua M Abbott
- The Skaggs School of Pharmacy and Pharmaceutical Sciences, Department of Pharmaceutical Sciences, The University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Qiong Zhou
- The Skaggs School of Pharmacy and Pharmaceutical Sciences, Department of Pharmaceutical Sciences, The University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Hector Esquer
- The Skaggs School of Pharmacy and Pharmaceutical Sciences, Department of Pharmaceutical Sciences, The University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Laura Pike
- The Skaggs School of Pharmacy and Pharmaceutical Sciences, Department of Pharmaceutical Sciences, The University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Travis P Broneske
- The Skaggs School of Pharmacy and Pharmaceutical Sciences, Department of Pharmaceutical Sciences, The University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Sébastien Rinaldetti
- The Skaggs School of Pharmacy and Pharmaceutical Sciences, Department of Pharmaceutical Sciences, The University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Adedoyin D Abraham
- The Skaggs School of Pharmacy and Pharmaceutical Sciences, Department of Pharmaceutical Sciences, The University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Dominique A Ramirez
- Flint Animal Cancer Center and Department of Clinical Sciences, School of Biomedical Engineering, Colorado State University, Fort Collins, Colorado
| | - Paul J Lunghofer
- Flint Animal Cancer Center and Department of Clinical Sciences, School of Biomedical Engineering, Colorado State University, Fort Collins, Colorado
| | - Todd M Pitts
- The School of Medicine, Division of Medical Oncology, The University of Colorado Anschutz Medical Campus, Aurora, Colorado.,The University of Colorado Cancer Center, The University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Daniel P Regan
- Flint Animal Cancer Center and Department of Clinical Sciences, School of Biomedical Engineering, Colorado State University, Fort Collins, Colorado
| | - Aik Choon Tan
- The School of Medicine, Division of Medical Oncology, The University of Colorado Anschutz Medical Campus, Aurora, Colorado.,The University of Colorado Cancer Center, The University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Daniel L Gustafson
- Flint Animal Cancer Center and Department of Clinical Sciences, School of Biomedical Engineering, Colorado State University, Fort Collins, Colorado.,The University of Colorado Cancer Center, The University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Wells A Messersmith
- The School of Medicine, Division of Medical Oncology, The University of Colorado Anschutz Medical Campus, Aurora, Colorado.,The University of Colorado Cancer Center, The University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Daniel V LaBarbera
- The Skaggs School of Pharmacy and Pharmaceutical Sciences, Department of Pharmaceutical Sciences, The University of Colorado Anschutz Medical Campus, Aurora, Colorado. .,The University of Colorado Cancer Center, The University of Colorado Anschutz Medical Campus, Aurora, Colorado
| |
Collapse
|
8
|
He WP, Guo YY, Yang GP, Lai HL, Sun TT, Zhang ZW, Ouyang LL, Zheng Y, Tian LM, Li XH, You ZS, Xie D, Yang GF. CHD1L promotes EOC cell invasiveness and metastasis via the regulation of METAP2. Int J Med Sci 2020; 17:2387-2395. [PMID: 32922205 PMCID: PMC7484650 DOI: 10.7150/ijms.48615] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/24/2020] [Accepted: 08/16/2020] [Indexed: 11/06/2022] Open
Abstract
Chromodomain helicase DNA binding protein 1-like (CHD1L) gene has been proposed to play an oncogenic role in human hepatocellular carcinoma. Previously we reported that CHD1L overexpression is significantly associated with the metastasis proceeding of epithelial ovarian cancer (EOC), and may predict a poor prognosis in EOC patients. However, the potential oncogenic mechanisms by which CHD1L acts in EOC remain unclear. To elucidate the oncogenic function of CHD1L, we carried out a series of in vitro assays, with effects of CHD1L ectogenic overexpression and silencing being determined in EOC cell lines (HO8910, A2780 and ES2). Real-time PCR and Western blotting analyses were used to identify potential downstream targets of CHD1L in the process of EOC invasion and metastasis. In ovarian carcinoma HO8910 cell lines, ectopic overexpression of CHD1L substantially induced the invasive and metastasis ability of the cancer cells in vitro. In contrast, knockdown of CHD1L using shRNA inhibited cell invasion in vitro in ovarian carcinoma A2780 and ES2 cell lines. We also demonstrated that methionyl aminopeptidase 2 (METAP2) was a downstream target of CHD1L in EOC, and we found a significant, positive correlation between the expression of CHD1L and METAP2 in EOC tissues (P<0.05). Our findings indicate that CHD1L plays a potential role in the inducement of EOC cancer cell invasion and/or metastasis via the regulation of METAP2 expression and suggests that CHD1L inhibition may provide a potential target for therapeutic intervention in human EOC.
Collapse
Affiliation(s)
- Wei-Peng He
- Department of Gynecology, the First Affiliated Hospital, Sun Yat-Sen University, No. 58, Zhongshan Road II, 510080 Guangzhou, China
| | - Yun-Yun Guo
- Department of Gynecology, the First Affiliated Hospital, Sun Yat-Sen University, No. 58, Zhongshan Road II, 510080 Guangzhou, China
| | - Gui-Ping Yang
- Department of Gynecology, the First Affiliated Hospital, Sun Yat-Sen University, No. 58, Zhongshan Road II, 510080 Guangzhou, China
| | - Hui-Ling Lai
- Department of Gynecology, the First Affiliated Hospital, Sun Yat-Sen University, No. 58, Zhongshan Road II, 510080 Guangzhou, China
| | - Ting-Ting Sun
- Department of Gynecology, the First Affiliated Hospital, Sun Yat-Sen University, No. 58, Zhongshan Road II, 510080 Guangzhou, China
| | - Zu-Wei Zhang
- Department of Gynecology, the First Affiliated Hospital, Sun Yat-Sen University, No. 58, Zhongshan Road II, 510080 Guangzhou, China
| | - Ling-Long Ouyang
- Department of Gynecology, the First Affiliated Hospital, Sun Yat-Sen University, No. 58, Zhongshan Road II, 510080 Guangzhou, China
| | - Yu Zheng
- Department of Gynecology, the First Affiliated Hospital, Sun Yat-Sen University, No. 58, Zhongshan Road II, 510080 Guangzhou, China
| | - Li-Ming Tian
- Department of Gynecology, the First Affiliated Hospital, Sun Yat-Sen University, No. 58, Zhongshan Road II, 510080 Guangzhou, China
| | - Xiao-Hui Li
- Department of Gynecology, the First Affiliated Hospital, Sun Yat-Sen University, No. 58, Zhongshan Road II, 510080 Guangzhou, China
| | - Ze-Shan You
- Department of Gynecology, the First Affiliated Hospital, Sun Yat-Sen University, No. 58, Zhongshan Road II, 510080 Guangzhou, China
| | - Dan Xie
- State Key Laboratory of Oncology in South China, Cancer Center, Sun Yat-Sen University, No. 651, Dongfeng Road East, 510060 Guangzhou, China
| | - Guo-Fen Yang
- Department of Gynecology, the First Affiliated Hospital, Sun Yat-Sen University, No. 58, Zhongshan Road II, 510080 Guangzhou, China
| |
Collapse
|
9
|
Li S, Chai Y, Ding Y, Yuan T, Wu C, Huang C. CHD1L is associated with poor survival and promotes the proliferation and metastasis of intrahepatic cholangiocarcinoma. Oncol Rep 2019; 42:657-669. [PMID: 31173252 PMCID: PMC6610041 DOI: 10.3892/or.2019.7174] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2018] [Accepted: 05/17/2019] [Indexed: 12/29/2022] Open
Abstract
Chromodomain helicase/ATPase DNA-binding protein 1-like gene (CHD1L) is a new oncogene which has been confirmed to be crucial to the progression of many solid tumors. In the present study, the expression of CHD1L was found to be upregulated in intrahepatic cholangiocarcinoma (ICC), which was significantly associated with histological differentiation (P=0.011), vascular invasion (P=0.002), lymph node metastasis (P=0.008) and TNM stage (P=0.001). Kaplan-Meier survival analysis revealed that ICC patients with positive CHD1L expression had shorter overall and disease-free survival than those with negative CHD1L expression. Functional study found that CHD1L exhibited strong oncogenic roles, including increased cell growth by CCK-8 assay, colony formation by plate colony formation assay, G1/S transition by flow cytometry and tumor formation in nude mice. In addition, RNAi-mediated silencing of CHD1L inhibited ICC invasion and metastasis by wound healing, Transwell migration and Matrigel invasion assays in vitro and in vivo. Collectively, our results show that CHD1L is upregulated and promotes the proliferation and metastasis of ICC cells. CHD1L acts as an oncogene and may be a prognostic factor or therapeutic target for patients with ICC.
Collapse
Affiliation(s)
- Shimiao Li
- Department of Hepatobiliary Surgery, Jiangxi Provincial People's Hospital, Nanchang, Jiangxi 330006, P.R. China
| | - Yi Chai
- Department of Neurosurgery, Shangrao People's Hospital, Shangrao, Jiangxi 334000, P.R. China
| | - Yanbao Ding
- Department of Hepatobiliary Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Tinghao Yuan
- Department of Hepatobiliary Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Changwen Wu
- Department of Urology Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Changwen Huang
- Department of Hepatobiliary Surgery, Jiangxi Provincial People's Hospital, Nanchang, Jiangxi 330006, P.R. China
| |
Collapse
|
10
|
Wang W, Wu J, Fei X, Chen W, Li Y, Shen K, Zhu L. CHD1L promotes cell cycle progression and cell motility by up-regulating MDM2 in breast cancer. Am J Transl Res 2019; 11:1581-1592. [PMID: 30972184 PMCID: PMC6456556] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2018] [Accepted: 01/14/2019] [Indexed: 06/09/2023]
Abstract
Chromodomain helicase/ATPase DNA-binding protein 1-like (CHD1L) gene is a novel oncogene amplified in many solid tumors. We investigated its role in breast cancer. CHD1L was over-expressed in 49.1% (57/116) breast cancer patients. Overexpression of CHD1L was significantly associated with younger age at diagnosis (P = 0.016), lymph node involvement (P = 0.040), higher tumor grade (P = 0.027), higher proliferation rate (P = 0.007) and shorter disease-free survival rate (77.2% vs. 91.5%, P = 0.037). A cDNA microarray analysis identified MDM2 as an important downstream target of CHD1L. And MDM2/p53 signaling pathway was showed to be significantly modulated by CHD1L. Further in vitro study showed that overexpression of CHD1L can promote tumorigenesis, metastasis, invasion and cell cycle progress. In vivo study confirmed the tumorigenesis ability of CHD1L. shRNA-mediated CHD1L silencing could abolishes the tumor-promotion effect of CHD1L in vitro and in vivo. In conclusion, CHD1L may promote the progress of breast cancer cells via the MDM2/p53 signaling pathway. This study identified CHD1L as a prognostic factor for breast cancer and MDM2 might be used as a potential target for therapeutic intervention in CHD1L overexpression breast cancer.
Collapse
Affiliation(s)
- Wei Wang
- Comprehensive Breast Health Center, Ruijin Hospital, School of Medicine, Shanghai Jiaotong University197 Ruijin Second Road, Shanghai 200025, P. R. China
| | - Jiayi Wu
- Comprehensive Breast Health Center, Ruijin Hospital, School of Medicine, Shanghai Jiaotong University197 Ruijin Second Road, Shanghai 200025, P. R. China
| | - Xiaochun Fei
- Pathology Department, Ruijin Hospital, School of Medicine, Shanghai Jiaotong University197 Ruijin Second Road, Shanghai 200025, P. R. China
| | - Weiguo Chen
- Comprehensive Breast Health Center, Ruijin Hospital, School of Medicine, Shanghai Jiaotong University197 Ruijin Second Road, Shanghai 200025, P. R. China
| | - Yafen Li
- Comprehensive Breast Health Center, Ruijin Hospital, School of Medicine, Shanghai Jiaotong University197 Ruijin Second Road, Shanghai 200025, P. R. China
| | - Kunwei Shen
- Comprehensive Breast Health Center, Ruijin Hospital, School of Medicine, Shanghai Jiaotong University197 Ruijin Second Road, Shanghai 200025, P. R. China
| | - Li Zhu
- Comprehensive Breast Health Center, Ruijin Hospital, School of Medicine, Shanghai Jiaotong University197 Ruijin Second Road, Shanghai 200025, P. R. China
| |
Collapse
|
11
|
CHD1L contributes to cisplatin resistance by upregulating the ABCB1-NF-κB axis in human non-small-cell lung cancer. Cell Death Dis 2019; 10:99. [PMID: 30718500 PMCID: PMC6362241 DOI: 10.1038/s41419-019-1371-1] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2018] [Revised: 11/26/2018] [Accepted: 01/09/2019] [Indexed: 12/13/2022]
Abstract
Chromodomain helicase/ATPase DNA binding protein 1-like gene (CHD1L) is a recently identified gene associated with malignant tumor progression and patient chemotherapy resistance in human hepatocellular carcinoma (HCC). Previously, we found an association between CHD1L overexpression and poor patient survival in non-small-cell lung cancer (NSCLC). However, little is known about the relationship between CHD1L expression and chemotherapy resistance of NSCLC. By employing immunohistochemistry, we analyzed the expression of CHD1L in NSCLC samples and elucidated the roles and mechanism of CHD1L in NSCLC chemoresistance. We found that the increased expression of CHD1L is positively correlated with a shorter survival time of patients who had received chemotherapy after surgery. We also found that the expression of CHD1L was increased after cisplatin treatment in A549 cells. Conversely, the depletion of CHD1L in cisplatin-resistance cells increased the cell sensitivity to cisplatin, indicating that CHD1L plays a critical role in cisplatin resistance of NSCLC cells. Importantly, we identified the ATP-Binding Cassette Sub-Family B Member (ABCB1) gene as a potential downstream target of CHD1L in NSCLC cells. Knocking down ABCB1 coupled with ectopic expression of CHD1L enhanced the effect of cisplatin on NSCLC cells apoptosis. In addition, overexpressed CHD1L increase the transcription of c-Jun which targeted directly to the promoter of ABCB1. Our data demonstrate that CHD1L could induce cisplatin resistance in NSCLC via c-Jun-ABCB1-NF-κB axis, and may serve as a novel predictive marker and the potential therapeutic target for cisplatin resistance in NSCLC.
Collapse
|
12
|
Liu W, Xu J, Zhang C. Prognostic role of chromodomain helicase DNA binding protein 1-like protein in human solid cancers: A meta-analysis. Medicine (Baltimore) 2018; 97:e11522. [PMID: 30024537 PMCID: PMC6086487 DOI: 10.1097/md.0000000000011522] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/05/2022] Open
Abstract
BACKGROUND Chromodomain helicase DNA binding protein 1-like (CHD1L) played vital roles in tumorigenesis and development. Its aberrant expression was reported to be related to progression and prognosis in various tumors. However, no consensus on the prognostic value of CHD1L protein has been made. This meta-analysis was aimed to assess the clinical significance of CHD1L protein in human solid tumors. METHODS Web of Science, PubMed, Embase, China National Knowledge Infrastructure (CNKI), and Wanfang databases were extensively searched to retrieve publications that reported the association between CHD1L expression and cancer prognosis. Hazard ratios (HRs) or odds ratios (ORs) with their 95% confidence intervals (95% CIs) were applied to assess the strength of the associations through Stata statistical software version 12.0 or Revman software 5.3, respectively. RESULT A total of 14 studies were screened according to the inclusion criteria. The pooled results revealed patients with higher CHD1L expression manifested with decreased overall survival (OS) (HR: 1.59, 95% CI: 1.29-1.89, P < .001) and poorer disease-free survival (DFS) (HR: 1.66, 95% CI: 1.17-2.15, P < .001). The prognostic value of CHD1L protein for OS was further confirmed by performing subgroup meta-analysis. Furthermore, the pooled results revealed a positive correlation of CHD1L protein expression with tumor depth (OR: 1.87, 95% CI: 1.48-2.37), lymph node metastasis (OR: 1.46, 95% CI: 1.01-2.11), and distant metastasis (OR: 1.86, 95% CI: 1.45-2.38). CONCLUSION CHD1L overexpression was associated with poor prognosis and advanced clinicopathological features, CHD1L may be a valuable biomarker for prognostication of cancer patients.
Collapse
|
13
|
Hua J, Li S, Huang C. Clinical significance of chromodomain helicase/ATPase DNA binding protein 1-like and human mutL homolog 1 gene expression in cholangiocarcinoma. Oncol Lett 2018; 16:2989-2994. [PMID: 30127888 PMCID: PMC6096073 DOI: 10.3892/ol.2018.9043] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2015] [Accepted: 06/06/2018] [Indexed: 12/18/2022] Open
Abstract
Cholangiocarcinoma is a highly malignant form of gastrointestinal cancer with an unfavorable prognosis. The novel oncogene chromodomain helicase/ATPase DNA binding protein 1-like (CHD1L) has been confirmed to serve a vital role in numerous types of cancer, including liver cancer. Mismatch repair (MMR) is a common DNA repair process that contributes to the preservation of the integrity and stability of genetic substances. Human mutL homolog 1 gene (hMLH1) is an important MMR protein family member. The present study aimed to evaluate the pathological and clinical features of cholangiocarcinoma, and to investigate the clinical significance of CHD1L and hMLH1 expression in cholangiocarcinoma. A total of 108 samples from cholangiocarcinoma tumor tissues and 60 samples from normal bile duct tissue were obtained from patients admitted to The Second Affiliated Hospital of Nanchang University between May 2005 and May 2014. All cholangiocarcinoma cases were pathologically confirmed. The expression of CHD1L and hMLH1 was examined by immunohistochemistry analysis. The expression of CHD1L in cholangiocarcinoma (94.44%) was significantly higher than in normal bile duct tissues (40.00%). CHD1L expression was associated with gallstone history, serum carbohydrate antigen 19-9 (CA19-9) level and Tumor-Node-Metastasis (TNM) stage (P<0.05). hMLH1 expression in cholangiocarcinoma (77.78%) was significantly lower than in normal bile duct tissues (96.67%), and was associated with gender, age, serum CA19-9 level, the presence of hepatitis B virus surface antigen, TNM stage and tumor diameter (P<0.05). Kaplan-Meier survival curve analysis indicated that the 3-year accumulative survival rates for CHD1L-positive and -negative patients differed significantly (P<0.05; 17.90 and 83.33%, respectively). There was no statistically significant difference (P>0.05) between the 3-year accumulate survival rates for hMLH1-positive and -negative patients (38.90 and 33.30%, respectively). High CHD1L expression and low hMLH1 expression levels were observed in patients with cholangiocarcinoma, and their abnormal expression patterns were associated with the progression of malignancy and an unfavorable disease prognosis. Therefore, CHD1L and hMLH1 may be potential prognostic biomarkers for cholangiocarcinoma.
Collapse
Affiliation(s)
- Jingwen Hua
- Department of Oncological Surgery, Xinyi People's Hospital, Xuzhou, Jiangsu 221400, P.R. China
| | - Shiniao Li
- Department of Hepatobiliary Surgery, Jiangxi Provincial People's Hospital, Nanchang, Jiangxi 330006, P.R. China
| | - Changwen Huang
- Department of Hepatobiliary Surgery, Jiangxi Provincial People's Hospital, Nanchang, Jiangxi 330006, P.R. China
| |
Collapse
|
14
|
Liu ZH, Zhang Q, Ding YJ, Ren YH, Yang HP, Xi Q, Cheng YN, Miao GL, Liu HK, Li CX, Yan WQ, Li Y, Xue Z, Zhang L, Li XY, Zhao CL, Da Y, Wu XZ, Chen JQ, Zhang R, Li ZG. Overexpression of CHD1L is associated with poor survival and aggressive tumor biology in esophageal carcinoma. Oncotarget 2017; 8:74178-74187. [PMID: 29088777 PMCID: PMC5650332 DOI: 10.18632/oncotarget.18830] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2017] [Accepted: 06/04/2017] [Indexed: 12/26/2022] Open
Abstract
Esophageal carcinoma (EC) is a malignancy with high metastatic potential. Chromosomal helicase/ATPase DNA binding protein 1-like (CHD1L) gene is a newly identified oncogene located at Chr1q21, and it is amplified in many solid tumors. However, the status of CHD1L protein expression in EC and its clinical significance is uncertain. This study was designed to investigate the significance of CHD1L expression in human EC and its biological function in EC cells. The expression of CHD1L was examined by immunohistochemistry in 191 surgically resected ECs. The associations between CHD1L expression and clinical pathological parameters and the prognostic value of CHD1L were analyzed. Western blot analysis showed that CHD1L was overexpressed in EC cell lines. In addition, positive CHD1L expression was strongly related to advanced clinical stage (P<0.01), and lymph node metastasis (P<0.01) of EC. The Kaplan-Meier curve indicated that high expression of CHD1L may result in poor prognosis of EC patients (P<0.01), and multivariate analysis showed that CHD1L overexpression was an independent predictor of overall survival. Furthermore, suppression of CHD1L in EC cells increased apoptosis and decreased cell proliferation invasion ability. Our results suggest that CHD1L is a target oncogene with the potential to serve as a novel prognostic biomarker in EC pathogenesis.
Collapse
Affiliation(s)
- Ze-Han Liu
- Nankai Clinical College, Tianjin Medical University, Tianjin, P.R. China
| | - Qi Zhang
- Institute of Integrative Medicine Therapy for Acute Abdominal Diseases of Tianjin, Nankai Hospital, Tianjin, P. R. China
- Laboratory of Immunology and Inflammation, Department of Immunology, Key Laboratory of Immune Microenvironment and Diseases of Educational Ministry of China, Basic Medical College, Tianjin Medical University, Tianjin, P.R. China
| | - Yi-Jie Ding
- First Central Clinical College, Tianjin Medical University, Tianjin, P.R. China
| | - Ying-Hui Ren
- Laboratory of Immunology and Inflammation, Department of Immunology, Key Laboratory of Immune Microenvironment and Diseases of Educational Ministry of China, Basic Medical College, Tianjin Medical University, Tianjin, P.R. China
| | - Hui-Peng Yang
- Laboratory of Immunology and Inflammation, Department of Immunology, Key Laboratory of Immune Microenvironment and Diseases of Educational Ministry of China, Basic Medical College, Tianjin Medical University, Tianjin, P.R. China
| | - Qing Xi
- Laboratory of Immunology and Inflammation, Department of Immunology, Key Laboratory of Immune Microenvironment and Diseases of Educational Ministry of China, Basic Medical College, Tianjin Medical University, Tianjin, P.R. China
| | - Ying-Nan Cheng
- Laboratory of Immunology and Inflammation, Department of Immunology, Key Laboratory of Immune Microenvironment and Diseases of Educational Ministry of China, Basic Medical College, Tianjin Medical University, Tianjin, P.R. China
| | - Guo-Lin Miao
- Laboratory of Immunology and Inflammation, Department of Immunology, Key Laboratory of Immune Microenvironment and Diseases of Educational Ministry of China, Basic Medical College, Tianjin Medical University, Tianjin, P.R. China
| | - Hong-Kun Liu
- Laboratory of Immunology and Inflammation, Department of Immunology, Key Laboratory of Immune Microenvironment and Diseases of Educational Ministry of China, Basic Medical College, Tianjin Medical University, Tianjin, P.R. China
| | - Cai-Xia Li
- Institute of Integrative Medicine Therapy for Acute Abdominal Diseases of Tianjin, Nankai Hospital, Tianjin, P. R. China
| | - Wen-Qiang Yan
- Department of Thoracic Surgery, Nankai Hospital, Nankai District, Tianjin, P. R. China
| | - Yan Li
- Laboratory of Immunology and Inflammation, Department of Immunology, Key Laboratory of Immune Microenvironment and Diseases of Educational Ministry of China, Basic Medical College, Tianjin Medical University, Tianjin, P.R. China
| | - Zhenyi Xue
- Laboratory of Immunology and Inflammation, Department of Immunology, Key Laboratory of Immune Microenvironment and Diseases of Educational Ministry of China, Basic Medical College, Tianjin Medical University, Tianjin, P.R. China
| | - Lijuan Zhang
- Laboratory of Immunology and Inflammation, Department of Immunology, Key Laboratory of Immune Microenvironment and Diseases of Educational Ministry of China, Basic Medical College, Tianjin Medical University, Tianjin, P.R. China
| | - Xin-Ye Li
- General Hospital, Tianjin Medical University, Tianjin, P.R. China
| | - Chen-Long Zhao
- General Hospital, Tianjin Medical University, Tianjin, P.R. China
| | - Yurong Da
- Laboratory of Immunology and Inflammation, Department of Immunology, Key Laboratory of Immune Microenvironment and Diseases of Educational Ministry of China, Basic Medical College, Tianjin Medical University, Tianjin, P.R. China
| | - Xian-Zhong Wu
- Institute of Integrative Medicine Therapy for Acute Abdominal Diseases of Tianjin, Nankai Hospital, Tianjin, P. R. China
| | - Jun-Qiang Chen
- Department of Radiotherapy, Fujian Provincial Tumor Hospital, Affiliated Tumor Hospital of Fujian Medical University, Fuzhou, P.R. China
| | - Rongxin Zhang
- Laboratory of Immunology and Inflammation, Department of Immunology, Key Laboratory of Immune Microenvironment and Diseases of Educational Ministry of China, Basic Medical College, Tianjin Medical University, Tianjin, P.R. China
- Laboratory of Immunology and Inflammation, Guangdong Pharmaceutical University, Guangzhou, P.R. China
| | - Zhi-Gang Li
- Institute of Integrative Medicine Therapy for Acute Abdominal Diseases of Tianjin, Nankai Hospital, Tianjin, P. R. China
- General Hospital, Tianjin Medical University, Tianjin, P.R. China
- Hainan Cancer Hospital, Affiliated Cancer Hospital of Hainan Medical College, Haikou City, P.R. China
| |
Collapse
|
15
|
He LR, Ma NF, Chen JW, Li BK, Guan XY, Liu MZ, Xie D. Overexpression of CHD1L is positively associated with metastasis of lung adenocarcinoma and predicts patients poor survival. Oncotarget 2016; 6:31181-90. [PMID: 26360781 PMCID: PMC4741596 DOI: 10.18632/oncotarget.5070] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2015] [Accepted: 08/14/2015] [Indexed: 12/13/2022] Open
Abstract
CHD1L (chromodomain helicase/ATPase DNA binding protein 1-like gene) has been demonstrated as an oncogene in hepatocellular carcinoma (HCC), however, the role of CHD1L in non-small-cell lung cancer (NSCLC) tumorigenesis hasn't been elucidated. In this study, the expression and amplification status of CHD1L were examined by immunohistochemistry and fluorescence in situ hybridization respectively in 248 surgically resected NSCLCs. The associations between CHD1L expression and clinicopathologic features and the prognostic value of CHD1L were analyzed. Overexpression and amplification of CHD1L was found in 42.1% and 17.7% of NSCLCs, respectively. The frequency of CHD1L overexpression (53.2% vs. 28.1%, P = 0.002) and amplification (25.2% vs. 8.2%, P = 0.020) in adenocarcinoma (ADC), was much higher than that in squamous cell carcinoma (SCC). CHD1L overexpression was associated closely with ascending pN status (P < 0.001), advanced clinical stage (P = 0.001) and tumor distant metastasis (P = 0.001) in ADCs, but not in SCCs. For the whole cohort and ADC patients, univariate survival analysis demonstrated a significant association of CHD1L overexpression with shortened survival; and in multivariate analysis, CHD1L overexpression was evaluated as a independent predictor for overall survival and distant metastasis free survival. These results suggested that overexpression of CHD1L is positively associated with tumor metastasis of lung ADC, and might serve as a novel prognostic biomarker and potential therapeutic target for lung ADC patients.
Collapse
Affiliation(s)
- Li-Ru He
- The State Key Laboratory of Oncology in South China, Sun Yat-Sen University Cancer Center, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China.,Department of Radiation Oncology, Sun Yat-Sen University Cancer Center, Guangzhou, China
| | - Ning-Fang Ma
- Department of Histology and Embryology, State Key Laboratory of Respiratory Disease, Guangzhou Medical University, Guangzhou, China
| | - Jie-Wei Chen
- The State Key Laboratory of Oncology in South China, Sun Yat-Sen University Cancer Center, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China.,Department of Pathology, Sun Yat-Sen University Cancer Center, Guangzhou, China
| | - Bin-Kui Li
- The State Key Laboratory of Oncology in South China, Sun Yat-Sen University Cancer Center, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Xin-Yuan Guan
- The State Key Laboratory of Oncology in South China, Sun Yat-Sen University Cancer Center, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China.,Departments of Clinical Oncology, The University of Hong Kong, Hong Kong, China
| | - Meng-Zhong Liu
- The State Key Laboratory of Oncology in South China, Sun Yat-Sen University Cancer Center, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China.,Department of Radiation Oncology, Sun Yat-Sen University Cancer Center, Guangzhou, China
| | - Dan Xie
- The State Key Laboratory of Oncology in South China, Sun Yat-Sen University Cancer Center, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China.,Department of Pathology, Sun Yat-Sen University Cancer Center, Guangzhou, China
| |
Collapse
|