1
|
Nadeau A, Ouellet M, Béland R, Mercier C, Robillard S, Lizotte F, Despatis MA, Bentzinger CF, Geraldes P. Endothelial SHP-1 regulates diabetes-induced abnormal collateral vessel formation and endothelial cell senescence. J Mol Cell Cardiol 2025; 202:50-63. [PMID: 40068773 DOI: 10.1016/j.yjmcc.2025.03.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/06/2024] [Revised: 03/01/2025] [Accepted: 03/05/2025] [Indexed: 03/16/2025]
Abstract
BACKGROUND Critical limb ischemia is a major cause of peripheral arterial disease and morbidity affecting patients with diabetes. Diabetes-induced premature senescence of endothelial cells (EC) has been proposed as a mechanism leading to impaired ischemia-driven angiogenesis. We showed that hyperglycemia induced expression of the protein tyrosine phosphatase SHP-1, which reduced angiogenic factor activity in ischemic muscle of diabetic mice. Here, we evaluate the impact of SHP-1 deletion on EC function and senescence. METHODS Ligation of the femoral artery was performed in nondiabetic (NDM) and 3 months diabetic (DM) mice with EC-specific deletion of SHP-1. Cell migration, proliferation and protein expression were evaluated in EC exposed to normal (NG) or high glucose (HG) concentrations. Gastrocnemius and tibial artery of patients with diabetes were collected and analyzed. RESULTS Blood flow reperfusion and limb function were reduced by 43 % and 82 %, respectively in DM mice as compared to NDM mice. EC-specific deletion of SHP-1 in DM mice restored blood flow reperfusion by 60 %, and limb function by 86 %, while capillary density was similar to NDM mice. Moreover, ablation of SHP-1 in EC prevented diabetes-induced expression of the senescence markers p53 and p21 and counteracted Nrf2 downregulation. In EC, elevated expression of beta-galactosidase, p21 and p53, and suppression of Nrf2 and VEGF actions were observed in EC exposed to HG levels and human muscle and artery of patients with diabetes, effects that were reversed by overexpression of dominant negative SHP-1. CONCLUSION SHP-1 in EC is a central effector of diabetes-induced senescence and induces aberrant collateral vessel formation and blood flow reperfusion. Reduced SHP-1 expression counteracts these pathologic features.
Collapse
Affiliation(s)
- Alexandre Nadeau
- Research Center of the Centre Hospitalier Universitaire de Sherbrooke, Québec, Canada
| | - Marike Ouellet
- Research Center of the Centre Hospitalier Universitaire de Sherbrooke, Québec, Canada
| | - Raphaël Béland
- Research Center of the Centre Hospitalier Universitaire de Sherbrooke, Québec, Canada
| | - Clément Mercier
- Research Center of the Centre Hospitalier Universitaire de Sherbrooke, Québec, Canada
| | - Stéphanie Robillard
- Research Center of the Centre Hospitalier Universitaire de Sherbrooke, Québec, Canada
| | - Farah Lizotte
- Research Center of the Centre Hospitalier Universitaire de Sherbrooke, Québec, Canada
| | | | - C Florian Bentzinger
- Research Center of the Centre Hospitalier Universitaire de Sherbrooke, Québec, Canada; Department of Pharmacology and Physiology, Québec, Canada
| | - Pedro Geraldes
- Research Center of the Centre Hospitalier Universitaire de Sherbrooke, Québec, Canada; Department of Medicine, Université de Sherbrooke, Québec, Canada.
| |
Collapse
|
2
|
Wu M, Wu B, Huang X, Wang Z, Zhu M, Zhu Y, Yu L, Liu J. Inhibition of the FEN1-PBX1 axis elicits cellular senescence in breast cancer via the increased intracellular reactive oxygen species levels. J Transl Med 2025; 23:248. [PMID: 40022092 PMCID: PMC11871692 DOI: 10.1186/s12967-025-06216-9] [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: 09/18/2024] [Accepted: 02/10/2025] [Indexed: 03/03/2025] Open
Abstract
BACKGROUND Cellular senescence is a state of irreversible cell growth arrest. As such, senescence induction is viewed as an efficacious countermeasure in cancer treatment. Flap endonuclease 1 (FEN1) has been reported to participate in tumor growth, metastasis and immunomodulation. However, the role of FEN1 in cellular senescence of breast cancer and its molecular mechanism remains unclear. METHODS In vitro assessments of breast cancer cell senescence and apoptosis were conducted using CCK-8 assay, cell cycle assay, senescence-associated β-galactosidase (SA-β-gal) staining, and cleaved caspase-3 staining. Western blot, dihydroethidium (DHE) staining, RNA-sequencing, quantitative real-time polymerase chain reaction (qRT-PCR), rescue experiments, and dual-luciferase reporter assay were performed to explore the potential target of FEN1. Co-Immunoprecipitation (Co-IP), Chromatin immunoprecipitation (ChIP)-qPCR assay, and immunostaining were used to evaluate the interaction between FEN1 and Pre-B-cell leukemia homeobox transcription factor 1 (PBX1). A xenograft mouse model was employed to validate the effect of FEN1 on breast cancer cell senescence and apoptosis. RESULTS Functional analysis demonstrated that FEN1 suppressed both senescence and apoptosis of breast cancer cells in vitro, while in vivo experiments demonstrated moderate therapeutic effects. Further studies indicated that FEN1 deficiency promoted the aforementioned effects by increasing intracellular reactive oxygen species (ROS) levels. RNA-sequencing and qRT-PCR assays revealed that FEN1 knockdown enhanced the expressions of several senescence-associated secretory phenotype (SASP) factors and resulted in decreased PBX1 level. The rescue experiments by PBX1 overexpression verified that PBX1 mediated the senescence and apoptosis of breast cancer cells induced by FEN1 inhibition. In detail, FEN1 downregulation inhibited the transcription activity of PBX1, which was partially restored by itself overexpression. Of note, FEN1 directly interacted with PBX1. Furthermore, immunostaining illustrated the colocalization of FEN1 and PBX1 in breast cancer cells and tissues. In our local breast cancer cohort, a positive correlation was identified between the expression levels of FEN1 and PBX1. CONCLUSIONS Knockdown of FEN1 facilitates breast cancer cell senescence through PBX1 down-regulation mediating increase in intracellular ROS levels. This study reveals FEN1 as a negative regulator of cellular senescence and provides support for pro-senescence cancer therapy. Given that FEN1 knockdown exhibited only moderate in vivo effects, these findings underscore the necessity of combining it with senolytic therapy to enhance therapeutic efficacy.
Collapse
Affiliation(s)
- Min Wu
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, 225009, China.
- Jiangsu Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Senile Diseases, Yangzhou University, Yangzhou, 225009, China.
| | - Benmeng Wu
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, 225009, China
- Jiangsu Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Senile Diseases, Yangzhou University, Yangzhou, 225009, China
| | - Xiaoshan Huang
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, 225009, China
- Jiangsu Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Senile Diseases, Yangzhou University, Yangzhou, 225009, China
| | - Zirui Wang
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, 225009, China
- Jiangsu Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Senile Diseases, Yangzhou University, Yangzhou, 225009, China
| | - Miaolin Zhu
- Department of Pathology, Jiangsu Cancer Hospital, Nanjing, 210018, China
| | - Yaqin Zhu
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, 225009, China
- Jiangsu Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Senile Diseases, Yangzhou University, Yangzhou, 225009, China
| | - Lin Yu
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, 225009, China
- Jiangsu Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Senile Diseases, Yangzhou University, Yangzhou, 225009, China
| | - Jingjing Liu
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, 225009, China.
- Jiangsu Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Senile Diseases, Yangzhou University, Yangzhou, 225009, China.
| |
Collapse
|
3
|
Romdhoni AC, Rajanagara AS, Albab CF, Waskito LA, Wibowo IN, Mohamad Yunus MR. The Role of Cyclin d1 in Radiotherapy Resistance of Advance Stage Nasopharyngeal Carcinoma: A Systematic Review. Asian Pac J Cancer Prev 2024; 25:2211-2218. [PMID: 39068551 PMCID: PMC11480630 DOI: 10.31557/apjcp.2024.25.7.2211] [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: 11/13/2023] [Indexed: 07/30/2024] Open
Abstract
OBJECTIVE One of the biggest therapy challenges for nasopharyngeal cancer (NPC) is still radioresistance. The radioresistance in NPC is thought to be caused by cyclin D1 overexpression. The purpose of this study was to determine how cyclin D1 contributes to radiation resistance in NPC. METHODS Adhering to the PRISMA guidelines, we systematically reviewed studies on cyclin D1-associated radioresistance in NPC from 2012 until 2023. From our search, 15 studies were included. RESULTS Cyclin D1's role in radiotherapy resistance is elucidated through several mechanisms, notably SHP-1 and B-catenin. Overexpression of SHP-1 led to an increase in cyclin D1, a higher proportion of cells in the S-phase, and radioresistance. Conversely, inhibiting β-catenin and cyclin D1 expression enhances radiation sensitivity. CONCLUSION In conclusion, Cyclin D1 has a strong correlation with radiation resistance; downregulation of the protein increases radiosensitivity, while overexpression of the protein promotes radioresistance.
Collapse
Affiliation(s)
- Achmad Chusnu Romdhoni
- Department of Otorhinolaryngology-Head and Neck Surgery, Faculty of Medicine, Universitas Airlangga, Indonesia.
| | | | | | - Langgeng Agung Waskito
- Department of Internal Medicine, Faculty of Medicine, Universitas Airlangga, Surabaya, East Java, 60286, Indonesia.
- Otorhinolaryngologist Head and Neck Surgeon, Faculty of Medicine, Univerisiti Kebangsaan, Malaysia.
| | | | - Mohd Razif Mohamad Yunus
- Otorhinolaryngologist Head and Neck Surgeon, Faculty of Medicine, Univerisiti Kebangsaan, Malaysia.
| |
Collapse
|
4
|
Meiners F, Hinz B, Boeckmann L, Secci R, Sueto S, Kuepfer L, Fuellen G, Barrantes I. Computational identification of natural senotherapeutic compounds that mimic dasatinib based on gene expression data. Sci Rep 2024; 14:6286. [PMID: 38491064 PMCID: PMC10943199 DOI: 10.1038/s41598-024-55870-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2023] [Accepted: 02/28/2024] [Indexed: 03/18/2024] Open
Abstract
The major risk factor for chronic disease is chronological age, and age-related chronic diseases account for the majority of deaths worldwide. Targeting senescent cells that accumulate in disease-related tissues presents a strategy to reduce disease burden and to increase healthspan. The senolytic combination of the tyrosine-kinase inhibitor dasatinib and the flavonol quercetin is frequently used in clinical trials aiming to eliminate senescent cells. Here, our goal was to computationally identify natural senotherapeutic repurposing candidates that may substitute dasatinib based on their similarity in gene expression effects. The natural senolytic piperlongumine (a compound found in long pepper), and the natural senomorphics parthenolide, phloretin and curcumin (found in various edible plants) were identified as potential substitutes of dasatinib. The gene expression changes underlying the repositioning highlight apoptosis-related genes and pathways. The four compounds, and in particular the top-runner piperlongumine, may be combined with quercetin to obtain natural formulas emulating the dasatinib + quercetin formula.
Collapse
Affiliation(s)
- Franziska Meiners
- Institute for Biostatistics and Informatics in Medicine and Ageing Research, Rostock University Medical Center, Rostock, Germany
| | - Burkhard Hinz
- Institute of Pharmacology and Toxicology, Rostock University Medical Center, Rostock, Germany
| | - Lars Boeckmann
- Clinic and Policlinic for Dermatology and Venerology, University Medical Center Rostock, Strempelstr. 13, 18057, Rostock, Germany
| | - Riccardo Secci
- Institute for Biostatistics and Informatics in Medicine and Ageing Research, Rostock University Medical Center, Rostock, Germany
| | - Salem Sueto
- Institute for Biostatistics and Informatics in Medicine and Ageing Research, Rostock University Medical Center, Rostock, Germany
| | - Lars Kuepfer
- Institute for Systems Medicine with Focus on Organ Interaction, University Hospital RWTH Aachen, Aachen, Germany
| | - Georg Fuellen
- Institute for Biostatistics and Informatics in Medicine and Ageing Research, Rostock University Medical Center, Rostock, Germany.
| | - Israel Barrantes
- Institute for Biostatistics and Informatics in Medicine and Ageing Research, Rostock University Medical Center, Rostock, Germany
| |
Collapse
|
5
|
Tang X, Qi C, Zhou H, Liu Y. Critical roles of PTPN family members regulated by non-coding RNAs in tumorigenesis and immunotherapy. Front Oncol 2022; 12:972906. [PMID: 35957898 PMCID: PMC9360549 DOI: 10.3389/fonc.2022.972906] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2022] [Accepted: 07/04/2022] [Indexed: 12/22/2022] Open
Abstract
Since tyrosine phosphorylation is reversible and dynamic in vivo, the phosphorylation state of proteins is controlled by the opposing roles of protein tyrosine kinases (PTKs) and protein tyrosine phosphatase (PTPs), both of which perform critical roles in signal transduction. Of these, intracellular non-receptor PTPs (PTPNs), which belong to the largest class I cysteine PTP family, are essential for the regulation of a variety of biological processes, including but not limited to hematopoiesis, inflammatory response, immune system, and glucose homeostasis. Additionally, a substantial amount of PTPNs have been identified to hold crucial roles in tumorigenesis, progression, metastasis, and drug resistance, and inhibitors of PTPNs have promising applications due to striking efficacy in antitumor therapy. Hence, the aim of this review is to summarize the role played by PTPNs, including PTPN1/PTP1B, PTPN2/TC-PTP, PTPN3/PTP-H1, PTPN4/PTPMEG, PTPN6/SHP-1, PTPN9/PTPMEG2, PTPN11/SHP-2, PTPN12/PTP-PEST, PTPN13/PTPL1, PTPN14/PEZ, PTPN18/PTP-HSCF, PTPN22/LYP, and PTPN23/HD-PTP, in human cancer and immunotherapy and to comprehensively describe the molecular pathways in which they are implicated. Given the specific roles of PTPNs, identifying potential regulators of PTPNs is significant for understanding the mechanisms of antitumor therapy. Consequently, this work also provides a review on the role of non-coding RNAs (ncRNAs) in regulating PTPNs in tumorigenesis and progression, which may help us to find effective therapeutic agents for tumor therapy.
Collapse
Affiliation(s)
- Xiaolong Tang
- Department of Clinical Laboratory Diagnostics, Binzhou Medical University, Binzhou, China
| | - Chumei Qi
- Department of Clinical Laboratory, Dazhou Women and Children’s Hospital, Dazhou, China
| | - Honghong Zhou
- Key Laboratory of RNA Biology, Center for Big Data Research in Health, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
- *Correspondence: Honghong Zhou, ; Yongshuo Liu,
| | - Yongshuo Liu
- Biomedical Pioneering Innovation Center (BIOPIC), Beijing Advanced Innovation Center for Genomics, Peking-Tsinghua Center for Life Sciences, Peking University Genome Editing Research Center, State Key Laboratory of Protein and Plant Gene Research, School of Life Sciences, Peking University, Beijing, China
- *Correspondence: Honghong Zhou, ; Yongshuo Liu,
| |
Collapse
|
6
|
Zhan Y, Fan S. Multiple Mechanisms Involving in Radioresistance of Nasopharyngeal Carcinoma. J Cancer 2020; 11:4193-4204. [PMID: 32368302 PMCID: PMC7196263 DOI: 10.7150/jca.39354] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2019] [Accepted: 02/04/2020] [Indexed: 02/07/2023] Open
Abstract
Nasopharyngeal carcinoma (NPC) is the malignant tumor with ethnic and geographical distribution preference. Although intensity-modulated radiotherapy (IMRT)-based radiotherapy combined with chemotherapy and targeted therapy has dramatically improved the overall survival of NPC patients, there are still some patients suffering from recurrent tumors and the prognosis is poor. Multiple mechanisms may be responsible for radioresistance of NPC, such as cancer stem cells (CSCs) existence, gene mutation or aberrant expression of genes, epigenetic modification of genes, abnormal activation of certain signaling pathways, alteration of tumor microenvironment, stress granules (SGs) formation, etc. We conduct a comprehensive review of the published literatures focusing on the causes of radioresistance, retrospect the regulation mechanisms following radiation, and discuss future directions of overcoming the resistance to radiation.
Collapse
Affiliation(s)
- Yuting Zhan
- Department of Pathology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Songqing Fan
- Department of Pathology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| |
Collapse
|
7
|
Avelar RA, Ortega JG, Tacutu R, Tyler EJ, Bennett D, Binetti P, Budovsky A, Chatsirisupachai K, Johnson E, Murray A, Shields S, Tejada-Martinez D, Thornton D, Fraifeld VE, Bishop CL, de Magalhães JP. A multidimensional systems biology analysis of cellular senescence in aging and disease. Genome Biol 2020; 21:91. [PMID: 32264951 PMCID: PMC7333371 DOI: 10.1186/s13059-020-01990-9] [Citation(s) in RCA: 215] [Impact Index Per Article: 43.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2019] [Accepted: 03/08/2020] [Indexed: 01/10/2023] Open
Abstract
BACKGROUND Cellular senescence, a permanent state of replicative arrest in otherwise proliferating cells, is a hallmark of aging and has been linked to aging-related diseases. Many genes play a role in cellular senescence, yet a comprehensive understanding of its pathways is still lacking. RESULTS We develop CellAge (http://genomics.senescence.info/cells), a manually curated database of 279 human genes driving cellular senescence, and perform various integrative analyses. Genes inducing cellular senescence tend to be overexpressed with age in human tissues and are significantly overrepresented in anti-longevity and tumor-suppressor genes, while genes inhibiting cellular senescence overlap with pro-longevity and oncogenes. Furthermore, cellular senescence genes are strongly conserved in mammals but not in invertebrates. We also build cellular senescence protein-protein interaction and co-expression networks. Clusters in the networks are enriched for cell cycle and immunological processes. Network topological parameters also reveal novel potential cellular senescence regulators. Using siRNAs, we observe that all 26 candidates tested induce at least one marker of senescence with 13 genes (C9orf40, CDC25A, CDCA4, CKAP2, GTF3C4, HAUS4, IMMT, MCM7, MTHFD2, MYBL2, NEK2, NIPA2, and TCEB3) decreasing cell number, activating p16/p21, and undergoing morphological changes that resemble cellular senescence. CONCLUSIONS Overall, our work provides a benchmark resource for researchers to study cellular senescence, and our systems biology analyses reveal new insights and gene regulators of cellular senescence.
Collapse
Affiliation(s)
- Roberto A Avelar
- Integrative Genomics of Ageing Group, Institute of Ageing and Chronic Disease, University of Liverpool, Liverpool, L7 8TX, UK
| | - Javier Gómez Ortega
- Integrative Genomics of Ageing Group, Institute of Ageing and Chronic Disease, University of Liverpool, Liverpool, L7 8TX, UK
- School of Biological Sciences, Monash University, Melbourne, VIC, 3800, Australia
| | - Robi Tacutu
- Integrative Genomics of Ageing Group, Institute of Ageing and Chronic Disease, University of Liverpool, Liverpool, L7 8TX, UK
- Computational Biology of Aging Group, Institute of Biochemistry, Romanian Academy, 060031, Bucharest, Romania
- Chronos Biosystems SRL, 060117, Bucharest, Romania
| | - Eleanor J Tyler
- Centre for Cell Biology and Cutaneous Research, Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, E1 2AT, UK
| | - Dominic Bennett
- Integrative Genomics of Ageing Group, Institute of Ageing and Chronic Disease, University of Liverpool, Liverpool, L7 8TX, UK
| | - Paolo Binetti
- Integrative Genomics of Ageing Group, Institute of Ageing and Chronic Disease, University of Liverpool, Liverpool, L7 8TX, UK
| | - Arie Budovsky
- Research and Development Authority, Barzilai Medical Center, Ashkelon, Israel
| | - Kasit Chatsirisupachai
- Integrative Genomics of Ageing Group, Institute of Ageing and Chronic Disease, University of Liverpool, Liverpool, L7 8TX, UK
| | - Emily Johnson
- Integrative Genomics of Ageing Group, Institute of Ageing and Chronic Disease, University of Liverpool, Liverpool, L7 8TX, UK
| | - Alex Murray
- Integrative Genomics of Ageing Group, Institute of Ageing and Chronic Disease, University of Liverpool, Liverpool, L7 8TX, UK
| | - Samuel Shields
- Integrative Genomics of Ageing Group, Institute of Ageing and Chronic Disease, University of Liverpool, Liverpool, L7 8TX, UK
| | - Daniela Tejada-Martinez
- Integrative Genomics of Ageing Group, Institute of Ageing and Chronic Disease, University of Liverpool, Liverpool, L7 8TX, UK
- Doctorado en Ciencias mención Ecología y Evolución, Instituto de Ciencias Ambientales y Evolutivas, Facultad de Ciencias, Universidad Austral de Chile, Independencia 631, Valdivia, Chile
| | - Daniel Thornton
- Integrative Genomics of Ageing Group, Institute of Ageing and Chronic Disease, University of Liverpool, Liverpool, L7 8TX, UK
| | - Vadim E Fraifeld
- The Shraga Segal Department of Microbiology, Immunology and Genetics, Faculty of Health Sciences, Center for Multidisciplinary Research on Aging, Ben-Gurion University of the Negev, 8410501, Beer Sheva, Israel
| | - Cleo L Bishop
- Centre for Cell Biology and Cutaneous Research, Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, E1 2AT, UK.
| | - João Pedro de Magalhães
- Integrative Genomics of Ageing Group, Institute of Ageing and Chronic Disease, University of Liverpool, Liverpool, L7 8TX, UK.
| |
Collapse
|
8
|
Zhang Y, Chen F, Xiao X, Pan W, Yuan Q, Cao J. Chrysin inhibits sphere formation in SMMC-7721 cells via modulation of SHP-1/STAT3 signaling pathway. Cancer Manag Res 2019; 11:2977-2985. [PMID: 31114345 PMCID: PMC6497861 DOI: 10.2147/cmar.s193647] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2018] [Accepted: 02/27/2019] [Indexed: 12/17/2022] Open
Abstract
Background: Chrysin is a natural flavonoid which has been identified as a candidate anti-cancer agent due to its inhibitory effect on a variety of cancer cells, including targeted inhibition of sphere formation in hepatocellular carcinoma (HCC) cell lines. However, the mechanism by which chrysin modulates HCC spheres remains unclear. Materials and methods: In this study, we investigate the effect of chrysin on the regulation of SHP-1 and its downstream signal molecule STAT3 to explain the mechanism by which chrysin inhibits sphere formation of HCC cell lines. Results: Here, we found that SHP-1 protein expression was markedly down-regulated in the spheres from both SMMC-7721 and MHCC97H cells. Chrysin significantly inhibited sphere formation and upregulated the expression of SHP-1 protein in both SMMC-7721 and MHCC97H cells, as well as reduced p-STAT3 and Twist1 expressions in SMMC-7721 cells. Furthermore, knockdown of SHP-1 in SMMC-7721 cells resulted in the induction of p-STAT3 and Twist1 protein expression and antagonizing the inhibitory effect of chrysin on sphere formation in SMMC-7721 cells. Conclusion: Overall, the study findings demonstrated that chrysin acts as a candidate for the treatment of HCC through modulating SHP-1/STAT3 signaling pathway.
Collapse
Affiliation(s)
- Yanqin Zhang
- Department of Pharmacy, Xiangya Hospital of Central South University, Changsha, 410008, People's Republic of China.,Department of Pharmacy, Hunan Food and Drug Vocational College, Changsha, 410208, People's Republic of China
| | - Feng Chen
- Department of Pharmacy, Xiangya Hospital of Central South University, Changsha, 410008, People's Republic of China
| | - Xinghua Xiao
- Department of Pharmacy, Xiangya Hospital of Central South University, Changsha, 410008, People's Republic of China
| | - Weinan Pan
- Department of Pharmacy, Hunan Food and Drug Vocational College, Changsha, 410208, People's Republic of China
| | - Qing Yuan
- Department of Pharmaceutical Science, Medical College, Hunan Normal University, Changsha 410013, People's Republic of China
| | - Jianguo Cao
- Department of Pharmaceutical Science, Medical College, Hunan Normal University, Changsha 410013, People's Republic of China
| |
Collapse
|
9
|
Shi C, Guan Y, Zeng L, Liu G, Zhu Y, Xu H, Lu Y, Liu J, Guo J, Feng X, Zhao X, Jiang W, Li G, Li G, Dai Y, Jin F, Li W, Zhou W. High COX-2 expression contributes to a poor prognosis through the inhibition of chemotherapy-induced senescence in nasopharyngeal carcinoma. Int J Oncol 2018; 53:1138-1148. [PMID: 29956730 PMCID: PMC6065426 DOI: 10.3892/ijo.2018.4462] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2018] [Accepted: 06/11/2018] [Indexed: 02/05/2023] Open
Abstract
Resistance to radiotherapy and chemotherapy currently represents one of the major reasons for therapeutic failure in nasopharyngeal carcinoma (NPC). However, the mechanisms underlying resistance to chemotherapy in NPC remain unclear. In this study, cell counting assay, cell cycle assay and senescence associated β-galactosidase activity were performed to evaluate cell growth, proliferation and senescence, respectively. We found that the aberrant expression of cyclooxygenase-2 (COX-2) was associated with a poor outcome and recurrance in patients with NPC. In NPC cells, COX-2 overexpression increased cell proliferation, inhibited cellular senescence and resulted in chemoresistance, while the knockdown of COX-2 reduced cell proliferation, promoted cellular senescence and overcame chemoresistance. Furthermore, fibroblasts from COX-2 knockout mice exhibited cellular senescence, particularly when treated with chemotherapeutic agents. Mechanistically, COX-2 interacted with p53 protein and inhibited cellular senescence, which resulted in chemotherapeutic resistance. On the whole, these findings indicate that COX-2 may play a critical role in chemotherapeutic resistance in NPC via the inhibition of chemotherapy-induced senescence via the inactivation of p53. This study provides experimental evidence for the preclinical value of increasing chemotherapy-induced senescence by targeting COX-2 as an effective antitumor treatment in patients with recurrent NPC.
Collapse
MESH Headings
- Animals
- Antineoplastic Agents/pharmacology
- Antineoplastic Agents/therapeutic use
- Benzothiazoles/pharmacology
- Biomarkers, Tumor
- Carcinoma/drug therapy
- Carcinoma/mortality
- Carcinoma/pathology
- Cell Line, Tumor
- Cell Proliferation
- Cellular Senescence/drug effects
- Cyclooxygenase 2/genetics
- Cyclooxygenase 2/metabolism
- Drug Resistance, Neoplasm
- Female
- Fibroblasts
- Gene Expression Profiling
- Humans
- Male
- Mice
- Mice, Inbred BALB C
- Mice, Inbred C57BL
- Mice, Knockout
- Mice, Nude
- Middle Aged
- Nasopharyngeal Carcinoma
- Nasopharyngeal Neoplasms/drug therapy
- Nasopharyngeal Neoplasms/mortality
- Nasopharyngeal Neoplasms/pathology
- Neoplasm Recurrence, Local/epidemiology
- Neoplasm Recurrence, Local/pathology
- Primary Cell Culture
- RNA Interference
- RNA, Small Interfering/metabolism
- Specific Pathogen-Free Organisms
- Survival Analysis
- Toluene/analogs & derivatives
- Toluene/pharmacology
- Tumor Suppressor Protein p53/antagonists & inhibitors
- Tumor Suppressor Protein p53/metabolism
Collapse
Affiliation(s)
- Chen Shi
- Cancer Center, The First Hospital of Jilin University, Changchun, Jilin 130021
| | - Yongjun Guan
- Cancer Research Institute; Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education; National Health and Family Planning Commission, Central South University, Changsha, Hunan 410008
| | - Liang Zeng
- Department of Pathology, Guangzhou Women and Children's Medical Center, Guangzhou, Guangdong 510623
| | - Guizhu Liu
- Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Chinese Academy of Sciences, Shanghai 200030
| | - Yinghong Zhu
- Cancer Research Institute; Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education; National Health and Family Planning Commission, Central South University, Changsha, Hunan 410008
| | - He Xu
- Cancer Center, The First Hospital of Jilin University, Changchun, Jilin 130021
- Cancer Research Institute; Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education; National Health and Family Planning Commission, Central South University, Changsha, Hunan 410008
| | - Yichen Lu
- Cancer Center, The First Hospital of Jilin University, Changchun, Jilin 130021
- Cancer Research Institute; Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education; National Health and Family Planning Commission, Central South University, Changsha, Hunan 410008
| | - Jiabin Liu
- Cancer Research Institute; Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education; National Health and Family Planning Commission, Central South University, Changsha, Hunan 410008
| | - Jiaojiao Guo
- Cancer Research Institute; Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education; National Health and Family Planning Commission, Central South University, Changsha, Hunan 410008
| | - Xiangling Feng
- Xiangya School of Public Health, Central South University, Changsha, Hunan 410008
| | - Xinying Zhao
- Xiangya School of Public Health, Central South University, Changsha, Hunan 410008
| | - Weihong Jiang
- Department of Otolaryngology, Xiangya Hospital, Central South University, Changsha, Hunan 410008
| | - Guancheng Li
- Cancer Research Institute; Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education; National Health and Family Planning Commission, Central South University, Changsha, Hunan 410008
| | - Guiyuan Li
- Cancer Research Institute; Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education; National Health and Family Planning Commission, Central South University, Changsha, Hunan 410008
| | - Yun Dai
- Laboratory of Cancer Precision Medicine, The First Hospital of Jilin University, Changchun, Jilin 130021, P.R. China
| | - Fengyan Jin
- Cancer Center, The First Hospital of Jilin University, Changchun, Jilin 130021
| | - Wei Li
- Cancer Center, The First Hospital of Jilin University, Changchun, Jilin 130021
| | - Wen Zhou
- Cancer Research Institute; Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education; National Health and Family Planning Commission, Central South University, Changsha, Hunan 410008
| |
Collapse
|
10
|
Liu SL, Tang LQ, Chen QY, Lin HX, Yang Q, Zhu Q, Wen W, Guo SS, Liu LT, Li Y, Xie HJ, Tang QN, Sun XS, Liang YJ, Li XY, Yan JJ, Lin C, Lan XW, Mai HQ, Guo L. The prognosis of neck residue nasopharyngeal carcinoma (NPC) patients: results from a case-cohort study. J Cancer 2018; 9:1765-1772. [PMID: 29805702 PMCID: PMC5968764 DOI: 10.7150/jca.24573] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2017] [Accepted: 03/03/2018] [Indexed: 02/05/2023] Open
Abstract
Background: To assess the prognosis of neck residue nasopharyngeal carcinoma (NPC) patients and the efficacy of neck dissection in the treatment of these patients. Methods: We recruited 68 neck residue NPC patients. For each neck residue patient we had three matched NPC patients without neck residue as controls (n = 204). The primary endpoint was progression-free survival (PFS). The Cox proportional hazards model was used to calculate hazard ratios (HRs) with 95% confidence intervals (CIs), and multivariable analysis was used to test the independent statistical significance of NPC patients. Results: Compared to controls, the neck residue patients showed significantly lower 3-year PFS (46.7% vs. 87.6%; P < 0.001). Multivariable analysis showed that neck residue was an independent prognostic factor for PFS. Conclusions: NPC patients who had pathologically proven neck residue are associated with poor prognosis. Management with neck dissection alone seems not to be sufficient for these patients.
Collapse
Affiliation(s)
- Sai-Lan Liu
- Sun Yat-sen University Cancer Center; State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine.,Department of Nasopharyngeal Carcinoma; Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University
| | - Lin-Quan Tang
- Sun Yat-sen University Cancer Center; State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine.,Department of Nasopharyngeal Carcinoma; Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University
| | - Qiu-Yan Chen
- Sun Yat-sen University Cancer Center; State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine.,Department of Nasopharyngeal Carcinoma; Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University
| | - Huan-Xin Lin
- Sun Yat-sen University Cancer Center; State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine
| | - Qi Yang
- Sun Yat-sen University Cancer Center; State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine.,Department of Nasopharyngeal Carcinoma; Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University
| | - Qian Zhu
- Sun Yat-sen University Cancer Center; State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine
| | - Wen Wen
- Sun Yat-sen University Cancer Center; State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine.,Department of Nasopharyngeal Carcinoma; Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University
| | - Shan-Shan Guo
- Sun Yat-sen University Cancer Center; State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine.,Department of Nasopharyngeal Carcinoma; Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University
| | - Li-Ting Liu
- Sun Yat-sen University Cancer Center; State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine.,Department of Nasopharyngeal Carcinoma; Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University
| | - Yang Li
- Sun Yat-sen University Cancer Center; State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine.,Department of Nasopharyngeal Carcinoma; Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University
| | - Hao-Jun Xie
- Sun Yat-sen University Cancer Center; State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine.,Department of Nasopharyngeal Carcinoma; Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University
| | - Qing-Nan Tang
- Sun Yat-sen University Cancer Center; State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine.,Department of Nasopharyngeal Carcinoma; Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University
| | - Xue-Song Sun
- Sun Yat-sen University Cancer Center; State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine.,Department of Nasopharyngeal Carcinoma; Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University
| | - Yu-Jing Liang
- Sun Yat-sen University Cancer Center; State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine.,Department of Nasopharyngeal Carcinoma; Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University
| | - Xiao-Yun Li
- Sun Yat-sen University Cancer Center; State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine.,Department of Nasopharyngeal Carcinoma; Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University
| | - Jin-Jie Yan
- Sun Yat-sen University Cancer Center; State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine.,Department of Nasopharyngeal Carcinoma; Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University
| | - Chao Lin
- Sun Yat-sen University Cancer Center; State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine.,Department of Nasopharyngeal Carcinoma; Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University
| | - Xiao-Wen Lan
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University.,Department of Radiation Oncology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University
| | - Hai-Qiang Mai
- Sun Yat-sen University Cancer Center; State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine.,Department of Nasopharyngeal Carcinoma; Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University
| | - Ling Guo
- Sun Yat-sen University Cancer Center; State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine.,Department of Nasopharyngeal Carcinoma; Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University
| |
Collapse
|
11
|
Wei F, Tang L, He Y, Wu Y, Shi L, Xiong F, Gong Z, Guo C, Li X, Liao Q, Zhang W, Ni Q, Luo J, Li X, Li Y, Peng C, Chen X, Li G, Xiong W, Zeng Z. BPIFB1 (LPLUNC1) inhibits radioresistance in nasopharyngeal carcinoma by inhibiting VTN expression. Cell Death Dis 2018; 9:432. [PMID: 29568064 PMCID: PMC5864881 DOI: 10.1038/s41419-018-0409-0] [Citation(s) in RCA: 66] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2017] [Revised: 02/15/2018] [Accepted: 02/16/2018] [Indexed: 12/13/2022]
Abstract
Bactericidal/permeability-increasing-fold-containing family B member 1 (BPIFB1, previously named LPLUNC1) is highly expressed in the nasopharynx and significantly downregulated in nasopharyngeal carcinoma (NPC). Low expression is also associated with poor prognosis in patients with NPC. Radiotherapy is a routine treatment for NPC; however, radioresistance is a major cause of treatment failure. Thus, we aimed to investigate the role of BPIFB1 in the radioresponse of NPC. Colony formation and cell survival results showed that BPIFB1 sensitized NPC cells to ionizing radiation. VTN, a previously identified BPIFB1-binding protein, was shown to induce cell proliferation and survival, G2/M phase arrest, DNA repair, activation of the ATM-Chk2 and ATR-Chk1 pathways, and anti-apoptotic effects after exposure to radiation, facilitating NPC cell radioresistance. However, BPIFB1 inhibited this VTN-mediated radioresistance, ultimately improving NPC radiosensitivity. In conclusion, this study is the first to demonstrate the functions of BPIFB1 and VTN in the NPC radioresponse. Our findings indicated that promoting BPIFB1 expression and targeting VTN might represent new therapeutic strategies for NPC.
Collapse
Affiliation(s)
- Fang Wei
- The Key Laboratory of Carcinogenesis of the Chinese Ministry of Health, Xiangya Hospital, Central South University, Changsha, Hunan, China
- The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan, China
- Hunan Key Laboratory of Nonresolving Inflammation and Cancer, Disease Genome Research Center, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Le Tang
- The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan, China
| | - Yi He
- The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan, China
- Hunan Key Laboratory of Translational Radiation Oncology, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Yingfen Wu
- The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan, China
| | - Lei Shi
- The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan, China
| | - Fang Xiong
- The Key Laboratory of Carcinogenesis of the Chinese Ministry of Health, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Zhaojian Gong
- The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan, China
| | - Can Guo
- The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan, China
| | - Xiayu Li
- The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan, China
- Hunan Key Laboratory of Nonresolving Inflammation and Cancer, Disease Genome Research Center, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Qianjin Liao
- The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan, China
- Hunan Key Laboratory of Translational Radiation Oncology, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Wenling Zhang
- The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan, China
| | - Qianxi Ni
- Hunan Key Laboratory of Translational Radiation Oncology, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Jia Luo
- Hunan Key Laboratory of Translational Radiation Oncology, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Xiaoling Li
- The Key Laboratory of Carcinogenesis of the Chinese Ministry of Health, Xiangya Hospital, Central South University, Changsha, Hunan, China
- The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan, China
- Hunan Key Laboratory of Nonresolving Inflammation and Cancer, Disease Genome Research Center, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Yong Li
- The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan, China
- Department of Cancer Biology, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Cong Peng
- The Key Laboratory of Carcinogenesis of the Chinese Ministry of Health, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Xiang Chen
- The Key Laboratory of Carcinogenesis of the Chinese Ministry of Health, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Guiyuan Li
- The Key Laboratory of Carcinogenesis of the Chinese Ministry of Health, Xiangya Hospital, Central South University, Changsha, Hunan, China
- The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan, China
- Hunan Key Laboratory of Nonresolving Inflammation and Cancer, Disease Genome Research Center, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Wei Xiong
- The Key Laboratory of Carcinogenesis of the Chinese Ministry of Health, Xiangya Hospital, Central South University, Changsha, Hunan, China.
- The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan, China.
- Hunan Key Laboratory of Nonresolving Inflammation and Cancer, Disease Genome Research Center, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China.
| | - Zhaoyang Zeng
- The Key Laboratory of Carcinogenesis of the Chinese Ministry of Health, Xiangya Hospital, Central South University, Changsha, Hunan, China.
- The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan, China.
- Hunan Key Laboratory of Nonresolving Inflammation and Cancer, Disease Genome Research Center, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China.
| |
Collapse
|
12
|
Li X, Zhang C, Bian Q, Gao N, Zhang X, Meng Q, Wu S, Wang S, Xia Y, Chen R. Integrative functional transcriptomic analyses implicate specific molecular pathways in pulmonary toxicity from exposure to aluminum oxide nanoparticles. Nanotoxicology 2016; 10:957-969. [PMID: 26830206 DOI: 10.3109/17435390.2016.1149632] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2015] [Accepted: 01/27/2016] [Indexed: 11/13/2022]
Abstract
Gene expression profiling has developed rapidly in recent years and it can predict and define mechanisms underlying chemical toxicity. Here, RNA microarray and computational technology were used to show that aluminum oxide nanoparticles (Al2O3 NPs) were capable of triggering up-regulation of genes related to the cell cycle and cell death in a human A549 lung adenocarcinoma cell line. Gene expression levels were validated in Al2O3 NPs exposed A549 cells and mice lung tissues, most of which showed consistent trends in regulation. Gene-transcription factor network analysis coupled with cell- and animal-based assays demonstrated that the genes encoding PTPN6, RTN4, BAX and IER play a role in the biological responses induced by the nanoparticle exposure, which caused cell death and cell cycle arrest in the G2/S phase. Further, down-regulated PTPN6 expression demonstrated a core role in the network, thus expression level of PTPN6 was rescued by plasmid transfection, which showed ameliorative effects of A549 cells against cell death and cell cycle arrest. These results demonstrate the feasibility of using gene expression profiling to predict cellular responses induced by nanomaterials, which could be used to develop a comprehensive knowledge of nanotoxicity.
Collapse
Affiliation(s)
- Xiaobo Li
- a Key Laboratory of Environmental Medicine Engineering, Ministry of Education , School of Public Health, Southeast University , Nanjing , China
| | - Chengcheng Zhang
- a Key Laboratory of Environmental Medicine Engineering, Ministry of Education , School of Public Health, Southeast University , Nanjing , China
| | - Qian Bian
- b Department of Toxicology and Function Assessment , Jiangsu Provincial Center for Disease Prevention and Control , Nanjing , China
| | - Na Gao
- c Institute of Bioinformatics, Heinrich Heine University , Dusseldorf , Germany
| | - Xin Zhang
- a Key Laboratory of Environmental Medicine Engineering, Ministry of Education , School of Public Health, Southeast University , Nanjing , China
| | - Qingtao Meng
- a Key Laboratory of Environmental Medicine Engineering, Ministry of Education , School of Public Health, Southeast University , Nanjing , China
| | - Shenshen Wu
- a Key Laboratory of Environmental Medicine Engineering, Ministry of Education , School of Public Health, Southeast University , Nanjing , China
| | - Shizhi Wang
- a Key Laboratory of Environmental Medicine Engineering, Ministry of Education , School of Public Health, Southeast University , Nanjing , China
| | - Yankai Xia
- d Key Laboratory of Modern Toxicology of Ministry of Education , School of Public Health, Nanjing Medical University , Nanjing , China , and
| | - Rui Chen
- a Key Laboratory of Environmental Medicine Engineering, Ministry of Education , School of Public Health, Southeast University , Nanjing , China
- e State Key Laboratory of Bioelectronics , Southeast University , Nanjing , China
| |
Collapse
|
13
|
Platinum-zoledronate complex blocks gastric cancer cell proliferation by inducing cell cycle arrest and apoptosis. Tumour Biol 2016; 37:10981-92. [PMID: 26891667 DOI: 10.1007/s13277-016-4977-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2015] [Accepted: 02/04/2016] [Indexed: 12/16/2022] Open
Abstract
A series of novel dinuclear platinum complexes based on the bisphosphonate ligands have been synthesized and characterized in our recent study. For the purpose of discovering the pharmacology and action mechanisms of this kind of compounds, the most potent compound [Pt(en)]2ZL was selected for systematic investigation. In the present study, the inhibition effect on the human gastric cancer cell lines SGC7901 and action mechanism of [Pt(en)]2ZL were investigated. The traditional 3-[4,5-dimethyl-2-thiazolyl]-2,5-diphenyl-2-tetrazolium bromide (MTT) assay and colony formation assay were carried out to study the effect of [Pt(en)]2ZL on the cell viability and proliferation capacity, respectively. The senescence-associated β-galactosidase staining and immunofluorescence staining were also performed to assess the cell senescence and microtubule polymerization. Fluorescence staining and flow cytometry (FCM) were used to monitor the cell cycle distribution and apoptosis, and Western blot analysis was applied to examine the expression of several apoptosis-related proteins. The results demonstrated that [Pt(en)]2ZL exhibited remarkable cytotoxicity and anti-proliferative effects on the SGC7901 cells in a dose- and time-dependent manner, and it also induced cell senescence and abnormal microtubule assembly. The cell apoptosis and cell cycle arrest induced by [Pt(en)]2ZL were also observed with the fluorescence staining and FCM. The expressions of cell cycle regulators (p53, p21, cyclin D1, cyclin E, and cyclin-dependent kinase (CDK)2) and apoptosis-related proteins (Bcl-2, Bax, caspase-3, poly ADP ribose polymerase (PARP), and survivin) were regulated by the treatment of [Pt(en)]2ZL, resulting in the cell cycle arrest and apoptosis. Therefore, [Pt(en)]2ZL exerted anti-tumor effect on the gastric cancer via inducing cell cycle arrest at G1/S phase and apoptosis.
Collapse
|