1
|
Zhu C, Ke S, Li Y, Zhang W, Che Y, Zhang R, Huang P, Xu T. Targeting DEP domain containing 1 in anaplastic thyroid carcinoma: Implications for stemness regulation and malignant phenotype suppression. Heliyon 2024; 10:e27150. [PMID: 38449652 PMCID: PMC10915564 DOI: 10.1016/j.heliyon.2024.e27150] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Revised: 02/11/2024] [Accepted: 02/25/2024] [Indexed: 03/08/2024] Open
Abstract
Background Anaplastic thyroid carcinoma (ATC), a rare but highly aggressive endocrine malignancy, is characterized by a significant presence of cancer stem-like cells (CSCs). These CSCs, known for their self-renewal and differentiation capacities, contribute to various aggressive tumor properties, including recurrence, metastasis, heterogeneity, multidrug resistance, and radiation resistance. Despite their critical role, the regulatory mechanisms of CSCs in ATC remain poorly elucidated, posing challenges in effectively targeting these cells for treatment. Methods To delve into this, we employed the single sample gene set enrichment analysis (ssGSEA) algorithm to evaluate the stemness of samples in combined datasets. Samples were then classified into high and low stemness subgroups based on their average stemness scores. Differential gene expression between these subgroups was analyzed. We further explored the association of candidate genes with patient prognosis. Additionally, we conducted gene set enrichment analysis (GSEA) and a series of cell biology experiments to validate the role of DEP domain-containing protein 1 (DEPDC1) in fostering CSC-like traits and regulating the malignant phenotypes of ATC. Results Our investigation demonstrated that DEPDC1 was significantly upregulated in CSCs and is abundantly expressed in ATC tissues. In vitro assays revealed that knockdown of DEPDC1 markedly inhibited tumor sphere formation and attenuated the proliferation, invasion, and migration of ATC cells. This silencing also resulted in reduced expression of stemness markers associated with CSCs. Furthermore, our GSEA findings linked high DEPDC1 expression to cell cycle progression and the maintenance of tumor cell stemness, with DEPDC1 knockdown disrupting these signaling pathways. Collectively, our results position DEPDC1 as a pivotal regulator of CSC-like characteristics in ATC, where aberrant DEPDC1 expression amplifies stemness properties and fuels the cancer's aggressive behavior. Consequently, DEPDC1 emerges as a promising therapeutic target for ATC management. In summary, this study underscores the pivotal role of DEPDC1 in modulating CSC-like features in ATC, offering new avenues for targeted therapy in this challenging malignancy.
Collapse
Affiliation(s)
- Chaozhuang Zhu
- Institute of Pharmacology, Department of Pharmaceutical Sciences, Zhejiang University of Technology, Hangzhou, China
| | - Shuwei Ke
- Institute of Pharmacology, Department of Pharmaceutical Sciences, Zhejiang University of Technology, Hangzhou, China
| | - Ying Li
- Institute of Pharmacology, Department of Pharmaceutical Sciences, Zhejiang University of Technology, Hangzhou, China
| | - Wanli Zhang
- Institute of Pharmacology, Department of Pharmaceutical Sciences, Zhejiang University of Technology, Hangzhou, China
| | - Yulu Che
- Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Ruidan Zhang
- Institute of Pharmacology, Department of Pharmaceutical Sciences, Zhejiang University of Technology, Hangzhou, China
| | - Ping Huang
- Center for Clinical Pharmacy, Cancer Center, Department of Pharmacy, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, China
- Key Laboratory of Endocrine Gland Diseases of Zhejiang Province, China
| | - Tong Xu
- Center for Clinical Pharmacy, Cancer Center, Department of Pharmacy, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, China
| |
Collapse
|
2
|
Muzyka L, Goff NK, Choudhary N, Koltz MT. Systematic Review of Molecular Targeted Therapies for Adult-Type Diffuse Glioma: An Analysis of Clinical and Laboratory Studies. Int J Mol Sci 2023; 24:10456. [PMID: 37445633 DOI: 10.3390/ijms241310456] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Revised: 06/05/2023] [Accepted: 06/16/2023] [Indexed: 07/15/2023] Open
Abstract
Gliomas are the most common brain tumor in adults, and molecularly targeted therapies to treat gliomas are becoming a frequent topic of investigation. The current state of molecular targeted therapy research for adult-type diffuse gliomas has yet to be characterized, particularly following the 2021 WHO guideline changes for classifying gliomas using molecular subtypes. This systematic review sought to characterize the current state of molecular target therapy research for adult-type diffuse glioma to better inform scientific progress and guide next steps in this field of study. A systematic review was conducted in accordance with PRISMA guidelines. Studies meeting inclusion criteria were queried for study design, subject (patients, human cell lines, mice, etc.), type of tumor studied, molecular target, respective molecular pathway, and details pertaining to the molecular targeted therapy-namely the modality, dose, and duration of treatment. A total of 350 studies met the inclusion criteria. A total of 52 of these were clinical studies, 190 were laboratory studies investigating existing molecular therapies, and 108 were laboratory studies investigating new molecular targets. Further, a total of 119 ongoing clinical trials are also underway, per a detailed query on clinicaltrials.gov. GBM was the predominant tumor studied in both ongoing and published clinical studies as well as in laboratory analyses. A few studies mentioned IDH-mutant astrocytomas or oligodendrogliomas. The most common molecular targets in published clinical studies and clinical trials were protein kinase pathways, followed by microenvironmental targets, immunotherapy, and cell cycle/apoptosis pathways. The most common molecular targets in laboratory studies were also protein kinase pathways; however, cell cycle/apoptosis pathways were the next most frequent target, followed by microenvironmental targets, then immunotherapy pathways, with the wnt/β-catenin pathway arising in the cohort of novel targets. In this systematic review, we examined the current evidence on molecular targeted therapy for adult-type diffuse glioma and discussed its implications for clinical practice and future research. Ultimately, published research falls broadly into three categories-clinical studies, laboratory testing of existing therapies, and laboratory identification of novel targets-and heavily centers on GBM rather than IDH-mutant astrocytoma or oligodendroglioma. Ongoing clinical trials are numerous in this area of research as well and follow a similar pattern in tumor type and targeted pathways as published clinical studies. The most common molecular targets in all study types were protein kinase pathways. Microenvironmental targets were more numerous in clinical studies, whereas cell cycle/apoptosis were more numerous in laboratory studies. Immunotherapy pathways are on the rise in all study types, and the wnt/β-catenin pathway is increasingly identified as a novel target.
Collapse
Affiliation(s)
- Logan Muzyka
- Department of Neurosurgery, Dell Medical School, The University of Texas at Austin, 1501 Red River Street, Austin, TX 78712, USA
| | - Nicolas K Goff
- Department of Neurosurgery, Dell Medical School, The University of Texas at Austin, 1501 Red River Street, Austin, TX 78712, USA
| | - Nikita Choudhary
- Department of Neurosurgery, Dell Medical School, The University of Texas at Austin, 1501 Red River Street, Austin, TX 78712, USA
| | - Michael T Koltz
- Department of Neurosurgery, Dell Medical School, The University of Texas at Austin, 1501 Red River Street, Austin, TX 78712, USA
| |
Collapse
|
3
|
Shen L, Li H, Liu R, Zhou C, Bretches M, Gong X, Lu L, Zhang Y, Zhao K, Ning B, Yang SY, Zhang A. DEPDC1 as a crucial factor in the progression of human osteosarcoma. Cancer Med 2023; 12:5798-5808. [PMID: 36479633 PMCID: PMC10028160 DOI: 10.1002/cam4.5340] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Revised: 06/22/2022] [Accepted: 07/03/2022] [Indexed: 12/12/2022] Open
Abstract
OBJECTIVE Novel therapeutic strategies are emerging with the increased understanding of the underlying mechanisms of human osteosarcoma. This current study tends to decipher the potentially critical role of DEP domain-containing 1 (DEPDC1), a tumor-related gene, during the progression of osteosarcoma. METHODS Bioinformatics analysis of 25,035 genes from the National Center for Biotechnology Information (NCBI) databases was performed to screen differentially expressed genes between osteosarcoma and normal control groups, complemented by the examination of 85 clinical osteosarcoma specimens. Furthermore, the manipulation of DEPDC1 expression levels by using silencing RNA (siRNA) or lentiviral vector intervention on human osteosarcoma cells was performed to reveal its role and interactions in in vitro and in vivo settings. RESULTS Gene expression profile analysis and immunohistochemical (IHC) examination suggested that DEPDC1 is highly expressed in human osteosarcoma cells and tumor tissue. The silencing of DEPDC1 arrested osteosarcoma cell proliferation, promoted apoptosis, and ceased tumor metastasis. Studies involving clinical human osteosarcoma cases exhibited a strong correlation of DEPDC1 over-expressed osteosarcoma specimens with a reduced patient survival rate. CONCLUSIONS Collectively, this study demonstrated that DEPDC1 is a critical driver in the promotion of osteosarcoma progression and results in poor patient prognosis. Genetically targeting or pharmacologically inhibiting DEPDC1 may serve as a promising strategy for treating human osteosarcoma.
Collapse
Affiliation(s)
- Lin Shen
- Department of Orthopaedics, Central Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Han Li
- Department of Endocrinology, The First Affiliated Hospital of Xiamen University, Xiamen, Fujian, China
| | - Ronghan Liu
- Department of Orthopaedics, Central Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Chendan Zhou
- Department of Pediatrics, Qilu Hospital, Shandong University, Jinan, China
| | - Morgan Bretches
- Department of Orthopaedic Surgery, University of Kansas School of Medicine-Wichita, Wichita, Kansas, USA
- Department of Biological Sciences, Wichita State University, Wichita, Kansas, USA
| | - Xuan Gong
- Department of Biological Sciences, Wichita State University, Wichita, Kansas, USA
- University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Laitong Lu
- Department of Orthopaedics, Central Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Ying Zhang
- Department of Orthopaedics, Central Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Kai Zhao
- Department of Orthopaedics, Central Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Bin Ning
- Department of Orthopaedics, Central Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Shang-You Yang
- Department of Orthopaedic Surgery, University of Kansas School of Medicine-Wichita, Wichita, Kansas, USA
- Department of Biological Sciences, Wichita State University, Wichita, Kansas, USA
| | - Aijun Zhang
- Department of Pediatrics, Qilu Hospital, Shandong University, Jinan, China
| |
Collapse
|
4
|
Wang Y, Wu J, Luo W, Zhang H, Shi G, Shen Y, Zhu Y, Ma C, Dai B, Ye D, Zhu Y. ALPK2 acts as tumor promotor in development of bladder cancer through targeting DEPDC1A. Cell Death Dis 2021; 12:661. [PMID: 34210956 PMCID: PMC8249393 DOI: 10.1038/s41419-021-03947-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2019] [Revised: 06/08/2021] [Accepted: 06/16/2021] [Indexed: 12/30/2022]
Abstract
Bladder cancer is one of the most common malignant tumors in the urinary system. The development and improvement of treatment efficiency require the deepening of the understanding of its molecular mechanism. This study investigated the role of ALPK2, which is rarely studied in malignant tumors, in the development of bladder cancer. Our results showed the upregulation of ALPK2 in bladder cancer, and data mining of TCGA database showed the association between ALPK2 and pathological parameters of patients with bladder cancer. In vitro and in vivo experiments demonstrated that knockdown of ALPK2 could inhibit bladder cancer development through regulating cell proliferation, cell apoptosis, and cell migration. Additionally, DEPDC1A is identified as a potential downstream of ALPK2 with direct interaction, whose overexpression/downregulation can inhibit/promote the malignant behavioral of bladder cancer cells. Moreover, the overexpression of DEPDC1A can rescue the inhibitory effects of ALPK2 knockdown on bladder cancer. In conclusion, ALPK2 exerts a cancer-promoting role in the development of bladder cancer by regulating DEPDC1A, which may become a promising target to improve the treatment strategy of bladder cancer.
Collapse
Affiliation(s)
- Yuchen Wang
- Department of Urology, Fundan University Shanghai Cancer Center, No. 270 Dongan Road, 200032, Shanghai, People's Republic of China
- Department of Oncology, Shanghai Medical College, Fudan University, No. 270 Dongan Road, 200032, Shanghai, People's Republic of China
| | - Jie Wu
- Department of Urology, Fundan University Shanghai Cancer Center, No. 270 Dongan Road, 200032, Shanghai, People's Republic of China
- Department of Oncology, Shanghai Medical College, Fudan University, No. 270 Dongan Road, 200032, Shanghai, People's Republic of China
| | - Wenjie Luo
- Department of Urology, Fundan University Shanghai Cancer Center, No. 270 Dongan Road, 200032, Shanghai, People's Republic of China
- Department of Oncology, Shanghai Medical College, Fudan University, No. 270 Dongan Road, 200032, Shanghai, People's Republic of China
| | - Hailiang Zhang
- Department of Urology, Fundan University Shanghai Cancer Center, No. 270 Dongan Road, 200032, Shanghai, People's Republic of China
- Department of Oncology, Shanghai Medical College, Fudan University, No. 270 Dongan Road, 200032, Shanghai, People's Republic of China
| | - Guohai Shi
- Department of Urology, Fundan University Shanghai Cancer Center, No. 270 Dongan Road, 200032, Shanghai, People's Republic of China
- Department of Oncology, Shanghai Medical College, Fudan University, No. 270 Dongan Road, 200032, Shanghai, People's Republic of China
| | - Yijun Shen
- Department of Urology, Fundan University Shanghai Cancer Center, No. 270 Dongan Road, 200032, Shanghai, People's Republic of China
- Department of Oncology, Shanghai Medical College, Fudan University, No. 270 Dongan Road, 200032, Shanghai, People's Republic of China
| | - Yao Zhu
- Department of Urology, Fundan University Shanghai Cancer Center, No. 270 Dongan Road, 200032, Shanghai, People's Republic of China
- Department of Oncology, Shanghai Medical College, Fudan University, No. 270 Dongan Road, 200032, Shanghai, People's Republic of China
| | - Chunguang Ma
- Department of Urology, Fundan University Shanghai Cancer Center, No. 270 Dongan Road, 200032, Shanghai, People's Republic of China
- Department of Oncology, Shanghai Medical College, Fudan University, No. 270 Dongan Road, 200032, Shanghai, People's Republic of China
| | - Bo Dai
- Department of Urology, Fundan University Shanghai Cancer Center, No. 270 Dongan Road, 200032, Shanghai, People's Republic of China
- Department of Oncology, Shanghai Medical College, Fudan University, No. 270 Dongan Road, 200032, Shanghai, People's Republic of China
| | - Dingwei Ye
- Department of Urology, Fundan University Shanghai Cancer Center, No. 270 Dongan Road, 200032, Shanghai, People's Republic of China.
- Department of Oncology, Shanghai Medical College, Fudan University, No. 270 Dongan Road, 200032, Shanghai, People's Republic of China.
| | - Yiping Zhu
- Department of Urology, Fundan University Shanghai Cancer Center, No. 270 Dongan Road, 200032, Shanghai, People's Republic of China.
- Department of Oncology, Shanghai Medical College, Fudan University, No. 270 Dongan Road, 200032, Shanghai, People's Republic of China.
| |
Collapse
|
5
|
Li Y, Tian Y, Zhong W, Wang N, Wang Y, Zhang Y, Zhang Z, Li J, Ma F, Zhao Z, Peng Y. Artemisia argyi Essential Oil Inhibits Hepatocellular Carcinoma Metastasis via Suppression of DEPDC1 Dependent Wnt/β-Catenin Signaling Pathway. Front Cell Dev Biol 2021; 9:664791. [PMID: 34268303 PMCID: PMC8276134 DOI: 10.3389/fcell.2021.664791] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2021] [Accepted: 04/01/2021] [Indexed: 11/19/2022] Open
Abstract
The tumor metastasis is the major hurdle for the treatment of advanced hepatocellular carcinoma (HCC), due in part to the lack of effective systemic treatments. DEPDC1, a novel oncoantigen upregulated in HCC, is thought to be a molecular-target for novel therapeutic drugs. Artemisia argyi is a traditional Chinese medicine with anti-inflammatory and anti-tumor activities. This study investigated the potential therapeutic benefits of Artemisia argyi essential oil (AAEO) in suppressing metastasis of HCC by targeting DEPDC1. Assessment of AAEO cytotoxicity was performed by MTT assay. Anti-metastatic effects of AAEO were investigated in vitro using wound healing and transwell assays. The HepG2 cells were transduced with lentiviral vector containing luciferase (Luc). A metastasis model of nude mice was established by tail vein injection of HepG2-Luc cells. The nude mice were treated with AAEO (57.5, 115, and 230 mg/kg) or sorafenib (40 mg/kg). Metastasis of HCC cells was monitored via in vivo bioluminescence imaging. After treatment for 21 days, tissues were collected for histological examination and immunohistochemistry analysis. Gene and protein levels were determined by real-time quantitative PCR and western blotting. The results revealed that AAEO significantly inhibits the migration and invasion in vitro in a concentration-dependent manner. In vivo assays further confirmed that AAEO markedly inhibits HCC metastasis into lung, brain, and femur tissues and exhibits low toxicity. Our results suggested that AAEO significantly downregulates the mRNA and protein expression of DEPDC1. Also, AAEO attenuated Wnt/β-catenin signaling through reduction of Wnt1 and β-catenin production. Moreover, AAEO prevented epithelial-mesenchymal transition (EMT) by downregulation of vimentin and upregulation of E-cadherin. Furthermore, we found that DEPDC1 promoted HCC migration and invasion via Wnt/β-catenin signaling pathway and EMT. These results demonstrate that AAEO effectively inhibits HCC metastasis via attenuating Wnt/β-catenin signaling and inhibiting EMT by suppressing DEPDC1 expression. Thus, AAEO likely acts as a novel inhibitor of the DEPDC1 dependent Wnt/β-catenin signaling pathway.
Collapse
Affiliation(s)
- Yanli Li
- Henan Key Laboratory for Pharmacology of Liver Diseases, Institute of Medical and Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, China
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, China
| | - Yang Tian
- Henan Key Laboratory for Pharmacology of Liver Diseases, Institute of Medical and Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, China
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, China
| | - Wei Zhong
- Department of Stomatology, People’s Hospital of Zhengzhou, Zhengzhou, China
| | - Ning Wang
- Henan Key Laboratory for Pharmacology of Liver Diseases, Institute of Medical and Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, China
| | - Yafeng Wang
- Henan Key Laboratory for Pharmacology of Liver Diseases, Institute of Medical and Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, China
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, China
| | - Yan Zhang
- Henan Key Laboratory for Pharmacology of Liver Diseases, Institute of Medical and Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, China
| | - Zhuangli Zhang
- Henan Key Laboratory for Pharmacology of Liver Diseases, Institute of Medical and Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, China
| | - Jianbo Li
- Henan Key Laboratory for Pharmacology of Liver Diseases, Institute of Medical and Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, China
| | - Fang Ma
- Henan Key Laboratory for Pharmacology of Liver Diseases, Institute of Medical and Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, China
| | - Zhihong Zhao
- Henan Key Laboratory for Pharmacology of Liver Diseases, Institute of Medical and Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, China
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, China
| | - Youmei Peng
- Henan Key Laboratory for Pharmacology of Liver Diseases, Institute of Medical and Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, China
| |
Collapse
|
6
|
Shen J, Xi M. [DEPDC1 is Highly Expressed in Lung Adenocarcinoma and Promotes Tumor Cell Proliferation]. ZHONGGUO FEI AI ZA ZHI = CHINESE JOURNAL OF LUNG CANCER 2021; 24:453-460. [PMID: 34134185 PMCID: PMC8317098 DOI: 10.3779/j.issn.1009-3419.2021.103.06] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
背景与目的 肺癌是目前世界范围内造成死亡人数最多的癌症之一,而肺腺癌是其主要分型。DEPDC1(DEP domain containing 1)被证实与多数肿瘤的发生发展密切相关,DEPDC1在肺腺癌中过表达已被初步证实,本研究旨在探究DEPDC1的表达与肺腺癌临床预后的关系,对DEPDC1作为肺腺癌潜在生物标志物和治疗靶点的可能性进行初步探讨。 方法 利用生物信息学网站GEPIA搜集相关信息,在线分析DEPDC1表达与患者预后生存关系。收集本医院患者资料,对收集的样本进行免疫组化染色,并进行统计学分析。随后,体外培养肺腺癌系细胞,通过Western blot与逆转录定量聚合酶链反应(reverse transcription-quantitative polymerase chain reaction, RT-qPCR)验证敲低效率,再进行细胞增殖的实验。 结果 DEPDC1在肺腺癌组织中表达显著高于癌旁正常组织,DEPDC1在肺腺癌组织中高表达,DEPDC1高表达与肺腺癌的肿瘤大小临床分期相关,敲低DEPDC1可抑制A549和H1975细胞增殖。 结论 DEPDC1在肺腺癌的进展演变中扮演着重要角色,有望成为肺腺癌重要的治疗靶点和一个潜在的新的生物标志物。
Collapse
Affiliation(s)
- Jian Shen
- Department of Rheumatology and Immunology, The Third Affiliated Hospital of Jinzhou Medical University, 121001 Jinzhou, China
| | - Mengmeng Xi
- Department of Respiratory Medicine, Jinzhou Women and Infant Hospital, 121001 Jinzhou, China
| |
Collapse
|
7
|
Gong Z, Chu H, Chen J, Jiang L, Gong B, Zhu P, Zhang C, Wang Z, Zhang W, Wang J, Li C, Zhao H. DEPDC1 upregulation promotes cell proliferation and predicts poor prognosis in patients with gastric cancer. Cancer Biomark 2021; 30:299-307. [PMID: 33361586 DOI: 10.3233/cbm-201760] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
BACKGROUND Previous studies revealed that DEP domain containing 1 (DEPDC1) is involved in the carcinogenesis and progression of several types of human cancer. However the role of DEPDC1 in gastric cancer has not been studied. OBJECTIVE The objective of this study was to study the expression and pathophysiological function of DEPDC1 in gastric cancer. METHODS DEPDC1 expression in gastric adenocarcinoma cells was examined with Western blot and qRT-PCR. Clinical pathological features of patients were determined by immunohistochemistry. The effect of DEPDC1 expression on cell proliferation was studied by in vitro cell proliferation assay; and cell cycle influence was assessed by flow cytometry. Survival curves were plotted using Kaplan-Meier. RESULTS DEPDC1 was overexpressed in gastric adenocarcinoma tissues compared with the paired adjacent normal gastric tissues, in accordance with mRNA level downloaded from GEPIA database. DEPDC1 expression level was significantly associated with cancer metastasis and differentiation. DEPDC1 upregulation caused cell cycle accelerating from G1 to S phase, and it was correlated with poorer overall survival. CONCLUSION Therefore, DEPDC1 upregulation in gastric adenocarcinoma is associated with tumor development and poor clinical outcomes of the patients, implying DEPDC1 might be a potential therapeutic target against gastric cancer.
Collapse
Affiliation(s)
- Zhaohua Gong
- Department of Oncology, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, Shandong, China.,Department of Oncology, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, Shandong, China
| | - Hongjin Chu
- Central Laboratory, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, Shandong, China.,Department of Oncology, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, Shandong, China
| | - Jian Chen
- Department of Oncology, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, Shandong, China
| | - Lixin Jiang
- Department of Gastrointestinal Surgery, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, Shandong, China
| | - Benjiao Gong
- Central Laboratory, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, Shandong, China
| | - Peng Zhu
- Central Laboratory, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, Shandong, China
| | - Chenglin Zhang
- Central Laboratory, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, Shandong, China
| | - Zhixin Wang
- Central Laboratory, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, Shandong, China
| | - Wendi Zhang
- Central Laboratory, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, Shandong, China
| | - Jiahui Wang
- Central Laboratory, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, Shandong, China
| | - Chen Li
- Department of Radiotherapy, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, Shandong, China
| | - Huishan Zhao
- Reproductive Medicine Centre, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, Shandong, China
| |
Collapse
|
8
|
Zador Z, Landry AP, Haibe-Kains B, Cusimano MD. Meta-gene markers predict meningioma recurrence with high accuracy. Sci Rep 2020; 10:18028. [PMID: 33093491 PMCID: PMC7582964 DOI: 10.1038/s41598-020-74482-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2020] [Accepted: 09/21/2020] [Indexed: 11/13/2022] Open
Abstract
Meningiomas, the most common adult brain tumors, recur in up to half of cases. This requires timely intervention and therefore accurate risk assessment of recurrence is essential. Our current practice relies heavily on histological grade and extent of surgical excision to predict meningioma recurrence. However, prediction accuracy can be as poor as 50% for low or intermediate grade tumors which constitute the majority of cases. Moreover, attempts to find molecular markers to predict their recurrence have been impeded by low or heterogenous genetic signal. We therefore sought to apply systems-biology approaches to transcriptomic data to better predict meningioma recurrence. We apply gene co-expression networks to a cohort of 252 adult patients from the publicly available genetic repository Gene Expression Omnibus. Resultant gene clusters (“modules”) were represented by the first principle component of their expression, and their ability to predict recurrence assessed with a logistic regression model. External validation was done using two independent samples: one merged microarray-based cohort with a total of 108 patients and one RNA-seq-based cohort with 145 patients, using the same modules. We used the bioinformatics database Enrichr to examine the gene ontology associations and driver transcription factors of each module. Using gene co-expression analysis, we were able predict tumor recurrence with high accuracy using a single module which mapped to cell cycle-related processes (AUC of 0.81 ± 0.09 and 0.77 ± 0.10 in external validation using microarray and RNA-seq data, respectively). This module remained predictive when controlling for WHO grade in all cohorts, and was associated with several cancer-associated transcription factors which may serve as novel therapeutic targets for patients with this disease. With the easy accessibility of gene panels in healthcare diagnostics, our results offer a basis for routine molecular testing in meningioma management and propose potential therapeutic targets for future research.
Collapse
Affiliation(s)
- Zsolt Zador
- Division of Neurosurgery, Department of Surgery, St. Michael's Hospital, Toronto, ON, Canada.
| | - Alexander P Landry
- Division of Neurosurgery, Department of Surgery, St. Michael's Hospital, Toronto, ON, Canada.
| | - Benjamin Haibe-Kains
- Department of Computer Science, University of Toronto, Toronto, ON, Canada.,Department of Medical Biophysics, University of Toronto, Toronto, ON, Canada.,Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada.,Ontario Institute for Cancer Research, Toronto, ON, Canada.,Vector Institute, Toronto, ON, Canada
| | - Michael D Cusimano
- Division of Neurosurgery, Department of Surgery, St. Michael's Hospital, Toronto, ON, Canada
| |
Collapse
|
9
|
Mishra NK, Niu M, Southekal S, Bajpai P, Elkholy A, Manne U, Guda C. Identification of Prognostic Markers in Cholangiocarcinoma Using Altered DNA Methylation and Gene Expression Profiles. Front Genet 2020; 11:522125. [PMID: 33193605 PMCID: PMC7606733 DOI: 10.3389/fgene.2020.522125] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2019] [Accepted: 08/21/2020] [Indexed: 12/30/2022] Open
Abstract
Background Cholangiocarcinoma (CCA) is a rare disease, but it is amongst the most lethal cancers with a median survival under 1 year. Variations in DNA methylation and gene expression have been extensively studied in other cancers for their role in pathogenesis and disease prognosis, but these studies are very limited in CCA. This study focusses on the identification of DNA methylation and gene expression prognostic biomarkers using multi-omics data of CCA tumors from The Cancer Genome Atlas (TCGA). Method We have conducted a genome-wide analysis of differential DNA methylation and gene/miRNA expression using data from 36 CCA tumor and 9 normal samples from TCGA. The impact of DNA methylation in promoters and long-range distal enhancers on the regulation and expression of CCA-associated genes was examined using linear regression. Next, we conducted network analyses on genes which are regulated by DNA methylation as well as by miRNA. Finally, we performed Kaplan–Meier and Cox proportional hazards regression analyses in order to identify the role of selected methylation sites and specific genes and miRNAs in patient survival. We also performed real-time quantitative PCR (qPCR) to confirm the change in gene expression in CCA patients’ tumor and adjacent normal samples. Results Altered DNA methylation was observed on 12,259 CpGs across all chromosomes, of which 78% were hypermethylated. We observed a strong negative relationship between promoter hypermethylation and corresponding gene expression in 92% of the CpGs. Differential expression analyses revealed altered expression patterns in 3,305 genes and 101 miRNAs. Finally, we identified 17 differentially methylated promoter CpGs, 72 differentially expressed genes, and two miRNAs that are likely associated with patient survival. Pathway analysis suggested that cell division, bile secretion, amino acid metabolism, PPAR signaling, hippo signaling were highly affected by gene expression and DNA methylation alterations. The qPCR analysis further confirmed that MDK, HNF1B, PACS1, and GLUD1 are differentially expressed in CCA. Conclusion Based on the survival analysis, we conclude that DEPDC1, FUT4, MDK, PACS1, PIWIL4 genes, miR-22, miR-551b microRNAs, and cg27362525 and cg26597242 CpGs can strongly support their use as prognostic markers of CCA.
Collapse
Affiliation(s)
- Nitish Kumar Mishra
- Department of Genetics, Cell Biology and Anatomy, University of Nebraska Medical Center, Omaha, NE, United States
| | - Meng Niu
- Department of Genetics, Cell Biology and Anatomy, University of Nebraska Medical Center, Omaha, NE, United States
| | - Siddesh Southekal
- Department of Genetics, Cell Biology and Anatomy, University of Nebraska Medical Center, Omaha, NE, United States
| | - Prachi Bajpai
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Amr Elkholy
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Upender Manne
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Chittibabu Guda
- Department of Genetics, Cell Biology and Anatomy, University of Nebraska Medical Center, Omaha, NE, United States
| |
Collapse
|
10
|
Chen X, Guo ZQ, Cao D, Chen Y, Chen J. Knockdown of DEPDC1B inhibits the development of glioblastoma. Cancer Cell Int 2020; 20:310. [PMID: 32684847 PMCID: PMC7362545 DOI: 10.1186/s12935-020-01404-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2019] [Accepted: 07/06/2020] [Indexed: 11/10/2022] Open
Abstract
Background Glioblastoma (GBM) is the most common primary malignant brain tumor in adults with a poor prognosis. DEPDC1B (DEP domain-containing protein 1B) has been shown to be associated with some types of malignancies. However, the role and underlying regulatory mechanisms of DEPDC1B in GBM remain elusive. Methods In this research, the expression level of DEPDC1B in GBM tissues was detected by IHC. The DEPDC1B knockdown cell line was constructed, identified by qRT-PCR and western blot and used to construct the xenotransplantation mice model and intracranial xenograft model. MTT assay, colony formation assay, flow cytometry, and Transwell assay were used to detected cell proliferation, apoptosis and migration. Results The results proved that DEPDC1B was significantly upregulated in tumor tissues, and silencing DEPDC1B could inhibit proliferation, migration and promote apoptosis of GBM cell. In addition, human apoptosis antibody array detection showed that after DEPDC1B knockdown, the expression of apoptosis-related proteins was downregulated, such as IGFBP-2, Survivin, N-cadherin, Vimentin and Snail. Finally, we indicated that knockdown of DEPDC1B significantly inhibited tumor growth in vivo. Conclusions In summary, DEPDC1B was involved in the development and progression of GBM, which may be a potential therapeutic target and bring a breakthrough in the treatment.
Collapse
Affiliation(s)
- Xu Chen
- Department of Neurosurgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Jiefang Ave, 1095, Wuhan, 430030 China
| | - Zheng-Qian Guo
- Department of Neurosurgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Jiefang Ave, 1095, Wuhan, 430030 China
| | - Dan Cao
- Department of Neurosurgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Jiefang Ave, 1095, Wuhan, 430030 China
| | - Yong Chen
- Department of Neurosurgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Jiefang Ave, 1095, Wuhan, 430030 China
| | - Jian Chen
- Department of Neurosurgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Jiefang Ave, 1095, Wuhan, 430030 China
| |
Collapse
|
11
|
Wang Q, Jiang S, Liu J, Ma G, Zheng J, Zhang Y. DEP Domain Containing 1 Promotes Proliferation, Invasion, and Epithelial-Mesenchymal Transition in Colorectal Cancer by Enhancing Expression of Suppressor of Zest 12. Cancer Biother Radiopharm 2020; 36:36-44. [PMID: 32343606 DOI: 10.1089/cbr.2019.3460] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Objective: DEP domain containing 1 (DEPDC1), aberrantly upregulated in various tumors, has been shown to be involved in the occurrence and development of tumors. This study aims to investigate pathophysiological roles of DEPDC1 in colorectal cancer (CRC). Materials and Methods: Expression level of DEPDC1 and suppressor of zest 12 (SUZ12) in CRC tissues and cell lines were analyzed by quantitative real-time polymerase chain reaction and immunohistochemistry. Staining with 5-bromo-2-deoxyuridine staining and colony formation assays were conducted to evaluate cell proliferation. Transwell or wound healing assay to evaluate invasion or migration, respectively. The effect on epithelial-mesenchymal transition (EMT) of CRC was determined by Western blot. Results: DEPDC1 and SUZ12 were increased in CRC tissues and cell lines. Silence of DEPDC1 suppressed cell proliferation, migration, and invasion of CRC. Moreover, DEPDC1 knockdown suppressed EMT of CRC. Mechanistically, the authors demonstrated silencing DEPDC1 decreased protein expression of SUZ12 and led to a remarkable reduction of trimethylation on the lysine 27 residue of histone H3 (H3K27Me3). Inhibitory ability of DEPDC1 knockdown on CRC progression was reversed by overexpression of SUZ12. Conclusions: DEPDC1 promoted CRC progression through regulation of SUZ12-mediated H3K27Me3, illuminating a novel DEPDC1-SUZ12 molecular axis as regulator in CRC progression and suggesting potential implications in treatment of CRC.
Collapse
Affiliation(s)
- Qiang Wang
- Department of Surgical Oncology, Mindong Hospital Affiliated to Fujian Medical University, Ningde, China
| | - Shijian Jiang
- Department of Surgical Oncology, Mindong Hospital Affiliated to Fujian Medical University, Ningde, China
| | - Jianchao Liu
- Department of General Surgery, Liaocheng People's Hospital, Liaocheng, China
| | - Genshun Ma
- Department of General Surgery, Liaocheng People's Hospital, Liaocheng, China
| | - Jianrui Zheng
- Department of Pathology, Mindong Hospital Affiliated to Fujian Medical University, Ningde, China
| | - Yajun Zhang
- Department of General Surgery, Liaocheng People's Hospital, Liaocheng, China
| |
Collapse
|
12
|
Zhou C, Wang P, Tu M, Huang Y, Xiong F, Wu Y. DEPDC1 promotes cell proliferation and suppresses sensitivity to chemotherapy in human hepatocellular carcinoma. Biosci Rep 2019; 39:BSR20190946. [PMID: 31189746 PMCID: PMC6620382 DOI: 10.1042/bsr20190946] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2019] [Revised: 06/07/2019] [Accepted: 06/12/2019] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND Hepatocellular carcinoma (HCC) is one of the major causes of tumor-related morbidity and mortality worldwide. Accumulating evidence has revealed that aberrant expression of crucial cancer-related genes contributes to hepatocellular carcinogenesis. This study aimed to characterize the biological role of DEP domain containing 1 (DEPDC1), a novel cancer-related gene, in HCC and illuminate the potential molecular mechanisms involved. MATERIALS AND METHODS Quantitative real-time PCR (qRT-PCR), Western blotting and immunohistochemical (IHC) staining were used to characterize the expression patterns of DEPDC1 in tumorous tissues and adjacent normal tissues. Kaplan-Meier survival analysis was launched to evaluate the relationship between DEPDC1 expression and overall survival. CCK8 assay, colony formation and flow cytometry were performed to investigate the effects of DEPDC1 on HCC cell viability, clonogenic capability and cell apoptosis. Murine xenograft models were established to determine the effect of DEPDC1 on tumor growth in vivo SP600125, a JNK specific inhibitor, was applied to carriy out mechanistic studies. RESULTS DEPDC1 was significantly up-regulated in HCC tissues compared with para-cancerous tissues. Besides, patients with high DEPDC1 expression experienced a significantly shorter overall survival. Functional investigations demonstrated that DEPDC1 overexpression facilitated HCC cell proliferation and suppressed cell apoptosis, whereas DEPDC1 depletion inhibited cell proliferation and promoted cell apoptosis. Furthermore, DEPDC1 ablation suppressed tumorigenecity of HCC cells in murine xenograft models. Mechanistic studies uncovered that JNK signaling pathway mediated the promoting effects of DEPDC1 on HCC cell viability and chemotherapy resistance. CONCLUSION Collectively, our data may provide some evidence for DEPDC1 as a candidate therapeutic target for HCC.
Collapse
Affiliation(s)
- Chao Zhou
- Department of Gastroenterology, Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, Chengdu 610072, China
| | - Pu Wang
- Department of Gastroenterology, Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, Chengdu 610072, China
| | - Mengtian Tu
- Department of Gastroenterology, Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, Chengdu 610072, China
| | - Yi Huang
- Department of Laboratory Medicine, Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, Chengdu 610072, China
| | - Fei Xiong
- Department of Gastroenterology, Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, Chengdu 610072, China
| | - Yue Wu
- Department of Pharmacy, Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, Chengdu 610072, China
| |
Collapse
|
13
|
Guo W, Li H, Liu H, Ma X, Yang S, Wang Z. DEPDC1 drives hepatocellular carcinoma cell proliferation, invasion and angiogenesis by regulating the CCL20/CCR6 signaling pathway. Oncol Rep 2019; 42:1075-1089. [PMID: 31322256 PMCID: PMC6667871 DOI: 10.3892/or.2019.7221] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2019] [Accepted: 07/03/2019] [Indexed: 12/16/2022] Open
Abstract
DEP domain containing 1 (DEPDC1) functions as an oncogene in hepatocellular carcinoma (HCC). However, the underlying mechanism of DEPDC1 remains largely unknown. The present study revealed that DEPDC1 knockdown inhibited HCC cell proliferation, colony formation and invasion in vitro and suppressed the growth of HCC xenografts in vivo. Furthermore, DEPDC1 overexpression promoted HCC cell proliferation, colony formation and invasion. DNA microarray, reverse transcription-quantitative-PCR and western blotting results demonstrated that DEPDC1 knockdown in Huh-7 significantly inhibited the expression of chemokine (C-C motif) ligand 20 (CCL20) and chemokine (C-C motif) receptor 6 (CCR6). In addition, the expression of CCL20 and CCR6 were upregulated in HCC tissues and cell lines, and were positively correlated with DEPDC1 expression. CCL20 or CCR6 knockdown via small interfering RNA reversed the effects of DEPDC1 overexpression in HCC cells. Furthermore, it was revealed that conditioned medium from DEPDC1 upregulated Li-7 and Hep3B cells led to angiogenesis in vitro, whereas CCL20 knockdown in Li-7 and Hep3B cells or CCR6 knockdown in human umbilical vein endothelial cells reversed the angiogenic effect of DEPDC1 overexpression. In conclusion, DEPDC1 facilitated cell proliferation, invasion and angiogenesis via the CCL20/CCR6 pathway in HCC.
Collapse
Affiliation(s)
- Wubin Guo
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, P.R. China
| | - Hui Li
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, P.R. China
| | - Huan Liu
- Research Laboratory of Biomedical Engineering, The TCM Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, P.R. China
| | - Xin Ma
- Department of General Surgery, The TCM Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, P.R. China
| | - Sijin Yang
- Department of Cardiovascular Medicine, The TCM Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, P.R. China
| | - Ziwei Wang
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, P.R. China
| |
Collapse
|
14
|
Hao S, Tian W, Chen Y, Wang L, Jiang Y, Gao B, Luo D. MicroRNA-374c-5p inhibits the development of breast cancer through TATA-box binding protein associated factor 7-mediated transcriptional regulation of DEP domain containing 1. J Cell Biochem 2019; 120:15360-15368. [PMID: 31162714 PMCID: PMC6771484 DOI: 10.1002/jcb.28803] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2019] [Accepted: 04/12/2019] [Indexed: 12/15/2022]
Abstract
Breast cancer is the most pervasive cancer tormenting women, with increasing incidence and mortality rates year after year. MicroRNAs (miRNAs) with abnormal expression has various effects in biological processes and progression in diverse tumors. Nevertheless, it is vitally crucial for us to inspect more underlying molecular mechanisms for the therapy of patients with breast cancer. In the paper, we inquired the expression level and potential regulation mechanism of miR‐374c‐5p in breast cancer. Our research found out that miR‐374c‐5p was low‐level expressed in breast cancer. Upregulation of miR‐374c‐5p repressed cell proliferation, migration, and also epithelial‐mesenchymal transition (EMT), and induced cell apoptosis of breast cancer cells. Further, we concluded that miR‐374c‐5p interacted with TAF7 and downregulated its expression. Moreover, miR‐374c‐5p modulated DEP domain containing 1 (DEPDC1) through mediating TAF7. Finally, rescue assays represented that miR‐374c‐5p suppressed breast cancer development via TAF7‐mediated transcriptional regulation of DEPDC1. We uncovered that overexpressed miR‐374c‐5p inhibited the development of breast cancer via TAF7‐regulated transcriptional regulation of DEPDC1, which may be a novel and vital proportion of cancer diagnosis and treatment strategies.
Collapse
Affiliation(s)
- Shuai Hao
- Department of Breast, Thyroid Surgery, Daping Hospital, Army Medical University, Chongqing, China
| | - Wuguo Tian
- Department of Breast, Thyroid Surgery, Daping Hospital, Army Medical University, Chongqing, China
| | - Yi Chen
- Department of Breast, Thyroid Surgery, Daping Hospital, Army Medical University, Chongqing, China
| | - Lingli Wang
- Department of Breast, Thyroid Surgery, Daping Hospital, Army Medical University, Chongqing, China
| | - Yan Jiang
- Department of Breast, Thyroid Surgery, Daping Hospital, Army Medical University, Chongqing, China
| | - Bo Gao
- Department of Breast, Thyroid Surgery, Daping Hospital, Army Medical University, Chongqing, China
| | - Donglin Luo
- Department of Breast, Thyroid Surgery, Daping Hospital, Army Medical University, Chongqing, China
| |
Collapse
|
15
|
Zhao H, Yu M, Sui L, Gong B, Zhou B, Chen J, Gong Z, Hao C. High Expression of DEPDC1 Promotes Malignant Phenotypes of Breast Cancer Cells and Predicts Poor Prognosis in Patients With Breast Cancer. Front Oncol 2019; 9:262. [PMID: 31032225 PMCID: PMC6473048 DOI: 10.3389/fonc.2019.00262] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2018] [Accepted: 03/22/2019] [Indexed: 12/15/2022] Open
Abstract
DEP domain containing 1 (DEPDC1) is a novel tumor-associated gene, which is aberrantly expressed in multiple types of cancer and involves in tumorigenesis and cancer progression. Here, we examined the functional involvement and underlying mechanism of DEPDC1 in breast cancer. In this study, the immunohistochemistry results demonstrated that DEPDC1 was high-expressed in breast cancer tissues compared with the paired adjacent normal breast tissues, and its tendency at protein level was consistent with mRNA level from TCGA data. Moreover, DEPDC1 mRNA level revealed the strongest association with poor prognosis and development in breast cancer. In vitro assays showed that DEPDC1 overexpression resulted in significant promotion of proliferation by regulating cell cycle in MCF-7 cells, whilst an opposite effect was found in the MDA-MB-231 cells with DEPDC1 deletion. Notably, further investigation indicated DEPDC1's ability of promoting breast cancer cells migration and invasion. In addition, we discovered that DEPDC1 caused hyper-activation of PI3K/AKT/mTOR signaling in breast cancer cells. Therefore, the increased DEPDC1 expression in breast cancer is correlated with disease progression and poor survival, which suggested that DEPDC1 might be a potential therapeutic target against this disease.
Collapse
Affiliation(s)
- Huishan Zhao
- Reproductive Medicine Centre, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, China
| | - Mingwei Yu
- Department of Orthopedics, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, China
| | - Laijian Sui
- Department of Orthopedics, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, China
| | - Benjiao Gong
- Central Laboratory, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, China
| | - Bo Zhou
- Department of Special Education, Binzhou Medical University, Yantai, China
| | - Jian Chen
- Central Laboratory, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, China.,Department of Oncology, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, China
| | - Zhaohua Gong
- Department of Oncology, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, China
| | - Cuifang Hao
- Reproductive Medicine Centre, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, China
| |
Collapse
|
16
|
Kikuchi R, Ueda R, Saito K, Shibao S, Nagashima H, Tamura R, Morimoto Y, Sasaki H, Noji S, Kawakami Y, Yoshida K, Toda M. A Pilot Study of Vaccine Therapy with Multiple Glioma Oncoantigen/Glioma Angiogenesis-Associated Antigen Peptides for Patients with Recurrent/Progressive High-Grade Glioma. J Clin Med 2019; 8:jcm8020263. [PMID: 30791546 PMCID: PMC6406695 DOI: 10.3390/jcm8020263] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2019] [Revised: 02/13/2019] [Accepted: 02/14/2019] [Indexed: 01/01/2023] Open
Abstract
High-grade gliomas (HGGs) carry a dismal prognosis despite current treatments. We previously confirmed the safety and immunogenicity of a vaccine treatment targeting tumor angiogenesis with synthetic peptides, for vascular endothelial growth factor receptor (VEGFR) epitopes in recurrent HGG patients. In this study, we evaluated a novel vaccine therapy targeting not only tumor vasculature but also tumor cells, using multiple glioma oncoantigen (GOA)/glioma angiogenesis-associated antigen (GAAA) peptides in HLA-A2402+ recurrent/progressive HGG patients. The vaccine included peptide epitopes from four GOAs (LY6K, DEPDC1, KIF20A, and FOXM1) and two GAAAs (VEGFR1 and VEGFR2). Ten patients received subcutaneous vaccinations. The primary endpoint was the safety of the treatment. T-lymphocyte responses against GOA/GAAA epitopes and treatment response were evaluated secondarily. The treatment was well tolerated without any severe systemic adverse events. The vaccinations induced immunoreactivity to at least three vaccine-targeted GOA/GAAA in all six evaluable patients. The median overall survival time in all patients was 9.2 months. Five achieved progression-free status lasting at least six months. Two recurrent glioblastoma patients demonstrated stable disease. One patient with anaplastic oligoastrocytoma achieved complete response nine months after the vaccination. Taken together, this regimen was well tolerated and induced robust GOA/GAAA-specific T-lymphocyte responses in recurrent/progressive HGG patients.
Collapse
Affiliation(s)
- Ryogo Kikuchi
- Department of Neurosurgery, Hiratsuka City Hospital, Hiratsuka, Kanagawa 254-0019, Japan.
- Department of Neurosurgery, Keio University School of Medicine, Shinjuku, Tokyo 160-8587, Japan.
| | - Ryo Ueda
- Department of Neurosurgery, Keio University School of Medicine, Shinjuku, Tokyo 160-8587, Japan.
- Department of Neurosurgery, Kawasaki Municipal Hospital, Kawasaki, Kanagawa 210-0013, Japan.
| | - Katsuya Saito
- Department of Neurosurgery, Ashikaga Red Cross Hospital, Ashikaga, Tochigi 326-0843, Japan.
| | - Shunsuke Shibao
- Department of Neurosurgery, Ashikaga Red Cross Hospital, Ashikaga, Tochigi 326-0843, Japan.
| | - Hideaki Nagashima
- Department of Neurosurgery, Keio University School of Medicine, Shinjuku, Tokyo 160-8587, Japan.
| | - Ryota Tamura
- Department of Neurosurgery, Keio University School of Medicine, Shinjuku, Tokyo 160-8587, Japan.
| | - Yukina Morimoto
- Department of Neurosurgery, Keio University School of Medicine, Shinjuku, Tokyo 160-8587, Japan.
| | - Hikaru Sasaki
- Department of Neurosurgery, Keio University School of Medicine, Shinjuku, Tokyo 160-8587, Japan.
| | - Shinobu Noji
- Division of Cellular Signaling, Institute for Advanced Medical Research, Keio University School of Medicine, Shinjku, Tokyo 160-8587, Japan.
| | - Yutaka Kawakami
- Division of Cellular Signaling, Institute for Advanced Medical Research, Keio University School of Medicine, Shinjku, Tokyo 160-8587, Japan.
| | - Kazunari Yoshida
- Department of Neurosurgery, Keio University School of Medicine, Shinjuku, Tokyo 160-8587, Japan.
| | - Masahiro Toda
- Department of Neurosurgery, Keio University School of Medicine, Shinjuku, Tokyo 160-8587, Japan.
| |
Collapse
|