1
|
Chutoe C, Inson I, Krobthong S, Phueakphud N, Khunluck T, Wongtrakoongate P, Charoenphandhu N, Lertsuwan K. Combinatorial effects of cannabinoid receptor 1 and 2 agonists on characteristics and proteomic alteration in MDA-MB-231 breast cancer cells. PLoS One 2024; 19:e0312851. [PMID: 39527598 PMCID: PMC11554208 DOI: 10.1371/journal.pone.0312851] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Accepted: 10/14/2024] [Indexed: 11/16/2024] Open
Abstract
Breast cancer is the most common cancer diagnosed in women worldwide. However, the effective treatment for breast cancer progression is still being sought. The activation of cannabinoid receptor (CB) has been shown to negatively affect breast cancer cell survival. Our previous study also reported that breast cancer cells responded to various combinations of CB1 and CB2 agonists differently. Nonetheless, the mechanism underlying this effect and whether this phenomenon can be seen in other cancer characteristics remain unknown. Therefore, this study aims to further elucidate the effects of highly selective CB agonists and their combination on triple-negative breast cancer proliferation, cell cycle progression, invasion, lamellipodia formation as well as proteomic profile of MDA-MB-231 breast cancer cells. The presence of CB agonists, specifically a 2:1 (ACEA: GW405833) combination, prominently inhibited colony formation and induced the S-phase cell cycle arrest in MDA-MB-231 cells. Furthermore, cell invasion ability and lamellipodia formation of MDA-MB-231 were also attenuated by the exposure of CB agonists and their 2:1 combination ratio. Our proteomic analysis revealed proteomic profile alteration in MDA-MB-231 upon CB exposure that potentially led to breast cancer suppression, such as ZPR1/SHC1/MAPK-mediated cell proliferation and AXL/VAV2/RAC1-mediated cell motility pathways. Our findings showed that selective CB agonists and their combination suppressed breast cancer characteristics in MDA-MB-231 cells. The exposure of CB agonists also altered the proteomic profile of MDA-MB-231, which could lead to cell proliferation and motility suppression.
Collapse
Affiliation(s)
- Chartinun Chutoe
- Department of Biochemistry, Faculty of Science, Mahidol University, Bangkok, Thailand
| | - Ingon Inson
- Department of Biochemistry, Faculty of Science, Mahidol University, Bangkok, Thailand
| | - Sucheewin Krobthong
- Center for Neuroscience, Faculty of Science, Mahidol University, Bangkok, Thailand
| | - Nut Phueakphud
- Department of Biochemistry, Faculty of Science, Mahidol University, Bangkok, Thailand
- Center for Neuroscience, Faculty of Science, Mahidol University, Bangkok, Thailand
| | - Tueanjai Khunluck
- Center of Calcium and Bone Research (COCAB), Faculty of Science, Mahidol University, Bangkok, Thailand
- HRH Princess Chulabhorn College of Medical Science, Chulabhorn Royal Academy, Bangkok, Thailand
| | - Patompon Wongtrakoongate
- Department of Biochemistry, Faculty of Science, Mahidol University, Bangkok, Thailand
- Center for Neuroscience, Faculty of Science, Mahidol University, Bangkok, Thailand
| | - Narattaphol Charoenphandhu
- Center of Calcium and Bone Research (COCAB), Faculty of Science, Mahidol University, Bangkok, Thailand
- Department of Physiology, Faculty of Science, Mahidol University, Bangkok, Thailand
- Institute of Molecular Biosciences, Mahidol University, Nakhon Pathom, Thailand
- The Academy of Science, The Royal Society of Thailand, Bangkok, Thailand
| | - Kornkamon Lertsuwan
- Department of Biochemistry, Faculty of Science, Mahidol University, Bangkok, Thailand
- Center of Calcium and Bone Research (COCAB), Faculty of Science, Mahidol University, Bangkok, Thailand
| |
Collapse
|
2
|
Reffai A, Hori M, Adusumilli R, Bermudez A, Bouzoubaa A, Pitteri S, Bennani Mechita M, Mallick P. A Proteomic Analysis of Nasopharyngeal Carcinoma in a Moroccan Subpopulation. Cancers (Basel) 2024; 16:3282. [PMID: 39409902 PMCID: PMC11476039 DOI: 10.3390/cancers16193282] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2024] [Revised: 09/14/2024] [Accepted: 09/19/2024] [Indexed: 10/20/2024] Open
Abstract
BACKGROUND Nasopharyngeal carcinoma (NPC) is a distinct cancer of the head and neck that is highly prevalent in Southeast Asia and North Africa. Though an extensive analysis of environmental and genetic contributors has been performed, very little is known about the proteome of this disease. A proteomic analysis of formalin-fixed paraffin-embedded (FFPE) tissues can provide valuable information on protein expression and molecular patterns for both increasing our understanding of the disease and for biomarker discovery. To date, very few NPC proteomic studies have been performed, and none focused on patients from Morocco and North Africa. METHODS Label-free Liquid Chromatography-Tandem Mass Spectrometry (LC-MS/MS) was used to perform a proteomic analysis of FFPE tissue samples from a cohort of 41 NPC tumor samples of Morocco and North Africa origins. The LC-MS/MS data from this cohort were analyzed alongside 21 healthy controls using MaxQuant 2.4.2.0. A differential expression analysis was performed using the MSstats package in R. Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) functional annotations were carried out using the DAVID bioinformatic tool. RESULTS 3341 proteins were identified across our NPC cases, revealing three main clusters and five DEPs with prognostic significance. The sex disparity of NPC was investigated from a proteomic perspective in which 59 DEPs were found between males and females, with significantly enriched terms associated with the immune response and gene expression. Furthermore, 26 DEPs were observed between patients with early and advanced stages of NPC with a significant cluster related to the immune response, implicating up-regulated DEPs such as IGHA, IGKC, and VAT1. Across both datasets, 6532 proteins were quantified between NPC patients and healthy controls. Among them, 1507 differentially expressed proteins (DEPs) were observed. GO and KEGG pathway analyses showed enriched terms of DEPs related to increased cellular activity, cell proliferation, and survival. PI3K and MAPK proteins as well as RAC1 BCL2 and PPIA were found to be overexpressed between cancer tissues and healthy controls. EBV infection was also one of the enriched pathways implicating its latent genes like LMP1 and LMP2 that activate several proteins and signaling pathways including NF-Kappa B, MAPK, and JAK-STAT pathways. CONCLUSION Our findings unveil the proteomic landscape of NPC for the first time in the Moroccan population. These studies additionally may provide a foundation for identifying potential biomarkers. Further research is still needed to help develop tools for the early diagnosis and treatment of NPC in Moroccan and North African populations.
Collapse
Affiliation(s)
- Ayman Reffai
- Intelligent Automation and BioMed Genomics Laboratory, Biology Department, Faculty of Sciences and Techniques of Tangier, Abdelmalek Essaadi University-Tetouan, Tangier 90000, Morocco
- Canary Center for Cancer Early Detection, School of Medicine, Stanford University, Stanford, CA 94305, USA
| | - Michelle Hori
- Canary Center for Cancer Early Detection, School of Medicine, Stanford University, Stanford, CA 94305, USA
| | - Ravali Adusumilli
- Canary Center for Cancer Early Detection, School of Medicine, Stanford University, Stanford, CA 94305, USA
| | - Abel Bermudez
- Canary Center for Cancer Early Detection, School of Medicine, Stanford University, Stanford, CA 94305, USA
| | | | - Sharon Pitteri
- Canary Center for Cancer Early Detection, School of Medicine, Stanford University, Stanford, CA 94305, USA
| | - Mohcine Bennani Mechita
- Intelligent Automation and BioMed Genomics Laboratory, Biology Department, Faculty of Sciences and Techniques of Tangier, Abdelmalek Essaadi University-Tetouan, Tangier 90000, Morocco
| | - Parag Mallick
- Canary Center for Cancer Early Detection, School of Medicine, Stanford University, Stanford, CA 94305, USA
| |
Collapse
|
3
|
Franci L, Vallini G, Bertolino FM, Cicaloni V, Inzalaco G, Cicogni M, Tinti L, Calabrese L, Barone V, Salvini L, Rubegni P, Galvagni F, Chiariello M. MAPK15 controls cellular responses to oxidative stress by regulating NRF2 activity and expression of its downstream target genes. Redox Biol 2024; 72:103131. [PMID: 38555711 PMCID: PMC10998232 DOI: 10.1016/j.redox.2024.103131] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Accepted: 03/22/2024] [Indexed: 04/02/2024] Open
Abstract
Oxidation processes in mitochondria and different environmental insults contribute to unwarranted accumulation of reactive oxygen species (ROS). These, in turn, rapidly damage intracellular lipids, proteins, and DNA, ultimately causing aging and several human diseases. Cells have developed different and very effective systems to control ROS levels. Among these, removal of excessive amounts is guaranteed by upregulated expression of various antioxidant enzymes, through activation of the NF-E2-Related Factor 2 (NRF2) protein. Here, we show that Mitogen Activated Protein Kinase 15 (MAPK15) controls the transactivating potential of NRF2 and, in turn, the expression of its downstream target genes. Specifically, upon oxidative stress, MAPK15 is necessary to increase NRF2 expression and nuclear translocation, by inducing its activating phosphorylation, ultimately supporting transactivation of cytoprotective antioxidant genes. Lungs are continuously exposed to oxidative damages induced by environmental insults such as air pollutants and cigarette smoke. Interestingly, we demonstrate that MAPK15 is very effective in supporting NRF2-dependent antioxidant transcriptional response to cigarette smoke of epithelial lung cells. Oxidative damage induced by cigarette smoke indeed represents a leading cause of disability and death worldwide by contributing to the pathogenesis of different chronic respiratory diseases and lung cancer. Therefore, the development of novel therapeutic strategies able to modulate cellular responses to oxidative stress would be highly beneficial. Our data contribute to the necessary understanding of the molecular mechanisms behind such responses and identify new potentially actionable targets.
Collapse
Affiliation(s)
- Lorenzo Franci
- Istituto di Fisiologia Clinica (IFC), Consiglio Nazionale Delle Ricerche (CNR), Siena, Italy; Core Research Laboratory (CRL), Istituto per lo Studio, la Prevenzione e la Rete Oncologica (ISPRO), Siena, Italy.
| | - Giulia Vallini
- Department of Medical Biotechnologies, University of Siena, Siena, Italy; Section of Dermatology, Department of Medical, Surgical and Neurological Science, University of Siena, Italy.
| | - Franca Maria Bertolino
- Istituto di Fisiologia Clinica (IFC), Consiglio Nazionale Delle Ricerche (CNR), Siena, Italy; Core Research Laboratory (CRL), Istituto per lo Studio, la Prevenzione e la Rete Oncologica (ISPRO), Siena, Italy; Department of Medical Biotechnologies, University of Siena, Siena, Italy.
| | | | - Giovanni Inzalaco
- Istituto di Fisiologia Clinica (IFC), Consiglio Nazionale Delle Ricerche (CNR), Siena, Italy; Core Research Laboratory (CRL), Istituto per lo Studio, la Prevenzione e la Rete Oncologica (ISPRO), Siena, Italy; Department of Medical Biotechnologies, University of Siena, Siena, Italy.
| | | | - Laura Tinti
- Toscana Life Sciences Foundation, Siena, Italy.
| | - Laura Calabrese
- Section of Dermatology, Department of Medical, Surgical and Neurological Science, University of Siena, Italy.
| | - Virginia Barone
- Department of Molecular and Developmental Medicine, University of Siena, Siena, Italy.
| | | | - Pietro Rubegni
- Section of Dermatology, Department of Medical, Surgical and Neurological Science, University of Siena, Italy.
| | - Federico Galvagni
- Department of Biotechnology, Chemistry and Pharmacy, University of Siena, Italy.
| | - Mario Chiariello
- Istituto di Fisiologia Clinica (IFC), Consiglio Nazionale Delle Ricerche (CNR), Siena, Italy; Core Research Laboratory (CRL), Istituto per lo Studio, la Prevenzione e la Rete Oncologica (ISPRO), Siena, Italy.
| |
Collapse
|
4
|
Hadwiger JA, Aranda RG, Fatima S. Atypical MAP kinases - new insights and directions from amoeba. J Cell Sci 2023; 136:jcs261447. [PMID: 37850857 PMCID: PMC10617611 DOI: 10.1242/jcs.261447] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2023] Open
Abstract
Mitogen-activated protein kinases (MAPKs) have been the focus of many studies over the past several decades, but the understanding of one subgroup of MAPKs, orthologs of MAPK15, known as atypical MAPKs, has lagged behind others. In most organisms, specific activating signals or downstream responses of atypical MAPK signaling pathways have not yet been identified even though these MAPKs are associated with many eukaryotic processes, including cancer and embryonic development. In this Review, we discuss recent studies that are shedding new light on both the regulation and function of atypical MAPKs in different organisms. In particular, the analysis of the atypical MAPK in the amoeba Dictyostelium discoideum has revealed important roles in chemotactic responses and gene regulation. The rapid and transient phosphorylation of the atypical MAPK in these responses suggest a highly regulated activation mechanism in vivo despite the ability of atypical MAPKs to autophosphorylate in vitro. Atypical MAPK function can also impact the activation of other MAPKs in amoeba. These advances are providing new perspectives on possible MAPK roles in animals that have not been previously considered, and this might lead to the identification of potential targets for regulating cell movement in the treatment of diseases.
Collapse
Affiliation(s)
- Jeffrey A. Hadwiger
- Department of Microbiology and Molecular Genetics, Oklahoma State University, Stillwater, OK 74078-3020, USA
| | - Ramee G. Aranda
- Department of Microbiology and Molecular Genetics, Oklahoma State University, Stillwater, OK 74078-3020, USA
| | - Saher Fatima
- Department of Microbiology and Molecular Genetics, Oklahoma State University, Stillwater, OK 74078-3020, USA
| |
Collapse
|
5
|
Fan H, Li J, Manuel AM, Zhao Z. Enzalutamide-induced signatures revealed by epigenetic plasticity using single-cell multi-omics sequencing in prostate cancer. MOLECULAR THERAPY. NUCLEIC ACIDS 2023; 31:648-661. [PMID: 36910711 PMCID: PMC9995291 DOI: 10.1016/j.omtn.2023.02.022] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Accepted: 02/15/2023] [Indexed: 02/19/2023]
Abstract
Prostate cancer is morphologically and molecularly heterogeneous, which poses obstacles for early diagnosis and treatment. Advancements in understanding the heterogeneity of prostate cancer will help navigate through these challenges and ultimately benefit patients. In this study, we integrated single-cell sequencing for transposase-accessible chromatin and whole transcriptome in prostate cancer cell lines, aiming to decode the epigenetic plasticity upon enzalutamide (ENZ) treatment. By comparing the cell populations representing early-treatment response or resistance to the initial tumor cells, we identified seven signature gene sets; they present consistent trends of chromatin closing co-occurred with down-regulated genes during early response and chromatin opening with up-regulated genes upon maintaining drug resistance. In the molecular signatures, we found genes ZNF337, MAPK15, and ESRRG are favorable in progression-free prognosis during early response, while genes CCDC150, CCDC18, and POC1A marked poor prognosis underpinning the pre-existing drug resistance in The Cancer Genome Atlas prostate adenocarcinoma cohort. Ultimately, drug-target analyses nominated combinatory drug candidates to either enhance early-treatment response or potentially overcome ENZ resistance. Together, our integrative, single-cell multi-omics approach in pre-clinical models is effective in identifying informative signatures from complex molecular events, illustrating diverse drug responses in prostate cancer, and invoking novel combinatory drug strategies to inform clinical decision making.
Collapse
Affiliation(s)
- Huihui Fan
- Center for Precision Health, School of Biomedical Informatics, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA.,Department of Neurology, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Jinze Li
- Environmental and Occupational Health Sciences, School of Public Health, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Astrid M Manuel
- Center for Precision Health, School of Biomedical Informatics, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Zhongming Zhao
- Center for Precision Health, School of Biomedical Informatics, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA.,Human Genetics Center, School of Public Health, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA.,MD Anderson Cancer Center, University of Texas Health Graduate School of Biomedical Sciences, Houston, TX 77030, USA
| |
Collapse
|
6
|
Arjmand B, Rezaei-Tavirani M, Hamzeloo-Moghadam M, Razzaghi Z, Khodadoost M, Okhovatian F, Zamanian-Azodi M, Ansari M. Hypofractionated Radiation Versus Conventional Fractionated Radiation: A Network Analysis. J Lasers Med Sci 2022; 13:e39. [PMID: 36743138 PMCID: PMC9841380 DOI: 10.34172/jlms.2022.39] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2022] [Accepted: 08/04/2022] [Indexed: 01/27/2023]
Abstract
Introduction: Conventional fractionation (CF) and hypofractionation (HF) are two radiotherapy methods against cancer, which are applied in medicine. Understanding the efficacy and molecular mechanism of the two methods implies more investigations. In the present study, proteomic findings about the mentioned methods relative to the controls were analyzed via network analysis. Methods: The significant differentially expressed proteins (DEPs) of prostate cancer (PCa) cell line DU145 in response to CF and HF radiation therapy versus controls were extracted from the literature. The protein-protein interaction (PPI) networks were constructed via the STRING database via Cytoscape software. The networks were analyzed by "NetworkAnalyzer" to determine hub DEPs. Results: 126 and 63 significant DEPs were identified for treated DU145 with CF and HF radiation respectively. The PPI networks were constructed by the queried DEPs plus 100 first neighbors. ALB, CD44, THBS1, EPCAM, F2, KRT19, and MCAM were highlighted as common hubs. VTM, OCLN, HSPB1, FLNA, AHSG, and SERPINC1 appeared as the discriminator hub between the studied cells. Conclusion: 70% of the hubs were common between CF and HF conditions, and they induced radio-resistance activity in the survived cells. Six central proteins which discriminate the function of the two groups of the irradiated cells were introduced. On the basis of these findings, it seems that DU145-CF cells, relative to the DU145-UF cells, are more radio-resistant.
Collapse
Affiliation(s)
- Babak Arjmand
- Cell Therapy and Regenerative Medicine Research Center, Endocrinology and Metabolism Molecular-Cellular Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Mostafa Rezaei-Tavirani
- Proteomics Research Center, Faculty of Paramedical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran,Correspondence to Mostafa Rezaei-Tavirani, E
| | - Maryam Hamzeloo-Moghadam
- Traditional Medicine and Materia Medical Research Center, Department of Traditional Pharmacy, School of Traditional Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Zahra Razzaghi
- Laser Application in Medical Sciences Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mahmood Khodadoost
- Traditional Medicine and Materia Medical Research Center, Department of Traditional Pharmacy, School of Traditional Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Farshad Okhovatian
- Physiotherapy Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mona Zamanian-Azodi
- Proteomics Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mojtaba Ansari
- Faculty of Medicine, Imam Hosein Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
7
|
Franci L, Tubita A, Bertolino FM, Palma A, Cannino G, Settembre C, Rasola A, Rovida E, Chiariello M. MAPK15 protects from oxidative stress-dependent cellular senescence by inducing the mitophagic process. Aging Cell 2022; 21:e13620. [PMID: 35642724 PMCID: PMC9282834 DOI: 10.1111/acel.13620] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Revised: 04/06/2022] [Accepted: 04/12/2022] [Indexed: 01/18/2023] Open
Abstract
Mitochondria are the major source of reactive oxygen species (ROS), whose aberrant production by dysfunctional mitochondria leads to oxidative stress, thus contributing to aging as well as neurodegenerative disorders and cancer. Cells efficiently eliminate damaged mitochondria through a selective type of autophagy, named mitophagy. Here, we demonstrate the involvement of the atypical MAP kinase family member MAPK15 in cellular senescence, by preserving mitochondrial quality, thanks to its ability to control mitophagy and, therefore, prevent oxidative stress. We indeed demonstrate that reduced MAPK15 expression strongly decreases mitochondrial respiration and ATP production, while increasing mitochondrial ROS levels. We show that MAPK15 controls the mitophagic process by stimulating ULK1‐dependent PRKN Ser108 phosphorylation and inducing the recruitment of damaged mitochondria to autophagosomal and lysosomal compartments, thus leading to a reduction of their mass, but also by participating in the reorganization of the mitochondrial network that usually anticipates their disposal. Consequently, MAPK15‐dependent mitophagy protects cells from accumulating nuclear DNA damage due to mitochondrial ROS and, consequently, from senescence deriving from this chronic DNA insult. Indeed, we ultimately demonstrate that MAPK15 protects primary human airway epithelial cells from senescence, establishing a new specific role for MAPK15 in controlling mitochondrial fitness by efficient disposal of old and damaged organelles and suggesting this kinase as a new potential therapeutic target in diverse age‐associated human diseases.
Collapse
Affiliation(s)
- Lorenzo Franci
- Istituto di Fisiologia Clinica (IFC) Consiglio Nazionale delle Ricerche (CNR) Siena Italy
- Core Research Laboratory (CRL) Istituto per lo Studio la Prevenzione e la Rete Oncologica (ISPRO) Siena Italy
- Department of Medical Biotechnologies University of Siena Siena Italy
| | - Alessandro Tubita
- Department of Experimental and Clinical Biomedical Sciences University of Firenze Firenze Italy
| | - Franca Maria Bertolino
- Istituto di Fisiologia Clinica (IFC) Consiglio Nazionale delle Ricerche (CNR) Siena Italy
- Core Research Laboratory (CRL) Istituto per lo Studio la Prevenzione e la Rete Oncologica (ISPRO) Siena Italy
| | - Alessandro Palma
- Department of Onco‐hematology, Gene and Cell Therapy Bambino Gesù Children’s Hospital–IRCCS Rome Italy
| | - Giuseppe Cannino
- Department of Biomedical Sciences University of Padova Padova Italy
| | - Carmine Settembre
- Telethon Institute of Genetics and Medicine (TIGEM) Pozzuoli Italy
- Department of Clinical Medicine and Surgery University of Napoli Federico II Napoli Italy
| | - Andrea Rasola
- Department of Biomedical Sciences University of Padova Padova Italy
| | - Elisabetta Rovida
- Department of Experimental and Clinical Biomedical Sciences University of Firenze Firenze Italy
| | - Mario Chiariello
- Istituto di Fisiologia Clinica (IFC) Consiglio Nazionale delle Ricerche (CNR) Siena Italy
- Core Research Laboratory (CRL) Istituto per lo Studio la Prevenzione e la Rete Oncologica (ISPRO) Siena Italy
| |
Collapse
|
8
|
Luo H, Ge H. Application of Proteomics in the Discovery of Radiosensitive Cancer Biomarkers. Front Oncol 2022; 12:852791. [PMID: 35280744 PMCID: PMC8904368 DOI: 10.3389/fonc.2022.852791] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Accepted: 02/04/2022] [Indexed: 12/21/2022] Open
Abstract
Radiation therapy remains an important component of cancer treatment. Gene-encoded proteins were the actual executors of cellular functions. Proteomic was a novel technology that can systematically analysis protein composition and measure their levels of change, this was a high throughput method, and were the import tools in the post genomic era. In recent years, rapid progress of proteomic have been made in the study of cancer mechanism, diagnosis, and treatment. This article elaborates current advances and future directions of proteomics in the discovery of radiosensitive cancer biomarkers.
Collapse
Affiliation(s)
- Hui Luo
- Department of Radiation Oncology, The Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou, China
| | - Hong Ge
- Department of Radiation Oncology, The Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou, China
| |
Collapse
|
9
|
Exploring the potential mechanism of Rhodomyrtus tomentosa (Ait.) Hassk fruit phenolic rich extract on ameliorating nonalcoholic fatty liver disease by integration of transcriptomics and metabolomics profiling. Food Res Int 2022; 151:110824. [PMID: 34980375 DOI: 10.1016/j.foodres.2021.110824] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Accepted: 11/23/2021] [Indexed: 12/16/2022]
Abstract
Nonalcoholic fatty liver disease (NAFLD), as the commonest form of chronic liver disease, is accompanied by liver oxidative stress and inflammatory responses. Rhodomyrtus tomentosa (Ait.) Hassk fruit phenolic rich extract (RTE) possesses multiple pharmacological effects in management of chronic diseases. In this study, the liver-protective effect of RTE on mice with high-fat-diet (HFD)-induced NAFLD was investigated for the first time, and the underlying molecular mechanism was explored via integration of transcriptomics and metabolomics. The results showed that RTE mitigated liver damage, which was evidenced by declined inflammatory cell infiltration in liver, decreased liver function markers, oxidative stress indexes, lipid profile levels and inflammatory cytokines levels. The differential metabolites by metabonomics illustrated supplementation of RTE affected metabolomics pathways including tryptophan metabolism, alanine, aspartate and glutamate metabolism, D-glutamine and D-glutamate metabolism, cysteine and methionine metabolism, arginine and proline metabolism, which are all involved in oxidative stress and inflammation. Furthermore, the five differential expression genes (DEGs) through liver transcriptomics were screened and recognized, namely Tnfrsf21, Ifit1, Inhbb, Mapk15 and Gadd45g, which revealed that HFD induced Cytokine-cytokine receptor interaction pathway, NF-κB signaling pathway NOD-like receptor pathway, TNF signaling pathway. Integrated analysis of transcriptomics and metabolomics confirmed the supplementation of RTE had significantly regulatory effects on the metabolic pathways involved in inflammatory responses. Additionally, RT-PCR and western blot authenticated RTE intervention regulated the mRNA levels of liver genes involved in inflammation response and inhibited the liver endotoxin-TLR4-NF-κB pathway triggered by HFD, thus alleviating NAFLD. Our findings strongly support the possibility that RTE can be regarded as a potential therapeutic method for obesity-associated NAFLD.
Collapse
|
10
|
Yan H, Zhou Y, Chen Z, Yan X, Zhu L. Long non-coding RNA HCG11 enhances osteosarcoma phenotypes by sponging miR-1245b-5p that directly inhibits plakophilin 2. Bioengineered 2021; 13:140-154. [PMID: 34949159 PMCID: PMC8805843 DOI: 10.1080/21655979.2021.2010367] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Long non-coding RNA (lncRNA) HCG11 can regulate various cancers through the ceRNA network. However, its role in osteosarcoma (OS) remains unknown. The HOS and Saos-2 cell lines were used for in vitro analyses. HCG11 and plakophilin 2 (PKP2) silencers, a miR-1245b-5p mimic, and a miR-1245b-5p inhibitor were utilized for the regulation analysis of lncRNA HCG11, miR-1245b-5p, and PKP2. Cell Counting Kit-8, wound healing, and transwell assays were used for cell proliferation, migration, and invasion analyses, and caspase-3 activity assay was used to measure cell apoptosis. The expression levels of lncRNA HCG11, miR-1245b-5p, and PKP2 were evaluated by quantitative real-time PCR and Western blotting. The distribution of lncRNA HCG11 was assessed using the RNA-FISH assay. The sponging and targeting roles of HCG11 and PKP2 on miR-1245b-5p were confirmed by dual-luciferase reporter analysis. An RNA immunoprecipitation assay was used to assess the binding between lncRNA HCG11 and miRNA-1245b-5p. We found that the lncRNA HCG11 was significantly upregulated in OS. LncRNA HCG11 silencing inhibits OS progression by repressing cell proliferation, migration, and invasion, and promoting cell apoptosis. RNA-FISH analysis indicated that lncRNA HCG11 was located in the cytoplasm. Mechanistic experiments showed that lncRNA HCG11 sponges miR-1245b-5p and negatively regulates miR-1245b-5p expression. Upregulated lncRNA HCG11 promotes proliferation, migration, and invasion, and inhibits apoptosis by inhibiting miR-1245b-5p in OS cells. PKP2 was verified as a target gene of miR-1245b-5p. Upregulated PKP2 promotes proliferation, migration, and invasion, and inhibits apoptosis by inhibiting miR-1245b-5p in OS. In conclusion, the HCG11/miR-1245b-5p/PKP2 axis promotes OS expression by promoting cell proliferation, migration, and invasion, and inhibiting apoptosis.
Collapse
Affiliation(s)
- Hao Yan
- Department of Spinal Surgery, Hubei 672 Orthopaedics Hospital of Integrated Chinese and Western Medicine, Wuhan, Hubei, China
| | - Yong Zhou
- Department of Oncology, Hubei Provincial Hospital of TCM, Wuhan, Hubei, China
| | - Zhujiang Chen
- Department of Spinal Surgery, Hubei 672 Orthopaedics Hospital of Integrated Chinese and Western Medicine, Wuhan, Hubei, China
| | - Xiaokang Yan
- Department of Orthopaedics, Hubei 672 Orthopaedics Hospital of Integrated Chinese and Western Medicine, Wuhan, Hubei, China
| | - Ling Zhu
- Department of Spinal Surgery, Hubei 672 Orthopaedics Hospital of Integrated Chinese and Western Medicine, Wuhan, Hubei, China
| |
Collapse
|
11
|
Shen L, Li C, Chen F, Shen L, Li Z, Li N. CRISPR/Cas9 genome-wide screening identifies LUC7L2 that promotes radioresistance via autophagy in nasopharyngeal carcinoma cells. Cell Death Discov 2021; 7:392. [PMID: 34907164 PMCID: PMC8671510 DOI: 10.1038/s41420-021-00783-8] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Revised: 11/23/2021] [Accepted: 12/01/2021] [Indexed: 12/18/2022] Open
Abstract
Radioresistance emerges as the major obstacle to nasopharyngeal carcinoma (NPC) treatment, further understanding of underlying mechanisms is necessary to overcome the radioresistance and improve the therapeutic effect. In this study, we first identified a candidate radioresistant-related gene LUC7L2 via CRISPR/Cas9 high-throughput screening and quantitative proteomic approach. Overexpression of LUC7L2 in NPC cells promoted cell viability following exposure to ionizing radiation (IR), while knockdown of LUC7L2 significantly slowed down the DNA replication and impaired cell survival, sensitized NPC-radioresistant cells to IR. Using immunoprecipitation assay, we found SQSTM1, an autophagy receptor, was a potential binding partner of LUC7L2. Down-regulation of LUC7L2 in NPC-radioresistant cells led to reduction of SQSTM1 expression and enhancement of autophagy level. Furthermore, LUC7L2 knockdown in combination with autophagy inhibitor, chloroquine (CQ), resulted in more NPC-radioresistant cell death. Besides, LUC7L2 was obviously distributed in NPC tissues, and high LUC7L2 expression correlated with shorter survival in NPC patients. Our data suggest that LUC7L2 plays a huge part in regulating radioresistance of NPC cells, and serves as a promising therapeutic target in re-sensitizing NPC to radiotherapy.
Collapse
Affiliation(s)
- Lin Shen
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, Hunan Province, 410008, China
| | - Chao Li
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, Hunan Province, 410008, China
| | - Fang Chen
- Hunan Key Laboratory of Molecular Precision Medicine, Xiangya Hospital, Central South University, Changsha, Hunan Province, 410008, China
| | - Liangfang Shen
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, Hunan Province, 410008, China
| | - Zhanzhan Li
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, Hunan Province, 410008, China
| | - Na Li
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, Hunan Province, 410008, China.
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan Province, 410008, China.
| |
Collapse
|
12
|
Cheng Y, Wang G, Zhao L, Dai S, Han J, Hu X, Zhou C, Wang F, Ma H, Li B, Meng Z. Periplocymarin Induced Colorectal Cancer Cells Apoptosis Via Impairing PI3K/AKT Pathway. Front Oncol 2021; 11:753598. [PMID: 34900704 PMCID: PMC8655334 DOI: 10.3389/fonc.2021.753598] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Accepted: 11/04/2021] [Indexed: 12/18/2022] Open
Abstract
Colorectal cancer (CRC) is one of the most common cancers worldwide, and approximately one-third of CRC patients present with metastatic disease. Periplocymarin (PPM), a cardiac glycoside isolated from Periploca sepium, is a latent anticancer compound. The purpose of this study was to explore the effect of PPM on CRC cells. CRC cells were treated with PPM and cell viability was evaluated by CCK-8 assay. Flow cytometry and TUNEL staining were performed to assess cell cycle and apoptosis. Quantitative proteomics has been used to check the proteins differentially expressed by using tandem mass tag (TMT) labeling and liquid chromatography–tandem mass spectrometry. Bioinformatic analysis was undertaken to identify the biological processes that these differentially expressed proteins are involved in. Gene expression was analyzed by western blotting. The effect of PPM in vivo was primarily checked in a subcutaneous xenograft mouse model of CRC, and the gene expression of tumor was checked by histochemistry staining. PPM could inhibit the proliferation of CRC cells in a dose-dependent manner, induce cell apoptosis and promote G0/G1 cell cycle arrest. A total of 539 proteins were identified differentially expressed following PPM treatment, where among those there were 286 genes upregulated and 293 downregulated. PPM treatment caused a pro-apoptosis gene expression profile both in vivo and in vitro, and impaired PI3K/AKT signaling pathway might be involved. In addition, PPM treatment caused less detrimental effects on blood cell, hepatic and renal function in mice, and the anti-cancer effect was found exaggerated by PPM+5-FU combination treatment. PPM may perform anti-CRC effects by promoting cell apoptosis and this might be achieved by targeting PI3K/AKT pathway. PPM might be a safe and promising anti-cancer drug that needs to be further studied.
Collapse
Affiliation(s)
- Yi Cheng
- Department of Dermatology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Guiying Wang
- Department of General Surgery, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China.,Department of Gastrointestinal Surgery, The Third Hospital of Hebei Medical University, Shijiazhuang, China
| | - Lianmei Zhao
- Scientific Research Center, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Suli Dai
- Scientific Research Center, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Jing Han
- Department of Medical Oncology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Xuhua Hu
- Department of General Surgery, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Chaoxi Zhou
- Department of General Surgery, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Feifei Wang
- Department of General Surgery, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Hongqing Ma
- Department of General Surgery, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Baokun Li
- Department of General Surgery, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Zesong Meng
- Department of General Surgery, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| |
Collapse
|
13
|
Jung I, Kim M, Rhee S, Lim S, Kim S. MONTI: A Multi-Omics Non-negative Tensor Decomposition Framework for Gene-Level Integrative Analysis. Front Genet 2021; 12:682841. [PMID: 34567063 PMCID: PMC8461247 DOI: 10.3389/fgene.2021.682841] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Accepted: 08/12/2021] [Indexed: 11/13/2022] Open
Abstract
Multi-omics data is frequently measured to enrich the comprehension of biological mechanisms underlying certain phenotypes. However, due to the complex relations and high dimension of multi-omics data, it is difficult to associate omics features to certain biological traits of interest. For example, the clinically valuable breast cancer subtypes are well-defined at the molecular level, but are poorly classified using gene expression data. Here, we propose a multi-omics analysis method called MONTI (Multi-Omics Non-negative Tensor decomposition for Integrative analysis), which goal is to select multi-omics features that are able to represent trait specific characteristics. Here, we demonstrate the strength of multi-omics integrated analysis in terms of cancer subtyping. The multi-omics data are first integrated in a biologically meaningful manner to form a three dimensional tensor, which is then decomposed using a non-negative tensor decomposition method. From the result, MONTI selects highly informative subtype specific multi-omics features. MONTI was applied to three case studies of 597 breast cancer, 314 colon cancer, and 305 stomach cancer cohorts. For all the case studies, we found that the subtype classification accuracy significantly improved when utilizing all available multi-omics data. MONTI was able to detect subtype specific gene sets that showed to be strongly regulated by certain omics, from which correlation between omics types could be inferred. Furthermore, various clinical attributes of nine cancer types were analyzed using MONTI, which showed that some clinical attributes could be well explained using multi-omics data. We demonstrated that integrating multi-omics data in a gene centric manner improves detecting cancer subtype specific features and other clinical features, which may be used to further understand the molecular characteristics of interest. The software and data used in this study are available at: https://github.com/inukj/MONTI.
Collapse
Affiliation(s)
- Inuk Jung
- Department of Computer Science and Engineering, Kyungpook National University, Daegu, South Korea
| | - Minsu Kim
- Computing and Computational Sciences Directorate, Oak Ridge National Laboratory, Oak Ridge, TN, United States
| | - Sungmin Rhee
- Department of Computer Science and Engineering, Seoul National University, Seoul, South Korea
| | - Sangsoo Lim
- Interdisciplinary Program in Bioinformatics, Seoul National University, Gwanak-Gu, Seoul, South Korea
| | - Sun Kim
- Computing and Computational Sciences Directorate, Oak Ridge National Laboratory, Oak Ridge, TN, United States.,Department of Computer Science and Engineering, Seoul National University, Seoul, South Korea.,Interdisciplinary Program in Bioinformatics, Seoul National University, Gwanak-Gu, Seoul, South Korea
| |
Collapse
|
14
|
Serum CXCL5 level is associated with tumor progression in penile cancer. Biosci Rep 2021; 41:227614. [PMID: 33458757 PMCID: PMC7843497 DOI: 10.1042/bsr20202133] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Revised: 01/09/2021] [Accepted: 01/14/2021] [Indexed: 12/01/2022] Open
Abstract
Chemokine (C-X-C motif) ligand 5 is an important regulator of tumor progression in many cancers, and could serve as potential serum cancer biomarker. Our initial analysis identified CXCL5 as a cancer-related gene highly expressed in PC. Patients with PC exhibited markedly higher preoperative serum CXCL5 levels compared with that in healthy individuals (P<0.001). The area under the curve (AUC) was 0.880 with the sensitivity of 84.0%, and specificity of 80.4% to distinguish PC. Serum CXCL5 levels were also significantly decreased following tumor resection in patients with PC (P=0.001). Preoperative serum CXCL5 level was significantly associated with clinicopathological characteristics including T stage (P=0.001), nodal status (P<0.001), and pelvic lymph node metastasis (P=0.018). Cox regression analysis showed that serum CXCL5 level could serve as an independent prognostic factor for disease-free survival with a HR of 6.363 (95% CI: 2.185–18.531, P=0.001). CXCL5 and its receptor CXCR2 exhibited correlated expression pattern in PC tissues. Differential CXCL5 expression was observed in normal penile tissues, PC cell lines, and their culture supernatants. Furthermore, knockdown of CXCL5 or CXCR2 expression markedly suppressed malignant phenotypes (cell proliferation, clonogenesis, apoptosis escape, migration, and invasion), attenuated STAT3 and AKT signaling, and reduced MMP2/9 secretion in PC cell lines. In conclusion, our findings revealed that serum CXCL5 level might serve as a potential diagnostic and prognostic cancer biomarker for penile cancer. Autocrine CXCL5/CXCR2 signaling might activate multiple downstream oncogenic signaling pathways (STAT3, AKT, MMP2/9) to promote malignant progression of PC, which may warrant further investigation in the future.
Collapse
|
15
|
Shen L, Li Z, Shen L. Quantitative Tyrosine Phosphoproteomic Analysis of Resistance to Radiotherapy in Nasopharyngeal Carcinoma Cells. Cancer Manag Res 2020; 12:12667-12678. [PMID: 33328764 PMCID: PMC7733897 DOI: 10.2147/cmar.s260028] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2020] [Accepted: 11/02/2020] [Indexed: 01/01/2023] Open
Abstract
BACKGROUND Radioresistance poses a major challenge in nasopharyngeal carcinoma (NPC) treatment. Protein tyrosine phosphorylation has emerged as a key device in the control of resistance to therapy in cancer cells. METHODS Using tandem mass tag (TMT) labeling and phospho-antibody affinity enrichment followed by high-resolution LC-MS/MS analysis, quantitative tyrosine phosphorylome analysis was performed in CNE2 (parental) and its radioresistant subline CNE2-IR. RESULTS Altogether, 233 tyrosine phosphorylation sites in 179 protein groups were identified, among which 179 sites in 140 proteins were quantified. Among the quantified proteins, 38 tyrosine phosphorylation proteins are up-regulated and 18 tyrosine phosphorylation proteins are down-regulated in CNE2-IR vs CNE2. Increased tyrosine phosphorylation in multiple receptor/protein tyrosine kinases (EPHA2, EGFR, IGF1R, ABL1 and LYN) was identified in CNE2-IR vs CNE2 cells. Intensive bioinformatic analyses revealed robust activation of multiple biological processes/pathways including E-cadherin stabilization, cell-cell adhesion, and cell junction organization in radioresistant CNE2-IR cells. Specifically, we observed that the CNE2 cells incubated with EphrinA1-Fc exhibited higher EPHA2 Y772 phosphorylation and lower E-cadherin expression, as compared with PBS control. Furthermore, an ATP-competitive EPHA2 RTK inhibitor (ALW-II-41-27, ALW) reduced EPHA2 Y772 phosphorylation and increased the expression of E-cadherin in CNE2-IR cells. Colony formation analysis showed that EFNA1 (EFNA1 is the ligand of EPHA2) treatment in CNE2 significantly promoted colony formation after 6Gy irradiation; while incubation with EPHA2 inhibitor ALW-II-41-27 in CNE2-IR cells impaired colony formation after irradiation, as compared with solvent control (DMSO). CONCLUSION In conclusion, phosphoproteomic approach allowed us to link tyrosine kinases signaling with radioresistance in NPC. Further studies are necessary to delineate the molecular function of EPHA2/E-cadherin signaling in radioresistant NPC and to explore rational combination therapy and its underlying mechanism.
Collapse
Affiliation(s)
- Lin Shen
- Department of Oncology, Xiangya Hospital, Central South University, Changsha410008, People’s Republic of China
| | - Zhanzhan Li
- Department of Oncology, Xiangya Hospital, Central South University, Changsha410008, People’s Republic of China
| | - Liangfang Shen
- Department of Oncology, Xiangya Hospital, Central South University, Changsha410008, People’s Republic of China
| |
Collapse
|
16
|
Proteomic approaches to investigate gammaherpesvirus biology and associated tumorigenesis. Adv Virus Res 2020; 109:201-254. [PMID: 33934828 DOI: 10.1016/bs.aivir.2020.10.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
The DNA viruses, Kaposi's sarcoma-associated herpesvirus (KSHV) and Epstein-Barr virus (EBV), are members of the gammaherpesvirus subfamily, a group of viruses whose infection is associated with multiple malignancies, including cancer. The primary host for these viruses is humans and, like all herpesviruses, infection with these pathogens is lifelong. Due to the persistence of gammaherpesvirus infection and the potential for cancer formation in infected individuals, there is a driving need to understand not only the biology of these viruses and how they remain undetected in host cells but also the mechanism(s) by which tumorigenesis occurs. One of the methods that has provided much insight into these processes is proteomics. Proteomics is the study of all the proteins that are encoded by a genome and allows for (i) identification of existing and novel proteins derived from a given genome, (ii) interrogation of protein-protein interactions within a system, and (iii) discovery of druggable targets for the treatment of malignancies. In this chapter, we explore how proteomics has contributed to our current understanding of gammaherpesvirus biology and their oncogenic processes, as well as the clinical applications of proteomics for the detection and treatment of gammaherpesvirus-associated cancers.
Collapse
|
17
|
Zhao Y, Zhou Q, Li N, Shen L, Li Z. Paranasal Sinus Invasion Should Be Classified as T4 Disease in Advanced Nasopharyngeal Carcinoma Patients Receiving Radiotherapy. Front Oncol 2020; 10:01465. [PMID: 33240800 PMCID: PMC7677568 DOI: 10.3389/fonc.2020.01465] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Accepted: 07/09/2020] [Indexed: 01/20/2023] Open
Abstract
In this study, we explored the association between paranasal sinus invasion and prognosis in patients with advanced nasopharyngeal carcinoma (NPC, (T3/T4N0-3M0), and we assessed the possibility of considering paranasal sinus invasion a T category in the 8th edition of the American Joint Committee on Cancer staging system. We enrolled 352 NPC patients who received intensity-modulated radiotherapy between 2008 and 2012. Clinical characteristics and follow-up data were collected. The incidence of paranasal sinus invasion was 36.4% (128 of 352 patients). Multivariate cox regression analysis indicated that paranasal sinus invasion and cervical lymphatic metastasis were independent negative prognostic factors for overall survival (OS, P=0.024, P=0.012), progression-free survival (PFS, P=0.007, P=0.007), and distant metastasis-free survival (DMFS, P=0.001, P=0.000). The gross tumor volume of the nasopharynx was an independent negative prognostic factor for OS (P=0.013). Cox regression analysis indicated that there were no significant differences in OS, PFS, DMFS, or local relapse-free survival (LRFS) between NPC patients with T4 stage disease and those with T3 and paranasal sinus invasion (P>0.05). The updated T + N staging system slightly improved the prediction of LRFS (0.649, 95% CI: 0.553-0.745) in NPC patients compared to the AJCC system (0.640, 95% CI: 0.545-0.736; P=0.023). Paranasal sinus invasion is independently associated with a poor prognosis in NPC patients. Thus, we recommend that the AJCC staging system upgrade paranasal sinus invasion to the T4 classification.
Collapse
Affiliation(s)
- Yajie Zhao
- Department of Nuclear Medicine, Xiangya Hospital, Central South University, Changsha, China
| | - Qin Zhou
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, China
| | - Na Li
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, China
| | - Liangfang Shen
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, China
| | - Zhanzhan Li
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
18
|
Mo M, Tong S, Huang W, Cai Y, Zu X, Hu X. High serum CCL20 is associated with tumor progression in penile cancer. J Cancer 2020; 11:6812-6822. [PMID: 33123272 PMCID: PMC7591991 DOI: 10.7150/jca.48939] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Accepted: 09/14/2020] [Indexed: 12/12/2022] Open
Abstract
Serum cancer biomarker has been proven to be very valuable in cancer diagnosis, disease monitoring and prognosis assessment, despite there is still a lack of serum biomarker for penile cancer (PC). Our initial analysis on public GEO dataset identified CCL20 as a top C-C motif ligand (CCL) gene enriched in PC. The patients with PC exhibited markedly higher preoperative serum CCL20 level than healthy control. The area under the curve (AUC) was 0.855 with the sensitivity of 72.4%, and specificity of 93.5% to distinguish PC. Preoperative serum CCL20 level was significantly associated with clinicopathological characteristics including T stage (P=0.005), nodal status (P=0.008), and pelvic lymph node metastasis (P=0.007). PC Patients with high serum CCL20 level had shorter disease-free survival compared to those with low level (P<0.001). Cox regression analysis showed that serum CCL20 level could serve as an independent prognostic factor for disease-free survival with a HR of 3.980 (95% CI: 1.209-13.098, P=0.023). Furthermore, CCL20 expression was observed in PC tissues and cell lines. Knockdown of CCL20 expression markedly suppressed malignant phenotypes (cell proliferation, clonogenesis, apoptosis escape, migration and invasion), attenuated STAT3 and AKT signaling and reduced MMP2/9 secretion in PC cell lines. Consistently, CCL20 and its receptor CCR6 exhibited correlated expression pattern in PC tissues. In conclusion, serum CCL20 level might serve as a potential diagnostic and prognostic cancer biomarker for PC. CCL20 might activate multiple downstream oncogenic signaling pathways (STAT3, AKT, MMP2/9) to promote malignant progression of PC, which may warrant further investigation in the future.
Collapse
Affiliation(s)
- Miao Mo
- Department of Urology, Xiangya Hospital, Central South University, Changsha, Hunan 410008, P.R. China
| | - Shiyu Tong
- Department of Urology, Xiangya Hospital, Central South University, Changsha, Hunan 410008, P.R. China
| | - Wei Huang
- Xiangya School of Medicine, Central South University, Changsha, Hunan 410008, P.R. China
| | - Yi Cai
- Department of Urology, Xiangya Hospital, Central South University, Changsha, Hunan 410008, P.R. China
| | - Xiongbing Zu
- Department of Urology, Xiangya Hospital, Central South University, Changsha, Hunan 410008, P.R. China
| | - Xiheng Hu
- Department of Urology, Xiangya Hospital, Central South University, Changsha, Hunan 410008, P.R. China
| |
Collapse
|
19
|
Pouliquen DL, Boissard A, Coqueret O, Guette C. Biomarkers of tumor invasiveness in proteomics (Review). Int J Oncol 2020; 57:409-432. [PMID: 32468071 PMCID: PMC7307599 DOI: 10.3892/ijo.2020.5075] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2020] [Accepted: 05/07/2020] [Indexed: 12/13/2022] Open
Abstract
Over the past two decades, quantitative proteomics has emerged as an important tool for deciphering the complex molecular events involved in cancers. The number of references involving studies on the cancer metastatic process has doubled since 2010, while the last 5 years have seen the development of novel technologies combining deep proteome coverage capabilities with quantitative consistency and accuracy. To highlight key findings within this huge amount of information, the present review identified a list of tumor invasive biomarkers based on both the literature and data collected on a biocollection of experimental cell lines, tumor models of increasing invasiveness and tumor samples from patients with colorectal or breast cancer. Crossing these different data sources led to 76 proteins of interest out of 1,245 mentioned in the literature. Information on these proteins can potentially be translated into clinical prospects, since they represent potential targets for the development and evaluation of innovative therapies, alone or in combination. Herein, a systematical review of the biology of each of these proteins, including their specific subcellular/extracellular or multiple localizations is presented. Finally, as an important advantage of quantitative proteomics is the ability to provide data on all these molecules simultaneously in cell pellets, body fluids or paraffin‑embedded sections of tumors/invaded tissues, the significance of some of their interconnections is discussed.
Collapse
Affiliation(s)
| | - Alice Boissard
- Paul Papin ICO Cancer Center, CRCINA, Inserm, Université d'Angers, F‑44000 Nantes, France
| | | | - Catherine Guette
- Paul Papin ICO Cancer Center, CRCINA, Inserm, Université d'Angers, F‑44000 Nantes, France
| |
Collapse
|
20
|
Jun F, Peng Z, Zhang Y, Shi D. Quantitative Proteomic Profiling Identifies SOX8 as Novel Regulator of Drug Resistance in Gestational Trophoblastic Neoplasia. Front Oncol 2020; 10:557. [PMID: 32411596 PMCID: PMC7198745 DOI: 10.3389/fonc.2020.00557] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2019] [Accepted: 03/27/2020] [Indexed: 12/15/2022] Open
Abstract
The development of drug resistance remains one of the major challenges to current chemotherapeutic regimens in gestational trophoblastic neoplasia (GTN). Further understanding on the mechanisms of drug resistance would help to develop more effective therapy to treat GTN. Herein, tandem mass tag-based (TMT) quantitative proteomic technique was used to establish drug resistance-related proteomic profiles in chemoresistant GTN cell models (JEG3/MTX, JEG3/VP16, JEG3/5-Fu). In total, we identified 5,704 protein groups, among which 4,997 proteins were quantified in JEG3 and its chemoresistant sublines. Bioinformatics analysis revealed that multiple biological processes/molecular pathways/signaling networks were involved in the regulation of drug resistance in chemoresistant JEG3 sublines. SOX8 was upregulated in all the three chemoresistant sublines, and its function was further investigated. Knockdown of SOX8 significantly reduced cell viability, impaired soft agar clonogenesis, and increased caspase-3 activities after drug treatment in JEG3 chemoresistant sublines. In addition, over-expression of SOX8 promoted cell survival, enhanced soft agar clonogenesis, and attenuated caspase-3 activities after drug treatment in GTN cells. Importantly, SOX8 might be a potential regulator of reactive oxygen species (ROS) homeostasis, as SOX8 regulated the expression of antioxidant enzymes (GPX1, HMOX1) and reduced drug-induced ROS accumulation in GTN cell models. Collectively, SOX8 might promote drug resistance through attenuating the accumulation of ROS induced by chemotherapeutic drugs in GTN cells. Targeting SOX8 might be useful to sensitize GTN cells to chemotherapy.
Collapse
Affiliation(s)
- Fu Jun
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, China.,Department of Neurosurgery, Sanbo Brain Hospital, Capital Medical University, Beijing, China
| | - Zheng Peng
- Department of Gynecology and Obstetrics, Xiangya Hospital, Central South University, Changsha, China.,Gynecological Oncology Research and Engineering Center of Hunan Province, Changsha, China
| | - Yi Zhang
- Department of Gynecology and Obstetrics, Xiangya Hospital, Central South University, Changsha, China.,Gynecological Oncology Research and Engineering Center of Hunan Province, Changsha, China
| | - Dazun Shi
- Department of Gynecology and Obstetrics, Xiangya Hospital, Central South University, Changsha, China.,Gynecological Oncology Research and Engineering Center of Hunan Province, Changsha, China
| |
Collapse
|
21
|
Su Z, Yang B, Zeng Z, Zhu S, Wang C, Lei S, Jiang Y, Lin L. Metastasis-associated gene MAPK15 promotes the migration and invasion of osteosarcoma cells via the c-Jun/MMPs pathway. Oncol Lett 2020; 20:99-112. [PMID: 32565938 PMCID: PMC7285714 DOI: 10.3892/ol.2020.11544] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2019] [Accepted: 01/14/2020] [Indexed: 12/12/2022] Open
Abstract
Osteosarcoma (OS) is the most common and destructive primary bone malignancy to affect children and adolescents. Metastases remain the primary cause of death in patients with OS. In the present study, weight gene co-expressed network analysis (WGCNA) and differentially-expressed gene analysis were used to identify key genes associated with the metastasis of OS. Reverse transcription-quantitative PCR and immunohistochemical staining were then used to detect the expression levels of these key genes in OS tissues, and to determine the hub genes of interest. Wound-healing and transwell assays, in addition to a lung metastasis model, were used to detect the effects of the hub genes on OS cell proliferation and metastasis in vitro and in vivo. Using WGCNA and differential expression analysis, deleted in lung and esophageal cancer protein 1 (DLEC1), Forkhead box J1 (FOXJ1) and mitogen-activated protein kinase 15 (MAPK15) were predicted to be key metastasis-associated genes, and highly expressed in metastatic OS tissues; among them, the protein and mRNA expression levels of MAPK15 were most significantly increased in our OS tissues from patients who exhibited metastases at diagnosis, and thus MAPK15 was determined to be a metastasis-associated hub gene to further study. Furthermore, inhibiting MAPK15 expression significantly decreased OS cell metastasis in vitro and in vivo, as well as suppressing c-Jun/matrix metalloproteinase (MMP)-associated pathways. Overexpression of MAPK15 activated the c-Jun/MMPs pathway and promoted OS cell metastasis, while inhibition of c-Jun blocked this effect. Taken together, MAPK15 was indicated to be an OS metastasis-associated gene, and was confirmed to promote the migration and invasion of OS cells via the c-Jun/MMP pathway. MAPK15 may therefore be an effective target for the treatment of OS.
Collapse
Affiliation(s)
- Zexin Su
- Department of Joint Surgery, Huadu District People's Hospital, Southern Medical University, Guangzhou, Guangdong 510800, P.R. China
| | - Bingsheng Yang
- Department of Orthopaedics, Zhujiang Hospital, Southern Medical University, Haizhu, Guangzhou, Guangdong 510282, P.R. China
| | - Zhirui Zeng
- Guizhou Provincial Key Laboratory of Pathogenesis and Drug Research on Common Chronic Diseases, Department of Physiology, School of Basic Medicine, Guizhou Medical University, Guiyang, Guizhou 550009, P.R. China
| | - Shuang Zhu
- Department of Orthopaedics, Zhujiang Hospital, Southern Medical University, Haizhu, Guangzhou, Guangdong 510282, P.R. China
| | - Chenyang Wang
- Department of Neurosurgery, Zhujiang Hospital, Neurosurgery Institute of Guangdong Province, Key Laboratory on Brain Function Repair and Rehabilitation, Southern Medical University, Guangzhou, Guangdong 510282, P.R. China
| | - Shan Lei
- Guizhou Provincial Key Laboratory of Pathogenesis and Drug Research on Common Chronic Diseases, Department of Physiology, School of Basic Medicine, Guizhou Medical University, Guiyang, Guizhou 550009, P.R. China
| | - Yongfa Jiang
- Department of Joint Surgery, Huadu District People's Hospital, Southern Medical University, Guangzhou, Guangdong 510800, P.R. China
| | - Lijun Lin
- Department of Orthopaedics, Zhujiang Hospital, Southern Medical University, Haizhu, Guangzhou, Guangdong 510282, P.R. China
| |
Collapse
|
22
|
Jun F, Peng Z, Zhang Y, Shi D. Quantitative proteomic analysis identifies novel regulators of methotrexate resistance in choriocarcinoma. Gynecol Oncol 2020; 157:268-279. [DOI: 10.1016/j.ygyno.2020.01.013] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2019] [Revised: 12/19/2019] [Accepted: 01/06/2020] [Indexed: 01/09/2023]
|
23
|
Abstract
Introduction: Nasopharyngeal carcinoma (NPC) is a distinct head and neck squamous cell carcinoma in its etiological association of Epstein-Barr virus (EBV) infection, hidden anatomical location, remarkable racial and geographical distribution, and high incidence of locoregional recurrence or metastasis. Thanks to the advancements in proteomics in recent decades, more understanding of the disease etiology, carcinogenesis, and progression has been gained, potentially deciphering the molecular characteristics of the malignancy. Areas covered: In this review, we provide an overview of the proteomic aberrations that are likely involved or drive NPC development and progression, focusing on the contributions of major EBV-encoded factors, intercommunication with environment, protein features of high metastasis and therapy resistance, and protein-protein interactions that allow NPC cells to evade immune recognition and elimination. Finally, multistep carcinogenesis and subtypes of NPC from a proteomic perspective are inquired. Expert commentary: Proteomic studies have covered various aspects involved in NPC pathogenesis, yet much remains to be uncovered. Coherent study designs, optimal conditions for obtaining high-quality data, and compelling interpretation are critical in ensuring the emergence of good science out of NPC proteomics. NPC proteogenomics and proteoform analysis are two promising fields to promote the application of the proteomic findings from bench to bedside.
Collapse
Affiliation(s)
- Zhefeng Xiao
- a NHC Key Laboratory of Cancer Proteomics , Xiangya Hospital, Central South University , Changsha , P. R. China
| | - Zhuchu Chen
- a NHC Key Laboratory of Cancer Proteomics , Xiangya Hospital, Central South University , Changsha , P. R. China
| |
Collapse
|
24
|
Bai F, Tu T, Qin F, Ma Y, Liu N, Liu Y, Liao X, Zhou S, Liu Q. Quantitative proteomics of changes in succinylated proteins expression profiling in left appendages tissue from valvular heart disease patients with atrial fibrillation. Clin Chim Acta 2019; 495:345-354. [PMID: 31059701 DOI: 10.1016/j.cca.2019.05.002] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2019] [Revised: 04/15/2019] [Accepted: 05/02/2019] [Indexed: 01/02/2023]
Abstract
BACKGROUND Previous studies have suggested that proteomic modifications are closely associated with cardiovascular diseases. The aim of this study was to identify potential mechanisms by profiling the changes in succinylated protein expression in left appendage tissues from patients with valvular heart disease and atrial fibrillation (AF). METHODS Using dimethyl labeling for relative and absolute quantification-coupled high-performance liquid chromatography-tandem mass spectrometry, we analyzed the proteomics profiles and succinylation events in 18 left atrial appendage tissue samples from patients who underwent cardiac valvular surgery, including nine patients with permanent AF and nine patients with sinus rhythm (SR). RESULTS In total, after setting the quantification ratio > 1.3 and < 1:1.3 representing the up- and downregulated cutoff values, respectively, 132 proteins were classified as targets of upregulation and 117 proteins as targets of downregulation. Within these proteins, 246 sites exhibited upregulated succinylation and 45 sites exhibited downregulated succinylation. Protein-protein interaction networks showed that the proteins exhibiting lysine succinylation and AF status were highly enriched in energy metabolism, extracellular matrix-related, and cellular structure-related proteins. These results were confirmed by western blot. CONCLUSIONS The differences in succinylation level of energy metabolism-related proteins indicates the possible involvement of these proteins in AF of valvular heart disease patients, and provide insight for further analysis of their biological functions.
Collapse
Affiliation(s)
- Fan Bai
- Department of Cardiology, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China
| | - Tao Tu
- Department of Cardiology, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China
| | - Fen Qin
- Department of Cardiology, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China
| | - Yingxu Ma
- Department of Cardiology, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China
| | - Na Liu
- Department of Cardiology, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China
| | - Yaozhong Liu
- Department of Cardiology, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China
| | - Xiaobo Liao
- Department of Cardiovascular Surgery, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China
| | - Shenghua Zhou
- Department of Cardiology, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China
| | - Qiming Liu
- Department of Cardiology, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China.
| |
Collapse
|
25
|
Peng H, Li Z, Fu J, Zhou R. Growth and differentiation factor 15 regulates PD-L1 expression in glioblastoma. Cancer Manag Res 2019; 11:2653-2661. [PMID: 31114328 PMCID: PMC6497826 DOI: 10.2147/cmar.s192095] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2018] [Accepted: 02/20/2019] [Indexed: 12/21/2022] Open
Abstract
Background: Gliomablastoma multiforme (GBM) is the most fatal form of all brain cancers in human with no successful treatment available. Programmed death-ligand 1 (PD-L1) is a coinhibitory ligand predominantly expressed by tumor cells. Growth differentiation factors (GDFs) are a subfamily of proteins belonging to the transforming growth factor beta superfamily that have functions predominantly in tissue development and cancer. Purpose: To investigat the expression of GDFs in GBMs, and explored the potential regulatory role of GDFs on PD-L1 expression in GBMs. Methods: GEO2R program were analyzed for the mRNA expression data of GDFs in GSE4290 dataset. Analysis of TCGA GBM datasets were further determined the relationship between GDFs and PD-L1. Western blot Western blot was used to detect the expression of PD-L1 in GBM cell lines. Results: GDFs displayed differential patterns of expression with GDF15 and myostatin (MSTN) highly enriched in GBM tissues. We also identified GDF15 as a novel regulator that induces PD-L1 expression in GBM cells. Consistently, GDF15 expression correlated with PD-L1 in TCGA GBM dataset. Further, GDF15 enhanced PD-L1 expression via Smad2/3 pathway in GBM cell line U87, U251 and SHG44, which was inhibited by Smad2/3 inhibitor SIS3. Knockdown of GDF15 attenuated Smad2/3 signaling and reduced PD-L1 expression in A172 and GIC6 glioma cells. Conclusion: GDF15 might be a novel regulator of PD-L1 expression in GBMs; targeting GDF15/PD-L1 pathway might be a promising therapeutic approach for GBM patients.
Collapse
Affiliation(s)
- Haiqin Peng
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, People’s Republic of China
| | - Zhanzhan Li
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, People’s Republic of China
| | - Jun Fu
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, People’s Republic of China
| | - Rongrong Zhou
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, People’s Republic of China
| |
Collapse
|