1
|
Liu W, Sun Y, Zhang Y, Yin D. The causal relationships between inflammatory cytokines, blood metabolites, and thyroid cancer: a two-step Mendelian randomization analysis. Discov Oncol 2025; 16:301. [PMID: 40072746 PMCID: PMC11904021 DOI: 10.1007/s12672-025-02029-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Accepted: 03/03/2025] [Indexed: 03/14/2025] Open
Abstract
BACKGROUND Thyroid cancer is a prevalent malignant tumor, especially with a higher incidence in women. Tumor microenvironment changes induced by inflammation and alterations in metabolic characteristics are critical in the development of thyroid cancer. Nevertheless, their causal relationships remain unclear. METHODS We utilized thyroid cancer GWAS data from the Global Biobank Meta-Analysis Initiative and GWAS data of 91 inflammatory cytokines and 1400 blood metabolites obtained from the GWAS Catalog to evaluate the causality between inflammatory cytokines, blood metabolites, and thyroid cancer using Mendelian randomization (MR). Initially, we identified inflammatory cytokines having a significant causal effect on thyroid cancer. Subsequently, for the identified positive blood metabolites, we applied a two-step mediation MR method to examine their mediating role in the causal effect of specific inflammatory cytokines on thyroid cancer. RESULTS Our forward MR analysis identified suggestive associations between 7 inflammatory cytokines and thyroid cancer risks, and found that tumor necrosis factor ligand superfamily member 14 (TNFSF14) (IVW-OR: 1.25, 95% CI 1.10-1.42, p = 0.0004) is a significant risk factor in thyroid cancer, and this causal relationship remained significant after Bonferroni correction. The reverse MR analysis identified suggestive causal associations between thyroid cancer and 3 inflammatory cytokines and ruled out the reverse causality between TNFSF14 and thyroid cancer. Then, we identified suggestive associations between 35 blood metabolites and 24 blood metabolite ratios with thyroid cancer, and found that 5-hydroxymethyl-2-furoylcarnitine (IVW-OR: 1.38, 95% CI 1.19-1.61, p = 0.00003) is a significant risk factor for thyroid cancer, with this causality remaining significant after Bonferroni correction. Finally, our two-step MR analysis indicated that Lactosyl-N-palmitoyl-sphingosine (d18:1/16:0) and X-12013 have a mediating effect in the causal relationship between TNFSF14 and thyroid cancer, with mediation proportions of 8.55% and 5.78%, respectively. Our MR analysis did not identify significant heterogeneity or horizontal pleiotropy. CONCLUSION This study identified some inflammatory cytokines and blood metabolites associated with thyroid cancer risk and revealed the mediating role of specific blood metabolites between TNFSF14 and thyroid cancer, highlighting the critical role of inflammatory and metabolic pathways in the pathogenesis of thyroid cancer.
Collapse
Affiliation(s)
- Weihao Liu
- Department of Thyroid Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Yuxiao Sun
- Department of Thyroid Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Yifei Zhang
- Department of Thyroid Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Detao Yin
- Department of Thyroid Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China.
- Engineering Research Center of Multidisciplinary Diagnosis and Treatment of Thyroid Cancer of Henan Province, Zhengzhou, 450052, China.
| |
Collapse
|
2
|
Li Y, Ding Z, Cheng T, Hu Y, Zhong F, Ren S, Wang S. Single-cell transcriptomics analysis reveals dynamic changes and prognostic signature in tumor microenvironment of PDAC. Sci Rep 2025; 15:4025. [PMID: 39894886 PMCID: PMC11788434 DOI: 10.1038/s41598-025-86950-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2024] [Accepted: 01/15/2025] [Indexed: 02/04/2025] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a highly lethal malignant tumor characterized by a complex tumor microenvironment (TME) with significant heterogeneity, posing immense challenges for devising effective therapeutic strategies. This study aims to elucidate the dynamic changes in the TME during PDAC progression and develop a prognostic model using single-cell RNA sequencing (scRNA-seq) data. We utilized a previously published comprehensive dataset comprising 31 samples (including 8 PDAC I, 9 PDAC II, 6 PDAC III, and 8 PDAC IV) to characterize the changes in TME composition with PDAC progression through advanced scRNA-seq analysis. We found that as cancer progresses, immune cells gradually become a predominant component in late-stage PDAC. We defined a novel Treg and exhausted T cell signature gene, TNFRSF4. Additionally, we identified a prognostic gene set (RPS10, MIF, MT-ATP6, CSTB, IFI30, NPC2, BTG1, CTSD, FCGR2A, SEC61G, IER3, HSPB1, HMOX1, and ZFP36L1) and differentiated high-risk from low-risk PDAC patients based on median risk score threshold. Based on these findings, we developed a novel prognostic model that identifies poorer prognosis in high-risk groups. Furthermore, our analysis revealed significant interactions between cells at different stages of PDAC and identified three promising therapeutic agents (XR-11576, Ixabepilone, and AMONAFIDE) based on correlated genes. Finally, molecular docking studies validated their potential by confirming stable binding with key protein targets. This study not only provides insights into the evolving TME of PDAC but also offers a new prognostic model and potential therapeutic strategies, contributing to improved management and treatment of this aggressive cancer.
Collapse
Affiliation(s)
- Yongsheng Li
- School of Life Science and Food Engineering, Huaiyin Institute of Technology, Huai'an, China
| | - Zhilong Ding
- Department of Hepatobiliary Surgery, The Affiliated Huaian Hospital of Xuzhou Medical University and Huai'an Second People's Hospital, Huai'an, Jiangsu, China
| | - Tingxin Cheng
- School of Life Science and Food Engineering, Huaiyin Institute of Technology, Huai'an, China
| | - Yihuai Hu
- School of Life Science and Food Engineering, Huaiyin Institute of Technology, Huai'an, China
| | - Fei Zhong
- Department of Laboratory Medicine, The Affiliated Huaian Hospital of Xuzhou Medical University and Huai'an Second People's Hospital, Huai'an, Jiangsu, China.
| | - Shiying Ren
- School of Life Science and Food Engineering, Huaiyin Institute of Technology, Huai'an, China.
| | - Shiyan Wang
- School of Life Science and Food Engineering, Huaiyin Institute of Technology, Huai'an, China.
| |
Collapse
|
3
|
Hou M, Yue M, Han X, Sun T, Zhu Y, Li Z, Han J, Zhao B, Tu M, An Y. Comparative analysis of BAG1 and BAG2: Insights into their structures, functions and implications in disease pathogenesis. Int Immunopharmacol 2024; 143:113369. [PMID: 39405938 DOI: 10.1016/j.intimp.2024.113369] [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: 07/19/2024] [Revised: 09/22/2024] [Accepted: 10/06/2024] [Indexed: 10/30/2024]
Abstract
As BAG family members, Bcl-2 associated athanogene family protein 1 (BAG1) and 2 (BAG2) are implicated in multiple cellular processes, including apoptosis, autophagy, protein folding and homeostasis. Although structurally similar, they considerably differ in many ways. Unlike BAG2, BAG1 has four isoforms (BAG1L, BAG1M, BAG1S and BAG1 p29) displaying different expression features and functional patterns. BAG1 and BAG2 play different cellular functions by interacting with different molecules to participate in the regulation of various diseases, including cancer/tumor and neurodegenerative diseases. Commonly, BAG1 acts as a protective factor to predict a good prognosis of patients with some types of cancer or a risk factor in some other cancers, while BAG2 is regarded as a risk factor to promote cancer/tumor progression. In neurodegenerative diseases, BAG2 commonly acts as a neuroprotective factor. In this review, we summarized the differences in molacular structure and biological function between BAG1 and BAG2, as well as the influences of them on pathogenesis of diseases, and explore the prospects for their clinical therapy application by specifying the activators and inhibitors of BAG1 and BAG2, which might provide a better understanding of the underlying pathogenesis and developing the targeted therapy strategies for diseases.
Collapse
Affiliation(s)
- Mengwen Hou
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Henan University, Kaifeng 475004, China; School of Stomatology, Henan University, Kaifeng 475004, China
| | - Man Yue
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Henan University, Kaifeng 475004, China; School of Stomatology, Henan University, Kaifeng 475004, China
| | - Xu Han
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Henan University, Kaifeng 475004, China; School of Stomatology, Henan University, Kaifeng 475004, China
| | - Tiantian Sun
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Henan University, Kaifeng 475004, China; School of Stomatology, Henan University, Kaifeng 475004, China
| | - Yonghao Zhu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Henan University, Kaifeng 475004, China; School of Stomatology, Henan University, Kaifeng 475004, China
| | - Zhihao Li
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Henan University, Kaifeng 475004, China; Henan Provincial Engineering Center for Tumor Molecular Medicine, Kaifeng Key Laboratory of Cell Signal Transduction, Henan University, Kaifeng 475004, China
| | - Jiayang Han
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Henan University, Kaifeng 475004, China; School of Stomatology, Henan University, Kaifeng 475004, China
| | - Binbin Zhao
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Henan University, Kaifeng 475004, China; School of Stomatology, Henan University, Kaifeng 475004, China
| | - Mengjie Tu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Henan University, Kaifeng 475004, China; School of Stomatology, Henan University, Kaifeng 475004, China
| | - Yang An
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Henan University, Kaifeng 475004, China; Henan Provincial Engineering Center for Tumor Molecular Medicine, Kaifeng Key Laboratory of Cell Signal Transduction, Henan University, Kaifeng 475004, China.
| |
Collapse
|
4
|
Chen Y, Jin C, Cui J, Diao Y, Wang R, Xu R, Yao Z, Wu W, Li X. Single-cell sequencing and bulk RNA data reveal the tumor microenvironment infiltration characteristics of disulfidptosis related genes in breast cancer. J Cancer Res Clin Oncol 2023; 149:12145-12164. [PMID: 37428249 DOI: 10.1007/s00432-023-05109-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Accepted: 06/30/2023] [Indexed: 07/11/2023]
Abstract
BACKGROUND Immunotherapy, represented by immune checkpoint inhibitors, has made significant progress in the treatment of cancer. Numerous studies have demonstrated that antitumor therapies targeting cell death exhibit synergistic effects with immunotherapy. Disulfidptosis is a recently discovered form of cell death, and its potential influence on immunotherapy, similar to other regulated cell death processes, requires further investigation. The prognostic value of disulfidptosis in breast cancer and its role in the immune microenvironment has not been investigated. METHODS High dimensional weighted gene coexpression network analysis (hdWGCNA) and Weighted co-expression network analysis (WGCNA) methods were employed to integrate breast cancer single-cell sequencing data and bulk RNA data. These analyses aimed to identify genes associated with disulfidptosis in breast cancer. Risk assessment signature was constructed using Univariate Cox and least absolute shrinkage and selection operator (LASSO) analyses. RESULTS In this study, we constructed a risk signature by disulfidptosis-related genes to predict overall survival and immunotherapy response in BRCA patients. The risk signature demonstrated robust prognostic power and accurately predicted survival compared to traditional clinicopathological features. It also effectively predicted the response to immunotherapy in patients with breast cancer. Through cell communication analysis in additional single-cell sequencing data, we identified TNFRSF14 as a key regulatory gene. Combining TNFRSF14 targeting and immune checkpoint inhibition to induce disulfidptosis in tumor cells could potentially suppress tumor proliferation and enhance survival in patients with BRCA.
Collapse
Affiliation(s)
- Yongxing Chen
- Department of Epidemiology and Health Statistics, Dalian Medical University, Dalian, China
| | - Chenxin Jin
- Department of Epidemiology and Health Statistics, Dalian Medical University, Dalian, China
| | - Jiaxue Cui
- Department of Epidemiology and Health Statistics, Dalian Medical University, Dalian, China
| | - Yizhuo Diao
- Department of Epidemiology and Health Statistics, Dalian Medical University, Dalian, China
| | - Ruiqi Wang
- Department of Epidemiology and Health Statistics, Dalian Medical University, Dalian, China
| | - Rongxuan Xu
- Department of Epidemiology and Health Statistics, Dalian Medical University, Dalian, China
| | - Zhihan Yao
- Department of Epidemiology and Health Statistics, Dalian Medical University, Dalian, China
| | - Wei Wu
- Department of Epidemiology and Health Statistics, Dalian Medical University, Dalian, China
| | - Xiaofeng Li
- Department of Epidemiology and Health Statistics, Dalian Medical University, Dalian, China.
| |
Collapse
|
5
|
Kumar B, Prasad P, Singh R, Sahu RK, Singh A, Magani SJ, Hedau S. Role of identified proteins in the proteome profiles of CDK4/6 inhibitor-resistant breast cancer cell lines. Mol Omics 2023. [PMID: 36938944 DOI: 10.1039/d2mo00285j] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/09/2023]
Abstract
Abemaciclib (Ab) and palbociclib (Pb) are CDK4/6 inhibitors used to cure advanced breast cancer (BC). However, acquired resistance is a major challenge. The molecular mechanisms and signature proteins of therapy resistance for Ab and Pb drugs need to be explored. Here we developed resistant cells for Ab and Pb drugs in MCF-7 cell lines and explored the mechanisms and signature proteins of therapy resistance in BC. Proteome profiling was performed using the label-free proteome-orbitrap-fusion-MS-MS technique. Gene ontology (GO)-terms, KEGG pathways and network analysis were performed for the proteome data. Drug-resistant cells showed increased drug tolerance, enhanced colony formation potential and an increased gap-healing tendency for the respective drug. Up-regulation of survival genes (BCL-2 and MCL-1) and down-regulation of apoptosis inducers were observed. Drug-resistance markers (MDR-1 and ABCG2 (BCRP)) along with ESR-1, CDK4, CDK6, and cyclin-D1 genes were up-regulated in resistant cells. A total of 237 and 239 proteins were found to be differentially expressed in the Ab and Pb-resistant cells, respectively. Down-regulated proteins induce apoptosis signalling and nucleotide metabolisms and restrict EGFR signalling; however, up-regulated proteins induce Erk, wnt-β-catenin, VEGFR-PI3K-AKT, glucose transportation, and hypoxia signalling pathways and regulate hydrogen peroxide signalling pathways. The panel of identified proteins associated with these pathways might have characteristics of molecular signature and new drug targets for overcoming drug resistance in breast cancer.
Collapse
Affiliation(s)
- Binayak Kumar
- Division of Molecular Oncology, ICMR-National Institute of Cancer Prevention and Research, I-7, Sector-39, Noida, Uttar Pradesh, 201301, India.
| | - Peeyush Prasad
- Department of Research, Sir Ganga Ram Hospital, 110060, New Delhi, India
| | - Ragini Singh
- Division of Molecular Oncology, ICMR-National Institute of Cancer Prevention and Research, I-7, Sector-39, Noida, Uttar Pradesh, 201301, India.
| | - Ram Krishna Sahu
- Division of Molecular Oncology, ICMR-National Institute of Cancer Prevention and Research, I-7, Sector-39, Noida, Uttar Pradesh, 201301, India.
| | - Ashutosh Singh
- Department of Life Sciences, Shiv Nadar University, NH-91, Tahsil-Dadri, Distt-Gautam Budhaa Nagar, Uttar Pradesh, 201314, India.
| | - Srikrishna Jayadev Magani
- Department of Life Sciences, Shiv Nadar University, NH-91, Tahsil-Dadri, Distt-Gautam Budhaa Nagar, Uttar Pradesh, 201314, India.
| | - Suresh Hedau
- Division of Molecular Oncology, ICMR-National Institute of Cancer Prevention and Research, I-7, Sector-39, Noida, Uttar Pradesh, 201301, India.
| |
Collapse
|
6
|
Han M, Sun Y, Zhao W, Xiang G, Wang X, Jiang Z, Xue Z, Zhou W. Comprehensive characterization of TNFSF14/LIGHT with implications in prognosis and immunotherapy of human gliomas. Front Immunol 2022; 13:1025286. [PMID: 36341396 PMCID: PMC9632349 DOI: 10.3389/fimmu.2022.1025286] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Accepted: 09/26/2022] [Indexed: 07/02/2024] Open
Abstract
Glioblastoma multiforme (GBM) is a common central neural system malignant tumor among adults. Alongside its microscopic spread, immunosuppression in the tumor microenvironment also induces its refractoriness, which makes immunotherapy for GBM particularly important. Unfortunately, traditional immune checkpoint inhibitors (ICIs) often show limited therapeutic effects in GBM clinical trials, and new therapeutic strategies or targets are urgently needed. TNFSF14/LIGHT is a novel immune checkpoint molecule that plays essential roles in both innate and acquired immunity. Despite recent advances in our understanding of the function of TNFSF14/LIGHT in a variety of cancer types, the clinical and immunological importance of TNFSF14/LIGHT in human gliomas has not been fully explained. Here, we employed a comprehensive in silico analysis with publicly available data to analyze the molecular and immune characteristics of TNFSF14/LIGHT to explore its feasibility as an immunotherapy target. Totally, 2215 glioma cases were enrolled in the current study. Immunohistochemistry staining based on patient tissues (n = 34) was performed for the validation. TNFSF14/LIGHT was expressed higher in higher-WHO-grade gliomas and mesenchymal subtypes, and it was sensitive as a prognostic marker in GBM and low-grade glioma (LGG). A nomogram prognostic model was established based on TNFSF14/LIGHT expression together with other risk factors. Additionally, Gene Ontology and pathway analysis revealed that TNFSF14/LIGHT participated in T-cell activities and inflammatory processes. Moreover, analysis based on the structure and interactions of TNFSF14/LIGHT revealed its mutation sites in tumors as well as crucial interacting proteins. Analysis of IMvigor210 indicated the role of TNFSF14/LIGHT in immunotherapy. Altogether, our results reveal an underlying role of TNFSF14/LIGHT as an immunotherapy target in GBM.
Collapse
Affiliation(s)
- Mingzhi Han
- Department of Neurosurgery, Qilu Hospital, Cheeloo College of Medicine and Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, China
- Jinan Microecological Biomedicine Shandong Laboratory, Jinan, China
- Shandong Key Laboratory of Brain Function Remodeling, Jinan, China
- Medical Integration and Practice Center, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Yanfei Sun
- Department of Neurosurgery, Qilu Hospital, Cheeloo College of Medicine and Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, China
| | - Wenbo Zhao
- Department of Neurosurgery, Qilu Hospital, Cheeloo College of Medicine and Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, China
| | - Guo Xiang
- Department of Neurosurgery, Qilu Hospital, Cheeloo College of Medicine and Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, China
| | - Xu Wang
- Department of Neurosurgery, Qilu Hospital, Cheeloo College of Medicine and Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, China
| | - Zheng Jiang
- Department of Neurosurgery, Qilu Hospital, Cheeloo College of Medicine and Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, China
| | - Zhiwei Xue
- Department of Neurosurgery, Qilu Hospital, Cheeloo College of Medicine and Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, China
| | - Wei Zhou
- Department of Radiation Oncology, Qilu Hospital, Shandong University, Jinan, China
| |
Collapse
|
7
|
Fu CC, Huang L, Xu LF, Jiang LH, Li HL, Liao S, Yue J, Lian C, Yang XG, Long YM. Serological biomarkers in autoimmune GFAP astrocytopathy. Front Immunol 2022; 13:957361. [PMID: 35983033 PMCID: PMC9378990 DOI: 10.3389/fimmu.2022.957361] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Accepted: 07/04/2022] [Indexed: 11/13/2022] Open
Abstract
Autoimmune glial fibrillary acidic protein astrocytopathy (GFAP-A) is a newly defined meningoencephalomyelitis. The pathogenesis of GFAP-A is not well understood. The present study measured the expression levels of 200 serological cytokines in GFAP-A patients, NMOSD patients and healthy controls (HCs). The correlations between serum cytokine levels and clinical information in GFAP-A patients were analyzed. A total of 147 serological proteins were differentially expressed in GFAP-A patients compared to HCs, and 33 of these proteins were not observed in NMOSD patients. Serum levels of EG-VEGF negatively correlated with GFAP antibody titers, MIP-3 alpha positively correlated with clinical severity in GFAP-A patients, and LIGHT positively correlated with WBC counts and protein levels in the CSF of GFAP-A patients. These results suggest that GFAP and AQP4 astrocytopathy share some common pathology related to TNF signaling. Serum MIP 3 alpha may be a biomarker to assess clinical severity and a potential target for therapy of autoimmune GFAP astrocytopathy.
Collapse
Affiliation(s)
- Cong-Cong Fu
- Department of Neurology, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
- Department of Neurology, Institute of Neuroscience, Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Lu Huang
- Department of Neurology, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
- Department of Neurology, Institute of Neuroscience, Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Lu-Fen Xu
- Department of Neurology, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
- Department of Neurology, Institute of Neuroscience, Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Li-Hong Jiang
- Department of Neurology, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
- Department of Neurology, Institute of Neuroscience, Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Hui-Lu Li
- Department of Neurology, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
- Department of Neurology, Institute of Neuroscience, Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Sha Liao
- Department of Neurology, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
- Department of Neurology, Institute of Neuroscience, Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Jiajia Yue
- Department of Neurology, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
- Department of Neurology, Institute of Neuroscience, Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Chun Lian
- Department of Neurology, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
- Department of Neurology, Institute of Neuroscience, Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Xin-Guang Yang
- Department of Neurology, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
- Department of Neurology, Institute of Neuroscience, Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
- Department of Neurology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
- *Correspondence: You-Ming Long, ; Xin-Guang Yang,
| | - You-Ming Long
- Department of Neurology, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
- Department of Neurology, Institute of Neuroscience, Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
- *Correspondence: You-Ming Long, ; Xin-Guang Yang,
| |
Collapse
|
8
|
HES5-mediated repression of LIGHT transcription may contribute to apoptosis in hepatocytes. Cell Death Discov 2021; 7:308. [PMID: 34689159 PMCID: PMC8542050 DOI: 10.1038/s41420-021-00707-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Revised: 10/04/2021] [Accepted: 10/06/2021] [Indexed: 11/26/2022] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) is prototypical form of metabolic syndrome and has become a global pandemic. Hepatocytes undergo apoptosis in the pathogenesis of NAFLD. We report that the lymphokine LIGHT/TNFSF14 was upregulated in the murine NAFLD livers and in hepatocytes treated with free fatty acids (palmitate, PA). LIGHT knockdown or neutralization attenuated PA-induced apoptosis of hepatocytes. Similarly, knockdown or blockade of LTβR, the receptor for LIGHT, ameliorated apoptosis in hepatocytes exposed to PA. Ingenuity pathway analysis (IPA) revealed several Notch-related transcription factors as upstream regulators of LIGHT, of which HES5 expression was downregulated paralleling LIGHT induction in the pathogenesis of NAFLD. HES5 knockdown enhanced whereas HES5 over-expression weakened LIGHT induction in hepatocytes. HES5 was found to directly bind to the LIGHT promoter and repress LIGHT transcription. Mechanistically, HES5 interacted with SIRT1 to deacetylate histone H3/H4 on the LIGHT promoter to repress LIGHT transcription. SIRT1 knockdown or inhibition offset the effect of HES5 over-expression on LIGHT transcription and hepatocyte apoptosis. In conclusion, our data unveil a novel mechanism that might contribute to excessive apoptosis in hepatocyte exposed to free fatty acids.
Collapse
|
9
|
Silva C, Andrade N, Guimarães JT, Patrício E, Martel F. The in vitro effect of the diabetes-associated markers insulin, leptin and oxidative stress on cellular characteristics promoting breast cancer progression is GLUT1-dependent. Eur J Pharmacol 2021; 898:173980. [PMID: 33647254 DOI: 10.1016/j.ejphar.2021.173980] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2020] [Revised: 02/15/2021] [Accepted: 02/23/2021] [Indexed: 01/09/2023]
Abstract
Obesity and type 2 diabetes mellitus (T2DM) associate with increased incidence and mortality from many cancers, including breast cancer. The mechanisms involved in this relation remain poorly understood. Our study aimed to investigate the in vitro effect of high levels of glucose, insulin, leptin, TNF-α, INF-γ and oxidative stress (induced with tert-butylhydroperoxide (TBH)), which are associated with T2DM, upon glucose uptake by breast cancer (MCF-7 and MDA-MB-231) and non-cancer (MCF-12A) cells and to correlate this effect with their effects upon cellular characteristics associated with cancer progression (cell proliferation, viability, migration, angiogenesis and apoptosis). 3H-DG uptake was markedly inhibited by a selective GLUT1 inhibitor (BAY-876) in all cell lines, proving that 3H-DG uptake is mainly GLUT1-mediated. TBH (2.5 μM), insulin (50 nM), leptin (500 ng/ml) and INF-y (100 ng/ml) stimulate GLUT1-mediated 3H-DG (1 mM) uptake by both ER-positive and triple-negative breast cancer cell lines. TBH and leptin, but not insulin and INF-γ, increase GLUT1 mRNA levels. Insulin and leptin (in both ER-positive and triple-negative breast cancer cell lines) and TBH (in the triple-negative cell line) have a proproliferative effect and leptin possesses a cytoprotective effect in both breast cancer cell lines that can contribute to cancer progression. The effects of TBH, insulin, leptin and INF-γ upon breast cancer cell proliferation and viability are GLUT1-dependent. In conclusion, T2DM-associated characteristics induce changes in GLUT1-mediated glucose uptake that can contribute to cancer progression. Moreover, we conclude that BAY-876 can be a strong candidate for development of a new effective anticancer agent against breast cancer.
Collapse
Affiliation(s)
- Cláudia Silva
- Unit of Biochemistry, Department of Biomedicine, Faculty of Medicine, University of Porto, Porto, Portugal; Instituto de Investigação e Inovação Em Saúde (i3S), University of Porto, Porto, Portugal
| | - Nelson Andrade
- Unit of Biochemistry, Department of Biomedicine, Faculty of Medicine, University of Porto, Porto, Portugal; Instituto de Investigação e Inovação Em Saúde (i3S), University of Porto, Porto, Portugal; REQUIMTE/LAQV, Department of Chemical Sciences, Faculty of Pharmacy, University of Porto, Portugal
| | - João Tiago Guimarães
- Unit of Biochemistry, Department of Biomedicine, Faculty of Medicine, University of Porto, Porto, Portugal; Department of Clinical Pathology, São João Hospital Centre, Porto, Portugal; Institute of Public Health, University of Porto, Porto, Portugal
| | - Emília Patrício
- Department of Clinical Pathology, São João Hospital Centre, Porto, Portugal
| | - Fátima Martel
- Unit of Biochemistry, Department of Biomedicine, Faculty of Medicine, University of Porto, Porto, Portugal; Instituto de Investigação e Inovação Em Saúde (i3S), University of Porto, Porto, Portugal.
| |
Collapse
|
10
|
Skeate JG, Otsmaa ME, Prins R, Fernandez DJ, Da Silva DM, Kast WM. TNFSF14: LIGHTing the Way for Effective Cancer Immunotherapy. Front Immunol 2020; 11:922. [PMID: 32499782 PMCID: PMC7243824 DOI: 10.3389/fimmu.2020.00922] [Citation(s) in RCA: 62] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2020] [Accepted: 04/21/2020] [Indexed: 12/21/2022] Open
Abstract
Tumor necrosis factor superfamily member 14 (LIGHT) has been in pre-clinical development for over a decade and shows promise as a modality of enhancing treatment approaches in the field of cancer immunotherapy. To date, LIGHT has been used to combat cancer in multiple tumor models where it can be combined with other immunotherapy modalities to clear established solid tumors as well as treat metastatic events. When LIGHT molecules are delivered to or expressed within tumors they cause significant changes in the tumor microenvironment that are primarily driven through vascular normalization and generation of tertiary lymphoid structures. These changes can synergize with methods that induce or support anti-tumor immune responses, such as checkpoint inhibitors and/or tumor vaccines, to greatly improve immunotherapeutic strategies against cancer. While investigators have utilized multiple vectors to LIGHT-up tumor tissues, there are still improvements needed and components to be found within a human tumor microenvironment that may impede translational efforts. This review addresses the current state of this field.
Collapse
Affiliation(s)
- Joseph G Skeate
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| | - Mikk E Otsmaa
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| | - Ruben Prins
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| | - Daniel J Fernandez
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| | - Diane M Da Silva
- Department of Obstetrics and Gynecology, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States.,Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA, United States
| | - W Martin Kast
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States.,Department of Obstetrics and Gynecology, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States.,Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA, United States
| |
Collapse
|
11
|
The Nuclear Protein HOXB13 Enhances Methylmercury Toxicity by Inducing Oncostatin M and Promoting Its Binding to TNFR3 in Cultured Cells. Cells 2019; 9:cells9010045. [PMID: 31878059 PMCID: PMC7017003 DOI: 10.3390/cells9010045] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2019] [Revised: 12/20/2019] [Accepted: 12/21/2019] [Indexed: 02/06/2023] Open
Abstract
Homeobox protein B13 (HOXB13), a transcription factor, is related to methylmercury toxicity; however, the downstream factors involved in enhancing methylmercury toxicity remain unknown. We performed microarray analysis to search for downstream factors whose expression is induced by methylmercury via HOXB13 in human embryonic kidney cells (HEK293), which are useful model cells for analyzing molecular mechanisms. Methylmercury induced the expression of oncostatin M (OSM), a cytokine of the interleukin-6 family, and this was markedly suppressed by HOXB13 knockdown. OSM knockdown also conferred resistance to methylmercury in HEK293 cells, and no added methylmercury resistance was observed when both HOXB13 and OSM were knocked down. Binding of HOXB13 to the OSM gene promoter was increased by methylmercury, indicating the involvement of HOXB13 in the enhancement of its toxicity. Because addition of recombinant OSM to the medium enhanced methylmercury toxicity in OSM-knockdown cells, extracellularly released OSM was believed to enhance methylmercury toxicity via membrane receptors. We discovered tumor necrosis factor receptor (TNF) receptor 3 (TNFR3) to be a potential candidate involved in the enhancement of methylmercury toxicity by OSM. This toxicity mechanism was also confirmed in mouse neuronal stem cells. We report, for the first time, that HOXB13 is involved in enhancement of methylmercury toxicity via OSM-expression induction and that the synthesized OSM causes cell death by binding to TNFR3 extracellularly.
Collapse
|
12
|
Dou JW, Shang RG, Lei XQ, Li KL, Guo ZZ, Ye K, Yang XJ, Li YW, Zhou YY, Yao J, Huang Q. Total saponins of Bolbostemma paniculatum (maxim.) Franquet exert antitumor activity against MDA-MB-231 human breast cancer cells via inhibiting PI3K/Akt/mTOR pathway. BMC COMPLEMENTARY AND ALTERNATIVE MEDICINE 2019; 19:304. [PMID: 31703679 PMCID: PMC6842232 DOI: 10.1186/s12906-019-2708-0] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/30/2019] [Accepted: 10/02/2019] [Indexed: 01/21/2023]
Abstract
BACKGROUND The aim of the present study was to examine the effects of the Bolbostemma paniculatum (Maxim.) Franquet (BP) active compound, BP total saponins (BPTS), on MDA-MB-231 cells, and investigate the underlying mechanism regarding BPTS-mediated attenuation of the PI3K/Akt/mTOR pathway. METHODS The effect of BPTS on cytotoxicity, induction of apoptosis and migration on MDA-MB-231 cells at three different concentrations was investigated. A CCK-8 assay, wound-healing assay and flow cytometry were used to demonstrate the effects of BPTS. Additionally, expression of the primary members of the PI3K/Akt/mTOR signaling pathway was assessed using western blotting. To verify the underlying mechanisms, a PI3K inhibitor and an mTOR inhibitor were used. RESULTS BPTS inhibited proliferation of MDA-MB-231 cells with an IC50 value of 10 μg/mL at 48 h. BPTS inhibited migration of MDA-MB-231 cells, and the western blot results demonstrated that BPTS reduced p-PI3K, p-Akt and p-mTOR protein expression levels in MDA-MB-231 cells. Additionally, the results were confirmed using a PI3K inhibitor and an mTOR inhibitor. BPTS decreased proliferation and migration of MDA-MB-231 cells possibly through inhibiting the PI3K/Akt/mTOR signaling pathway. CONCLUSIONS The results highlight the therapeutic potential of BPTS for treating patients with triple-negative breast cancer.
Collapse
Affiliation(s)
- Jian-Wei Dou
- School of Pharmacy, Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, People's Republic of China
- Shaanxi Key Laboratory of "Qiyao" Resources And Anti-tumor Activities, Xi'an, Shaanxi, 710061, People's Republic of China
| | - Rong-Guo Shang
- School of Pharmacy, Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, People's Republic of China
- Shaanxi Key Laboratory of "Qiyao" Resources And Anti-tumor Activities, Xi'an, Shaanxi, 710061, People's Republic of China
| | - Xiao-Qin Lei
- Department of Ophthalmology, Affiliated Guangren Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710004, People's Republic of China
- Department of Ophthalmology, Xi'an No.4 Hospital, Xi'an, Shaanxi, 710004, People's Republic of China
| | - Kang-Le Li
- School of Pharmacy, Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, People's Republic of China
- Shaanxi Key Laboratory of "Qiyao" Resources And Anti-tumor Activities, Xi'an, Shaanxi, 710061, People's Republic of China
| | - Zhan-Zi Guo
- School of Basic Medicine, Gansu University of Chinese Medicine, Lanzhou, Gansu, 730000, People's Republic of China
| | - Kai Ye
- School of Basic Medicine, Gansu University of Chinese Medicine, Lanzhou, Gansu, 730000, People's Republic of China
| | - Xiao-Juan Yang
- School of Basic Medicine, Gansu University of Chinese Medicine, Lanzhou, Gansu, 730000, People's Republic of China
| | - Yu-Wei Li
- Department of Ophthalmology, Affiliated Guangren Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710004, People's Republic of China
- Department of Ophthalmology, Xi'an No.4 Hospital, Xi'an, Shaanxi, 710004, People's Republic of China
| | - Yun-Yun Zhou
- Department of Ophthalmology, Affiliated Guangren Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710004, People's Republic of China
- Department of Ophthalmology, Xi'an No.4 Hospital, Xi'an, Shaanxi, 710004, People's Republic of China
| | - Jia Yao
- Xi'an Hospital of Traditional Chinese Medicine Affiliated to Shaanxi University of Chinese Medicine, Xi'an, Shaanxi, 710021, People's Republic of China
| | - Qian Huang
- Xi'an Hospital of Traditional Chinese Medicine Affiliated to Shaanxi University of Chinese Medicine, Xi'an, Shaanxi, 710021, People's Republic of China.
| |
Collapse
|
13
|
Alabdulmon W, Alhomaidan HT, Rasheed Z, Madar IH, Alasmael N, Alkhatib S, Al Ssadh H. CD74 a Potential Therapeutic Target for Breast Cancer Therapy: Interferon Gamma Up-regulates its Expression in CAMA-1 and MDA-MB-231 Cancer Cells. ACTA ACUST UNITED AC 2018. [DOI: 10.3923/ijcr.2018.58.69] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
|
14
|
High throughput screening of cytokines, chemokines and matrix metalloproteinases in wound fluid induced by mammary surgery. Oncotarget 2016; 6:29296-310. [PMID: 26313265 PMCID: PMC4745727 DOI: 10.18632/oncotarget.4828] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2015] [Accepted: 07/29/2015] [Indexed: 12/28/2022] Open
Abstract
Objective To clarify the composition of wound fluid (WF) and investigate the impact of WF on breast cancer cell lines. Methods The proliferation and migration of WF-treated breast cancer cells MDA-MB-231 and MCF-7 were assessed with colony formation test, MTT cell proliferation test and scratch wound test. The quantitative profiles of WF were analyzed using Bio-Plex Pro kits. Results The proliferation and migration of WF-treated breast cancer cells were significantly higher than that of untreated cells. Fifteen cytokines, 29 chemokines and 9 matrix metalloproteinases (MMPs) were assessed in WF. The concentrations of these factors were influenced by post-surgery days, neoadjuvant chemotherapy (NAC), TNM stage, pathological type and molecular subtype. The WF harvested from patients underwent NAC showed significant higher profiles of interleukin-1β (IL-1β), IL-4, IL-6, IL-17F, IL-21, IL-23, IL-25, IL-31, Interferon γ (IFNγ), CD40 ligand (CD40L), tumor necrosis factor α (TNFα), CXCL1, CXCL2, CXCL5, CCL3, CCL7 and CCL20. Conclusions Surgery-induced WF promotes the proliferation and migration of breast cancer cells. The composition of WF is influenced by various clinical features and provides potential therapeutic targets to control local recurrence and tumor progression.
Collapse
|
15
|
Zheng QY, Cao ZH, Hu XB, Li GQ, Dong SF, Xu GL, Zhang KQ. LIGHT/IFN-γ triggers β cells apoptosis via NF-κB/Bcl2-dependent mitochondrial pathway. J Cell Mol Med 2016; 20:1861-71. [PMID: 27241100 PMCID: PMC5020636 DOI: 10.1111/jcmm.12876] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2015] [Accepted: 03/08/2016] [Indexed: 01/14/2023] Open
Abstract
LIGHT recruits and activates naive T cells in the islets at the onset of diabetes. IFN‐γ secreted by activated T lymphocytes is involved in beta cell apoptosis. However, whether LIGHT sensitizes IFNγ‐induced beta cells destruction remains unclear. In this study, we used the murine beta cell line MIN6 and primary islet cells as models for investigating the underlying cellular mechanisms involved in LIGHT/IFNγ – induced pancreatic beta cell destruction. LIGHT and IFN‐γ synergistically reduced MIN6 and primary islet cells viability; decreased cell viability was due to apoptosis, as demonstrated by a significant increase in Annexin V+ cell percentage, detected by flow cytometry. In addition to marked increases in cytochrome c release and NF‐κB activation, the combination of LIGHT and IFN‐γ caused an obvious decrease in expression of the anti‐apoptotic proteins Bcl‐2 and Bcl‐xL, but an increase in expression of the pro‐apoptotic proteins Bak and Bax in MIN6 cells. Accordingly, LIGHT deficiency led to a decrease in NF‐κB activation and Bak expression, and peri‐insulitis in non‐obese diabetes mice. Inhibition of NF‐κB activation with the specific NF‐κB inhibitor, PDTC (pyrrolidine dithiocarbamate), reversed Bcl‐xL down‐regulation and Bax up‐regulation, and led to a significant increase in LIGHT‐ and IFN‐γ‐treated cell viability. Moreover, cleaved caspase‐9, ‐3, and PARP (poly (ADP‐ribose) polymerase) were observed after LIGHT and IFN‐γ treatment. Pretreatment with caspase inhibitors remarkably attenuated LIGHT‐ and IFNγ‐induced cell apoptosis. Taken together, our results indicate that LIGHT signalling pathway combined with IFN‐γ induces beta cells apoptosis via an NF‐κB/Bcl2‐dependent mitochondrial pathway.
Collapse
Affiliation(s)
- Quan-You Zheng
- Department of Nephrology, Southwest Hospital, Third Military Medical University, Chongqing, China
| | - Zhao-Hui Cao
- Department of Nephrology, Southwest Hospital, Third Military Medical University, Chongqing, China.,Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, School of Pharmacy and Biosciences, University of South China, Hengyang, China.,Department of Immunology, School of Basic Medical Sciences, Third Military Medical University, Chongqing, China
| | - Xiao-Bo Hu
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, School of Pharmacy and Biosciences, University of South China, Hengyang, China
| | - Gui-Qing Li
- Department of Immunology, School of Basic Medical Sciences, Third Military Medical University, Chongqing, China
| | - Shi-Fang Dong
- Department of Immunology, School of Basic Medical Sciences, Third Military Medical University, Chongqing, China
| | - Gui-Lian Xu
- Department of Immunology, School of Basic Medical Sciences, Third Military Medical University, Chongqing, China
| | - Ke-Qin Zhang
- Department of Nephrology, Southwest Hospital, Third Military Medical University, Chongqing, China.
| |
Collapse
|
16
|
Kubatka P, Kapinová A, Kružliak P, Kello M, Výbohová D, Kajo K, Novák M, Chripková M, Adamkov M, Péč M, Mojžiš J, Bojková B, Kassayová M, Stollárová N, Dobrota D. Antineoplastic effects of Chlorella pyrenoidosa in the breast cancer model. Nutrition 2014; 31:560-9. [PMID: 25770318 DOI: 10.1016/j.nut.2014.08.010] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2013] [Revised: 05/30/2014] [Accepted: 08/25/2014] [Indexed: 02/05/2023]
Abstract
OBJECTIVES There has been considerable interest in both clinical and preclinical research about the role of phytochemicals in the reduction of risk for cancer in humans. The aim of this study was to determine the antineoplastic effects of Chlorella pyrenoidosa in experimental breast cancer in vivo and in vitro. METHODS In this experiment, the antineoplastic effects of C. pyrenoidosa in the chemoprevention of N-methyl-N-nitrosourea-induced mammary carcinogenesis in female rats were evaluated. Chlorella powder was administered through diet at concentrations of 0.3% and 3%. The experiment was terminated 14 wk after carcinogen administration. At autopsy, mammary tumors were removed and prepared for histopathological and immunohistochemical analysis. In vitro cytotoxicity assay, parameters of apoptosis, and proliferation after chlorella treatment in human breast adenocarcinoma (MCF-7) cells were carried out. RESULTS Basic parameters of experimental carcinogenesis, mechanism of action (biomarkers of apoptosis, proliferation, and angiogenesis), chosen metabolic variables, and side effects after long-term chlorella treatment in animals were assessed. Chlorella at higher concentration suppressed tumor frequency by 61% (P < 0.02) and lengthened tumor latency by 12.5 d (P < 0.02) in comparison with the controls. Immunohistochemical analysis of rat tumor cells showed caspase-7 expression increase by 73.5% (P < 0.001) and vascular endothelial growth factor receptor-2 expression decrease by 19% (P = 0.07) after chlorella treatment. In a parallel in vitro study, chlorella significantly decreased survival of MCF-7 cells in a dose-dependent manner. In chlorella-treated MCF-7 cells, a significant increase in cells having sub-G0/G1 DNA content and significant increase of early apoptotic and late apoptotic/necrotic cells after annexin V/PI staining assay were found. Decreases in mitochondrial membrane potential and increasing reactive oxygen species generation were observed in the chlorella-treated MCF-7 cells. CONCLUSIONS This study is the first report on the antineoplastic effects of C. pyrenoidosa in experimental breast cancer in vivo and in vitro.
Collapse
Affiliation(s)
- Peter Kubatka
- Department of Medical Biology, Jessenius Faculty of Medicine, Comenius University, Martin, Slovakia.
| | - Andrea Kapinová
- Department of Medical Biochemistry, Jessenius Faculty of Medicine, Comenius University, Martin, Slovakia
| | - Peter Kružliak
- International Clinical Research Center, St. Anne's University Hospital and Masaryk University, Brno, Czech Republic
| | - Martin Kello
- Department of Pharmacology, Faculty of Medicine, P. J. Šafárik University, Košice, Slovakia
| | - Desanka Výbohová
- Department of Anatomy, Jessenius Faculty of Medicine, Comenius University, Martin, Slovakia
| | - Karol Kajo
- Department of Pathology, Slovak Medical University and St. Elisabeth Oncology Institute, Bratislava, Slovakia
| | - Miroslav Novák
- International Clinical Research Center, St. Anne's University Hospital and Masaryk University, Brno, Czech Republic
| | - Martina Chripková
- Department of Pharmacology, Faculty of Medicine, P. J. Šafárik University, Košice, Slovakia
| | - Marián Adamkov
- Department of Histology and Embryology, Jessenius Faculty of Medicine, Comenius University, Martin, Slovakia
| | - Martin Péč
- Department of Medical Biology, Jessenius Faculty of Medicine, Comenius University, Martin, Slovakia
| | - Ján Mojžiš
- Department of Pharmacology, Faculty of Medicine, P. J. Šafárik University, Košice, Slovakia
| | - Bianka Bojková
- Department of Animal Physiology, Institute of Biological and Ecological Sciences, Faculty of Science, P. J. Šafárik University, Košice, Slovakia
| | - Monika Kassayová
- Department of Animal Physiology, Institute of Biological and Ecological Sciences, Faculty of Science, P. J. Šafárik University, Košice, Slovakia
| | - Nadežda Stollárová
- Department of Biology and Ecology, Faculty of Education, Catholic University in Ružomberok, Ružomberok, Slovakia
| | - Dušan Dobrota
- Department of Medical Biochemistry, Jessenius Faculty of Medicine, Comenius University, Martin, Slovakia
| |
Collapse
|
17
|
Liu LH, Zhou YJ, Ding L, Zhang SZ, Sun J, Cao JG. Induction of apoptosis by VB1 in breast cancer cells: the role of reactive oxygen species and Bcl-2 family proteins. Int J Mol Med 2013; 33:423-30. [PMID: 24276280 DOI: 10.3892/ijmm.2013.1567] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2013] [Accepted: 11/18/2013] [Indexed: 11/05/2022] Open
Abstract
We have previously reported that the EVn-50 mixture of vitexins (lignan compounds) containing the purified vitexin (neolignan) compound, 6-hydroxy-4(4-hydroxy-3-methoxyphenyl)-3-hydroxymethyl‑7-methoxy-3,4-dihydro-2-naphthaldehyde, termed VB1, exhibits potent anticancer activity through the induction of apoptosis in several types of cancer cells, including MDA-MB‑231 cells. However, the exact molecular mechanisms by which VB1 induces apoptosis in MDA-MB‑231 cells have not yet been fully elucidated. In this study, to our knowledge, we provide for the first time mechanistic evidence that VB1-induced apoptosis in the human breast cancer line, MDA-MB-231, is associated with the generation of reactive oxygen species (ROS), the activation of caspases and the modulation of the expression of myeloid leukemia cell differentiation protein 1 (Mcl‑1), B cell lymphoma‑2 (Bcl-2) and Bcl-2-associated X (Bax) proteins. The silencing of Mcl-1 by RNA interference enhanced VB1-induced apoptosis. In addition, VB1 did not induce ROS generation or apoptosis in the immortalized non‑cancerous breast cell line, MCF-10A. Our findings reveal a novel mechanism underlying VB1-induced apoptosis, and highlight VB1 as a promising candidate for the therapy of human breast cancer.
Collapse
Affiliation(s)
- Li-Hua Liu
- The First Hospital of Changsha, Changsha, Hunan 410005, P.R. China
| | - Ying-Jun Zhou
- School of Pharmacy, Central South University, Changsha, Hunan 410013, P.R. China
| | - Lan Ding
- The First Hospital of Changsha, Changsha, Hunan 410005, P.R. China
| | - Shun-Zhi Zhang
- The First Hospital of Changsha, Changsha, Hunan 410005, P.R. China
| | - Ji Sun
- The First Hospital of Changsha, Changsha, Hunan 410005, P.R. China
| | - Jian-Guo Cao
- Medical College, Hunan Normal University, Changsha, Hunan 410013, P.R. China
| |
Collapse
|
18
|
Maña P, Liñares D, Silva DG, Fordham S, Scheu S, Pfeffer K, Staykova M, Bertram EM. LIGHT (TNFSF14/CD258) Is a Decisive Factor for Recovery from Experimental Autoimmune Encephalomyelitis. THE JOURNAL OF IMMUNOLOGY 2013; 191:154-63. [DOI: 10.4049/jimmunol.1203016] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
|
19
|
KUO SHOUJEN, LIN HUIYI, CHIEN SUYU, CHEN DARREN. SIRT1 suppresses breast cancer growth through downregulation of the Bcl-2 protein. Oncol Rep 2013; 30:125-30. [DOI: 10.3892/or.2013.2470] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2012] [Accepted: 03/06/2013] [Indexed: 11/05/2022] Open
|
20
|
Hu X, Zimmerman MA, Bardhan K, Yang D, Waller JL, Liles GB, Lee JR, Pollock R, Lev D, Ware CF, Garber E, Bailly V, Browning JL, Liu K. Lymphotoxin β receptor mediates caspase-dependent tumor cell apoptosis in vitro and tumor suppression in vivo despite induction of NF-κB activation. Carcinogenesis 2013; 34:1105-14. [PMID: 23349015 DOI: 10.1093/carcin/bgt014] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Affiliation(s)
- Xiaolin Hu
- Department of Biochemistry and Molecular Biology, Medical College of Georgia, Georgia Health Sciences University, Augusta, GA 30912, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
21
|
Simpson NE, Tryndyak VP, Pogribna M, Beland FA, Pogribny IP. Modifying metabolically sensitive histone marks by inhibiting glutamine metabolism affects gene expression and alters cancer cell phenotype. Epigenetics 2012; 7:1413-20. [PMID: 23117580 PMCID: PMC3528696 DOI: 10.4161/epi.22713] [Citation(s) in RCA: 61] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
The interplay of metabolism and epigenetic regulatory mechanisms has become a focal point for a better understanding of cancer development and progression. In this study, we have acquired data supporting previous observations that demonstrate glutamine metabolism affects histone modifications in human breast cancer cell lines. Treatment of non-invasive epithelial (T-47D and MDA-MB-361) and invasive mesenchymal (MDA-MB-231 and Hs-578T) breast cancer cell lines with the glutaminase inhibitor, Compound 968, resulted in cytotoxicity in all cell lines, with the greatest effect being observed in MDA-MB-231 breast cancer cells. Compound 968-treatment induced significant downregulation of 20 critical cancer-related genes, the majority of which are anti-apoptotic and/or promote metastasis, including AKT, BCL2, BCL2L1, CCND1, CDKN3, ERBB2, ETS1, E2F1, JUN, KITLG, MYB, and MYC. Histone H3K4me3, a mark of transcriptional activation, was reduced at the promoters of all but one of these critical cancer genes. The decrease in histone H3K4me3 at global and gene-specific levels correlated with reduced expression of SETD1 and ASH2L, genes encoding the histone H3K4 methyltransferase complex. Further, the expression of other epigenetic regulatory genes, known to be downregulated during apoptosis (e.g., DNMT1, DNMT3B, SETD1 and SIRT1), was also downregulated by Compound 968. These changes in gene expression and histone modifications were accompanied by the activation of apoptosis, and decreased invasiveness and resistance of MDA-MB-231 cells to chemotherapeutic drug doxorubicin. The results of this study provide evidence to a link between cytotoxicity caused by inhibiting glutamine metabolism with alterations of the epigenome of breast cancer cells and suggest that modification of intracellular metabolism may enhance the efficiency of epigenetic therapy.
Collapse
Affiliation(s)
- Natalie E. Simpson
- Division of Biochemical Toxicology; National Center for Toxicological Research; Jefferson, AR USA
| | - Volodymyr P. Tryndyak
- Division of Biochemical Toxicology; National Center for Toxicological Research; Jefferson, AR USA
| | - Marta Pogribna
- Division of Biochemical Toxicology; National Center for Toxicological Research; Jefferson, AR USA
| | - Frederick A. Beland
- Division of Biochemical Toxicology; National Center for Toxicological Research; Jefferson, AR USA
| | - Igor P. Pogribny
- Division of Biochemical Toxicology; National Center for Toxicological Research; Jefferson, AR USA
| |
Collapse
|
22
|
KIM SOJUNG, KIM HYEONGJIN, KIM HYERI, LEE SEUNGHO, CHO SUNGDAE, CHOI CHANGSUN, NAM JEONGSEOK, JUNG JIYOUN. Antitumor actions of baicalein and wogonin in HT-29 human colorectal cancer cells. Mol Med Rep 2012; 6:1443-9. [DOI: 10.3892/mmr.2012.1085] [Citation(s) in RCA: 73] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2012] [Accepted: 09/11/2012] [Indexed: 02/06/2023] Open
|
23
|
Pasero C, Barbarat B, Just-Landi S, Bernard A, Aurran-Schleinitz T, Rey J, Eldering E, Truneh A, Costello RT, Olive D. A role for HVEM, but not lymphotoxin-beta receptor, in LIGHT-induced tumor cell death and chemokine production. Eur J Immunol 2009; 39:2502-14. [PMID: 19701890 DOI: 10.1002/eji.200939069] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
The TNF member LIGHT also known as TL4 or TNFSF14) can play a major role in cancer control via its two receptors; it induces tumor cell death through lymphotoxin-beta receptor (LT-betaR) and ligation to the herpes virus entry mediator (HVEM) amplifies the immune response. By studying the effect of LIGHT in the transcriptional profile of a lymphoid malignancy, we found that HVEM, but not LT-betaR, stimulation induces a significant increase in the expression of chemokine genes such as IL-8, and an unexpected upregulation of apoptotic genes. This had functional consequences, since LIGHT, or HVEM mAb, thus far known to costimulate T- and B-cell activation, induced chronic lymphocytic leukemia cell death. Many of the mediators involved were identified here, with an apoptotic pathway as demonstrated by caspases activation, decrease in mitochondrial membrane potential, upregulation of the pro-apoptotic protein Bax, but also a role of TRAIL. Moreover, HVEM induced endogenous TNF-alpha production and TNF-alpha enhanced HVEM-mediated cell death. HVEM function was mainly dependent on LIGHT, since other ligands like HSV-glycoprotein D and B and T lymphocyte attenuator were essentially ineffective. In conclusion, we describe a novel, as yet unknown killing effect of LIGHT through HVEM on a lymphoid malignancy, and combined with induction of chemokine release this may represent an additional tool to boost cancer immunotherapy.
Collapse
Affiliation(s)
- Christine Pasero
- INSERM UMR891, Centre de Recherche en Cancérologie de Marseille, Université de Méditerranée, Institut de Cancérologie et d'Immunologie de Marseille, Marseille, France
| | | | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Yang C, Yang S, Wood KB, Hornicek FJ, Schwab JH, Fondren G, Mankin H, Duan Z. Multidrug resistant osteosarcoma cell lines exhibit deficiency of GADD45alpha expression. Apoptosis 2009; 14:124-33. [PMID: 19052873 DOI: 10.1007/s10495-008-0282-x] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
To identify apoptosis genes involved in the multidrug resistance of osteosarcoma, a multidrug resistant human osteosarcoma cell line (U-2 OS MR) was established. Apoptosis gene microarray analysis demonstrated that GADD45alpha was significantly induced in U-2 OS cells after exposure to paclitaxel (P < 0.0001). However, the induction of GADD45alpha did not occur in U-2 OS MR cells. Subsequent analysis by Western blot confirmed that the expression of GADD45alpha could be significantly induced by paclitaxel and doxorubicin in U-2 OS cells but not in U-2 OS MR cells. Furthermore, the paclitaxel or doxorubicin treated U-2 OS and KH OS cells have a higher percentage of apoptotic cells when compared with U-2 OS MR and KH OS R2 cells treated with the same drugs. When GADD45alpha was transfected into U-2 OS MR or KH OS R2 and expressed, the cells became more sensitive to chemotherapeutic drugs. These results suggest that GADD45alpha may play a role in drug-induced apoptosis, as well as multidrug resistance in osteosarcoma cells.
Collapse
Affiliation(s)
- Cao Yang
- Department of Orthopaedic Surgery, Massachusetts General Hospital, Boston, MA 02114, USA
| | | | | | | | | | | | | | | |
Collapse
|
25
|
Yang D, Stewart TJ, Smith KK, Georgi D, Abrams SI, Liu K. Downregulation of IFN-gammaR in association with loss of Fas function is linked to tumor progression. Int J Cancer 2008; 122:350-62. [PMID: 17918178 DOI: 10.1002/ijc.23090] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
The host immune system functions as an intrinsic surveillance network in the recognition and destruction of tumor cells, and it has been demonstrated that lymphocytes and IFN-gamma are the primary tumor suppressors of the immune system. However, the immune system can concurrently select for tumor variants with reduced immunogenicity and aggressive phenotypes. We report here that tumor escape variants that have survived CTL adoptive immunotherapy exhibited decreased expression levels of both Fas and IFN-gammaR in vitro. Furthermore, examination of spontaneously arising mouse primary mammary carcinoma and lung metastases revealed that both Fas and IFN-gammaR protein levels were dramatically lower in lung metastases than in primary tumors in vivo. Functional disruption of either the Fas- or the IFN-gamma signaling pathway enhanced the colonization efficiency of preexisting metastatic tumor cells, whereas disruption of both Fas and IFN-gammaR pathways resulted in synergistic augmentation of the colonization efficiency of the preexisting metastatic tumor cells, as determined by experimental lung metastases assay. Gene expression profiling revealed that altered expression of genes involved in immediate IFN-gammaR signaling, the interferon primary response, apoptosis and tumor colonization is associated with loss of IFN-gammaR function and enhanced metastatic potential. Interestingly, disruption of IFN-gammaR function did not alter tumor cell susceptibility to CTL-mediated cytotoxicity, but is linked to enhanced infiltration of endogenous T cells in the tumor microenvironment in vivo. These findings suggest that coordinate downregulation of Fas and IFN-gammaR, 2 key components of cancer immunosurveillance system on tumor cells, leads to a more aggressive metastatic phenotype.
Collapse
Affiliation(s)
- Dafeng Yang
- Department of Biochemistry and Molecular Biology, Medical College of Georgia, Augusta, GA 30912, USA
| | | | | | | | | | | |
Collapse
|
26
|
Luparello C, Sirchia R, Lo Sasso B. Midregion PTHrP regulates Rip1 and caspase expression in MDA-MB231 breast cancer cells. Breast Cancer Res Treat 2007; 111:461-74. [PMID: 18030616 DOI: 10.1007/s10549-007-9816-0] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2007] [Accepted: 11/08/2007] [Indexed: 11/28/2022]
Abstract
It was previously reported that the midregion PTHrP domain (38-94)-amide restrains growth and invasion "in vitro", causes striking toxicity and accelerates death of some breast cancer cell lines, the most responsive being MDA-MB231 whose tumorigenesis was also attenuated "in vivo". In addition, we have demonstrated that midregion PTHrP is imported in the nucleoplasm of cultured MDA-MB231 cells, and that "in vitro" it can bind chromatin of metaphase spread preparations and also an isolated 20-mer oligonucleotide, thereby appearing endowed with a putative transcription factor-like DNA-binding ability. Here, we examined whether PTHrP (38-94)-amide was able to modulate the expression of genes encoding for apoptosis factors and caspases. Employing a combination of conventional and semi-quantitative multiplex PCR techniques, antisense oligonucleotide (asODN) transfections, proliferation/invasion assays and protein analyses, here we report that PTHrP treatment induces the up-regulation of Bcl-xS, Bad and Rip1 and switches-on the expression of caspase-2, -5, -6, -7 and -8 in MDA-MB231 cells. Moreover, we demonstrate for the first time that asODN-induced under-expression of Rip1 can lead to a more pronounced up-regulation of some caspases due, at least in part, to JNK inactivation, thus providing a new example of factor involved in the transcriptional regulation of the apoptotic enzymes.
Collapse
Affiliation(s)
- Claudio Luparello
- Dipartimento di Biologia Cellulare e dello Sviluppo, Università di Palermo, Viale delle Scienze, 90128, Palermo, Italy.
| | | | | |
Collapse
|
27
|
Lukashev M, LePage D, Wilson C, Bailly V, Garber E, Lukashin A, Ngam-ek A, Zeng W, Allaire N, Perrin S, Xu X, Szeliga K, Wortham K, Kelly R, Bottiglio C, Ding J, Griffith L, Heaney G, Silverio E, Yang W, Jarpe M, Fawell S, Reff M, Carmillo A, Miatkowski K, Amatucci J, Crowell T, Prentice H, Meier W, Violette SM, Mackay F, Yang D, Hoffman R, Browning JL. Targeting the lymphotoxin-beta receptor with agonist antibodies as a potential cancer therapy. Cancer Res 2007; 66:9617-24. [PMID: 17018619 DOI: 10.1158/0008-5472.can-06-0217] [Citation(s) in RCA: 80] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The lymphotoxin-beta receptor (LT beta R) is a tumor necrosis factor receptor family member critical for the development and maintenance of various lymphoid microenvironments. Herein, we show that agonistic anti-LT beta R monoclonal antibody (mAb) CBE11 inhibited tumor growth in xenograft models and potentiated tumor responses to chemotherapeutic agents. In a syngeneic colon carcinoma tumor model, treatment of the tumor-bearing mice with an agonistic antibody against murine LT beta R caused increased lymphocyte infiltration and necrosis of the tumor. A pattern of differential gene expression predictive of cellular and xenograft response to LT beta R activation was identified in a panel of colon carcinoma cell lines and when applied to a panel of clinical colorectal tumor samples indicated 35% likelihood a tumor response to CBE11. Consistent with this estimate, CBE11 decreased tumor size and/or improved long-term animal survival with two of six independent orthotopic xenografts prepared from surgical colorectal carcinoma samples. Targeting of LT beta R with agonistic mAbs offers a novel approach to the treatment of colorectal and potentially other types of cancers.
Collapse
Affiliation(s)
- Matvey Lukashev
- Department of Immunobiology, Biogen Idec, Cambridge, MA 02142, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Oxelius VA. Personal glimpses of Robert A. Good. Immunol Res 2007. [DOI: 10.1007/s12026-007-0008-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
29
|
García-Tuñón I, Ricote M, Ruiz A A, Fraile B, Paniagua R, Royuela M. Influence of IFN-gamma and its receptors in human breast cancer. BMC Cancer 2007; 7:158. [PMID: 17697357 PMCID: PMC1976422 DOI: 10.1186/1471-2407-7-158] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2007] [Accepted: 08/14/2007] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Interferons are a group of proteins that trigger multiple responses including prevention of viral replication, inhibition of cell growth, and modulation of cell differentiation. In different mammary carcinoma cell lines IFNgamma induces growth arrest at mid-G1. At the present there are no in vivo studies in human breast. The aim of this study was to investigate the expression patterns of IFNgamma and its two receptors (IFNgamma-Ralpha and IFNgamma-Rbeta) by Western blot and immunohistochemistry, in order to elucidate its role in the different types of human breast cancer (in situ and infiltrative). METHODS Immunohistochemical and semiquantitative study of IFNgamma, its receptors types (IFNgamma-Ralpha and IFNgamma-Rbeta), cell proliferation (proliferating cell nuclear antigen, also named PCNA), and apoptosis (TUNEL method) was carried between the three breast groups (fibrocystic lesions, in situ tumors and infiltrating tumors). RESULTS In the three groups of patients, IFNgamma and IFNgamma-Ralpha immunoreactions appeared in the cytoplasm while IFNgamma-Rbeta also was found in the nucleus. The optical density to IFNgamma was higher in in situ carcinoma than in benign and infiltrating tumors. When we observed IFNgamma-Ralpha, the optical density was lower in infiltrating carcinoma than in benign and in situ tumors (the higher density). To IFNgamma-Rbeta, the optical density was similar in the three group samples. In tumor samples PCNA and TUNEL index was significantly higher; than in benign diseases. PCNA index increased with the malignance. No significant differences were found between cancer types to TUNEL. IFNgamma could be a potential therapeutic tool in breast cancer. However, tumor cells are able to escape from the control of this cytokine in the early tumor stages; this is probably due to a decreased expression of IFNgamma, or also to an alteration of either its receptors or some transduction elements. CONCLUSION We conclude that the decrease in the % positive samples that expressed IFNgamma and IFNgamma-Ralpha together with the nuclear localization of IFNgamma-Rbeta, could be a tumoral cell response, although perhaps insufficient to inhibit the uncontrolled cell proliferation. Perhaps, IFNgamma might be unable to activate p21 to stop the cell cycle, suggesting a possible participation in breast cancer development.
Collapse
Affiliation(s)
- Ignacio García-Tuñón
- Department of Cell Biology and Genetics. University of Alcalá, E-28871. Alcalá de Henares, Madrid, Spain
| | - Mónica Ricote
- Department of Cell Biology and Genetics. University of Alcalá, E-28871. Alcalá de Henares, Madrid, Spain
| | - Antonio Ruiz A
- Department of Pathology, Hospital Príncipe de Asturias, E-28871 Alcalá de Henares, Madrid, Spain
| | - Benito Fraile
- Department of Cell Biology and Genetics. University of Alcalá, E-28871. Alcalá de Henares, Madrid, Spain
| | - Ricardo Paniagua
- Department of Cell Biology and Genetics. University of Alcalá, E-28871. Alcalá de Henares, Madrid, Spain
| | - Mar Royuela
- Department of Cell Biology and Genetics. University of Alcalá, E-28871. Alcalá de Henares, Madrid, Spain
| |
Collapse
|
30
|
Schwarz BT, Wang F, Shen L, Clayburgh DR, Su L, Wang Y, Fu YX, Turner JR. LIGHT signals directly to intestinal epithelia to cause barrier dysfunction via cytoskeletal and endocytic mechanisms. Gastroenterology 2007; 132:2383-94. [PMID: 17570213 PMCID: PMC2709832 DOI: 10.1053/j.gastro.2007.02.052] [Citation(s) in RCA: 140] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/15/2007] [Accepted: 02/15/2007] [Indexed: 01/05/2023]
Abstract
BACKGROUND & AIMS LIGHT (lymphotoxin-like inducible protein that competes with glycoprotein D for herpes virus entry on T cells) is a tumor necrosis factor core family member that regulates T-cell activation and causes experimental inflammatory bowel disease. Additional data suggest that LIGHT may be involved in the pathogenesis of human inflammatory bowel disease. The aim of this study was to determine if LIGHT is capable of signaling directly to intestinal epithelia and to define the mechanisms and consequences of such signaling. METHODS The effects of LIGHT and interferon-gamma on barrier function, cytoskeletal regulation, and tight junction structure were assessed in mice and intestinal epithelial monolayers. RESULTS LIGHT induced barrier loss in cultured epithelia via myosin II regulatory light chain (MLC) phosphorylation; both barrier loss and MLC phosphorylation were reversed by MLC kinase (MLCK) inhibition. Pretreatment with interferon-gamma, which induced lymphotoxin beta receptor (LT beta R) expression, was required for these effects, and neither barrier dysfunction nor intestinal epithelial MLC phosphorylation occurred in LT beta R knockout mice. In cultured monolayers, endocytosis of the tight junction protein occludin correlated with barrier loss. Internalized occludin colocalized with caveolin-1. LIGHT-induced occludin endocytosis and barrier loss were both prevented by inhibition of caveolar endocytosis. CONCLUSIONS T cell-derived LIGHT activates intestinal epithelial LT beta R to disrupt barrier function. This requires MLCK activation and caveolar endocytosis. These data suggest a novel role for LIGHT in disease pathogenesis and suggest that inhibition of MLCK-dependent caveolar endocytosis may represent an approach to restoring barrier function in inflammatory bowel disease.
Collapse
Affiliation(s)
- Brad T. Schwarz
- Department of Pathology, The University of Chicago, Chicago IL, 60637
| | - Fengjun Wang
- Department of Pathology, The University of Chicago, Chicago IL, 60637
- State Key Laboratory of Trauma, Burns and Combined Injury, Institute of Burn Research, Southwest Hospital, Third Military Medical University, Chongqing, China
| | - Le Shen
- Department of Pathology, The University of Chicago, Chicago IL, 60637
| | | | - Liping Su
- Department of Pathology, The University of Chicago, Chicago IL, 60637
| | - Yingmin Wang
- Department of Pathology, The University of Chicago, Chicago IL, 60637
| | - Yang-Xin Fu
- Department of Pathology, The University of Chicago, Chicago IL, 60637
| | - Jerrold R. Turner
- Department of Pathology, The University of Chicago, Chicago IL, 60637
| |
Collapse
|
31
|
Pierer M, Brentano F, Rethage J, Wagner U, Hantzschel H, Gay RE, Gay S, Kyburz D. The TNF superfamily member LIGHT contributes to survival and activation of synovial fibroblasts in rheumatoid arthritis. Rheumatology (Oxford) 2007; 46:1063-70. [PMID: 17426140 DOI: 10.1093/rheumatology/kem063] [Citation(s) in RCA: 53] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
OBJECTIVES The TNF superfamily member LIGHT has a T-cell co-stimulatory role and has previously been associated with inflammation and autoimmunity. To investigate its role in rheumatoid arthritis (RA), a disease where activated T cells contribute in a prominent way, we have analysed the expression of LIGHT and its receptors in RA and analysed its effects on synovial fibroblasts in vitro. METHODS The expression of LIGHT was measured in synovial tissues and fluids and the receptors of LIGHT were detected on synovial fibroblasts derived from patients with RA and osteoarthritis (OA). The effects of recombinant LIGHT on the production of proinflammatory cytokines and proteases and on the apoptosis of synovial fibroblasts was assessed. RESULTS LIGHT mRNA was present in synovial tissues of patients with RA but not with OA. Correspondingly, soluble LIGHT protein could be detected in RA synovial fluid samples at much higher levels than in synovial fluid from patients with OA. Immunohistochemical detection of LIGHT and analysis of synovial fluid cells by flow cytometry revealed CD4 T cells as the major source of LIGHT in the rheumatoid joint. Synovial fibroblasts from RA patients were found to express the LIGHT receptors HVEM and LTbetaR. Recombinant LIGHT induced RA synovial fibroblasts to upregulate MMP-9 mRNA, CD54 and IL-6 in an NF-kappaB-dependent fashion. In vitro, exposure of cultured synovial fibroblasts to LIGHT reduced FAS-mediated apoptosis significantly, without affecting the rate of spontaneous apoptosis. CONCLUSIONS The results provide evidence for a novel T-cell-dependent activation of synovial fibroblasts by LIGHT in joints of patients with RA, contributing to an inflammatory and destructive phenotype.
Collapse
MESH Headings
- Aged
- Apoptosis/drug effects
- Arthritis, Rheumatoid/immunology
- Arthritis, Rheumatoid/metabolism
- Arthritis, Rheumatoid/pathology
- CD4-Positive T-Lymphocytes/chemistry
- CD4-Positive T-Lymphocytes/metabolism
- Cell Proliferation
- Cell Survival
- Cells, Cultured
- Female
- Fibroblasts/metabolism
- Fibroblasts/pathology
- Flow Cytometry
- Humans
- Immunohistochemistry
- Intercellular Adhesion Molecule-1/analysis
- Interleukin-6/analysis
- Leukotriene B4/analysis
- Leukotriene B4/metabolism
- Male
- Matrix Metalloproteinase 9/analysis
- Middle Aged
- NF-kappa B/analysis
- NF-kappa B/metabolism
- Osteoarthritis/immunology
- Osteoarthritis/metabolism
- Osteoarthritis/pathology
- RNA, Messenger/analysis
- Receptors, Tumor Necrosis Factor, Member 14/analysis
- Receptors, Tumor Necrosis Factor, Member 14/metabolism
- Recombinant Proteins/pharmacology
- Reverse Transcriptase Polymerase Chain Reaction
- Statistics, Nonparametric
- Synovial Fluid/chemistry
- Synovial Fluid/immunology
- Synovial Fluid/metabolism
- Synovial Membrane/immunology
- Synovial Membrane/metabolism
- Synovial Membrane/pathology
- Tumor Necrosis Factor Ligand Superfamily Member 14/analysis
- Tumor Necrosis Factor Ligand Superfamily Member 14/genetics
- Tumor Necrosis Factor Ligand Superfamily Member 14/metabolism
Collapse
Affiliation(s)
- M Pierer
- Center of Experimental Rheumatology, Department of Rheumatology, University Hospital of Zurich, Gloriastrasse 25, 8091 Zurich, Switzerland
| | | | | | | | | | | | | | | |
Collapse
|
32
|
Soond SM, Carroll C, Townsend PA, Sayan E, Melino G, Behrmann I, Knight RA, Latchman DS, Stephanou A. STAT1 regulates p73-mediated Bax gene expression. FEBS Lett 2007; 581:1217-26. [PMID: 17346710 DOI: 10.1016/j.febslet.2007.02.049] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2007] [Revised: 02/16/2007] [Accepted: 02/22/2007] [Indexed: 10/23/2022]
Abstract
Although signal transducer and activator of transcription 1 (STAT1) mediated regulation of p53 transcription and apoptosis has been previously reported, modulation of other members of the p53 family of transcription factors remains poorly understood. In this study, we found that STAT1 and TA-p73 can interact directly and that p73-mediated Bax promoter activity was observed to be reduced by STAT1 expression in a p53-independent manner for which STAT1 Tyrosine-701 and Serine-727 are key residues. This study presents the first report physically linking STAT1 and TA-p73 signalling and highlights the modulation of the Bax promoter in the context of IFN-gamma stimulation.
Collapse
Affiliation(s)
- Surinder M Soond
- Medical Molecular Biology Unit, Institute of Child Health, University College London, 30 Guilford Street, London WC1N 1EH, UK.
| | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Xu Y, Tamada K, Chen L. LIGHT-related molecular network in the regulation of innate and adaptive immunity. Immunol Res 2007; 37:17-32. [PMID: 17496344 DOI: 10.1007/bf02686093] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/1999] [Revised: 11/30/1999] [Accepted: 11/30/1999] [Indexed: 12/12/2022]
Abstract
The LIGHT-related molecular network is composed of at least seven interacting receptors and ligands. Recent studies reveal that this network has profound immune regulatory functions for both innate and adaptive immunity. Experimental data support the concept that this network may also play roles in the pathogenesis of human diseases including cancer, infection, transplantation tolerance, and autoimmune diseases. In this review, we attempt to dissect each molecular interaction in detail and assemble them in the context of their roles in the pathogenesis and possible therapeutic potential in human diseases.
Collapse
Affiliation(s)
- Yanhui Xu
- Molecular Biology Graduate Program, Mayo Clinic College of Medicine, Rochester, MN, USA
| | | | | |
Collapse
|
34
|
You RI, Chen MC, Wang HW, Chou YC, Lin CH, Hsieh SL. Inhibition of Lymphotoxin-β Receptor–Mediated Cell Death by Survivin-ΔEx3. Cancer Res 2006; 66:3051-61. [PMID: 16540654 DOI: 10.1158/0008-5472.can-05-2479] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
TNFSF14/LIGHT is a member of the tumor necrosis factor superfamily that binds to lymphotoxin-beta receptor (LTbetaR) to induce cell death via caspase-dependent and caspase-independent pathways. It has been shown that cellular inhibitor of apoptosis protein-1 inhibits cell death by binding to LTbetaR-TRAF2/TRAF3 complexes and caspases. In this study, we found that both Kaposi's sarcoma-associated herpesvirus K7 (KSHV-K7), a viral inhibitor of apoptosis protein, and the structurally related protein survivin-DeltaEx3 could inhibit LTbetaR-mediated caspase-3 activation. However, only survivin-DeltaEx3 could protect cells from LTbetaR-mediated cell death. The differential protective effects of survivin-DeltaEx3 and KSHV-K7 can be attributed to the fact that survivin-DeltaEx3, but not KSHV-K7, is able to maintain mitochondrial membrane potential and inhibit second mitochondria-derived activator of caspase/DIABLO release. Moreover, survivin-DeltaEx3 is able to inhibit production of reactive oxygen species and can translocate from nucleus to cytosol to associate with apoptosis signal-regulating kinase 1 after activation of LTbetaR. Furthermore, survivin-DeltaEx3 protects LTbetaR-mediated cell death in caspase-3-deficient MCF-7 cells. Thus, survivin-DeltaEx3 is able to regulate both caspase-dependent and caspase-independent pathways, whereas inhibition of caspase-independent pathway is both sufficient and necessary for its protective effect on LTbetaR-mediated cell death.
Collapse
Affiliation(s)
- Ren-In You
- Institute and Department of Microbiology and Immunology, National Yang-Ming University, Taipei, Taiwan
| | | | | | | | | | | |
Collapse
|
35
|
Danforth DN, Zhu Y. Conversion of Fas-resistant to Fas-sensitive MCF-7 Breast Cancer Cells by the Synergistic Interaction of Interferon-γ and all-TransRetinoic Acid. Breast Cancer Res Treat 2005; 94:81-91. [PMID: 16136269 DOI: 10.1007/s10549-005-7491-6] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
The membrane receptor Fas (Apo-1/CD95) is an important initiator of programmed cell death induced by anti-Fas antibody or Fas ligand. MCF-7 human breast cancer cells have low levels of Fas receptor (FasR) and are resistant to anti-FasR antibody mediated apoptosis, however two naturally occurring substances, interferon and all-trans retinoic acid (AT), act synergistically to enhance antiproliferative processes in these cells, suggesting this combination may also be an effective means for enhancing FasR expression. When this was studied, it was found that IFN-gamma and AT in combination acted synergistically to induce expression of FasR mRNA and FasR protein in a time-dependent and dose-dependent manner. This induction required continuous protein synthesis, and STAT1 protein, but not PKR or TR1 protein, was induced in a manner quantitatively and temporally related to FasR protein induction, and consistent with STAT1 mediation of the synergistic effect of IFN-gamma and AT on FasR expression. FasR-induced cells were resistant to stimulation of apoptosis by anti-FasR antibody, however treatment with cycloheximide rendered these cells sensitive to antibody-induced apoptosis, suggesting endogenous blockade to signaling. These cells did not express caspase 3, or FLIP(L), but strongly expressed the endogenous inhibitor of apoptosis Bcl-2, indicating a type II Fas signaling pathway. Expression of these proteins was not modulated by IFN/AT, however treatment of Fas-induced cells with Bcl-2 specific small interfering RNA (SiRNA) downregulated Bcl-2 protein expression and rendered these cells sensitive to the cytotoxic effects of anti-Fas antibody. These findings indicate that IFN-gamma+AT in combination modulate Fas signaling and provide a novel mechanism for the promotion of cell death in breast cancer cells.
Collapse
Affiliation(s)
- David N Danforth
- Surgery Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA.
| | | |
Collapse
|
36
|
Mortarini R, Scarito A, Nonaka D, Zanon M, Bersani I, Montaldi E, Pennacchioli E, Patuzzo R, Santinami M, Anichini A. Constitutive expression and costimulatory function of LIGHT/TNFSF14 on human melanoma cells and melanoma-derived microvesicles. Cancer Res 2005; 65:3428-36. [PMID: 15833878 DOI: 10.1158/0008-5472.can-04-3239] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Neoplastic cells are thought to have defective expression of costimulatory molecules. However, in this study, we show that human melanoma cells express LIGHT/TNFSF14, a ligand of herpesvirus entry mediator on T cells and of lymphotoxin beta receptor on stromal cells. In vitro, melanoma cells stained for LIGHT in the intracellular compartment, with weak or negative cell surface expression. However, LIGHT was expressed on tumor-derived microvesicles released from melanoma cells. In vivo, LIGHT was found in metastatic lesions, and the extent of lymphotoxin beta receptor expression on the stromal cells was significantly associated with a "brisk" T-cell infiltrate in the neoplastic tissue. In the lesions with a brisk T-cell infiltrate, stromal cells surrounding the tumor also stained for the T-cell attractant chemokine CCL21. The intratumoral T lymphocytes frequently expressed herpesvirus entry mediator and were characterized by a differentiated phenotype. Coculture of lymphocytes with LIGHT(+) melanoma-derived microvesicles or even with LIGHT(+) melanoma cells in the presence of interleukin-2 costimulated LIGHT-dependent CD3(+)CD8(+) T-cell proliferation. However, lymphocyte coculture with LIGHT(+) microvesicles in the presence of interleukin-2 was also associated with an apoptotic response as documented by increased binding of Annexin V by CD3(+)CD8(+) T cells. These data suggest that LIGHT constitutively expressed in human melanoma cells and microvesicles may contribute to regulate T-cell responses to tumor cells.
Collapse
Affiliation(s)
- Roberta Mortarini
- Human Tumor Immunobiology Unit, Department of Experimental Oncology, Istituto Nazionale per lo Studio e la Cura dei Tumori, Via Venezian 1, 20133 Milan, Italy
| | | | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Nishida M, Nasu K, Ueda T, Fukuda J, Takai N, Miyakawa I. Endometriotic cells are resistant to interferon-gamma-induced cell growth inhibition and apoptosis: a possible mechanism involved in the pathogenesis of endometriosis. Mol Hum Reprod 2005; 11:29-34. [PMID: 15579658 DOI: 10.1093/molehr/gah133] [Citation(s) in RCA: 69] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
In order to evaluate the involvement of cell proliferation and apoptosis in the pathogenesis of endometriosis, we investigated the effects of interferon-gamma (IFN-gamma) on cell growth inhibition and apoptosis of cultured ovarian endometriotic cyst stromal cells (ECSC), eutopic endometrial stromal cells with endometriosis (ESCwE) and normal endometrial stromal cells (NESC) by modified methylthiazoletetrazolium assay, 5-bromo-2'-deoxyuridine incorporation assay and internucleosomal DNA fragmentation assay. The expression of apoptosis-related molecules and IFN-gamma receptor 1 was also examined in ECSC, ESCwE and NESC using western blot analysis. IFN-gamma significantly inhibited cell proliferation and DNA synthesis of ESCwE and NESC, and induced apoptosis of these cells. In contrast, IFN-gamma did not show apparent effects on the viable cell number, DNA synthesis, or apoptosis of ECSC. An up-regulated expression of Bcl-2 and Bcl-X(L) proteins was observed in ECSC in comparison with ESCwE and NESC, whereas the levels of Bax, Bad, Fas and Fas ligand proteins in ECSC were similar to those in ESCwE and NESC. IFN-gamma receptor 1 expression was detected in ECSC, ESCwE and NESC. Enhanced expression of anti-apoptotic molecules in the ectopic endometrial cells may contribute to the development of endometriosis by conferring resistance to cytokine-induced apoptosis and increasing the chance that these cells will survive and implant outside the uterus. Further investigations on the regulation of cell proliferation in both the endometriotic and the normal endometrium may be important for the elucidation of the pathogenesis of endometriosis.
Collapse
Affiliation(s)
- Masakazu Nishida
- Department of Obstetrics and Gynecology, Faculty of Medicine, Oita University, Hasama-machi, Oita 879-5593, Japan
| | | | | | | | | | | |
Collapse
|
38
|
Zhang MC, Liu HP, Demchik LL, Zhai YF, Yang DJ. LIGHT sensitizes IFNγ–mediated apoptosis of HT-29 human carcinoma cells through both death receptor and mitochondria pathways. Cell Res 2004; 14:117-24. [PMID: 15115612 DOI: 10.1038/sj.cr.7290210] [Citation(s) in RCA: 40] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
LIGHT [homologous to lymphotoxins, shows inducible expression, and competes with herpes simplex virus glycoprotein D for herpes virus entry mediator (HVEM/TR2)] is a new member of TNF superfamily. The HT-29 colon cancer cell line is the most sensitive to LIGHT-induced, IFNg-mediated apoptosis among the cell lines we have examined so far. Besides downregulation of Bcl-XL, upregulation of Bak, and activation of both PARP [poly (ADP-ribose) polymerase] and DFF45 (DNA fragmentation factor), LIGHT-induced, IFNg-mediated apoptosis of HT-29 cells involves extensive caspase activation. Caspase-8 and caspase-9 activation, as shown by their cleavages appeared as early as 24 h after treatment, whereas caspase-3 and caspase-7 activation, as shown by their cleavages occurred after 72 h of LIGHT treatment. Caspase-3 inhibitor Z-DEVD-FMK (benzyloxycarbonyl-Asp-Glu-Val-Asp-fluoromethylketone) and a broad range caspase inhibitor Z-VAD-FMK (benzyloxycarbonyl-Val-Ala-Asp fluoromethylketone) were able to block LIGHT-induced, IFNg-mediated apoptosis of HT-29 cells. The activity of caspase-3, which is one of the major executioner caspases, was found to be inhibited by both Z-DEVD-MFK and Z-VAD-FMK. These results suggest that LIGHT-induced, IFNg-mediated apoptosis of HT-29 cells is caspase-dependent, and LIGHT signaling is mediated through both death receptor and mitochondria pathways.
Collapse
Affiliation(s)
- Man Chao Zhang
- Division of Hematology/Oncology, Department of Internal Medicine, the University of Michigan, Ann Arbor, MI 48109-0934, USA.
| | | | | | | | | |
Collapse
|
39
|
Takahashi K, Loo G. Disruption of mitochondria during tocotrienol-induced apoptosis in MDA-MB-231 human breast cancer cells. Biochem Pharmacol 2004; 67:315-24. [PMID: 14698044 DOI: 10.1016/j.bcp.2003.07.015] [Citation(s) in RCA: 103] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Tocotrienols, which are Vitamin E isoforms, are known to inhibit the growth of human breast cancer cells due partly to apoptosis. However, the characterization of tocotrienol-induced apoptosis is incomplete, particularly what happens during the initiation phase that precedes execution of the cells. The objective of this study was to clarify the apoptotic effects of tocotrienols, with especial emphasis in determining if the mitochondria-mediated death pathway is activated when human breast cancer cells are incubated with a specific tocotrienol isomer. During incubation with gamma-tocotrienol, MDA-MB-231 human breast cancer cells showed membrane blebbing, and apoptotic bodies were present. Upon 4',6-diamidino-2-phenylindole staining of the cells, chromatin condensation and fragmentation were observed. Additionally, the annexin V-binding assay detected the translocation of membrane phospholipid during earlier analysis of the cells. Taken together, these results further establish that gamma-tocotrienol can induce apoptosis in human breast cancer cells. To help elucidate how gamma-tocotrienol induced the apoptosis, some important parameters related to the mitochondria-mediated death pathway were examined next. In gamma-tocotrienol-treated cells, the mitochondria were disrupted. Collapse of the mitochondrial membrane potential was detected, and cytochrome c was released later from mitochondria. However, expression of Bax and Bcl-2 (mRNA and protein) did not change. Furthermore, poly-(ADP-ribose)-polymerase cleavage was not detected, suggesting that caspases were not involved in the gamma-tocotrienol-induced apoptosis. These results imply that cytochrome c is not the critical protein released from mitochondria that triggers gamma-tocotrienol-induced apoptosis in MDA-MB-231 cells.
Collapse
Affiliation(s)
- Keiko Takahashi
- Cellular and Molecular Nutrition Research Laboratory, Graduate Program in Nutrition, The University of North Carolina at Greensboro, Greensboro, NC 27402-6170, USA
| | | |
Collapse
|
40
|
Chang YH, Chao Y, Hsieh SL, Lin WW. Mechanism of LIGHT/interferon-?-induced cell death in HT-29 cells. J Cell Biochem 2004; 93:1188-202. [PMID: 15486969 DOI: 10.1002/jcb.20282] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
LIGHT is a member of tumor necrosis factor (TNF) superfamily, and previous studies have indicated that in the presence of interferon-gamma (IFN-gamma), LIGHT through LTbetaR signaling can induce cell death with features unlike classic apoptosis. In present study, we investigated the mechanism of LIGHT/IFN-gamma-induced cell death in HT-29 cells, where the cell death was profoundly induced when sub-toxic concentrations of LIGHT and IFN-gamma were co-treated. LIGHT/IFN-gamma-induced cell death was accompanied by DNA fragmentation and slight LDH release. This effect was not affected by caspase, JNK nor cathepsin B inhibitors, but was partially prevented by p38 mitogen-activated protein kinase (MAPK) and poly (ADP-ribose) polymerase (PARP) inhibitors, and abolished by aurintricarboxylic acid (ATA), which is an inhibitor of endonuclease and STATs signaling of IFN-gamma. Immunobloting reveals that LIGHT/IFN-gamma could induce p38 MAPK activity, Bak and Fas expression, but down-regulate Mcl-1. Besides, LIGHT/IFN-gamma could not activate caspase-3 and -9, but decreased mitochondrial membrane potential. Although LIGHT could not affect IFN-gamma-induced STAT1 phosphorylation and transactivation activity, which was required for the sensitization of cell death, survival NF-kappaB signaling of LIGHT was inhibited by IFN-gamma. These data suggest that co-presence of LIGHT and IFN-gamma can induce an integrated interaction in signaling pathways, which lead to mitochondrial dysfunction and mix-type cell death, not involving caspase activation.
Collapse
Affiliation(s)
- Ying-Hsin Chang
- Department of Pharmacology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | | | | | | |
Collapse
|
41
|
Zhang M, Guo R, Zhai Y, Fu XY, Yang D. Light stimulates IFNgamma-mediated intercellular adhesion molecule-1 upregulation of cancer cells. Hum Immunol 2003; 64:416-26. [PMID: 12651068 DOI: 10.1016/s0198-8859(03)00026-0] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Intercellular adhesion molecule-1 (ICAM-1) works as one of the ligands for activating the killing activity of natural killer (NK) cells and cancer specific cytotoxic T lymphocytes (CTL). Expression of ICAM-1 enhances lymphocyte adhesion to the cancer cells in vivo. Cancer cell lines express significantly lower level of ICAM-1 than that of normal epithelium or benign cells. Overexpression of LIGHT (LIGHT: homologous to lymphotoxins, indicating inducible expression, and competes with herpes simplex virus glycoprotein D for herpes virus entry mediator [HVEM/TR2]) in MDA-MB-231 human breast cancer cells was observed to suppress tumor growth in vivo. In order to elucidate the mechanisms how LIGHT overexpression could trigger tumor suppression, the expression level of a panel of cell surface makers CD54, CD56, CD95, and CD119 was investigated in a group of cancer cells. Flow cytometry analysis results demonstrate that LIGHT gene expression in cancer cells can greatly increase ICAM-1 expression level, IFNgamma alone can stimulate cancer cells to express ICAM-1, which can be highly augmented by LIGHT in a dose-dependent manner. This upregulation of ICAM-1 expression is not only at ICAM-1 protein trafficking level on cell surface as demonstrated by flow cytometry analysis, but also at ICAM-1 total protein level as confirmed by Western blot. There is no difference of expression level among these cancer cell lines for the other three cell surface markers: CD56, CD95 (Fas), and CD119. It was confirmed that LIGHT enhancement upregulation of ICAM-1 expression is at least STAT1 and JAK1 dependent by using STAT1-deficient U3A and JAK1-deficient E2A4 cells. These findings suggest that LIGHT-induced inhibition of tumor growth is highly correlated with its upregulation of ICAM-1 expression.
Collapse
Affiliation(s)
- Manchao Zhang
- Structure Biology and Cancer Drug Discovery Program, Lombardi Cancer Center and Department of Oncology, Washington, DC, USA.
| | | | | | | | | |
Collapse
|