1
|
Staffeldt L, Maar H, Beimdiek J, Chambers S, Riecken K, von Itzstein M, Buettner FFR, Everest-Dass A, Lange T. Depletion of β1,6-N-acetylglucosaminyltransferase reduces E-selectin binding capacity and migratory potential of human gastrointestinal adenocarcinoma cells. Neoplasia 2025; 59:101083. [PMID: 39547084 PMCID: PMC11609255 DOI: 10.1016/j.neo.2024.101083] [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: 09/02/2024] [Revised: 10/22/2024] [Accepted: 11/05/2024] [Indexed: 11/17/2024]
Abstract
The commonly altered glycosylation of tumor cells is a hallmark of tumor progression and metastasis formation. One prominent example is the interaction of sialylated glycans at the tumor cell surface with endothelial (E)-selectin as an early event of an adhesion cascade that enables extravasation of circulating tumor cells (CTCs) into distant tissues. In a previous study, we identified GCNT3 (mucin-type core2/ core4 β1,6-N-acetylglucosaminyltransferase) highly over-expressed in gastrointestinal adenocarcinoma cells that facilitate the canonical E-selectin ligands sialyl-Lewis A and X (sLeA/X) for E-selectin binding and endothelial adhesion. Here we show that shRNA-mediated, stable depletion of GCNT3 reduced sLeA (tumor marker CA19-9) presentation on two out of three tested human gastrointestinal adenocarcinoma cell lines, concurrently showing reduced static E-selectin binding. Significant effects of GCNT3 depletion on dynamic, shear-resistant tumor cell adhesion on immobilized E-selectin as well as endothelial cells were only partially and inconsistently observable as were effects on tumor cell proliferation (2D) or 3D colony formation. Nevertheless, tumor cell migration was consistently reduced upon GCNT3 depletion in all tested cell lines. Detailed glycome analyses revealed that GCNT3 depletion caused cell line-specific alterations in N- and O-glycans as well as glycosphingolipids, collectively mainly associating with decreased Core-2 structures resulting in varied abundance of sialylation and Lewis antigen with consistent phenotypic changes. Distinctive N- and O-glycosylation features were found to be inherent to specific cell types. These findings suggest GCNT3 products as possible carriers of sLeA and static E-selectin binding sites as well as common pro-migratory glycans in human gastrointestinal cancer.
Collapse
Affiliation(s)
- Lisa Staffeldt
- Institute of Anatomy and Experimental Morphology, University Cancer Center Hamburg, University Medical Center Hamburg-Eppendorf, 20241, Hamburg, Germany
| | - Hanna Maar
- Institute of Anatomy and Experimental Morphology, University Cancer Center Hamburg, University Medical Center Hamburg-Eppendorf, 20241, Hamburg, Germany; Institute of Anatomy I, Jena University Hospital, 07743, Jena, Germany; Comprehensive Cancer Center Central Germany (CCCG)
| | - Julia Beimdiek
- Institute of Clinical Biochemistry, Hannover Medical School, 30625, Hannover, Germany; Proteomics, Institute of Theoretical Medicine, Faculty of Medicine, University of Augsburg, Augsburg, Germany
| | - Samuel Chambers
- Institute for Glycomics, Griffith University, Gold Coast Campus, Gold Coast, QLD4222, Australia
| | - Kristoffer Riecken
- Research Department Cell and Gene Therapy, Department of Stem Cell Transplantation, University Medical Center Hamburg-Eppendorf, 20246, Hamburg
| | - Mark von Itzstein
- Institute for Glycomics, Griffith University, Gold Coast Campus, Gold Coast, QLD4222, Australia
| | - Falk F R Buettner
- Institute of Anatomy and Experimental Morphology, University Cancer Center Hamburg, University Medical Center Hamburg-Eppendorf, 20241, Hamburg, Germany; Institute of Clinical Biochemistry, Hannover Medical School, 30625, Hannover, Germany; Proteomics, Institute of Theoretical Medicine, Faculty of Medicine, University of Augsburg, Augsburg, Germany
| | - Arun Everest-Dass
- Institute of Anatomy and Experimental Morphology, University Cancer Center Hamburg, University Medical Center Hamburg-Eppendorf, 20241, Hamburg, Germany; Institute for Glycomics, Griffith University, Gold Coast Campus, Gold Coast, QLD4222, Australia
| | - Tobias Lange
- Institute of Anatomy and Experimental Morphology, University Cancer Center Hamburg, University Medical Center Hamburg-Eppendorf, 20241, Hamburg, Germany; Institute of Anatomy I, Jena University Hospital, 07743, Jena, Germany; Comprehensive Cancer Center Central Germany (CCCG).
| |
Collapse
|
2
|
Antwi SO, Siaw ADJ, Armasu SM, Frank JA, Yan IK, Ahmed FY, Izquierdo-Sanchez L, Boix L, Rojasti A, Banales JM, Reig M, Stål P, Romero Gómez M, Wangensteen KJ, Singal AG, Roberts LR, Patel T. Genome-wide DNA methylation markers associated with metabolic liver cancer. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.11.15.24317378. [PMID: 39606355 PMCID: PMC11601684 DOI: 10.1101/2024.11.15.24317378] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/29/2024]
Abstract
Background and Aims Metabolic liver disease is the fastest rising cause of hepatocellular carcinoma (HCC) worldwide, but the underlying molecular processes that drive HCC development in the setting of metabolic perturbations are unclear. We investigated the role of aberrant DNA methylation in metabolic HCC development in a multicenter international study. Methods We used a case-control design, frequency-matched on age, sex, and study site. Genome-wide profiling of peripheral blood leukocyte DNA was performed using the 850k EPIC array. Cell type proportions were estimated from the methylation data. The study samples were split 80% and 20% for training and validation. Differential methylation analysis was performed with adjustment for cell type, and we generated area under the receiver-operating curves (ROC-AUC). Results We enrolled 272 metabolic HCC patients and 316 control patients with metabolic liver disease from six sites. Fifty-five differentially methylated CpGs were identified; 33 hypermethylated and 22 hypomethylated in cases versus controls. The panel of 55 CpGs discriminated between cases and controls with AUC=0.79 (95%CI=0.71-0.87), sensitivity=0.77 (95%CI=0.66-0.89), and specificity=0.74 (95%CI=0.64-0.85). The 55-CpG classifier panel performed better than a base model that comprised age, sex, race, and diabetes mellitus (AUC=0.65, 95%CI=0.55-0.75, sensitivity=0.62 (95%CI=0.49-0.75) and specificity=0.64 (95%CI=0.52-0.75). A multifactorial model that combined the 55 CpGs with age, sex, race, and diabetes, yielded AUC=0.78 (95%CI=0.70-0.86), sensitivity=0.81 (95%CI=0.71-0.92), and specificity=0.67 (95%CI=0.55-0.78). Conclusions A panel of 55 blood leukocyte DNA methylation markers differentiates patients with metabolic HCC from control patients with benign metabolic liver disease, with a slightly higher sensitivity when combined with demographic and clinical information.
Collapse
Affiliation(s)
- Samuel O. Antwi
- Division of Epidemiology, Department of Quantitative Health Sciences, Mayo Clinic, Jacksonville, FL, USA
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Mayo Clinic, Jacksonville, FL, USA
| | - Ampem Darko Jnr. Siaw
- Division of Epidemiology, Department of Quantitative Health Sciences, Mayo Clinic, Jacksonville, FL, USA
| | - Sebastian M. Armasu
- Division of Clinical Trials and Biostatistics, Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN, USA
| | - Jacob A. Frank
- Division of Clinical Trials and Biostatistics, Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN, USA
| | - Irene K. Yan
- Department of Cancer Biology, Mayo Clinic, Jacksonville, FL, USA
| | - Fowsiyo Y. Ahmed
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Mayo Clinic, Rochester, MN, USA
| | - Laura Izquierdo-Sanchez
- Department of Liver and Gastrointestinal Diseases, Biogipuzkoa Health Research Institute-Donostia University Hospital, University of the Basque Country (UPV/EHU), CIBERehd, San Sebastian, Spain
| | - Loreto Boix
- BCLC Group, Liver Unit, ICMDM, IDIBAPS, Hospital Clinic of Barcelona, University of Barcelona, Barcelona, Spain. Centro de Investigación Biomédica en Red en Enfermedades Hepáticas y Digestivas (CIBEREHD), Madrid, Spain; Barcelona University, Barcelona, Spain
| | - Angela Rojasti
- SeLiver Group, UCM Digestive Diseases, Institute of Biomedicine of Seville (IBiS), Virgen del Rocio University Hospital/CSIC/University of Seville, Seville, Spain
- Hepatic and Digestive Diseases Networking Biomedical Research Centre (CIBERehd), Madrid, Spain
| | - Jesus M. Banales
- Department of Liver and Gastrointestinal Diseases, Biogipuzkoa Health Research Institute-Donostia University Hospital, University of the Basque Country (UPV/EHU), CIBERehd, San Sebastian, Spain
- Department of Biochemistry and Genetics, School of Sciences, University of Navarra, Pamplona, Spain
- Ikerbasque, Basque Foundation for Science, Bilbao, Spain
| | - Maria Reig
- BCLC Group, Liver Unit, ICMDM, IDIBAPS, Hospital Clinic of Barcelona, University of Barcelona, Barcelona, Spain. Centro de Investigación Biomédica en Red en Enfermedades Hepáticas y Digestivas (CIBEREHD), Madrid, Spain; Barcelona University, Barcelona, Spain
| | - Per Stål
- Department of Upper GI Diseases, Karolinska University Hospital, Department of Medicine Huddinge, Karolinska Institutet, Stockholm, Sweden
| | - Manuel Romero Gómez
- SeLiver Group, UCM Digestive Diseases, Institute of Biomedicine of Seville (IBiS), Virgen del Rocio University Hospital/CSIC/University of Seville, Seville, Spain
- Hepatic and Digestive Diseases Networking Biomedical Research Centre (CIBERehd), Madrid, Spain
| | - Kirk J. Wangensteen
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Mayo Clinic, Rochester, MN, USA
| | - Amit G. Singal
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Lewis R. Roberts
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Mayo Clinic, Rochester, MN, USA
| | - Tushar Patel
- Department of Cancer Biology, Mayo Clinic, Jacksonville, FL, USA
- Department of Transplantation, Mayo Clinic, Jacksonville, FL, USA
| |
Collapse
|
3
|
Cao L, Liu M, Ma X, Rong P, Zhang J, Wang W. Comprehensive scRNA-seq Analysis and Identification of CD8_+T Cell Related Gene Markers for Predicting Prognosis and Drug Resistance of Hepatocellular Carcinoma. Curr Med Chem 2024; 31:2414-2430. [PMID: 37936457 DOI: 10.2174/0109298673274578231030065454] [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: 07/28/2023] [Revised: 09/26/2023] [Accepted: 10/23/2023] [Indexed: 11/09/2023]
Abstract
BACKGROUND Tumor heterogeneity of immune infiltration of cells plays a decisive role in hepatocellular carcinoma (HCC) therapy response and prognosis. This study investigated the effect of different subtypes of CD8+T cells on the HCC tumor microenvironment about its prognosis. METHODS Single-cell RNA sequencing, transcriptome, and single-nucleotide variant data from LUAD patients were obtained based on the GEO, TCGA, and HCCD18 databases. CD8+ T cells-associated subtypes were identified by consensus clustering analysis, and genes with the highest correlation with prognostic CD8+ T cell subtypes were identified using WGCNA. The ssGSEA and ESTIMATE algorithms were used to calculate pathway enrichment scores and immune cell infiltration levels between different subtypes. Finally, the TIDE algorithm, CYT score, and tumor responsiveness score were utilized to predict patient response to immunotherapy. RESULTS We defined 3 CD8+T cell clusters (CD8_0, CD8_1, CD8_2) based on the scRNA- seq dataset (GSE149614). Among, CD8_2 was prognosis-related risk factor with HCC. We screened 30 prognosis genes from CD8_2, and identified 3 molecular subtypes (clust1, clust2, clust3). Clust1 had better survival outcomes, higher gene mutation, and enhanced immune infiltration. Furthermore, we identified a 12 genes signature (including CYP7A1, SPP1, MSC, CXCL8, CXCL1, GCNT3, TMEM45A, SPP2, ME1, TSPAN13, S100A9, and NQO1) with excellent prediction performance for HCC prognosis. In addition, High-score patients with higher immune infiltration benefited less from immunotherapy. The sensitivity of low-score patients to multiple drugs including Parthenolide and Shikonin was significantly higher than that of high-score patients. Moreover, high-score patients had increased oxidative stress pathways scores, and the RiskScore was closely associated with oxidative stress pathways scores. And the nomogram had good clinical utility. CONCLUSION To predict the survival outcome and immunotherapy response for HCC, we developed a 12-gene signature based on the heterogeneity of the CD8+ T cells.
Collapse
Affiliation(s)
- Lu Cao
- The Institute for Cell Transplantation and Gene Therapy, the Third Xiangya Hospital, Central South University, Changsha, 410005, China
- Department of Radiology, the Third Xiangya Hospital, Central South University, Changsha, 410005, China
- Postdoctoral Research Station of Special Medicine, The Third Xiangya Hospital, Changsha, 410005, China
| | - Muqi Liu
- The Institute for Cell Transplantation and Gene Therapy, the Third Xiangya Hospital, Central South University, Changsha, 410005, China
- Department of Radiology, the Third Xiangya Hospital, Central South University, Changsha, 410005, China
| | - Xiaoqian Ma
- The Institute for Cell Transplantation and Gene Therapy, the Third Xiangya Hospital, Central South University, Changsha, 410005, China
- Department of Radiology, the Third Xiangya Hospital, Central South University, Changsha, 410005, China
| | - Pengfei Rong
- The Institute for Cell Transplantation and Gene Therapy, the Third Xiangya Hospital, Central South University, Changsha, 410005, China
- Department of Radiology, the Third Xiangya Hospital, Central South University, Changsha, 410005, China
| | - Juan Zhang
- The Institute for Cell Transplantation and Gene Therapy, the Third Xiangya Hospital, Central South University, Changsha, 410005, China
- Department of Radiology, the Third Xiangya Hospital, Central South University, Changsha, 410005, China
| | - Wei Wang
- The Institute for Cell Transplantation and Gene Therapy, the Third Xiangya Hospital, Central South University, Changsha, 410005, China
- Department of Radiology, the Third Xiangya Hospital, Central South University, Changsha, 410005, China
| |
Collapse
|
4
|
Yang X, Tai Y, Ma Y, Xu Z, Hao J, Han D, Li J, Deng X. Cecum microbiome and metabolism characteristics of Silky Fowl and White Leghorn chicken in late laying stages. Front Microbiol 2022; 13:984654. [PMID: 36338096 PMCID: PMC9633115 DOI: 10.3389/fmicb.2022.984654] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2022] [Accepted: 09/05/2022] [Indexed: 01/08/2023] Open
Abstract
Cecal microflora plays a key role in the production performance and immune function of chickens. White Leghorn (WL) is a well-known commercial layer line chicken with high egg production rate. In contrast, Silky Fowl (SF), a Chinese native chicken variety, has a low egg production rate, but good immune performance. This study analyzed the composition of cecal microbiota, metabolism, and gene expression in intestinal tissue of these varieties and the correlations among them. Significant differences were observed in the cecal microbes: Bacteroides was significantly enriched in WL, whereas Veillonellaceae and Parabacteroides were significantly enriched in SF. Carbohydrate biosynthesis and metabolism pathways were significantly upregulated in WL cecum, which might provide more energy to the host, leading to persistently high levels of egg production. The higher Parabacteroides abundance in SF increased volicitin content, enhanced α-linolenic acid metabolism, and significantly negatively correlated with metabolites of propanoate metabolism and carbohydrate metabolism. Genes related to lipid metabolism, immunity, and melanogenesis were significantly upregulated in the SF cecum, regulating lipid metabolism, and participating in the immune response, while genes related to glucose metabolism and bile acid metabolism were expressed at higher levels in WL, benefiting energy support. This study provided a mechanism for intestinal microorganisms and metabolic pathways to regulate chicken egg-laying performance and immunity.
Collapse
Affiliation(s)
- Xue Yang
- Key Laboratory of Animal Genetics, Breeding and Reproduction of the Ministry of Agriculture, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Yurong Tai
- Key Laboratory of Animal Genetics, Breeding and Reproduction of the Ministry of Agriculture, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Yuhao Ma
- Key Laboratory of Animal Genetics, Breeding and Reproduction of the Ministry of Agriculture, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Zihan Xu
- Key Laboratory of Animal Genetics, Breeding and Reproduction of the Ministry of Agriculture, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Jiaqi Hao
- Key Laboratory of Animal Genetics, Breeding and Reproduction of the Ministry of Agriculture, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Deping Han
- College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Junying Li
- Key Laboratory of Animal Genetics, Breeding and Reproduction of the Ministry of Agriculture, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Xuemei Deng
- Key Laboratory of Animal Genetics, Breeding and Reproduction of the Ministry of Agriculture, College of Animal Science and Technology, China Agricultural University, Beijing, China
| |
Collapse
|
5
|
Yamamoto D, Sasaki K, Kosaka T, Oya M, Sato T. Functional analysis of GCNT3 for cell migration and EMT of castration-resistant prostate cancer cells. Glycobiology 2022; 32:897-908. [PMID: 35867813 DOI: 10.1093/glycob/cwac044] [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: 03/11/2022] [Revised: 06/23/2022] [Accepted: 06/28/2022] [Indexed: 11/13/2022] Open
Abstract
Castration-resistant prostate cancer (CRPC) is a malignant tumor that is resistant to androgen deprivation therapy. Treatments for CRPC are limited, and no diagnostic markers are currently available. O-glycans are known to play an important role in cell proliferation, migration, invasion, and metastasis of cancer cells. However, the differences in the O-glycan expression profiles for normal prostate cancer (PCa) cells compared to CRPC cells have not yet been investigated. In this study, the saccharide primer method was employed to analyze the O-glycans expressed in CRPC cells. Expression levels of core 4-type O-glycans were significantly increased in CRPC cells. Furthermore, the expression level of N-Acetylglucosaminyltransferase 3 (GCNT3), a core 4-type O-glycan synthase gene, was increased in CRPC cells. The expression of core 4-type O-glycans and GCNT3 was presumed to be regulated by androgen deprivation. GCNT3 knockdown induced cell migration and epithelial-mesenchymal transition (EMT). These observations elucidate the mechanism of acquisition of castration resistance in PCa and offer new possibilities for the development of diagnostic markers and therapeutic targets in the treatment of PCa.
Collapse
Affiliation(s)
- Daiki Yamamoto
- Department of Biosciences and Informatics, Faculty of Science and Technology, Keio University, Kanagawa, Japan
| | - Katsumasa Sasaki
- Department of Biosciences and Informatics, Faculty of Science and Technology, Keio University, Kanagawa, Japan
| | - Takeo Kosaka
- Department of Urology, Keio University School of Medicine, Tokyo, Japan
| | - Mototsugu Oya
- Department of Urology, Keio University School of Medicine, Tokyo, Japan
| | - Toshinori Sato
- Department of Biosciences and Informatics, Faculty of Science and Technology, Keio University, Kanagawa, Japan
| |
Collapse
|
6
|
Yuan B, Zhao X, Wang X, Liu E, Liu C, Zong Y, Jiang Y, Hou M, Chen Y, Chen L, Zhang Y, Wang H, Fu J. Patient-derived organoids for personalized gallbladder cancer modelling and drug screening. Clin Transl Med 2022; 12:e678. [PMID: 35075805 PMCID: PMC8786696 DOI: 10.1002/ctm2.678] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2021] [Revised: 11/15/2021] [Accepted: 11/30/2021] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Gallbladder carcinoma (GBC) is a relatively rare but highly aggressive cancer with late clinical detection and a poor prognosis. However, the lack of models with features consistent with human gallbladder tumours has hindered progress in pathogenic mechanisms and therapies. METHODS We established organoid lines derived from human GBC as well as normal gallbladder and benign gallbladder adenoma (GBA) tissues. The histopathology signatures of organoid cultures were identified by H&E staining, immunohistochemistry and immunofluorescence. The genetic and transcriptional features of organoids were analysed by whole-exome sequencing and RNA sequencing. A set of compounds targeting the most active signalling pathways in GBCs were screened for their ability to suppress GBC organoids. The antitumour effects of candidate compounds, CUDC-101 and CUDC-907, were evaluated in vitro and in vivo. RESULTS The established organoids were cultured stably for more than 6 months and closely recapitulated the histopathology, genetic and transcriptional features, and intratumour heterogeneity of the primary tissues at the single-cell level. Notably, expression profiling analysis of the organoids revealed a set of genes that varied across the three subtypes and thus may participate in the malignant progression of gallbladder diseases. More importantly, we found that the dual PI3K/HDAC inhibitor CUDC-907 significantly restrained the growth of various GBC organoids with minimal toxicity to normal gallbladder organoids. CONCLUSIONS Patient-derived organoids are potentially a useful platform to explore molecular pathogenesis of gallbladder tumours and discover personalized drugs.
Collapse
Affiliation(s)
- Bo Yuan
- International Cooperation Laboratory on Signal TransductionMinistry of Education Key Laboratory on Signaling Regulation and Targeting Therapy of Liver CancerShanghai Key Laboratory of Hepato‐biliary Tumor BiologyEastern Hepatobiliary Surgery HospitalSecond Military Medical UniversityShanghaiChina
| | - Xiaofang Zhao
- Research Center for OrganoidsThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenanChina
| | - Xiang Wang
- Second Department of Biliary SurgeryEastern Hepatobiliary Surgery HospitalSecond Military Medical UniversityShanghaiChina
| | - Erdong Liu
- School of Life SciencesFudan UniversityShanghaiChina
| | - Chunliang Liu
- International Cooperation Laboratory on Signal TransductionMinistry of Education Key Laboratory on Signaling Regulation and Targeting Therapy of Liver CancerShanghai Key Laboratory of Hepato‐biliary Tumor BiologyEastern Hepatobiliary Surgery HospitalSecond Military Medical UniversityShanghaiChina
| | - Yali Zong
- School of Life SciencesFudan UniversityShanghaiChina
| | - Youhai Jiang
- Division of Life Sciences and MedicineCancer Research CenterThe First Affiliated Hospital of USTCUniversity of Science and Technology of ChinaHefeiAnhuiChina
| | - Minghui Hou
- Research Center for OrganoidsThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenanChina
| | - Yao Chen
- International Cooperation Laboratory on Signal TransductionMinistry of Education Key Laboratory on Signaling Regulation and Targeting Therapy of Liver CancerShanghai Key Laboratory of Hepato‐biliary Tumor BiologyEastern Hepatobiliary Surgery HospitalSecond Military Medical UniversityShanghaiChina
| | - Lei Chen
- International Cooperation Laboratory on Signal TransductionMinistry of Education Key Laboratory on Signaling Regulation and Targeting Therapy of Liver CancerShanghai Key Laboratory of Hepato‐biliary Tumor BiologyEastern Hepatobiliary Surgery HospitalSecond Military Medical UniversityShanghaiChina
| | - Yongjie Zhang
- Second Department of Biliary SurgeryEastern Hepatobiliary Surgery HospitalSecond Military Medical UniversityShanghaiChina
| | - Hongyang Wang
- International Cooperation Laboratory on Signal TransductionMinistry of Education Key Laboratory on Signaling Regulation and Targeting Therapy of Liver CancerShanghai Key Laboratory of Hepato‐biliary Tumor BiologyEastern Hepatobiliary Surgery HospitalSecond Military Medical UniversityShanghaiChina
| | - Jing Fu
- International Cooperation Laboratory on Signal TransductionMinistry of Education Key Laboratory on Signaling Regulation and Targeting Therapy of Liver CancerShanghai Key Laboratory of Hepato‐biliary Tumor BiologyEastern Hepatobiliary Surgery HospitalSecond Military Medical UniversityShanghaiChina
| |
Collapse
|
7
|
Chatterjee S, Ugonotti J, Lee LY, Everest-Dass A, Kawahara R, Thaysen-Andersen M. Trends in oligomannosylation and α1,2-mannosidase expression in human cancers. Oncotarget 2021; 12:2188-2205. [PMID: 34676051 PMCID: PMC8522845 DOI: 10.18632/oncotarget.28064] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Accepted: 08/18/2021] [Indexed: 02/05/2023] Open
Abstract
Aberrant protein glycosylation is a prominent cancer feature. While many tumour-associated glycoepitopes have been reported, advances in glycoanalytics continue to uncover new associations between glycosylation and cancer. Guided by a comprehensive literature survey suggesting that oligomannosylation (Man5–9 GlcNAc2) is a widespread and often regulated glycosignature in human cancers, we here revisit a valuable compilation of nearly 500 porous graphitized carbon LC-MS/MS N-glycomics datasets acquired across 11 human cancer types to systematically test for oligomannose-cancer associations. Firstly, the quantitative glycomics data obtained across 34 cancerous cell lines demonstrated that oligomannosylation is a pan-cancer feature spanning in a wide abundance range. In keeping with literature, our quantitative glycomics data of tumour and matching control tissues and new MALDI-MS imaging data of tissue microarrays showed a strong cancer-associated elevation of oligomannosylation in both basal cell (p = 1.78 × 10–12) and squamous cell (p = 1.23 × 10–11) skin cancer and colorectal cancer (p = 8.0 × 10–4). The glycomics data also indicated that some cancer types including gastric and liver cancer exhibit unchanged or reduced oligomannose levels, observations also supported by literature and MALDI-MS imaging data. Finally, expression data from public cancer repositories indicated that several α1,2-mannosidases are regulated in tumour tissues suggesting that these glycan-processing enzymes may contribute to the cancer-associated modulation of oligomannosylation. This omics-centric study has compiled robust glycomics and enzyme expression data revealing interesting molecular trends that open avenues to better understand the role of oligomannosylation in human cancers.
Collapse
Affiliation(s)
| | - Julian Ugonotti
- Department of Molecular Sciences, Macquarie University, Sydney, Australia
| | - Ling Y Lee
- Department of Molecular Sciences, Macquarie University, Sydney, Australia
| | | | - Rebeca Kawahara
- Department of Molecular Sciences, Macquarie University, Sydney, Australia.,Joint senior authors
| | - Morten Thaysen-Andersen
- Department of Molecular Sciences, Macquarie University, Sydney, Australia.,Biomolecular Discovery Research Centre (BDRC), Macquarie University, Sydney, Australia.,Joint senior authors
| |
Collapse
|
8
|
Zhang J, Ten Dijke P, Wuhrer M, Zhang T. Role of glycosylation in TGF-β signaling and epithelial-to-mesenchymal transition in cancer. Protein Cell 2021; 12:89-106. [PMID: 32583064 PMCID: PMC7862465 DOI: 10.1007/s13238-020-00741-7] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2020] [Accepted: 05/29/2020] [Indexed: 12/14/2022] Open
Abstract
Glycosylation is a common posttranslational modification on membrane-associated and secreted proteins that is of pivotal importance for regulating cell functions. Aberrant glycosylation can lead to uncontrolled cell proliferation, cell-matrix interactions, migration and differentiation, and has been shown to be involved in cancer and other diseases. The epithelial-to-mesenchymal transition is a key step in the metastatic process by which cancer cells gain the ability to invade tissues and extravasate into the bloodstream. This cellular transformation process, which is associated by morphological change, loss of epithelial traits and gain of mesenchymal markers, is triggered by the secreted cytokine transforming growth factor-β (TGF-β). TGF-β bioactivity is carefully regulated, and its effects on cells are mediated by its receptors on the cell surface. In this review, we first provide a brief overview of major types of glycans, namely, N-glycans, O-glycans, glycosphingolipids and glycosaminoglycans that are involved in cancer progression. Thereafter, we summarize studies on how the glycosylation of TGF-β signaling components regulates TGF-β secretion, bioavailability and TGF-β receptor function. Then, we review glycosylation changes associated with TGF-β-induced epithelial-to-mesenchymal transition in cancer. Identifying and understanding the mechanisms by which glycosylation affects TGF-β signaling and downstream biological responses will facilitate the identification of glycans as biomarkers and enable novel therapeutic approaches.
Collapse
Affiliation(s)
- Jing Zhang
- Oncode Institute and Cell Chemical Biology, Leiden University Medical Center, 2300 RC, Leiden, The Netherlands
| | - Peter Ten Dijke
- Oncode Institute and Cell Chemical Biology, Leiden University Medical Center, 2300 RC, Leiden, The Netherlands.
| | - Manfred Wuhrer
- Center for Proteomics and Metabolomics, Leiden University Medical Center, Leiden, The Netherlands
| | - Tao Zhang
- Center for Proteomics and Metabolomics, Leiden University Medical Center, Leiden, The Netherlands
| |
Collapse
|
9
|
Sun H, Chang J, Ye M, Weng W, Zhang M, Ni S, Tan C, Huang D, Wang L, Du X, Xu MD, Sheng W. GCNT4 is Associated with Prognosis and Suppress Cell Proliferation in Gastric Cancer. Onco Targets Ther 2020; 13:8601-8613. [PMID: 32922038 PMCID: PMC7457769 DOI: 10.2147/ott.s248997] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2020] [Accepted: 08/03/2020] [Indexed: 11/23/2022] Open
Abstract
Background GCNT4 is a member of the glucosaminyl (N-acetyl) transferases family that has been implicated in multiple human malignancies. However, the role of GCNT4 in gastric cancer (GC) is unknown. In this present study, we aimed to explore the role and clinicopathological correlation of GCNT4 in GC. Materials and Methods We first evaluated the dysregulation of GCNT4 in The Cancer Genome Atlas (TCGA) and then we performed RT-qPCR and immunohistochemistry to validate the results in a cohort of in-house patients. The clinicopathological correlation and function of GCNT4 in GC were also analysed. Results GCNT4 was found to be significantly downregulated in GC. In addition, GCNT4 expression correlated with tumour depth, nervous invasion and pathological tumor-node-metastasis (pTNM) stage. Moreover, lower GCNT4 levels conferred poor overall survival (OS) and disease-free survival (DFS) to GC patients. Multivariate Cox regression analysis revealed that GCNT4 protein expression is an independent prognostic factor for OS in patients with GC. Further functional experimental results revealed that overexpression of GCNT4 appears to halt GC cell proliferation and the cell cycle. Conclusion Altogether, these findings indicated that GCNT4 regulates the GC cell cycle and have important implications for the selection of therapeutic targets to prevent tumour proliferation.
Collapse
Affiliation(s)
- Hui Sun
- Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai 200032, People's Republic of China.,Department of Pathology, Eye, Ear, Nose and Throat Hospital, Fudan University, Shanghai 200031, People's Republic of China
| | - Jinjia Chang
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, People's Republic of China.,Department of Medical Oncology, Fudan University Shanghai Cancer Center, Fudan University, Shanghai 200032, People's Republic of China
| | - Min Ye
- Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai 200032, People's Republic of China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, People's Republic of China.,Institute of Pathology, Fudan University, Shanghai 200032, People's Republic of China
| | - Weiwei Weng
- Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai 200032, People's Republic of China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, People's Republic of China.,Institute of Pathology, Fudan University, Shanghai 200032, People's Republic of China
| | - Meng Zhang
- Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai 200032, People's Republic of China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, People's Republic of China.,Institute of Pathology, Fudan University, Shanghai 200032, People's Republic of China
| | - Shujuan Ni
- Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai 200032, People's Republic of China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, People's Republic of China.,Institute of Pathology, Fudan University, Shanghai 200032, People's Republic of China
| | - Cong Tan
- Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai 200032, People's Republic of China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, People's Republic of China.,Institute of Pathology, Fudan University, Shanghai 200032, People's Republic of China
| | - Dan Huang
- Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai 200032, People's Republic of China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, People's Republic of China.,Institute of Pathology, Fudan University, Shanghai 200032, People's Republic of China
| | - Lei Wang
- Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai 200032, People's Republic of China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, People's Republic of China.,Institute of Pathology, Fudan University, Shanghai 200032, People's Republic of China
| | - Xiang Du
- Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai 200032, People's Republic of China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, People's Republic of China.,Institute of Pathology, Fudan University, Shanghai 200032, People's Republic of China
| | - Mi-Die Xu
- Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai 200032, People's Republic of China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, People's Republic of China.,Institute of Pathology, Fudan University, Shanghai 200032, People's Republic of China
| | - Weiqi Sheng
- Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai 200032, People's Republic of China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, People's Republic of China.,Institute of Pathology, Fudan University, Shanghai 200032, People's Republic of China
| |
Collapse
|
10
|
Rasheduzzaman M, Kulasinghe A, Dolcetti R, Kenny L, Johnson NW, Kolarich D, Punyadeera C. Protein glycosylation in head and neck cancers: From diagnosis to treatment. Biochim Biophys Acta Rev Cancer 2020; 1874:188422. [PMID: 32853734 DOI: 10.1016/j.bbcan.2020.188422] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Revised: 08/20/2020] [Accepted: 08/21/2020] [Indexed: 12/24/2022]
Abstract
Glycosylation is the most common post-translational modification (PTM) of proteins. Malignant tumour cells frequently undergo an alteration in surface protein glycosylation. This phenomenon is also common in cancers of the head and neck, most of which are squamous cell carcinomas (HNSCC). It affects cell functions, including proliferation, motility and invasiveness, thus increasing the propensity to metastasise. HNSCC represents the sixth most frequent malignancy worldwide. These neoplasms, which arise from the mucous membranes of the various anatomical subsites of the upper aero-digestive tract, are heterogeneous in terms of aetiology and clinico-pathologic features. With current treatments, only about 50% of HNSCC patients survive beyond 5-years. Therefore, there is the pressing need to dissect NHSCC heterogeneity to inform treatment choices. In particular, reliable biomarkers of predictive and prognostic value are eagerly needed. This review describes the current state of the art and bio-pathological meaning of glycosylation signatures associated with HNSCC and explores the possible role of tumour specific glycoproteins as potential biomarkers and attractive therapeutic targets. We have also compiled data relating to altered glycosylation and the nature of glycoproteins as tools for the identification of circulating tumour cells (CTCs) in the new era of liquid biopsy.
Collapse
Affiliation(s)
- Mohammad Rasheduzzaman
- Saliva and Liquid Biopsy Translational Laboratory, The School of Biomedical Sciences, Institute of Health and Biomedical Innovation, Queensland University of Technology, Kelvin Grove, QLD, Australia; Translational Research Institute, Woolloongabba, QLD, Australia
| | - Arutha Kulasinghe
- Saliva and Liquid Biopsy Translational Laboratory, The School of Biomedical Sciences, Institute of Health and Biomedical Innovation, Queensland University of Technology, Kelvin Grove, QLD, Australia; Translational Research Institute, Woolloongabba, QLD, Australia
| | - Riccardo Dolcetti
- Translational Research Institute, Woolloongabba, QLD, Australia.; The University of Queensland Diamantina Institute, 37 Kent Street Woolloongabba, QLD 4102, Australia
| | - Liz Kenny
- Department of Radiation Oncology, Cancer Care Services, Royal Brisbane and Women's Hospital, Joyce Tweddell Building, Herston, QLD, 4029, Australia
| | - Newell W Johnson
- Menzies Health Institute Queensland, Griffith University, Southport, Queensland, Australia; Faculty of Dentistry, Oral and Craniofacial Sciences, King's College, London, United Kingdom
| | - Daniel Kolarich
- Institute for Glycomics, Griffith University, Gold Coast, QLD, Australia; ARC Centre of Excellence for Nanoscale BioPhotonics, Griffith University, QLD, Australia.
| | - Chamindie Punyadeera
- Saliva and Liquid Biopsy Translational Laboratory, The School of Biomedical Sciences, Institute of Health and Biomedical Innovation, Queensland University of Technology, Kelvin Grove, QLD, Australia; Translational Research Institute, Woolloongabba, QLD, Australia..
| |
Collapse
|
11
|
Hamester F, Legler K, Wichert B, Kelle N, Eylmann K, Rossberg M, Ding Y, Kürti S, Schmalfeldt B, Milde-Langosch K, Oliveira-Ferrer L. Prognostic relevance of the Golgi mannosidase MAN1A1 in ovarian cancer: impact of N-glycosylation on tumour cell aggregation. Br J Cancer 2019; 121:944-953. [PMID: 31659304 PMCID: PMC6889143 DOI: 10.1038/s41416-019-0607-2] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2019] [Revised: 09/20/2019] [Accepted: 10/01/2019] [Indexed: 11/10/2022] Open
Abstract
Background Maturation of complex N-glycans involves the action of Golgi mannosidases and plays a major role in cancer progression. We recently showed a favourable prognostic role of α-mannosidase MAN1A1 in breast cancer mainly caused by alteration of certain adhesion molecules. Methods We analysed the protein expression of MAN1A1 in ovarian cancer (n = 204) using western blot and studied the impact of MAN1A1 itself and of MAN1A1-related glycosylation on the prognostic relevance of two adhesion molecules. Functional consequences of mannosidase inhibition using kifunensine and MAN1A1 knock out were investigated in ovarian cancer cells in vitro. Results Patients with high MAN1A1 expression in tumours showed significantly shorter RFS than those with low-MAN1A1 levels. Moreover, high MAN1A1 expression correlated significantly with advanced stage, lymph node involvement and distant metastasis. Further, the glycosylated adhesion molecule ALCAM reveals a significant adverse prognostic effect only in the presence of high MAN1A1 expression. In spheroid-formation assays, mannosidase inhibition and especially MAN1A1 knock out led to strong reduction of tumour cell aggregation. Conclusions Our study demonstrates the unfavourable prognostic role of MAN1A1 in ovarian cancer, probably caused by an altered ability of spheroid formation, and the strong influence of this glycosylation enzyme on the prognostic impact of ALCAM.
Collapse
Affiliation(s)
- Fabienne Hamester
- Department of Gynecology, University Medical Centre Hamburg-Eppendorf, Martinistr. 52, Hamburg, 20246, Germany
| | - Karen Legler
- Department of Gynecology, University Medical Centre Hamburg-Eppendorf, Martinistr. 52, Hamburg, 20246, Germany
| | - Beatrice Wichert
- Department of Gynecology, University Medical Centre Hamburg-Eppendorf, Martinistr. 52, Hamburg, 20246, Germany
| | - Nicole Kelle
- Department of Gynecology, University Medical Centre Hamburg-Eppendorf, Martinistr. 52, Hamburg, 20246, Germany
| | - Kathrin Eylmann
- Department of Gynecology, University Medical Centre Hamburg-Eppendorf, Martinistr. 52, Hamburg, 20246, Germany
| | - Maila Rossberg
- Department of Gynecology, University Medical Centre Hamburg-Eppendorf, Martinistr. 52, Hamburg, 20246, Germany
| | - Yi Ding
- Department of Gynecology, University Medical Centre Hamburg-Eppendorf, Martinistr. 52, Hamburg, 20246, Germany
| | - Sascha Kürti
- Department of Gynecology, University Medical Centre Hamburg-Eppendorf, Martinistr. 52, Hamburg, 20246, Germany
| | - Barbara Schmalfeldt
- Department of Gynecology, University Medical Centre Hamburg-Eppendorf, Martinistr. 52, Hamburg, 20246, Germany
| | - Karin Milde-Langosch
- Department of Gynecology, University Medical Centre Hamburg-Eppendorf, Martinistr. 52, Hamburg, 20246, Germany
| | - Leticia Oliveira-Ferrer
- Department of Gynecology, University Medical Centre Hamburg-Eppendorf, Martinistr. 52, Hamburg, 20246, Germany.
| |
Collapse
|
12
|
Myricetin inhibits migration and invasion of hepatocellular carcinoma MHCC97H cell line by inhibiting the EMT process. Oncol Lett 2019; 18:6614-6620. [PMID: 31788118 PMCID: PMC6865832 DOI: 10.3892/ol.2019.10998] [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: 12/23/2017] [Accepted: 08/17/2019] [Indexed: 12/15/2022] Open
Abstract
The recurrence and metastasis of hepatocellular carcinoma (HCC) are a major concern in current research. Epithelial-mesenchymal transition (EMT) is the leading cause underlying the high mobility and invasiveness of tumor cells. Myricetin is a natural flavonol with various pharmacological activities. The effects of myricetin on the migration and invasion of HCC MHCC97H cells were evaluated in the present study. Wound healing, Transwell migration and invasion assays were used to examine cell migration and invasion. Western blot analysis and reverse transcription-quantitative polymerase chain reaction (RT-qPCR) were used to examine the expression of epithelial (E)-cadherin, neural (N)-cadherin and vimentin. The present study aimed to investigate the effects of myricetin on the migration and invasion of HCC MHCC97H cells. It was indicated that myricetin decreased the viability of MHCC97H cells in a concentration and time-dependent manner, and inhibited MHCC97H cells migration and invasion. As the concentration of myricetin increased, filopodia and lamellipodia in cells weakened and cells were arranged more closely. RT-qPCR and western blotting revealed that myricetin upregulated E-cadherin expression and downregulated N-cadherin. Collectively, the results of the present study demonstrate that myricetin may inhibit the migration and invasion of HCC MHCC97H cells by inhibiting the EMT process.
Collapse
|
13
|
Tax G, Lia A, Santino A, Roversi P. Modulation of ERQC and ERAD: A Broad-Spectrum Spanner in the Works of Cancer Cells? JOURNAL OF ONCOLOGY 2019; 2019:8384913. [PMID: 31662755 PMCID: PMC6791201 DOI: 10.1155/2019/8384913] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/20/2019] [Accepted: 08/27/2019] [Indexed: 12/21/2022]
Abstract
Endoplasmic reticulum glycoprotein folding quality control (ERQC) and ER-associated degradation (ERAD) preside over cellular glycoprotein secretion and maintain steady glycoproteostasis. When cells turn malignant, cancer cell plasticity is affected and supported either by point mutations, preferential isoform selection, altered expression levels, or shifts to conformational equilibria of a secreted glycoprotein. Such changes are crucial in mediating altered extracellular signalling, metabolic behavior, and adhesion properties of cancer cells. It is therefore conceivable that interference with ERQC and/or ERAD can be used to selectively damage cancers. Indeed, inhibitors of the late stages of ERAD are already in the clinic against cancers such as multiple myeloma. Here, we review recent advances in our understanding of the complex relationship between glycoproteostasis and cancer biology and discuss the potential of ERQC and ERAD modulators for the selective targeting of cancer cell plasticity.
Collapse
Affiliation(s)
- Gábor Tax
- Leicester Institute of Structural and Chemical Biology, Department of Molecular and Cell Biology, University of Leicester, Henry Wellcome Building, Lancaster Road, Leicester LE1 7RH, UK
| | - Andrea Lia
- Leicester Institute of Structural and Chemical Biology, Department of Molecular and Cell Biology, University of Leicester, Henry Wellcome Building, Lancaster Road, Leicester LE1 7RH, UK
- Institute of Sciences of Food Production, C.N.R. Unit of Lecce, via Monteroni, I-73100 Lecce, Italy
| | - Angelo Santino
- Institute of Sciences of Food Production, C.N.R. Unit of Lecce, via Monteroni, I-73100 Lecce, Italy
| | - Pietro Roversi
- Leicester Institute of Structural and Chemical Biology, Department of Molecular and Cell Biology, University of Leicester, Henry Wellcome Building, Lancaster Road, Leicester LE1 7RH, UK
| |
Collapse
|
14
|
Chevalier F, Cuyas L, Jouhet J, Gros VR, Chiarenza S, Secco D, Whelan J, Seddiki K, Block MA, Nussaume L, Marechal E. Interplay between Jasmonic Acid, Phosphate Signaling and the Regulation of Glycerolipid Homeostasis in Arabidopsis. PLANT & CELL PHYSIOLOGY 2019; 60:1260-1273. [PMID: 30753691 DOI: 10.1093/pcp/pcz027] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/02/2018] [Accepted: 01/29/2019] [Indexed: 05/25/2023]
Abstract
Jasmonic acid (JA) biosynthesis and signaling are activated in Arabidopsis cultivated in phosphate (Pi) deprived conditions. This activation occurs mainly in photosynthetic tissues and is less important in roots. In leaves, the enhanced biosynthesis of JA coincides with membrane glycerolipid remodeling triggered by the lack of Pi. We addressed the possible role of JA on the dynamics and magnitude of glycerolipid remodeling in response to Pi deprivation and resupply. Based on combined analyses of gene expression, JA biosynthesis and glycerolipid remodeling in wild-type Arabidopsis and in the coi1-16 mutant, JA signaling seems important in the determination of the basal levels of phosphatidylcholine, phosphatidic acid (PA), monogalactosyldiacylglycerol (MGDG) and digalactosyldiacylglycerol. JA impact on MGDG steady state level and fluctuations seem contradictory. In the coi1-16 mutant, the steady state level of MGDG is higher, possibly due to a higher level of PA in the mutant, activating MGD1, and to an increased expression of MGD3. These results support a possible impact of JA in limiting the overall content of this lipid. Concerning lipid variations, upon Pi deprivation, JA seems rather associated with a specific MGDG increase. Following Pi resupply, whereas the expression of glycerolipid remodeling genes returns to basal level, JA biosynthesis and signaling genes are still upregulated, likely due to a JA-induced positive feedback remaining active. Distinct impacts on enzymes synthesizing MGDG, that is, downregulating MGD3, possibly activating MGD1 expression and limiting the activation of MGD1 via PA, might allow JA playing a role in a sophisticated fine tuning of galactolipid variations.
Collapse
Affiliation(s)
- Florian Chevalier
- Laboratoire de Physiologie Cellulaire et V�g�tale, Unit� mixe de recherche 5168 CNRS, CEA, INRA, Universit� Grenoble Alpes, IRIG, CEA Grenoble, 17, rue des Martyrs, Grenoble, France
| | - Laura Cuyas
- Laboratoire de Biologie V�g�tale et Microbiologie Environnementale, Unit� mixte de recherche 7265 CNRS, CEA, Universit� Aix-Marseille, Institut de Biosciences et Biotechnologies d'Aix-Marseille, CEA Cadarache, Saint-Paul-lez-Durance, France
- Centre Mondial de l'Innovation, Groupe Roullier, 18 avenue Franklin Roosevelt, Saint-Malo, France
| | - Juliette Jouhet
- Laboratoire de Physiologie Cellulaire et V�g�tale, Unit� mixe de recherche 5168 CNRS, CEA, INRA, Universit� Grenoble Alpes, IRIG, CEA Grenoble, 17, rue des Martyrs, Grenoble, France
| | - Valï Rie Gros
- Laboratoire de Physiologie Cellulaire et V�g�tale, Unit� mixe de recherche 5168 CNRS, CEA, INRA, Universit� Grenoble Alpes, IRIG, CEA Grenoble, 17, rue des Martyrs, Grenoble, France
| | - Serge Chiarenza
- Laboratoire de Biologie V�g�tale et Microbiologie Environnementale, Unit� mixte de recherche 7265 CNRS, CEA, Universit� Aix-Marseille, Institut de Biosciences et Biotechnologies d'Aix-Marseille, CEA Cadarache, Saint-Paul-lez-Durance, France
| | - David Secco
- Department of Animal, Plant and Soil Sciences, Australian Research Council Centre of Excellence in Plant Energy Biology, School of Life Sciences, La Trobe University, Bundoora, Victoria, Australia
| | - James Whelan
- Department of Animal, Plant and Soil Sciences, Australian Research Council Centre of Excellence in Plant Energy Biology, School of Life Sciences, La Trobe University, Bundoora, Victoria, Australia
| | - Khawla Seddiki
- Laboratoire de Physiologie Cellulaire et V�g�tale, Unit� mixe de recherche 5168 CNRS, CEA, INRA, Universit� Grenoble Alpes, IRIG, CEA Grenoble, 17, rue des Martyrs, Grenoble, France
| | - Maryse A Block
- Laboratoire de Physiologie Cellulaire et V�g�tale, Unit� mixe de recherche 5168 CNRS, CEA, INRA, Universit� Grenoble Alpes, IRIG, CEA Grenoble, 17, rue des Martyrs, Grenoble, France
| | - Laurent Nussaume
- Laboratoire de Biologie V�g�tale et Microbiologie Environnementale, Unit� mixte de recherche 7265 CNRS, CEA, Universit� Aix-Marseille, Institut de Biosciences et Biotechnologies d'Aix-Marseille, CEA Cadarache, Saint-Paul-lez-Durance, France
| | - Eric Marechal
- Laboratoire de Physiologie Cellulaire et V�g�tale, Unit� mixe de recherche 5168 CNRS, CEA, INRA, Universit� Grenoble Alpes, IRIG, CEA Grenoble, 17, rue des Martyrs, Grenoble, France
| |
Collapse
|
15
|
MicroRNA in Lung Cancer Metastasis. Cancers (Basel) 2019; 11:cancers11020265. [PMID: 30813457 PMCID: PMC6406837 DOI: 10.3390/cancers11020265] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2019] [Revised: 02/17/2019] [Accepted: 02/18/2019] [Indexed: 12/12/2022] Open
Abstract
Tumor metastasis is a hallmark of cancer, with distant metastasis frequently developing in lung cancer, even at initial diagnosis, resulting in poor prognosis and high mortality. However, available biomarkers cannot reliably predict cancer spreading sites. The metastatic cascade involves highly complicated processes including invasion, migration, angiogenesis, and epithelial-to-mesenchymal transition that are tightly controlled by various genetic expression modalities along with interaction between cancer cells and the extracellular matrix. In particular, microRNAs (miRNAs), a group of small non-coding RNAs, can influence the transcriptional and post-transcriptional processes, with dysregulation of miRNA expression contributing to the regulation of cancer metastasis. Nevertheless, although miRNA-targeted therapy is widely studied in vitro and in vivo, this strategy currently affords limited feasibility and a few miRNA-targeted therapies for lung cancer have entered into clinical trials to date. Advances in understanding the molecular mechanism of metastasis will thus provide additional potential targets for lung cancer treatment. This review discusses the current research related to the role of miRNAs in lung cancer invasion and metastasis, with a particular focus on the different metastatic lesions and potential miRNA-targeted treatments for lung cancer with the expectation that further exploration of miRNA-targeted therapy may establish a new spectrum of lung cancer treatments.
Collapse
|
16
|
Hou J, Wang L, Wu D. The root of Actinidia chinensis inhibits hepatocellular carcinomas cells through LAMB3. Cell Biol Toxicol 2018; 34:321-332. [PMID: 29127567 DOI: 10.1007/s10565-017-9416-7] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2017] [Accepted: 10/05/2017] [Indexed: 12/14/2022]
Abstract
The root of Actinidia chinensis, as traditional Chinese medicine, has been shown to inhibit cell proliferation in numerous cancer cells. However, the mechanisms underlying its inhibitory activity remain unclear. Death rates of hepatocellular carcinoma (HCC) are increasing, but therapies for advanced HCC are not well developed. We choose the extract from root of Actinidia chinensis (ERAC) to treat the HCC cell lines in vitro, displaying distinct effects on cell proliferation, S-phase cell cycle arrest, and apoptosis. LAMB3, the gene encoding laminin subunit beta-3, plays a key role in the proliferation suppression and S-phase cell cycle arrest of HepG2 cells treated with ERAC. The downstream genes ITGA3, CCND2, and TP53 in LAMB3 pathway show the same response to ERAC as LAMB3. Thus, LAMB3 pathways, along with extracellular matrix-receptor interaction, pathways in cancer, and focal adhesion, are involved in the ERAC-induced suppressive response in HepG2.
Collapse
Affiliation(s)
- Jiayun Hou
- Zhongshan Hospital Institute of Clinical Science, Shanghai Institute of Clinical Bioinformatics; Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Lingyan Wang
- Zhongshan Hospital Institute of Clinical Science, Shanghai Institute of Clinical Bioinformatics; Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Duojiao Wu
- Zhongshan Hospital Institute of Clinical Science, Shanghai Institute of Clinical Bioinformatics; Zhongshan Hospital, Fudan University, Shanghai, 200032, China.
| |
Collapse
|
17
|
Balcik-Ercin P, Cetin M, Yalim-Camci I, Odabas G, Tokay N, Sayan AE, Yagci T. Genome-wide analysis of endogenously expressed ZEB2 binding sites reveals inverse correlations between ZEB2 and GalNAc-transferase GALNT3 in human tumors. Cell Oncol (Dordr) 2018; 41:379-393. [PMID: 29516288 DOI: 10.1007/s13402-018-0375-7] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/22/2018] [Indexed: 10/17/2022] Open
Abstract
BACKGROUND ZEB2 is a transcriptional repressor that regulates epithelial-to-mesenchymal transition (EMT) through binding to bipartite E-box motifs in gene regulatory regions. Despite the abundant presence of E-boxes within the human genome and the multiplicity of pathophysiological processes regulated during ZEB2-induced EMT, only a small fraction of ZEB2 targets has been identified so far. Hence, we explored genome-wide ZEB2 binding by chromatin immunoprecipitation-sequencing (ChIP-seq) under endogenous ZEB2 expression conditions. METHODS For ChIP-Seq we used an anti-ZEB2 monoclonal antibody, clone 6E5, in SNU398 hepatocellular carcinoma cells exhibiting a high endogenous ZEB2 expression. The ChIP-Seq targets were validated using ChIP-qPCR, whereas ZEB2-dependent expression of target genes was assessed by RT-qPCR and Western blotting in shRNA-mediated ZEB2 silenced SNU398 cells and doxycycline-induced ZEB2 overexpressing colorectal carcinoma DLD1 cells. Changes in target gene expression were also assessed using primary human tumor cDNA arrays in conjunction with RT-qPCR. Additional differential expression and correlation analyses were performed using expO and Human Protein Atlas datasets. RESULTS Over 500 ChIP-Seq positive genes were annotated, and intervals related to these genes were found to include the ZEB2 binding motif CACCTG according to TOMTOM motif analysis in the MEME Suite database. Assessment of ZEB2-dependent expression of target genes in ZEB2-silenced SNU398 cells and ZEB2-induced DLD1 cells revealed that the GALNT3 gene serves as a ZEB2 target with the highest, but inversely correlated, expression level. Remarkably, GALNT3 also exhibited the highest enrichment in the ChIP-qPCR validation assays. Through the analyses of primary tumor cDNA arrays and expO datasets a significant differential expression and a significant inverse correlation between ZEB2 and GALNT3 expression were detected in most of the tumors. We also explored ZEB2 and GALNT3 protein expression using the Human Protein Atlas dataset and, again, observed an inverse correlation in all analyzed tumor types, except malignant melanoma. In contrast to a generally negative or weak ZEB2 expression, we found that most tumor tissues exhibited a strong or moderate GALNT3 expression. CONCLUSIONS Our observation that ZEB2 negatively regulates a GalNAc-transferase (GALNT3) that is involved in O-glycosylation adds another layer of complexity to the role of ZEB2 in cancer progression and metastasis. Proteins glycosylated by GALNT3 may be exploited as novel diagnostics and/or therapeutic targets.
Collapse
Affiliation(s)
- Pelin Balcik-Ercin
- Department of Molecular Biology and Genetics, Laboratory of Molecular Oncology, Gebze Technical University, C2-Building, 41400, Gebze-Kocaeli, Turkey
| | - Metin Cetin
- Department of Molecular Biology and Genetics, Laboratory of Molecular Oncology, Gebze Technical University, C2-Building, 41400, Gebze-Kocaeli, Turkey
| | - Irem Yalim-Camci
- Department of Molecular Biology and Genetics, Laboratory of Molecular Oncology, Gebze Technical University, C2-Building, 41400, Gebze-Kocaeli, Turkey
| | - Gorkem Odabas
- Department of Molecular Biology and Genetics, Laboratory of Molecular Oncology, Gebze Technical University, C2-Building, 41400, Gebze-Kocaeli, Turkey
| | - Nurettin Tokay
- Department of Molecular Biology and Genetics, Laboratory of Molecular Oncology, Gebze Technical University, C2-Building, 41400, Gebze-Kocaeli, Turkey
| | - A Emre Sayan
- Faculty of Medicine, Cancer Sciences, University of Southampton, Somers Building, Tremona Road, Southampton, SO16 6YD, UK
| | - Tamer Yagci
- Department of Molecular Biology and Genetics, Laboratory of Molecular Oncology, Gebze Technical University, C2-Building, 41400, Gebze-Kocaeli, Turkey.
| |
Collapse
|
18
|
Phoomak C, Silsirivanit A, Park D, Sawanyawisuth K, Vaeteewoottacharn K, Wongkham C, Lam EWF, Pairojkul C, Lebrilla CB, Wongkham S. O-GlcNAcylation mediates metastasis of cholangiocarcinoma through FOXO3 and MAN1A1. Oncogene 2018; 37:5648-5665. [PMID: 29915392 PMCID: PMC6151127 DOI: 10.1038/s41388-018-0366-1] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2017] [Revised: 05/23/2018] [Accepted: 05/25/2018] [Indexed: 02/06/2023]
Abstract
The leading cause of death in cancer patients is metastasis, for which an effective treatment is still necessary. During metastasis, cancer cells aberrantly express several glycans that are correlated with poor patient outcome. This study was aimed toward exploring the effects of O-GlcNAcylation on membranous N-glycans that are associated with the progression of cholangiocarcinoma (CCA). Global O-GlcNAcylation in CCA cells was depleted using specific siRNA against O-GlcNAc transferase (OGT), which transfers GlcNAc to the acceptor proteins. Using an HPLC-Chip/Time-of-Flight (Chip/TOF) MS system, the N-glycans associated with O-GlcNAcylation were identified by comparing the membranous N-glycans of siOGT-treated cells with those of scramble siRNA-treated cells. In parallel, the membranous N-glycans of the parental cells (KKU-213 and KKU-214) were compared with those of the highly metastatic cells (KKU-213L5 and KKU-214L5). Together, these data revealed that high mannose (Hex9HexNAc2) and biantennary complex (Hex5HexNAc4Fuc1NeuAc1) N-linked glycans correlated positively with metastasis. We subsequently demonstrate that suppression of O-GlcNAcylation decreased the expression of these two N-glycans, suggesting that O-GlcNAcylation mediates their levels in CCA. In addition, the ability of highly metastatic cells to migrate and invade was reduced by the presence of Pisum Sativum Agglutinin (PSA), a mannose-specific lectin, further indicating the association of high mannose type N-glycans with CCA metastasis. The molecular mechanism of O-GlcNAc-mediated progression of CCA was shown to proceed via a series of signaling events, involving the activation of Akt/Erk (i), an increase in FOXO3 phosphorylation (ii), which results in the reduction of MAN1A1 expression (iii) and thus the accumulation of Hex9HexNAc2 N-glycans (iv). This study demonstrates for the first time the association between O-GlcNAcylation, high mannose type N-glycans, and the progression of CCA metastasis, suggesting a novel therapeutic target for treatment of metastatic CCA.
Collapse
Affiliation(s)
- Chatchai Phoomak
- Department of Biochemistry, Faculty of Medicine, Khon Kaen University, Khon Kaen, 40002, Thailand.,Cholangiocarcinoma Research Institute, Khon Kaen University, Khon Kaen, 40002, Thailand
| | - Atit Silsirivanit
- Department of Biochemistry, Faculty of Medicine, Khon Kaen University, Khon Kaen, 40002, Thailand.,Cholangiocarcinoma Research Institute, Khon Kaen University, Khon Kaen, 40002, Thailand
| | - Dayoung Park
- Department of Chemistry, University of California, Davis, CA, 95616, USA.,Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, 02115, USA
| | - Kanlayanee Sawanyawisuth
- Department of Biochemistry, Faculty of Medicine, Khon Kaen University, Khon Kaen, 40002, Thailand.,Cholangiocarcinoma Research Institute, Khon Kaen University, Khon Kaen, 40002, Thailand
| | - Kulthida Vaeteewoottacharn
- Department of Biochemistry, Faculty of Medicine, Khon Kaen University, Khon Kaen, 40002, Thailand.,Cholangiocarcinoma Research Institute, Khon Kaen University, Khon Kaen, 40002, Thailand
| | - Chaisiri Wongkham
- Department of Biochemistry, Faculty of Medicine, Khon Kaen University, Khon Kaen, 40002, Thailand.,Cholangiocarcinoma Research Institute, Khon Kaen University, Khon Kaen, 40002, Thailand
| | - Eric W-F Lam
- Department of Surgery and Cancer, Imperial Centre for Translational and Experimental Medicine, Imperial College London, London, W12 0NN, UK
| | - Chawalit Pairojkul
- Cholangiocarcinoma Research Institute, Khon Kaen University, Khon Kaen, 40002, Thailand.,Department of Pathology, Faculty of Medicine, Khon Kaen University, Khon Kaen, 40002, Thailand
| | - Carlito B Lebrilla
- Department of Chemistry, University of California, Davis, CA, 95616, USA.
| | - Sopit Wongkham
- Department of Biochemistry, Faculty of Medicine, Khon Kaen University, Khon Kaen, 40002, Thailand. .,Cholangiocarcinoma Research Institute, Khon Kaen University, Khon Kaen, 40002, Thailand.
| |
Collapse
|
19
|
Conte M, Lupette J, Seddiki K, Meï C, Dolch LJ, Gros V, Barette C, Rébeillé F, Jouhet J, Maréchal E. Screening for Biologically Annotated Drugs That Trigger Triacylglycerol Accumulation in the Diatom Phaeodactylum. PLANT PHYSIOLOGY 2018; 177:532-552. [PMID: 29535162 PMCID: PMC6001342 DOI: 10.1104/pp.17.01804] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/22/2017] [Accepted: 03/06/2018] [Indexed: 05/03/2023]
Abstract
Microalgae are a promising feedstock for the production of triacylglycerol (TAG) for a variety of potential applications, ranging from food and human health to biofuels and green chemistry. However, obtaining high TAG yields is challenging. A phenotypic assay for the accumulation of oil droplets was developed to screen a library of 1,200 drugs, annotated with pharmacology information, to select compounds that trigger TAG accumulation in the diatom Phaeodactylum tricornutum Using this screen, we identified 34 molecules acting in a dose-dependent manner. Previously characterized targets of these compounds include cell division and cell signaling effectors, membrane receptors and transporters, and sterol metabolism. Among the five compounds possibly acting on sterol metabolism, we focused our study on ethynylestradiol, a synthetic form of estrogen that is used in contraceptive pills and known for its ecological impact as an endocrine disruptor. Ethynylestradiol impaired the production of very-long-chain polyunsaturated fatty acids, destabilized the galactolipid versus phospholipid balance, and triggered the recycling of fatty acids from membrane lipids to TAG. The P. tricornutum transcriptomic response to treatment with ethynylestradiol was consistent with the reallocation of carbon from sterols to acetyl-coenzyme A and TAG. The mode of action and catabolism of ethynylestradiol are unknown but might involve several up-regulated cytochrome P450 proteins. A fatty acid elongase, Δ6-ELO-B1, might be involved in the impairment of very-long-chain polyunsaturated fatty acids and fatty acid turnover. This phenotypic screen opens new perspectives for the exploration of novel bioactive molecules, potential target genes, and pathways controlling TAG biosynthesis. It also unraveled the sensitivity of diatoms to endocrine disruptors, highlighting an impact of anthropogenic pollution on phytoplankton.
Collapse
Affiliation(s)
- Melissa Conte
- Laboratoire de Physiologie Cellulaire et Végétale, Centre National de la Recherche Scientifique, Commissariat à l'Energie Atomique, Institut National de la Recherche Agronomique, Université Grenoble Alpes, Institut de Biosciences et Biotechnologies de Grenoble, Commissariat à l'Energie Atomique-Grenoble, 38000 Grenoble, France
| | - Josselin Lupette
- Laboratoire de Physiologie Cellulaire et Végétale, Centre National de la Recherche Scientifique, Commissariat à l'Energie Atomique, Institut National de la Recherche Agronomique, Université Grenoble Alpes, Institut de Biosciences et Biotechnologies de Grenoble, Commissariat à l'Energie Atomique-Grenoble, 38000 Grenoble, France
| | - Khawla Seddiki
- Laboratoire de Physiologie Cellulaire et Végétale, Centre National de la Recherche Scientifique, Commissariat à l'Energie Atomique, Institut National de la Recherche Agronomique, Université Grenoble Alpes, Institut de Biosciences et Biotechnologies de Grenoble, Commissariat à l'Energie Atomique-Grenoble, 38000 Grenoble, France
| | - Coline Meï
- Laboratoire de Physiologie Cellulaire et Végétale, Centre National de la Recherche Scientifique, Commissariat à l'Energie Atomique, Institut National de la Recherche Agronomique, Université Grenoble Alpes, Institut de Biosciences et Biotechnologies de Grenoble, Commissariat à l'Energie Atomique-Grenoble, 38000 Grenoble, France
| | - Lina-Juana Dolch
- Laboratoire de Physiologie Cellulaire et Végétale, Centre National de la Recherche Scientifique, Commissariat à l'Energie Atomique, Institut National de la Recherche Agronomique, Université Grenoble Alpes, Institut de Biosciences et Biotechnologies de Grenoble, Commissariat à l'Energie Atomique-Grenoble, 38000 Grenoble, France
| | - Valérie Gros
- Laboratoire de Physiologie Cellulaire et Végétale, Centre National de la Recherche Scientifique, Commissariat à l'Energie Atomique, Institut National de la Recherche Agronomique, Université Grenoble Alpes, Institut de Biosciences et Biotechnologies de Grenoble, Commissariat à l'Energie Atomique-Grenoble, 38000 Grenoble, France
| | - Caroline Barette
- Laboratoire de Biologie à Grande Echelle, Commissariat à l'Energie Atomique, INSERM, Université Grenoble Alpes, Institut de Biosciences et Biotechnologies de Grenoble, Commissariat à l'Energie Atomique-Grenoble, 38000 Grenoble, France
| | - Fabrice Rébeillé
- Laboratoire de Physiologie Cellulaire et Végétale, Centre National de la Recherche Scientifique, Commissariat à l'Energie Atomique, Institut National de la Recherche Agronomique, Université Grenoble Alpes, Institut de Biosciences et Biotechnologies de Grenoble, Commissariat à l'Energie Atomique-Grenoble, 38000 Grenoble, France
| | - Juliette Jouhet
- Laboratoire de Physiologie Cellulaire et Végétale, Centre National de la Recherche Scientifique, Commissariat à l'Energie Atomique, Institut National de la Recherche Agronomique, Université Grenoble Alpes, Institut de Biosciences et Biotechnologies de Grenoble, Commissariat à l'Energie Atomique-Grenoble, 38000 Grenoble, France
| | - Eric Maréchal
- Laboratoire de Physiologie Cellulaire et Végétale, Centre National de la Recherche Scientifique, Commissariat à l'Energie Atomique, Institut National de la Recherche Agronomique, Université Grenoble Alpes, Institut de Biosciences et Biotechnologies de Grenoble, Commissariat à l'Energie Atomique-Grenoble, 38000 Grenoble, France
| |
Collapse
|
20
|
The role of glycosyltransferase enzyme GCNT3 in colon and ovarian cancer prognosis and chemoresistance. Sci Rep 2018; 8:8485. [PMID: 29855486 PMCID: PMC5981315 DOI: 10.1038/s41598-018-26468-4] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2018] [Accepted: 04/19/2018] [Indexed: 12/17/2022] Open
Abstract
Glycosyltransferase enzyme GCNT3, has been proposed as a biomarker for prognosis in colorectal cancer (CRC). Our study goes in depth into the molecular basis of GCNT3 role in tumorigenesis and drug resistance, and it explores its potential role as biomarker in epithelial ovarian cancer (EOC). High levels of GCNT3 are associated with increased sensibility to 5-fluoracil in metastatic cells. Accordingly, GCNT3 re-expression leads to the gain of anti-carcinogenic cellular properties by reducing cell growth, invasion and by changing metabolic capacities. Integrated transcriptomic and proteomic analyses reveal that GCNT3 is linked to cellular cycle, mitosis and proliferation, response to drugs and metabolism pathways. The vascular epithelial growth factor A (VEGFA) arises as an attractive partner of GCNT3 functions in cell invasion and resistance. Finally, GCNT3 expression was analyzed in a cohort of 56 EOC patients followed by a meta-analysis of more than one thousand patients. This study reveals that GCNT3 might constitute a prognostic factor also in EOC, since its overexpression is associated with better clinical outcome and response to initial therapy. GCNT3 emerges as an essential glycosylation-related molecule in CRC and EOC progression, with potential interest as a predictive biomarker of response to chemotherapy.
Collapse
|
21
|
Reduced mannosidase MAN1A1 expression leads to aberrant N-glycosylation and impaired survival in breast cancer. Br J Cancer 2018; 118:847-856. [PMID: 29381688 PMCID: PMC5877434 DOI: 10.1038/bjc.2017.472] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2017] [Revised: 11/28/2017] [Accepted: 11/29/2017] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Alterations in protein glycosylation have been related to malignant transformation and tumour progression. We recently showed that low mRNA levels of Golgi alpha-mannosidase MAN1A1 correlate with poor prognosis in breast cancer patients. METHODS We analysed the role of MAN1A1 on a protein level using western blot analysis (n=105) and studied the impact of MAN1A1-related glycosylation on the prognostic relevance of adhesion molecules involved in breast cancer using microarray data (n=194). Functional consequences of mannosidase inhibition using the inhibitor kifunensine or MAN1A1 silencing were investigated in breast cancer cells in vitro. RESULTS Patients with low/moderate MAN1A1 expression in tumours showed significantly shorter disease-free intervals than those with high MAN1A1 levels (P=0.005). Moreover, low MAN1A1 expression correlated significantly with nodal status, grading and brain metastasis. At an mRNA level, membrane proteins ALCAM and CD24 were only significantly prognostic in tumours with high MAN1A1 expression. In vitro, reduced MAN1A1 expression or mannosidase inhibition led to a significantly increased adhesion of breast cancer cells to endothelial cells. CONCLUSIONS Our study demonstrates the prognostic role of MAN1A1 in breast cancer by affecting the adhesive properties of tumour cells and the strong influence of this glycosylation enzyme on the prognostic impact of some adhesion proteins.
Collapse
|
22
|
Dolch LJ, Lupette J, Tourcier G, Bedhomme M, Collin S, Magneschi L, Conte M, Seddiki K, Richard C, Corre E, Fourage L, Laeuffer F, Richards R, Reith M, Rébeillé F, Jouhet J, McGinn P, Maréchal E. Nitric Oxide Mediates Nitrite-Sensing and Acclimation and Triggers a Remodeling of Lipids. PLANT PHYSIOLOGY 2017; 175:1407-1423. [PMID: 28924015 PMCID: PMC5664477 DOI: 10.1104/pp.17.01042] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/31/2017] [Accepted: 09/13/2017] [Indexed: 05/20/2023]
Abstract
Nitric oxide (NO) is an intermediate of the nitrogen cycle, an industrial pollutant, and a marker of climate change. NO also acts as a gaseous transmitter in a variety of biological processes. The impact of environmental NO needs to be addressed. In diatoms, a dominant phylum in phytoplankton, NO was reported to mediate programmed cell death in response to diatom-derived polyunsaturated aldehydes. Here, using the Phaeodactylum Pt1 strain, 2E,4E-decadienal supplied in the micromolar concentration range led to a nonspecific cell toxicity. We reexamined NO biosynthesis and response in Phaeodactylum NO inhibits cell growth and triggers triacylglycerol (TAG) accumulation. Feeding experiments indicate that NO is not produced from Arg but via conversion of nitrite by the nitrate reductase. Genome-wide transcriptional analysis shows that NO up-regulates the expression of the plastid nitrite reductase and genes involved in the subsequent incorporation of ammonium into amino acids, via both Gln synthesis and Orn-urea pathway. The phosphoenolpyruvate dehydrogenase complex is also up-regulated, leading to the production of acetyl-CoA, which can feed TAG accumulation upon exposure to NO. Transcriptional reprogramming leading to higher TAG content is balanced with a decrease of monogalactosyldiacylglycerol (MGDG) in the plastid via posttranslational inhibition of MGDG synthase enzymatic activity by NO. Intracellular and transient NO emission acts therefore at the basis of a nitrite-sensing and acclimating system, whereas a long exposure to NO can additionally induce a redirection of carbon to neutral lipids and a stress response.
Collapse
Affiliation(s)
- Lina-Juana Dolch
- Laboratoire de Physiologie Cellulaire et Végétale, Unité mixte de recherche 5168 CNRS - CEA - INRA - Université Grenoble Alpes, Institut de Biosciences Biotechnologies de Grenoble, CEA Grenoble, 17 rue des Martyrs, 38054, Grenoble Cedex 9, France
| | - Josselin Lupette
- Laboratoire de Physiologie Cellulaire et Végétale, Unité mixte de recherche 5168 CNRS - CEA - INRA - Université Grenoble Alpes, Institut de Biosciences Biotechnologies de Grenoble, CEA Grenoble, 17 rue des Martyrs, 38054, Grenoble Cedex 9, France
| | - Guillaume Tourcier
- Laboratoire de Physiologie Cellulaire et Végétale, Unité mixte de recherche 5168 CNRS - CEA - INRA - Université Grenoble Alpes, Institut de Biosciences Biotechnologies de Grenoble, CEA Grenoble, 17 rue des Martyrs, 38054, Grenoble Cedex 9, France
| | - Mariette Bedhomme
- Laboratoire de Physiologie Cellulaire et Végétale, Unité mixte de recherche 5168 CNRS - CEA - INRA - Université Grenoble Alpes, Institut de Biosciences Biotechnologies de Grenoble, CEA Grenoble, 17 rue des Martyrs, 38054, Grenoble Cedex 9, France
- Total Refining Chemicals, Tour Michelet, 24 Cours Michelet - La Défense 10, 92069 Paris La Défense Cedex, France
| | - Séverine Collin
- Total Refining Chemicals, Tour Michelet, 24 Cours Michelet - La Défense 10, 92069 Paris La Défense Cedex, France
| | - Leonardo Magneschi
- Laboratoire de Physiologie Cellulaire et Végétale, Unité mixte de recherche 5168 CNRS - CEA - INRA - Université Grenoble Alpes, Institut de Biosciences Biotechnologies de Grenoble, CEA Grenoble, 17 rue des Martyrs, 38054, Grenoble Cedex 9, France
| | - Melissa Conte
- Laboratoire de Physiologie Cellulaire et Végétale, Unité mixte de recherche 5168 CNRS - CEA - INRA - Université Grenoble Alpes, Institut de Biosciences Biotechnologies de Grenoble, CEA Grenoble, 17 rue des Martyrs, 38054, Grenoble Cedex 9, France
| | - Khawla Seddiki
- Laboratoire de Physiologie Cellulaire et Végétale, Unité mixte de recherche 5168 CNRS - CEA - INRA - Université Grenoble Alpes, Institut de Biosciences Biotechnologies de Grenoble, CEA Grenoble, 17 rue des Martyrs, 38054, Grenoble Cedex 9, France
| | - Christelle Richard
- Laboratoire de Physiologie Cellulaire et Végétale, Unité mixte de recherche 5168 CNRS - CEA - INRA - Université Grenoble Alpes, Institut de Biosciences Biotechnologies de Grenoble, CEA Grenoble, 17 rue des Martyrs, 38054, Grenoble Cedex 9, France
| | - Erwan Corre
- Station Biologique de Roscoff, CNRS - Université Pierre et Marie Curie, Analyses and Bioinformatics for Marine Science, 29680 Roscoff, France
| | - Laurent Fourage
- Total Refining Chemicals, Tour Michelet, 24 Cours Michelet - La Défense 10, 92069 Paris La Défense Cedex, France
| | - Frédéric Laeuffer
- Total Refining Chemicals, Tour Michelet, 24 Cours Michelet - La Défense 10, 92069 Paris La Défense Cedex, France
| | - Robert Richards
- National Research Council of Canada, Aquatic and Crop Resource Development, 1411 Oxford Street, Halifax, Nova Scotia B3H3Z1, Canada
| | - Michael Reith
- National Research Council of Canada, Aquatic and Crop Resource Development, 1411 Oxford Street, Halifax, Nova Scotia B3H3Z1, Canada
| | - Fabrice Rébeillé
- Laboratoire de Physiologie Cellulaire et Végétale, Unité mixte de recherche 5168 CNRS - CEA - INRA - Université Grenoble Alpes, Institut de Biosciences Biotechnologies de Grenoble, CEA Grenoble, 17 rue des Martyrs, 38054, Grenoble Cedex 9, France
| | - Juliette Jouhet
- Laboratoire de Physiologie Cellulaire et Végétale, Unité mixte de recherche 5168 CNRS - CEA - INRA - Université Grenoble Alpes, Institut de Biosciences Biotechnologies de Grenoble, CEA Grenoble, 17 rue des Martyrs, 38054, Grenoble Cedex 9, France
| | - Patrick McGinn
- National Research Council of Canada, Aquatic and Crop Resource Development, 1411 Oxford Street, Halifax, Nova Scotia B3H3Z1, Canada
| | - Eric Maréchal
- Laboratoire de Physiologie Cellulaire et Végétale, Unité mixte de recherche 5168 CNRS - CEA - INRA - Université Grenoble Alpes, Institut de Biosciences Biotechnologies de Grenoble, CEA Grenoble, 17 rue des Martyrs, 38054, Grenoble Cedex 9, France
| |
Collapse
|
23
|
Aguirre-Portolés C, Fernández LP, Ramírez de Molina A. Precision Nutrition for Targeting Lipid Metabolism in Colorectal Cancer. Nutrients 2017; 9:nu9101076. [PMID: 28956850 PMCID: PMC5691693 DOI: 10.3390/nu9101076] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2017] [Revised: 09/20/2017] [Accepted: 09/25/2017] [Indexed: 12/16/2022] Open
Abstract
Cancer is a multistage and multifactorial condition with genetic and environmental factors modulating tumorogenesis and disease progression. Nevertheless, cancer is preventable, as one third of cancer deaths could be avoided by modifying key risk factors. Nutrients can directly affect fundamental cellular processes and are considered among the most important risk factors in colorectal cancer (CRC). Red and processed meat, poultry consumption, fiber, and folate are the best-known diet components that interact with colorectal cancer susceptibility. In addition, the direct association of an unhealthy diet with obesity and dysbiosis opens new routes in the understanding of how daily diet nutrients could influence cancer prognosis. In the “omics” era, traditional nutrition has been naturally evolved to precision nutrition where technical developments have contributed to a more accurate discipline. In this sense, genomic and transcriptomic studies have been extensively used in precision nutrition approaches. However, the relation between CRC carcinogenesis and nutrition factors is more complex than originally expected. Together with classical diet-nutrition-related genes, nowadays, lipid-metabolism-related genes have acquired relevant interest in precision nutrition studies. Lipids regulate very diverse cellular processes from ATP synthesis and the activation of essential cell-signaling pathways to membrane organization and plasticity. Therefore, a wide range of tumorogenic steps can be influenced by lipid metabolism, both in primary tumours and distal metastasis. The extent to which genetic variants, together with the intake of specific dietary components, affect the risk of CRC is currently under investigation, and new therapeutic or preventive applications must be explored in CRC models. In this review, we will go in depth into the study of co-occurring events, which orchestrate CRC tumorogenesis and are essential for the evolution of precision nutrition paradigms. Likewise, we will discuss the application of precision nutrition approaches to target lipid metabolism in CRC.
Collapse
Affiliation(s)
- Cristina Aguirre-Portolés
- Molecular Oncology and Nutritional Genomics of Cancer Group, IMDEA Food Institute, CEI UAM + CSIC, Carretera de Cantoblanco 8, E-28049 Madrid, Spain.
| | - Lara P Fernández
- Molecular Oncology and Nutritional Genomics of Cancer Group, IMDEA Food Institute, CEI UAM + CSIC, Carretera de Cantoblanco 8, E-28049 Madrid, Spain.
| | - Ana Ramírez de Molina
- Molecular Oncology and Nutritional Genomics of Cancer Group, IMDEA Food Institute, CEI UAM + CSIC, Carretera de Cantoblanco 8, E-28049 Madrid, Spain.
| |
Collapse
|
24
|
Zhang J, Lu S, Zhou Y, Meng K, Chen Z, Cui Y, Shi Y, Wang T, He QY. Motile hepatocellular carcinoma cells preferentially secret sugar metabolism regulatory proteins via exosomes. Proteomics 2017; 17. [PMID: 28590090 DOI: 10.1002/pmic.201700103] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2017] [Revised: 05/14/2017] [Accepted: 05/29/2017] [Indexed: 01/29/2023]
Abstract
Exosomes are deliverers of critically functional proteins, capable of transforming target cells in numerous cancers, including hepatocellular carcinoma (HCC). We hypothesize that the motility of HCC cells can be featured by comparative proteome of exosomes. Hence, we performed the super-SILAC-based MS analysis on the exosomes secreted by three human HCC cell lines, including the non-motile Hep3B cell, and the motile 97H and LM3 cells. More than 1400 exosomal proteins were confidently quantified in each MS analysis with highly biological reproducibility. We justified that 469 and 443 exosomal proteins represented differentially expressed proteins (DEPs) in the 97H/Hep3B and LM3/Hep3B comparisons, respectively. These DEPs focused on sugar metabolism-centric canonical pathways per ingenuity pathway analysis, which was consistent with the gene ontology analysis on biological process enrichment. These pathways included glycolysis I, gluconeogenesis I and pentose phosphate pathways; and the DEPs enriched in these pathways could form a tightly connected network. By analyzing the relative abundance of proteins and translating mRNAs, we found significantly positive correlation between exosomes and cells. The involved exosomal proteins were again focusing on sugar metabolism. In conclusion, motile HCC cells tend to preferentially export more sugar metabolism-associated proteins via exosomes that differentiate them from non-motile HCC cells.
Collapse
Affiliation(s)
- Jing Zhang
- Key Laboratory of Functional Protein Research of Guangdong Higher Education Institutes, Institute of Life and Health Engineering, College of Life Science and Technology, Jinan University, Guangzhou, P. R. China
| | - Shaohua Lu
- Key Laboratory of Functional Protein Research of Guangdong Higher Education Institutes, Institute of Life and Health Engineering, College of Life Science and Technology, Jinan University, Guangzhou, P. R. China
| | - Ye Zhou
- Key Laboratory of Functional Protein Research of Guangdong Higher Education Institutes, Institute of Life and Health Engineering, College of Life Science and Technology, Jinan University, Guangzhou, P. R. China
| | - Kun Meng
- Key Laboratory of Functional Protein Research of Guangdong Higher Education Institutes, Institute of Life and Health Engineering, College of Life Science and Technology, Jinan University, Guangzhou, P. R. China
| | - Zhipeng Chen
- Key Laboratory of Functional Protein Research of Guangdong Higher Education Institutes, Institute of Life and Health Engineering, College of Life Science and Technology, Jinan University, Guangzhou, P. R. China
| | - Yizhi Cui
- Key Laboratory of Functional Protein Research of Guangdong Higher Education Institutes, Institute of Life and Health Engineering, College of Life Science and Technology, Jinan University, Guangzhou, P. R. China
| | - Yunfeng Shi
- Key Laboratory of Functional Protein Research of Guangdong Higher Education Institutes, Institute of Life and Health Engineering, College of Life Science and Technology, Jinan University, Guangzhou, P. R. China
| | - Tong Wang
- Key Laboratory of Functional Protein Research of Guangdong Higher Education Institutes, Institute of Life and Health Engineering, College of Life Science and Technology, Jinan University, Guangzhou, P. R. China
| | - Qing-Yu He
- Key Laboratory of Functional Protein Research of Guangdong Higher Education Institutes, Institute of Life and Health Engineering, College of Life Science and Technology, Jinan University, Guangzhou, P. R. China
| |
Collapse
|
25
|
Tu HC, Hsiao YC, Yang WY, Tsai SL, Lin HK, Liao CY, Lu JW, Chou YT, Wang HD, Yuh CH. Up-regulation of golgi α-mannosidase IA and down-regulation of golgi α-mannosidase IC activates unfolded protein response during hepatocarcinogenesis. Hepatol Commun 2017; 1:230-247. [PMID: 29404456 PMCID: PMC5721452 DOI: 10.1002/hep4.1032] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/14/2016] [Revised: 02/27/2017] [Accepted: 03/09/2017] [Indexed: 12/27/2022] Open
Abstract
α‐1,2 mannosidases, key enzymes in N‐glycosylation, are required for the formation of mature glycoproteins in eukaryotes. Aberrant regulation of α‐1,2 mannosidases can result in cancer, although the underlying mechanisms are unclear. Here, we report the distinct roles of α‐1,2 mannosidase subtypes (MAN1A, MAN1B, ERMAN1, MAN1C) in the formation of hepatocellular carcinoma (HCC). Clinicopathological analyses revealed that the clinical stage, tumor size, α‐fetoprotein level, and invasion status were positively correlated with the expression levels of MAN1A1, MAN1B1, and MAN1A2. In contrast, the expression of MAN1C1 was decreased as early as stage I of HCC. Survival analyses showed that high MAN1A1, MAN1A2, and MAN1B1 expression levels combined with low MAN1C1 expression levels were significantly correlated with shorter overall survival rates. Functionally, the overexpression of MAN1A1 promoted proliferation, migration, and transformation as well as in vivo migration in zebrafish. Conversely, overexpression of MAN1C1 reduced the migration ability both in vitro and in vivo, decreased the colony formation ability, and shortened the S phase of the cell cycle. Furthermore, the expression of genes involved in cell cycle/proliferation and migration was increased in MAN1A1‐overexpressing cells but decreased in MAN1C1‐overexpressing cells. MAN1A1 activated the expression of key regulators of the unfolded protein response (UPR), while treatment with endoplasmic reticulum stress inhibitors blocked the expression of MAN1A1‐activated genes. Using the MAN1A1 liver‐specific overexpression zebrafish model, we observed steatosis and inflammation at earlier stages and HCC formation at a later stage accompanied by the increased expression of the UPR modulator binding immunoglobulin protein (BiP). These data suggest that the up‐regulation of MAN1A1 activates the UPR and might initiate metastasis. Conclusion: MAN1A1 represents a novel oncogene while MAN1C1 plays a role in tumor suppression in hepatocarcinogenesis. (Hepatology Communications 2017;1:230‐247)
Collapse
Affiliation(s)
- Hsiao-Chen Tu
- Institute of Molecular and Genomic Medicine National Health Research Institutes Zhunan Miaoli Taiwan.,Institute of Biotechnology National Tsing-Hua University Hsinchu Taiwan
| | - Yung-Chun Hsiao
- Institute of Molecular and Genomic Medicine National Health Research Institutes Zhunan Miaoli Taiwan.,Institute of Biotechnology National Tsing-Hua University Hsinchu Taiwan
| | - Wan-Yu Yang
- Institute of Molecular and Genomic Medicine National Health Research Institutes Zhunan Miaoli Taiwan
| | - Shin-Lin Tsai
- Institute of Molecular and Genomic Medicine National Health Research Institutes Zhunan Miaoli Taiwan
| | - Hua-Kuo Lin
- Institute of Molecular and Genomic Medicine National Health Research Institutes Zhunan Miaoli Taiwan
| | - Chong-Yi Liao
- Institute of Molecular and Genomic Medicine National Health Research Institutes Zhunan Miaoli Taiwan.,Institute of Biotechnology National Tsing-Hua University Hsinchu Taiwan
| | - Jeng-Wei Lu
- Institute of Molecular and Genomic Medicine National Health Research Institutes Zhunan Miaoli Taiwan.,Department of Life Sciences National Central University Jhongli City Taoyuan Taiwan
| | - Yu-Ting Chou
- Institute of Molecular and Genomic Medicine National Health Research Institutes Zhunan Miaoli Taiwan.,Institute of Biotechnology National Tsing-Hua University Hsinchu Taiwan
| | - Horng-Dar Wang
- Institute of Biotechnology National Tsing-Hua University Hsinchu Taiwan
| | - Chiou-Hwa Yuh
- Institute of Molecular and Genomic Medicine National Health Research Institutes Zhunan Miaoli Taiwan.,Institute of Bioinformatics and Structural Biology National Tsing-Hua University Hsinchu Taiwan.,Department of Biological Science and Technology National Chiao Tung University Hsinchu Taiwan.,Kaohsiung Medical University Kaohsiung Taiwan
| |
Collapse
|
26
|
Systems approach to characterize the metabolism of liver cancer stem cells expressing CD133. Sci Rep 2017; 7:45557. [PMID: 28367990 PMCID: PMC5377334 DOI: 10.1038/srep45557] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2016] [Accepted: 02/28/2017] [Indexed: 01/29/2023] Open
Abstract
Liver cancer stem cells (LCSCs) have attracted attention because they cause therapeutic resistance in hepatocellular carcinoma (HCC). Understanding the metabolism of LCSCs can be a key to developing therapeutic strategy, but metabolic characteristics have not yet been studied. Here, we systematically analyzed and compared the global metabolic phenotype between LCSCs and non-LCSCs using transcriptome and metabolome data. We also reconstructed genome-scale metabolic models (GEMs) for LCSC and non-LCSC to comparatively examine differences in their metabolism at genome-scale. We demonstrated that LCSCs exhibited an increased proliferation rate through enhancing glycolysis compared with non-LCSCs. We also confirmed that MYC, a central point of regulation in cancer metabolism, was significantly up-regulated in LCSCs compared with non-LCSCs. Moreover, LCSCs tend to have less active fatty acid oxidation. In this study, the metabolic characteristics of LCSCs were identified using integrative systems analysis, and these characteristics could be potential cures for the resistance of liver cancer cells to anticancer treatments.
Collapse
|
27
|
Oliveira-Ferrer L, Legler K, Milde-Langosch K. Role of protein glycosylation in cancer metastasis. Semin Cancer Biol 2017; 44:141-152. [PMID: 28315783 DOI: 10.1016/j.semcancer.2017.03.002] [Citation(s) in RCA: 186] [Impact Index Per Article: 23.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2017] [Revised: 03/08/2017] [Accepted: 03/13/2017] [Indexed: 02/06/2023]
Abstract
Although altered glycosylation has been detected in human cancer cells decades ago, only investigations in the last years have enormously increased our knowledge about the details of protein glycosylation and its role in tumour progression. Many proteins, which are heavily glycosylated, i.e. adhesion proteins or proteases, play an important role in cancer metastasis that represents the crucial and frequently life-threatening step in progression of most tumour types. Compared to normal tissue, tumour cells often show altered glycosylation patters with appearance of new tumour-specific antigens. In this review, we give an overview about the role of glycosylation in tumour metastasis, describing recent results about O-glycans, N-glycans and glycosaminoglycans. We show that glycan structures, glycosylated proteins and glycosylation enzymes have influence on different steps of the metastatic process, including epithelial-mesenchymal transition (EMT), migration, invasion/intravasation and extravasation of tumour cells. Regarding the important role of cancer metastasis for patients survival, further knowledge about the consequences of altered glycosylation patterns in tumour cells is needed which might eventually lead to the development of novel therapeutic approaches.
Collapse
Affiliation(s)
| | - Karen Legler
- Department of Gynecology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Karin Milde-Langosch
- Department of Gynecology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| |
Collapse
|
28
|
Rao CV, Janakiram NB, Mohammed A. Molecular Pathways: Mucins and Drug Delivery in Cancer. Clin Cancer Res 2017; 23:1373-1378. [PMID: 28039261 PMCID: PMC6038927 DOI: 10.1158/1078-0432.ccr-16-0862] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2016] [Revised: 11/29/2016] [Accepted: 11/30/2016] [Indexed: 12/12/2022]
Abstract
Over the past few decades, clinical and preclinical studies have clearly demonstrated the role of mucins in tumor development. It is well established that mucins form a barrier impeding drug access to target sites, leading to cancer chemoresistance. Recently gained knowledge regarding core enzyme synthesis has opened avenues to explore the possibility of disrupting mucin synthesis to improve drug efficacy. Cancer cells exploit aberrant mucin synthesis to efficiently mask the epithelial cells and ensure survival under hostile tumor microenvironment conditions. However, O-glycan synthesis enzyme core 2 beta 1,6 N-acetylglucosaminyltransferase (GCNT3/C2GnT-2) is overexpressed in Kras-driven mouse and human cancer, and inhibition of GCNT3 has been shown to disrupt mucin synthesis. This previously unrecognized developmental pathway might be responsible for aberrant mucin biosynthesis and chemoresistance. In this Molecular Pathways article, we briefly discuss the potential role of mucin synthesis in cancers, ways to improve drug delivery and disrupt mucin mesh to overcome chemoresistance by targeting mucin synthesis, and the unique opportunity to target the GCNT3 pathway for the prevention and treatment of cancers. Clin Cancer Res; 23(6); 1373-8. ©2016 AACR.
Collapse
Affiliation(s)
- Chinthalapally V Rao
- Center for Cancer Prevention and Drug Development, Hematology and Oncology Section, Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma.
| | - Naveena B Janakiram
- Center for Cancer Prevention and Drug Development, Hematology and Oncology Section, Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
| | - Altaf Mohammed
- Center for Cancer Prevention and Drug Development, Hematology and Oncology Section, Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma.
| |
Collapse
|
29
|
Klasić M, Krištić J, Korać P, Horvat T, Markulin D, Vojta A, Reiding KR, Wuhrer M, Lauc G, Zoldoš V. DNA hypomethylation upregulates expression of the MGAT3 gene in HepG2 cells and leads to changes in N-glycosylation of secreted glycoproteins. Sci Rep 2016; 6:24363. [PMID: 27073020 PMCID: PMC4829869 DOI: 10.1038/srep24363] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2015] [Accepted: 03/23/2016] [Indexed: 12/12/2022] Open
Abstract
Changes in N-glycosylation of plasma proteins are observed in many types of cancer, nevertheless, few studies suggest the exact mechanism involved in aberrant protein glycosylation. Here we studied the impact of DNA methylation on the N-glycome in the secretome of the HepG2 cell line derived from hepatocellular carcinoma (HCC). Since the majority of plasma glycoproteins originate from the liver, the HepG2 cells represent a good model for glycosylation changes in HCC that are detectable in blood, which is an easily accessible analytic material in a clinical setting. Two different concentrations of 5-aza-2′-deoxycytidine (5-aza-2dC) differentially affected global genome methylation and induced different glycan changes. Around twenty percent of 84 glyco-genes analysed changed expression level after the 5-aza-2dC treatment as a result of global genome hypomethylation. A correlation study between the changes in glyco-gene expression and the HepG2 glycosylation profile suggests that the MGAT3 gene might be responsible for the glycan changes consistently induced by both doses of 5-aza-2dC. Core-fucosylated tetra-antennary structures were decreased in quantity likely as a result of hypomethylated MGAT3 gene promoter followed by increased expression of this gene.
Collapse
Affiliation(s)
- Marija Klasić
- University of Zagreb Faculty of Science, Zagreb, Croatia
| | | | - Petra Korać
- University of Zagreb Faculty of Science, Zagreb, Croatia
| | | | - Dora Markulin
- University of Zagreb Faculty of Science, Zagreb, Croatia
| | | | - Karli R Reiding
- Center for Proteomics and Metabolomics, Leiden University Medical Center, Leiden, The Netherlands
| | - Manfred Wuhrer
- Center for Proteomics and Metabolomics, Leiden University Medical Center, Leiden, The Netherlands.,Division of BioAnalytical Chemistry, VU University Amsterdam, Amsterdam, The Netherlands
| | - Gordan Lauc
- Genos Glycoscience Research Laboratory, Zagreb, Croatia.,University of Zagreb Faculty of Pharmacy and Biochemistry, Zagreb, Croatia
| | - Vlatka Zoldoš
- University of Zagreb Faculty of Science, Zagreb, Croatia
| |
Collapse
|
30
|
Gao HJ, Chen YJ, Zuo D, Xiao MM, Li Y, Guo H, Zhang N, Chen RB. Quantitative proteomic analysis for high-throughput screening of differential glycoproteins in hepatocellular carcinoma serum. Cancer Biol Med 2015; 12:246-54. [PMID: 26487969 PMCID: PMC4607824 DOI: 10.7497/j.issn.2095-3941.2015.0010] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
OBJECTIVE Hepatocellular carcinoma (HCC) is a leading cause of cancer-related deaths. Novel serum biomarkers are required to increase the sensitivity and specificity of serum screening for early HCC diagnosis. This study employed a quantitative proteomic strategy to analyze the differential expression of serum glycoproteins between HCC and normal control serum samples. METHODS Lectin affinity chromatography (LAC) was used to enrich glycoproteins from the serum samples. Quantitative mass spectrometric analysis combined with stable isotope dimethyl labeling and 2D liquid chromatography (LC) separations were performed to examine the differential levels of the detected proteins between HCC and control serum samples. Western blot was used to analyze the differential expression levels of the three serum proteins. RESULTS A total of 2,280 protein groups were identified in the serum samples from HCC patients by using the 2D LC-MS/MS method. Up to 36 proteins were up-regulated in the HCC serum, whereas 19 proteins were down-regulated. Three differential glycoproteins, namely, fibrinogen gamma chain (FGG), FOS-like antigen 2 (FOSL2), and α-1,6-mannosylglycoprotein 6-β-N-acetylglucosaminyltransferase B (MGAT5B) were validated by Western blot. All these three proteins were up-regulated in the HCC serum samples. CONCLUSION A quantitative glycoproteomic method was established and proven useful to determine potential novel biomarkers for HCC.
Collapse
Affiliation(s)
- Hua-Jun Gao
- 1 Research Center of Basic Medical Sciences & School of Medical Laboratory, Tianjin Medical University, Tianjin 300070, China ; 2 Laboratory of Cancer Cell Biology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin Key Laboratory of Cancer Prevention and Therapy, Tianjin 300060, China
| | - Ya-Jing Chen
- 1 Research Center of Basic Medical Sciences & School of Medical Laboratory, Tianjin Medical University, Tianjin 300070, China ; 2 Laboratory of Cancer Cell Biology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin Key Laboratory of Cancer Prevention and Therapy, Tianjin 300060, China
| | - Duo Zuo
- 1 Research Center of Basic Medical Sciences & School of Medical Laboratory, Tianjin Medical University, Tianjin 300070, China ; 2 Laboratory of Cancer Cell Biology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin Key Laboratory of Cancer Prevention and Therapy, Tianjin 300060, China
| | - Ming-Ming Xiao
- 1 Research Center of Basic Medical Sciences & School of Medical Laboratory, Tianjin Medical University, Tianjin 300070, China ; 2 Laboratory of Cancer Cell Biology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin Key Laboratory of Cancer Prevention and Therapy, Tianjin 300060, China
| | - Ying Li
- 1 Research Center of Basic Medical Sciences & School of Medical Laboratory, Tianjin Medical University, Tianjin 300070, China ; 2 Laboratory of Cancer Cell Biology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin Key Laboratory of Cancer Prevention and Therapy, Tianjin 300060, China
| | - Hua Guo
- 1 Research Center of Basic Medical Sciences & School of Medical Laboratory, Tianjin Medical University, Tianjin 300070, China ; 2 Laboratory of Cancer Cell Biology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin Key Laboratory of Cancer Prevention and Therapy, Tianjin 300060, China
| | - Ning Zhang
- 1 Research Center of Basic Medical Sciences & School of Medical Laboratory, Tianjin Medical University, Tianjin 300070, China ; 2 Laboratory of Cancer Cell Biology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin Key Laboratory of Cancer Prevention and Therapy, Tianjin 300060, China
| | - Rui-Bing Chen
- 1 Research Center of Basic Medical Sciences & School of Medical Laboratory, Tianjin Medical University, Tianjin 300070, China ; 2 Laboratory of Cancer Cell Biology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin Key Laboratory of Cancer Prevention and Therapy, Tianjin 300060, China
| |
Collapse
|
31
|
Xiong Y, Jia M, Yuan J, Zhang C, Zhu Y, Kuang X, Lan L, Wang X. STAT3‑regulated long non‑coding RNAs lnc‑7SK and lnc‑IGF2‑AS promote hepatitis C virus replication. Mol Med Rep 2015; 12:6738-44. [PMID: 26328522 PMCID: PMC4626162 DOI: 10.3892/mmr.2015.4278] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2014] [Accepted: 07/31/2015] [Indexed: 12/16/2022] Open
Abstract
Long non‑coding RNAs (lncRNAs) are a class of RNAs that do not code protein but are important in diverse biological processes. In previous years, with the application of high‑throughput sequencing, a large number of lncRNAs associated with virus infections have been identified and intensively investigated, however, there are few studies examining the association between lncRNAs and HCV replication. Previous studies have demonstrated that signal transducer and activator of transcription 3 (STAT3) is activated by the hepatitis C virus (HCV) and in turn increases the replication of HCV. However, the detailed molecular mechanism is only partially understood. In the present study, using human lncRNA polymerase chain reaction (PCR) arrays, it was identified that lnc‑IGF2‑AS, lnc‑7SK, lnc‑SChLAP1 and lnc‑SRA1 are upregulated by STAT3. In addition, among these four lncRNAs, only lnc‑IGF2‑AS and lnc‑7SK were involved in HCV replication. Transfection of siRNA lnc‑7SK and siRNA lnc‑IGF2‑AS partially inhibited the replication of HCV in Huh7 cells. Data also indicated that when transfected with siRNA lnc‑7SK and siRNA lnc‑IGF2‑AS, the expression of phosphatidylinositol 4‑phosphate (PI4P), which was identified to be associated with HCV replication, was reduced. Thus, the present study identified two new types of lncRNAs, lnc‑IGF2‑AS and lnc‑7SK, which can be upregulated by STAT3 and are involved in HCV replication by regulating PI4P.
Collapse
Affiliation(s)
- Yulin Xiong
- Key Laboratory of Infectious Disease Research, Institute of Infectious Diseases of Chinese PLA, Southwest Hospital, Third Military Medical University, Chongqing 400038, P.R. China
| | - Ming Jia
- Key Laboratory of Infectious Disease Research, Institute of Infectious Diseases of Chinese PLA, Southwest Hospital, Third Military Medical University, Chongqing 400038, P.R. China
| | - Jing Yuan
- Key Laboratory of Infectious Disease Research, Institute of Infectious Diseases of Chinese PLA, Southwest Hospital, Third Military Medical University, Chongqing 400038, P.R. China
| | - Changjiang Zhang
- Key Laboratory of Infectious Disease Research, Institute of Infectious Diseases of Chinese PLA, Southwest Hospital, Third Military Medical University, Chongqing 400038, P.R. China
| | - Yan Zhu
- Key Laboratory of Infectious Disease Research, Institute of Infectious Diseases of Chinese PLA, Southwest Hospital, Third Military Medical University, Chongqing 400038, P.R. China
| | - Xuemei Kuang
- Key Laboratory of Infectious Disease Research, Institute of Infectious Diseases of Chinese PLA, Southwest Hospital, Third Military Medical University, Chongqing 400038, P.R. China
| | - Lin Lan
- Key Laboratory of Infectious Disease Research, Institute of Infectious Diseases of Chinese PLA, Southwest Hospital, Third Military Medical University, Chongqing 400038, P.R. China
| | - Xiaohong Wang
- Key Laboratory of Infectious Disease Research, Institute of Infectious Diseases of Chinese PLA, Southwest Hospital, Third Military Medical University, Chongqing 400038, P.R. China
| |
Collapse
|
32
|
Xiong Y, Yuan J, Zhang C, Zhu Y, Kuang X, Lan L, Wang X. The STAT3-regulated long non-coding RNA Lethe promote the HCV replication. Biomed Pharmacother 2015; 72:165-71. [PMID: 26054691 DOI: 10.1016/j.biopha.2015.04.019] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2015] [Accepted: 04/27/2015] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Long non-coding RNAs (lncRNAs) are a class of RNAs that do not code protein but play important roles in diverse biological processes. In recent years, with the application of high-throughput sequencing, a great deal of lncRNAs associated with virus infections have been discovered and intensively studied, but there are few studies about the relationship between lncRNAs and HCV replication. METHODS We identify that several lncRNAs can be upregulated and downregulated by phosphorylated STAT3 by using human PCR array method. And among these lncRNAs, lnc-Lethe was involved in the HCV replication. Transfection of siRNA Lethe partially blocked the replication of HCV in Huh7 cells. RESULTS In the present study, we have established that phosphorylated STAT3 can promote the HCV replication. Data also indicated that when transfected with siRNA Lethe, the expression levels of PKR, OAS and IRF1, which were all ISGs, were all up regulated. CONCLUSIONS Based on our findings from Lethe knockdown, we have identified that Lethe, which was upregulated by activated STAT3, may promoting the replication of HCV through a negative regulatory mechanism of type I IFN response.
Collapse
Affiliation(s)
- Yulin Xiong
- Institute of Infectious Diseases of Chinese PLA, Southwest Hospital, Third Military Medical University, Gaotanyan Centre Street No. 30, Shapingba district, Chongqing 400038, China
| | - Jing Yuan
- Institute of Infectious Diseases of Chinese PLA, Southwest Hospital, Third Military Medical University, Gaotanyan Centre Street No. 30, Shapingba district, Chongqing 400038, China
| | - Changjiang Zhang
- Institute of Infectious Diseases of Chinese PLA, Southwest Hospital, Third Military Medical University, Gaotanyan Centre Street No. 30, Shapingba district, Chongqing 400038, China
| | - Yan Zhu
- Institute of Infectious Diseases of Chinese PLA, Southwest Hospital, Third Military Medical University, Gaotanyan Centre Street No. 30, Shapingba district, Chongqing 400038, China
| | - Xuemei Kuang
- Institute of Infectious Diseases of Chinese PLA, Southwest Hospital, Third Military Medical University, Gaotanyan Centre Street No. 30, Shapingba district, Chongqing 400038, China
| | - Lin Lan
- Institute of Infectious Diseases of Chinese PLA, Southwest Hospital, Third Military Medical University, Gaotanyan Centre Street No. 30, Shapingba district, Chongqing 400038, China
| | - Xiaohong Wang
- Institute of Infectious Diseases of Chinese PLA, Southwest Hospital, Third Military Medical University, Gaotanyan Centre Street No. 30, Shapingba district, Chongqing 400038, China.
| |
Collapse
|