1
|
Chen Y, Wang C, Hu S, Liu X. HRS Facilitates Newcastle Disease Virus Replication in Tumor Cells by Promoting Viral Budding. Int J Mol Sci 2024; 25:10060. [PMID: 39337546 PMCID: PMC11432301 DOI: 10.3390/ijms251810060] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Revised: 09/13/2024] [Accepted: 09/17/2024] [Indexed: 09/30/2024] Open
Abstract
Newcastle disease virus (NDV) is a highly pathogenic avian infectious disease agent and also a promising oncolytic virus with broad application prospects. The Endosomal Sorting Complex Required for Transport (ESCRT) machinery has been increasingly recognized for its crucial role in the life cycles of enveloped viruses, influencing processes such as viral entry, replication, and budding. In this study, we employed an RNA interference screening approach to identify key ESCRT components that regulate NDV replication in tumor cells. qPCR, immunofluorescence, and Western blot assays demonstrated that knockdown of HRS, CHMP4A, CHMP4B, and CHMP4C significantly impaired NDV replication in HeLa cells, with HRS exhibiting the most pronounced inhibitory effect. Additionally, HRS knockout significantly inhibited viral budding and suppressed NDV-induced cell death in HeLa cells. Notably, NDV infection was shown to significantly upregulate HRS gene and protein expression in a time-dependent manner. In conclusion, this study systematically identifies critical ESCRT components involved in NDV replication within tumor cells, with a particular focus on the role of HRS in promoting NDV's replication by promoting viral budding, offering new insights for the development of NDV-based oncolytic therapies.
Collapse
Affiliation(s)
- Yu Chen
- Animal Infectious Disease Laboratory, College of Veterinary Medicine, Yangzhou University, Yangzhou 225012, China
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonosis, Yangzhou University, Yangzhou 225012, China
- Jiangsu Key Laboratory of Zoonosis, Yangzhou University, Yangzhou 225012, China
| | - Chunxuan Wang
- Animal Infectious Disease Laboratory, College of Veterinary Medicine, Yangzhou University, Yangzhou 225012, China
| | - Shunlin Hu
- Animal Infectious Disease Laboratory, College of Veterinary Medicine, Yangzhou University, Yangzhou 225012, China
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonosis, Yangzhou University, Yangzhou 225012, China
- Jiangsu Key Laboratory of Zoonosis, Yangzhou University, Yangzhou 225012, China
| | - Xiufan Liu
- Animal Infectious Disease Laboratory, College of Veterinary Medicine, Yangzhou University, Yangzhou 225012, China
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonosis, Yangzhou University, Yangzhou 225012, China
- Jiangsu Key Laboratory of Zoonosis, Yangzhou University, Yangzhou 225012, China
| |
Collapse
|
2
|
Hussain T, Chai L, Wang Y, Zhang Q, Wang J, Shi W, Wang Q, Li M, Xie X. Activation of PPAR-γ prevents TERT-mediated pulmonary vascular remodeling in MCT-induced pulmonary hypertension. Heliyon 2023; 9:e14173. [PMID: 36938425 PMCID: PMC10015197 DOI: 10.1016/j.heliyon.2023.e14173] [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: 11/06/2022] [Revised: 02/22/2023] [Accepted: 02/23/2023] [Indexed: 03/06/2023] Open
Abstract
Background It has been demonstrated that elevated telomerase reverse transcriptase (TERT) expression or activity is implicated in pulmonary hypertension (PH). In addition, activation of peroxisome-proliferator-activated receptor γ (PPAR-γ) has been found to prevent PH progression. However, the molecular mechanism responsible for the protective effect of PPAR-γ activation on TERT expression in the pathogenesis of PH remains unknown. This study was performed to address these issues. Methods Intraperitoneal injection of monocrotaline (MCT) was used to establish PH. BIBR1532 was applied to inhibit the activity of telomerase. The right ventricular systolic pressure (RVSP) and histological analysis were used to detect the development of PH. The protein levels of p-Akt, t-Akt, c-Myc and TERT were determined by western blotting. Pharmacological inhibition of TERT by BIBR1532 effectively suppressed RVSP, RVHI and the WT% in MCT-induced PH rats. Results Pharmacological inhibition of Akt/c-Myc pathway by LY294002 diminished TERT upregulation, RVSP, RVHI and WT% in MCT-PH rats. Activation of PPAR-γ by pioglitazone inhibited p-Akt and c-Myc expressions and further downregulated TERT, thus to reduced RVSP, RVHI and WT% in MCT-treated PH rats. Conclusions In conclusion, TERT upregulation contributes to PH development in MCT-treated rats. Activation of PPAR-γ prevents pulmonary arterial remodeling through Akt/c-Myc/TERT axis suppression.
Collapse
Affiliation(s)
- Tafseel Hussain
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, PR China
| | - Limin Chai
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, PR China
| | - Yan Wang
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, PR China
| | - Qianqian Zhang
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, PR China
| | - Jian Wang
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, PR China
| | - Wenhua Shi
- Department of Respiratory and Critical Care Medicine, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, PR China
| | - Qingting Wang
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, PR China
| | - Manxiang Li
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, PR China
| | - Xinming Xie
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, PR China
- Corresponding author. Department of Respiratory and Critical Care Medicine, the First Affiliated Hospital of Xi'an Jiaotong University, No. 277, West Yanta Road, Xi'an, Shaanxi 710061, PR China.
| |
Collapse
|
3
|
Chen H, Wei L, Luo M, Wang X, Zhu C, Huang H, Liu X, Lu H, Zhong Y. LINC00324 suppresses apoptosis and autophagy in nasopharyngeal carcinoma through upregulation of PAD4 and activation of the PI3K/AKT signaling pathway. Cell Biol Toxicol 2022; 38:995-1011. [PMID: 34322788 DOI: 10.1007/s10565-021-09632-x] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Accepted: 07/13/2021] [Indexed: 01/25/2023]
Abstract
BACKGROUND Nasopharyngeal carcinoma (NPC) has high incidence in Southern China and is derived from the mucosal epithelium of the nasopharynx. Accumulating evidence has revealed that peptidyl arginine deiminase 4 (PAD4) exerts carcinogenic effect on certain cancers. We designed this study to probe the specific role that PAD4 plays in NPC and its molecular mechanism. METHODS PAD4 expression in NPC cells was detected by RT-qPCR analysis. MTT, colony formation, flow cytometry, TUNEL staining, and LC3-II punctuation experiments were done to probe into the biological functions of PAD4 on NPC cellular behaviors in vitro. Subsequently, the upstream regulatory mechanism of PAD4 was investigated by luciferase reporter, RNA pull-down, and RIP assays. The impact of PAD4 on NPC tumor growth in mice was assessed by in vivo xenograft tumor assay. RESULTS PAD4 was upregulated in NPC cells. PAD4 knockdown suppressed proliferative ability and promoted apoptosis and autophagy in NPC cells. Additionally, PAD4 expression was negatively regulated by microRNA 3164 (miR-3164). LINC00324 positively upregulated PAD4 expression by interacting with miR-3164 and recruiting HuR protein. The LINC00324/miR-3164/PAD4 axis modulated the PI3K/AKT pathway in NPC cells. Moreover, PAD4 upregulation countervailed the influences of LINC00324 deficiency on NPC cell proliferation, apoptosis, and autophagy and on NPC tumor growth in mice. CONCLUSION LINC00324 promoted NPC malignancy by upregulation of PAD4 to activate the PI3K/AKT pathway.
Collapse
Affiliation(s)
- Hao Chen
- Department of Radiation Oncology, People's Hospital of Guangxi Zhuang Autonomous Region, No. 6 Taoyuan Road, Nanning, 530021, Guangxi, China
| | - Lining Wei
- Department of Endoscopy, The Affiliated Tumor Hospital of Guangxi Medical University, Nanning, 530021, Guangxi, China
| | - Min Luo
- Department of Oncology, The Third Affiliated Hospital of Guangxi Medical University, Nanning, 530031, Guangxi, China
| | - Xiaochen Wang
- Department of Oncology, The Third Affiliated Hospital of Guangxi Medical University, Nanning, 530031, Guangxi, China
| | - Chaohua Zhu
- Department of Radiation Oncology, People's Hospital of Guangxi Zhuang Autonomous Region, No. 6 Taoyuan Road, Nanning, 530021, Guangxi, China
| | - Huixian Huang
- Department of Radiation Oncology, People's Hospital of Guangxi Zhuang Autonomous Region, No. 6 Taoyuan Road, Nanning, 530021, Guangxi, China
| | - Xu Liu
- Department of Radiation Oncology, People's Hospital of Guangxi Zhuang Autonomous Region, No. 6 Taoyuan Road, Nanning, 530021, Guangxi, China
| | - Heming Lu
- Department of Radiation Oncology, People's Hospital of Guangxi Zhuang Autonomous Region, No. 6 Taoyuan Road, Nanning, 530021, Guangxi, China.
| | - Yahua Zhong
- Department of Radiation and Medical Oncology, Zhongnan Hospital of Wuhan University, 169 Donghu Road, Wuchang District, Wuhan, 430000, Hubei, China.
| |
Collapse
|
4
|
Phan NN, Liu S, Wang CY, Hsu HP, Lai MD, Li CY, Chen CF, Chiao CC, Yen MC, Sun Z, Jiang JZ. Overexpressed gene signature of EPH receptor A/B family in cancer patients-comprehensive analyses from the public high-throughput database. INTERNATIONAL JOURNAL OF CLINICAL AND EXPERIMENTAL PATHOLOGY 2020; 13:1220-1242. [PMID: 32509099 PMCID: PMC7270671] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 02/23/2020] [Accepted: 03/27/2020] [Indexed: 06/11/2023]
Abstract
Although a previous study suggested that erythropoietin-producing hepatoma (EPH) receptors play important roles in tumor progression and the overexpression of EPHs in cancer patients is related to poor prognoses, high-throughput gene expression profiling of EPH family members in different types and subtypes of cancers has so far not been conducted. We herein carried out a series of bioinformatic analyses on expressive profiles of every EPH member across 21 different types of clinical cancers versus matched normal tissues gathered from the Oncomine platform. We validated these results by protein expression study of all EPHs family members by The Human Protein Atlas repository. Our results uncovered the overexpression of most EPH subunits in numerous cancer types, especially the dramatic overexpression of six EPHs members, namely EPHA1, EPHA2, EPHA3, EPHA4 and EPHB1, EPHB2, EPHB3, EPHB4 in bladder, colorectal, esophageal, gastric, and prostate cancers. Furthermore, EPHB2 was specifically highly expressed in cervical cancer, EPHA3 in liver cancer, and EPHB1 in uterine cancer. Collectively, expressive profiles of these EPHs were confirmed and correlated with different cancer subtypes as potential biomarkers. This study provides useful information for further studies on cancer development and clinical treatments.
Collapse
Affiliation(s)
- Nam Nhut Phan
- NTT Institute of Hi-Technology, Nguyen Tat Thanh UniversityHo Chi Minh City 700000, Vietnam
| | - Shirui Liu
- Department of Mechanical Engineering, School of Engineering, San Francisco State UniversitySan Francisco, CA 94143, USA
- Department of Mechanical Design Manufacturing and Automation, College of Mechanical Engineering, Taiyuan University of Science and TechnologyShanxi 030024, People’s Republic of China
| | - Chih-Yang Wang
- Department of Biochemistry and Molecular Biology, Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung UniversityTainan 11031, Taiwan
- PhD Program for Cancer Molecular Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical UniversityTaipei 11031, Taiwan
- Graduate Institute of Cancer Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical UniversityTaipei 11031, Taiwan
| | - Hui-Ping Hsu
- Department of Surgery, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung UniversityTainan 704, Taiwan
| | - Ming-Derg Lai
- Department of Biochemistry and Molecular Biology, Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung UniversityTainan 11031, Taiwan
| | - Chung-Yen Li
- Department of Biochemistry and Molecular Biology, Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung UniversityTainan 11031, Taiwan
| | - Chien-Fu Chen
- School of Chinese Medicine for Post-Baccalaureate, I-Shou UniversityKaohsiung 84001, Taiwan
| | - Chung-Chieh Chiao
- School of Chinese Medicine for Post-Baccalaureate, I-Shou UniversityKaohsiung 84001, Taiwan
| | - Meng-Chi Yen
- Department of Emergency Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical UniversityKaohsiung 80708, Taiwan
| | - Zhengda Sun
- Department of Radiology, University of CaliforniaSan Francisco, San Francisco, CA 94143, USA
| | - Jia-Zhen Jiang
- Emergency Department, Huashan Hospital North, Fudan UniversityShanghai 201508, People’s Republic of China
| |
Collapse
|
5
|
EPHA3 enhances macrophage autophagy and apoptosis by disrupting the mTOR signaling pathway in mice with endometriosis. Biosci Rep 2019; 39:BSR20182274. [PMID: 31262977 PMCID: PMC6667729 DOI: 10.1042/bsr20182274] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2018] [Revised: 05/27/2019] [Accepted: 06/18/2019] [Indexed: 12/28/2022] Open
Abstract
Background: Endometriosis is a chronic fibrotic disease characterized by agonizing pelvic pain and low fertility, mainly affecting middle-aged women. The aim of the present study is to assess the potential effects of erythropoietin-producing hepatocellular carcinoma A3 (EPHA3) on endometriosis, with emphasis on the autophagy and apoptosis of macrophages via inhibition of the mammalian target of rapamycin (mTOR) signaling pathway. Methods: The mouse models of endometriosis were established followed by culturing the macrophages and macrophage transfection via the EPHA3 vector, siRNA EPHA3, and RAPA (an inhibitor of the mTOR signaling pathway). The expression of EPHA3, related factors in the mTOR signaling pathway, macrophage autophagy (autophagy-related gene 3 (Atg3), light chain 3-I (LC3-I), light chain 3-II (LC3-II) and Beclin1) and apoptosis (B-cell lymphoma-2 (bcl-2), bax and fas) were all detected and documented, respectively. The changes of autophagic lysosomes and the apoptosis of macrophages in each group following transfection were also inspected and detected. Results: The results of the in silico analysis ascertained EPHA3 to be a candidate gene of endometriosis. After successful modeling, the uterine tissues of endometriosis mice presented with a low expression of EPHA3 and activated mTOR signaling pathway. Overexpression of EPHA3 inhibited the activation of the mTOR signaling pathway, down-regulated bcl-2 expression, up-regulated the expression of Atg3, LC3-II/LC3-I, Beclin1, bax and fas, and also promoted the autophagy and apoptosis of macrophages in endometriosis mice. Conclusion: Altogether, EPHA3 could potentially promote the autophagy and apoptosis of macrophages in endometriosis via inhibition of the mTOR signaling pathway, highlighting the potential of EPHA3 as the target to treat endometriosis.
Collapse
|
6
|
Sun Y, Wang S, Yang H, Wu J, Li S, Qiao G, Wang S, Wang X, Zhou X, Osada T, Hobeika A, Morse MA, Ren J, Lyerly HK. Impact of synchronized anti-PD-1 with Ad-CEA vaccination on inhibition of colon cancer growth. Immunotherapy 2019; 11:953-966. [PMID: 31192764 DOI: 10.2217/imt-2019-0055] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Aim: The purpose of this study was to determine whether addition of anti-PD-1 antibody increased the immunogenicity and anti-tumor activity of Ad-CEA vaccination in a murine model of colon cancer. Methods: Ad-CEA was administered prior to implantation of MC-38-CEA cells followed by administration of anti-PD-1 antibody. CEA-specific T-cell responses were measured by flow cytometry and ELISPOT. Dynamic co-culture of splenocytes with tumor cells was conducted to analyze anti-tumor activities. Tumor infiltration by lymphocytes was measured by IHC. Tumor volume and overall survival were also recorded. Results: Ad-CEA combined with anti-PD-1 antibody showed greater anti-tumor activity compared with either alone. The combination also increased T-cell infiltration but decreased Tregs. Conclusion: Combining Ad-CEA vaccination with anti-PD-1 antibody enhanced anti-tumor activity and immune responses.
Collapse
Affiliation(s)
- Yuanyuan Sun
- Department of Medical Oncology, Beijing Key Laboratory for Therapeutic Cancer Vaccines, Capital Medical University Cancer Center, Beijing Shijitan Hospital, Capital Medical University, Beijing, 100038, China
| | - Suya Wang
- Department of Medical Oncology, Beijing Key Laboratory for Therapeutic Cancer Vaccines, Capital Medical University Cancer Center, Beijing Shijitan Hospital, Capital Medical University, Beijing, 100038, China
| | - Hainan Yang
- Department of Medical Oncology, Beijing Key Laboratory for Therapeutic Cancer Vaccines, Capital Medical University Cancer Center, Beijing Shijitan Hospital, Capital Medical University, Beijing, 100038, China
| | - Jiangping Wu
- Department of Medical Oncology, Beijing Key Laboratory for Therapeutic Cancer Vaccines, Capital Medical University Cancer Center, Beijing Shijitan Hospital, Capital Medical University, Beijing, 100038, China
| | - Sha Li
- Department of Medical Oncology, Beijing Key Laboratory for Therapeutic Cancer Vaccines, Capital Medical University Cancer Center, Beijing Shijitan Hospital, Capital Medical University, Beijing, 100038, China
| | - Guoliang Qiao
- Department of Medical Oncology, Beijing Key Laboratory for Therapeutic Cancer Vaccines, Capital Medical University Cancer Center, Beijing Shijitan Hospital, Capital Medical University, Beijing, 100038, China
| | - Shuo Wang
- Department of Medical Oncology, Beijing Key Laboratory for Therapeutic Cancer Vaccines, Capital Medical University Cancer Center, Beijing Shijitan Hospital, Capital Medical University, Beijing, 100038, China
| | - Xiaoli Wang
- Department of Medical Oncology, Beijing Key Laboratory for Therapeutic Cancer Vaccines, Capital Medical University Cancer Center, Beijing Shijitan Hospital, Capital Medical University, Beijing, 100038, China
| | - Xinna Zhou
- Department of Medical Oncology, Beijing Key Laboratory for Therapeutic Cancer Vaccines, Capital Medical University Cancer Center, Beijing Shijitan Hospital, Capital Medical University, Beijing, 100038, China
| | - Takuya Osada
- Department of Surgery, Duke University Medical Center, Durham, NC 27710, USA
| | - Amy Hobeika
- Department of Surgery, Duke University Medical Center, Durham, NC 27710, USA
| | - Michael A Morse
- Department of Medicine, Duke University Medical Center, Durham, NC 27710, USA.,Department of Surgery, Duke University Medical Center, Durham, NC 27710, USA
| | - Jun Ren
- Department of Medical Oncology, Beijing Key Laboratory for Therapeutic Cancer Vaccines, Capital Medical University Cancer Center, Beijing Shijitan Hospital, Capital Medical University, Beijing, 100038, China.,Department of Surgery, Duke University Medical Center, Durham, NC 27710, USA
| | - Herbert Kim Lyerly
- Department of Surgery, Duke University Medical Center, Durham, NC 27710, USA
| |
Collapse
|
7
|
Hong Z, Mao X, You J, Liu Z, Shi Y. An Evaluation of HER2-Positive Ovarian Carcinoma Xenografts: From a Novel Therapy to a Noninvasive Monitoring Method. Cancer Biother Radiopharm 2018; 33:411-419. [PMID: 30052070 DOI: 10.1089/cbr.2018.2516] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Affiliation(s)
- Zhihui Hong
- Department of Nuclear Medicine, The Second Affiliated Hospital of Soochow University, Suzhou City, People's Republic of China
| | - Xinping Mao
- Division of Medical Imageology, GanSu University of Chinese Medicine, Lanzhou City, People's Republic of China
| | - Jiaxi You
- Department of Nuclear Medicine, The Second Affiliated Hospital of Soochow University, Suzhou City, People's Republic of China
| | - Zengli Liu
- Department of Nuclear Medicine, The Second Affiliated Hospital of Soochow University, Suzhou City, People's Republic of China
| | - Yizhen Shi
- Department of Nuclear Medicine, The Second Affiliated Hospital of Soochow University, Suzhou City, People's Republic of China
| |
Collapse
|
8
|
Zhang J, Ding M, Xu K, Mao L, Zheng J. shRNA-armed conditionally replicative adenoviruses: a promising approach for cancer therapy. Oncotarget 2018; 7:29824-34. [PMID: 26980708 PMCID: PMC5045436 DOI: 10.18632/oncotarget.8035] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2015] [Accepted: 02/15/2016] [Indexed: 12/16/2022] Open
Abstract
The small-interfering RNAs (siRNAs) have been employed to knockdown the expression of cancer-associated genes and shown some promise in cancer therapy. However, synthetic siRNA duplexes or plasmid mediated delivery of siRNAs have several problems, such as short half-life, low transfection efficiency and cytotoxicity associated with transfection. Conditionally replicating adenovirus (CRAds) as the delivery vector for short hairpin RNAs (shRNAs) could overcome these limitations and have shown augmented anti-tumor effects in experimental studies and preclinical trials. In this review, we summarize recent progress in the development of CRAds-shRNA for cancer treatment. Combination of CRAds-shRNA with chemotherapeutics, radiation, dendritic cells, monoclonal antibodies and small-molecule inhibitors will be necessary to eradicate cancer cells and cancer stem cells and achieve superior outcomes. The use of CRAd platform for efficient delivery of shRNAs and foreign genes will open a new avenue for cancer therapy.
Collapse
Affiliation(s)
- Jie Zhang
- Jiangsu Key Laboratory of Biological Cancer Therapy, Xuzhou Medical College, Xuzhou, China
| | - Meng Ding
- Jiangsu Key Laboratory of Biological Cancer Therapy, Xuzhou Medical College, Xuzhou, China
| | - Kai Xu
- Jiangsu Key Laboratory of Biological Cancer Therapy, Xuzhou Medical College, Xuzhou, China
| | - Lijun Mao
- Jiangsu Key Laboratory of Biological Cancer Therapy, Xuzhou Medical College, Xuzhou, China.,Department of Urinary Surgery, The Affiliated Hospital of Xuzhou Medical College, Xuzhou, China
| | - Junian Zheng
- Jiangsu Key Laboratory of Biological Cancer Therapy, Xuzhou Medical College, Xuzhou, China
| |
Collapse
|
9
|
Howells A, Marelli G, Lemoine NR, Wang Y. Oncolytic Viruses-Interaction of Virus and Tumor Cells in the Battle to Eliminate Cancer. Front Oncol 2017; 7:195. [PMID: 28944214 PMCID: PMC5596080 DOI: 10.3389/fonc.2017.00195] [Citation(s) in RCA: 94] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2017] [Accepted: 08/15/2017] [Indexed: 12/13/2022] Open
Abstract
Oncolytic viruses (OVs) are an emerging treatment option for many cancer types and have recently been the focus of extensive research aiming to develop their therapeutic potential. The ultimate aim is to design a virus which can effectively replicate within the host, specifically target and lyse tumor cells and induce robust, long lasting tumor-specific immunity. There are a number of viruses which are either naturally tumor-selective or can be modified to specifically target and eliminate tumor cells. This means they are able to infect only tumor cells and healthy tissue remains unharmed. This specificity is imperative in order to reduce the side effects of oncolytic virotherapy. These viruses can also be modified by various methods including insertion and deletion of specific genes with the aim of improving their efficacy and safety profiles. In this review, we have provided an overview of the various virus species currently being investigated for their oncolytic potential and the positive and negative effects of a multitude of modifications used to increase their infectivity, anti-tumor immunity, and treatment safety, in particular focusing on the interaction of tumor cells and OVs.
Collapse
Affiliation(s)
- Anwen Howells
- Centre for Molecular Oncology, Barts Cancer Institute, Queen Mary University of London, London, United Kingdom
| | - Giulia Marelli
- Centre for Molecular Oncology, Barts Cancer Institute, Queen Mary University of London, London, United Kingdom
| | - Nicholas R Lemoine
- Centre for Molecular Oncology, Barts Cancer Institute, Queen Mary University of London, London, United Kingdom.,National Centre for International Research in Cell and Gene Therapy, Sino-British Research Centre for Molecular Oncology, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Yaohe Wang
- Centre for Molecular Oncology, Barts Cancer Institute, Queen Mary University of London, London, United Kingdom.,National Centre for International Research in Cell and Gene Therapy, Sino-British Research Centre for Molecular Oncology, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, China
| |
Collapse
|
10
|
Liu Y, Zheng Y. Bach1 siRNA attenuates bleomycin-induced pulmonary fibrosis by modulating oxidative stress in mice. Int J Mol Med 2016; 39:91-100. [PMID: 27959382 PMCID: PMC5179191 DOI: 10.3892/ijmm.2016.2823] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2015] [Accepted: 12/02/2016] [Indexed: 01/08/2023] Open
Abstract
Oxidative stress plays an essential role in inflammation and fibrosis. Bach1 is an important transcriptional repressor that acts by modulating oxidative stress and represents a potential target in the treatment of pulmonary fibrosis (PF). In this study, we knocked down Bach1 using adenovirus-mediated small interfering RNA (siRNA) to determine whether the use of Bach1 siRNA is an effective therapeutic strategy in mice with bleomycin (BLM)‑induced PF. Mouse lung fibroblasts (MLFs) were incubated with transforming growth factor (TGF)-β1 (5 ng/ml) and subsequently infected with recombined adenovirus-like Bach1 siRNA1 and Bach1 siRNA2, while an empty adenovirus vector was used as the negative control. The selected Bach1 siRNA with higher interference efficiency was used for the animal experiments. A mouse model of BLM-induced PF was established, and Bach1 siRNA (1x109 pfu) was administered to the mice via the tail vein. The results revealed that the Bach1 mRNA and protein levels were significantly downregulated by Bach1 siRNA. Furthermore, the MLFs infected with Bach1 siRNA exhibited increased mRNA and protein expression levels of heme oxygenase-1 and glutathione peroxidase 1, but decreased levels of TGF-β1 and interleukin-6 in the cell supernatants compared with the cells exposed to TGF-β1 alone. Bach1 knockdown by siRNA also enhanced the expression of antioxidant factors, but suppressed that of fibrosis‑related cytokines in mice compared with the BLM group. Finally, the inflammatory infiltration of alveolar and interstitial cells and the destruction of lung structure were significantly attenuated in the mide administered Bach1 siRNA compared with those in the BLM group. On the whole, our findings demonstrate that Bach1 siRNA exerts protective effects against BLM-induced PF in mice. Our data may provide the basis for the development of novel targeted therapeutic strategies for PF.
Collapse
Affiliation(s)
- Yuan Liu
- Department of Rheumatology and Immunology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing 100069, P.R. China
| | - Yi Zheng
- Department of Rheumatology and Immunology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing 100069, P.R. China
| |
Collapse
|
11
|
Malanga D, De Marco C, Guerriero I, Colelli F, Rinaldo N, Scrima M, Mirante T, De Vitis C, Zoppoli P, Ceccarelli M, Riccardi M, Ravo M, Weisz A, Federico A, Franco R, Rocco G, Mancini R, Rizzuto A, Gulletta E, Ciliberto G, Viglietto G. The Akt1/IL-6/STAT3 pathway regulates growth of lung tumor initiating cells. Oncotarget 2016; 6:42667-86. [PMID: 26486080 PMCID: PMC4767462 DOI: 10.18632/oncotarget.5626] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2015] [Accepted: 09/09/2015] [Indexed: 01/13/2023] Open
Abstract
Here we report that the PI3K/Akt1/IL-6/STAT3 signalling pathway regulates generation and stem cell-like properties of Non-Small Cell Lung Cancer (NSCLC) tumor initiating cells (TICs). Mutant Akt1, mutant PIK3CA or PTEN loss enhances formation of lung cancer spheroids (LCS), self-renewal, expression of stemness markers and tumorigenic potential of human immortalized bronchial cells (BEAS-2B) whereas Akt inhibition suppresses these activities in established (NCI-H460) and primary NSCLC cells. Matched microarray analysis of Akt1-interfered cells and LCSs identified IL-6 as a critical target of Akt signalling in NSCLC TICs. Accordingly, suppression of Akt in NSCLC cells decreases IL-6 levels, phosphorylation of IkK and IkB, NF-kB transcriptional activity, phosphorylation and transcriptional activity of STAT3 whereas active Akt1 up-regulates them. Exposure of LCSs isolated from NSCLC cells to blocking anti-IL-6 mAbs, shRNA to IL-6 receptor or to STAT3 markedly reduces the capability to generate LCSs, to self-renew and to form tumors, whereas administration of IL-6 to Akt-interfered cells restores the capability to generate LCSs. Finally, immunohistochemical studies in NSCLC patients demonstrated a positive correlative trend between activated Akt, IL-6 expression and STAT3 phosphorylation (n = 94; p < 0.05). In conclusion, our data indicate that aberrant Akt signalling contributes to maintaining stemness in lung cancer TICs through a NF-kB/IL-6/STAT3 pathway and provide novel potential therapeutic targets for eliminating these malignant cells in NSCLC.
Collapse
Affiliation(s)
- Donatella Malanga
- Dipartimento di Medicina Sperimentale e Clinica, Università Magna Graecia, Catanzaro, Italy
| | - Carmela De Marco
- Dipartimento di Medicina Sperimentale e Clinica, Università Magna Graecia, Catanzaro, Italy.,Biogem scarl, Istituto di Ricerche Genetiche, Ariano Irpino (Avellino), Italy
| | - Ilaria Guerriero
- Biogem scarl, Istituto di Ricerche Genetiche, Ariano Irpino (Avellino), Italy
| | - Fabiana Colelli
- Biogem scarl, Istituto di Ricerche Genetiche, Ariano Irpino (Avellino), Italy
| | - Nicola Rinaldo
- Biogem scarl, Istituto di Ricerche Genetiche, Ariano Irpino (Avellino), Italy
| | - Marianna Scrima
- Dipartimento di Medicina Sperimentale e Clinica, Università Magna Graecia, Catanzaro, Italy.,Biogem scarl, Istituto di Ricerche Genetiche, Ariano Irpino (Avellino), Italy
| | - Teresa Mirante
- Dipartimento di Scienze Mediche e Chirurgiche, Università Magna Graecia, Catanzaro, Italy
| | - Claudia De Vitis
- IRCCS Istituto Nazionale Tumori Fondazione G. Pascale, Napoli, Italy
| | - Pietro Zoppoli
- Biogem scarl, Istituto di Ricerche Genetiche, Ariano Irpino (Avellino), Italy
| | - Michele Ceccarelli
- Dipartimento di Medicina Sperimentale e Clinica, Università Magna Graecia, Catanzaro, Italy.,Dipartimento di Scienze e Tecnologie, Università del Sannio, Benevento, Italy
| | - Miriam Riccardi
- Dipartimento di Medicina Sperimentale e Clinica, Università Magna Graecia, Catanzaro, Italy.,Biogem scarl, Istituto di Ricerche Genetiche, Ariano Irpino (Avellino), Italy
| | - Maria Ravo
- Dipartimento di Medicina e Chirurgia, Università di Salerno, Baronissi, Italy
| | - Alessandro Weisz
- Dipartimento di Medicina e Chirurgia, Università di Salerno, Baronissi, Italy
| | - Antonella Federico
- Dipartimento di Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università Federico II, Napoli, Italy
| | - Renato Franco
- IRCCS Istituto Nazionale Tumori Fondazione G. Pascale, Napoli, Italy
| | - Gaetano Rocco
- IRCCS Istituto Nazionale Tumori Fondazione G. Pascale, Napoli, Italy
| | - Rita Mancini
- Dipartimento di Medicina Clinica e Molecolare, Università di Roma "La Sapienza" Ospedale S. Andrea, Roma, Italy
| | - Antonia Rizzuto
- Dipartimento di Scienze Mediche e Chirurgiche, Università Magna Graecia, Catanzaro, Italy
| | - Elio Gulletta
- Dipartimento di Scienze della Salute, Università Magna Graecia, Catanzaro, Italy
| | - Gennaro Ciliberto
- IRCCS Istituto Nazionale Tumori Fondazione G. Pascale, Napoli, Italy
| | - Giuseppe Viglietto
- Dipartimento di Medicina Sperimentale e Clinica, Università Magna Graecia, Catanzaro, Italy.,Biogem scarl, Istituto di Ricerche Genetiche, Ariano Irpino (Avellino), Italy
| |
Collapse
|
12
|
Nascimento AV, Gattacceca F, Singh A, Bousbaa H, Ferreira D, Sarmento B, Amiji MM. Biodistribution and pharmacokinetics of Mad2 siRNA-loaded EGFR-targeted chitosan nanoparticles in cisplatin sensitive and resistant lung cancer models. Nanomedicine (Lond) 2016; 11:767-81. [PMID: 26980454 PMCID: PMC4910968 DOI: 10.2217/nnm.16.14] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2015] [Accepted: 01/26/2016] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND The present study focuses on biodistribution profile and pharmacokinetic parameters of EGFR-targeted chitosan nanoparticles (TG CS nanoparticles) for siRNA/cisplatin combination therapy of lung cancer. MATERIAL & METHODS Mad2 siRNA was encapsulated in EGFR targeted and nontargeted (NTG) CS nanoparticles by electrostatic interaction. The biodistribution of the nanoparticles was assessed qualitatively and quantitatively in cisplatin (DDP) sensitive and resistant lung cancer xenograft model. RESULTS TG nanoparticles showed a consistent and preferential tumor targeting ability with rapid clearance from the plasma to infiltrate and sustain within the tumor up to 96 h. They exhibit a sixfold higher tumor targeting efficiency compared with the NTG nanoparticles. CONCLUSION TG nanoparticles present as an attractive drug delivery platform for RNAi therapeutics against NSCLC.
Collapse
Affiliation(s)
- Ana Vanessa Nascimento
- CESPU, Instituto de Investigação e Formação Avançada em Ciências e Tecnologias da Saúde, Gandra, Portugal
- Laboratory of Pharmaceutical Technology, Faculty of Pharmacy, University of Porto, Portugal
- Department of Pharmaceutical Sciences, School of Pharmacy, Bouvé College of Health Sciences, Northeastern University, Boston, MA, USA
| | - Florence Gattacceca
- Institut de Recherche en Cancérologie de Montpellier IRCM, INSERM U1194, ICM, Université de Montpellier, Montpellier, France
| | - Amit Singh
- Department of Pharmaceutical Sciences, School of Pharmacy, Bouvé College of Health Sciences, Northeastern University, Boston, MA, USA
| | - Hassan Bousbaa
- CESPU, Instituto de Investigação e Formação Avançada em Ciências e Tecnologias da Saúde, Gandra, Portugal
- Centro Interdisciplinar de Investigação Marinha e Ambiental (CIIMAR/CIMAR), Universidade do Porto, Portugal
| | - Domingos Ferreira
- Laboratory of Pharmaceutical Technology, Faculty of Pharmacy, University of Porto, Portugal
| | - Bruno Sarmento
- CESPU, Instituto de Investigação e Formação Avançada em Ciências e Tecnologias da Saúde, Gandra, Portugal
- I3S, Instituto de Investigação e Inovação em Saúde and INEB – Instituto de Engenharia Biomédica, Universidade do Porto, Portugal
| | - Mansoor M Amiji
- Department of Pharmaceutical Sciences, School of Pharmacy, Bouvé College of Health Sciences, Northeastern University, Boston, MA, USA
- Faculty of Pharmacy, King Abdulaziz University, Jeddah, Saudi Arabia
| |
Collapse
|
13
|
Shi J, Sun J, Wu M, Hu N, Hu Y. Hepatitis A virus-encoded miRNAs attenuate the accumulation of viral genomic RNAs in infected cells. Virus Genes 2016; 52:317-24. [DOI: 10.1007/s11262-016-1306-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2016] [Accepted: 02/16/2016] [Indexed: 11/30/2022]
|