1
|
Elliott WJ, Gurramkonda N, Guda MR, Tsung AJ, Velpula KK. Survivin Interference and SurVaxM as an Adjunct Therapy for Glioblastoma Multiforme. Cells 2025; 14:755. [PMID: 40422257 DOI: 10.3390/cells14100755] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2025] [Revised: 04/15/2025] [Accepted: 04/18/2025] [Indexed: 05/28/2025] Open
Abstract
Glioblastoma, IDH wild-type WHO Grade IV, is a devastating diagnosis in pediatric and adult populations with a poor prognosis and median overall survival of less than two years. Despite the advent of the Stupp protocol and advances in neurosurgical tumor resection techniques, there has been minimal change to both the quantity and quality of life in individuals diagnosed. Provided the extensive research on survivin's association with glioblastoma tumor microenvironment, this review suggests that priming the individual's immune systems to the tumor-promoting protein may reduce tumor burden through multiple mechanisms, including the arrest of the G2/M phase, microtubule dysfunction, induction of autophagy, and ultimately activation of apoptosis in glioblastoma cells. SurVaxM, a multiple peptide, survivin-specific vaccine, may assist in tumor cell destruction by eliciting the production of cytotoxic T-cells specific to survivin-expression glioblastoma tumors. Although phase I and II clinical trials suggest relatively safe adverse effects and potential efficacy, additional research is necessary to evaluate further how this vaccine may compare to standard treatment.
Collapse
Affiliation(s)
- Willie James Elliott
- Department of Cancer Biology and Pharmacology, University of Illinois College of Medicine Peoria, Peoria, IL 61605, USA
| | - Nandini Gurramkonda
- Department of Cancer Biology and Pharmacology, University of Illinois College of Medicine Peoria, Peoria, IL 61605, USA
| | - Maheedhara R Guda
- Department of Cancer Biology and Pharmacology, University of Illinois College of Medicine Peoria, Peoria, IL 61605, USA
| | - Andrew J Tsung
- Department of Cancer Biology and Pharmacology, University of Illinois College of Medicine Peoria, Peoria, IL 61605, USA
- Department of Neurosurgery, University of Illinois College of Medicine Peoria, Peoria, IL 61605, USA
- Illinois Neurological Institute, Peoria, IL 61605, USA
| | - Kiran K Velpula
- Department of Cancer Biology and Pharmacology, University of Illinois College of Medicine Peoria, Peoria, IL 61605, USA
- Department of Neurosurgery, University of Illinois College of Medicine Peoria, Peoria, IL 61605, USA
- Department of Pediatrics, University of Illinois College of Medicine Peoria, Peoria, IL 61605, USA
| |
Collapse
|
2
|
Palomer X, Salvador JM, Griñán-Ferré C, Barroso E, Pallàs M, Vázquez-Carrera M. GADD45A: With or without you. Med Res Rev 2024; 44:1375-1403. [PMID: 38264852 DOI: 10.1002/med.22015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Revised: 12/11/2023] [Accepted: 01/09/2024] [Indexed: 01/25/2024]
Abstract
The growth arrest and DNA damage inducible (GADD)45 family includes three small and ubiquitously distributed proteins (GADD45A, GADD45B, and GADD45G) that regulate numerous cellular processes associated with stress signaling and injury response. Here, we provide a comprehensive review of the current literature investigating GADD45A, the first discovered member of the family. We first depict how its levels are regulated by a myriad of genotoxic and non-genotoxic stressors, and through the combined action of intricate transcriptional, posttranscriptional, and even, posttranslational mechanisms. GADD45A is a recognized tumor suppressor and, for this reason, we next summarize its role in cancer, as well as the different mechanisms by which it regulates cell cycle, DNA repair, and apoptosis. Beyond these most well-known actions, GADD45A may also influence catabolic and anabolic pathways in the liver, adipose tissue and skeletal muscle, among others. Not surprisingly, GADD45A may trigger AMP-activated protein kinase activity, a master regulator of metabolism, and is known to act as a transcriptional coregulator of numerous nuclear receptors. GADD45A has also been reported to display a cytoprotective role by regulating inflammation, fibrosis and oxidative stress in several organs and tissues, and is regarded an important contributor for the development of heart failure. Overall data point to that GADD45A may play an important role in metabolic, neurodegenerative and cardiovascular diseases, and also autoimmune-related disorders. Thus, the potential mechanisms by which dysregulation of GADD45A activity may contribute to the progression of these diseases are also reviewed below.
Collapse
Affiliation(s)
- Xavier Palomer
- Department of Pharmacology, Toxicology and Therapeutic Chemistry, Faculty of Pharmacy and Food Sciences, University of Barcelona, Barcelona, Spain
- Institute of Biomedicine of the University of Barcelona (IBUB), University of Barcelona, Barcelona, Spain
- Spanish Biomedical Research Center in Diabetes and Associated Metabolic Diseases (CIBERDEM)-Instituto de Salud Carlos III, Madrid, Spain
- Pediatric Research Institute-Hospital Sant Joan de Déu, Esplugues de Llobregat, Barcelona, Spain
| | - Jesús M Salvador
- Department of Immunology and Oncology, Centro Nacional de Biotecnología/CSIC, Madrid, Spain
| | - Christian Griñán-Ferré
- Department of Pharmacology, Toxicology and Therapeutic Chemistry, Faculty of Pharmacy and Food Sciences, University of Barcelona, Barcelona, Spain
- Institut de Neurociències, Universitat de Barcelona (NeuroUB), Barcelona, Spain
- Spanish Biomedical Research Center in Neurodegenerative Diseases (CIBERNED)-Instituto de Salud Carlos III, Madrid, Spain
| | - Emma Barroso
- Department of Pharmacology, Toxicology and Therapeutic Chemistry, Faculty of Pharmacy and Food Sciences, University of Barcelona, Barcelona, Spain
- Institute of Biomedicine of the University of Barcelona (IBUB), University of Barcelona, Barcelona, Spain
- Spanish Biomedical Research Center in Diabetes and Associated Metabolic Diseases (CIBERDEM)-Instituto de Salud Carlos III, Madrid, Spain
- Pediatric Research Institute-Hospital Sant Joan de Déu, Esplugues de Llobregat, Barcelona, Spain
| | - Mercè Pallàs
- Department of Pharmacology, Toxicology and Therapeutic Chemistry, Faculty of Pharmacy and Food Sciences, University of Barcelona, Barcelona, Spain
- Institut de Neurociències, Universitat de Barcelona (NeuroUB), Barcelona, Spain
- Spanish Biomedical Research Center in Neurodegenerative Diseases (CIBERNED)-Instituto de Salud Carlos III, Madrid, Spain
| | - Manuel Vázquez-Carrera
- Department of Pharmacology, Toxicology and Therapeutic Chemistry, Faculty of Pharmacy and Food Sciences, University of Barcelona, Barcelona, Spain
- Institute of Biomedicine of the University of Barcelona (IBUB), University of Barcelona, Barcelona, Spain
- Spanish Biomedical Research Center in Diabetes and Associated Metabolic Diseases (CIBERDEM)-Instituto de Salud Carlos III, Madrid, Spain
- Pediatric Research Institute-Hospital Sant Joan de Déu, Esplugues de Llobregat, Barcelona, Spain
| |
Collapse
|
3
|
Patel K, Murray MG, Whelan KA. Roles for GADD45 in Development and Cancer. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2022; 1360:23-39. [DOI: 10.1007/978-3-030-94804-7_2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
|
4
|
Peng J, Wu A, Yu X, Zhong Q, Deng X, Zhu Y. Combined Network Pharmacology and Cytology Experiments to Identify Potential Anti-Breast Cancer Targets and Mechanisms of Delphinidin. Nutr Cancer 2021; 74:2591-2606. [PMID: 34875956 DOI: 10.1080/01635581.2021.2012582] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Delphinidin is a type of anthocyanin monomer with antioxidant, anti-inflammatory, and anti-tumor effects. However, the biological mechanisms underlying its anti-breast cancer activity have not been thoroughly studied. We further studied the effect of delphinidin on breast cancer cells through comprehensive network pharmacology, cellular and molecular experiments. We acquired the know therapeutic targets of delphinidin and obtained differentially expressed genes (DEGs) of breast cancer using RTCGA. We used topological analysis to screen out the 106 core targets of delphinium anti-breast cancer and performed functional analysis. These genes were mainly enriched in the pathways in cancer, Progesterone-mediated oocyte maturation and cell cycle. Then, by taking the intersection of the three analyzed data sets, important core targets (EGFR, TOP2A and PTGS2) were obtained and molecular-docking was performed to validate the result. Additionally, In Vitro experiments, MCF-7 and BT-474 cell proliferation was inhibited in a dose-dependent manner by delphinidin and the expressions of EGFR, TOP2A and PTGS were reduced. Moreover, delphinidin influenced cell cycle, the expressions of cdk1 and cyclin B1 were reduced. Furthermore, delphinidin induced apoptosis by activating the MAPK-Signaling pathway. Collectively, our findings suggested that delphinidin may offer effective approaches in breast cancer prevention and therapy.Supplemental data for this article is available online at http://dx.doi.org/10.1080/01635581.2021.2012582.
Collapse
Affiliation(s)
- Jiayuan Peng
- Department of Pathology, Leshan People's Hospital, Leshan, China.,Department of Basic Medical Science, Chengdu Medical College, Chengdu, China
| | - Ailin Wu
- Department of Basic Medical Science, Chengdu Medical College, Chengdu, China
| | - Xiaoping Yu
- Department of Public Health, Chengdu Medical College, Chengdu, China
| | - Qian Zhong
- Department of Pharmacy, Chengdu Medical College, Chengdu, China
| | - Xiaoming Deng
- Department of Basic Medical Science, Chengdu Medical College, Chengdu, China
| | - Yanfeng Zhu
- Department of Public Health, Chengdu Medical College, Chengdu, China
| |
Collapse
|
5
|
Wu A, Zhu Y, Han B, Peng J, Deng X, Chen W, Du J, Ou Y, Peng X, Yu X. Delphinidin induces cell cycle arrest and apoptosis in HER-2 positive breast cancer cell lines by regulating the NF-κB and MAPK signaling pathways. Oncol Lett 2021; 22:832. [PMID: 34712357 PMCID: PMC8548810 DOI: 10.3892/ol.2021.13093] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2020] [Accepted: 06/25/2021] [Indexed: 12/23/2022] Open
Abstract
Delphinidin is an anthocyanidin monomer, commonly found in vegetables and fruits, and has demonstrated antitumor effects in the HER-2-positive MDA-MB-453 breast cancer cell line, with low cytotoxicity on normal breast cells. However, the direct functional mechanisms underlying the effect of delphinidin on HER-2-positive breast cancer cells has not been fully characterized. In the present study, it was found that delphinidin could induce G2/M phase cell cycle arrest by inhibiting the protein expression level of cyclin B1 and Cdk1 in HER-2-positive breast cancer cell lines. In addition, delphinidin promoted the mitochondrial apoptosis pathway by inhibiting the ERK and NF-κB signaling pathway and activating the JNK signaling pathway. Therefore, delphinidin markedly suppressed the viability of the HER-2-positive breast cancer cell lines by modulating the cell cycle and inducing apoptosis. Overall, the findings from the present study demonstrated that delphinidin treatment could induce the mitochondrial apoptosis pathway in human HER-2-positive breast cancer cell lines, providing an experimental basis for the prevention and treatment of HER-2-positive breast cancer by flavonoids.
Collapse
Affiliation(s)
- Ailin Wu
- Basic Medical School, Chengdu Medical College, Chengdu, Sichuan 610500, P.R. China.,Ministry of Science and Technology, The Second Affiliated Hospital of Chengdu Medical College, China National Nuclear Corporation 416 Hospital, Chengdu, Sichuan 610051, P.R. China
| | - Yanfeng Zhu
- Graduate School, Chengdu Medical College, Chengdu, Sichuan 610500, P.R. China
| | - Bin Han
- School of Public Health, Chengdu Medical College, Chengdu, Sichuan 610500, P.R. China
| | - Jiayuan Peng
- Basic Medical School, Chengdu Medical College, Chengdu, Sichuan 610500, P.R. China
| | - Xiaomin Deng
- Basic Medical School, Chengdu Medical College, Chengdu, Sichuan 610500, P.R. China
| | - Wei Chen
- Basic Medical School, Chengdu Medical College, Chengdu, Sichuan 610500, P.R. China
| | - Jingchang Du
- School of Public Health, Chengdu Medical College, Chengdu, Sichuan 610500, P.R. China
| | - Yu Ou
- Division of Planning and Finance, Chengdu Medical College, Chengdu, Sichuan 610500, P.R. China
| | - Xiaoli Peng
- School of Public Health, Chengdu Medical College, Chengdu, Sichuan 610500, P.R. China
| | - Xiaoping Yu
- Graduate School, Chengdu Medical College, Chengdu, Sichuan 610500, P.R. China
| |
Collapse
|
6
|
Addison R, Weatherhead SC, Pawitri A, Smith GR, Rider A, Grantham HJ, Cockell SJ, Reynolds NJ. Therapeutic wavelengths of ultraviolet B radiation activate apoptotic, circadian rhythm, redox signalling and key canonical pathways in psoriatic epidermis. Redox Biol 2021; 41:101924. [PMID: 33812333 PMCID: PMC8050411 DOI: 10.1016/j.redox.2021.101924] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Revised: 02/22/2021] [Accepted: 02/26/2021] [Indexed: 01/09/2023] Open
Abstract
Ultraviolet B radiation (UVB) exerts pleiotropic effects on human skin. DNA damage response and repair pathways are activated by UVB; if damage cannot be repaired, apoptosis ensues. Although cumulative UVB exposure predisposes to skin cancer, UVB phototherapy is widely used as an effective treatment for psoriasis. Previous studies defined the therapeutic action spectrum of UVB and showed that psoriasis is resistant to apoptosis. This study aimed to investigate early molecular responses within psoriasis plaques following irradiation with single equi-erythemogenic doses of clinically-effective (311 nm, narrow-band) compared to clinically-ineffective (290 nm) UVB. Forty-eight micro-dissected epidermal samples from 20 psoriatic patients were analyzed using microarrays. Our bioinformatic analysis compared gene expression between 311 nm irradiated, 290 nm irradiated and control psoriasis epidermis to specifically identify 311 nm UVB differentially expressed genes (DEGs) and their upstream regulatory pathways. Key DEGs and pathways were validated by immunohistochemical analysis. There was a dynamic induction and repression of 311 nm UVB DEGs between 6 h and 18 h, only a limited number of DEGs maintained their designated expression status between time-points. Key disease and function pathways included apoptosis, cell death, cell migration and leucocyte chemotaxis. DNA damage response pathways, NRF2-mediated oxidative stress response and P53 signalling were key nodes, interconnecting apoptosis and cell cycle arrest. Interferon signalling, dendritic cell maturation, granulocyte adhesion and atherosclerotic pathways were also differentially regulated. Consistent with these findings, top transcriptional regulators of 311 nm UVB DEGs related to: a) apoptosis, DNA damage response and cell cycle control; b) innate/acquired immune regulation and inflammation; c) hypoxia/redox response and angiogenesis; d) circadian rhythmicity; f) EGR/AP1 signalling and keratinocyte differentiation; and g) mitochondrial biogenesis. This research provides important insights into the molecular targets of 311 nm UVB, underscoring key roles for apoptosis and cell death. These and the other key pathways delineated may be central to the therapeutic effects of 311 nm in psoriasis.
Collapse
Affiliation(s)
- Rachel Addison
- Institute of Translational and Clinical Medicine, Faculty of Medical Sciences, Framlington Place, Newcastle University, Newcastle Upon Tyne, UK
| | - Sophie C Weatherhead
- Institute of Translational and Clinical Medicine, Faculty of Medical Sciences, Framlington Place, Newcastle University, Newcastle Upon Tyne, UK; Department of Dermatology, Royal Victoria Infirmary, Newcastle Hospitals NHS Foundation Trust, Newcastle Upon Tyne, UK
| | - Anandika Pawitri
- Institute of Translational and Clinical Medicine, Faculty of Medical Sciences, Framlington Place, Newcastle University, Newcastle Upon Tyne, UK
| | - Graham R Smith
- Bioinformatics Support Unit, Faculty of Medical Sciences, Framlington Place, Newcastle University, Newcastle Upon Tyne, UK
| | - Ashley Rider
- Institute of Translational and Clinical Medicine, Faculty of Medical Sciences, Framlington Place, Newcastle University, Newcastle Upon Tyne, UK
| | - Henry J Grantham
- Institute of Translational and Clinical Medicine, Faculty of Medical Sciences, Framlington Place, Newcastle University, Newcastle Upon Tyne, UK; Department of Dermatology, Royal Victoria Infirmary, Newcastle Hospitals NHS Foundation Trust, Newcastle Upon Tyne, UK
| | - Simon J Cockell
- Bioinformatics Support Unit, Faculty of Medical Sciences, Framlington Place, Newcastle University, Newcastle Upon Tyne, UK
| | - Nick J Reynolds
- Institute of Translational and Clinical Medicine, Faculty of Medical Sciences, Framlington Place, Newcastle University, Newcastle Upon Tyne, UK; Department of Dermatology, Royal Victoria Infirmary, Newcastle Hospitals NHS Foundation Trust, Newcastle Upon Tyne, UK.
| |
Collapse
|
7
|
Wu K, Chen X, Feng J, Zhang S, Xu Y, Zhang J, Wu Q, You M, Xia B, Ma S. Capilliposide C from Lysimachia capillipes Restores Radiosensitivity in Ionizing Radiation-Resistant Lung Cancer Cells Through Regulation of ERRFI1/EGFR/STAT3 Signaling Pathway. Front Oncol 2021; 11:644117. [PMID: 33869036 PMCID: PMC8047471 DOI: 10.3389/fonc.2021.644117] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2020] [Accepted: 03/12/2021] [Indexed: 12/25/2022] Open
Abstract
Aims Radiation therapy is used as the primary treatment for lung cancer. Unfortunately, radiation resistance remains to be the major clinic problem for lung cancer patients. Lysimachia capillipes capilliposide C (LC-C), an extract from LC Hemsl, has demonstrated multiple anti-cancer effects in several types of cancer. Here, we investigated the potential therapeutic impacts of LC-C on radiosensitivity in lung cancer cells and their underlying mechanisms. Methods Non-small cell lung cancer cell lines were initially irradiated to generate ionizing radiation (IR)-resistant lung cancer cell lines. RNA-seq analysis was used to examine the whole-transcriptome alteration in IR-resistant lung cancer cells treated with or without LC-C, and the differentially expressed genes with most significance were verified by RT-qPCR. Colony formation assays were performed to determine the effect of LC-C and the target gene ErbB receptor feedback inhibitor 1 (ERRFI1) on radiosensitivity of IR-resistant lung cancer cells. In addition, effects of ERRFI1 on cell cycle distribution, DNA damage repair activity were assessed by flow cytometry and γ-H2AX immunofluorescence staining respectively. Western blotting was performed to identify the activation of related signaling pathways. Tumor xenograft experiments were conducted to observe the effect of LC-C and ERRFI1 on radiosensitivity of IR-resistant lung cancer cells in vivo. Results Compared with parental cells, IR-resistant lung cancer cells were more resistant to radiation. LC-C significantly enhanced the effect of radiation in IR-resistant lung cancer cells both in vitro and in vivo and validated ERRFI1 as a candidate downstream gene by RNA-seq. Forced expression of ERRFI1 alone could significantly increase the radiosensitivity of IR-resistant lung cancer cells, while silencing of ERRFI1 attenuated the radiosensitizing function of LC-C. Accordingly, LC-C and ERRFI1 effectively inhibited IR-induced DNA damage repair, and ERRFI1 significantly induced G2/M checkpoint arrest. Additional investigations revealed that down-regulation of EGFR/STAT3 pathway played an important role in radiosensitization between ERRFI1 and LC-C. Furthermore, the high expression level of ERRFI1 was associated with high overall survival rates in lung cancer patients. Conclusions Treatment of LC-C may serve as a promising therapeutic strategy to overcome the radiation resistance and ERRFI1 may be a potential therapeutic target in NSCLC.
Collapse
Affiliation(s)
- Kan Wu
- Department of Thoracic Oncology, Key Laboratory of Clinical Cancer Pharmacology and Toxicology Research of Zhejiang Province, Affiliated Hangzhou Cancer Hospital, Zhejiang University School of Medicine, Zhejiang University Cancer Center, Hangzhou, China
| | - Xueqin Chen
- Department of Thoracic Oncology, Key Laboratory of Clinical Cancer Pharmacology and Toxicology Research of Zhejiang Province, Affiliated Hangzhou Cancer Hospital, Zhejiang University School of Medicine, Zhejiang University Cancer Center, Hangzhou, China.,Department of Thoracic Oncology, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Zhejiang University Cancer Center, Hangzhou, China
| | - Jianguo Feng
- Zhejiang Cancer Research Institute, Cancer Hospital of the University of Chinese Academy of Sciences, Zhejiang Cancer Hospital, Hangzhou, China
| | - Shirong Zhang
- Center for Translational Medicine, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Zhejiang University Cancer Center, Hangzhou, China
| | - Yasi Xu
- Center for Translational Medicine, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Zhejiang University Cancer Center, Hangzhou, China
| | - Jingjing Zhang
- Center for Translational Medicine, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Zhejiang University Cancer Center, Hangzhou, China
| | - Qiong Wu
- Center for Translational Medicine, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Zhejiang University Cancer Center, Hangzhou, China
| | - Mingliang You
- Hangzhou Cancer Institute, Affiliated Hangzhou Cancer Hospital, Zhejiang University School of Medicine, Zhejiang University Cancer Center, Hangzhou, China
| | - Bing Xia
- Department of Thoracic Oncology, Key Laboratory of Clinical Cancer Pharmacology and Toxicology Research of Zhejiang Province, Affiliated Hangzhou Cancer Hospital, Zhejiang University School of Medicine, Zhejiang University Cancer Center, Hangzhou, China.,Department of Oncology, Jiande Second People's Hospital, Hangzhou, China
| | - Shenglin Ma
- Department of Thoracic Oncology, Key Laboratory of Clinical Cancer Pharmacology and Toxicology Research of Zhejiang Province, Affiliated Hangzhou Cancer Hospital, Zhejiang University School of Medicine, Zhejiang University Cancer Center, Hangzhou, China
| |
Collapse
|
8
|
Chander Y, Kumar R, Khandelwal N, Singh N, Shringi BN, Barua S, Kumar N. Role of p38 mitogen-activated protein kinase signalling in virus replication and potential for developing broad spectrum antiviral drugs. Rev Med Virol 2021; 31:1-16. [PMID: 33450133 DOI: 10.1002/rmv.2217] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2020] [Revised: 12/21/2020] [Accepted: 12/23/2020] [Indexed: 12/11/2022]
Abstract
Mitogen-activated protein kinases (MAPKs) play a key role in complex cellular processes such as proliferation, development, differentiation, transformation and apoptosis. Mammals express at least four distinctly regulated groups of MAPKs which include extracellular signal-related kinases (ERK)-1/2, p38 proteins, Jun amino-terminal kinases (JNK1/2/3) and ERK5. p38 MAPK is activated by a wide range of cellular stresses and modulates activity of several downstream kinases and transcription factors which are involved in regulating cytoskeleton remodeling, cell cycle modulation, inflammation, antiviral response and apoptosis. In viral infections, activation of cell signalling pathways is part of the cellular defense mechanism with the basic aim of inducing an antiviral state. However, viruses can exploit enhanced cell signalling activities to support various stages of their replication cycles. Kinase activity can be inhibited by small molecule chemical inhibitors, so one strategy to develop antiviral drugs is to target these cellular signalling pathways. In this review, we provide an overview on the current understanding of various cellular and viral events regulated by the p38 signalling pathway, with a special emphasis on targeting these events for antiviral drug development which might identify candidates with broad spectrum activity.
Collapse
Affiliation(s)
- Yogesh Chander
- National Centre for Veterinary Type Cultures, ICAR-National Research Centre on Equines, Hisar, Haryana, India.,Department of Bio and Nano Technology, Guru Jambeshwar University of Science and Technology, Hisar, Haryana, India
| | - Ram Kumar
- National Centre for Veterinary Type Cultures, ICAR-National Research Centre on Equines, Hisar, Haryana, India.,Department of Veterinary Microbiology and Biotechnology, Rajasthan University of Veterinary and Animal Sciences, Bikaner, India
| | - Nitin Khandelwal
- National Centre for Veterinary Type Cultures, ICAR-National Research Centre on Equines, Hisar, Haryana, India.,Department of Biotechnology, GLA University, Mathura, India
| | - Namita Singh
- Department of Bio and Nano Technology, Guru Jambeshwar University of Science and Technology, Hisar, Haryana, India
| | - Brij Nandan Shringi
- Department of Veterinary Microbiology and Biotechnology, Rajasthan University of Veterinary and Animal Sciences, Bikaner, India
| | - Sanjay Barua
- National Centre for Veterinary Type Cultures, ICAR-National Research Centre on Equines, Hisar, Haryana, India
| | - Naveen Kumar
- National Centre for Veterinary Type Cultures, ICAR-National Research Centre on Equines, Hisar, Haryana, India
| |
Collapse
|
9
|
Liu JY, Fu WQ, Zheng XJ, Li W, Ren LW, Wang JH, Yang C, Du GH. Avasimibe exerts anticancer effects on human glioblastoma cells via inducing cell apoptosis and cell cycle arrest. Acta Pharmacol Sin 2021; 42:97-107. [PMID: 32451414 PMCID: PMC7921416 DOI: 10.1038/s41401-020-0404-8] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/08/2019] [Accepted: 03/19/2020] [Indexed: 01/11/2023]
Abstract
Glioblastoma (GBM) is the most common and lethal primary brain tumor in adults, but there is no effective drug available for GBM. Avasimibe is a potent inhibitor of acyl-coenzyme A: cholesterol acyltransferase-1 (ACAT-1), which was used to treat atherosclerosis. Experimental evidence and bioinformatics have shown that avasimibe has anticancer activity. In this study we investigated the anticancer effects of avasimibe on human glioblastoma cells and the underlying mechanisms. Our results showed that avasimibe dose-dependently inhibited the proliferation of U251 and U87 human glioblastoma cells with IC50 values of 20.29 and 28.27 μM, respectively, at 48 h. Avasimibe (7.5, 15, 30 μM) decreased the DNA synthesis, and inhibited the colony formation of the tumor cells. Treatment of avasimibe also dose-dependently increased the apoptotic rate of tumor cells, decreased the mitochondrial membrane potential, induced the activity of caspase-3/7, and increased the protein expression of cleaved caspase-9, cleaved PARP and Bax in U251 and U87 cells. RNA-sequencing analyses revealed that avasimibe suppressed the expression of CDK2, cyclin E1, CDK4, cyclin D, CDK1, cyclin B1, Aurora A, and PLK1, while induced the expression of p53, p21, p27, and GADD45A, which was validated by Western blot analysis. These results demonstrated that avasimibe induced mitochondria-dependent apoptosis in glioblastoma cells, which was associated with arresting the cell cycle at G0/G1 phase and G2/M phase by regulating the p53/p21 pathway, p53/GADD45A and Aurora A/PLK1 signaling pathways. In U87 xenograft nude mice model, administration of avasimibe (15, 30 mg·kg-1·d-1, ip, for 18 days) dose-dependently inhibit the tumor growth. Taken together, our results demonstrated that avasimibe might be a promising chemotherapy drug in the treatment of GBM.
Collapse
Affiliation(s)
- Jin-Yi Liu
- The State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Beijing, 100050, China
- Key Laboratory of Drug Target Research and Drug Screen, Institute of Materia Medica, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, 100050, China
| | - Wei-Qi Fu
- The State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Beijing, 100050, China
- Key Laboratory of Drug Target Research and Drug Screen, Institute of Materia Medica, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, 100050, China
| | - Xiang-Jin Zheng
- The State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Beijing, 100050, China
- Key Laboratory of Drug Target Research and Drug Screen, Institute of Materia Medica, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, 100050, China
| | - Wan Li
- The State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Beijing, 100050, China
- Key Laboratory of Drug Target Research and Drug Screen, Institute of Materia Medica, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, 100050, China
| | - Li-Wen Ren
- The State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Beijing, 100050, China
- Key Laboratory of Drug Target Research and Drug Screen, Institute of Materia Medica, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, 100050, China
| | - Jin-Hua Wang
- The State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Beijing, 100050, China.
- Key Laboratory of Drug Target Research and Drug Screen, Institute of Materia Medica, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, 100050, China.
| | - Cui Yang
- Ethnic Drug Screening & Pharmacology Center, Key Laboratory of Chemistry in Ethnic Medicinal Resources, State Ethnic Affairs Commission & Ministry of Education, Yunnan Minzu University, Kunming, 650500, China.
| | - Guan-Hua Du
- The State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Beijing, 100050, China.
- Key Laboratory of Drug Target Research and Drug Screen, Institute of Materia Medica, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, 100050, China.
| |
Collapse
|
10
|
Cucurbitacin D Induces G2/M Phase Arrest and Apoptosis via the ROS/p38 Pathway in Capan-1 Pancreatic Cancer Cell Line. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2020; 2020:6571674. [PMID: 33029168 PMCID: PMC7527894 DOI: 10.1155/2020/6571674] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/13/2020] [Revised: 08/18/2020] [Accepted: 09/12/2020] [Indexed: 02/07/2023]
Abstract
Pancreatic cancer has a poor prognosis with a five-year survival rate of less than 10%. Moreover, chemotherapy is mostly rendered ineffective owing to chemotherapy resistance and cytotoxicity. Therefore, the development of effective therapeutic strategies and novel drugs against pancreatic cancer is an urgent need. Cucurbitacin D (CuD), a plant steroid derived from Trichosanthes kirilowii, is an anticancer agent effective against various cancer cell lines. However, the anticancer activity and molecular mechanism of CuD in pancreatic cancer remain unknown. Therefore, we aimed to investigate the anticancer activity and molecular mechanism of CuD in the human pancreatic cancer cell line, Capan-1. CuD induced cell cycle arrest at the G2/M phase, apoptosis, and reactive oxygen species generation in Capan-1 cell line. In addition, CuD induced the activation of the p38 MAPK signaling pathway that regulates apoptosis, which was also inhibited by N-acetyl-L-cysteine and the p38 inhibitor SB203580. These data suggest that CuD induces cell cycle arrest and apoptosis via the ROS/p38 pathway in Capan-1 pancreatic cancer cell line; hence, CuD is a promising candidate that should be explored further for its effectiveness as an anticancer agent against pancreatic cancer.
Collapse
|
11
|
Yang X, Chen X, Xia C, Li S, Zhu L, Xu C. Comparative analysis of the expression profiles of genes related to the Gadd45α signaling pathway in four kinds of liver diseases. Histol Histopathol 2020; 35:949-960. [PMID: 32298459 DOI: 10.14670/hh-18-218] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Gadd45α (growth arrest and DNA damage inducible alpha) is a member of a group of genes whose transcript levels are increased following stressful conditions that lead to growth arrest and treatment with agents that lead to DNA damage. Gadd45α is upregulated in liver cirrhosis (LC), hepatic cancer (HC), acute liver failure (AHF) and non-alcoholic fatty liver disease(NAFLD). Here, we investigated the essential differences in the Gadd45α signaling pathway in these diseases at the transcriptional level. The results showed that 44, 46, 71 and 27 genes significant changes in these diseases, and the H-cluster showed that the expression of the Gadd45α signaling-related genes was significantly different in the four liver diseases. DAVID functional analysis showed that the Gadd45α signaling pathway-related genes were mainly involved in cell adhesion and migration, cell proliferation, apoptosis, stress and inflammatory responses, etc. Ingenuity pathway analysis (IPA) software was used to predict the functions of the Gadd45α signaling-related genes, and the results indicated that there were significant changes in cell differentiation, DNA damage repair, autophagy, apoptosis and necrosis. Gadd45α signaling pathway is involved in four kinds of liver disease and regulates a variety of activities via P38 MAPK, NF-κB, mTOR/STAT3, P21, PCNA, PI3K/Akt and other signaling pathways. Modulation of Gadd45α may be exploited to prevent the progression of liver disease, and to identify specific treatments for different stages of liver disease. In summary, the Gadd45α signaling pathway is involved in four kinds of liver disease and regulates a variety of physiological activities through various signaling pathways.
Collapse
Affiliation(s)
- Xianguang Yang
- College of Life Science, Henan Normal University, Xinxiang, Henan Province, China. .,State Key Laboratory Cultivation Base for Cell Differentiation Regulation and Henan Bioengineering Key Laboratory, Henan Normal University, Xinxiang, Henan Province, China
| | - Xuelin Chen
- State Key Laboratory Cultivation Base for Cell Differentiation Regulation and Henan Bioengineering Key Laboratory, Henan Normal University, Xinxiang, Henan Province, China.,College of Life Science, Henan Normal University, Xinxiang, Henan Province, China
| | - Cong Xia
- State Key Laboratory Cultivation Base for Cell Differentiation Regulation and Henan Bioengineering Key Laboratory, Henan Normal University, Xinxiang, Henan Province, China.,College of Life Science, Henan Normal University, Xinxiang, Henan Province, China
| | - Shuaihong Li
- State Key Laboratory Cultivation Base for Cell Differentiation Regulation and Henan Bioengineering Key Laboratory, Henan Normal University, Xinxiang, Henan Province, China.,College of Life Science, Henan Normal University, Xinxiang, Henan Province, China
| | - Lin Zhu
- State Key Laboratory Cultivation Base for Cell Differentiation Regulation and Henan Bioengineering Key Laboratory, Henan Normal University, Xinxiang, Henan Province, China.,College of Life Science, Henan Normal University, Xinxiang, Henan Province, China
| | - Cunshuan Xu
- State Key Laboratory Cultivation Base for Cell Differentiation Regulation and Henan Bioengineering Key Laboratory, Henan Normal University, Xinxiang, Henan Province, China.,College of Life Science, Henan Normal University, Xinxiang, Henan Province, China
| |
Collapse
|
12
|
Guo F, Xia T, Zhang Y, Ma X, Yan Z, Hao S, Han Y, Ma R, Zhou Y, Du X. Menstrual blood derived mesenchymal stem cells combined with Bushen Tiaochong recipe improved chemotherapy-induced premature ovarian failure in mice by inhibiting GADD45b expression in the cell cycle pathway. Reprod Biol Endocrinol 2019; 17:56. [PMID: 31311554 PMCID: PMC6636150 DOI: 10.1186/s12958-019-0499-2] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/15/2019] [Accepted: 07/06/2019] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND To investigate the therapeutic effects of menstrual blood derived mesenchymal stem cells (MB-MSCs) combined with Bushen Tiaochong recipe (BSTCR) on epirubicin induced premature ovarian failure (POF) in mice. METHODS Twenty-four female C57BL/6 mice of 6-8 weeks were intraperitoneally injected with epirubicin to induce POF, and then they were randomized into 4 groups of 6 mice each and treated with PBS, MB-MSCs, BSTCR, and MB-MSCs combined with BSTCR, respectively. Six mice of the same age were used as controls. Vaginal smear, TUNEL and hematoxylin-eosin staining were to observe estrous cycles, ovarian cell apoptosis and follicles. Enzyme-linked immunosorbent analysis determined serum estradiol, follicle-stimulating hormone (FSH) and anti-Müllerian hormone (AMH) levels. RT-qPCR and Western Blot analysis were to determine GADD45b, CyclinB1, CDC2 and pCDC2 expressions. RESULTS Epirubicin treatment resulted in a decrease in the number of primordial, primary, secondary and antral follicles, an increase in the number of atretic follicles and ovarian cell apoptosis, a decrease in estradiol and AMH levels, an increase in FSH levels, and estrous cycle arrest. However, MB-MSCs combined with BSTCR rescued epirubicin induced POF through down-regulating GADD45b and pCDC2 expressions, and up-regulating CyclinB1 and CDC2 expressions. The combined treatment showed better therapeutic efficacy than BSTCR or MB-MSCs alone. CONCLUSIONS MB-MSCs combined with BSTCR improved the ovarian function of epirubicin induced POF mice, which might be related to the inhibition of GADD45b expression and the promotion of CyclinB1 and CDC2 expressions. The combined treatment had better therapeutic efficacy than BSTCR or MB-MSCs alone.
Collapse
Affiliation(s)
- Fengyi Guo
- 0000 0000 9792 1228grid.265021.2Department of Obstetrics & Gynecology, General Hospital, Tianjin Medical University, NO.154, Anshan Road, Heping District, Tianjin, 300052 People’s Republic of China
| | - Tian Xia
- 0000 0004 1799 2712grid.412635.7Reproductive Center, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, No. 88 Chang Ling Street, Xi Qing district, Tianjin, 300112 China
| | - Yedan Zhang
- 0000 0000 9792 1228grid.265021.2Department of Obstetrics & Gynecology, General Hospital, Tianjin Medical University, NO.154, Anshan Road, Heping District, Tianjin, 300052 People’s Republic of China
| | - Xiaotong Ma
- 0000 0000 9792 1228grid.265021.2Department of Obstetrics & Gynecology, General Hospital, Tianjin Medical University, NO.154, Anshan Road, Heping District, Tianjin, 300052 People’s Republic of China
| | - Zhongrui Yan
- 0000 0000 9792 1228grid.265021.2Department of Obstetrics & Gynecology, General Hospital, Tianjin Medical University, NO.154, Anshan Road, Heping District, Tianjin, 300052 People’s Republic of China
| | - Shaohua Hao
- 0000 0000 9792 1228grid.265021.2Department of Obstetrics & Gynecology, General Hospital, Tianjin Medical University, NO.154, Anshan Road, Heping District, Tianjin, 300052 People’s Republic of China
| | - Yali Han
- 0000 0000 9792 1228grid.265021.2Department of Obstetrics & Gynecology, General Hospital, Tianjin Medical University, NO.154, Anshan Road, Heping District, Tianjin, 300052 People’s Republic of China
| | - Ruihong Ma
- 0000 0004 1799 2712grid.412635.7Reproductive Center, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, No. 88 Chang Ling Street, Xi Qing district, Tianjin, 300112 China
| | - Yuan Zhou
- grid.461843.cState Key Laboratory of Experimental Hematology, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, 300020 China
| | - Xue Du
- 0000 0000 9792 1228grid.265021.2Department of Obstetrics & Gynecology, General Hospital, Tianjin Medical University, NO.154, Anshan Road, Heping District, Tianjin, 300052 People’s Republic of China
| |
Collapse
|
13
|
Lin SY, Dan X, Du XX, Ran CL, Lu X, Ren SJ, Tang ZT, Yin LZ, He CL, Yuan ZX, Fu HL, Zhao XL, Shu G. Protective Effects of Salidroside against Carbon Tetrachloride (CCl 4)-Induced Liver Injury by Initiating Mitochondria to Resist Oxidative Stress in Mice. Int J Mol Sci 2019; 20:E3187. [PMID: 31261843 PMCID: PMC6651463 DOI: 10.3390/ijms20133187] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2019] [Revised: 06/22/2019] [Accepted: 06/24/2019] [Indexed: 12/14/2022] Open
Abstract
The antioxidant effect of salidroside has been proven, but its role in liver injury is poorly understood. In this study, we aimed to evaluate the protective effects and mechanism of salidroside on liver injury induced by carbon tetrachloride (CCl4) in vivo. Mice were pretreated with salidroside (60 mg/kg, intraperitoneally injected, i.p.) once per day for 14 consecutive days and then administered with CCl4 (15.95 g/kg, i.p.) for 24 h to produce a liver injury model. Salidroside attenuated hepatic transaminase elevation in serum and ameliorated liver steatosis and necrosis, thereby suggesting its protective effect on the liver. Salidroside antagonized CCl4-induced toxicity by equilibrating antioxidation system, thereby inhibiting reactive oxygen species accumulation, and restoring mitochondrial structure and function. Salidroside exerts antioxidant and liver-protective effects by selectively inhibiting the activation of genes, including growth arrest and DNA -damage-inducible 45 α (Gadd45a), mitogen-activated protein kinase 7 (Mapk7), and related RAS viral oncogene homolog 2 (Rras2), which induce oxidative stress in the mitogen-activated protein kinase pathway. These results revealed that salidroside can protect the liver from CCl4-induced injury by resisting oxidative stress and protecting mitochondrial function.
Collapse
Affiliation(s)
- Shi-Yu Lin
- Department of Pharmacy, Veterinary Medicine College of Sichuan Agricultural University, Chengdu 611130, China
| | - Xu Dan
- Department of Pharmacy, Veterinary Medicine College of Sichuan Agricultural University, Chengdu 611130, China
| | - Xia-Xia Du
- Department of Pharmacy, Veterinary Medicine College of Sichuan Agricultural University, Chengdu 611130, China
| | - Chong-Lin Ran
- Department of Pharmacy, Veterinary Medicine College of Sichuan Agricultural University, Chengdu 611130, China
| | - Xu Lu
- Department of Pharmacy, Veterinary Medicine College of Sichuan Agricultural University, Chengdu 611130, China
| | - Shao-Jun Ren
- Department of Pharmacy, Veterinary Medicine College of Sichuan Agricultural University, Chengdu 611130, China
| | - Zi-Ting Tang
- Department of Pharmacy, Veterinary Medicine College of Sichuan Agricultural University, Chengdu 611130, China
| | - Li-Zi Yin
- Department of Pharmacy, Veterinary Medicine College of Sichuan Agricultural University, Chengdu 611130, China
| | - Chang-Liang He
- Department of Pharmacy, Veterinary Medicine College of Sichuan Agricultural University, Chengdu 611130, China
| | - Zhi-Xiang Yuan
- Department of Pharmacy, Veterinary Medicine College of Sichuan Agricultural University, Chengdu 611130, China
| | - Hua-Lin Fu
- Department of Pharmacy, Veterinary Medicine College of Sichuan Agricultural University, Chengdu 611130, China
| | - Xiao-Ling Zhao
- Department of Animal Science, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China
| | - Gang Shu
- Department of Pharmacy, Veterinary Medicine College of Sichuan Agricultural University, Chengdu 611130, China.
| |
Collapse
|
14
|
Camilleri-Robles C, Serras F, Corominas M. Role of D-GADD45 in JNK-Dependent Apoptosis and Regeneration in Drosophila. Genes (Basel) 2019; 10:378. [PMID: 31109086 PMCID: PMC6562583 DOI: 10.3390/genes10050378] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2019] [Revised: 05/14/2019] [Accepted: 05/16/2019] [Indexed: 12/11/2022] Open
Abstract
The GADD45 proteins are induced in response to stress and have been implicated in the regulation of several cellular functions, including DNA repair, cell cycle control, senescence, and apoptosis. In this study, we investigate the role of D-GADD45 during Drosophila development and regeneration of the wing imaginal discs. We find that higher expression of D-GADD45 results in JNK-dependent apoptosis, while its temporary expression does not have harmful effects. Moreover, D-GADD45 is required for proper regeneration of wing imaginal discs. Our findings demonstrate that a tight regulation of D-GADD45 levels is required for its correct function both, in development and during the stress response after cell death.
Collapse
Affiliation(s)
- Carlos Camilleri-Robles
- Departament de Genètica, Microbiologia i Estadística, Facultat de Biologia and Institut de Biomedicina (IBUB), Universitat de Barcelona, Barcelona 08028, Spain.
| | - Florenci Serras
- Departament de Genètica, Microbiologia i Estadística, Facultat de Biologia and Institut de Biomedicina (IBUB), Universitat de Barcelona, Barcelona 08028, Spain.
| | - Montserrat Corominas
- Departament de Genètica, Microbiologia i Estadística, Facultat de Biologia and Institut de Biomedicina (IBUB), Universitat de Barcelona, Barcelona 08028, Spain.
| |
Collapse
|
15
|
Li C, Ming Y, Wang Z, Xu Q, Yao L, Xu D, Tang Y, Lei X, Li X, Mao Y. GADD45α alleviates acetaminophen-induced hepatotoxicity by promoting AMPK activation. Cell Mol Life Sci 2019; 76:129-145. [PMID: 30151693 PMCID: PMC11105285 DOI: 10.1007/s00018-018-2912-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2018] [Revised: 07/31/2018] [Accepted: 08/22/2018] [Indexed: 02/08/2023]
Abstract
As an analgesic and antipyretic drug, acetaminophen (APAP) is commonly used and known to be safe at therapeutic doses. In many countries, the overuse of APAP provokes acute liver injury and even liver failure. APAP-induced liver injury (AILI) is the most used experimental model of drug-induced liver injury (DILI). Here, we have demonstrated elevated levels of growth arrest and DNA damage-inducible 45α (GADD45α) in the livers of patients with DILI/AILI, in APAP-injured mouse livers and in APAP-treated hepatocytes. GADD45α exhibited a protective effect against APAP-induced liver injury and alleviated the accumulation of small lipid droplets in vitro and in vivo. We found that GADD45α promoted the activation of AMP-activated protein kinase α and induced fatty acid beta-oxidation, tricarboxylic acid cycle (TCA) and glycogenolysis-related gene expression after APAP exposure. Liquid chromatography-mass spectrometry (LC-MS) analysis showed that GADD45α increased the levels of TCA cycle metabolites. Co-immunoprecipitation analysis showed that Ppp2cb, a catalytic subunit of protein phosphatase 2A, could interact directly with GADD45α. Our results indicate that hepatocyte GADD45α might represent a therapeutic target to prevent and rescue liver injury caused by APAP.
Collapse
Affiliation(s)
- Chunmin Li
- Division of Gastroenterology and Hepatology, School of Medicine, Shanghai Institute of Digestive Disease, Renji Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Yanan Ming
- Division of Gastroenterology and Hepatology, School of Medicine, Shanghai Institute of Digestive Disease, Renji Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Zhengyang Wang
- Department of Pathology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, Henan, China
| | - Qingling Xu
- Department of Hepatology, Mengchao Hepatobiliary Hospital, Fujian Medical University, Fuzhou, Fujian, China
| | - Lvfeng Yao
- Department of Hepatology, Mengchao Hepatobiliary Hospital, Fujian Medical University, Fuzhou, Fujian, China
| | - Dongke Xu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fudan University, Shanghai, 200032, China
| | - Yingyue Tang
- Division of Gastroenterology and Hepatology, School of Medicine, Shanghai Institute of Digestive Disease, Renji Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Xiaohong Lei
- Division of Gastroenterology and Hepatology, School of Medicine, Shanghai Institute of Digestive Disease, Renji Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Xiaobo Li
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fudan University, Shanghai, 200032, China.
| | - Yimin Mao
- Division of Gastroenterology and Hepatology, School of Medicine, Shanghai Institute of Digestive Disease, Renji Hospital, Shanghai Jiao Tong University, Shanghai, China.
| |
Collapse
|
16
|
Fang Y, Xu XY, Shen Y, Li J. Molecular cloning and functional analysis of Growth arrest and DNA damage-inducible 45 aa and ab (Gadd45aa and Gadd45 ab) in Ctenopharyngodon idella. FISH & SHELLFISH IMMUNOLOGY 2018; 77:187-193. [PMID: 29605506 DOI: 10.1016/j.fsi.2018.03.051] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/25/2017] [Revised: 03/14/2018] [Accepted: 03/28/2018] [Indexed: 06/08/2023]
Abstract
The Gadd45aa and Gadd45 ab genes are members of the Gadd45 family, which are critically involved in immunological and apoptosis functions. In this study, we isolated and characterized Gadd45aa and Gadd45 ab cDNA from grass carp (Ctenopharyngodon idella) (designated CiGadd45aa and CiGadd45 ab). The CiGadd45aa and CiGadd45 ab fragments spanned 1272 bp/1248 bp, which contained 474 bp/480 bp open reading frames encoding 157/159 amino acid proteins. BLAST analysis revealed that CiGadd45aa and CiGadd45 ab shared high similarity with known Gadd45a sequences. qRT-PCR analysis showed widespread and abundant expression of CiGadd45aa in gill, intestine, kidney, brain, blood, skin and fin, but low in liver, spleen, head kidney, heart, and muscle. CiGadd45 ab was expressed highly in liver, spleen and blood but at low levels in gill, intestine, kidney, head kidney, heart, brain, skin, muscle, and fin. Following challenge of grass carp with Aeromonas hydrophila, CiGadd45aa and CiGadd45 ab expression was upregulated. In immune-relevant tissues and MAPK family genes (p38, JNK and ERK) were upregulated by CiGadd45aa and CiGadd45 ab overexpression and partly downregulated by interfered in the CIK grass carp kidney cell line. In addition, transcription of the cytokine-encoding il-8 gene was upregulated/downregulated by CiGadd45aa and CiGadd45 ab overexpression and interference. These results suggest that CiGadd45aa and CiGadd45 ab play roles in innate immune responses against A. hydrophila in grass carp.
Collapse
Affiliation(s)
- Yuan Fang
- Key Laboratory of Genetic Resources for Freshwater Aquaculture and Fisheries, Shanghai Ocean University, Ministry of Agriculture, Shanghai, 201306, China
| | - Xiao-Yan Xu
- Key Laboratory of Genetic Resources for Freshwater Aquaculture and Fisheries, Shanghai Ocean University, Ministry of Agriculture, Shanghai, 201306, China; Shanghai Engineering Research Center of Aquaculture, Shanghai Ocean University, Shanghai, China; National Demonstration Center for Experimental Fisheries Science Education, Shanghai Ocean University, Shanghai, China
| | - Yubang Shen
- Key Laboratory of Genetic Resources for Freshwater Aquaculture and Fisheries, Shanghai Ocean University, Ministry of Agriculture, Shanghai, 201306, China; Chinese Academy of Fishery Sciences, Key Laboratory of Freshwater Fisheries and Germplasm Resources Utilization, Wuxi, 214081, China; Shanghai Engineering Research Center of Aquaculture, Shanghai Ocean University, Shanghai, China; National Demonstration Center for Experimental Fisheries Science Education, Shanghai Ocean University, Shanghai, China
| | - Jiale Li
- Key Laboratory of Genetic Resources for Freshwater Aquaculture and Fisheries, Shanghai Ocean University, Ministry of Agriculture, Shanghai, 201306, China; Shanghai Engineering Research Center of Aquaculture, Shanghai Ocean University, Shanghai, China; National Demonstration Center for Experimental Fisheries Science Education, Shanghai Ocean University, Shanghai, China.
| |
Collapse
|
17
|
Zhang B, Deng C, Wang L, Zhou F, Zhang S, Kang W, Zhan P, Chen J, Shen S, Guo H, Zhang M, Wang Y, Zhang F, Zhang W, Xiao J, Kong B, Friess H, Zhuge Y, Yan H, Zou X. Upregulation of UBE2Q1 via gene copy number gain in hepatocellular carcinoma promotes cancer progression through β-catenin-EGFR-PI3K-Akt-mTOR signaling pathway. Mol Carcinog 2018; 57:201-215. [PMID: 29027712 DOI: 10.1002/mc.22747] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2017] [Revised: 08/22/2017] [Accepted: 09/29/2017] [Indexed: 01/20/2023]
Abstract
Hepatocellular carcinoma (HCC) is the most common type of liver cancer and represents a highly malignant tumor with a poor prognosis. Therapeutic modalities for HCC are limited and generally ineffective. UBE2Q1 is a putative E2 ubiquitin conjugating enzyme, and has been shown to be overexpressed in various types of cancers including HCC. How UBE2Q1 contributes to hepatocarcinogenesis remains unknown. Here, we show that UBE2Q1 is up-regulated in HCC cell lines and in a subset of human HCC tissues. Up-regulation of UBE2Q1 in primary HCC tumors was significantly correlated with shorter overall survival and disease-free survival. Mechanistically, we showed that the frequent up-regulation of UBE2Q1 in HCCs was attributed to the recurrent UBE2Q1 gene copy gain at chromosome 1q21. Functionally, we showed that knockdown of UBE2Q1 reduced HCC cell proliferation, promoted apoptosis via induction of GADD45α, and suppressed orthotopic tumorigenicity both in vitro and in vivo. Inactivation of UBE2Q1 also impeded HCC cell migration and invasion in vitro through regulating EMT process, and suppressed HCC metastasis in vivo. Interestingly, our data revealed a role of UBE2Q1 in the regulation of β-catenin-EGFR-PI3K-Akt-mTOR signaling pathway. Our findings indicate that UBE2Q1 is a candidate oncogene involved in HCC development and progression and therefore a potential therapeutic target in applicable HCC patients.
Collapse
Affiliation(s)
- Bin Zhang
- Department of Gastroenterology, the Affiliated Drum Tower Hospital of Nanjing University, Medical School, Nanjing, Jiangsu, China
| | - Chao Deng
- Department of Gastroenterology, the Affiliated Drum Tower Hospital of Nanjing University, Medical School, Nanjing, Jiangsu, China
| | - Lei Wang
- Department of Gastroenterology, the Affiliated Drum Tower Hospital of Nanjing University, Medical School, Nanjing, Jiangsu, China
| | - Fan Zhou
- Department of Gastroenterology, the Affiliated Drum Tower Hospital of Nanjing University, Medical School, Nanjing, Jiangsu, China
| | - Shu Zhang
- Department of Gastroenterology, the Affiliated Drum Tower Hospital of Nanjing University, Medical School, Nanjing, Jiangsu, China
| | - Wei Kang
- Department of Anatomical and Cellular Pathology, State Key Laboratory of Oncology in South China, Institute of Digestive Disease, Partner State Key Laboratory of Digestive Disease, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Ping Zhan
- Department of Respiratory Medicine, Jinling Hospital, Nanjing University, Medical School, Nanjing, Jiangsu, China
| | - Juan Chen
- The Key Laboratory of Molecular Biology on Infectious Diseases, Ministry of Education, The Second Affiliated Hospital of Chongqing Medical University, Chongqing Medical University, Chongqing, China
| | - Shanshan Shen
- Department of Gastroenterology, the Affiliated Drum Tower Hospital of Nanjing University, Medical School, Nanjing, Jiangsu, China
| | - Huimin Guo
- Department of Gastroenterology, the Affiliated Drum Tower Hospital of Nanjing University, Medical School, Nanjing, Jiangsu, China
| | - Ming Zhang
- Department of Gastroenterology, the Affiliated Drum Tower Hospital of Nanjing University, Medical School, Nanjing, Jiangsu, China
| | - Yi Wang
- Department of Gastroenterology, the Affiliated Drum Tower Hospital of Nanjing University, Medical School, Nanjing, Jiangsu, China
| | - Feng Zhang
- Department of Gastroenterology, the Affiliated Drum Tower Hospital of Nanjing University, Medical School, Nanjing, Jiangsu, China
| | - Wei Zhang
- Department of Gastroenterology, the Affiliated Drum Tower Hospital of Nanjing University, Medical School, Nanjing, Jiangsu, China
| | - Jiangqiang Xiao
- Department of Gastroenterology, the Affiliated Drum Tower Hospital of Nanjing University, Medical School, Nanjing, Jiangsu, China
| | - Bo Kong
- Department of Gastroenterology, the Affiliated Drum Tower Hospital of Nanjing University, Medical School, Nanjing, Jiangsu, China
- Department of Surgery, Technical University of Munich, Munich, Germany
| | - Helmut Friess
- Department of Surgery, Technical University of Munich, Munich, Germany
| | - Yuzheng Zhuge
- Department of Gastroenterology, the Affiliated Drum Tower Hospital of Nanjing University, Medical School, Nanjing, Jiangsu, China
| | - Hongli Yan
- Department of Laboratory Medicine, Changhai Hospital, the Second Military Medical University, Shanghai, China
| | - Xiaoping Zou
- Department of Gastroenterology, the Affiliated Drum Tower Hospital of Nanjing University, Medical School, Nanjing, Jiangsu, China
| |
Collapse
|
18
|
Mukherjee K, Sha X, Magimaidas A, Maifrede S, Skorski T, Bhatia R, Hoffman B, Liebermann DA. Gadd45a deficiency accelerates BCR-ABL driven chronic myelogenous leukemia. Oncotarget 2017; 8:10809-10821. [PMID: 28086219 PMCID: PMC5355225 DOI: 10.18632/oncotarget.14580] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2016] [Accepted: 11/23/2016] [Indexed: 12/26/2022] Open
Abstract
The Gadd45a stress sensor gene is a member in the Gadd45 family of genes that includes Gadd45b & Gadd45g. To investigate the effect of GADD45A in the development of CML, syngeneic wild type lethally irradiated mice were reconstituted with either wild type or Gadd45a null myeloid progenitors transduced with a retroviral vector expressing the 210-kD BCR-ABL fusion oncoprotein. Loss of Gadd45a was observed to accelerate BCR-ABL driven CML resulting in the development of a more aggressive disease, a significantly shortened median mice survival time, and increased BCR-ABL expressing leukemic stem/progenitor cells (GFP+Lin- cKit+Sca+). GADD45A deficient progenitors expressing BCR-ABL exhibited increased proliferation and decreased apoptosis relative to WT counterparts, which was associated with enhanced PI3K-AKT-mTOR-4E-BP1 signaling, upregulation of p30C/EBPa expression, and hyper-activation of p38 and Stat5. Furthermore, Gadd45a expression in samples obtained from CML patients was upregulated in more indolent chronic phase CML samples and down regulated in aggressive accelerated phase CML and blast crisis CML. These results provide novel evidence that Gadd45a functions as a suppressor of BCR/ABL driven leukemia and may provide a unique prognostic marker of CML progression.
Collapse
Affiliation(s)
- Kaushiki Mukherjee
- Fels Institute for Cancer Research and Molecular Biology, Philadelphia, PA, USA
| | - Xiaojin Sha
- Fels Institute for Cancer Research and Molecular Biology, Philadelphia, PA, USA
| | - Andrew Magimaidas
- Fels Institute for Cancer Research and Molecular Biology, Philadelphia, PA, USA.,Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Silvia Maifrede
- Fels Institute for Cancer Research and Molecular Biology, Philadelphia, PA, USA.,Department of Microbiology and Immunology, Temple University, Philadelphia, PA, USA
| | - Tomasz Skorski
- Fels Institute for Cancer Research and Molecular Biology, Philadelphia, PA, USA.,Department of Microbiology and Immunology, Temple University, Philadelphia, PA, USA
| | - Ravi Bhatia
- Division of Hematology and Oncology, University of Alabama, Tuscaloosa, AL, USA
| | - Barbara Hoffman
- Fels Institute for Cancer Research and Molecular Biology, Philadelphia, PA, USA.,Department of Medical Genetics and Molecular Biochemistry, Temple University, Philadelphia, PA, USA
| | - Dan A Liebermann
- Fels Institute for Cancer Research and Molecular Biology, Philadelphia, PA, USA.,Department of Medical Genetics and Molecular Biochemistry, Temple University, Philadelphia, PA, USA
| |
Collapse
|
19
|
Sadowska A, Nynca A, Ruszkowska M, Paukszto L, Myszczynski K, Orlowska K, Swigonska S, Molcan T, Jastrzebski JP, Ciereszko RE. Transcriptional profiling of porcine granulosa cells exposed to 2,3,7,8-tetrachlorodibenzo-p-dioxin. CHEMOSPHERE 2017; 178:368-377. [PMID: 28340459 DOI: 10.1016/j.chemosphere.2017.03.055] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/30/2016] [Revised: 03/06/2017] [Accepted: 03/12/2017] [Indexed: 06/06/2023]
Abstract
2,3,7,8-Tetrachlorodibenzo-p-dioxin (TCDD) is a toxic man-made chemical compound contaminating the environment. An exposure of living organisms to TCDD may result in numerous disorders, including reproductive pathologies. The aim of the current study was to examine the effects of TCDD on the transcriptome of porcine granulosa cell line AVG-16. By employing next-generation sequencing (NGS) we aimed to identify genes potentially involved in the mechanism of TCDD action and toxicity in porcine granulosa cells. The AVG-16 cells were treated with TCDD (100 nM) for 3, 12 or 24 h, and afterwards total cellular RNA was isolated and sequenced. In TCDD-treated cells we identified 141 differentially expressed genes (DEGs; padjusted < 0.05 and log2 fold change ≥1.0). The DEGs were assigned to GO term, covering biological processes, molecular functions and cellular components. Due to the large number of genes with altered expression, in the current study we analyzed only the genes involved in follicular growth, development and functioning. The obtained results showed that TCDD may affect ovarian follicle fate by influencing granulosa cell cycle, proliferation and DNA repair. The demonstrated over-time changes in the quantity and quality of genes being affected by TCDD treatment showed that the effects of TCDD on granulosa cells changed dramatically between 3-, 12- and 24-h of cell culture. This finding indicate that timing of gene expression measurement is critical for drawing correct conclusions on detailed relationships between the TCDD-affected genes and resulting intracellular processes. These conclusions have to be confirmed and extended by research involving proteomic and functional studies.
Collapse
Affiliation(s)
- Agnieszka Sadowska
- Department of Animal Physiology, Faculty of Biology and Biotechnology, University of Warmia and Mazury in Olsztyn, Poland.
| | - Anna Nynca
- Laboratory of Molecular Diagnostics, Faculty of Biology and Biotechnology, University of Warmia and Mazury in Olsztyn, Poland
| | - Monika Ruszkowska
- Department of Animal Physiology, Faculty of Biology and Biotechnology, University of Warmia and Mazury in Olsztyn, Poland
| | - Lukasz Paukszto
- Department of Plant Physiology, Genetics and Biotechnology, Faculty of Biology and Biotechnology, University of Warmia and Mazury in Olsztyn, Poland
| | - Kamil Myszczynski
- Department of Botany and Nature Protection, Faculty of Biology and Biotechnology, University of Warmia and Mazury in Olsztyn, Poland
| | - Karina Orlowska
- Department of Animal Physiology, Faculty of Biology and Biotechnology, University of Warmia and Mazury in Olsztyn, Poland
| | - Sylwia Swigonska
- Laboratory of Molecular Diagnostics, Faculty of Biology and Biotechnology, University of Warmia and Mazury in Olsztyn, Poland
| | - Tomasz Molcan
- Department of Animal Physiology, Faculty of Biology and Biotechnology, University of Warmia and Mazury in Olsztyn, Poland
| | - Jan P Jastrzebski
- Department of Plant Physiology, Genetics and Biotechnology, Faculty of Biology and Biotechnology, University of Warmia and Mazury in Olsztyn, Poland
| | - Renata E Ciereszko
- Department of Animal Physiology, Faculty of Biology and Biotechnology, University of Warmia and Mazury in Olsztyn, Poland; Laboratory of Molecular Diagnostics, Faculty of Biology and Biotechnology, University of Warmia and Mazury in Olsztyn, Poland
| |
Collapse
|
20
|
GADD45a Regulates Olaquindox-Induced DNA Damage and S-Phase Arrest in Human Hepatoma G2 Cells via JNK/p38 Pathways. Molecules 2017; 22:molecules22010124. [PMID: 28098804 PMCID: PMC6155949 DOI: 10.3390/molecules22010124] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2016] [Revised: 12/28/2016] [Accepted: 01/09/2017] [Indexed: 01/09/2023] Open
Abstract
Olaquindox, a quinoxaline 1,4-dioxide derivative, is widely used as a feed additive in many countries. The potential genotoxicity of olaquindox, hence, is of concern. However, the proper mechanism of toxicity was unclear. The aim of the present study was to investigate the effect of growth arrest and DNA damage 45 alpha (GADD45a) on olaquindox-induced DNA damage and cell cycle arrest in HepG2 cells. The results showed that olaquindox could induce reactive oxygen species (ROS)-mediated DNA damage and S-phase arrest, where increases of GADD45a, cyclin A, Cdk 2, p21 and p53 protein expression, decrease of cyclin D1 and the activation of phosphorylation-c-Jun N-terminal kinases (p-JNK), phosphorylation-p38 (p-p38) and phosphorylation-extracellular signal-regulated kinases (p-ERK) were involved. However, GADD45a knockdown cells treated with olaquindox could significantly decrease cell viability, exacerbate DNA damage and increase S-phase arrest, associated with the marked activation of p-JNK, p-p38, but not p-ERK. Furthermore, SP600125 and SB203580 aggravated olaquindox-induced DNA damage and S-phase arrest, suppressed the expression of GADD45a. Taken together, these findings revealed that GADD45a played a protective role in olaquindox treatment and JNK/p38 pathways may partly contribute to GADD45a regulated olaquindox-induced DNA damage and S-phase arrest. Our findings increase the understanding on the molecular mechanisms of olaquindox.
Collapse
|
21
|
Wingert S, Rieger MA. Terminal differentiation induction as DNA damage response in hematopoietic stem cells by GADD45A. Exp Hematol 2016; 44:561-6. [PMID: 27262218 DOI: 10.1016/j.exphem.2016.04.006] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2016] [Revised: 04/06/2016] [Accepted: 04/07/2016] [Indexed: 12/17/2022]
Abstract
Hematopoietic stem cells (HSCs) sustain lifelong blood cell regeneration by balancing their ability for self-renewal with their ability to differentiate into all blood cell types. To prevent organ exhaustion and malignant transformation, long-lived HSCs, in particular, must be protected from exogenous and endogenous stress, which cause severe DNA damage. When DNA is damaged, distinct DNA repair mechanisms and cell fate controls occur in adult HSCs compared with committed cells. Growth arrest and DNA damage-inducible 45 alpha (GADD45A) is known to coordinate a variety of cellular stress responses, indicating the molecule is an important stress mediator. So far, the function of GADD45A in hematopoietic stem and progenitor cells is controversial and appears highly dependent on the cell type and stress stimulus. Recent studies have analyzed its role in cell fate decision control of prospectively isolated HSCs and have revealed unexpected functions of GADD45A, as discussed here. The upregulation of GADD45A by DNA damage-causing conditions results in enhanced HSC differentiation, probably to efficiently eliminate aberrant HSCs from the system. These findings, in concert with a few studies on other stem cell systems, have led us to propose DNA damage-induced differentiation as a novel DNA damage response mechanism in stem cells that circumvents the fatal consequences of cumulative DNA damage in the stem cell compartment.
Collapse
Affiliation(s)
- Susanne Wingert
- LOEWE Center for Cell and Gene Therapy and Department of Medicine, Hematology/Oncology, Goethe University, Frankfurt am Main, Germany
| | - Michael A Rieger
- LOEWE Center for Cell and Gene Therapy and Department of Medicine, Hematology/Oncology, Goethe University, Frankfurt am Main, Germany; German Cancer Consortium (DKTK), Heidelberg, Germany; German Cancer Research Center (DKFZ), Heidelberg, Germany.
| |
Collapse
|
22
|
Yong KJ, Milenic DE, Baidoo KE, Brechbiel MW. Mechanisms of Cell Killing Response from Low Linear Energy Transfer (LET) Radiation Originating from (177)Lu Radioimmunotherapy Targeting Disseminated Intraperitoneal Tumor Xenografts. Int J Mol Sci 2016; 17:ijms17050736. [PMID: 27196891 PMCID: PMC4881558 DOI: 10.3390/ijms17050736] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2016] [Revised: 04/28/2016] [Accepted: 05/07/2016] [Indexed: 12/23/2022] Open
Abstract
Radiolabeled antibodies (mAbs) provide efficient tools for cancer therapy. The combination of low energy β(-)-emissions (500 keVmax; 130 keVave) along with a γ-emission for imaging makes (177)Lu (T1/2 = 6.7 day) a suitable radionuclide for radioimmunotherapy (RIT) of tumor burdens possibly too large to treat with α-particle radiation. RIT with (177)Lu-trastuzumab has proven to be effective for treatment of disseminated HER2 positive peritoneal disease in a pre-clinical model. To elucidate mechanisms originating from this RIT therapy at the molecular level, tumor bearing mice (LS-174T intraperitoneal xenografts) were treated with (177)Lu-trastuzumab comparatively to animals treated with a non-specific control, (177)Lu-HuIgG, and then to prior published results obtained using (212)Pb-trastuzumab, an α-particle RIT agent. (177)Lu-trastuzumab induced cell death via DNA double strand breaks (DSB), caspase-3 apoptosis, and interfered with DNA-PK expression, which is associated with the repair of DNA non-homologous end joining damage. This contrasts to prior results, wherein (212)Pb-trastuzumab was found to down-regulate RAD51, which is involved with homologous recombination DNA damage repair. (177)Lu-trastuzumab therapy was associated with significant chromosomal disruption and up-regulation of genes in the apoptotic process. These results suggest an inhibition of the repair mechanism specific to the type of radiation damage being inflicted by either high or low linear energy transfer radiation. Understanding the mechanisms of action of β(-)- and α-particle RIT comparatively through an in vivo tumor environment offers real information suitable to enhance combination therapy regimens involving α- and β(-)-particle RIT for the management of intraperitoneal disease.
Collapse
Affiliation(s)
- Kwon Joong Yong
- Radioimmune & Inorganic Chemistry Section, Radiation Oncology Branch, National Cancer Institute, National Institutes of Health, 10 Center Drive MSC-1002, Bethesda, MD 20892, USA.
| | - Diane E Milenic
- Radioimmune & Inorganic Chemistry Section, Radiation Oncology Branch, National Cancer Institute, National Institutes of Health, 10 Center Drive MSC-1002, Bethesda, MD 20892, USA.
| | - Kwamena E Baidoo
- Radioimmune & Inorganic Chemistry Section, Radiation Oncology Branch, National Cancer Institute, National Institutes of Health, 10 Center Drive MSC-1002, Bethesda, MD 20892, USA.
| | - Martin W Brechbiel
- Radioimmune & Inorganic Chemistry Section, Radiation Oncology Branch, National Cancer Institute, National Institutes of Health, 10 Center Drive MSC-1002, Bethesda, MD 20892, USA.
| |
Collapse
|
23
|
Martin TM, Plautz SA, Pannier AK. Temporal endogenous gene expression profiles in response to polymer-mediated transfection and profile comparison to lipid-mediated transfection. J Gene Med 2015; 17:33-53. [PMID: 25663627 DOI: 10.1002/jgm.2822] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2014] [Revised: 02/01/2015] [Accepted: 02/03/2015] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Design of efficient nonviral gene delivery systems is limited by the rudimentary understanding of specific molecules that facilitate transfection. METHODS Polyplexes using 25-kDa polyethylenimine (PEI) and plasmid-encoding green fluorescent protein (GFP) were delivered to HEK 293T cells. After treating cells with polyplexes, microarrays were used to identify endogenous genes differentially expressed between treated and untreated cells (2 h of exposure) or between flow-separated transfected cells (GFP+) and treated, untransfected cells (GFP-) at 8, 16 and 24 h after lipoplex treatment. Cell priming studies were conducted using pharmacologic agents to alter endogenous levels of the identified differentially expressed genes to determine effect on transfection levels. Differentially expressed genes in polyplex-mediated transfection were compared with those differentially expressed in lipoplex transfection to identify DNA carrier-dependent molecular factors. RESULTS Differentially expressed genes were RGS1, ARHGAP24, PDZD2, SNX24, GSN and IGF2BP1 after 2 h; RAP1A and ACTA1 after 8 h; RAP1A, WDR78 and ACTA1 after 16 h; and RAP1A, SCG5, ATF3, IREB2 and ACTA1 after 24 h. Pharmacologic studies altering endogenous levels for ARHGAP24, GSN, IGF2BP1, PDZD2 and RGS1 were able to increase or decrease transgene production. Comparing differentially expressed genes for polyplexes and lipoplexes, no common genes were identified at the 2-h time point, whereas, after the 8-h time point, RAP1A, ATF3 and HSPA6 were similarly expressed. SCG5 and PGAP1 were only upregulated in polyplex-transfected cells. CONCLUSIONS The identified genes and pharmacologic agents provide targets for improving transfection systems, although polyplex or lipoplex dependencies must be considered.
Collapse
Affiliation(s)
- Timothy M Martin
- Department of Pharmaceutical Sciences, Durham Research Center II, University of Nebraska-Medical Center, Omaha, NE, USA
| | | | | |
Collapse
|
24
|
Yang P, Ni F, Deng RQ, Qiang G, Zhao H, Yang MZ, Wang XY, Xu YZ, Chen L, Chen DL, Chen ZJ, Kan LX, Wang SY. MiR-362-5p promotes the malignancy of chronic myelocytic leukaemia via down-regulation of GADD45α. Mol Cancer 2015; 14:190. [PMID: 26545365 PMCID: PMC4636774 DOI: 10.1186/s12943-015-0465-3] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2015] [Accepted: 11/01/2015] [Indexed: 12/18/2022] Open
Abstract
Background MicroRNAs (miR, miRNAs) play pivotal roles in numerous physiological and pathophysiological contexts. We investigated whether miR-362-5p act as an oncogene in chronic myeloid leukaemia (CML) and aimed to understand its potential underlying mechanisms. Methods We compared the miR-362-5p expression levels between CML and non-CML cell lines, and between fresh blood samples from CML patients and normal healthy controls using quantitative real-time PCR (qPCR). Cell counting kit-8 (CCK-8) and Annexin V-FITC/PI analyses were used to measure the effects of miR-362-5p on proliferation and apoptosis, and Transwell assays were used to evaluate migration and invasion. A xenograft model was used to examine in vivo tumourigenicity. The potential target of miR-362-5p was confirmed by a luciferase reporter assay, qPCR and western blotting. Involvement of the JNK1/2 and P38 pathways was investigated by western blotting. Results miR-362-5p was up-regulated in CML cell lines and fresh blood samples from CML patients, and was associated with Growth arrest and DNA damage-inducible (GADD)45α down-regulation. Inhibition of miR-362-5p simultaneously repressed tumour growth and up-regulated GADD45α expression in a xenograft model. Consistently, the knockdown of GADD45α expression partially neutralized the effects of miR-362-5p inhibition. Furthermore study suggested that GADD45α mediated downstream the effects of miR-362-5p, which might indirectly regulates the activation of the JNK1/2 and P38 signalling pathways. Conclusion miR-362-5p acts as an oncomiR that down-regulates GADD45α, which consequently activates the JNK1/2 and P38 signalling. This finding provides novel insights into CML leukaemogenesis and may help identify new diagnostic and therapeutic targets. Electronic supplementary material The online version of this article (doi:10.1186/s12943-015-0465-3) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Peng Yang
- Department of Pathophysiology, School of Basic Medical Science, Anhui Medical University, 81 MeiShan Road, Hefei, Anhui, 230032, PR China. .,Department of Transfusion, the First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, 230022, PR China.
| | - Fang Ni
- Department of Pathophysiology, School of Basic Medical Science, Anhui Medical University, 81 MeiShan Road, Hefei, Anhui, 230032, PR China.
| | - Rui-Qing Deng
- Department of Pathophysiology, School of Basic Medical Science, Anhui Medical University, 81 MeiShan Road, Hefei, Anhui, 230032, PR China.
| | - Guo Qiang
- Department of Pathophysiology, School of Basic Medical Science, Anhui Medical University, 81 MeiShan Road, Hefei, Anhui, 230032, PR China.
| | - Hua Zhao
- Department of Pathophysiology, School of Basic Medical Science, Anhui Medical University, 81 MeiShan Road, Hefei, Anhui, 230032, PR China.
| | - Ming-Zhen Yang
- Department of Haematology, the First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, 230022, PR China.
| | - Xin-Yi Wang
- Department of Pathophysiology, School of Basic Medical Science, Anhui Medical University, 81 MeiShan Road, Hefei, Anhui, 230032, PR China. .,Department of Clinical Medicine, Anhui Medical University, Hefei, Anhui, 230032, PR China.
| | - You-Zhi Xu
- Department of Pathophysiology, School of Basic Medical Science, Anhui Medical University, 81 MeiShan Road, Hefei, Anhui, 230032, PR China.
| | - Li Chen
- Department of Pathophysiology, School of Basic Medical Science, Anhui Medical University, 81 MeiShan Road, Hefei, Anhui, 230032, PR China.
| | - Dan-Lei Chen
- Department of Pathophysiology, School of Basic Medical Science, Anhui Medical University, 81 MeiShan Road, Hefei, Anhui, 230032, PR China.
| | - Zhi-Jun Chen
- Department of Pathophysiology, School of Basic Medical Science, Anhui Medical University, 81 MeiShan Road, Hefei, Anhui, 230032, PR China.
| | - Li-Xin Kan
- Department of Pathophysiology, School of Basic Medical Science, Anhui Medical University, 81 MeiShan Road, Hefei, Anhui, 230032, PR China.
| | - Si-Ying Wang
- Department of Pathophysiology, School of Basic Medical Science, Anhui Medical University, 81 MeiShan Road, Hefei, Anhui, 230032, PR China.
| |
Collapse
|
25
|
Xu G, Zhang L, Ma A, Qian Y, Ding Q, Liu Y, Wang B, Yang Z, Liu Y. SIP1 is a downstream effector of GADD45G in senescence induction and growth inhibition of liver tumor cells. Oncotarget 2015; 6:33636-47. [PMID: 26378039 PMCID: PMC4741791 DOI: 10.18632/oncotarget.5602] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2015] [Accepted: 08/26/2015] [Indexed: 01/17/2023] Open
Abstract
Cellular senescence evasion caused by the inactivation of tumor suppressive programs is implicated in tumor initiation and therapeutic resistance. Our previous study has shown that the downregulation of growth arrest and DNA damage 45G (GADD45G) contributes to senescence bypass in hepatocellular carcinoma (HCC). Here, we report that the Smad-interacting protein-1 (SIP1) is transcriptionally activated and functions critically in the GADD45G-induced tumor cell senescence. Knockdown of SIP1 significantly abrogates the suppressive effects of GADD45G on the growth of xenografted liver tumor in vivo. The essential role of SIP1 in GADD45G activities is further validated in the model of the proteasome inhibitor MG132-induced cell senescence. We further show that JNK but not p38 MAPK activation is involved in the GADD45G-mediated SIP1 upregulation, and that JNK inhibition counteracts the GADD45G-induced cellular senescence. More importantly, we show that GADD45G and SIP1 expression are coincidently downregulated in primary human HCC tissues. Together, our results establish that the dowregulation of GADD45G-SIP1 axis may contribute to cellular senescence evasion and HCC development.
Collapse
Affiliation(s)
- Guiqin Xu
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Li Zhang
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Aihui Ma
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Yu Qian
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Qi Ding
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Yun Liu
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Boshi Wang
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Zhaojuan Yang
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Yongzhong Liu
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| |
Collapse
|
26
|
The GADD45A (1506T>C) Polymorphism Is Associated with Ovarian Cancer Susceptibility and Prognosis. PLoS One 2015; 10:e0138692. [PMID: 26422378 PMCID: PMC4589388 DOI: 10.1371/journal.pone.0138692] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2015] [Accepted: 09/02/2015] [Indexed: 11/20/2022] Open
Abstract
GADD45A (growth arrest and DNA damage 45 A) is the first stress-inducible gene identified to be a target of p53. However, no studies to date have assessed variants of the GADD45 gene and their potential relationship to tumor susceptibility. We investigated the association of the GADD45A (1506T>C) polymorphism with ovarian cancer development in 258 ovarian cancer patients and 332 age-matched healthy women as controls using sequence analysis. We found a statistically significant difference in the GADD45A (1506T>C) genotype distributions between the case and control groups (TT vs. TC vs. CC, P = 0.0021) and found that variant 1506T>C was significantly associated with an increased risk of ovarian cancer (P<0.001, OR = 1.71, 95% CI [1.28–2.29]). We observed a statistically significant effect between tumor histology (P = 0.032) and CA125 status (P = 0.021). Carrying the C allele (TC+CC) was associated with an increased risk of positive CA125 (OR = 3.20, 95% CI [1.15–8.71). Carrying the T allele (TT+TC) showed a significant correlation with both higher GADD45A mRNA expression and longer ovarian cancer RFS (relapse-free survival) and OS (overall survival). We are the first group to demonstrate that the GADD45A (1506T>C) polymorphism is associated with ovarian cancer susceptibility and prognosis. These data suggest that GADD45A (1506T>C) is a new tumor susceptibility gene and could be a useful molecular marker for assessing ovarian cancer risk and for predicting ovarian cancer patient prognosis.
Collapse
|
27
|
Barış İC, Caner V, Şen Türk N, Sarı İ, Hacıoğlu S, Doğu MH, Çetin O, Tepeli E, Can Ö, Bağcı G, Keskin A. Possible Role of GADD45γ Methylation in Diffuse Large B-Cell Lymphoma: Does It Affect the Progression and Tissue Involvement? Turk J Haematol 2015; 32:295-303. [PMID: 25912017 PMCID: PMC4805329 DOI: 10.4274/tjh.2014.0174] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Objective: Diffuse large B-cell lymphoma (DLBCL) is the most common type of non-Hodgkin lymphoma among adults and is characterized by heterogeneous clinical, immunophenotypic, and genetic features. Different mechanisms deregulating cell cycle and apoptosis play a role in the pathogenesis of DLBCL. Growth arrest DNA damage-inducible 45 (GADD45γ) is an important gene family involved in these mechanisms. The aims of this study are to determine the frequency of GADD45γ methylation, to evaluate the correlation between GADD45γ methylation and protein expression, and to investigate the relation between methylation status and clinicopathologic parameters in DLBCL tissues and reactive lymphoid node tissues from patients with reactive lymphoid hyperplasia. Materials and Methods: Thirty-six tissue samples of DLBCL and 40 nonmalignant reactive lymphoid node tissues were analyzed in this study. Methylation-sensitive high-resolution melting analysis was used for the determination of GADD45γ methylation status. The GADD45γ protein expression was determined by immunohistochemistry. Results: GADD45γ methylation was frequent (50.0%) in DLBCL. It was also significantly higher in advanced-stage tumors compared with early-stage (p=0.041). In contrast, unmethylated GADD45γ was associated with nodal involvement as the primary anatomical site (p=0.040). Conclusion: The results of this study show that, in contrast to solid tumors, the frequency of GADD45γ methylation is higher and this epigenetic alteration of GADD45γ may be associated with progression in DLBCL. In addition, nodal involvement is more likely to be present in patients with unmethylated GADD45γ.
Collapse
Affiliation(s)
| | - Vildan Caner
- Pamukkale University Faculty of Medicine, Department of Medical Biology, Denizli, Turkey Phone: +90 258 296 24 94 E-mail: ,
| | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
|
29
|
Gallium maltolate inhibits human cutaneous T-cell lymphoma tumor development in mice. J Invest Dermatol 2014; 135:877-884. [PMID: 25371972 DOI: 10.1038/jid.2014.476] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2014] [Revised: 09/22/2014] [Accepted: 10/20/2014] [Indexed: 01/07/2023]
Abstract
Cutaneous T-cell lymphomas (CTCLs) represent a heterogeneous group of non-Hodgkin's lymphoma characterized by an accumulation of malignant CD4 T cells in the skin. The group IIIa metal salt, gallium nitrate, is known to have antineoplastic activity against B-cell lymphoma in humans, but its activity in CTCLs has not been elaborated in detail. Herein, we examined the antineoplastic efficacy of a gallium compound, gallium maltolate (GaM), in vitro and in vivo with murine models of CTCLs. GaM inhibited cell growth and induced apoptosis of cultured CTCL cells. In human CTCL xenograft models, peritumoral injection of GaM limited the growth of CTCL cells, shown by fewer tumor formations, smaller tumor sizes, and decreased neovascularization in tumor microenvironment. To identify key signaling pathways that have a role in GaM-mediated reduction of tumor growth, we analyzed inflammatory cytokines, as well as signal transduction pathways in CTCL cells treated by GaM. IFN-γ-induced chemokines and IL-13 were found to be notably increased in GaM-treated CTCL cells. However, immunosuppressive cytokines, such as IL-10, were decreased with GaM treatment. Interestingly, both oxidative stress and p53 pathways were involved in GaM-induced cytotoxicity. These results warrant further investigation of GaM as a therapeutic agent for CTCLs.
Collapse
|
30
|
A systems biology approach to the analysis of subset-specific responses to lipopolysaccharide in dendritic cells. PLoS One 2014; 9:e100613. [PMID: 24949855 PMCID: PMC4065045 DOI: 10.1371/journal.pone.0100613] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2013] [Accepted: 05/28/2014] [Indexed: 12/24/2022] Open
Abstract
Dendritic cells (DCs) are critical for regulating CD4 and CD8 T cell immunity, controlling Th1, Th2, and Th17 commitment, generating inducible Tregs, and mediating tolerance. It is believed that distinct DC subsets have evolved to control these different immune outcomes. However, how DC subsets mount different responses to inflammatory and/or tolerogenic signals in order to accomplish their divergent functions remains unclear. Lipopolysaccharide (LPS) provides an excellent model for investigating responses in closely related splenic DC subsets, as all subsets express the LPS receptor TLR4 and respond to LPS in vitro. However, previous studies of the LPS-induced DC transcriptome have been performed only on mixed DC populations. Moreover, comparisons of the in vivo response of two closely related DC subsets to LPS stimulation have not been reported in the literature to date. We compared the transcriptomes of murine splenic CD8 and CD11b DC subsets after in vivo LPS stimulation, using RNA-Seq and systems biology approaches. We identified subset-specific gene signatures, which included multiple functional immune mediators unique to each subset. To explain the observed subset-specific differences, we used a network analysis approach. While both DC subsets used a conserved set of transcription factors and major signalling pathways, the subsets showed differential regulation of sets of genes that ‘fine-tune’ the network Hubs expressed in common. We propose a model in which signalling through common pathway components is ‘fine-tuned’ by transcriptional control of subset-specific modulators, thus allowing for distinct functional outcomes in closely related DC subsets. We extend this analysis to comparable datasets from the literature and confirm that our model can account for cell subset-specific responses to LPS stimulation in multiple subpopulations in mouse and man.
Collapse
|
31
|
Zhang L, Yang Z, Liu Y. GADD45 proteins: roles in cellular senescence and tumor development. Exp Biol Med (Maywood) 2014; 239:773-778. [PMID: 24872428 DOI: 10.1177/1535370214531879] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
The growth arrest and DNA damage 45 (GADD45) family genes regulate DNA repair, cell cycle, cell survival, apoptosis, senescence, and DNA demethylation in the cells under various stress stimuli, such as oxidative stress, UV radiation, and oncogenic stress. Recent studies have provided important insights regarding how different oncogenic stresses activate GADD45 signaling pathway and lead to disparate influences on tumor initiation. In this review, we discuss the deregulation and cellular function of GADD45 proteins in the context of cancer development. We also highlight recent advances in exploring the tumor suppressive function of GADD45 proteins-triggered cellular senescence.
Collapse
Affiliation(s)
- Li Zhang
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200240, China
| | - Zhaojuan Yang
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200240, China
| | - Yongzhong Liu
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200240, China
| |
Collapse
|
32
|
Hsu YC, Huang TY, Chen MJ. Therapeutic ROS targeting of GADD45γ in the induction of G2/M arrest in primary human colorectal cancer cell lines by cucurbitacin E. Cell Death Dis 2014; 5:e1198. [PMID: 24763055 PMCID: PMC4001305 DOI: 10.1038/cddis.2014.151] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2014] [Revised: 02/21/2014] [Accepted: 03/10/2014] [Indexed: 02/06/2023]
Abstract
Cucurbitacin E (CuE) or α-elaterin is a natural compound previously shown to be an antifeedant as well as a potent chemopreventive agent against several types of cancer. The present study investigated the anticancer effects of CuE on colorectal cancer (CRC) using primary cell lines isolated from five CRC patients in Taiwan, Specifically, we explored the anti-proliferation and cell cycle G2/M arrest induced by CuE in CRC cells. MPM-2 flow cytometry tests show that CuE-treated cells accumulated in metaphase (CuE 2.5-7.5 μM). Results further indicate that CuE produced G2/M arrest as well as the downregulation of CDC2 and cyclin B1 expression and dissociation. Both effects increased proportionally with the dose of CuE; however, the inhibition of proliferation, arrest of mitosis, production of reactive oxygen species (ROS), and loss of mitochondrial membrane potential (ΔΨm) were found to be dependent on the quantity of CuE used to treat the cancer cells. In addition, cell cycle arrest in treated cells coincided with the activation of the gene GADD45(α, β, γ). Incubation with CuE resulted in the binding of GADD45γ to CDC2, which suggests that the delay in CuE-induced mitosis is regulated by the overexpression of GADD45γ. Our findings suggest that, in addition to the known effects on cancer prevention, CuE may have antitumor activities in established CRC.
Collapse
Affiliation(s)
- Y-C Hsu
- Graduate Institute of Medical Science, College of Health Sciences, Chang Jung Christian University, Tainan, Taiwan
- Innovative Research Center of Medicine, College of Health Sciences, Chang Jung Christian University, Tainan, Taiwan
| | - T-Y Huang
- Department of Neurosurgery, Tainan Sin-Lau Hospital, Tainan, Taiwan
| | - M-J Chen
- Division of Traumatology, Department of Surgery, Chi Mei Medical Center, Tainan, Taiwan
- Department of Sports Management, College of Leisure and Recreation Management, Chia Nan University of Pharmacy and Science, Tainan, Taiwan
| |
Collapse
|
33
|
Anticancer and multidrug resistance-reversal effects of solanidine analogs synthetized from pregnadienolone acetate. Molecules 2014; 19:2061-76. [PMID: 24549231 PMCID: PMC6271930 DOI: 10.3390/molecules19022061] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2013] [Revised: 01/29/2014] [Accepted: 02/06/2014] [Indexed: 12/18/2022] Open
Abstract
A set of solanidine analogs with antiproliferative properties were recently synthetized from pregnadienolone acetate, which occurs in Nature. The aim of the present study was an in vitro characterization of their antiproliferative action and an investigation of their multidrug resistance-reversal activity on cancer cells. Six of the compounds elicited the accumulation of a hypodiploid population of HeLa cells, indicating their apoptosis-inducing character, and another one caused cell cycle arrest at the G2/M phase. The most effective agents inhibited the activity of topoisomerase I, as evidenced by plasmid supercoil relaxation assays. One of the most potent analogs down-regulated the expression of cell-cycle related genes at the mRNA level, including tumor necrosis factor alpha and S-phase kinase-associated protein 2, and induced growth arrest and DNA damage protein 45 alpha. Some of the investigated compounds inhibited the ABCB1 transporter and caused rhodamine-123 accumulation in murine lymphoma cells transfected by human MDR1 gene, expressing the efflux pump (L5178). One of the most active agents in this aspect potentiated the antiproliferative action of doxorubicin without substantial intrinsic cytostatic capacity. The current results indicate that the modified solanidine skeleton is a suitable substrate for the rational design and synthesis of further innovative drug candidates with anticancer activities.
Collapse
|
34
|
Ma L, Liu J, Liu L, Duan G, Wang Q, Xu Y, Xia F, Shan J, Shen J, Yang Z, Bie P, Cui Y, Bian XW, Prieto J, Avila MA, Qian C. Overexpression of the transcription factor MEF2D in hepatocellular carcinoma sustains malignant character by suppressing G2-M transition genes. Cancer Res 2014; 74:1452-62. [PMID: 24390737 DOI: 10.1158/0008-5472.can-13-2171] [Citation(s) in RCA: 74] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The underlying molecular pathogenesis in hepatocellular carcinoma remains poorly understood. The transcription factor MEF2D promotes survival in various cell types and it seems to function as an oncogene in leukemia. However, its potential contributions to solid cancers have not been explored. In this study, we investigated MEF2D expression and function in hepatocellular carcinoma, finding that MEF2D elevation in hepatocellular carcinoma clinical specimens was associated with poor prognosis. MEF2D-positive primary hepatocellular carcinoma cells displayed a faster proliferation rate compared with MEF2D-negative cells, and silencing or promoting MEF2D expression in these settings limited or accelerated cell proliferation, respectively. Notably, MEF2D-silencing abolished hepatocellular carcinoma tumorigenicity in mouse xenograft models. Mechanistic investigations revealed that MEF2D-silencing triggered G2-M arrest in a manner associated with direct downregulation of the cell-cycle regulatory genes RPRM, GADD45A, GADD45B, and CDKN1A. Furthermore, we identified MEF2D as an authentic target of miR-122, the reduced expression of which in hepatocellular carcinoma may be responsible for MEF2D upregulation. Together, our results identify MEF2D as a candidate oncogene in hepatocellular carcinoma and a potential target for hepatocellular carcinoma therapy.
Collapse
Affiliation(s)
- Leina Ma
- Authors' Affiliations: Institute of Pathology and Southwest Cancer Center; Institute of Hepatobiliary Surgery, Southwest Hospital, Third Military Medical University, Chongqing, China; Division of Hepatology and Gene Therapy, CIMA, University of Navarra, Pamplona; and CIBERehd. Instituto de Salud Carlos III, Madrid, Spain
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Sripayap P, Nagai T, Uesawa M, Kobayashi H, Tsukahara T, Ohmine K, Muroi K, Ozawa K. Mechanisms of resistance to azacitidine in human leukemia cell lines. Exp Hematol 2013; 42:294-306.e2. [PMID: 24368162 DOI: 10.1016/j.exphem.2013.12.004] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2013] [Revised: 11/28/2013] [Accepted: 12/16/2013] [Indexed: 12/31/2022]
Abstract
The DNA methylation inhibitor azacitidine (5-azacytidine) is used against myelodysplastic syndrome and acute myeloid leukemia, but drug resistance is an ongoing, intractable problem. To investigate resistance mechanisms, we generated two azacitidine-resistant cell lines, THP-1/AR and HL60/AR, and studied genetic disparities between them and their corresponding parental lines. In cells treated with azacitidine, significant mitotic variations were noted in parental cells which were absent in resistant cells, suggesting that resistance arises from negating azacitidine-mediated activation of apoptosis signaling and reestablishing G2/M checkpoint. Importantly, both resistant cell lines have common point mutations in the uridine-cytidine kinase 2 (UCK2) gene, which encodes the rate-limiting enzyme of the azacitidine activation pathway. Forced expression of mutated UCK2 in parental THP-1 cells abrogated azacitidine-induced apoptosis, whereas overexpression of wild type UCK2 in resistant THP-1/AR cells restored sensitivity to azacitidine, implying that UCK2 gene mutations perturb azacitidine activation and advance azacitidine resistance. Our study provides new insights into azacitidine resistance and establishes models useful in developing effective strategies to overcome it.
Collapse
Affiliation(s)
| | - Tadashi Nagai
- Division of Hematology, Jichi Medical University, Tochigi, Japan.
| | - Mitsuyo Uesawa
- Division of Hematology, Jichi Medical University, Tochigi, Japan
| | | | - Tomonori Tsukahara
- Division of Genetic Therapeutics, Center for Molecular Medicine, Jichi Medical University, Tochigi, Japan
| | - Ken Ohmine
- Division of Hematology, Jichi Medical University, Tochigi, Japan
| | - Kazuo Muroi
- Division of Cell Transplantation and Transfusion, Jichi Medical University, Tochigi, Japan
| | - Keiya Ozawa
- Division of Hematology, Jichi Medical University, Tochigi, Japan
| |
Collapse
|
36
|
Fontes-Oliveira CC, Busquets S, Fuster G, Ametller E, Figueras M, Olivan M, Toledo M, López-Soriano FJ, Qu X, Demuth J, Stevens P, Varbanov A, Wang F, Isfort RJ, Argilés JM. A differential pattern of gene expression in skeletal muscle of tumor-bearing rats reveals dysregulation of excitation-contraction coupling together with additional muscle alterations. Muscle Nerve 2013; 49:233-48. [DOI: 10.1002/mus.23893] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2012] [Revised: 04/19/2013] [Accepted: 04/24/2013] [Indexed: 12/31/2022]
Affiliation(s)
- Cibely Cristine Fontes-Oliveira
- Cancer Research Group, Departament de Bioquímica i Biologia Molecular, Facultat de Biologia; Universitat de Barcelona; Diagonal 643 Barcelona 08028 Spain
| | - Sílvia Busquets
- Cancer Research Group, Departament de Bioquímica i Biologia Molecular, Facultat de Biologia; Universitat de Barcelona; Diagonal 643 Barcelona 08028 Spain
- Institut de Biomedicina de la Universitat de Barcelona; Barcelona Spain
| | - Gemma Fuster
- Cancer Research Group, Departament de Bioquímica i Biologia Molecular, Facultat de Biologia; Universitat de Barcelona; Diagonal 643 Barcelona 08028 Spain
| | - Elisabet Ametller
- Cancer Research Group, Departament de Bioquímica i Biologia Molecular, Facultat de Biologia; Universitat de Barcelona; Diagonal 643 Barcelona 08028 Spain
| | - Maite Figueras
- Cancer Research Group, Departament de Bioquímica i Biologia Molecular, Facultat de Biologia; Universitat de Barcelona; Diagonal 643 Barcelona 08028 Spain
| | - Mireia Olivan
- Cancer Research Group, Departament de Bioquímica i Biologia Molecular, Facultat de Biologia; Universitat de Barcelona; Diagonal 643 Barcelona 08028 Spain
| | - Míriam Toledo
- Cancer Research Group, Departament de Bioquímica i Biologia Molecular, Facultat de Biologia; Universitat de Barcelona; Diagonal 643 Barcelona 08028 Spain
| | - Francisco J. López-Soriano
- Cancer Research Group, Departament de Bioquímica i Biologia Molecular, Facultat de Biologia; Universitat de Barcelona; Diagonal 643 Barcelona 08028 Spain
- Institut de Biomedicina de la Universitat de Barcelona; Barcelona Spain
| | - Xiaoyan Qu
- Procter & Gamble; Mason Business Center; 8700 Mason-Montgomery Road Mason Ohio 45040 USA
| | - Jeffrey Demuth
- Procter & Gamble; Mason Business Center; 8700 Mason-Montgomery Road Mason Ohio 45040 USA
| | - Paula Stevens
- Procter & Gamble; Mason Business Center; 8700 Mason-Montgomery Road Mason Ohio 45040 USA
| | - Alex Varbanov
- Procter & Gamble; Mason Business Center; 8700 Mason-Montgomery Road Mason Ohio 45040 USA
| | - Feng Wang
- Procter & Gamble; Mason Business Center; 8700 Mason-Montgomery Road Mason Ohio 45040 USA
| | - Robert J. Isfort
- Procter & Gamble; Mason Business Center; 8700 Mason-Montgomery Road Mason Ohio 45040 USA
| | - Josep M. Argilés
- Cancer Research Group, Departament de Bioquímica i Biologia Molecular, Facultat de Biologia; Universitat de Barcelona; Diagonal 643 Barcelona 08028 Spain
- Institut de Biomedicina de la Universitat de Barcelona; Barcelona Spain
| |
Collapse
|
37
|
Martin TM, Plautz SA, Pannier AK. Network analysis of endogenous gene expression profiles after polyethyleneimine-mediated DNA delivery. J Gene Med 2013; 15:142-54. [PMID: 23526566 DOI: 10.1002/jgm.2704] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2012] [Revised: 03/08/2013] [Accepted: 03/15/2013] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND DNA delivery systems, which transport exogenous DNA to cells, have applications that include gene therapy, tissue engineering and medical devices. Although the cationic nonviral DNA carrier polyethyleneimine (PEI) has been widely studied, the molecular factors and pathways underlying PEI-mediated DNA transfer remain largely unknown, preventing the design of more efficient delivery systems. METHODS HEK 293 T cells were treated with polyplexes formed with PEI and pEGFPLuc encoding for green fluorescent protein (GFP). Transfected cells expressing GFP were flow-separated from treated, untransfected cells. Gene expression profiles were obtained using Affymetrix HG-U133 2.0 microarrays and differentially expressed genes were identified using R/Bioconductor. Gene network analysis using EGAN (exploratory gene association network) bioinformatics tools was then used to find interaction among genes and enriched gene ontology (GO) terms related to transfection. Genes identified by this method were perturbed using pharmacologic activators or inhibitors to assess their effect on DNA transfer. RESULTS Microarray analysis comparing transfected cells to untransfected cells revealed 215 genes to be differentially expressed, with the majority enriched to GO processes including metabolism, response to stimulus, cell cycle, biological regulation and cellular component organization or biogenesis pathways. Gene network analysis revealed a coordinated induction of RAP1A, SCG5, PGAP1, ATF3 and NEB genes implicated in cell stress, cell cycle and cytoskeletal processes. Altering pathways with pharmacologic agents confirmed the potential role of RAP1A, SCG5 and ATF3 in transfection. CONCLUSIONS Microarray and gene network analyses of the sorted, transfected cell population can identify potential mediators of transfection, providing a basis for the design of improved delivery systems.
Collapse
Affiliation(s)
- Timothy M Martin
- Department of Biological Systems Engineering, University of Nebraska-Lincoln, Lincoln, NE, USA
| | | | | |
Collapse
|
38
|
Yong KJ, Milenic DE, Baidoo KE, Kim YS, Brechbiel MW. Gene expression profiling upon (212) Pb-TCMC-trastuzumab treatment in the LS-174T i.p. xenograft model. Cancer Med 2013; 2:646-53. [PMID: 24403230 PMCID: PMC3892796 DOI: 10.1002/cam4.132] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2013] [Revised: 07/30/2013] [Accepted: 08/14/2013] [Indexed: 02/03/2023] Open
Abstract
Recent studies have demonstrated that therapy with (212) Pb-TCMC-trastuzumab resulted in (1) induction of apoptosis, (2) G2/M arrest, and (3) blockage of double-strand DNA damage repair in LS-174T i.p. (intraperitoneal) xenografts. To further understand the molecular basis of the cell killing efficacy of (212) Pb-TCMC-trastuzumab, gene expression profiling was performed with LS-174T xenografts 24 h after exposure to (212) Pb-TCMC-trastuzumab. DNA damage response genes (84) were screened using a quantitative real-time polymerase chain reaction array (qRT-PCR array). Differentially regulated genes were identified following exposure to (212) Pb-TCMC-trastuzumab. These included genes involved in apoptosis (ABL, GADD45α, GADD45γ, PCBP4, and p73), cell cycle (ATM, DDIT3, GADD45α, GTSE1, MKK6, PCBP4, and SESN1), and damaged DNA binding (DDB) and repair (ATM and BTG2). The stressful growth arrest conditions provoked by (212) Pb-TCMC-trastuzumab were found to induce genes involved in apoptosis and cell cycle arrest in the G2/M phase. The expression of genes involved in DDB and single-strand DNA breaks was also enhanced by (212) Pb-TCMC-trastuzumab while no modulation of genes involved in double-strand break repair was apparent. Furthermore, the p73/GADD45 signaling pathway mediated by p38 kinase signaling may be involved in the cellular response, as evidenced by the enhanced expression of genes and proteins of this pathway. These results further support the previously described cell killing mechanism by (212) Pb-TCMC-trastuzumab in the same LS-174T i.p. xenograft. Insight into these mechanisms could lead to improved strategies for rational application of radioimmunotherapy using α-particle emitters.
Collapse
Affiliation(s)
- Kwon J Yong
- Radioimmune and Inorganic Chemistry Section, Radiation Oncology Branch, National Cancer Institute, National Institutes of HealthBethesda, Maryland
| | - Diane E Milenic
- Radioimmune and Inorganic Chemistry Section, Radiation Oncology Branch, National Cancer Institute, National Institutes of HealthBethesda, Maryland
| | - Kwamena E Baidoo
- Radioimmune and Inorganic Chemistry Section, Radiation Oncology Branch, National Cancer Institute, National Institutes of HealthBethesda, Maryland
| | - Young-Seung Kim
- Radioimmune and Inorganic Chemistry Section, Radiation Oncology Branch, National Cancer Institute, National Institutes of HealthBethesda, Maryland
| | - Martin W Brechbiel
- Radioimmune and Inorganic Chemistry Section, Radiation Oncology Branch, National Cancer Institute, National Institutes of HealthBethesda, Maryland
| |
Collapse
|
39
|
Meyer C, Sims AH, Morgan K, Harrison B, Muir M, Bai J, Faratian D, Millar RP, Langdon SP. Transcript and protein profiling identifies signaling, growth arrest, apoptosis, and NF-κB survival signatures following GNRH receptor activation. Endocr Relat Cancer 2013; 20. [PMID: 23202794 PMCID: PMC3573841 DOI: 10.1530/erc-12-0192] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
GNRH significantly inhibits proliferation of a proportion of cancer cell lines by activating GNRH receptor (GNRHR)-G protein signaling. Therefore, manipulation of GNRHR signaling may have an under-utilized role in treating certain breast and ovarian cancers. However, the precise signaling pathways necessary for the effect and the features of cellular responses remain poorly defined. We used transcriptomic and proteomic profiling approaches to characterize the effects of GNRHR activation in sensitive cells (HEK293-GNRHR, SCL60) in vitro and in vivo, compared to unresponsive HEK293. Analyses of gene expression demonstrated a dynamic response to the GNRH superagonist Triptorelin. Early and mid-phase changes (0.5-1.0 h) comprised mainly transcription factors. Later changes (8-24 h) included a GNRH target gene, CGA, and up- or downregulation of transcripts encoding signaling and cell division machinery. Pathway analysis identified altered MAPK and cell cycle pathways, consistent with occurrence of G(2)/M arrest and apoptosis. Nuclear factor kappa B (NF-κB) pathway gene transcripts were differentially expressed between control and Triptorelin-treated SCL60 cultures. Reverse-phase protein and phospho-proteomic array analyses profiled responses in cultured cells and SCL60 xenografts in vivo during Triptorelin anti-proliferation. Increased phosphorylated NF-κB (p65) occurred in SCL60 in vitro, and p-NF-κB and IκBε were higher in treated xenografts than controls after 4 days Triptorelin. NF-κB inhibition enhanced the anti-proliferative effect of Triptorelin in SCL60 cultures. This study reveals details of pathways interacting with intense GNRHR signaling, identifies potential anti-proliferative target genes, and implicates the NF-κB survival pathway as a node for enhancing GNRH agonist-induced anti-proliferation.
Collapse
Affiliation(s)
| | | | - Kevin Morgan
- Medical Research Council Human Reproductive Sciences UnitQueen's Medical Research Institute47 Little France Crescent, Edinburgh, EH16 4TJUK
| | | | | | | | | | - Robert P Millar
- Centre for Integrative PhysiologyUniversity of EdinburghEdinburgh, EH8 9XDUK
- Mammal Research InstituteUniversity Pretoria and UCT/MRC Receptor Biology Unit, University of Cape TownCape TownSouth Africa
| | | |
Collapse
|
40
|
Moskalev A, Plyusnina E, Shaposhnikov M, Shilova L, Kazachenok A, Zhavoronkov A. The role of D-GADD45 in oxidative, thermal and genotoxic stress resistance. Cell Cycle 2012; 11:4222-41. [PMID: 23095639 PMCID: PMC3524218 DOI: 10.4161/cc.22545] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
There is a relationship between various cellular stress factors and aging. In earlier studies, we demonstrated that overexpression of the D-GADD45 gene increases the life span of Drosophila melanogaster. In this study, we investigate the relationship between D-GADD45 activity and resistance to oxidative, genotoxic and thermal stresses as well as starvation. In most cases, flies with constitutive and conditional D-GADD45 overexpression in the nervous system were more stress-resistant than ones without overexpression. At the same time, most of the studied stress factors increased D-GADD45 expression in the wild-type strain. The lifespan-extending effect of D-GADD45 overexpression was also retained after exposure to chronic and acute gamma-irradiation, with doses of 40 сGy and 30 Gy, respectively. However, knocking out D-GADD45 resulted in a significant reduction in lifespan, lack of radiation hormesis and radioadaptive response. A dramatic decrease in the spontaneous level of D-GADD45 expression was observed in the nervous system as age progressed, which may be one of the causes of the age-related deterioration of organismal stress resistance. Thus, D-GADD45 expression is activated by most of the studied stress factors, and D-GADD45 overexpression resulted in an increase of stress resistance.
Collapse
Affiliation(s)
- Alexey Moskalev
- Laboratory of Molecular Radiobiology and Gerontology, Institute of Biology, Komi Science Center of Russian Academy of Sciences, Syktyvkar, Russia.
| | | | | | | | | | | |
Collapse
|
41
|
Park SW, Won KJ, Lee YS, Kim HS, Kim YK, Lee HW, Kim B, Lee BH, Kim JH, Kim DK. Increased HoxB4 Inhibits Apoptotic Cell Death in Pro-B Cells. THE KOREAN JOURNAL OF PHYSIOLOGY & PHARMACOLOGY : OFFICIAL JOURNAL OF THE KOREAN PHYSIOLOGICAL SOCIETY AND THE KOREAN SOCIETY OF PHARMACOLOGY 2012; 16:265-71. [PMID: 22915992 PMCID: PMC3419762 DOI: 10.4196/kjpp.2012.16.4.265] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/12/2012] [Revised: 07/04/2012] [Accepted: 07/19/2012] [Indexed: 02/07/2023]
Abstract
HoxB4, a homeodomain-containing transcription factor, is involved in the expansion of hematopoietic stem cells and progenitor cells in vivo and in vitro, and plays a key role in regulating the balance between hematopoietic stem cell renewal and cell differentiation. However, the biological activity of HoxB4 in other cells has not been reported. In this study, we investigated the effect of overexpressed HoxB4 on cell survival under various conditions that induce death, using the Ba/F3 cell line. Analysis of phenotypical characteristics showed that HoxB4 overexpression in Ba/F3 cells reduced cell size, death, and proliferation rate. Moreover, the progression from early to late apoptotic stages was inhibited in Ba/F3 cells subjected to HoxB4 overexpression under removal of interleukin-3-mediated signal, leading to the induction of cell cycle arrest at the G2/M phase and attenuated cell death by Fas protein stimulation in vitro. Furthermore, apoptotic cell death induced by doxorubicin-treated G2/M phase cell-cycle arrest also decreased with HoxB4 overexpression in Ba/F3 cells. From these data, we suggest that HoxB4 may play an important role in the regulation of pro-B cell survival under various apoptotic death environments.
Collapse
Affiliation(s)
- Sung-Won Park
- Department of Biomedical Science, College of Life Science, CHA University, Seongnam 463-712, Korea
| | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Frau M, Simile MM, Tomasi ML, Demartis MI, Daino L, Seddaiu MA, Brozzetti S, Feo CF, Massarelli G, Solinas G, Feo F, Lee JS, Pascale RM. An expression signature of phenotypic resistance to hepatocellular carcinoma identified by cross-species gene expression analysis. Cell Oncol (Dordr) 2012; 35:163-173. [PMID: 22434528 PMCID: PMC4517440 DOI: 10.1007/s13402-011-0067-z] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/27/2011] [Indexed: 01/08/2023] Open
Abstract
BACKGROUND AND AIMS Hepatocarcinogenesis is under polygenic control. We analyzed gene expression patterns of dysplastic liver nodules (DNs) and hepatocellular carcinomas (HCCs) chemically-induced in F344 and BN rats, respectively susceptible and resistant to hepatocarcinogenesis. METHODS Expression profiles were performed by microarray and validated by quantitative RT-PCR and Western blot. RESULTS Cluster analysis revealed two distinctive gene expression patterns, the first of which included normal liver of both strains and BN nodules, and the second one F344 nodules and HCC of both strains. We identified a signature predicting DN and HCC progression, characterized by highest expression of oncosuppressors Csmd1, Dmbt1, Dusp1, and Gnmt, in DNs, and Bhmt, Dmbt1, Dusp1, Gadd45g, Gnmt, Napsa, Pp2ca, and Ptpn13 in HCCs of resistant rats. Integrated gene expression data revealed highest expression of proliferation-related CTGF, c-MYC, and PCNA, and lowest expression of BHMT, DMBT1, DUSP1, GADD45g, and GNMT, in more aggressive rat and human HCC. BHMT, DUSP1, and GADD45g expression predicted patients' survival. CONCLUSIONS Our results disclose, for the first time, a major role of oncosuppressor genes as effectors of genetic resistance to hepatocarcinogenesis. Comparative functional genomic analysis allowed discovering an evolutionarily conserved gene expression signature discriminating HCC with different propensity to progression in rat and human.
Collapse
Affiliation(s)
- Maddalena Frau
- Department of Clinical and Experimental Medicine, Division of Experimental Pathology and Oncology, University of Sassari, Sassari, Italy
| | - Maria M. Simile
- Department of Clinical and Experimental Medicine, Division of Experimental Pathology and Oncology, University of Sassari, Sassari, Italy
| | - Maria L. Tomasi
- Department of Clinical and Experimental Medicine, Division of Experimental Pathology and Oncology, University of Sassari, Sassari, Italy
| | - Maria I. Demartis
- Department of Clinical and Experimental Medicine, Division of Experimental Pathology and Oncology, University of Sassari, Sassari, Italy
| | - Lucia Daino
- Department of Clinical and Experimental Medicine, Division of Experimental Pathology and Oncology, University of Sassari, Sassari, Italy
| | - Maria A. Seddaiu
- Department of Clinical and Experimental Medicine, Division of Experimental Pathology and Oncology, University of Sassari, Sassari, Italy
| | - Stefania Brozzetti
- Department of Surgery “Pietro Valdoni”, University of Rome “La Sapienza”, Rome, Italy
| | - Claudio F. Feo
- Department of Clinical and Experimental Medicine, Division of Surgery, University of Sassari, Sassari, Italy
| | - Giovanni Massarelli
- Department of Clinical and Experimental Medicine and Oncology, Division of Morbid Anatomy, University of Sassari, Sassari, Italy
| | - Giuliana Solinas
- Department of Clinical and Experimental Medicine, Division of Experimental Pathology and Oncology, University of Sassari, Sassari, Italy
| | - Francesco Feo
- Department of Clinical and Experimental Medicine, Division of Experimental Pathology and Oncology, University of Sassari, Sassari, Italy
| | - Ju-Seog Lee
- MD Anderson Cancer Center, University of Texas, Huston, TX, USA
| | - Rosa M. Pascale
- Department of Clinical and Experimental Medicine, Division of Experimental Pathology and Oncology, University of Sassari, Sassari, Italy
| |
Collapse
|
43
|
Tamura RE, de Vasconcellos JF, Sarkar D, Libermann TA, Fisher PB, Zerbini LF. GADD45 proteins: central players in tumorigenesis. Curr Mol Med 2012; 12:634-51. [PMID: 22515981 PMCID: PMC3797964 DOI: 10.2174/156652412800619978] [Citation(s) in RCA: 246] [Impact Index Per Article: 18.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2011] [Revised: 11/23/2011] [Accepted: 12/15/2011] [Indexed: 12/18/2022]
Abstract
The Growth Arrest and DNA Damage-inducible 45 (GADD45) proteins have been implicated in regulation of many cellular functions including DNA repair, cell cycle control, senescence and genotoxic stress. However, the pro-apoptotic activities have also positioned GADD45 as an essential player in oncogenesis. Emerging functional evidence implies that GADD45 proteins serve as tumor suppressors in response to diverse stimuli, connecting multiple cell signaling modules. Defects in the GADD45 pathway can be related to the initiation and progression of malignancies. Moreover, induction of GADD45 expression is an essential step for mediating anti-cancer activity of multiple chemotherapeutic drugs and the absence of GADD45 might abrogate their effects in cancer cells. In this review, we present a comprehensive discussion of the functions of GADD45 proteins, linking their regulation to effectors of cell cycle arrest, DNA repair and apoptosis. The ramifications regarding their roles as essential and central players in tumor growth suppression are also examined. We also extensively review recent literature to clarify how different chemotherapeutic drugs induce GADD45 gene expression and how its up-regulation and interaction with different molecular partners may benefit cancer chemotherapy and facilitate novel drug discovery.
Collapse
Affiliation(s)
- Rodrigo Esaki Tamura
- International Centre for Genetic Engineering and Biotechnology, and Medical Biochemistry Division, University of Cape Town, Cape Town, South Africa
| | - Jaíra Ferreira de Vasconcellos
- Centro Infantil Boldrini, Molecular Biology Laboratory, Campinas, Brazil
- State University of Campinas, Faculty of Medical Sciences, Department of Medical Genetics, Campinas, Brazil
| | - Devanand Sarkar
- Department of Human and Molecular Genetics, VCU Institute of Molecular Medicine, VCU Massey Cancer Center, Virginia Commonwealth University, School of Medicine, Richmond, VA
| | - Towia A Libermann
- BIDMC Genomics and Proteomics Center, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts
| | - Paul B Fisher
- Department of Human and Molecular Genetics, VCU Institute of Molecular Medicine, VCU Massey Cancer Center, Virginia Commonwealth University, School of Medicine, Richmond, VA
| | - Luiz Fernando Zerbini
- International Centre for Genetic Engineering and Biotechnology, and Medical Biochemistry Division, University of Cape Town, Cape Town, South Africa
- BIDMC Genomics and Proteomics Center, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
44
|
Kamino H, Negishi M. The nuclear receptor constitutive active/androstane receptor arrests DNA-damaged human hepatocellular carcinoma Huh7 cells at the G2/M phase. Mol Carcinog 2012; 51:206-12. [PMID: 21557330 PMCID: PMC3242902 DOI: 10.1002/mc.20783] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2011] [Revised: 03/18/2011] [Accepted: 03/24/2011] [Indexed: 11/11/2022]
Abstract
Here, we have demonstrated that xenobiotic activation of the nuclear receptor (CAR, NR1I3) can result in arresting DNA-damaged human hepatocellular carcinoma Huh7 cells at the G2/M phase. Huh7 cells over-expressing CAR were either treated with dimethyl sulfoxide, the CAR activator TCPOBOP (1,4-bis[2-(3,5-dichloropyridyloxy)]benzene; androstenol, 16,(5α)-androsten-3α-OL), or repressor androstenol; these treatments were then followed by adriamycin treatment to damage DNA. FACS analysis revealed that CAR-activation by TCPOBOP increased the rate of arrested Huh7 cells at the G2/M phase (4N DNA content) after DNA damage by adriamycin. This increase correlated with the increase of cell viability in TCPOBOP-treated Huh7 cells, as determined by MTT assays. Real-time polymerase chain reaction analysis determined that, as regulated by CAR, the growth arrest and DNA damage-inducible γ (GADD45γ) and Cyclin G2 genes increased and decreased, respectively, as TCPOBOP increased the number of Huh7 cells arrested at the G2/M phase. Thus, the results suggest that CAR regulates cell cycle, increasing G2/M arrest, and delaying the death of DNA-damaged cells.
Collapse
Affiliation(s)
- Hiroki Kamino
- Laboratory of Reproductive and Developmental Toxicology, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, North Carolina 27709, USA
| | | |
Collapse
|
45
|
Cury-Boaventura MF, Torrinhas RSMDM, Godoy ABPD, Curi R, Waitzberg DL. Human Leukocyte Death After a Preoperative Infusion of Medium/Long-Chain Triglyceride and Fish Oil Parenteral Emulsions. JPEN J Parenter Enteral Nutr 2012; 36:677-84. [DOI: 10.1177/0148607111432759] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Affiliation(s)
| | | | | | - Rui Curi
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Dan Linetzky Waitzberg
- Department of Gastroenterology, Faculty of Medicine, University of São Paulo (LIM 35), São Paulo, Brazil
| |
Collapse
|
46
|
Zhang C, Wang J, Lü G, Li J, Lu X, Mantion G, Vuitton DA, Wen H, Lin R. Hepatocyte proliferation/growth arrest balance in the liver of mice during E. multilocularis infection: a coordinated 3-stage course. PLoS One 2012; 7:e30127. [PMID: 22253905 PMCID: PMC3254660 DOI: 10.1371/journal.pone.0030127] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2011] [Accepted: 12/12/2011] [Indexed: 12/24/2022] Open
Abstract
Background Alveolar echinococcosis (AE) is characterized by the tumor-like growth of Echinococcus (E.) multilocularis. Very little is known on the influence of helminth parasites which develop in the liver on the proliferation/growth arrest metabolic pathways in the hepatocytes of the infected liver over the various stages of infection. Methodology/Principal Findings Using Western blot analysis, qPCR and immunohistochemistry, we measured the levels of MAPKs activation, Cyclins, PCNA, Gadd45β, Gadd45γ, p53 and p21 expression in the murine AE model, from day 2 to 360 post-infection. Within the early (day 2–60) and middle (day60–180) stages, CyclinB1 and CyclinD1 gene expression increased up to day30 and then returned to control level after day60; Gadd45β, CyclinA and PCNA increased all over the period; ERK1/2 was permanently activated. Meanwhile, p53, p21 and Gadd45γ gene expression, and caspase 3 activation, gradually increased in a time-dependent manner. In the late stage (day180–360), p53, p21 and Gadd45γ gene expression were significantly higher in infected mice; JNK and caspase 3 were activated. TUNEL analysis showed apoptosis of hepatocytes. No significant change in CyclinE, p53 mRNA and p-p38 expression were observed at any time. Conclusions Our data support the concept of a sequential activation of metabolic pathways which 1) would first favor parasitic, liver and immune cell proliferation and survival, and thus promote metacestode fertility and tolerance by the host, and 2) would then favor liver damage/apoptosis, impairment in protein synthesis and xenobiotic metabolism, as well as promote immune deficiency, and thus contribute to the dissemination of the protoscoleces after metacestode fertility has been acquired. These findings give a rational explanation to the clinical observations of hepatomegaly and of unexpected survival of AE patients after major hepatic resections, and of chronic liver injury, necrosis and of hepatic failure at an advanced stage and in experimental animals.
Collapse
Affiliation(s)
- Chuanshan Zhang
- State Key Laboratory Incubation Base of Major Diseases in Xinjiang and Xinjiang Key Laboratory of Echinococcosis, First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, China
| | - Junhua Wang
- State Key Laboratory Incubation Base of Major Diseases in Xinjiang and Xinjiang Key Laboratory of Echinococcosis, First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, China
| | - Guodong Lü
- State Key Laboratory Incubation Base of Major Diseases in Xinjiang and Xinjiang Key Laboratory of Echinococcosis, First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, China
| | - Jing Li
- State Key Laboratory Incubation Base of Major Diseases in Xinjiang and Xinjiang Key Laboratory of Echinococcosis, First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, China
| | - Xiaomei Lu
- State Key Laboratory Incubation Base of Major Diseases in Xinjiang and Xinjiang Key Laboratory of Echinococcosis, First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, China
| | - Georges Mantion
- World Health Organization-Collaborating Centre for the Prevention and Treatment of Human Echinococcosis, Department of Digestive Surgery; Jean Minjoz Hospital, University of Franche-Comté and University Hospital, Besançon, France
| | - Dominique A. Vuitton
- World Health Organization-Collaborating Centre for the Prevention and Treatment of Human Echinococcosis, Department of Digestive Surgery; Jean Minjoz Hospital, University of Franche-Comté and University Hospital, Besançon, France
- Research Unit EA 3181 “Epithelial Carcinogenesis: Predictive and Prognostic Factors,” University of Franche-Comté, Besançon, France
| | - Hao Wen
- State Key Laboratory Incubation Base of Major Diseases in Xinjiang and Xinjiang Key Laboratory of Echinococcosis, First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, China
- * E-mail: (RL); (HW)
| | - Renyong Lin
- State Key Laboratory Incubation Base of Major Diseases in Xinjiang and Xinjiang Key Laboratory of Echinococcosis, First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, China
- * E-mail: (RL); (HW)
| |
Collapse
|
47
|
Moskalev AA, Smit-McBride Z, Shaposhnikov MV, Plyusnina EN, Zhavoronkov A, Budovsky A, Tacutu R, Fraifeld VE. Gadd45 proteins: relevance to aging, longevity and age-related pathologies. Ageing Res Rev 2012; 11:51-66. [PMID: 21986581 PMCID: PMC3765067 DOI: 10.1016/j.arr.2011.09.003] [Citation(s) in RCA: 113] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2011] [Revised: 09/25/2011] [Accepted: 09/27/2011] [Indexed: 12/12/2022]
Abstract
The Gadd45 proteins have been intensively studied, in view of their important role in key cellular processes. Indeed, the Gadd45 proteins stand at the crossroad of the cell fates by controlling the balance between cell (DNA) repair, eliminating (apoptosis) or preventing the expansion of potentially dangerous cells (cell cycle arrest, cellular senescence), and maintaining the stem cell pool. However, the biogerontological aspects have not thus far received sufficient attention. Here we analyzed the pathways and modes of action by which Gadd45 members are involved in aging, longevity and age-related diseases. Because of their pleiotropic action, a decreased inducibility of Gadd45 members may have far-reaching consequences including genome instability, accumulation of DNA damage, and disorders in cellular homeostasis - all of which may eventually contribute to the aging process and age-related disorders (promotion of tumorigenesis, immune disorders, insulin resistance and reduced responsiveness to stress). Most recently, the dGadd45 gene has been identified as a longevity regulator in Drosophila. Although further wide-scale research is warranted, it is becoming increasingly clear that Gadd45s are highly relevant to aging, age-related diseases (ARDs) and to the control of life span, suggesting them as potential therapeutic targets in ARDs and pro-longevity interventions.
Collapse
Affiliation(s)
- Alexey A Moskalev
- Group of Molecular Radiobiology and Gerontology, Institute of Biology, Komi Science Center of Russian Academy of Sciences.
| | | | | | | | | | | | | | | |
Collapse
|
48
|
Vincristine potentiates the anti-proliferative effect of an aurora kinase inhibitor, VE-465, in myeloid leukemia cells. Biochem Pharmacol 2011; 82:1884-90. [PMID: 21971583 DOI: 10.1016/j.bcp.2011.09.015] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2011] [Revised: 09/09/2011] [Accepted: 09/12/2011] [Indexed: 12/25/2022]
Abstract
Aurora kinases play an essential role in the regulation of mitosis. The kinases are overexpressed in a variety of cancer cells and are involved in tumorgenesis. Although aurora kinase inhibitors are potential agents for treatment of leukemia, the establishment of efficacious combination therapies is an attractive approach for making good use of these agents. In this study, we examined the effects of a specific aurora kinase inhibitor, VE-465, in combination with various conventional anti-leukemia agents, including doxorubicin, daunorubicin, idarubicin, mitoxantron, cytosine arabinoside, vincristine and etoposide, on acute myeloid leukemia cell lines (HL60, U937, THP-1 and KY821), chronic myeloid leukemia cell lines (KCL22, K562 and KU812) and primary leukemia cells. We found that a combination of VE-465 and vincristine had a synergistic/additive inhibitory effect on the growth of leukemia cells. VE-465 initially increased G2/M-phase cells, followed by induction of sub-G1 cells. Vincristine enhanced this effect of VE-465. The combination of VE-465 and vincristine increased the levels of cleaved caspase 3, cleaved caspase 7, cleaved caspase 9, cleaved PARP and Phospho-Chk2, suggesting that the combination caused Chk2-mediated activation of the G2/M checkpoint, resulting in sequential induction of apoptosis. Interestingly, the combination markedly decreased the level of Phospho-ERK1/2, suggesting that the combination alters a network of cellular signaling pathways. In contrast, combinations of VE-465 and other agents showed no synergistic inhibitory effect but rather had an antagonistic effect. In conclusion, our results indicate the utility of the combination of VE-465 and vincristine as a potential therapy for myeloid leukemia.
Collapse
|
49
|
Liu Y, Zhou Y, Liu L, Sun L, Zhang M, Liu Y, Li D. Maize ZmMEK1 is a single-copy gene. Mol Biol Rep 2011; 39:2957-66. [PMID: 21691709 DOI: 10.1007/s11033-011-1057-y] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2010] [Accepted: 06/08/2011] [Indexed: 11/24/2022]
Abstract
Mitogen-activated protein kinase (MAPK) cascade constitutes a conserved signaling module in eukaryotes. MAPK kinase (MAPKK) plays a crucial role in a MAPK cascade. ZmMEK1 is the first characterized MAPKK gene in maize. Although ZmMEK1 has been studied in detail in biochemical level, the genomic organization of ZmMEK1 gene is obscure. In this research, we clarified ZmMEK1 is a single-copy gene in the maize genome. Southern blot analysis using 3' specific region of ZmMEK1 cDNA as a probe revealed the presence of distinct single bands in each lane of EcoRI and HindIII. Although previous Southern blot analysis using full-length ZmMEK1 cDNA as a probe revealed several hybridizing bands, we showed here that all bands come from one genomic fragment corresponding to ZmMEK1 gene. Furthermore, ZmMEK1 was induced by PEG, abscisic acid (ABA), and salicylic acid (SA) and was down-regulated by NaCl.
Collapse
Affiliation(s)
- Yukun Liu
- State Key Laboratory of Crop Biology, Shandong Key Laboratory of Crop Biology, College of Life Sciences, Shandong Agricultural University, 61 DaiZong Street, Tai'an, 271018 Shandong, China
| | | | | | | | | | | | | |
Collapse
|
50
|
Nakayama Wong LS, Aung HH, Lamé MW, Wegesser TC, Wilson DW. Fine particulate matter from urban ambient and wildfire sources from California's San Joaquin Valley initiate differential inflammatory, oxidative stress, and xenobiotic responses in human bronchial epithelial cells. Toxicol In Vitro 2011; 25:1895-905. [PMID: 21703343 DOI: 10.1016/j.tiv.2011.06.001] [Citation(s) in RCA: 62] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2011] [Revised: 06/02/2011] [Accepted: 06/03/2011] [Indexed: 01/27/2023]
Abstract
Environmental particulate matter (PM) exposure has been correlated with pathogenesis of acute airway inflammatory disease such as asthma and COPD. PM size and concentration have been studied extensively, but the additional effects of particulate components such as biological material, transition metals, and polycyclic aromatic hydrocarbons could also impact initial disease pathogenesis. In this study, we compared urban ambient particulate matter (APM) collected from Fresno, California with wildfire (WF) particulate matter collected from Escalon, California on early transcriptional responses in human bronchial epithelial cells (HBE). Global gene expression profiling of APM treated HBE activated genes related to xenobiotic metabolism (CYP 1B1), endogenous ROS generation and response genes (DUOX1, SOD2, PTGS2) and pro-inflammatory responses associated with asthma or COPD such as IL-1α, IL-1β, IL-8, and CCL20. WF PM treatments also induced a pro-inflammatory gene response, but elicited a more robust xenobiotic metabolism and oxidative stress response. Inhibitor studies targeting endotoxin, ROS, and trace metals, found endotoxin inhibition had modest selective inhibition of inflammation while inhibition of hydrogen peroxide and transition metals had broad effects suggesting additional interactions with xenobiotic metabolism pathways. APM induced a greater inflammatory response while WF PM had more marked metabolism and ROS related responses.
Collapse
Affiliation(s)
- L S Nakayama Wong
- School of Veterinary Medicine, Department of Pathology, Microbiology, and Immunology, University of California, Davis, CA 95616, United States
| | | | | | | | | |
Collapse
|