1
|
Lakshman AH, Wright ES. EvoWeaver: large-scale prediction of gene functional associations from coevolutionary signals. Nat Commun 2025; 16:3878. [PMID: 40274827 PMCID: PMC12022180 DOI: 10.1038/s41467-025-59175-6] [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: 02/05/2025] [Accepted: 04/09/2025] [Indexed: 04/26/2025] Open
Abstract
The known universe of uncharacterized proteins is expanding far faster than our ability to annotate their functions through laboratory study. Computational annotation approaches rely on similarity to previously studied proteins, thereby ignoring unstudied proteins. Coevolutionary approaches hold promise for injecting new information into our knowledge of the protein universe by linking proteins through 'guilt-by-association'. However, existing coevolutionary algorithms have insufficient accuracy and scalability to connect the entire universe of proteins. We present EvoWeaver, a method that weaves together 12 signals of coevolution to quantify the degree of shared evolution between genes. EvoWeaver accurately identifies proteins involved in protein complexes or separate steps of a biochemical pathway. We show the merits of EvoWeaver by partly reconstructing known biochemical pathways without any prior knowledge other than that available from genomic sequences. Applying EvoWeaver to 1545 gene groups from 8564 genomes reveals missing connections in popular databases and potentially undiscovered links between proteins.
Collapse
Affiliation(s)
- Aidan H Lakshman
- Department of Biomedical Informatics, University of Pittsburgh, Pittsburgh, PA, USA
| | - Erik S Wright
- Department of Biomedical Informatics, University of Pittsburgh, Pittsburgh, PA, USA.
- Center for Evolutionary Biology and Medicine, Pittsburgh, PA, USA.
| |
Collapse
|
2
|
Peng F, Feng Y, Yu S, He R, Wang H, Xie Y, Qin R. Pan-cancer analysis of B3GNT5 with potential implications for cancer immunotherapy and cancer stem cell stemness. PLoS One 2024; 19:e0314609. [PMID: 39671359 PMCID: PMC11642946 DOI: 10.1371/journal.pone.0314609] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Accepted: 11/11/2024] [Indexed: 12/15/2024] Open
Abstract
B3GNT5, a critical member of the β-1,3-N-acetylglucosaminyl transferase gene family involved in lactose and glycosphingolipids biosynthesis, has been documented to promote tumor-infiltrating T-cell responses. Our research utilized the Pan-Cancer dataset from The Cancer Genome Atlas (TCGA) to explore the functional role of B3GNT5. Our study demonstrated that the antibody-driven inhibition of B3GNT5 diminished T cell-mediated anti-tumor responses in both in vitro and in vivo settings. By analyzing RNA-seq data from Genotype-Tissue Expression (GTEx) and TCGA databases, we observed differential expression levels of B3GNT5 across various tumor types accompanied by an unfavorable prognostic correlation. We further utilized integrated clinical survival data from TCGA and immune cell infiltration scoring patterns to identify significant associations between B3GNT5 expression and immune checkpoints, cancer stemness, chemokines, chemokine receptors, and immune-activating genes. B3GNT5's expression was highly correlated with different immunoregulatory factors, including T cell infiltration, chemokine receptors, and activation genes. Subsequent experiments discovered that suppressing B3GNT5 expression in pancreatic adenocarcinoma cells significantly reduced their tumorigenicity by limiting sphere-forming ability and self-renewal capacity, thus underscoring B3GNT5's vital role as a prognostic factor in immune regulation across pan-cancer. Our findings suggest that B3GNT5 presents a viable target for cancer immunotherapy by enabling effective communication between cancer stem cells and immune cells during tumor treatment.
Collapse
Affiliation(s)
- Feng Peng
- Department of Biliary-Pancreatic Surgery, Affiliated Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yechen Feng
- Department of Biliary-Pancreatic Surgery, Affiliated Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Shuo Yu
- Department of Biliary-Pancreatic Surgery, Affiliated Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ruizhi He
- Department of Biliary-Pancreatic Surgery, Affiliated Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Hebin Wang
- Department of Biliary-Pancreatic Surgery, Affiliated Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yu Xie
- Department of Biliary-Pancreatic Surgery, Affiliated Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Renyi Qin
- Department of Biliary-Pancreatic Surgery, Affiliated Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
3
|
Kanack AJ, Prodoehl E, Ishihara-Aoki M, Aoki K, Dahms NM. Glycosphingolipids and their impact on platelet activity in a murine model of fabry disease. Sci Rep 2024; 14:29488. [PMID: 39604471 PMCID: PMC11603304 DOI: 10.1038/s41598-024-80633-6] [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: 05/13/2024] [Accepted: 11/21/2024] [Indexed: 11/29/2024] Open
Abstract
Fabry disease is an X-linked lysosomal storage disorder caused by deficiency of the lysosomal enzyme ⍺-galactosidase-A (⍺-Gal A), resulting in widespread accumulation of terminal galactose-containing glycosphingolipids (GSLs) and the impairment of multiple organ systems. Thrombotic events are common in Fabry patients, with strokes and heart attacks being significant contributors to a shortened lifespan in patients of both genders. Previously, we developed an ⍺-Gal A-knockout (KO) murine model that recapitulates most Fabry symptomologies and demonstrated that platelets from KO males become sensitized to agonist-mediated activation. In the current report, we used mass spectrometry, platelet-based assays and histology to define further the mechanisms linking GSL accumulation with thrombotic phenotypes in both sexes. Sera and platelets from ⍺-Gal A-KO females have elevated levels of Fabry-associated GSLs relative to wild-type females, but accumulated less of these GSLs than KO males. Correspondingly, KO females demonstrate a less severe thrombotic phenotypes than KO males. Notably, treatment of platelets from wild-type animals with globotriaosylceramide (Gb3) increased baseline platelet activation and aggregation. In contrast, several control GSLs did not stimulate platelet responses. These data suggest that chronically high concentrations of the Fabry-associated GSL, Gb3, contributes to the prothrombotic phenotypes experienced by Fabry patients by directly stimulating platelet activation.
Collapse
Affiliation(s)
- Adam J Kanack
- Department of Laboratory Medicine and Pathology, Mayo Clinic, 200 1st St. SW, Rochester, MN, 55905, USA.
| | - Eve Prodoehl
- Department of Biochemistry, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI, 53226, USA
| | - Mayumi Ishihara-Aoki
- Translational Metabolomics Shared Resource, Cancer Center, Medical College of Wisconsin, Milwaukee, WI, 53226, USA
| | - Kazuhiro Aoki
- Translational Metabolomics Shared Resource, Cancer Center, Medical College of Wisconsin, Milwaukee, WI, 53226, USA
- Department of Cell Biology, Neurobiology and Anatomy, Medical College of Wisconsin, Milwaukee, WI, 53226, USA
| | - Nancy M Dahms
- Department of Biochemistry, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI, 53226, USA.
| |
Collapse
|
4
|
Zhang X, Zeng B, Zhu H, Ma R, Yuan P, Chen Z, Su C, Liu Z, Yao X, Lawrence A, Liu Z, Zou J. Role of glycosphingolipid biosynthesis coregulators in malignant progression of thymoma. Int J Biol Sci 2023; 19:4442-4456. [PMID: 37781041 PMCID: PMC10535712 DOI: 10.7150/ijbs.83468] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2023] [Accepted: 08/14/2023] [Indexed: 10/03/2023] Open
Abstract
As the most common malignancy from mediastinum, the metabolic reprogramming of thymoma is important in its development. Nevertheless, the connection between the metabolic map and thymoma development is yet to be discovered. Thymoma was categorized into three subcategories by unsupervised clustering of molecular markers for metabolic pathway presentation in the TCGA dataset. Different genes and functions enriched were demonstrated through the utilization of metabolic Gene Ontology (GO) analysis. To identify the main contributors in the development of thymic malignancy, we utilized Gene Set Enrichment Analysis (GSEA), Gene Set Variation Analysis (GSVA), and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analysis. The prognosis of thymoma was evaluated by screening the essential pathways and genes using GSVA scores and machine learning classifiers. Furthermore, we integrated the transcriptomics findings with spectrum metabolomics investigation, detected through LC-MS/MS, in order to establish the essential controller network of metabolic reprogramming during thymoma progression. The thymoma prognosis is related to glycosphingolipid biosynthesis-lacto and neolacto series pathway, of what high B3GNT5 indicate poor survival. The investigation revealed that glycosphingolipid charts have a significant impact on metabolic dysfunction and could potentially serve as crucial targets in the clinical advancement of metabolic therapy.
Collapse
Affiliation(s)
- Xin Zhang
- Department of Thoracic Surgery, the First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, 510080, China
| | - Bo Zeng
- Department of Thoracic Surgery, the First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, 510080, China
| | - Haoshuai Zhu
- Department of Thoracic Surgery, the First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, 510080, China
| | - Rui Ma
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Disease, The Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510655, China
| | - Ping Yuan
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Disease, The Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510655, China
| | - Zhenguang Chen
- Department of Thoracic Surgery, the First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, 510080, China
| | - Chunhua Su
- Department of Thoracic Surgery, the First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, 510080, China
| | - Zhihao Liu
- Department of Thoracic Surgery, the First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, 510080, China
| | - Xiaojing Yao
- Department of Thoracic Surgery, the First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, 510080, China
| | - Aurora Lawrence
- School of Medicine, Stanford University, 450 Serra Mall, Stanford, CA 94305, USA
| | - Zhenguo Liu
- Department of Thoracic Surgery, the First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, 510080, China
| | - Jianyong Zou
- Department of Thoracic Surgery, the First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, 510080, China
| |
Collapse
|
5
|
Kent A, Crump LS, Davila E. Beyond αβ T cells: NK, iNKT, and γδT cell biology in leukemic patients and potential for off-the-shelf adoptive cell therapies for AML. Front Immunol 2023; 14:1202950. [PMID: 37654497 PMCID: PMC10465706 DOI: 10.3389/fimmu.2023.1202950] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2023] [Accepted: 07/24/2023] [Indexed: 09/02/2023] Open
Abstract
Acute myeloid leukemia (AML) remains an elusive disease to treat, let alone cure, even after highly intensive therapies such as stem cell transplants. Adoptive cell therapeutic strategies based on conventional alpha beta (αβ)T cells are an active area of research in myeloid neoplasms given their remarkable success in other hematologic malignancies, particularly B-cell-derived acute lymphoid leukemia, myeloma, and lymphomas. Several limitations have hindered clinical application of adoptive cell therapies in AML including lack of leukemia-specific antigens, on-target-off-leukemic toxicity, immunosuppressive microenvironments, and leukemic stem cell populations elusive to immune recognition and destruction. While there are promising T cell-based therapies including chimeric antigen receptor (CAR)-T designs under development, other cytotoxic lymphocyte cell subsets have unique phenotypes and capabilities that might be of additional benefit in AML treatment. Of particular interest are the natural killer (NK) and unconventional T cells known as invariant natural killer T (iNKT) and gamma delta (γδ) T cells. NK, iNKT, and γδT cells exhibit intrinsic anti-malignant properties, potential for alloreactivity, and human leukocyte-antigen (HLA)-independent function. Here we review the biology of each of these unconventional cytotoxic lymphocyte cell types and compare and contrast their strengths and limitations as the basis for adoptive cell therapies for AML.
Collapse
Affiliation(s)
- Andrew Kent
- Division of Medical Oncology, Department of Medicine, University of Colorado, Aurora, CO, United States
- Human Immunology and Immunotherapy Initiative, University of Colorado, Aurora, CO, United States
- Department of Medicine, University of Colorado Comprehensive Cancer Center, Aurora, CO, United States
| | | | - Eduardo Davila
- Division of Medical Oncology, Department of Medicine, University of Colorado, Aurora, CO, United States
- Human Immunology and Immunotherapy Initiative, University of Colorado, Aurora, CO, United States
- Department of Medicine, University of Colorado Comprehensive Cancer Center, Aurora, CO, United States
- Department of Medicine, University of Colorado, Aurora, CO, United States
| |
Collapse
|
6
|
Leng H, Zhang H, Li L, Zhang S, Wang Y, Chavda SJ, Galas-Filipowicz D, Lou H, Ersek A, Morris EV, Sezgin E, Lee YH, Li Y, Lechuga-Vieco AV, Tian M, Mi JQ, Yong K, Zhong Q, Edwards CM, Simon AK, Horwood NJ. Modulating glycosphingolipid metabolism and autophagy improves outcomes in pre-clinical models of myeloma bone disease. Nat Commun 2022; 13:7868. [PMID: 36550101 PMCID: PMC9780346 DOI: 10.1038/s41467-022-35358-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2021] [Accepted: 11/29/2022] [Indexed: 12/24/2022] Open
Abstract
Patients with multiple myeloma, an incurable malignancy of plasma cells, frequently develop osteolytic bone lesions that severely impact quality of life and clinical outcomes. Eliglustat, a U.S. Food and Drug Administration-approved glucosylceramide synthase inhibitor, reduced osteoclast-driven bone loss in preclinical in vivo models of myeloma. In combination with zoledronic acid, a bisphosphonate that treats myeloma bone disease, eliglustat provided further protection from bone loss. Autophagic degradation of TRAF3, a key step for osteoclast differentiation, was inhibited by eliglustat as evidenced by TRAF3 lysosomal and cytoplasmic accumulation. Eliglustat blocked autophagy by altering glycosphingolipid composition whilst restoration of missing glycosphingolipids rescued autophagy markers and TRAF3 degradation thus restoring osteoclastogenesis in bone marrow cells from myeloma patients. This work delineates both the mechanism by which glucosylceramide synthase inhibition prevents autophagic degradation of TRAF3 to reduce osteoclastogenesis as well as highlighting the clinical translational potential of eliglustat for the treatment of myeloma bone disease.
Collapse
Affiliation(s)
- Houfu Leng
- Kennedy Institute of Rheumatology, University of Oxford, Roosevelt Drive, Oxford, OX3 7FY, UK
| | - Hanlin Zhang
- Kennedy Institute of Rheumatology, University of Oxford, Roosevelt Drive, Oxford, OX3 7FY, UK
| | - Linsen Li
- Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Department of Pathophysiology, Shanghai Jiao Tong University School of Medicine, Shanghai, P.R. China
| | - Shuhao Zhang
- Kennedy Institute of Rheumatology, University of Oxford, Roosevelt Drive, Oxford, OX3 7FY, UK
- Computational Biology Department, Carnegie Mellon University, Pittsburgh, PA, 15217, USA
| | - Yanping Wang
- Institutes of Biology and Medical Sciences, Soochow University, Suzhou, P.R. China
| | - Selina J Chavda
- Department of Hematology, UCL Cancer Institute, University College London, London, UK
| | | | - Hantao Lou
- Ludwig Institute for Cancer Research, Nuffield Department of Medicine, University of Oxford, Oxford, OX3 7DQ, UK
| | - Adel Ersek
- Norwich Medical School, University of East Anglia, James Watson Road, Norwich, NR4 7UQ, UK
| | - Emma V Morris
- Nuffield Department of Surgical Sciences, Botnar Research Centre, University of Oxford, Old Road, Oxford, OX3 7LD, UK
| | - Erdinc Sezgin
- Science for Life Laboratory, Department of Women's and Children's Health, Karolinska Institute, Solna, Sweden
- MRC Weatherall Institute of Molecular Medicine, MRC Human Immunology Unit, Oxford, OX3 9DS, UK
| | - Yi-Hsuan Lee
- Kennedy Institute of Rheumatology, University of Oxford, Roosevelt Drive, Oxford, OX3 7FY, UK
- Norwich Medical School, University of East Anglia, James Watson Road, Norwich, NR4 7UQ, UK
| | - Yunsen Li
- Institutes of Biology and Medical Sciences, Soochow University, Suzhou, P.R. China
| | | | - Mei Tian
- Human Phenome Institute, Fudan University, 825 Zhangheng Road, Shanghai, P.R. China
| | - Jian-Qing Mi
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, RuiJin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, P.R. China
| | - Kwee Yong
- Department of Hematology, UCL Cancer Institute, University College London, London, UK
| | - Qing Zhong
- Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Department of Pathophysiology, Shanghai Jiao Tong University School of Medicine, Shanghai, P.R. China
| | - Claire M Edwards
- Nuffield Department of Surgical Sciences, Botnar Research Centre, University of Oxford, Old Road, Oxford, OX3 7LD, UK
- Nuffield Department of Orthopaedics, Rheumatology, and Musculoskeletal Sciences, Botnar Research Centre, University of Oxford, Old Road, Oxford, OX3 7LD, UK
| | - Anna Katharina Simon
- Kennedy Institute of Rheumatology, University of Oxford, Roosevelt Drive, Oxford, OX3 7FY, UK.
| | - Nicole J Horwood
- Kennedy Institute of Rheumatology, University of Oxford, Roosevelt Drive, Oxford, OX3 7FY, UK.
- Norwich Medical School, University of East Anglia, James Watson Road, Norwich, NR4 7UQ, UK.
| |
Collapse
|
7
|
Ung J, Tan SF, Fox TE, Shaw JJP, Vass LR, Costa-Pinheiro P, Garrett-Bakelman FE, Keng MK, Sharma A, Claxton DF, Levine RL, Tallman MS, Cabot MC, Kester M, Feith DJ, Loughran TP. Harnessing the power of sphingolipids: Prospects for acute myeloid leukemia. Blood Rev 2022; 55:100950. [PMID: 35487785 PMCID: PMC9475810 DOI: 10.1016/j.blre.2022.100950] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Revised: 03/31/2022] [Accepted: 04/04/2022] [Indexed: 11/02/2022]
Abstract
Acute myeloid leukemia (AML) is an aggressive, heterogenous malignancy characterized by clonal expansion of bone marrow-derived myeloid progenitor cells. While our current understanding of the molecular and genomic landscape of AML has evolved dramatically and opened avenues for molecularly targeted therapeutics to improve upon standard intensive induction chemotherapy, curative treatments are elusive, particularly in older patients. Responses to current AML treatments are transient and incomplete, necessitating the development of novel treatment strategies to improve outcomes. To this end, harnessing the power of bioactive sphingolipids to treat cancer shows great promise. Sphingolipids are involved in many hallmarks of cancer of paramount importance in AML. Leukemic blast survival is influenced by cellular levels of ceramide, a bona fide pro-death molecule, and its conversion to signaling molecules such as sphingosine-1-phosphate and glycosphingolipids. Preclinical studies demonstrate the efficacy of therapeutics that target dysregulated sphingolipid metabolism as well as their combinatorial synergy with clinically-relevant therapeutics. Thus, increased understanding of sphingolipid dysregulation may be exploited to improve AML patient care and outcomes. This review summarizes the current knowledge of dysregulated sphingolipid metabolism in AML, evaluates how pro-survival sphingolipids promote AML pathogenesis, and discusses the therapeutic potential of targeting these dysregulated sphingolipid pathways.
Collapse
Affiliation(s)
- Johnson Ung
- Department of Microbiology, Immunology, and Cancer Biology, University of Virginia School of Medicine, Charlottesville, VA, United States of America; Division of Hematology/Oncology, University of Virginia School of Medicine, Charlottesville, VA, United States of America; University of Virginia Cancer Center, Charlottesville, VA, United States of America
| | - Su-Fern Tan
- Division of Hematology/Oncology, University of Virginia School of Medicine, Charlottesville, VA, United States of America; University of Virginia Cancer Center, Charlottesville, VA, United States of America
| | - Todd E Fox
- University of Virginia Cancer Center, Charlottesville, VA, United States of America; Department of Pharmacology, University of Virginia School of Medicine, Charlottesville, VA, United States of America
| | - Jeremy J P Shaw
- University of Virginia Cancer Center, Charlottesville, VA, United States of America; Department of Experimental Pathology, University of Virginia School of Medicine, Charlottesville, VA, United States of America
| | - Luke R Vass
- University of Virginia Cancer Center, Charlottesville, VA, United States of America; Department of Experimental Pathology, University of Virginia School of Medicine, Charlottesville, VA, United States of America
| | - Pedro Costa-Pinheiro
- Cancer Biology, University of Pennsylvania, Philadelphia, PA, United States of America
| | - Francine E Garrett-Bakelman
- Division of Hematology/Oncology, University of Virginia School of Medicine, Charlottesville, VA, United States of America; University of Virginia Cancer Center, Charlottesville, VA, United States of America; Department of Biochemistry and Molecular Genetics, University of Virginia School of Medicine, Charlottesville, VA, United States of America
| | - Michael K Keng
- Division of Hematology/Oncology, University of Virginia School of Medicine, Charlottesville, VA, United States of America; University of Virginia Cancer Center, Charlottesville, VA, United States of America
| | - Arati Sharma
- Penn State Cancer Institute, Hershey, PA, United States of America
| | - David F Claxton
- Penn State Cancer Institute, Hershey, PA, United States of America
| | - Ross L Levine
- Leukemia Service, Memorial Sloan Kettering Cancer Center, New York, NY, United States of America; Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, United States of America
| | - Martin S Tallman
- Leukemia Service, Memorial Sloan Kettering Cancer Center, New York, NY, United States of America
| | - Myles C Cabot
- Department of Biochemistry and Molecular Biology, East Carolina University, Brody School of Medicine, Greenville, NC, United States of America; East Carolina Diabetes and Obesity Institute, East Carolina University, Brody School of Medicine, Greenville, NC, United States of America
| | - Mark Kester
- University of Virginia Cancer Center, Charlottesville, VA, United States of America; Department of Pharmacology, University of Virginia School of Medicine, Charlottesville, VA, United States of America
| | - David J Feith
- Division of Hematology/Oncology, University of Virginia School of Medicine, Charlottesville, VA, United States of America; University of Virginia Cancer Center, Charlottesville, VA, United States of America
| | - Thomas P Loughran
- Department of Microbiology, Immunology, and Cancer Biology, University of Virginia School of Medicine, Charlottesville, VA, United States of America; Division of Hematology/Oncology, University of Virginia School of Medicine, Charlottesville, VA, United States of America; University of Virginia Cancer Center, Charlottesville, VA, United States of America.
| |
Collapse
|
8
|
Miao Z, Cao Q, Liao R, Chen X, Li X, Bai L, Ma C, Deng X, Dai Z, Li J, Dong C. Elevated transcription and glycosylation of B3GNT5 promotes breast cancer aggressiveness. J Exp Clin Cancer Res 2022; 41:169. [PMID: 35526049 PMCID: PMC9077843 DOI: 10.1186/s13046-022-02375-5] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Accepted: 04/27/2022] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Basal-like breast cancer (BLBC) is the most aggressive subtype of breast cancer because of its aggressive biological characteristics and no effective targeted agents. However, the mechanism underlying its aggressive behavior remain poorly understood. β1,3-N-acetylglucosaminyltransferase V (B3GNT5) overexpression occurs specifically in BLBC. Here, we studied the possible molecular mechanisms of B3GBT5 promoting the aggressiveness of BLBC. METHODS The potential effects of B3GNT5 on breast cancer cells were tested by colony formation, mammosphere formation, cell proliferation assay, flow cytometry and Western blotting. The glycosylation patterns of B3GNT5 and associated functions were determined by Western blotting, quantitative real-time PCR and flow cytometry. The effect of B3GNT5 expression on BLBC was assessed by in vitro and in vivo tumorigenesis model. RESULTS In this study, we showed that B3GNT5 copy number amplification and hypomethylation of B3GNT5 promoter contributed to the overexpression of B3GNT5 in BLBC. Knockout of B3GNT5 strongly reduced surface expression of SSEA-1 and impeded cancer stem cell (CSC)-like properties of BLBC cells. Our results also showed that B3GNT5 protein was heavily N-glycosylated, which is critical for its protein stabilization. Clinically, elevated expression of B3GNT5 was correlated with high grade, large tumor size and poor survival, indicating poor prognosis of breast cancer patients. CONCLUSIONS Our work uncovers the critical association of B3GNT5 overexpression and glycosylation with enhanced CSCs properties in BLBC. These findings suggest that B3GNT5 has the potential to become a prognostic marker and therapeutic target for BLBC.
Collapse
Affiliation(s)
- Zhaorui Miao
- Department of Pathology and Pathophysiology, Department of Colorectal Surgery and Oncology, Key Laboratory of Cancer Prevention and Intervention, The Second Affiliated Hospital, Ministry of Education, Zhejiang University School of Medicine, 310058, Hangzhou, China
- Zhejiang Key Laboratory for Disease Proteomics, Zhejiang University School of Medicine, 310058, Hangzhou, China
| | - Qianhua Cao
- Department of Pathology and Pathophysiology, Department of Colorectal Surgery and Oncology, Key Laboratory of Cancer Prevention and Intervention, The Second Affiliated Hospital, Ministry of Education, Zhejiang University School of Medicine, 310058, Hangzhou, China
- Zhejiang Key Laboratory for Disease Proteomics, Zhejiang University School of Medicine, 310058, Hangzhou, China
| | - Ruocen Liao
- Department of Pathology and Pathophysiology, Department of Colorectal Surgery and Oncology, Key Laboratory of Cancer Prevention and Intervention, The Second Affiliated Hospital, Ministry of Education, Zhejiang University School of Medicine, 310058, Hangzhou, China
- Zhejiang Key Laboratory for Disease Proteomics, Zhejiang University School of Medicine, 310058, Hangzhou, China
| | - Xingyu Chen
- Department of Pathology and Pathophysiology, Department of Colorectal Surgery and Oncology, Key Laboratory of Cancer Prevention and Intervention, The Second Affiliated Hospital, Ministry of Education, Zhejiang University School of Medicine, 310058, Hangzhou, China
- Zhejiang Key Laboratory for Disease Proteomics, Zhejiang University School of Medicine, 310058, Hangzhou, China
| | - Xiaoli Li
- Abcam Plc, 1418-32 Moganshan Road, 311500, Hangzhou, China
| | - Longchang Bai
- Department of Pathology and Pathophysiology, Department of Colorectal Surgery and Oncology, Key Laboratory of Cancer Prevention and Intervention, The Second Affiliated Hospital, Ministry of Education, Zhejiang University School of Medicine, 310058, Hangzhou, China
- Zhejiang Key Laboratory for Disease Proteomics, Zhejiang University School of Medicine, 310058, Hangzhou, China
| | - Chenglong Ma
- Department of Pathology and Pathophysiology, Department of Colorectal Surgery and Oncology, Key Laboratory of Cancer Prevention and Intervention, The Second Affiliated Hospital, Ministry of Education, Zhejiang University School of Medicine, 310058, Hangzhou, China
- Zhejiang Key Laboratory for Disease Proteomics, Zhejiang University School of Medicine, 310058, Hangzhou, China
| | - Xinyue Deng
- Department of Breast Surgery, First Affiliated Hospital, Zhejiang University School of Medicine, 310058, Hangzhou, China
| | - Zhijun Dai
- Department of Breast Surgery, First Affiliated Hospital, Zhejiang University School of Medicine, 310058, Hangzhou, China.
| | - Jun Li
- Department of Pathology and Pathophysiology, Department of Colorectal Surgery and Oncology, Key Laboratory of Cancer Prevention and Intervention, The Second Affiliated Hospital, Ministry of Education, Zhejiang University School of Medicine, 310058, Hangzhou, China.
| | - Chenfang Dong
- Department of Pathology and Pathophysiology, Department of Colorectal Surgery and Oncology, Key Laboratory of Cancer Prevention and Intervention, The Second Affiliated Hospital, Ministry of Education, Zhejiang University School of Medicine, 310058, Hangzhou, China.
- Zhejiang Key Laboratory for Disease Proteomics, Zhejiang University School of Medicine, 310058, Hangzhou, China.
| |
Collapse
|
9
|
He X, Guan F, Lei L. Structure and function of glycosphingolipids on small extracellular vesicles. Glycoconj J 2022; 39:197-205. [PMID: 35201531 PMCID: PMC8866925 DOI: 10.1007/s10719-022-10052-0] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Revised: 02/12/2022] [Accepted: 02/16/2022] [Indexed: 12/22/2022]
Abstract
Extracellular vesicles (EVs) are membrane-delineated particles secreted by most types of cells under both normal and pathophysiological conditions. EVs are believed to mediate intercellular communication by serving as carriers of different bioactive ingredients, including proteins, nucleic acids and lipids. Glycoconjugates are complex molecules consisting of covalently linked carbohydrate with proteins or lipids. These glycoconjugates play essential roles in the sorting of vesicular protein and the uptake of small extracellular vesicles (30–100 nm, sEVs) into recipient cells. Glycosphingolipids (GSLs), one subtype of glycolipids, which are ubiquitous membrane components in almost all living organisms, are also commonly distributed on sEVs. However, the study of functional roles of GSLs on sEVs are far behind than other functional cargos. The purpose of this review is to highlight the importance of GSLs on sEVs. Initially, we described classification and structure of GSLs. Then, we briefly introduced the essential functions of GSLs, which are able to interact with functional membrane proteins, such as growth factor receptors, integrins and tetraspanins, to modulate cell growth, adhesion and cell motility. In addition, we discussed analytical methods for studying GSLs on sEVs. Finally, we focused on the function of GSLs on sEVs, including regulating the aggregation of extracellular α-synuclein (α-syn) or extracellular amyloid-β (Aβ) and influencing tumor cell malignancy.
Collapse
Affiliation(s)
- Xin He
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Provincial Key Laboratory of Biotechnology, College of Life Sciences, Northwest University, Xi'an, China
| | - Feng Guan
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Provincial Key Laboratory of Biotechnology, College of Life Sciences, Northwest University, Xi'an, China.
| | - Lei Lei
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Provincial Key Laboratory of Biotechnology, College of Life Sciences, Northwest University, Xi'an, China.
| |
Collapse
|
10
|
Wang D, Zhang T, Madunić K, de Waard AA, Blöchl C, Mayboroda OA, Griffioen M, Spaapen RM, Huber CG, Lageveen-Kammeijer GSM, Wuhrer M. Glycosphingolipid-Glycan Signatures of Acute Myeloid Leukemia Cell Lines Reflect Hematopoietic Differentiation. J Proteome Res 2022; 21:1029-1040. [PMID: 35168327 PMCID: PMC8981326 DOI: 10.1021/acs.jproteome.1c00911] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Aberrant expression of certain glycosphingolipids (GSLs) is associated with the differentiation of acute myeloid leukemia (AML) cells. However, the expression patterns of GSLs in AML are still poorly explored because of their complexity, the presence of multiple isomeric structures, and tedious analytical procedures. In this study, we performed an in-depth GSL glycan analysis of 19 AML cell lines using porous graphitized carbon liquid chromatography-mass spectrometry revealing strikingly different GSL glycan profiles between the various AML cell lines. The cell lines of the M6 subtype showed a high expression of gangliosides with α2,3-sialylation and Neu5Gc, while the M2 and M5 subtypes were characterized by high expression of (neo)lacto-series glycans and Lewis A/X antigens. Integrated analysis of glycomics and available transcriptomics data revealed the association of GSL glycan abundances with the transcriptomics expression of certain glycosyltransferases (GTs) and transcription factors (TFs). In addition, correlations were found between specific GTs and TFs. Our data reveal TFs GATA2, GATA1, and RUNX1 as candidate inducers of the expression of gangliosides and sialylation via regulation of the GTs ST3GAL2 and ST8SIA1. In conclusion, we show that GSL glycan expression levels are associated with hematopoietic AML classifications and TF and GT gene expression. Further research is needed to dissect the regulation of GSL expression and its role in hematopoiesis and associated malignancies.
Collapse
Affiliation(s)
- Di Wang
- Center for Proteomics and Metabolomics, Leiden University Medical Center, Postbus 9600, 2300 RC Leiden, The Netherlands
| | - Tao Zhang
- Center for Proteomics and Metabolomics, Leiden University Medical Center, Postbus 9600, 2300 RC Leiden, The Netherlands
| | - Katarina Madunić
- Center for Proteomics and Metabolomics, Leiden University Medical Center, Postbus 9600, 2300 RC Leiden, The Netherlands
| | - Antonius A de Waard
- Department of Immunopathology, Sanquin Research, 1066 CX Amsterdam, The Netherlands.,Landsteiner Laboratory, Amsterdam UMC, University of Amsterdam, 1066 CX Amsterdam, The Netherlands
| | - Constantin Blöchl
- Center for Proteomics and Metabolomics, Leiden University Medical Center, Postbus 9600, 2300 RC Leiden, The Netherlands.,Department of Biosciences, University of Salzburg, Hellbrunnerstrasse 34, 5020 Salzburg, Austria
| | - Oleg A Mayboroda
- Center for Proteomics and Metabolomics, Leiden University Medical Center, Postbus 9600, 2300 RC Leiden, The Netherlands
| | - Marieke Griffioen
- Department of Hematology, Leiden University Medical Center, Postbus 9600, 2300 RC Leiden, The Netherlands
| | - Robbert M Spaapen
- Department of Immunopathology, Sanquin Research, 1066 CX Amsterdam, The Netherlands.,Landsteiner Laboratory, Amsterdam UMC, University of Amsterdam, 1066 CX Amsterdam, The Netherlands
| | - Christian G Huber
- Department of Biosciences, University of Salzburg, Hellbrunnerstrasse 34, 5020 Salzburg, Austria
| | | | - Manfred Wuhrer
- Center for Proteomics and Metabolomics, Leiden University Medical Center, Postbus 9600, 2300 RC Leiden, The Netherlands
| |
Collapse
|
11
|
Pothukuchi P, Agliarulo I, Pirozzi M, Rizzo R, Russo D, Turacchio G, Nüchel J, Yang JS, Gehin C, Capolupo L, Hernandez-Corbacho MJ, Biswas A, Vanacore G, Dathan N, Nitta T, Henklein P, Thattai M, Inokuchi JI, Hsu VW, Plomann M, Obeid LM, Hannun YA, Luini A, D'Angelo G, Parashuraman S. GRASP55 regulates intra-Golgi localization of glycosylation enzymes to control glycosphingolipid biosynthesis. EMBO J 2021; 40:e107766. [PMID: 34516001 PMCID: PMC8521277 DOI: 10.15252/embj.2021107766] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Revised: 07/26/2021] [Accepted: 08/06/2021] [Indexed: 12/24/2022] Open
Abstract
The Golgi apparatus, the main glycosylation station of the cell, consists of a stack of discontinuous cisternae. Glycosylation enzymes are usually concentrated in one or two specific cisternae along the cis‐trans axis of the organelle. How such compartmentalized localization of enzymes is achieved and how it contributes to glycosylation are not clear. Here, we show that the Golgi matrix protein GRASP55 directs the compartmentalized localization of key enzymes involved in glycosphingolipid (GSL) biosynthesis. GRASP55 binds to these enzymes and prevents their entry into COPI‐based retrograde transport vesicles, thus concentrating them in the trans‐Golgi. In genome‐edited cells lacking GRASP55, or in cells expressing mutant enzymes without GRASP55 binding sites, these enzymes relocate to the cis‐Golgi, which affects glycosphingolipid biosynthesis by changing flux across metabolic branch points. These findings reveal a mechanism by which a matrix protein regulates polarized localization of glycosylation enzymes in the Golgi and controls competition in glycan biosynthesis.
Collapse
Affiliation(s)
- Prathyush Pothukuchi
- Institute of Biochemistry and Cell Biology, National Research Council of Italy, Rome, Italy
| | - Ilenia Agliarulo
- Institute of Biochemistry and Cell Biology, National Research Council of Italy, Rome, Italy
| | - Marinella Pirozzi
- Institute of Biochemistry and Cell Biology, National Research Council of Italy, Rome, Italy
| | - Riccardo Rizzo
- Institute of Biochemistry and Cell Biology, National Research Council of Italy, Rome, Italy
| | - Domenico Russo
- Institute of Biochemistry and Cell Biology, National Research Council of Italy, Rome, Italy
| | - Gabriele Turacchio
- Institute of Biochemistry and Cell Biology, National Research Council of Italy, Rome, Italy
| | - Julian Nüchel
- Medical Faculty, Center for Biochemistry, University of Cologne, Cologne, Germany
| | - Jia-Shu Yang
- Division of Rheumatology, Inflammation and Immunity, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Charlotte Gehin
- École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Laura Capolupo
- École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | | | - Ansuman Biswas
- National Center of Biological Sciences, Bengaluru, India
| | - Giovanna Vanacore
- Institute of Biochemistry and Cell Biology, National Research Council of Italy, Rome, Italy
| | - Nina Dathan
- Institute of Biochemistry and Cell Biology, National Research Council of Italy, Rome, Italy
| | - Takahiro Nitta
- Division of Glycopathology, Institute of Molecular Biomembrane and Glycobiology, Tohoku Medical and Pharmaceutical University, Sendai, Japan
| | - Petra Henklein
- Universitätsmedizin Berlin Institut für Biochemie Charité CrossOver Charitéplatz 1 / Sitz, Berlin, Germany
| | - Mukund Thattai
- National Center of Biological Sciences, Bengaluru, India
| | - Jin-Ichi Inokuchi
- Division of Glycopathology, Institute of Molecular Biomembrane and Glycobiology, Tohoku Medical and Pharmaceutical University, Sendai, Japan
| | - Victor W Hsu
- Division of Rheumatology, Inflammation and Immunity, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Markus Plomann
- Medical Faculty, Center for Biochemistry, University of Cologne, Cologne, Germany
| | - Lina M Obeid
- Stony Brook University Medical Center, Stony Brook, NY, USA
| | - Yusuf A Hannun
- Stony Brook University Medical Center, Stony Brook, NY, USA
| | - Alberto Luini
- Institute of Biochemistry and Cell Biology, National Research Council of Italy, Rome, Italy
| | - Giovanni D'Angelo
- Institute of Biochemistry and Cell Biology, National Research Council of Italy, Rome, Italy.,École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | | |
Collapse
|
12
|
Differential Regulation of Lacto-/Neolacto- Glycosphingolipid Biosynthesis Pathway Reveals Transcription Factors as Potential Candidates in Triple-Negative Breast Cancer. Cancers (Basel) 2021; 13:cancers13133330. [PMID: 34283051 PMCID: PMC8268693 DOI: 10.3390/cancers13133330] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2021] [Revised: 06/26/2021] [Accepted: 06/29/2021] [Indexed: 12/31/2022] Open
Abstract
Triple-negative breast cancer (TNBC) is an aggressive breast cancer with limited treatment options. Glycosylation has been implicated in cancer development, but TNBC-specific glycosylation pathways have not been examined. Here, we applied bioinformatic analyses on public datasets to discover TNBC-specific glycogenes and pathways, as well as their upstream regulatory mechanisms. Unsupervised clustering of 345 glycogene expressions in breast cancer datasets revealed a relative homogenous expression pattern in basal-like TNBC subtype. Differential expression analyses of the 345 glycogenes between basal-like TNBC (hereafter termed TNBC) and other BC subtypes, or normal controls, revealed 84 differential glycogenes in TNBC. Pathway enrichment showed two common TNBC-enriched pathways across all three datasets, cell cycle and lacto-/neolacto- glycosphingolipid (GSL) biosynthesis, while a total of four glycosylation-related pathways were significantly enriched in TNBC. We applied a selection criterion of the top 50% differential anabolic/catabolic glycogenes in the enriched pathways to define 34 TNBC-specific glycogenes. The lacto-/neolacto- GSL biosynthesis pathway was the most highly enriched, with seven glycogenes all up-regulated in TNBC. This data led us to investigate the hypothesis that a common upstream mechanism in TNBC up-regulates the lacto-/neolacto-GSL biosynthesis pathway. Using public multi-omic datasets, we excluded the involvement of copy-number alteration and DNA methylation, but identified three transcription factors (AR, GATA3 and ZNG622) that each target three candidate genes in the lacto-/neolacto- GSL biosynthesis pathway. Interestingly, a subset of TNBC has been reported to express AR and GATA3, and AR antagonists are being trialed for TNBC. Our findings suggest that AR and GATA3 may contribute to TNBC via GSL regulation, and provide a list of candidate glycogenes for further investigation.
Collapse
|
13
|
Jongsma MLM, Neefjes J, Spaapen RM. Playing hide and seek: Tumor cells in control of MHC class I antigen presentation. Mol Immunol 2021; 136:36-44. [PMID: 34082257 DOI: 10.1016/j.molimm.2021.05.009] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Revised: 04/07/2021] [Accepted: 05/18/2021] [Indexed: 12/15/2022]
Abstract
MHC class I (MHC-I) molecules present a blueprint of the intracellular proteome to T cells allowing them to control infection or malignant transformation. As a response, pathogens and tumor cells often downmodulate MHC-I mediated antigen presentation to escape from immune surveillance. Although the fundamental rules of antigen presentation are known in detail, the players in this system are not saturated and new modules of regulation have recently been uncovered. Here, we update the understanding of antigen presentation by MHC-I molecules and how this can be exploited by tumors to prevent exposure of the intracellular proteome. This knowledge can provide new ways to improve immune responses against tumors and pathogens.
Collapse
Affiliation(s)
- M L M Jongsma
- Department of Cell and Chemical Biology, Oncode Institute, Leiden University Medical Center, Leiden, the Netherlands
| | - J Neefjes
- Department of Cell and Chemical Biology, Oncode Institute, Leiden University Medical Center, Leiden, the Netherlands
| | - R M Spaapen
- Department of Immunopathology, Sanquin Research, Amsterdam, the Netherlands; Landsteiner Laboratory, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands; Cancer Center Amsterdam, Amsterdam, the Netherlands.
| |
Collapse
|
14
|
Jongsma MLM, de Waard AA, Raaben M, Zhang T, Cabukusta B, Platzer R, Blomen VA, Xagara A, Verkerk T, Bliss S, Kong X, Gerke C, Janssen L, Stickel E, Holst S, Plomp R, Mulder A, Ferrone S, Claas FHJ, Heemskerk MHM, Griffioen M, Halenius A, Overkleeft H, Huppa JB, Wuhrer M, Brummelkamp TR, Neefjes J, Spaapen RM. The SPPL3-Defined Glycosphingolipid Repertoire Orchestrates HLA Class I-Mediated Immune Responses. Immunity 2021; 54:132-150.e9. [PMID: 33271119 PMCID: PMC8722104 DOI: 10.1016/j.immuni.2020.11.003] [Citation(s) in RCA: 62] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2020] [Revised: 09/25/2020] [Accepted: 11/06/2020] [Indexed: 12/26/2022]
Abstract
HLA class I (HLA-I) glycoproteins drive immune responses by presenting antigens to cognate CD8+ T cells. This process is often hijacked by tumors and pathogens for immune evasion. Because options for restoring HLA-I antigen presentation are limited, we aimed to identify druggable HLA-I pathway targets. Using iterative genome-wide screens, we uncovered that the cell surface glycosphingolipid (GSL) repertoire determines effective HLA-I antigen presentation. We show that absence of the protease SPPL3 augmented B3GNT5 enzyme activity, resulting in upregulation of surface neolacto-series GSLs. These GSLs sterically impeded antibody and receptor interactions with HLA-I and diminished CD8+ T cell activation. Furthermore, a disturbed SPPL3-B3GNT5 pathway in glioma correlated with decreased patient survival. We show that the immunomodulatory effect could be reversed through GSL synthesis inhibition using clinically approved drugs. Overall, our study identifies a GSL signature that inhibits immune recognition and represents a potential therapeutic target in cancer, infection, and autoimmunity.
Collapse
Affiliation(s)
- Marlieke L M Jongsma
- Department of Immunopathology, Sanquin Research, Amsterdam, the Netherlands; Landsteiner Laboratory, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands; Cancer Center Amsterdam, Amsterdam, the Netherlands; Oncode Institute and Department of Cell and Chemical Biology, LUMC, Leiden, the Netherlands
| | - Antonius A de Waard
- Department of Immunopathology, Sanquin Research, Amsterdam, the Netherlands; Landsteiner Laboratory, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands; Cancer Center Amsterdam, Amsterdam, the Netherlands
| | - Matthijs Raaben
- Oncode Institute, Division of Biochemistry, the Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Tao Zhang
- Center for Proteomics and Metabolics, LUMC, Leiden, the Netherlands
| | - Birol Cabukusta
- Oncode Institute and Department of Cell and Chemical Biology, LUMC, Leiden, the Netherlands
| | - René Platzer
- Institut für Hygiene und Angewandte Immunologie, Vienna, Austria
| | - Vincent A Blomen
- Oncode Institute, Division of Biochemistry, the Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Anastasia Xagara
- Department of Immunopathology, Sanquin Research, Amsterdam, the Netherlands; Landsteiner Laboratory, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands; Cancer Center Amsterdam, Amsterdam, the Netherlands
| | - Tamara Verkerk
- Department of Immunopathology, Sanquin Research, Amsterdam, the Netherlands; Landsteiner Laboratory, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands; Cancer Center Amsterdam, Amsterdam, the Netherlands
| | - Sophie Bliss
- Department of Immunopathology, Sanquin Research, Amsterdam, the Netherlands; Landsteiner Laboratory, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands; Cancer Center Amsterdam, Amsterdam, the Netherlands
| | - Xiangrui Kong
- Department of Immunopathology, Sanquin Research, Amsterdam, the Netherlands; Landsteiner Laboratory, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands; Cancer Center Amsterdam, Amsterdam, the Netherlands
| | - Carolin Gerke
- Institute of Virology, Medical Center University of Freiburg, Freiburg, Germany; Faculty of Medicine, University of Freiburg, Freiburg, Germany; Spemann Graduate School of Biology and Medicine, University of Freiburg, Freiburg, Germany; Faculty of Biology, University of Freiburg, Freiburg, Germany
| | - Lennert Janssen
- Oncode Institute and Department of Cell and Chemical Biology, LUMC, Leiden, the Netherlands
| | - Elmer Stickel
- Oncode Institute, Division of Biochemistry, the Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Stephanie Holst
- Center for Proteomics and Metabolics, LUMC, Leiden, the Netherlands
| | - Rosina Plomp
- Center for Proteomics and Metabolics, LUMC, Leiden, the Netherlands
| | - Arend Mulder
- Department of Immunology, LUMC, Leiden, the Netherlands
| | - Soldano Ferrone
- Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | | | - Mirjam H M Heemskerk
- Department of Hematology, Leiden University Medical Center, Leiden, the Netherlands
| | - Marieke Griffioen
- Department of Hematology, Leiden University Medical Center, Leiden, the Netherlands
| | - Anne Halenius
- Institute of Virology, Medical Center University of Freiburg, Freiburg, Germany; Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Hermen Overkleeft
- Leiden Institute of Chemistry, Leiden University, Leiden, the Netherlands
| | - Johannes B Huppa
- Institut für Hygiene und Angewandte Immunologie, Vienna, Austria
| | - Manfred Wuhrer
- Center for Proteomics and Metabolics, LUMC, Leiden, the Netherlands
| | - Thijn R Brummelkamp
- Oncode Institute, Division of Biochemistry, the Netherlands Cancer Institute, Amsterdam, the Netherlands; CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria; Cancer Genomics Center, Amsterdam, the Netherlands
| | - Jacques Neefjes
- Oncode Institute and Department of Cell and Chemical Biology, LUMC, Leiden, the Netherlands
| | - Robbert M Spaapen
- Department of Immunopathology, Sanquin Research, Amsterdam, the Netherlands; Landsteiner Laboratory, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands; Cancer Center Amsterdam, Amsterdam, the Netherlands.
| |
Collapse
|
15
|
Deciphering the Importance of Glycosphingolipids on Cellular and Molecular Mechanisms Associated with Epithelial-to-Mesenchymal Transition in Cancer. Biomolecules 2021; 11:biom11010062. [PMID: 33418847 PMCID: PMC7824851 DOI: 10.3390/biom11010062] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2020] [Revised: 12/31/2020] [Accepted: 01/02/2021] [Indexed: 12/12/2022] Open
Abstract
Every living cell is covered with a dense and complex layer of glycans on the cell surface, which have important functions in the interaction between cells and their environment. Glycosphingolipids (GSLs) are glycans linked to lipid molecules that together with sphingolipids, sterols, and proteins form plasma membrane lipid rafts that contribute to membrane integrity and provide specific recognition sites. GSLs are subdivided into three major series (globo-, ganglio-, and neolacto-series) and are synthesized in a non-template driven process by enzymes localized in the ER and Golgi apparatus. Altered glycosylation of lipids are known to be involved in tumor development and metastasis. Metastasis is frequently linked with reversible epithelial-to-mesenchymal transition (EMT), a process involved in tumor progression, and the formation of new distant metastatic sites (mesenchymal-to-epithelial transition or MET). On a single cell basis, cancer cells lose their epithelial features to gain mesenchymal characteristics via mechanisms influenced by the composition of the GSLs on the cell surface. Here, we summarize the literature on GSLs in the context of reversible and cancer-associated EMT and discuss how the modification of GSLs at the cell surface may promote this process.
Collapse
|
16
|
Jeong HY, Park S, Kim H, Moon S, Lee S, Lee SH, Kim S. B3GNT5 is a novel marker correlated with stem-like phenotype and poor clinical outcome in human gliomas. CNS Neurosci Ther 2020; 26:1147-1154. [PMID: 32677340 PMCID: PMC7564194 DOI: 10.1111/cns.13439] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Revised: 06/23/2020] [Accepted: 06/24/2020] [Indexed: 12/14/2022] Open
Abstract
AIMS Glioblastoma multiforme (GBM) is the most lethal tumor with a median patient survival of 14 to 15 months. Glioma stem cells (GSCs) play a critical role in tumor initiation and therapeutic resistance in GBM. B3GNT5 has been suggested as the key glycosyltransferase in the biosynthesis of the (neo-) lacto series of glycosphingolipid. In this study, we evaluated the B3GNT5 expression in GSCs as well as the correlation with clinical data in GBM. METHODS The mRNA levels of B3GNT5 in normal astrocytes, four glioma cell lines, and four GSCs were evaluated using real-time PCR. Small interference RNAs (siRNAs) were used to inhibit B3GNT5 expression and analyze its ability to form neurospheres. Statistical analyses were conducted to determine the association with B3GNT5 expression and tumor grade and GBM subtypes as well as patient survival using public datasets. RESULTS B3GNT5 expression was significantly elevated in GSCs compared with normal astrocytes, glioma cell lines, and their matched differentiated tumor cells. Knockdown of B3GNT5 in GSCs decreased the neurosphere formation. Patients with high B3GNT5 expression had a short overall survival. B3GNT5 is correlated with classical and mesenchymal GBM subtypes. CONCLUSION The findings suggest the central role of B3GNT5 in regulating malignancy of GBM.
Collapse
Affiliation(s)
- Hang Yeon Jeong
- Department of Animal ScienceCollege of Agriculture and Life SciencesChonnam National UniversityGwangjuKorea
| | - Seo‐Young Park
- Department of Animal ScienceCollege of Agriculture and Life SciencesChonnam National UniversityGwangjuKorea
| | - Hyun‐Jin Kim
- Department of Animal ScienceCollege of Agriculture and Life SciencesChonnam National UniversityGwangjuKorea
| | - Seungju Moon
- Department of Animal ScienceCollege of Agriculture and Life SciencesChonnam National UniversityGwangjuKorea
| | - Seongsoo Lee
- Gwangju CenterKorea Basic Science InstituteGwangjuKorea
| | - Seung Ho Lee
- Department of Nano‐BioengineeringIncheon National UniversityIncheonKorea
| | - Sung‐Hak Kim
- Department of Animal ScienceCollege of Agriculture and Life SciencesChonnam National UniversityGwangjuKorea
- Gwangju CenterKorea Basic Science InstituteGwangjuKorea
| |
Collapse
|
17
|
Shimizu K, Iyoda T, Yamasaki S, Kadowaki N, Tojo A, Fujii SI. NK and NKT Cell-Mediated Immune Surveillance against Hematological Malignancies. Cancers (Basel) 2020; 12:cancers12040817. [PMID: 32231116 PMCID: PMC7226455 DOI: 10.3390/cancers12040817] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2020] [Revised: 03/24/2020] [Accepted: 03/26/2020] [Indexed: 02/07/2023] Open
Abstract
Recent cancer treatment modalities have been intensively focused on immunotherapy. The success of chimeric antigen receptor T cell therapy for treatment of refractory B cell acute lymphoblastic leukemia has pushed forward research on hematological malignancies. Among the effector types of innate lymphocytes, natural killer (NK) cells show great importance in immune surveillance against infectious and tumor diseases. Particularly, the role of NK cells has been argued in either elimination of target tumor cells or escape of tumor cells from immune surveillance. Therefore, an NK cell activation approach has been explored. Recent findings demonstrate that invariant natural killer T (iNKT) cells capable of producing IFN-γ when optimally activated can promptly trigger NK cells. Here, we review the role of NKT and/or NK cells and their interaction in anti-tumor responses by highlighting how innate immune cells recognize tumors, exert effector functions, and amplify adaptive immune responses. In addition, we discuss these innate lymphocytes in hematological disorders, particularly multiple myeloma and acute myeloid leukemia. The immune balance at different stages of both diseases is explored in light of disease progression. Various types of innate immunity-mediated therapeutic approaches, recent advances in clinical immunotherapies, and iNKT-mediated cancer immunotherapy as next-generation immunotherapy are then discussed.
Collapse
Affiliation(s)
- Kanako Shimizu
- Laboratory for Immunotherapy, RIKEN Center for Integrative Medical Sciences, 1-7-22, Suehiro-cho, Tsurumi-ku, Yokohama, Kanagawa 230-0045, Japan; (T.I.); (S.Y.)
- Correspondence: (K.S.); (S.-i.F.); Tel.: +81-45-503-7062 (K.S. & S.-i.F.); Fax: +81-45-503-7061 (K.S. & S.-i.F.)
| | - Tomonori Iyoda
- Laboratory for Immunotherapy, RIKEN Center for Integrative Medical Sciences, 1-7-22, Suehiro-cho, Tsurumi-ku, Yokohama, Kanagawa 230-0045, Japan; (T.I.); (S.Y.)
| | - Satoru Yamasaki
- Laboratory for Immunotherapy, RIKEN Center for Integrative Medical Sciences, 1-7-22, Suehiro-cho, Tsurumi-ku, Yokohama, Kanagawa 230-0045, Japan; (T.I.); (S.Y.)
| | - Norimitsu Kadowaki
- Department of Internal Medicine, Hematology, Rheumatology and Respiratory Medicine, Faculty of Medicine, Kagawa University, 1750-1 Ikenobe, Miki-cho, Kita-gun, Kagawa 761-0793, Japan;
| | - Arinobu Tojo
- Department of Hematology/Oncology, The Institute of Medical Science, The University of Tokyo, Minato, Tokyo 108-8639, Japan;
| | - Shin-ichiro Fujii
- Laboratory for Immunotherapy, RIKEN Center for Integrative Medical Sciences, 1-7-22, Suehiro-cho, Tsurumi-ku, Yokohama, Kanagawa 230-0045, Japan; (T.I.); (S.Y.)
- Correspondence: (K.S.); (S.-i.F.); Tel.: +81-45-503-7062 (K.S. & S.-i.F.); Fax: +81-45-503-7061 (K.S. & S.-i.F.)
| |
Collapse
|
18
|
Robinson JL, Kocabaş P, Wang H, Cholley PE, Cook D, Nilsson A, Anton M, Ferreira R, Domenzain I, Billa V, Limeta A, Hedin A, Gustafsson J, Kerkhoven EJ, Svensson LT, Palsson BO, Mardinoglu A, Hansson L, Uhlén M, Nielsen J. An atlas of human metabolism. Sci Signal 2020; 13:13/624/eaaz1482. [PMID: 32209698 DOI: 10.1126/scisignal.aaz1482] [Citation(s) in RCA: 217] [Impact Index Per Article: 43.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Genome-scale metabolic models (GEMs) are valuable tools to study metabolism and provide a scaffold for the integrative analysis of omics data. Researchers have developed increasingly comprehensive human GEMs, but the disconnect among different model sources and versions impedes further progress. We therefore integrated and extensively curated the most recent human metabolic models to construct a consensus GEM, Human1. We demonstrated the versatility of Human1 through the generation and analysis of cell- and tissue-specific models using transcriptomic, proteomic, and kinetic data. We also present an accompanying web portal, Metabolic Atlas (https://www.metabolicatlas.org/), which facilitates further exploration and visualization of Human1 content. Human1 was created using a version-controlled, open-source model development framework to enable community-driven curation and refinement. This framework allows Human1 to be an evolving shared resource for future studies of human health and disease.
Collapse
Affiliation(s)
- Jonathan L Robinson
- Department of Biology and Biological Engineering, Chalmers University of Technology, Kemivägen 10, SE-41258 Gothenburg, Sweden.,Wallenberg Center for Protein Research, Chalmers University of Technology, Kemivägen 10, SE-41258 Gothenburg, Sweden
| | - Pınar Kocabaş
- Department of Biology and Biological Engineering, Chalmers University of Technology, Kemivägen 10, SE-41258 Gothenburg, Sweden.,Wallenberg Center for Protein Research, Chalmers University of Technology, Kemivägen 10, SE-41258 Gothenburg, Sweden
| | - Hao Wang
- Department of Biology and Biological Engineering, Chalmers University of Technology, Kemivägen 10, SE-41258 Gothenburg, Sweden.,Wallenberg Center for Molecular and Translational Medicine, University of Gothenburg, Kemivägen 10, SE-41258 Gothenburg, Sweden.,Department of Biology and Biological Engineering, National Bioinformatics Infrastructure Sweden, Science for Life Laboratory, Chalmers University of Technology, Kemivägen 10, SE-41258 Gothenburg, Sweden
| | - Pierre-Etienne Cholley
- Department of Biology and Biological Engineering, National Bioinformatics Infrastructure Sweden, Science for Life Laboratory, Chalmers University of Technology, Kemivägen 10, SE-41258 Gothenburg, Sweden
| | - Daniel Cook
- Department of Biology and Biological Engineering, Chalmers University of Technology, Kemivägen 10, SE-41258 Gothenburg, Sweden
| | - Avlant Nilsson
- Department of Biology and Biological Engineering, Chalmers University of Technology, Kemivägen 10, SE-41258 Gothenburg, Sweden
| | - Mihail Anton
- Department of Biology and Biological Engineering, National Bioinformatics Infrastructure Sweden, Science for Life Laboratory, Chalmers University of Technology, Kemivägen 10, SE-41258 Gothenburg, Sweden
| | - Raphael Ferreira
- Department of Biology and Biological Engineering, Chalmers University of Technology, Kemivägen 10, SE-41258 Gothenburg, Sweden
| | - Iván Domenzain
- Department of Biology and Biological Engineering, Chalmers University of Technology, Kemivägen 10, SE-41258 Gothenburg, Sweden.,Wallenberg Center for Protein Research, Chalmers University of Technology, Kemivägen 10, SE-41258 Gothenburg, Sweden
| | - Virinchi Billa
- Department of Biology and Biological Engineering, Chalmers University of Technology, Kemivägen 10, SE-41258 Gothenburg, Sweden
| | - Angelo Limeta
- Department of Biology and Biological Engineering, Chalmers University of Technology, Kemivägen 10, SE-41258 Gothenburg, Sweden
| | - Alex Hedin
- Department of Biology and Biological Engineering, Chalmers University of Technology, Kemivägen 10, SE-41258 Gothenburg, Sweden
| | - Johan Gustafsson
- Department of Biology and Biological Engineering, Chalmers University of Technology, Kemivägen 10, SE-41258 Gothenburg, Sweden.,Wallenberg Center for Protein Research, Chalmers University of Technology, Kemivägen 10, SE-41258 Gothenburg, Sweden
| | - Eduard J Kerkhoven
- Department of Biology and Biological Engineering, Chalmers University of Technology, Kemivägen 10, SE-41258 Gothenburg, Sweden
| | - L Thomas Svensson
- Department of Biology and Biological Engineering, National Bioinformatics Infrastructure Sweden, Science for Life Laboratory, Chalmers University of Technology, Kemivägen 10, SE-41258 Gothenburg, Sweden
| | - Bernhard O Palsson
- Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark, DK-2800 Kgs. Lyngby, Denmark.,Department of Bioengineering, University of California, San Diego, La Jolla, CA 92093, USA.,Department of Pediatrics, University of California, San Diego, La Jolla, CA 92093, USA
| | - Adil Mardinoglu
- Department of Protein Science, Science for Life Laboratory, KTH-Royal Institute of Technology, SE-10044 Stockholm, Sweden.,Centre for Host-Microbiome Interactions, Faculty of Dentistry, Oral & Craniofacial Sciences, King's College London, London WC2R 2LS, UK
| | - Lena Hansson
- Department of Biology and Biological Engineering, National Bioinformatics Infrastructure Sweden, Science for Life Laboratory, Chalmers University of Technology, Kemivägen 10, SE-41258 Gothenburg, Sweden.,Novo Nordisk Research Centre Oxford, Oxford OX3 7FZ, UK
| | - Mathias Uhlén
- Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark, DK-2800 Kgs. Lyngby, Denmark.,Department of Protein Science, Science for Life Laboratory, KTH-Royal Institute of Technology, SE-10044 Stockholm, Sweden.,Wallenberg Center for Protein Research, KTH-Royal Institute of Technology, SE-10044 Stockholm, Sweden
| | - Jens Nielsen
- Department of Biology and Biological Engineering, Chalmers University of Technology, Kemivägen 10, SE-41258 Gothenburg, Sweden. .,Wallenberg Center for Protein Research, Chalmers University of Technology, Kemivägen 10, SE-41258 Gothenburg, Sweden.,Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark, DK-2800 Kgs. Lyngby, Denmark.,BioInnovation Institute, Ole Maaløes Vej 3, DK-2200 Copenhagen, Denmark
| |
Collapse
|
19
|
Soliman C, Chua JX, Vankemmelbeke M, McIntosh RS, Guy AJ, Spendlove I, Durrant LG, Ramsland PA. The terminal sialic acid of stage-specific embryonic antigen-4 has a crucial role in binding to a cancer-targeting antibody. J Biol Chem 2020. [DOI: 10.1016/s0021-9258(17)49911-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
|
20
|
Soliman C, Chua JX, Vankemmelbeke M, McIntosh RS, Guy AJ, Spendlove I, Durrant LG, Ramsland PA. The terminal sialic acid of stage-specific embryonic antigen-4 has a crucial role in binding to a cancer-targeting antibody. J Biol Chem 2019; 295:1009-1020. [PMID: 31831622 DOI: 10.1074/jbc.ra119.011518] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2019] [Revised: 12/05/2019] [Indexed: 01/06/2023] Open
Abstract
Cancer remains a leading cause of morbidity and mortality worldwide, requiring ongoing development of targeted therapeutics such as monoclonal antibodies. Carbohydrates on embryonic cells are often highly expressed in cancer and are therefore attractive targets for antibodies. Stage-specific embryonic antigen-4 (SSEA-4) is one such glycolipid target expressed in many cancers, including breast and ovarian carcinomas. Here, we defined the structural basis for recognition of SSEA-4 by a novel monospecific chimeric antibody (ch28/11). Five X-ray structures of ch28/11 Fab complexes with the SSEA-4 glycan headgroup, determined at 1.5-2.7 Å resolutions, displayed highly similar three-dimensional structures indicating a stable binding mode. The structures also revealed that by adopting a horseshoe-shaped conformation in a deep groove, the glycan headgroup likely sits flat against the membrane to allow the antibody to interact with SSEA-4 on cancer cells. Moreover, we found that the terminal sialic acid of SSEA-4 plays a dominant role in dictating the exquisite specificity of the ch28/11 antibody. This observation was further supported by molecular dynamics simulations of the ch28/11-glycan complex, which show that SSEA-4 is stabilized by its terminal sialic acid, unlike SSEA-3, which lacks this sialic acid modification. These high-resolution views of how a glycolipid interacts with an antibody may help to advance a new class of cancer-targeting immunotherapy.
Collapse
Affiliation(s)
- Caroline Soliman
- School of Science, RMIT University, Melbourne, Victoria 3083, Australia
| | - Jia Xin Chua
- Academic Department of Clinical Oncology, Division of Cancer and Stem Cells, School of Medicine, University of Nottingham, City Hospital Campus, Nottingham NG7 2RD, United Kingdom.,Scancell Ltd., Academic Department of Clinical Oncology, University of Nottingham, City Hospital Campus, Nottingham NG7 2RD, United Kingdom
| | - Mireille Vankemmelbeke
- Academic Department of Clinical Oncology, Division of Cancer and Stem Cells, School of Medicine, University of Nottingham, City Hospital Campus, Nottingham NG7 2RD, United Kingdom.,Scancell Ltd., Academic Department of Clinical Oncology, University of Nottingham, City Hospital Campus, Nottingham NG7 2RD, United Kingdom
| | - Richard S McIntosh
- Academic Department of Clinical Oncology, Division of Cancer and Stem Cells, School of Medicine, University of Nottingham, City Hospital Campus, Nottingham NG7 2RD, United Kingdom
| | - Andrew J Guy
- School of Science, RMIT University, Melbourne, Victoria 3083, Australia
| | - Ian Spendlove
- Academic Department of Clinical Oncology, Division of Cancer and Stem Cells, School of Medicine, University of Nottingham, City Hospital Campus, Nottingham NG7 2RD, United Kingdom
| | - Lindy G Durrant
- Academic Department of Clinical Oncology, Division of Cancer and Stem Cells, School of Medicine, University of Nottingham, City Hospital Campus, Nottingham NG7 2RD, United Kingdom.,Scancell Ltd., Academic Department of Clinical Oncology, University of Nottingham, City Hospital Campus, Nottingham NG7 2RD, United Kingdom
| | - Paul A Ramsland
- School of Science, RMIT University, Melbourne, Victoria 3083, Australia .,Department of Immunology, Central Clinical School, Monash University, Melbourne, Victoria 3800, Australia.,Department of Surgery Austin Health, University of Melbourne, Heidelberg, Victoria 3084, Australia
| |
Collapse
|
21
|
Chen X, Jin L, Jiang X, Guo L, Gu G, Xu L, Lu L, Wang F, Xiao M. Converting a β-N-acetylhexosaminidase into two trans-β-N-acetylhexosaminidases by domain-targeted mutagenesis. Appl Microbiol Biotechnol 2019; 104:661-673. [PMID: 31822984 DOI: 10.1007/s00253-019-10253-y] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2019] [Revised: 10/29/2019] [Accepted: 11/12/2019] [Indexed: 01/14/2023]
Abstract
We have recently derived a β-N-acetylhexosaminidase, BbhI, from Bifidobacterium bifidum JCM 1254, which could regioselectively synthesize GlcNAcβ1-3Galβ1-4Glc with a yield of 44.9%. Here, directed evolution of BbhI by domain-targeted mutagenesis was carried out. Firstly, the GH20 domain was selected for random mutagenesis using MEGAWHOP method and a small library of 1300 clones was created. A total of 734 colonies with reduced hydrolytic activity were isolated, and three mutants with elevated transglycosylation yields, GlcNAcβ1-3Galβ1-4Glc yields of 68.5%, 74.7%, and 81.1%, respectively, were obtained. Subsequently, nineteen independent mutants were constructed according to all the mutation sites in these three mutants. After transglycosylation analysis, Asp714 and Trp773 were identified as key residues for improvement in transglycosylation ability and were chosen for the second round of directed evolution by site-saturation mutagenesis. Two most efficient mutants D714T and W773R that acted as trans-β-N-acetylhexosaminidase were finally achieved. D714T with the substitution at the putative nucleophile assistant residue Asp714 by threonine showed high yield of 84.7% with unobserved hydrolysis towards transglycosylation product. W773R with arginine substitution at Trp773 residue locating at the entrance of catalytic cavity led to the yield up to 81.8%. The kcat/Km values of D714T and W773R for hydrolysis of pNP-β-GlcNAc displayed drastic decreases. NMR investigation of protein-substrate interaction revealed an invariable mode of the catalytic cavity of D714T, W773R, and WT BbhI. The collective motions of protein model showed the mutations Thr714 and Arg773 exerted little effect on the dynamics of the inside but a large effect on the dynamics of the outside of catalytic cavity.
Collapse
Affiliation(s)
- Xiaodi Chen
- State Key Lab of Microbial Technology, National Glycoengineering Research Center, Shandong Provincial Key Laboratory of Carbohydrate Chemistry and Glycobiology, Shandong University, Qingdao, 266237, People's Republic of China.,School of Pharmaceutical Sciences, Shandong University, Jinan, 250012, People's Republic of China
| | - Lan Jin
- State Key Lab of Microbial Technology, National Glycoengineering Research Center, Shandong Provincial Key Laboratory of Carbohydrate Chemistry and Glycobiology, Shandong University, Qingdao, 266237, People's Republic of China
| | - Xukai Jiang
- State Key Lab of Microbial Technology, National Glycoengineering Research Center, Shandong Provincial Key Laboratory of Carbohydrate Chemistry and Glycobiology, Shandong University, Qingdao, 266237, People's Republic of China
| | - Longcheng Guo
- State Key Lab of Microbial Technology, National Glycoengineering Research Center, Shandong Provincial Key Laboratory of Carbohydrate Chemistry and Glycobiology, Shandong University, Qingdao, 266237, People's Republic of China
| | - Guofeng Gu
- State Key Lab of Microbial Technology, National Glycoengineering Research Center, Shandong Provincial Key Laboratory of Carbohydrate Chemistry and Glycobiology, Shandong University, Qingdao, 266237, People's Republic of China
| | - Li Xu
- State Key Lab of Microbial Technology, National Glycoengineering Research Center, Shandong Provincial Key Laboratory of Carbohydrate Chemistry and Glycobiology, Shandong University, Qingdao, 266237, People's Republic of China
| | - Lili Lu
- School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, People's Republic of China
| | - Fengshan Wang
- School of Pharmaceutical Sciences, Shandong University, Jinan, 250012, People's Republic of China
| | - Min Xiao
- State Key Lab of Microbial Technology, National Glycoengineering Research Center, Shandong Provincial Key Laboratory of Carbohydrate Chemistry and Glycobiology, Shandong University, Qingdao, 266237, People's Republic of China.
| |
Collapse
|
22
|
Russo D, Capolupo L, Loomba JS, Sticco L, D'Angelo G. Glycosphingolipid metabolism in cell fate specification. J Cell Sci 2018; 131:131/24/jcs219204. [PMID: 30559216 DOI: 10.1242/jcs.219204] [Citation(s) in RCA: 60] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Glycosphingolipids (GSLs) are ubiquitous components of eukaryotic plasma membranes that consist of a ceramide backbone linked to a glycan moiety. Both the ceramide and the glycan parts of GSLs display structural variations that result in a remarkable repertoire of diverse compounds. This diversity of GSLs is exploited during embryogenesis, when different GSLs are produced at specific developmental stages and along several differentiation trajectories. Importantly, plasma membrane receptors interact with GSLs to modify their activities. Consequently, two otherwise identical cells can respond differently to the same stimulus owing to their different GSL composition. The metabolic reprograming of GSLs is in fact a necessary part of developmental programs, as its impairment results in developmental failure or tissue-specific defects. Moreover, single-cell variability is emerging as a fundamental player in development: GSL composition displays cell-to-cell variability in syngeneic cell populations owing to the regulatory gene expression circuits involved in microenvironment adaptation and in differentiation. Here, we discuss how GSLs are synthesized and classified and review the role of GSLs in the establishment and maintenance of cell identity. We further highlight the existence of the regulatory circuits that modify GSL pathways and speculate how GSL heterogeneity might contribute to developmental patterning.
Collapse
Affiliation(s)
- Domenico Russo
- Institute of Protein Biochemistry, National Research Council, Via P. Castellino 111, Napoli, Italy
| | - Laura Capolupo
- Institute of Protein Biochemistry, National Research Council, Via P. Castellino 111, Napoli, Italy.,Institute of Bioengineering, Laboratory of Lipid Cell Biology, École polytechnique fédérale de Lausanne (EPFL) CH-1015 Lausanne, Switzerland
| | - Jaipreet Singh Loomba
- Institute of Protein Biochemistry, National Research Council, Via P. Castellino 111, Napoli, Italy.,Institute of Bioengineering, Laboratory of Lipid Cell Biology, École polytechnique fédérale de Lausanne (EPFL) CH-1015 Lausanne, Switzerland
| | - Lucia Sticco
- Institute of Protein Biochemistry, National Research Council, Via P. Castellino 111, Napoli, Italy
| | - Giovanni D'Angelo
- Institute of Protein Biochemistry, National Research Council, Via P. Castellino 111, Napoli, Italy .,Institute of Bioengineering, Laboratory of Lipid Cell Biology, École polytechnique fédérale de Lausanne (EPFL) CH-1015 Lausanne, Switzerland
| |
Collapse
|
23
|
Plasma Gelsolin: Indicator of Inflammation and Its Potential as a Diagnostic Tool and Therapeutic Target. Int J Mol Sci 2018; 19:ijms19092516. [PMID: 30149613 PMCID: PMC6164782 DOI: 10.3390/ijms19092516] [Citation(s) in RCA: 91] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2018] [Revised: 08/14/2018] [Accepted: 08/18/2018] [Indexed: 12/19/2022] Open
Abstract
Gelsolin, an actin-depolymerizing protein expressed both in extracellular fluids and in the cytoplasm of a majority of human cells, has been recently implicated in a variety of both physiological and pathological processes. Its extracellular isoform, called plasma gelsolin (pGSN), is present in blood, cerebrospinal fluid, milk, urine, and other extracellular fluids. This isoform has been recognized as a potential biomarker of inflammatory-associated medical conditions, allowing for the prediction of illness severity, recovery, efficacy of treatment, and clinical outcome. A compelling number of animal studies also demonstrate a broad spectrum of beneficial effects mediated by gelsolin, suggesting therapeutic utility for extracellular recombinant gelsolin. In the review, we summarize the current data related to the potential of pGSN as an inflammatory predictor and therapeutic target, discuss gelsolin-mediated mechanisms of action, and highlight recent progress in the clinical use of pGSN.
Collapse
|
24
|
Zhuo D, Li X, Guan F. Biological Roles of Aberrantly Expressed Glycosphingolipids and Related Enzymes in Human Cancer Development and Progression. Front Physiol 2018; 9:466. [PMID: 29773994 PMCID: PMC5943571 DOI: 10.3389/fphys.2018.00466] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2017] [Accepted: 04/13/2018] [Indexed: 01/05/2023] Open
Abstract
Glycosphingolipids (GSLs), which consist of a hydrophobic ceramide backbone and a hydrophilic carbohydrate residue, are an important type of glycolipid expressed in surface membranes of all animal cells. GSLs play essential roles in maintenance of plasma membrane stability, in regulation of numerous cellular processes (including adhesion, proliferation, apoptosis, and recognition), and in modulation of signal transduction pathways. GSLs have traditionally been classified as ganglio-series, lacto-series, or globo-series on the basis of their diverse types of oligosaccharide chains. Structures and functions of specific GSLs are also determined by their oligosaccharide chains. Different cells and tissues show differential expression of GSLs, and changes in structures of GSL glycan moieties occur during development of numerous types of human cancer. Association of GSLs and/or related enzymes with initiation and progression of cancer has been documented in 100s of studies, and many such GSLs are useful markers or targets for cancer diagnosis or therapy. In this review, we summarize (i) recent studies on aberrant expression and distribution of GSLs in common human cancers (breast, lung, colorectal, melanoma, prostate, ovarian, leukemia, renal, bladder, gastric); (ii) biological functions of specific GSLs in these cancers.
Collapse
Affiliation(s)
- Dinghao Zhuo
- Key Laboratory of Carbohydrate Chemistry and Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi, China
| | - Xiang Li
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of China, College of Life Science, Northwest University, Xi'an, China
| | - Feng Guan
- Key Laboratory of Carbohydrate Chemistry and Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi, China
| |
Collapse
|
25
|
Wątek M, Durnaś B, Wollny T, Pasiarski M, Góźdź S, Marzec M, Chabowska A, Wolak P, Żendzian-Piotrowska M, Bucki R. Unexpected profile of sphingolipid contents in blood and bone marrow plasma collected from patients diagnosed with acute myeloid leukemia. Lipids Health Dis 2017; 16:235. [PMID: 29216917 PMCID: PMC5721620 DOI: 10.1186/s12944-017-0624-1] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2017] [Accepted: 11/27/2017] [Indexed: 01/21/2023] Open
Abstract
Background Impaired apoptotic pathways in leukemic cells enable them to grow in an uncontrolled way. Moreover, aberrations in the apoptotic pathways are the main factor of leukemic cells drug resistance. Methods To assess the presence of potential abnormalities that might promote dysfunction of leukemic cells growth, HPLC system was used to determine sphingosine (SFO), sphinganine (SFA), sphingosine-1-phosphate (S1P) and ceramide (CER) concentration in the blood collected from patients diagnose with acute myeloblastic leukemia (AML; n = 49) and compare to values of control (healthily) group (n = 51). Additionally, in AML group concentration of SFO, SFA, S1P and CER was determined in bone marrow plasma and compared to respective values in blood plasma. The concentration of S1P and CER binding protein – plasma gelsolin (GSN) was also assessed in collected samples using immunoblotting assay. Results We observed that in AML patients the average SFO, SFA and CER concentration in blood plasma was significantly higher (p < 0.001) compare to control group, when blood plasma S1P concentration was significantly lower (p < 0.001). At the same time the CER/S1P ratio in AML patient (44.5 ± 19.4) was about 54% higher compare to control group (20.9 ± 13.1). Interestingly the average concentration of S1P in blood plasma (196 ± 13 pmol/ml) was higher compare to its concentration in plasma collected from bone marrow (154 ± 21 pmol/ml). Conclusions We hypothesize that changes in profile of sphingolipids concentration and some of their binding protein partners such as GSN in extracellular environment of blood and bone marrow cells in leukemic patients can be targeted to develop new AML treatment method(s).
Collapse
Affiliation(s)
- Marzena Wątek
- Department of Hematology, Holy Cross Oncology Center of Kielce, Artwińskiego 3, 25-734, Kielce, Poland
| | - Bonita Durnaś
- Faculty of Medicine and Health Sciences of the Jan Kochanowski University in Kielce, Kielce, Poland
| | - Tomasz Wollny
- Department of Hematology, Holy Cross Oncology Center of Kielce, Artwińskiego 3, 25-734, Kielce, Poland
| | - Marcin Pasiarski
- Department of Hematology, Holy Cross Oncology Center of Kielce, Artwińskiego 3, 25-734, Kielce, Poland.,Faculty of Medicine and Health Sciences of the Jan Kochanowski University in Kielce, Kielce, Poland
| | - Stanisław Góźdź
- Department of Hematology, Holy Cross Oncology Center of Kielce, Artwińskiego 3, 25-734, Kielce, Poland.,Faculty of Medicine and Health Sciences of the Jan Kochanowski University in Kielce, Kielce, Poland
| | - Michał Marzec
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Anna Chabowska
- Regional Blood Transfusion Center in Bialystok, 15-950, Bialystok, Poland
| | - Przemysław Wolak
- Faculty of Medicine and Health Sciences of the Jan Kochanowski University in Kielce, Kielce, Poland
| | - Małgorzata Żendzian-Piotrowska
- Department of Hygiene, Epidemiology and Ergonomics Department Medical University of Bialystok, 15-222, Bialystok, Poland
| | - Robert Bucki
- Faculty of Medicine and Health Sciences of the Jan Kochanowski University in Kielce, Kielce, Poland. .,Department of Microbiological and Nanobiomedical Engineering, Medical University of Bialystok, 15-222, Bialystok, Poland.
| |
Collapse
|
26
|
Wang Z, Wen L, Zhu F, Wang Y, Xie Q, Chen Z, Li Y. Overexpression of ceramide synthase 1 increases C18-ceramide and leads to lethal autophagy in human glioma. Oncotarget 2017; 8:104022-104036. [PMID: 29262618 PMCID: PMC5732784 DOI: 10.18632/oncotarget.21955] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2017] [Accepted: 09/25/2017] [Indexed: 01/25/2023] Open
Abstract
Ceramide synthase 1 (CERS1) is the most highly expressed CERS in the central nervous system, and ceramide with an 18-carbon-containing fatty acid chain (C18-ceramide) in the brain plays important roles in signaling and sphingolipid development. However, the roles of CERS1 and C18-ceramide in glioma are largely unknown. In the present study, measured by electrospray ionization linear ion trap mass spectrometry, C18-ceramide was significantly lower in glioma tumor tissues compared with controls (P < 0.001), indicating that C18-ceramide might have a role in glioma. These roles were examined by reconstitution of C18-ceramide in U251 and A172 glioma cells via addition of exogenous C18-ceramide or overexpression of CERS1, which has been shown to specifically induce the generation of C18-ceramide. Overexpression of CERS1 or adding exogenous C18-ceramide inhibited cell viability and induced cell death by activating endoplasmic reticulum stress, which induced lethal autophagy and inhibited PI3K/AKT signal pathway in U251 and A172 glioma cells. Moreover, overexpression of CERS1 or adding exogenous C18-ceramide increased the sensitivity of U251 and A172 glioma cells to teniposide (VM-26). Thus, the combined therapy of CERS1/C18-ceramide and VM-26 may be a novel therapeutic strategy for the treatment of human glioma.
Collapse
Affiliation(s)
- Zheng Wang
- Institutes of Biology and Medical Sciences, Medical College, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Soochow University, Suzhou, China
| | - Lijun Wen
- Institutes of Biology and Medical Sciences, Medical College, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Soochow University, Suzhou, China
| | - Fei Zhu
- Institutes of Biology and Medical Sciences, Medical College, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Soochow University, Suzhou, China
| | - Yanping Wang
- Institutes of Biology and Medical Sciences, Medical College, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Soochow University, Suzhou, China
| | - Qing Xie
- Institutes of Biology and Medical Sciences, Medical College, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Soochow University, Suzhou, China
| | - Zijun Chen
- Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yunsen Li
- Institutes of Biology and Medical Sciences, Medical College, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Soochow University, Suzhou, China
| |
Collapse
|
27
|
Altered (neo-) lacto series glycolipid biosynthesis impairs α2-6 sialylation on N-glycoproteins in ovarian cancer cells. Sci Rep 2017; 7:45367. [PMID: 28358117 PMCID: PMC5371825 DOI: 10.1038/srep45367] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2016] [Accepted: 02/15/2017] [Indexed: 12/16/2022] Open
Abstract
The (neo-) lacto series glycosphingolipids (nsGSLs) comprise of glycan epitopes that are present as blood group antigens, act as primary receptors for human pathogens and are also increasingly associated with malignant diseases. Beta-1, 3-N-acetyl-glucosaminyl-transferase 5 (B3GNT5) is suggested as the key glycosyltransferase for the biosynthesis of nsGSLs. In this study, we investigated the impact of CRISPR-Cas9 -mediated gene disruption of B3GNT5 (∆B3GNT5) on the expression of glycosphingolipids and N-glycoproteins by utilizing immunostaining and glycomics-based PGC-UHPLC-ESI-QTOF-MS/MS profiling. ∆B3GNT5 cells lost nsGSL expression coinciding with reduction of α2-6 sialylation on N-glycoproteins. In contrast, disruption of B4GALNT1, a glycosyltransferase for ganglio series GSLs did not affect α2-6 sialylation on N-glycoproteins. We further profiled all known
α2-6 sialyltransferase-encoding genes and showed that the loss of α2-6 sialylation is due to silencing of ST6GAL1 expression in ∆B3GNT5 cells. These results demonstrate that nsGSLs are part of a complex network affecting N-glycosylation in ovarian cancer cells.
Collapse
|
28
|
Efficient and Regioselective Synthesis of β-GalNAc/GlcNAc-Lactose by a Bifunctional Transglycosylating β-N-Acetylhexosaminidase from Bifidobacterium bifidum. Appl Environ Microbiol 2016; 82:5642-52. [PMID: 27422836 DOI: 10.1128/aem.01325-16] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2016] [Accepted: 06/29/2016] [Indexed: 01/08/2023] Open
Abstract
UNLABELLED β-N-Acetylhexosaminidases have attracted interest particularly for oligosaccharide synthesis, but their use remains limited by the rarity of enzyme sources , low efficiency, and relaxed regioselectivity of transglycosylation. In this work, genes of 13 β-N-acetylhexosaminidases, including 5 from Bacteroides fragilis ATCC 25285, 5 from Clostridium perfringens ATCC 13124, and 3 from Bifidobacterium bifidum JCM 1254, were cloned and heterogeneously expressed in Escherichia coli The resulting recombinant enzymes were purified and screened for transglycosylation activity. A β-N-acetylhexosaminidase named BbhI, which belongs to glycoside hydrolase family 20 and was obtained from B. bifidum JCM 1254, possesses the bifunctional property of efficiently transferring both GalNAc and GlcNAc residues through β1-3 linkage to the Gal residue of lactose. The effects of initial substrate concentration, pH, temperature, and reaction time on transglycosylation activities of BbhI were studied in detail. With the use of 10 mM pNP-β-GalNAc or 20 mM pNP-β-GlcNAc as the donor and 400 mM lactose as the acceptor in phosphate buffer (pH 5.8), BbhI synthesized GalNAcβ1-3Galβ1-4Glc and GlcNAcβ1-3Galβ1-4Glc at maximal yields of 55.4% at 45°C and 4 h and 44.9% at 55°C and 1.5 h, respectively. The model docking of BbhI with lactose showed the possible molecular basis of strict regioselectivity of β1-3 linkage in β-N-acetylhexosaminyl lactose synthesis. IMPORTANCE Oligosaccharides play a crucial role in many biological events and therefore are promising potential therapeutic agents. However, their use is limited because large-scale production of oligosaccharides is difficult. The chemical synthesis requires multiple protecting group manipulations to control the regio- and stereoselectivity of glycosidic bonds. In comparison, enzymatic synthesis can produce oligosaccharides in one step by using glycosyltransferases and glycosidases. Given the lower price of their glycosyl donor and their broader acceptor specificity, glycosidases are more advantageous than glycosyltransferases for large-scale synthesis. β-N-Acetylhexosaminidases have attracted interest particularly for β-N-acetylhexosaminyl oligosaccharide synthesis, but their application is affected by having few enzyme sources, low efficiency, and relaxed regioselectivity of transglycosylation. In this work, we describe a microbial β-N-acetylhexosaminidase that exhibited strong transglycosylation activity and strict regioselectivity for β-N-acetylhexosaminyl lactose synthesis and thus provides a powerful synthetic tool to obtain biologically important GalNAcβ1-3Lac and GlcNAcβ1-3Lac.
Collapse
|
29
|
Wang K, Wang J, Sun T, Bian G, Pan W, Feng T, Wang P, Li Y, Dai J. Glycosphingolipid GM3 is Indispensable for Dengue Virus Genome Replication. Int J Biol Sci 2016; 12:872-83. [PMID: 27313500 PMCID: PMC4910605 DOI: 10.7150/ijbs.15641] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2016] [Accepted: 05/03/2016] [Indexed: 01/08/2023] Open
Abstract
Dengue virus (DENV) causes the most prevalent arthropod-borne viral disease of humans worldwide. Glycosphingolipids (GSLs) are involved in virus infection by regulating various steps of viral-host interaction. However, the distinct role of GSLs during DENV infection remains unclear. In this study, we used mouse melanoma B16 cells and their GSL-deficient mutant counterpart GM95 cells to study the influence of GSLs on DENV infection. Surprisingly, GM95 cells were highly resistant to DENV infection compared with B16 cells. Pretreatment of B16 cells with synthetase inhibitor of GM3, the most abundant GSLs in B16 cells, or silencing GM3 synthetase T3GAL5, significantly inhibited DENV infection. DENV attachment and endocytosis were not impaired in GM95 cells, but DENV genome replication was obviously inhibited in GM95 cells compared to B16 cells. Furthermore, GM3 was colocalized with DENV viral replication complex on endoplasmic reticulum (ER) inside the B16 cells. Finally, GM3 synthetase inhibitor significantly reduced the mortality rate of suckling mice that challenged with DENV by impairing the viral replication in mouse brain. Taken together, these data indicated that GM3 was not required for DENV attachment and endocytosis, however, essential for viral genome replication. Targeting GM3 could be a novel strategy to inhibit DENV infection.
Collapse
Affiliation(s)
- Kezhen Wang
- 1. Institutes of Biology and Medical Sciences, Jiangsu Key Laboratory of Infection and Immunity, Soochow University, Suzhou 215123, P.R. China
| | - Juanjuan Wang
- 1. Institutes of Biology and Medical Sciences, Jiangsu Key Laboratory of Infection and Immunity, Soochow University, Suzhou 215123, P.R. China
| | - Ta Sun
- 1. Institutes of Biology and Medical Sciences, Jiangsu Key Laboratory of Infection and Immunity, Soochow University, Suzhou 215123, P.R. China
| | - Gang Bian
- 1. Institutes of Biology and Medical Sciences, Jiangsu Key Laboratory of Infection and Immunity, Soochow University, Suzhou 215123, P.R. China
| | - Wen Pan
- 1. Institutes of Biology and Medical Sciences, Jiangsu Key Laboratory of Infection and Immunity, Soochow University, Suzhou 215123, P.R. China
| | - Tingting Feng
- 1. Institutes of Biology and Medical Sciences, Jiangsu Key Laboratory of Infection and Immunity, Soochow University, Suzhou 215123, P.R. China
| | - Penghua Wang
- 2. Department of Microbiology and Immunology, New York Medical College, Valhalla, NY 10595, USA
| | - Yunsen Li
- 1. Institutes of Biology and Medical Sciences, Jiangsu Key Laboratory of Infection and Immunity, Soochow University, Suzhou 215123, P.R. China
| | - Jianfeng Dai
- 1. Institutes of Biology and Medical Sciences, Jiangsu Key Laboratory of Infection and Immunity, Soochow University, Suzhou 215123, P.R. China
| |
Collapse
|
30
|
Binnington B, Nguyen L, Kamani M, Hossain D, Marks DL, Budani M, Lingwood CA. Inhibition of Rab prenylation by statins induces cellular glycosphingolipid remodeling. Glycobiology 2016; 26:166-80. [PMID: 26405105 PMCID: PMC4691287 DOI: 10.1093/glycob/cwv084] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2015] [Revised: 09/11/2015] [Accepted: 09/14/2015] [Indexed: 12/17/2022] Open
Abstract
Statins, which specifically inhibit HMG Co-A reductase, the rate-limiting step of cholesterol biosynthesis, are widely prescribed to reduce serum cholesterol and cardiac risk, but many other effects are seen. We now show an effect of these drugs to induce profound changes in the step-wise synthesis of glycosphingolipids (GSLs) in the Golgi. Glucosylceramide (GlcCer) was increased several-fold in all cell lines tested, demonstrating a widespread effect. Additionally, de novo or elevated lactotriaosylceramide (Lc3Cer; GlcNAcβ1-3Galβ1-4GlcCer) synthesis was observed in 70%. Western blot showed that GlcCer synthase (GCS) was elevated by statins, and GCS and Lc3Cer synthase (Lc3S) activities were increased; however, transcript was elevated for Lc3S only. Supplementation with the isoprenoid precursor, geranylgeranyl pyrophosphate (GGPP), a downstream product of HMG Co-A reductase, reversed statin-induced glycosyltransferase and GSL elevation. The Rab geranylgeranyl transferase inhibitor 3-PEHPC, but not specific inhibitors of farnesyl transferase, or geranylgeranyl transferase I, was sufficient to replicate statin-induced GlcCer and Lc3Cer synthesis, supporting a Rab prenylation-dependent mechanism. While total cholesterol was unaffected, the trans-Golgi network (TGN) cholesterol pool was dissipated and medial Golgi GCS partially relocated by statins. GSL-dependent vesicular retrograde transport of Verotoxin and cholera toxin to the Golgi/endoplasmic reticulum were blocked after statin or 3-PEHPC treatment, suggesting aberrant, prenylation-dependent vesicular traffic as a basis of glycosyltransferase increase and GSL remodeling. These in vitro studies indicate a previously unreported link between Rab prenylation and regulation of GCS activity and GlcCer metabolism.
Collapse
Affiliation(s)
- Beth Binnington
- Research Institute, Program in Molecular Structure and Function, The Hospital for Sick Children, 686 Bay St., Toronto, ON M5G 1X8, Canada
| | - Long Nguyen
- Research Institute, Program in Molecular Structure and Function, The Hospital for Sick Children, 686 Bay St., Toronto, ON M5G 1X8, Canada
| | - Mustafa Kamani
- Research Institute, Program in Molecular Structure and Function, The Hospital for Sick Children, 686 Bay St., Toronto, ON M5G 1X8, Canada Department of Biochemistry
| | - Delowar Hossain
- Research Institute, Program in Molecular Structure and Function, The Hospital for Sick Children, 686 Bay St., Toronto, ON M5G 1X8, Canada
| | - David L Marks
- Schulze Center for Novel Therapeutics, Division of Oncology Research, Mayo Clinic, Rochester, MN, USA
| | - Monique Budani
- Research Institute, Program in Molecular Structure and Function, The Hospital for Sick Children, 686 Bay St., Toronto, ON M5G 1X8, Canada Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Clifford A Lingwood
- Research Institute, Program in Molecular Structure and Function, The Hospital for Sick Children, 686 Bay St., Toronto, ON M5G 1X8, Canada Department of Biochemistry Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON M5S 1A8, Canada
| |
Collapse
|
31
|
Fair RJ, Hahm HS, Seeberger PH. Combination of automated solid-phase and enzymatic oligosaccharide synthesis provides access to α(2,3)-sialylated glycans. Chem Commun (Camb) 2015; 51:6183-5. [PMID: 25754251 DOI: 10.1039/c5cc01368b] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
A synthetic strategy combining automated solid-phase chemical synthesis and enzymatic sialylation was developed to access α(2,3)-sialylated glycans.
Collapse
Affiliation(s)
- Richard J Fair
- Department of Biomolecular Systems, Max-Planck-Institute of Colloids and Interfaces, Am Mühlenberg 1, 14476 Potsdam, Germany.
| | | | | |
Collapse
|
32
|
Wang R, Fang J, Ma H, Feng L, Lian M, Yang F, Wang H, Wang Q, Chen X. Effect of microRNA-203 on tumor growth in human hypopharyngeal squamous cell carcinoma. Mol Cell Biochem 2015; 405:97-104. [PMID: 25840888 DOI: 10.1007/s11010-015-2401-7] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2014] [Accepted: 03/27/2015] [Indexed: 01/27/2023]
Abstract
MicroRNAs (MiRNAs) have been recognized to regulate cancer initiation and progression in carcinogenesis as either oncogenes or tumor suppressor genes, but their role in hypopharyngeal cancer development is not clearly defined. To determine whether miRNA-203 can promote tumor growth in human hypopharyngeal squamous cell carcinoma, we conducted experiments on the functional study of miRNA-203 and identification of miRNA-203 regulated target genes in hypopharyngeal cancer cells. We found that cell proliferation and cell colony-forming increased more in the miRNA-203 up-regulated cancer cells than in the negative control cancer cells. Up-regulation of miRNA-203 accelerated cell cycle progression in hypopharyngeal cancer cells. TP63 and B3GNT5 mRNAs were identified and validated as targets of miRNA-203. However, transwell assay and wound scratch assay showed that miRNA-203 did not involve in invasion and metastasis in hypopharyngeal cancer cells. According to the results, we conclude that miRNA-203 can promote tumor growth in human hypopharyngeal squamous cell carcinoma. These results provide the convincing evidence for the first time that up-regulation of miRNA-203 contributes to the malignancy of hypopharyngeal squamous cell carcinoma, possibly through down-regulating TP63 and B3GNT5.
Collapse
Affiliation(s)
- Ru Wang
- Department of Otorhinolaryngology Head and Neck Surgery, Beijing Tongren Hospital, Capital Medical University, Beijing, 100730, China
| | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Zhu T, Xu L, Xu X, Wang Z, Zhu J, Xie Q, Zhang B, Wang Y, Ju L, He Y, Ye X, Zhou D, Li Y. Analysis of breast cancer-associated glycosphingolipids using electrospray ionization-linear ion trap quadrupole mass spectrometry. Carbohydr Res 2015; 402:189-99. [DOI: 10.1016/j.carres.2014.10.006] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2014] [Revised: 09/27/2014] [Accepted: 10/14/2014] [Indexed: 10/24/2022]
|
34
|
Xu X, Yu Y, Wang Z, Zhu T, Wang Y, Zhu J, Chen Z, He Y, Ju L, Li Y. Mass spectrometry based phospholipidomics of mammalian thymus and leukemia patients: implication for function of iNKT cells. Anal Bioanal Chem 2013; 405:5267-78. [PMID: 23595640 DOI: 10.1007/s00216-013-6923-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2012] [Revised: 03/06/2013] [Accepted: 03/15/2013] [Indexed: 10/27/2022]
Abstract
In previous studies phospholipids have been proved to be involved in biochemical, physiological, and pathological processes. As a special class of phospholipids, peroxisome-derived lipids (PDLs) have been proved to be potential ligands of invariant natural killer T (iNKT) cells in recent studies. Here, on the basis of phospholipidomics, we focused on the relative quantity of PDLs extracted from mammalian thymus or bone marrow using electrospray ionization mass spectrometry (MS). In phospholipid analysis, we identified 12 classes of phospholipids and accounted for their relative quantities by comparing their relative abundances in the MS(1) map. Our results show that PDLs are present in mammalian thymus as well as mouse spleen and liver. Interestingly, the relative quantity of PDLs extracted from human acute myeloid leukemia (AML) and acute lymphoblastic leukemia (ALL) bone marrows is higher than that extracted from bone marrow of healthy donors. Our results may help to explain the close correlation between PDLs and iNKT cell function in thymus, spleen, liver, and especially in leukemia patients. We think that our phospholipidomics work may reveal a function of iNKT cells.
Collapse
Affiliation(s)
- Xiukun Xu
- Laboratory of Cellular and Molecular Tumor Immunology, Institutes of Biology and Medical Sciences, Jiangsu Laboratory of Infection Immunity, Soochow University, 199 Ren-Ai Road, Suzhou 215123, China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Ersek A, Karadimitris A, Horwood NJ. Effect of glycosphingolipids on osteoclastogenesis and osteolytic bone diseases. Front Endocrinol (Lausanne) 2012; 3:106. [PMID: 22936926 PMCID: PMC3425772 DOI: 10.3389/fendo.2012.00106] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/09/2012] [Accepted: 08/12/2012] [Indexed: 12/21/2022] Open
Abstract
Alterations in glycosphingolipid (GSL) production results in lysosomal storage disorders associated with neurodegenerative changes. In Gaucher's disease, the patients also develop osteoporosis that is ameliorated upon treatment for the underlying defect in GSL metabolism. The role of GSLs in osteoclast and osteoblast formation is discussed here as well as the potential therapeutic uses of already approved drugs that limit GSL production in bone loss disorders such as multiple myeloma and periodontal disease.
Collapse
Affiliation(s)
- Adel Ersek
- Kennedy Institute of Rheumatology, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of OxfordLondon, UK
- Centre for Haematology, Department of Medicine, Imperial College London, Hammersmith HospitalLondon, UK
| | - Anastasios Karadimitris
- Centre for Haematology, Department of Medicine, Imperial College London, Hammersmith HospitalLondon, UK
| | - Nicole J. Horwood
- Kennedy Institute of Rheumatology, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of OxfordLondon, UK
- *Correspondence: Nicole J. Horwood, Kennedy Institute of Rheumatology, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, 65 Aspenlea Road, London W6 8LH, UK. e-mail:
| |
Collapse
|