1
|
Khorami-Sarvestani S, Hanash SM, Fahrmann JF, León-Letelier RA, Katayama H. Glycosylation in cancer as a source of biomarkers. Expert Rev Proteomics 2024:1-21. [PMID: 39376081 DOI: 10.1080/14789450.2024.2409224] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Revised: 08/12/2024] [Accepted: 09/17/2024] [Indexed: 10/09/2024]
Abstract
INTRODUCTION Glycosylation, the process of glycan synthesis and attachment to target molecules, is a crucial and common post-translational modification (PTM) in mammalian cells. It affects the protein's hydrophilicity, charge, solubility, structure, localization, function, and protection from proteolysis. Aberrant glycosylation in proteins can reveal new detection and therapeutic Glyco-biomarkers, which help to improve accurate early diagnosis and personalized treatment. This review underscores the pivotal role of glycans and glycoproteins as a source of biomarkers in human diseases, particularly cancer. AREAS COVERED This review delves into the implications of glycosylation, shedding light on its intricate roles in cancer-related cellular processes influencing biomarkers. It is underpinned by a thorough examination of literature up to June 2024 in PubMed, Scopus, and Google Scholar; concentrating on the terms: (Glycosylation[Title/Abstract]) OR (Glycan[Title/Abstract]) OR (glycoproteomics[Title/Abstract]) OR (Proteoglycans[Title/Abstract]) OR (Glycomarkers[Title/Abstract]) AND (Cancer[Title/Abstract]) AND ((Diagno*[Title/Abstract]) OR (Progno*[Title/Abstract])). EXPERT OPINION Glyco-biomarkers enhance early cancer detection, allow early intervention, and improve patient prognoses. However, the abundance and complex dynamic glycan structure may make their scientific and clinical application difficult. This exploration of glycosylation signatures in cancer biomarkers can provide a detailed view of cancer etiology and instill hope in the potential of glycosylation to revolutionize cancer research.
Collapse
Affiliation(s)
- Sara Khorami-Sarvestani
- Department of Clinical Cancer Prevention, the University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Samir M Hanash
- Department of Clinical Cancer Prevention, the University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Johannes F Fahrmann
- Department of Clinical Cancer Prevention, the University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Ricardo A León-Letelier
- Department of Clinical Cancer Prevention, the University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Hiroyuki Katayama
- Department of Clinical Cancer Prevention, the University of Texas MD Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|
2
|
Jia W, Yuan J, Zhang J, Li S, Lin W, Cheng B. Bioactive sphingolipids as emerging targets for signal transduction in cancer development. Biochim Biophys Acta Rev Cancer 2024; 1879:189176. [PMID: 39233263 DOI: 10.1016/j.bbcan.2024.189176] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2024] [Revised: 08/28/2024] [Accepted: 08/28/2024] [Indexed: 09/06/2024]
Abstract
Sphingolipids, crucial components of cellular membranes, play a vital role in maintaining cellular structure and signaling integrity. Disruptions in sphingolipid metabolism are increasingly implicated in cancer development. Key bioactive sphingolipids, such as ceramides, sphingosine-1-phosphate (S1P), ceramide-1-phosphate (C1P), and glycosphingolipids, profoundly impact tumor biology. They influence the behavior of tumor cells, stromal cells, and immune cells, affecting tumor aggressiveness, angiogenesis, immune modulation, and extracellular matrix remodeling. Furthermore, abnormal expression of sphingolipids and their metabolizing enzymes modulates the secretion of tumor-derived extracellular vesicles (TDEs), which are key players in creating an immunosuppressive tumor microenvironment, remodeling the extracellular matrix, and facilitating oncogenic signaling within in situ tumors and distant pre-metastatic niches (PMNs). Understanding the role of sphingolipids in the biogenesis of tumor-derived extracellular vesicles (TDEs) and their bioactive contents can pave the way for new biomarkers in cancer diagnosis and prognosis, ultimately enhancing comprehensive tumor treatment strategies.
Collapse
Affiliation(s)
- Wentao Jia
- Department of General Practice, The First Affiliated Hospital of Naval Medical University, Shanghai 200433, China; Oncology Department of Traditional Chinese Medicine, The First Affiliated Hospital of Naval Medical University, Shanghai 200433, China; Faculty of Traditional Chinese Medicine, Naval Medical University, Shanghai 200043, China
| | - Jiaying Yuan
- Department of Pulmonary and Critical Care Medicine, Tongji Hospital, School of Medicine, Tongji University, Shanghai 200065, China
| | - Jinbo Zhang
- Department of Pharmacy, Tianjin Rehabilitation and Recuperation Center, Joint Logistics Support Force, Tianjin 300000, China
| | - Shu Li
- Department of Gastroenterology, Baoshan Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 201900, China
| | - Wanfu Lin
- Oncology Department of Traditional Chinese Medicine, The First Affiliated Hospital of Naval Medical University, Shanghai 200433, China; Faculty of Traditional Chinese Medicine, Naval Medical University, Shanghai 200043, China.
| | - Binbin Cheng
- Oncology Department of Traditional Chinese Medicine, The First Affiliated Hospital of Naval Medical University, Shanghai 200433, China; Faculty of Traditional Chinese Medicine, Naval Medical University, Shanghai 200043, China.
| |
Collapse
|
3
|
Hasnat MA, Ohmi Y, Yesmin F, Kambe M, Kawamoto Y, Bhuiyan RH, Mizutani M, Hashimoto N, Tsuchida A, Ohkawa Y, Kaneko K, Tajima O, Furukawa K, Furukawa K. Crucial roles of exosomes secreted from ganglioside GD3/GD2-positive glioma cells in enhancement of the malignant phenotypes and signals of GD3/GD2-negative glioma cells. NAGOYA JOURNAL OF MEDICAL SCIENCE 2024; 86:435-451. [PMID: 39355364 PMCID: PMC11439612 DOI: 10.18999/nagjms.86.3.435] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Accepted: 12/08/2023] [Indexed: 10/03/2024]
Abstract
Neuroectoderm-derived tumors characteristically express gangliosides such as GD3 and GD2. Many studies have reported that gangliosides GD3/GD2 enhance malignant phenotypes of cancers. Recently, we reported that human gliomas expressing GD3/GD2 exhibited enhanced malignant phenotypes. Here, we investigated the function of GD3/GD2 in glioma cells and GD3/GD2-expressing glioma-derived exosomes. As reported previously, transfectant cells of human glioma U251 MG expressing GD3/GD2 showed enhanced cancer phenotypes compared with GD3/GD2-negative controls. When GD3/GD2-negative cells were treated with exosomes secreted from GD3/GD2-positive cells, clearly increased malignant properties were observed. Furthermore, increased phosphorylation of signaling molecules was detected after 5-15 min of exosome treatment, ie, higher tyrosine phosphorylation of platelet-derived growth factor receptor, focal adhesion kinase, and paxillin was found in treated cells than in controls. Phosphorylation of extracellular signal-regulated kinase-1/2 was also enhanced. Consequently, it is suggested that exosomes secreted from GD3/GD2-positive gliomas play important roles in enhancement of the malignant properties of glioma cells, leading to total aggravation of heterogenous cancer tissues, and also in the regulation of tumor microenvironments.
Collapse
Affiliation(s)
- Mohammad Abul Hasnat
- Department of Biomedical Sciences, College of Life and Health Sciences, Chubu University, Kasugai, Japan
- Department of Biochemistry and Molecular Biology, Shahjalal University of Science and Technology, Sylhet-3114, Bangladesh
| | - Yuhsuke Ohmi
- Department of Clinical Engineering, College of Life and Health Sciences, Chubu University, Kasugai, Japan
| | - Farhana Yesmin
- Department of Biomedical Sciences, College of Life and Health Sciences, Chubu University, Kasugai, Japan
| | - Mariko Kambe
- Department of Biomedical Sciences, College of Life and Health Sciences, Chubu University, Kasugai, Japan
| | - Yoshiyuki Kawamoto
- Department of Immunology, College of Life and Health Sciences, Chubu University, Kasugai, Japan
| | - Robiul H Bhuiyan
- Department of Biomedical Sciences, College of Life and Health Sciences, Chubu University, Kasugai, Japan
| | - Momoka Mizutani
- Department of Biomedical Sciences, College of Life and Health Sciences, Chubu University, Kasugai, Japan
| | - Noboru Hashimoto
- Department of Tissue Regeneration, Tokushima University School of Dentistry, Tokushima, Japan
| | - Akiko Tsuchida
- Laboratory of Glyco-bioengineering, The Noguchi Institute, Tokyo, Japan
| | - Yuki Ohkawa
- Osaka International Cancer Institute, Osaka, Japan
| | - Kei Kaneko
- Department of Biomedical Sciences, College of Life and Health Sciences, Chubu University, Kasugai, Japan
| | - Orie Tajima
- Department of Biomedical Sciences, College of Life and Health Sciences, Chubu University, Kasugai, Japan
| | - Keiko Furukawa
- Department of Biomedical Sciences, College of Life and Health Sciences, Chubu University, Kasugai, Japan
| | - Koichi Furukawa
- Department of Biomedical Sciences, College of Life and Health Sciences, Chubu University, Kasugai, Japan
| |
Collapse
|
4
|
AlMalki RH, Al-Nasrallah HK, Aldossry A, Barnawi R, Al-Khaldi S, Almozyan S, Al-Ansari MM, Ghebeh H, Abdel Rahman AM, Al-Alwan M. Comparative Analysis of Breast Cancer Metabolomes Highlights Fascin's Central Role in Regulating Key Pathways Related to Disease Progression. Int J Mol Sci 2024; 25:7891. [PMID: 39063133 PMCID: PMC11277536 DOI: 10.3390/ijms25147891] [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: 05/21/2024] [Revised: 07/16/2024] [Accepted: 07/17/2024] [Indexed: 07/28/2024] Open
Abstract
Omics technologies provide useful tools for the identification of novel biomarkers in many diseases, including breast cancer, which is the most diagnosed cancer in women worldwide. We and others have reported a central role for the actin-bundling protein (fascin) in regulating breast cancer disease progression at different levels. However, whether fascin expression promotes metabolic molecules that could predict disease progression has not been fully elucidated. Here, fascin expression was manipulated via knockdown (fascinKD+NORF) and rescue (fascinKD+FORF) in the naturally fascin-positive (fascinpos+NORF) MDA-MB-231 breast cancer cells. Whether fascin dysregulates metabolic profiles that are associated with disease progression was assessed using untargeted metabolomics analyses via liquid chromatography-mass spectrometry. Overall, 12,226 metabolic features were detected in the tested cell pellets. Fascinpos+NORF cell pellets showed 2510 and 3804 significantly dysregulated metabolites compared to their fascinKD+NORF counterparts. Fascin rescue (fascinKD+FORF) revealed 2710 significantly dysregulated cellular metabolites compared to fascinKD+NORF counterparts. A total of 101 overlapped cellular metabolites between fascinKD+FORF and fascinpos+NORF were significantly dysregulated in the fascinKD+NORF cells. Analysis of the significantly dysregulated metabolites by fascin expression revealed their involvement in the metabolism of sphingolipid, phenylalanine, tyrosine, and tryptophan biosynthesis, and pantothenate and CoA biosynthesis, which are critical pathways for breast cancer progression. Our findings of fascin-mediated alteration of metabolic pathways could be used as putative poor prognostic biomarkers and highlight other underlying mechanisms of fascin contribution to breast cancer progression.
Collapse
Affiliation(s)
- Reem H. AlMalki
- Metabolomics Section, Department of Clinical Genomics, Center for Genomics Medicine, King Faisal Specialist Hospital and Research Centre, Riyadh 11211, Saudi Arabia;
| | - Huda K. Al-Nasrallah
- Cell Therapy and Immunobiology Department, King Faisal Specialist Hospital and Research Centre, Riyadh 11211, Saudi Arabia; (H.K.A.-N.); (A.A.); (R.B.); (S.A.-K.); (S.A.); (H.G.)
| | - Alanoud Aldossry
- Cell Therapy and Immunobiology Department, King Faisal Specialist Hospital and Research Centre, Riyadh 11211, Saudi Arabia; (H.K.A.-N.); (A.A.); (R.B.); (S.A.-K.); (S.A.); (H.G.)
| | - Rayanah Barnawi
- Cell Therapy and Immunobiology Department, King Faisal Specialist Hospital and Research Centre, Riyadh 11211, Saudi Arabia; (H.K.A.-N.); (A.A.); (R.B.); (S.A.-K.); (S.A.); (H.G.)
| | - Samiyah Al-Khaldi
- Cell Therapy and Immunobiology Department, King Faisal Specialist Hospital and Research Centre, Riyadh 11211, Saudi Arabia; (H.K.A.-N.); (A.A.); (R.B.); (S.A.-K.); (S.A.); (H.G.)
- Applied Genomics Technologies Institute, Health Sector, King Abdulaziz City for Sciences and Technology, Riyadh 11442, Saudi Arabia
| | - Sheema Almozyan
- Cell Therapy and Immunobiology Department, King Faisal Specialist Hospital and Research Centre, Riyadh 11211, Saudi Arabia; (H.K.A.-N.); (A.A.); (R.B.); (S.A.-K.); (S.A.); (H.G.)
| | - Mysoon M. Al-Ansari
- Department of Molecular Oncology, Cancer Biology & Experimental Therapeutics Section, King Faisal Specialist Hospital and Research Centre, Riyadh 11211, Saudi Arabia;
- Department of Botany and Microbiology, College of Science, King Saud University, Riyadh 11451, Saudi Arabia
| | - Hazem Ghebeh
- Cell Therapy and Immunobiology Department, King Faisal Specialist Hospital and Research Centre, Riyadh 11211, Saudi Arabia; (H.K.A.-N.); (A.A.); (R.B.); (S.A.-K.); (S.A.); (H.G.)
- College of Medicine, Al-Faisal University, Riyadh 11533, Saudi Arabia
| | - Anas M. Abdel Rahman
- Metabolomics Section, Department of Clinical Genomics, Center for Genomics Medicine, King Faisal Specialist Hospital and Research Centre, Riyadh 11211, Saudi Arabia;
- College of Medicine, Al-Faisal University, Riyadh 11533, Saudi Arabia
| | - Monther Al-Alwan
- Cell Therapy and Immunobiology Department, King Faisal Specialist Hospital and Research Centre, Riyadh 11211, Saudi Arabia; (H.K.A.-N.); (A.A.); (R.B.); (S.A.-K.); (S.A.); (H.G.)
- College of Medicine, Al-Faisal University, Riyadh 11533, Saudi Arabia
| |
Collapse
|
5
|
Gorostegui M, Muñoz JP, Perez-Jaume S, Simao-Rafael M, Larrosa C, Garraus M, Salvador N, Lavarino C, Krauel L, Mañe S, Castañeda A, Mora J. Management of High-Risk Neuroblastoma with Soft-Tissue-Only Disease in the Era of Anti-GD2 Immunotherapy. Cancers (Basel) 2024; 16:1735. [PMID: 38730688 PMCID: PMC11083939 DOI: 10.3390/cancers16091735] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Accepted: 04/24/2024] [Indexed: 05/13/2024] Open
Abstract
Neuroblastoma presents with two patterns of disease: locoregional or systemic. The poor prognostic risk factors of locoregional neuroblastoma (LR-NB) include age, MYCN or MDM2-CDK4 amplification, 11q, histology, diploidy with ALK or TERT mutations, and ATRX aberrations. Anti-GD2 immunotherapy has significantly improved the outcome of high-risk (HR) NB and is mostly effective against osteomedullary minimal residual disease (MRD), but less so against soft tissue disease. The question is whether adding anti-GD2 monoclonal antibodies (mAbs) benefits patients with HR-NB compounded by only soft tissue. We reviewed 31 patients treated at SJD for HR-NB with no osteomedullary involvement at diagnosis. All tumors had molecular genetic features of HR-NB. The outcome after first-line treatment showed 25 (80.6%) patients achieving CR. Thirteen patients remain in continued CR, median follow-up 3.9 years. We analyzed whether adding anti-GD2 immunotherapy to first-line treatment had any prognostic significance. The EFS analysis using Cox models showed a HR of 0.20, p = 0.0054, and an 80% decrease in the risk of relapse in patients treated with anti-GD2 immunotherapy in the first line. Neither EFS nor OS were significantly different by CR status after first-line treatment. In conclusion, adding treatment with anti-GD2 mAbs at the stage of MRD helps prevent relapse that unequivocally portends poor survival.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | - Jaume Mora
- Pediatric Cancer Center Barcelona, Hospital Sant Joan de Déu, 08950 Barcelona, Spain; (M.G.); (J.P.M.); (M.S.-R.); (C.L.); (M.G.); (N.S.); (C.L.); (L.K.); (S.M.); (A.C.)
| |
Collapse
|
6
|
Philippova J, Shevchenko J, Sennikov S. GD2-targeting therapy: a comparative analysis of approaches and promising directions. Front Immunol 2024; 15:1371345. [PMID: 38558810 PMCID: PMC10979305 DOI: 10.3389/fimmu.2024.1371345] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Accepted: 02/26/2024] [Indexed: 04/04/2024] Open
Abstract
Disialoganglioside GD2 is a promising target for immunotherapy with expression primarily restricted to neuroectodermal and epithelial tumor cells. Although its role in the maintenance and repair of neural tissue is well-established, its functions during normal organism development remain understudied. Meanwhile, studies have shown that GD2 plays an important role in tumorigenesis. Its functions include proliferation, invasion, motility, and metastasis, and its high expression and ability to transform the tumor microenvironment may be associated with a malignant phenotype. Structurally, GD2 is a glycosphingolipid that is stably expressed on the surface of tumor cells, making it a suitable candidate for targeting by antibodies or chimeric antigen receptors. Based on mouse monoclonal antibodies, chimeric and humanized antibodies and their combinations with cytokines, toxins, drugs, radionuclides, nanoparticles as well as chimeric antigen receptor have been developed. Furthermore, vaccines and photoimmunotherapy are being used to treat GD2-positive tumors, and GD2 aptamers can be used for targeting. In the field of cell therapy, allogeneic immunocompetent cells are also being utilized to enhance GD2 therapy. Efforts are currently being made to optimize the chimeric antigen receptor by modifying its design or by transducing not only αβ T cells, but also γδ T cells, NK cells, NKT cells, and macrophages. In addition, immunotherapy can combine both diagnostic and therapeutic methods, allowing for early detection of disease and minimal residual disease. This review discusses each immunotherapy method and strategy, its advantages and disadvantages, and highlights future directions for GD2 therapy.
Collapse
Affiliation(s)
| | | | - Sergey Sennikov
- Laboratory of Molecular Immunology, Federal State Budgetary Scientific Institution Research Institute of Fundamental and Clinical Immunology, Novosibirsk, Russia
| |
Collapse
|
7
|
Li G, Wang H, Meftahpour V. Overall review of curative impact and barriers of CAR-T cells in osteosarcoma. EXCLI JOURNAL 2024; 23:364-383. [PMID: 38655095 PMCID: PMC11036068 DOI: 10.17179/excli2023-6760] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Accepted: 02/27/2024] [Indexed: 04/26/2024]
Abstract
Osteosarcoma (OS) is a rare form of cancer and primary bone malignancy in children and adolescents. Current therapies include surgery, chemotherapy, and amputation. Therefore, a new therapeutic strategy is needed to dramatically change cancer treatment. Recently, chimeric antigen receptor T cells (CAR-T cells) have been of considerable interest as it has provided auspicious results and patients suffering from low side effects after injection that resolve with current therapy. However, there are reports that cytokine release storm (CRS) can be observed in some patients. In addition, as researchers have faced problems that limit and suppress T cells, further studies are required to resolve these problems. In addition, to maximize the therapeutic benefit of CAR-T cell therapy, researchers have suggested that combination therapy could be better used to treat cancer by overcoming any problems and reducing side effects as much as possible. This review summarizes these problems, barriers, and the results of some studies on the evaluation of CAR-T cells in patients with osteosarcoma.
Collapse
Affiliation(s)
- Guilin Li
- Xinyang Vocational and Technical College, Xinyang Henan 464000 China
| | - Hong Wang
- Xinyang Vocational and Technical College, Xinyang Henan 464000 China
| | - Vafa Meftahpour
- Medical Immunology, Cellular and Molecular Research Center, Urmia University of Medical Sciences, Urmia, Iran
| |
Collapse
|
8
|
Nejatie A, Yee SS, Jeter A, Saragovi HU. The cancer glycocode as a family of diagnostic biomarkers, exemplified by tumor-associated gangliosides. Front Oncol 2023; 13:1261090. [PMID: 37954075 PMCID: PMC10637394 DOI: 10.3389/fonc.2023.1261090] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Accepted: 10/13/2023] [Indexed: 11/14/2023] Open
Abstract
One unexploited family of cancer biomarkers comprise glycoproteins, carbohydrates, and glycolipids (the Tumor Glycocode).A class of glycolipid cancer biomarkers, the tumor-marker gangliosides (TMGs) are presented here as potential diagnostics for detecting cancer, especially at early stages, as the biological function of TMGs makes them etiological. We propose that a quantitative matrix of the Cancer Biomarker Glycocode and artificial intelligence-driven algorithms will expand the menu of validated cancer biomarkers as a step to resolve some of the challenges in cancer diagnosis, and yield a combination that can identify a specific cancer, in a tissue-agnostic manner especially at early stages, to enable early intervention. Diagnosis is critical to reducing cancer mortality but many cancers lack efficient and effective diagnostic tests, especially for early stage disease. Ideal diagnostic biomarkers are etiological, samples are preferably obtained via non-invasive methods (e.g. liquid biopsy of blood or urine), and are quantitated using assays that yield high diagnostic sensitivity and specificity for efficient diagnosis, prognosis, or predicting response to therapy. Validated biomarkers with these features are rare. While the advent of proteomics and genomics has led to the identification of a multitude of proteins and nucleic acid sequences as cancer biomarkers, relatively few have been approved for clinical use. The use of multiplex arrays and artificial intelligence-driven algorithms offer the option of combining data of known biomarkers; however, for most, the sensitivity and the specificity are below acceptable criteria, and clinical validation has proven difficult. One strategic solution to this problem is to expand the biomarker families beyond those currently exploited. One unexploited family of cancer biomarkers comprise glycoproteins, carbohydrates, and glycolipids (the Tumor Glycocode). Here, we focus on a family of glycolipid cancer biomarkers, the tumor-marker gangliosides (TMGs). We discuss the diagnostic potential of TMGs for detecting cancer, especially at early stages. We include prior studies from the literature to summarize findings for ganglioside quantification, expression, detection, and biological function and its role in various cancers. We highlight the examples of TMGs exhibiting ideal properties of cancer diagnostic biomarkers, and the application of GD2 and GD3 for diagnosis of early stage cancers with high sensitivity and specificity. We propose that a quantitative matrix of the Cancer Biomarker Glycocode and artificial intelligence-driven algorithms will expand the menu of validated cancer biomarkers as a step to resolve some of the challenges in cancer diagnosis, and yield a combination that can identify a specific cancer, in a tissue-agnostic manner especially at early stages, to enable early intervention.
Collapse
Affiliation(s)
- Ali Nejatie
- Center for Translational Research, Lady Davis Research Institute-Jewish General Hospital, Montreal, QC, Canada
- Pharmacology and Therapeutics, McGill University, Montreal, QC, Canada
| | - Samantha S. Yee
- Department of Obstetrics and Gynecology, University of Chicago, Chicago, IL, United States
| | | | - Horacio Uri Saragovi
- Center for Translational Research, Lady Davis Research Institute-Jewish General Hospital, Montreal, QC, Canada
- Pharmacology and Therapeutics, McGill University, Montreal, QC, Canada
- Ophthalmology and Vision Science, McGill University, Montreal, QC, Canada
| |
Collapse
|
9
|
Lodewijk I, Dueñas M, Paramio JM, Rubio C. CD44v6, STn & O-GD2: promising tumor associated antigens paving the way for new targeted cancer therapies. Front Immunol 2023; 14:1272681. [PMID: 37854601 PMCID: PMC10579806 DOI: 10.3389/fimmu.2023.1272681] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Accepted: 09/14/2023] [Indexed: 10/20/2023] Open
Abstract
Targeted therapies are the state of the art in oncology today, and every year new Tumor-associated antigens (TAAs) are developed for preclinical research and clinical trials, but few of them really change the therapeutic scenario. Difficulties, either to find antigens that are solely expressed in tumors or the generation of good binders to these antigens, represent a major bottleneck. Specialized cellular mechanisms, such as differential splicing and glycosylation processes, are a good source of neo-antigen expression. Changes in these processes generate surface proteins that, instead of showing decreased or increased antigen expression driven by enhanced mRNA processing, are aberrant in nature and therefore more specific targets to elicit a precise anti-tumor therapy. Here, we present promising TAAs demonstrated to be potential targets for cancer monitoring, targeted therapy and the generation of new immunotherapy tools, such as recombinant antibodies and chimeric antigen receptor (CAR) T cell (CAR-T) or Chimeric Antigen Receptor-Engineered Natural Killer (CAR-NK) for specific tumor killing, in a wide variety of tumor types. Specifically, this review is a detailed update on TAAs CD44v6, STn and O-GD2, describing their origin as well as their current and potential use as disease biomarker and therapeutic target in a diversity of tumor types.
Collapse
Affiliation(s)
- Iris Lodewijk
- Biomedical Research Institute I+12, University Hospital “12 de Octubre”, Madrid, Spain
- Molecular Oncology Unit, CIEMAT (Centro de Investigaciones Energéticas, Medioambientales y Tecnológicas), Madrid, Spain
- Biomedical Research Networking Center on Oncology-CIBERONC, ISCIII, Madrid, Spain
| | - Marta Dueñas
- Biomedical Research Institute I+12, University Hospital “12 de Octubre”, Madrid, Spain
- Molecular Oncology Unit, CIEMAT (Centro de Investigaciones Energéticas, Medioambientales y Tecnológicas), Madrid, Spain
- Biomedical Research Networking Center on Oncology-CIBERONC, ISCIII, Madrid, Spain
| | - Jesus M. Paramio
- Biomedical Research Institute I+12, University Hospital “12 de Octubre”, Madrid, Spain
- Molecular Oncology Unit, CIEMAT (Centro de Investigaciones Energéticas, Medioambientales y Tecnológicas), Madrid, Spain
- Biomedical Research Networking Center on Oncology-CIBERONC, ISCIII, Madrid, Spain
| | - Carolina Rubio
- Biomedical Research Institute I+12, University Hospital “12 de Octubre”, Madrid, Spain
- Molecular Oncology Unit, CIEMAT (Centro de Investigaciones Energéticas, Medioambientales y Tecnológicas), Madrid, Spain
- Biomedical Research Networking Center on Oncology-CIBERONC, ISCIII, Madrid, Spain
| |
Collapse
|
10
|
Machy P, Mortier E, Birklé S. Biology of GD2 ganglioside: implications for cancer immunotherapy. Front Pharmacol 2023; 14:1249929. [PMID: 37670947 PMCID: PMC10475612 DOI: 10.3389/fphar.2023.1249929] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Accepted: 07/31/2023] [Indexed: 09/07/2023] Open
Abstract
Part of the broader glycosphingolipid family, gangliosides are composed of a ceramide bound to a sialic acid-containing glycan chain, and locate at the plasma membrane. Gangliosides are produced through sequential steps of glycosylation and sialylation. This diversity of composition is reflected in differences in expression patterns and functions of the various gangliosides. Ganglioside GD2 designates different subspecies following a basic structure containing three carbohydrate residues and two sialic acids. GD2 expression, usually restrained to limited tissues, is frequently altered in various neuroectoderm-derived cancers. While GD2 is of evident interest, its glycolipid nature has rendered research challenging. Physiological GD2 expression has been linked to developmental processes. Passing this stage, varying levels of GD2, physiologically expressed mainly in the central nervous system, affect composition and formation of membrane microdomains involved in surface receptor signaling. Overexpressed in cancer, GD2 has been shown to enhance cell survival and invasion. Furthermore, binding of antibodies leads to immune-independent cell death mechanisms. In addition, GD2 contributes to T-cell dysfunction, and functions as an immune checkpoint. Given the cancer-associated functions, GD2 has been a source of interest for immunotherapy. As a potential biomarker, methods are being developed to quantify GD2 from patients' samples. In addition, various therapeutic strategies are tested. Based on initial success with antibodies, derivates such as bispecific antibodies and immunocytokines have been developed, engaging patient immune system. Cytotoxic effectors or payloads may be redirected based on anti-GD2 antibodies. Finally, vaccines can be used to mount an immune response in patients. We review here the pertinent biological information on GD2 which may be of use for optimizing current immunotherapeutic strategies.
Collapse
Affiliation(s)
| | | | - Stéphane Birklé
- Nantes Université, Univ Angers, INSERM, CNRS, CRCI2NA, Nantes, France
| |
Collapse
|
11
|
Park JA, Cheung NKV. Promise and Challenges of T Cell Immunotherapy for Osteosarcoma. Int J Mol Sci 2023; 24:12520. [PMID: 37569894 PMCID: PMC10419531 DOI: 10.3390/ijms241512520] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Revised: 07/30/2023] [Accepted: 08/03/2023] [Indexed: 08/13/2023] Open
Abstract
The cure rate for metastatic or relapsed osteosarcoma has not substantially improved over the past decades despite the exploitation of multimodal treatment approaches, allowing long-term survival in less than 30% of cases. Patients with osteosarcoma often develop resistance to chemotherapeutic agents, where personalized targeted therapies should offer new hope. T cell immunotherapy as a complementary or alternative treatment modality is advancing rapidly in general, but its potential against osteosarcoma remains largely unexplored. Strategies incorporating immune checkpoint inhibitors (ICIs), chimeric antigen receptor (CAR) modified T cells, and T cell engaging bispecific antibodies (BsAbs) are being explored to tackle relapsed or refractory osteosarcoma. However, osteosarcoma is an inherently heterogeneous tumor, both at the intra- and inter-tumor level, with no identical driver mutations. It has a pro-tumoral microenvironment, where bone cells, stromal cells, neovasculature, suppressive immune cells, and a mineralized extracellular matrix (ECM) combine to derail T cell infiltration and its anti-tumor function. To realize the potential of T cell immunotherapy in osteosarcoma, an integrated approach targeting this complex ecosystem needs smart planning and execution. Herein, we review the current status of T cell immunotherapies for osteosarcoma, summarize the challenges encountered, and explore combination strategies to overcome these hurdles, with the ultimate goal of curing osteosarcoma with less acute and long-term side effects.
Collapse
Affiliation(s)
- Jeong A Park
- Department of Pediatrics, Inha University College of Medicine, Incheon 22212, Republic of Korea
| | - Nai-Kong V. Cheung
- Pediatrics, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA;
| |
Collapse
|
12
|
Larrosa C, Mora J, Cheung NK. Global Impact of Monoclonal Antibodies (mAbs) in Children: A Focus on Anti-GD2. Cancers (Basel) 2023; 15:3729. [PMID: 37509390 PMCID: PMC10378537 DOI: 10.3390/cancers15143729] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2023] [Revised: 07/16/2023] [Accepted: 07/17/2023] [Indexed: 07/30/2023] Open
Abstract
Monoclonal antibodies (mAbs), as the name implies, are clonal antibodies that bind to the same antigen. mAbs are broadly used as diagnostic or therapeutic tools for neoplasms, autoimmune diseases, allergic conditions, and infections. Although most mAbs are approved for treating adult cancers, few are applicable to childhood malignancies, limited mostly to hematological cancers. As for solid tumors, only anti-disialoganglioside (GD2) mAbs are approved specifically for neuroblastoma. Inequities of drug access have continued, affecting most therapeutic mAbs globally. To understand these challenges, a deeper dive into the complex transition from basic research to the clinic, or between marketing and regulatory agencies, is timely. This review focuses on current mAbs approved or under investigation in pediatric cancer, with special attention on solid tumors and anti-GD2 mAbs, and the hurdles that limit their broad global access. Beyond understanding the mechanisms of drug resistance, the continual discovery of next generation drugs safer for children and easier to administer, the discovery of predictive biomarkers to avoid futility should ease the acceptance by patient, health care professionals and regulatory agencies, in order to expand clinical utility. With a better integration into the multimodal treatment for each disease, protocols that align with the regional clinical practice should also improve acceptance and cost-effectiveness. Communication and collaboration between academic institutions, pharmaceutical companies, and regulatory agencies should help to ensure accessible, affordable, and sustainable health care for all.
Collapse
Affiliation(s)
- Cristina Larrosa
- Pediatric Cancer Center Barcelona, 08950 Barcelona, Spain; (C.L.); (J.M.)
| | - Jaume Mora
- Pediatric Cancer Center Barcelona, 08950 Barcelona, Spain; (C.L.); (J.M.)
| | - Nai-Kong Cheung
- Memorial Sloan-Kettering Cancer Center, New York, NY 10065, USA
| |
Collapse
|
13
|
El Malki K, Wehling P, Alt F, Sandhoff R, Zahnreich S, Ustjanzew A, Wilzius C, Brockmann MA, Wingerter A, Russo A, Beck O, Sommer C, Ottenhausen M, Frauenknecht KBM, Paret C, Faber J. Glucosylceramide Synthase Inhibitors Induce Ceramide Accumulation and Sensitize H3K27 Mutant Diffuse Midline Glioma to Irradiation. Int J Mol Sci 2023; 24:9905. [PMID: 37373053 DOI: 10.3390/ijms24129905] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2023] [Revised: 06/04/2023] [Accepted: 06/05/2023] [Indexed: 06/29/2023] Open
Abstract
H3K27M mutant (mut) diffuse midline glioma (DMG) is a lethal cancer with no effective cure. The glycosphingolipids (GSL) metabolism is altered in these tumors and could be exploited to develop new therapies. We tested the effect of the glucosylceramide synthase inhibitors (GSI) miglustat and eliglustat on cell proliferation, alone or in combination with temozolomide or ionizing radiation. Miglustat was included in the therapy protocol of two pediatric patients. The effect of H3.3K27 trimethylation on GSL composition was analyzed in ependymoma. GSI reduced the expression of the ganglioside GD2 in a concentration and time-dependent manner and increased the expression of ceramide, ceramide 1-phosphate, sphingosine, and sphingomyelin but not of sphingosine 1-phosphate. Miglustat significantly increased the efficacy of irradiation. Treatment with miglustat according to dose recommendations for patients with Niemann-Pick disease was well tolerated with manageable toxicities. One patient showed a mixed response. In ependymoma, a high concentration of GD2 was found only in the presence of the loss of H3.3K27 trimethylation. In conclusion, treatment with miglustat and, in general, targeting GSL metabolism may offer a new therapeutic opportunity and can be administered in close proximity to radiation therapy. Alterations in H3K27 could be useful to identify patients with a deregulated GSL metabolism.
Collapse
Affiliation(s)
- Khalifa El Malki
- Department of Pediatric Hematology/Oncology, Center for Pediatric and Adolescent Medicine, University Medical Center, Johannes Gutenberg-University Mainz, 55131 Mainz, Germany
- University Cancer Center (UCT), University Medical Center, Johannes Gutenberg-University Mainz, 55131 Mainz, Germany
| | - Pia Wehling
- Department of Pediatric Hematology/Oncology, Center for Pediatric and Adolescent Medicine, University Medical Center, Johannes Gutenberg-University Mainz, 55131 Mainz, Germany
- University Cancer Center (UCT), University Medical Center, Johannes Gutenberg-University Mainz, 55131 Mainz, Germany
| | - Francesca Alt
- Department of Pediatric Hematology/Oncology, Center for Pediatric and Adolescent Medicine, University Medical Center, Johannes Gutenberg-University Mainz, 55131 Mainz, Germany
- University Cancer Center (UCT), University Medical Center, Johannes Gutenberg-University Mainz, 55131 Mainz, Germany
| | - Roger Sandhoff
- Lipid Pathobiochemistry, German Cancer Research Center, 69120 Heidelberg, Germany
- Helmholtz-Institute for Translational Oncology Mainz (HI-TRON), 55131 Mainz, Germany
| | - Sebastian Zahnreich
- Department of Radiation Oncology and Radiation Therapy, University Medical Center, Johannes Gutenberg University Mainz, 55131 Mainz, Germany
| | - Arsenij Ustjanzew
- Institute of Medical Biostatistics, Epidemiology and Informatics (IMBEI), University Medical Center, Johannes Gutenberg-University Mainz, 55131 Mainz, Germany
| | - Carolin Wilzius
- Lipid Pathobiochemistry, German Cancer Research Center, 69120 Heidelberg, Germany
| | - Marc A Brockmann
- Department of Neuroradiology, University Medical Center, Johannes Gutenberg University Mainz, 55131 Mainz, Germany
| | - Arthur Wingerter
- Department of Pediatric Hematology/Oncology, Center for Pediatric and Adolescent Medicine, University Medical Center, Johannes Gutenberg-University Mainz, 55131 Mainz, Germany
- University Cancer Center (UCT), University Medical Center, Johannes Gutenberg-University Mainz, 55131 Mainz, Germany
| | - Alexandra Russo
- Department of Pediatric Hematology/Oncology, Center for Pediatric and Adolescent Medicine, University Medical Center, Johannes Gutenberg-University Mainz, 55131 Mainz, Germany
- University Cancer Center (UCT), University Medical Center, Johannes Gutenberg-University Mainz, 55131 Mainz, Germany
- German Cancer Consortium (DKTK), Site Frankfurt/Mainz, Germany, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| | - Olaf Beck
- Department of Pediatric Hematology/Oncology, Center for Pediatric and Adolescent Medicine, University Medical Center, Johannes Gutenberg-University Mainz, 55131 Mainz, Germany
- University Cancer Center (UCT), University Medical Center, Johannes Gutenberg-University Mainz, 55131 Mainz, Germany
| | - Clemens Sommer
- Institute of Neuropathology, University Medical Center, Johannes Gutenberg-University Mainz, 55131 Mainz, Germany
| | - Malte Ottenhausen
- Department of Neurosurgery, University Medical Center, Johannes Gutenberg-University Mainz, 55131 Mainz, Germany
| | - Katrin B M Frauenknecht
- Helmholtz-Institute for Translational Oncology Mainz (HI-TRON), 55131 Mainz, Germany
- Institute of Neuropathology, University Medical Center, Johannes Gutenberg-University Mainz, 55131 Mainz, Germany
- National Center of Pathology (NCP), Laboratoire National de Santé, 3555 Dudelange, Luxembourg
- Luxembourg Center of Neuropathology (LCNP), Laboratoire National de Santé, 3555 Dudelange, Luxembourg
| | - Claudia Paret
- Department of Pediatric Hematology/Oncology, Center for Pediatric and Adolescent Medicine, University Medical Center, Johannes Gutenberg-University Mainz, 55131 Mainz, Germany
- University Cancer Center (UCT), University Medical Center, Johannes Gutenberg-University Mainz, 55131 Mainz, Germany
- Helmholtz-Institute for Translational Oncology Mainz (HI-TRON), 55131 Mainz, Germany
- German Cancer Consortium (DKTK), Site Frankfurt/Mainz, Germany, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
- Research Center of Immunotherapy (FZI), University Medical Center, Johannes Gutenberg-University Mainz, 55131 Mainz, Germany
| | - Jörg Faber
- Department of Pediatric Hematology/Oncology, Center for Pediatric and Adolescent Medicine, University Medical Center, Johannes Gutenberg-University Mainz, 55131 Mainz, Germany
- University Cancer Center (UCT), University Medical Center, Johannes Gutenberg-University Mainz, 55131 Mainz, Germany
- Helmholtz-Institute for Translational Oncology Mainz (HI-TRON), 55131 Mainz, Germany
- German Cancer Consortium (DKTK), Site Frankfurt/Mainz, Germany, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| |
Collapse
|
14
|
Ju WS, Seo SY, Mun SE, Kim K, Yu JO, Ryu JS, Kim JS, Choo YK. 7,8-Dihydroxyflavone induces mitochondrial apoptosis and down-regulates the expression of ganglioside GD3 in malignant melanoma cells. Discov Oncol 2023; 14:36. [PMID: 36991237 PMCID: PMC10060447 DOI: 10.1007/s12672-023-00643-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Accepted: 03/22/2023] [Indexed: 03/31/2023] Open
Abstract
Malignant melanoma is a skin cancer with poor prognosis and high resistance to conventional treatment. 7,8-dihydroxyflavone (7,8-DHF) has shown anti-carcinogenic, anti-inflammatory, anti-oxidant, and pharmacological effects in several types of cancer. However, the relationship between ganglioside expression and the anti-cancer effects of 7,8-DHF in melanoma is not fully understood. In the present study, 7,8-DHF exhibits specific anti-proliferation, anti-migration, and G2/M phase cell-cycle arrest effects on both melanoma cancer cell lines, and induces mitochondrial dysfunction and apoptosis, making it a potent candidate for anti-melanoma treatment. Furthermore, we confirmed that 7,8-DHF significantly reduces the expression levels of ganglioside GD3 and its synthase, which are known to be closely involved in carcinogenesis. Taken together, our findings suggest that 7,8-DHF may be a potent anti-cancer drug candidate for the treatment of malignant melanoma.
Collapse
Affiliation(s)
- Won Seok Ju
- Department of Biological Science, College of Natural Sciences, Wonkwang University, 460, Iksan-daero, Iksan-si, Jeollabuk-do, 54538, Republic of Korea
- Animal Biotechnology Division, Rural Development Administration, National Institute of Animal Science, 1500 Kongjwipatjwi-ro, Iseo-myeon, Wanju-gun, Jeonbuk, 55365, Republic of Korea
| | - Sang Young Seo
- Department of Biological Science, College of Natural Sciences, Wonkwang University, 460, Iksan-daero, Iksan-si, Jeollabuk-do, 54538, Republic of Korea
| | - Seong-Eun Mun
- Department of Biological Science, College of Natural Sciences, Wonkwang University, 460, Iksan-daero, Iksan-si, Jeollabuk-do, 54538, Republic of Korea
| | - Kyongtae Kim
- Department of Biological Science, College of Natural Sciences, Wonkwang University, 460, Iksan-daero, Iksan-si, Jeollabuk-do, 54538, Republic of Korea
| | - Jin Ok Yu
- Department of Biological Science, College of Natural Sciences, Wonkwang University, 460, Iksan-daero, Iksan-si, Jeollabuk-do, 54538, Republic of Korea
| | - Jae-Sung Ryu
- Stem Cell Convergence Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), 125, Gwahak-ro, Yuseong-gu, Daejeon, 34141, Republic of Korea
| | - Ji-Su Kim
- Primate Resources Center (PRC), Korea Research Institute of Bioscience and Biotechnology (KRIBB), 181, Ipsin-gil, Jeongeup-si, Jeollabuk-do, 56216, Republic of Korea
| | - Young-Kug Choo
- Department of Biological Science, College of Natural Sciences, Wonkwang University, 460, Iksan-daero, Iksan-si, Jeollabuk-do, 54538, Republic of Korea.
- Institute for Glycoscience, Wonkwang University, 460, Iksan-daero, Iksan-si, Jeollabuk-do, 54538, Republic of Korea.
| |
Collapse
|
15
|
Yesmin F, Furukawa K, Kambe M, Ohmi Y, Bhuiyan RH, Hasnat MA, Mizutani M, Tajima O, Hashimoto N, Tsuchida A, Kaneko K, Furukawa K. Extracellular vesicles released from ganglioside GD2-expressing melanoma cells enhance the malignant properties of GD2-negative melanomas. Sci Rep 2023; 13:4987. [PMID: 36973292 PMCID: PMC10042834 DOI: 10.1038/s41598-023-31216-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Accepted: 03/08/2023] [Indexed: 03/29/2023] Open
Abstract
Exosomes (small extracellular vesicles: EVs) have attracted increasing attention from basic scientists and clinicians since they play important roles in cell-to-cell communication in various biological processes. Various features of EVs have been elucidated regarding their contents, generation and secretion mechanisms, and functions in inflammation, regeneration, and cancers. These vesicles are reported to contain proteins, RNAs, microRNAs, DNAs, and lipids. Although the roles of individual components have been rigorously studied, the presence and roles of glycans in EVs have rarely been reported. In particular, glycosphingolipids in EVs have not been investigated to date. In this study, the expression and function of a representative cancer-associated ganglioside, GD2, in malignant melanomas was investigated. Generally, cancer-associated gangliosides have been shown to enhance malignant properties and signals in cancers. Notably, EVs derived from GD2-expressing melanomas enhanced the malignant phenotypes of GD2-negative melanomas, such as cell growth, invasion, and cell adhesion, in a dose-dependent manner. The EVs also induced increased phosphorylation of signaling molecules such as EGF receptor and focal adhesion kinase. These results suggest that EVs released from cancer-associated ganglioside-expressing cells exert many functions that have been reported as a function of these gangliosides and regulate microenvironments, including total aggravation of heterogeneous cancer tissues, leading to more malignant and advanced cancer types.
Collapse
Affiliation(s)
- Farhana Yesmin
- Department of Biomedical Sciences, Chubu University College of Life and Health Sciences, Matsumoto 1200, Kasugai, Aichi, 487-8501, Japan
| | - Keiko Furukawa
- Department of Biomedical Sciences, Chubu University College of Life and Health Sciences, Matsumoto 1200, Kasugai, Aichi, 487-8501, Japan
| | - Mariko Kambe
- Department of Biomedical Sciences, Chubu University College of Life and Health Sciences, Matsumoto 1200, Kasugai, Aichi, 487-8501, Japan
| | - Yuhsuke Ohmi
- Department of Clinical Engineering, Chubu University College of Life and Health Sciences, Matsumoto 1200, Kasugai, Aichi, 487-8501, Japan
| | - Robiul Hasan Bhuiyan
- Department of Biomedical Sciences, Chubu University College of Life and Health Sciences, Matsumoto 1200, Kasugai, Aichi, 487-8501, Japan
- Department of Biochemistry and Molecular Biology, University of Chittagong, 4331, Chittagong, Bangladesh
| | - Mohammad Abul Hasnat
- Department of Biomedical Sciences, Chubu University College of Life and Health Sciences, Matsumoto 1200, Kasugai, Aichi, 487-8501, Japan
| | - Momoka Mizutani
- Department of Biomedical Sciences, Chubu University College of Life and Health Sciences, Matsumoto 1200, Kasugai, Aichi, 487-8501, Japan
| | - Orie Tajima
- Department of Biomedical Sciences, Chubu University College of Life and Health Sciences, Matsumoto 1200, Kasugai, Aichi, 487-8501, Japan
| | - Noboru Hashimoto
- Department of Tissue Regeneration, Tokushima University School of Dentistry, Kuramoto-Cho 3, Tokushima, 770-8504, Japan
| | - Akiko Tsuchida
- Laboratory of Glyco- Bioengineering, The Noguchi Institute, Itabashi, 173-0003, Japan
| | - Kei Kaneko
- Department of Biomedical Sciences, Chubu University College of Life and Health Sciences, Matsumoto 1200, Kasugai, Aichi, 487-8501, Japan
| | - Koichi Furukawa
- Department of Biomedical Sciences, Chubu University College of Life and Health Sciences, Matsumoto 1200, Kasugai, Aichi, 487-8501, Japan.
| |
Collapse
|
16
|
Possible regulation of ganglioside GD3 synthase gene expression with DNA methylation in human glioma cells. Glycoconj J 2023; 40:323-332. [PMID: 36897478 DOI: 10.1007/s10719-023-10108-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 02/02/2023] [Accepted: 02/21/2023] [Indexed: 03/11/2023]
Abstract
Gangliosides are expressed in nervous systems and some neuroectoderm-derived tumors at high levels and play pivotal roles. However, mechanisms for the regulation of glycosyltransferase genes responsible for the ganglioside synthesis are not well understood. In this study, we analyzed DNA methylation patterns of promoter regions of GD3 synthase (ST8SIA1) as well as mRNA levels and ganglioside expression using human glioma cell lines. Among 5 cell lines examined, 4 lines showed changes in the expression levels of related genes after treatment with 5-aza-dC. LN319 showed up-regulation of St8sia1 and increased b-series gangliosides after 5-aza-dC treatment, and an astrocytoma cell line, AS showed high expression of ST8SIA1 and b-series gangliosides persistently before and after 5-Aza-2'-deoxycytidine treatment. Using these 2 cell lines, DNA methylation patterns of the promoter regions of the gene were analyzed by bisulfite-sequencing. Consequently, 2 regions that were methylated before 5-Aza-2'-deoxycytidine treatment were demethylated in LN319 after the treatment, while those regions were persistently demethylated in AS. These 2 regions corresponded with sites defined as promoter regions by Luciferase assay. Taken together, it was suggested that ST8SIA1 gene is regulated by DNA methylation at the promoter regions, leading to the regulation of tumor phenotypes.
Collapse
|
17
|
Cao S, Hu X, Ren S, Wang Y, Shao Y, Wu K, Yang Z, Yang W, He G, Li X. The biological role and immunotherapy of gangliosides and GD3 synthase in cancers. Front Cell Dev Biol 2023; 11:1076862. [PMID: 36824365 PMCID: PMC9941352 DOI: 10.3389/fcell.2023.1076862] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2022] [Accepted: 01/26/2023] [Indexed: 02/10/2023] Open
Abstract
Gangliosides are a large subfamily of glycosphingolipids that broadly exist in the nervous system and interact with signaling molecules in the lipid rafts. GD3 and GD2 are two types of disialogangliosides (GDs) that include two sialic acid residues. The expression of GD3 and GD2 in various cancers is mostly upregulated and is involved in tumor proliferation, invasion, metastasis, and immune responses. GD3 synthase (GD3S, ST8SiaI), a subclass of sialyltransferases, regulates the biosynthesis of GD3 and GD2. GD3S is also upregulated in most tumors and plays an important role in the development and progression of tumors. Many clinical trials targeting GD2 are ongoing and various immunotherapy studies targeting gangliosides and GD3S are gradually attracting much interest and attention. This review summarizes the function, molecular mechanisms, and ongoing clinical applications of GD3, GD2, and GD3S in abundant types of tumors, which aims to provide novel targets for future cancer therapy.
Collapse
Affiliation(s)
- Shangqi Cao
- 1Department of Urology, Institute of Urology, West China Hospital, West China Medical School, Sichuan University, Chengdu, China
| | - Xu Hu
- 1Department of Urology, Institute of Urology, West China Hospital, West China Medical School, Sichuan University, Chengdu, China
| | - Shangqing Ren
- 2Robotic Minimally Invasive Surgery Center, Sichuan Academy of Medical Sciences and Sichuan Provincial Peoples Hospital, Chengdu, China
| | - Yaohui Wang
- 1Department of Urology, Institute of Urology, West China Hospital, West China Medical School, Sichuan University, Chengdu, China
| | - Yanxiang Shao
- 1Department of Urology, Institute of Urology, West China Hospital, West China Medical School, Sichuan University, Chengdu, China
| | - Kan Wu
- 1Department of Urology, Institute of Urology, West China Hospital, West China Medical School, Sichuan University, Chengdu, China
| | - Zhen Yang
- 3Department of Urology, Chengdu Second People’s Hospital, Chengdu, China
| | - Weixiao Yang
- 1Department of Urology, Institute of Urology, West China Hospital, West China Medical School, Sichuan University, Chengdu, China
| | - Gu He
- 4State Key Laboratory of Biotherapy and Department of Pharmacy, West China Hospital, Sichuan University and Collaborative Innovation Center of Biotherapy, Chengdu, Sichuan, China,*Correspondence: Gu He, ; Xiang Li,
| | - Xiang Li
- 1Department of Urology, Institute of Urology, West China Hospital, West China Medical School, Sichuan University, Chengdu, China,*Correspondence: Gu He, ; Xiang Li,
| |
Collapse
|
18
|
Yang J, Han L, Sha Y, Jin Y, Li Z, Gong B, Li J, Liu Y, Wang Y, Zhao Q. A novel ganglioside-related risk signature can reveal the distinct immune landscape of neuroblastoma and predict the immunotherapeutic response. Front Immunol 2022; 13:1061814. [PMID: 36605200 PMCID: PMC9807785 DOI: 10.3389/fimmu.2022.1061814] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2022] [Accepted: 12/05/2022] [Indexed: 12/24/2022] Open
Abstract
Introduction Gangliosides play an essential role in cancer development and progression. However, the involvement of gangliosides in the prognosis and tumor microenvironment (TME) of neuroblastoma is not entirely understood. Methods Consensus clustering analysis was performed to identify ganglioside-mediated molecular subtypes. LASSO-Cox analysis was conducted to identify independent prognostic genes, and a novel risk signature was constructed. The risk signature was validated internally and externally. We further explored the independent prognosis value, immune landscape, drug susceptibility, and tumor dedifferentiation of the risk signature. The role of the signature gene B3GALT4 in neuroblastoma was explored in vitro. Results Seventeen ganglioside-related genes were differentially expressed between INSS stage 4 and other stages, and two ganglioside-related clusters with distinct prognoses were identified. A novel risk signature integrating ten ganglioside-related prognostic genes was established. Across the train set and external validation sets, the risk signature presented high predictive accuracy and discrimination. The risk signature was an independent prognostic factor and constructed a nomogram combining multiple clinical characteristics. In the high-score group, the deficiency in antigen processing and presenting machinery, lack of immune cell infiltration, and escaping NK cells contributed substantially to immune escape. The low-score group was more responsive to immune checkpoint blockade therapy, while the high-score group showed substantial sensitivity to multiple chemotherapeutic drugs. Besides, the risk score was significantly positively correlated with the stemness index and reduced considerably in all-trans retinoic acid-treated neuroblastoma cell lines, indicating high dedifferentiation in the high-score group. Additionally, neuroblastoma cells with downregulation of B3GALT4 present with increased proliferation, invasion, and metastasis abilities in vitro. Conclusion The novel ganglioside-related risk signature highlights the role of ganglioside in neuroblastoma prognosis and immune landscape and helps optimize chemotherapy and immunotherapy for neuroblastoma.
Collapse
Affiliation(s)
- Jiaxing Yang
- Department of Pediatric Oncology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
- National Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
- Key Laboratory of Cancer Prevention and Therapy, Tianjin, China
- Tianjin’s Clinical Research Center for Cancer, Tianjin, China
| | - Lei Han
- National Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
- Key Laboratory of Cancer Prevention and Therapy, Tianjin, China
- Tianjin’s Clinical Research Center for Cancer, Tianjin, China
- Department of Cancer Molecular Diagnostics Core, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Yongliang Sha
- Department of Pediatric Oncology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
- National Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
- Key Laboratory of Cancer Prevention and Therapy, Tianjin, China
- Tianjin’s Clinical Research Center for Cancer, Tianjin, China
| | - Yan Jin
- Department of Pediatric Oncology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
- National Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
- Key Laboratory of Cancer Prevention and Therapy, Tianjin, China
- Tianjin’s Clinical Research Center for Cancer, Tianjin, China
| | - Zhongyuan Li
- Department of Pediatric Oncology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
- National Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
- Key Laboratory of Cancer Prevention and Therapy, Tianjin, China
- Tianjin’s Clinical Research Center for Cancer, Tianjin, China
| | - Baocheng Gong
- Department of Pediatric Oncology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
- National Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
- Key Laboratory of Cancer Prevention and Therapy, Tianjin, China
- Tianjin’s Clinical Research Center for Cancer, Tianjin, China
| | - Jie Li
- Department of Pediatric Oncology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
- National Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
- Key Laboratory of Cancer Prevention and Therapy, Tianjin, China
- Tianjin’s Clinical Research Center for Cancer, Tianjin, China
| | - Yun Liu
- Department of Pediatric Oncology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
- National Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
- Key Laboratory of Cancer Prevention and Therapy, Tianjin, China
- Tianjin’s Clinical Research Center for Cancer, Tianjin, China
| | - Yangyang Wang
- Department of Pediatric Oncology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
- National Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
- Key Laboratory of Cancer Prevention and Therapy, Tianjin, China
- Tianjin’s Clinical Research Center for Cancer, Tianjin, China
| | - Qiang Zhao
- Department of Pediatric Oncology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
- National Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
- Key Laboratory of Cancer Prevention and Therapy, Tianjin, China
- Tianjin’s Clinical Research Center for Cancer, Tianjin, China
| |
Collapse
|
19
|
Inagaki FF, Kato T, Furusawa A, Okada R, Wakiyama H, Furumoto H, Okuyama S, Choyke PL, Kobayashi H. Disialoganglioside GD2-Targeted Near-Infrared Photoimmunotherapy (NIR-PIT) in Tumors of Neuroectodermal Origin. Pharmaceutics 2022; 14:2037. [PMID: 36297471 PMCID: PMC9612122 DOI: 10.3390/pharmaceutics14102037] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Revised: 09/18/2022] [Accepted: 09/22/2022] [Indexed: 10/21/2023] Open
Abstract
Disialoganglioside (GD2) is a subtype of glycolipids that is highly expressed in tumors of neuroectodermal origins, such as neuroblastoma and osteosarcoma. Its limited expression in normal tissues makes GD2 a potential target for precision therapy. Several anti-GD2 monoclonal antibodies are currently in clinical use and have had moderate success. Near-infrared photoimmunotherapy (NIR-PIT) is a cancer therapy that arms antibodies with IRDye700DX (IR700) and then exposes this antibody-dye conjugate (ADC) to NIR light at a wavelength of 690 nm. NIR light irradiation induces a profound photochemical response in IR700, resulting in protein aggregates that lead to cell membrane damage and death. In this study, we examined the feasibility of GD2-targeted NIR-PIT. Although GD2, like other glycolipids, is only located in the outer leaflet of the cell membrane, the aggregates formation exerted sufficient physical force to disrupt the cell membrane and kill target cells in vitro. In in vivo studies, tumor growth was significantly inhibited after GD2-targeted NIR-PIT, resulting in prolonged survival. Following GD2-targeted NIR-PIT, activation of host immunity was observed. In conclusion, GD2-targeted NIR-PIT was similarly effective to the conventional protein-targeted NIR-PIT. This study demonstrates that membrane glycolipid can be a new target of NIR-PIT.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Hisataka Kobayashi
- Molecular Imaging Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| |
Collapse
|
20
|
Attenuation of Bone Formation through a Decrease in Osteoblasts in Mutant Mice Lacking the GM2/GD2 Synthase Gene. Int J Mol Sci 2022; 23:ijms23169044. [PMID: 36012308 PMCID: PMC9409452 DOI: 10.3390/ijms23169044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2022] [Revised: 08/05/2022] [Accepted: 08/11/2022] [Indexed: 11/17/2022] Open
Abstract
The ganglioside GD1a has been reported to promote the differentiation of mesenchymal stem cells to osteoblasts in cell culture systems. However, the involvement of gangliosides, including GD1a, in bone formation in vivo remains unknown; therefore, we herein investigated their roles in GM2/GD2 synthase-knockout (GM2/GD2S KO) mice without GD1a. The femoral cancellous bone mass was analyzed using three-dimensional micro-computed tomography. A histomorphometric analysis of bone using hematoxylin and eosin (HE) and tartrate-resistant acid phosphatase was performed to examine bone formation and resorption, respectively. Calcein double labeling was also conducted to evaluate bone formation. Although no significant differences were observed in bone mass or resorption between GM2/GD2S KO mice and wild-type (WT) mice, analyses of the parameters of bone formation using HE staining and calcein double labeling revealed less bone formation in GM2/GD2S KO mice than in WT mice. These results suggest that gangliosides play roles in bone formation.
Collapse
|
21
|
Furukawa K, Ohmi Y, Hamamura K, Kondo Y, Ohkawa Y, Kaneko K, Hashimoto N, Yesmin F, Bhuiyan RH, Tajima O, Furukawa K. Signaling domains of cancer-associated glycolipids. Glycoconj J 2022; 39:145-155. [PMID: 35315508 DOI: 10.1007/s10719-022-10051-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2021] [Revised: 02/06/2022] [Accepted: 02/15/2022] [Indexed: 12/16/2022]
Abstract
Immunotherapy of malignant cancers is now becoming one of representative approaches to overcome cancers. To construct strategies for immunotherapy, presence of tumor-specific antigens should be a major promise. A number of cancer specific- or cancer-associated antigens have been reported based on various experimental sets and various animal systems. The most reasonable strategy to define tumor-specific antigens might be "autologous typing" performed by Old's group, proposing three classes of tumor-antigens recognized by host immune systems of cancer patients. Namely, class 1, individual antigens that is present only in the patient's sample analyzed; class 2, shared antigens that can be found only in some group of cancers in some patients, but not in normal cells and tissues; class 3, universal antigens that are present in some cancers but also in normal cells and tissues with different densities. Sen Hakomori reported there were novel carbohydrates in cancers that could not be detected in normal cells mainly by biochemical approaches. Consequently, many of class 2 cancer-specific antigens have been revealed to be carbohydrate antigens, and been used for cancer diagnosis and treatment. Not only as cancer markers, but roles of those cancer-associated carbohydrates have also been recognized as functional molecules in cancer cells. In particular, roles of complex carbohydrates in the regulation of cell signaling on the cell surface microdomains, glycolipid-enriched microdomain (GEM)/rafts have been reported by Hakomori and many other researchers including us. The processes and present status of these studies on cancer-associated glycolipids were summarized.
Collapse
Affiliation(s)
- Koichi Furukawa
- Department of Biomedical Sciences, Chubu University College of Life and Health Sciences, Kasugai, Japan. .,Department of Molecular and Cellular Biochemistry, Nagoya University Graduate School of Medicine, Nagoya, Japan.
| | - Yuhsuke Ohmi
- Department of Clinical Engineering, Chubu University College of Life and Health Sciences, Kasugai, Japan
| | - Kazunori Hamamura
- Department of Pharmacology, Aichi Gakuin University School of Dentistry, Nisshin, Japan
| | - Yuji Kondo
- Department of Molecular and Cellular Biochemistry, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Yuki Ohkawa
- Department of Glyco-Oncology and Medical Biochemistry, Osaka International Institute, Osaka, Japan
| | - Kei Kaneko
- Department of Biomedical Sciences, Chubu University College of Life and Health Sciences, Kasugai, Japan
| | - Noboru Hashimoto
- Department of Tissue Regeneration, Tokushima University Graduate School Institute of Biomedical Sciences, Tokushima, Japan
| | - Farhana Yesmin
- Department of Biomedical Sciences, Chubu University College of Life and Health Sciences, Kasugai, Japan.,Department of Molecular and Cellular Biochemistry, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Robiul H Bhuiyan
- Department of Biomedical Sciences, Chubu University College of Life and Health Sciences, Kasugai, Japan
| | - Orie Tajima
- Department of Biomedical Sciences, Chubu University College of Life and Health Sciences, Kasugai, Japan
| | - Keiko Furukawa
- Department of Biomedical Sciences, Chubu University College of Life and Health Sciences, Kasugai, Japan
| |
Collapse
|
22
|
Glycosphingolipids in human embryonic stem cells and breast cancer stem cells, and potential cancer therapy strategies based on their structures and functions. Glycoconj J 2022; 39:177-195. [PMID: 35267131 DOI: 10.1007/s10719-021-10032-w] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2021] [Revised: 11/27/2021] [Accepted: 12/08/2021] [Indexed: 12/26/2022]
Abstract
Expression profiles of glycosphingolipids (GSLs) in human embryonic stem cell (hESC) lines and their differentiated embryoid body (EB) outgrowth cells, consisting of three germ layers, were surveyed systematically. Several globo- and lacto-series GSLs were identified in undifferentiated hESCs and during differentiation of hESCs to EB outgrowth cells, and core structure switching of these GSLs to gangliosides was observed. Such switching was attributable to altered expression of key glycosyltransferases (GTs) in GSL biosynthetic pathways, reflecting the unique stage-specific transitions and mechanisms characteristic of the differentiation process. Lineage-specific differentiation of hESCs was associated with further GSL alterations. During differentiation of undifferentiated hESCs to neural progenitor cells, core structure switching from globo- and lacto-series to primarily gangliosides (particularly GD3) was again observed. During differentiation to endodermal cells, alterations of GSL profiles were distinct from those in differentiation to EB outgrowth or neural progenitor cells, with high expression of Gb4Cer and low expression of stage-specific embryonic antigen (SSEA)-3, -4, or GD3 in endodermal cells. Again, such profile changes resulted from alterations of key GTs in GSL biosynthetic pathways. Novel glycan structures identified on hESCs and their differentiated counterparts presumably play functional roles in hESCs and related cancer or cancer stem cells, and will be useful as surface biomarkers. We also examined GSL expression profiles in breast cancer stem cells (CSCs), using a model of epithelial-mesenchymal transition (EMT)-induced human breast CSCs. We found that GD2 and GD3, together with their common upstream GTs, GD3 synthase (GD3S) and GD2/GM2 synthase, maintained stem cell phenotype in breast CSCs. Subsequent studies showed that GD3 was associated with epidermal growth factor receptor (EGFR), and activated EGFR signaling in breast CSCs and breast cancer cell lines. GD3S knockdown enhanced cytotoxicity of gefitinib (an EGFR kinase inhibitor) in resistant MDA-MB468 cells, both in vitro and in vivo. Our findings indicate that GD3S contributes to gefitinib resistance in EGFR-positive breast cancer cells, and is a potentially useful therapeutic target in drug-resistant breast cancers.
Collapse
|
23
|
Nishimaki H, Nakanishi Y, Yagasaki H, Masuda S. Multiple Immunofluorescence Imaging Analysis Reveals Differential Expression of Disialogangliosides GD3 and GD2 in Neuroblastomas. Pediatr Dev Pathol 2022; 25:141-154. [PMID: 34674560 DOI: 10.1177/10935266211048733] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND Peripheral neuroblastic tumors (pNTs) are the most common childhood extracranial solid tumors. There are several therapeutic strategies targeting disialoganglioside GD2. Disialoganglioside GD3 has become a potential target. However, the mechanism by which pNTs express GD3 and GD2 remains unclear. We investigated the combined expression status of GD3 and GD2 in pNTs and delineated their clinicopathological values. METHODS GD3 and GD2 expression was examined in pNT tissue samples (n = 35) using immunohistochemistry and multiple immunofluorescence imaging. RESULTS GD3 and GD2 expression was positive in 32/35 and 25/35 samples, respectively. Combinatorial analysis of GD3 and GD2 expression in neuroblastoma showed that both were heterogeneously expressed from cell to cell. There were higher numbers of GD3-positive and GD2-negative cells in the low-risk group than in the intermediate-risk (P = 0.014) and high-risk (P = 0.009) groups. Cases with high proportions of GD3-positive and GD2-negative cells were associated with the International Neuroblastoma Staging System stage (P = 0.004), Children's Oncology Group risk group (P = 0.001), and outcome (P = 0.019) and tended to have a higher overall survival rate. CONCLUSION We demonstrated that neuroblastomas from low-risk patients included more GD3-positive and GD2-negative cells than those from high-risk patients. Clarifying the heterogeneity of neuroblastoma aids in better understanding the biological characteristics and clinical behavior.
Collapse
Affiliation(s)
- Haruna Nishimaki
- Division of Oncologic Pathology, Department of Pathology and Microbiology, Nihon University School of Medicine, Itabashi-ku, Tokyo, Japan
| | - Yoko Nakanishi
- Division of Oncologic Pathology, Department of Pathology and Microbiology, Nihon University School of Medicine, Itabashi-ku, Tokyo, Japan
| | - Hiroshi Yagasaki
- Department of Pediatric and Child Health, Nihon University School of Medicine, Itabashi-ku, Tokyo, Japan
| | - Shinobu Masuda
- Division of Oncologic Pathology, Department of Pathology and Microbiology, Nihon University School of Medicine, Itabashi-ku, Tokyo, Japan
| |
Collapse
|
24
|
CAR T targets and microenvironmental barriers of osteosarcoma. Cytotherapy 2022; 24:567-576. [DOI: 10.1016/j.jcyt.2021.12.010] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Revised: 11/11/2021] [Accepted: 12/07/2021] [Indexed: 02/06/2023]
|
25
|
Yesmin F, Bhuiyan RH, Ohmi Y, Yamamoto S, Kaneko K, Ohkawa Y, Zhang P, Hamamura K, Cheung NKV, Kotani N, Honke K, Okajima T, Kambe M, Tajima O, Furukawa K, Furukawa K. Ganglioside GD2 Enhances the Malignant Phenotypes of Melanoma Cells by Cooperating with Integrins. Int J Mol Sci 2021; 23:ijms23010423. [PMID: 35008849 PMCID: PMC8745508 DOI: 10.3390/ijms23010423] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2021] [Revised: 12/12/2021] [Accepted: 12/28/2021] [Indexed: 11/16/2022] Open
Abstract
Gangliosides have been considered to modulate cell signals in the microdomain of the cell membrane, lipid/rafts, or glycolipid-enriched microdomain/rafts (GEM/rafts). In particular, cancer-associated gangliosides were reported to enhance the malignant properties of cancer cells. In fact, GD2-positive (GD2+) cells showed increased proliferation, invasion, and adhesion, compared with GD2-negative (GD2-) cells. However, the precise mechanisms by which gangliosides regulate cell signaling in GEM/rafts are not well understood. In order to analyze the roles of ganglioside GD2 in the malignant properties of melanoma cells, we searched for GD2-associating molecules on the cell membrane using the enzyme-mediated activation of radical sources combined with mass spectrometry, and integrin β1 was identified as a representative GD2-associating molecule. Then, we showed the physical association of GD2 and integrin β1 by immunoprecipitation/immunoblotting. Close localization was also shown by immuno-cytostaining and the proximity ligation assay. During cell adhesion, GD2+ cells showed multiple phospho-tyrosine bands, i.e., the epithelial growth factor receptor and focal adhesion kinase. The knockdown of integrin β1 revealed that the increased malignant phenotypes in GD2+ cells were clearly cancelled. Furthermore, the phosphor-tyrosine bands detected during the adhesion of GD2+ cells almost completely disappeared after the knockdown of integrin β1. Finally, immunoblotting to examine the intracellular distribution of integrins during cell adhesion revealed that large amounts of integrin β1 were localized in GEM/raft fractions in GD2+ cells before and just after cell adhesion, with the majority being localized in the non-raft fractions in GD2- cells. All these results suggest that GD2 and integrin β1 cooperate in GEM/rafts, leading to enhanced malignant phenotypes of melanomas.
Collapse
Affiliation(s)
- Farhana Yesmin
- Department of Biomedical Sciences, Chubu University College of Life and Health Sciences, Kasugai 487-8501, Japan; (F.Y.); (R.H.B.); (S.Y.); (K.K.); (Y.O.); (P.Z.); (M.K.); (O.T.); (K.F.)
- Department of Molecular Biochemistry, Nagoya University Graduate School of Medicine, Nagoya 466-0065, Japan;
| | - Robiul H. Bhuiyan
- Department of Biomedical Sciences, Chubu University College of Life and Health Sciences, Kasugai 487-8501, Japan; (F.Y.); (R.H.B.); (S.Y.); (K.K.); (Y.O.); (P.Z.); (M.K.); (O.T.); (K.F.)
| | - Yuhsuke Ohmi
- Department of Medical Technology, Chubu University College of Life and Health Sciences, Kasugai 487-8501, Japan;
| | - Satoko Yamamoto
- Department of Biomedical Sciences, Chubu University College of Life and Health Sciences, Kasugai 487-8501, Japan; (F.Y.); (R.H.B.); (S.Y.); (K.K.); (Y.O.); (P.Z.); (M.K.); (O.T.); (K.F.)
| | - Kei Kaneko
- Department of Biomedical Sciences, Chubu University College of Life and Health Sciences, Kasugai 487-8501, Japan; (F.Y.); (R.H.B.); (S.Y.); (K.K.); (Y.O.); (P.Z.); (M.K.); (O.T.); (K.F.)
| | - Yuki Ohkawa
- Department of Biomedical Sciences, Chubu University College of Life and Health Sciences, Kasugai 487-8501, Japan; (F.Y.); (R.H.B.); (S.Y.); (K.K.); (Y.O.); (P.Z.); (M.K.); (O.T.); (K.F.)
- Department of Glyco-Oncology and Medical Biochemistry, Osaka International Cancer Institute, Osaka 541-8567, Japan
| | - Pu Zhang
- Department of Biomedical Sciences, Chubu University College of Life and Health Sciences, Kasugai 487-8501, Japan; (F.Y.); (R.H.B.); (S.Y.); (K.K.); (Y.O.); (P.Z.); (M.K.); (O.T.); (K.F.)
- Department of Molecular Biochemistry, Nagoya University Graduate School of Medicine, Nagoya 466-0065, Japan;
| | - Kazunori Hamamura
- Department of Pharmacology, Aichi Gakuin University School of Dentistry, Nagoya 464-8650, Japan;
| | | | - Norihiro Kotani
- Department of Biochemistry, Saitama Medical University, Saitama 350-0495, Japan;
| | - Koichi Honke
- Department of Biochemistry, Kochi University School of Medicine, Nangoku 783-8505, Japan;
| | - Tetsuya Okajima
- Department of Molecular Biochemistry, Nagoya University Graduate School of Medicine, Nagoya 466-0065, Japan;
| | - Mariko Kambe
- Department of Biomedical Sciences, Chubu University College of Life and Health Sciences, Kasugai 487-8501, Japan; (F.Y.); (R.H.B.); (S.Y.); (K.K.); (Y.O.); (P.Z.); (M.K.); (O.T.); (K.F.)
| | - Orie Tajima
- Department of Biomedical Sciences, Chubu University College of Life and Health Sciences, Kasugai 487-8501, Japan; (F.Y.); (R.H.B.); (S.Y.); (K.K.); (Y.O.); (P.Z.); (M.K.); (O.T.); (K.F.)
| | - Keiko Furukawa
- Department of Biomedical Sciences, Chubu University College of Life and Health Sciences, Kasugai 487-8501, Japan; (F.Y.); (R.H.B.); (S.Y.); (K.K.); (Y.O.); (P.Z.); (M.K.); (O.T.); (K.F.)
| | - Koichi Furukawa
- Department of Biomedical Sciences, Chubu University College of Life and Health Sciences, Kasugai 487-8501, Japan; (F.Y.); (R.H.B.); (S.Y.); (K.K.); (Y.O.); (P.Z.); (M.K.); (O.T.); (K.F.)
- Department of Molecular Biochemistry, Nagoya University Graduate School of Medicine, Nagoya 466-0065, Japan;
- Correspondence: ; Tel.: +81-568-51-9512
| |
Collapse
|
26
|
Piazzesi A, Afsar SY, van Echten‐Deckert G. Sphingolipid metabolism in the development and progression of cancer: one cancer's help is another's hindrance. Mol Oncol 2021; 15:3256-3279. [PMID: 34289244 PMCID: PMC8637577 DOI: 10.1002/1878-0261.13063] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Revised: 06/17/2021] [Accepted: 07/19/2021] [Indexed: 11/27/2022] Open
Abstract
Cancer development is a multistep process in which cells must overcome a series of obstacles before they can become fully developed tumors. First, cells must develop the ability to proliferate unchecked. Once this is accomplished, they must be able to invade the neighboring tissue, as well as provide themselves with oxygen and nutrients. Finally, they must acquire the ability to detach from the newly formed mass in order to spread to other tissues, all the while evading an immune system that is primed for their destruction. Furthermore, increased levels of inflammation have been shown to be linked to the development of cancer, with sites of chronic inflammation being a common component of tumorigenic microenvironments. In this Review, we give an overview of the impact of sphingolipid metabolism in cancers, from initiation to metastatic dissemination, as well as discussing immune responses and resistance to treatments. We explore how sphingolipids can either help or hinder the progression of cells from a healthy phenotype to a cancerous one.
Collapse
Affiliation(s)
- Antonia Piazzesi
- LIMES Institute for Membrane Biology and Lipid BiochemistryUniversity of BonnGermany
| | - Sumaiya Yasmeen Afsar
- LIMES Institute for Membrane Biology and Lipid BiochemistryUniversity of BonnGermany
| | | |
Collapse
|
27
|
Mechanisms, Characteristics, and Treatment of Neuropathic Pain and Peripheral Neuropathy Associated with Dinutuximab in Neuroblastoma Patients. Int J Mol Sci 2021; 22:ijms222312648. [PMID: 34884452 PMCID: PMC8657961 DOI: 10.3390/ijms222312648] [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: 09/30/2021] [Revised: 11/13/2021] [Accepted: 11/18/2021] [Indexed: 12/21/2022] Open
Abstract
Prognosis of metastatic neuroblastoma is very poor. Its treatment includes induction chemotherapy, surgery, high-dose chemotherapy, radiotherapy, and maintenance with retinoic acid, associated with the anti-GD2 monoclonal antibody (ch14.18) dinutuximab. Immunotherapy determined a significant improvement in survival rate and is also utilized in relapsed and resistant neuroblastoma patients. Five courses of dinutuximab 100 mg/m2 are usually administered as a 10-day continuous infusion or over 5 consecutive days every 5 weeks. Dinutuximab targets the disialoganglioside GD2, which is highly expressed on neuroblastoma cells and minimally present on the surface of normal human neurons, peripheral pain fibers, and skin melanocytes. Anti GD2 antibodies bind to surface GD2 and determine the lysis of neuroblastoma cells induced by immune response via the antibody-dependent cellular cytotoxicity and the complement-dependent cytotoxicity. Dinutuximab has significant side effects, including neuropathic pain, peripheral neuropathy, hypersensitivity reactions, capillary leak syndrome, photophobia, and hypotension. The most important side effect is neuropathic pain, which is triggered by the same antibody–antigen immune response, but generates ectopic activity in axons, which results in hyperalgesia and spontaneous pain. Pain can be severe especially in the first courses of dinutuximab infusion, and requires the administration of gabapentin and continuous morphine infusion. This paper will focus on the incidence, mechanisms, characteristics, and treatment of neuropathic pain and peripheral neuropathy due to dinutuximab administration in neuroblastoma patients.
Collapse
|
28
|
Wiebel M, Kailayangiri S, Altvater B, Meltzer J, Grobe K, Kupich S, Rossig C. Surface expression of the immunotherapeutic target G D2 in osteosarcoma depends on cell confluency. Cancer Rep (Hoboken) 2021; 4:e1394. [PMID: 33811471 PMCID: PMC8551999 DOI: 10.1002/cnr2.1394] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Revised: 03/03/2021] [Accepted: 03/25/2021] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND Chimeric antigen receptor (CAR) T-cell therapy of pediatric sarcomas is challenged by the paucity of targetable cell surface antigens. A candidate target in osteosarcoma (OS) is the ganglioside GD2 , but heterogeneous expression of GD2 limits its value. AIM We aimed to identify mechanisms that upregulate GD2 target expression in OS. METHODS AND RESULTS GD2 surface expression in OS cells, studied by flow cytometry, was found to vary both among and within individual OS cell lines. Pharmacological approaches, including inhibition of the histone methyltransferase Enhancer of Zeste Homolog 2 (EZH2) and modulation of the protein kinase C, failed to increase GD2 expression. Instead, cell confluency was found to be associated with higher GD2 expression levels both in monolayer cultures and in tumor spheroids. The sensitivity of OS cells to targeting by GD2 -specific CAR T cells was compared in an in vitro cytotoxicity assay. Higher cell confluencies enhanced the sensitivity of OS cells to GD2 -antigen specific, CAR T-cell-mediated in vitro cytolysis. Mechanistic studies revealed that confluency-dependent upregulation of GD2 expression in OS cells is mediated by increased de novo biosynthesis, through a yet unknown mechanism. CONCLUSION Expression of GD2 in OS cell lines is highly variable and associated with increasing cell confluency in vitro. Strategies for selective upregulation of GD2 are needed to enable effective therapeutic targeting of this antigen in OS.
Collapse
Affiliation(s)
- Malena Wiebel
- Department of Pediatric Hematology and OncologyUniversity Children's Hospital MuensterMuensterGermany
| | - Sareetha Kailayangiri
- Department of Pediatric Hematology and OncologyUniversity Children's Hospital MuensterMuensterGermany
| | - Bianca Altvater
- Department of Pediatric Hematology and OncologyUniversity Children's Hospital MuensterMuensterGermany
| | - Jutta Meltzer
- Department of Pediatric Hematology and OncologyUniversity Children's Hospital MuensterMuensterGermany
| | - Kay Grobe
- Institute of Physiological Chemistry and PathobiochemistryUniversity of MuensterMuensterGermany
| | - Sabine Kupich
- Institute of Physiological Chemistry and PathobiochemistryUniversity of MuensterMuensterGermany
| | - Claudia Rossig
- Department of Pediatric Hematology and OncologyUniversity Children's Hospital MuensterMuensterGermany
- Cells‐in‐Motion Cluster of Excellence (EXC 1003 ‐ CiM)University of MuensterMuensterGermany
| |
Collapse
|
29
|
Zhang P, Ohkawa Y, Yamamoto S, Momota H, Kato A, Kaneko K, Natsume A, Farhana Y, Ohmi Y, Okajima T, Bhuiyan RH, Wakabayashi T, Furukawa K, Furukawa K. St8sia1-deficiency in mice alters tumor environments of gliomas, leading to reduced disease severity. NAGOYA JOURNAL OF MEDICAL SCIENCE 2021; 83:535-549. [PMID: 34552288 PMCID: PMC8438004 DOI: 10.18999/nagjms.83.3.535] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/18/2020] [Accepted: 12/18/2020] [Indexed: 12/29/2022]
Abstract
Ganglioside GD3/GD2 are over-expressed in various neuroectoderm-derived tumors. Previous studies indicated that GD3 is involved in the enhancement of cancer properties such as rapid growth and increased invasiveness. However, little is known about the functions of GD3/GD2 in glioma cells and glioma microenvironments. To clarify the functions of GD3/GD2 in gliomas, we used a mouse glioma model based on the RCAS/Gtv-a system. At first, we compared the gliomas size between wild-type (WT) and GD3 synthase (GD3S) knockout (KO) mice, showing a less malignant histology and slower tumor growth in GD3S-KO mice than in WT mice. Immunohistochemistry of glioma sections from WT and GD3S-KO mice revealed that reactive microglia/macrophages showed different localization patterns between the two genetic types of mice. CD68+ cells were more frequently stained inside glioma tissues of GD3S-KO mice, while they were stained mainly around glioma tissues in WT mice. The number of CD68+ cells markedly increased in tumor tissues of GD3S-KO mice at 2 weeks after injection of transfectant DF-1 cells. Furthermore, CD68+ cells in GD3S(-/-) glioma tissues expressed higher levels of inducible nitric oxide synthase. We observed higher expression levels of pro-inflammatory cytokine genes in primary-cultured glioma cells of WT mice than in GD3S-KO mice. DNA microarray data also revealed differential expression levels of various cytokines and chemokines in glioma tissues between WT and GD3S-KO mice. These results suggest that expression of GD3S allows glioma cells to promote polarization of microglia/macrophages towards M2-like phenotypes by modulating the expression levels of chemokines and cytokines.
Collapse
Affiliation(s)
- Pu Zhang
- Department of Biomedical Sciences, Chubu University College of Life and Health Sciences, Kasugai, Japan.,Department of Biochemistry II, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Yuki Ohkawa
- Department of Biomedical Sciences, Chubu University College of Life and Health Sciences, Kasugai, Japan.,Department of Neurosurgery, Nagoya University Graduate School of Medicine, Nagoya, Japan.,Department of Glyco-Oncology and Medical Biochemistry, Osaka International Cancer Institute
| | - Satoko Yamamoto
- Department of Biomedical Sciences, Chubu University College of Life and Health Sciences, Kasugai, Japan
| | - Hiroyuki Momota
- Department of Neurosurgery, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Akira Kato
- Department of Neurosurgery, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Kei Kaneko
- Department of Biomedical Sciences, Chubu University College of Life and Health Sciences, Kasugai, Japan
| | - Atsushi Natsume
- Department of Neurosurgery, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Yesmin Farhana
- Department of Biomedical Sciences, Chubu University College of Life and Health Sciences, Kasugai, Japan.,Department of Biochemistry II, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Yuhsuke Ohmi
- Department of Biomedical Sciences, Chubu University College of Life and Health Sciences, Kasugai, Japan
| | - Tetsuya Okajima
- Department of Biochemistry II, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Robiul H Bhuiyan
- Department of Biomedical Sciences, Chubu University College of Life and Health Sciences, Kasugai, Japan
| | - Toshihiko Wakabayashi
- Department of Neurosurgery, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Keiko Furukawa
- Department of Biomedical Sciences, Chubu University College of Life and Health Sciences, Kasugai, Japan
| | - Koichi Furukawa
- Department of Biomedical Sciences, Chubu University College of Life and Health Sciences, Kasugai, Japan.,Department of Biochemistry II, Nagoya University Graduate School of Medicine, Nagoya, Japan
| |
Collapse
|
30
|
Liu YJ, Li JP, Zhang Y, Nie MJ, Zhang YH, Liu SL, Zou X. FSTL3 is a Prognostic Biomarker in Gastric Cancer and is Correlated with M2 Macrophage Infiltration. Onco Targets Ther 2021; 14:4099-4117. [PMID: 34262295 PMCID: PMC8274543 DOI: 10.2147/ott.s314561] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Accepted: 06/22/2021] [Indexed: 12/31/2022] Open
Abstract
Purpose Follistatin-related gene 3 (FSTL3), an established oncogene, can modulate target gene expression via members of the transforming growth factor β (TGF-β) superfamily. The present study was conducted to evaluate the expression of FSTL3 in gastric cancer (GC) and to determine its prognostic significance. We also evaluated the possible mechanisms involved in the oncogenic role of FSTL3 in gastric carcinogenesis and development. Methods We obtained data from the Human Protein Atlas, MethSurv, cBioPortal, UALCAN, TIMER, GEPIA, STRING, GeneMANIA, ONCOMINE, and MEXPRESS databases and examined it using R software. RNAi was used to establish stable FSTL3-knockdown (shFSTL3) and overexpression (OE) cell strains. Western blot; enzyme-linked immunosorbent (ELISA); and immunohistochemical (ICH), immunofluorescence, and phalloidin staining were used for examining protein expression. Cell invasion and migration were determined using transwell and scratch-wound assays. After tumor-associated macrophage (TAM) generation, co-culturing of cancer cells with TAMs was performed to confirm the relationship between FSTL3 and TAMs. Results In GC patients, FSTL3 mRNA and protein levels were upregulated. FSTL3 expression was significantly linked to cancer stage as well as to pathological tumor grade in GC. Moreover, a high expression of FSTL3 was associated with a dismal survival duration in patients with GC. Furthermore, functional enrichment analysis demonstrated that FSTL3 overexpression could activate epithelial-mesenchymal transition (EMT) by promoting F-actin expression and BMP/SMAD signaling. Finally, immunofluorescence staining confirmed that the overexpression of FSTL3 promoted the proliferation of M2 TAMs. Conclusion Taken together, our findings suggest that FSTL3 may be involved in GC progression via the promotion of BMP/SMAD signaling-mediated EMT and M2 macrophage activation.
Collapse
Affiliation(s)
- Yuan-Jie Liu
- Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine, Nanjing, Jiangsu, 210029, People's Republic of China.,No. 1 Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, 210023, People's Republic of China
| | - Jie-Pin Li
- Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine, Nanjing, Jiangsu, 210029, People's Republic of China.,No. 1 Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, 210023, People's Republic of China.,Department of Oncology, Zhangjiagang TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Zhangjiagang, Jiangsu, 215600, People's Republic of China
| | - Ying Zhang
- Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine, Nanjing, Jiangsu, 210029, People's Republic of China.,No. 1 Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, 210023, People's Republic of China
| | - Meng-Jun Nie
- Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine, Nanjing, Jiangsu, 210029, People's Republic of China.,No. 1 Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, 210023, People's Republic of China
| | - Yong-Hua Zhang
- Department of Oncology, Zhangjiagang TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Zhangjiagang, Jiangsu, 215600, People's Republic of China
| | - Shen-Lin Liu
- Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine, Nanjing, Jiangsu, 210029, People's Republic of China.,No. 1 Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, 210023, People's Republic of China
| | - Xi Zou
- Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine, Nanjing, Jiangsu, 210029, People's Republic of China
| |
Collapse
|
31
|
Groux-Degroote S, Foulquier F, Cavdarli S, Delannoy P. [Reticular and Golgi glycosylation: Advances and associated diseases]. Med Sci (Paris) 2021; 37:609-617. [PMID: 34180820 DOI: 10.1051/medsci/2021082] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
Glycosylation is one of the essential modifications of proteins and lipids. It is carried out mainly in the endoplasmic reticulum and Golgi apparatus, and requires a specific molecular machinery associating several hundreds of glycosyltransferases, glycosidases, transporters and regulating proteins. Modifications of glycosylation are found in numerous diseases, notably in cancers. All types of glycosylation can be affected and this leads to dysfunctions of cellular metabolism. In this review, we present the current knowledge on the regulation of glycosylation mechanisms and illustrate how the alteration of these regulatory mechanisms can lead to abnormal protein and lipid glycosylation, and take part in the development of cancers.
Collapse
Affiliation(s)
- Sophie Groux-Degroote
- Univ. Lille, CNRS, UMR 8576 - UGSF - Unité de glycobiologie structurale et fonctionnelle, Avenue Mendeleïev, 59655 Villeneuve-d'Ascq, France
| | - François Foulquier
- Univ. Lille, CNRS, UMR 8576 - UGSF - Unité de glycobiologie structurale et fonctionnelle, Avenue Mendeleïev, 59655 Villeneuve-d'Ascq, France
| | - Sumeyye Cavdarli
- Univ. Lille, CNRS, UMR 8576 - UGSF - Unité de glycobiologie structurale et fonctionnelle, Avenue Mendeleïev, 59655 Villeneuve-d'Ascq, France
| | - Philippe Delannoy
- Univ. Lille, CNRS, UMR 8576 - UGSF - Unité de glycobiologie structurale et fonctionnelle, Avenue Mendeleïev, 59655 Villeneuve-d'Ascq, France
| |
Collapse
|
32
|
Gangliosides as Signaling Regulators in Cancer. Int J Mol Sci 2021; 22:ijms22105076. [PMID: 34064863 PMCID: PMC8150402 DOI: 10.3390/ijms22105076] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2021] [Revised: 05/07/2021] [Accepted: 05/07/2021] [Indexed: 02/06/2023] Open
Abstract
At the plasma membrane, gangliosides, a group of glycosphingolipids, are expressed along with glycosphingolipids, phospholipids, and cholesterol in so-called lipid rafts that interact with signaling receptors and related molecules. Most cancers present abnormalities in the intracellular signal transduction system involved in tumor growth, invasion, and metastasis. To date, the roles of gangliosides as regulators of signal transduction have been reported in several cancer types. Gangliosides can be expressed by the exogenous ganglioside addition, with their endogenous expression regulated at the enzymatic level by targeting specific glycosyltransferases. Accordingly, the relationship between changes in the composition of cell surface gangliosides and signal transduction has been investigated by controlling ganglioside expression. In cancer cells, several types of signaling molecules are positively or negatively regulated by ganglioside expression levels, promoting malignant properties. Moreover, antibodies against gangliosides have been shown to possess cytotoxic effects on ganglioside-expressing cancer cells. In the present review, we highlight the involvement of gangliosides in the regulation of cancer cell signaling, and we explore possible therapies targeting ganglioside-expressing cancer.
Collapse
|
33
|
Sarbu M, Petrica L, Clemmer DE, Vukelić Ž, Zamfir AD. Gangliosides of Human Glioblastoma Multiforme: A Comprehensive Mapping and Structural Analysis by Ion Mobility Tandem Mass Spectrometry. JOURNAL OF THE AMERICAN SOCIETY FOR MASS SPECTROMETRY 2021; 32:1249-1257. [PMID: 33900081 DOI: 10.1021/jasms.1c00088] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
Glioblastoma multiforme (GBM), a malignant, highly aggressive, grade IV brain tumor, which rapidly infiltrates into the nearby tissue, has drawn a significant amount of attention because of its poor prognosis and the limited treatment options available. In GBM, nearly all tumor cells exhibit aberrant cell-surface glycosylation patterns due to the alteration of their biosynthesis or postsynthesis modification process. Since gangliosides (GGs) are acknowledged as tumor-associated antigens, we have carried out here a comprehensive profiling of native ganglioside mixtures extracted and purified from GBM specimens. For this purpose, high performance ion mobility separation mass spectrometry (IMS MS) was thoroughly optimized to allow the discovery of GBM-specific structures and the assessment of their roles as tumor markers or possible associated antigens. GG separation by IMS according to the charge state, carbohydrate chain length, degree of sialylation, and ceramide composition led to the identification of no less than 160 distinct components, which represents 3-fold the number of structures identified before. The detected GGs and asialo-GGs were found characterized by a high heterogeneity in their ceramide and glycan compositions, encompassing up five Neu5Ac residues. The tumor was found dominated in equal and high proportions by GD3 and GT1 forms, with a particular incidence of C24:1 fatty acids in the ceramide. By the occurrence of only one mobility feature and the diagnostic fragment ions, the IMS tandem MS conducted using collision-induced dissociation (CID) disclosed for the first time the presence of GT1c(d18:1/24:1) newly proposed here as a potential GBM marker.
Collapse
Affiliation(s)
- Mirela Sarbu
- Department of Condensed Matter, National Institute for Research and Development in Electrochemistry and Condensed Matter, 300224, Timisoara, Romania
- Faculty of Physics, West University of Timisoara, 300223, Timisoara, Romania
| | - Ligia Petrica
- Department of Internal Medicine II - Division of Nephrology, County Emergency Hospital Timisoara and Centers for Molecular Research in Nephrology and Vascular Diseases, Translational Research and Systems and Cognitive Research in Neuropsychiatric Pathology, Faculty of Medicine, "Victor Babes" University of Medicine and Pharmacy, 300041, Timisoara, Romania
| | - David E Clemmer
- Department of Chemistry, The College of Arts and Science, Indiana University, Bloomington, Indiana 47405-7102, United States
| | - Željka Vukelić
- Department of Chemistry and Biochemistry, Faculty of Medicine, University of Zagreb, Zagreb 10000, Croatia
| | - Alina D Zamfir
- Department of Condensed Matter, National Institute for Research and Development in Electrochemistry and Condensed Matter, 300224, Timisoara, Romania
- Department of Technical and Natural Sciences, "Aurel Vlaicu" University of Arad, 310130, Arad, Romania
| |
Collapse
|
34
|
Fasanya HO, Dopico PJ, Yeager Z, Fan ZH, Siemann DW. Using a combination of gangliosides and cell surface vimentin as surface biomarkers for isolating osteosarcoma cells in microfluidic devices. J Bone Oncol 2021; 28:100357. [PMID: 33912384 PMCID: PMC8065304 DOI: 10.1016/j.jbo.2021.100357] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2021] [Revised: 02/18/2021] [Accepted: 03/01/2021] [Indexed: 12/28/2022] Open
Abstract
Background Osteosarcoma (OS) is the most common primary bone tumor and the third leading cause of pediatric cancer deaths. Liquid biopsies are an alternative to current diagnostic imaging modalities that can be used to monitor treatment efficacy and the development of metastases. This study addresses the use of novel biomarkers to detect circulating osteosarcoma cells. Procedures Flow cytometry was used to evaluate the relative expression of epithelial cell adhesion molecule (EpCAM), ganglioside 2 and 3 (GD2/3), and cell surface vimentin (CSV) on a panel of OS cell lines. A microfluidic device was used to affirm the efficacy of GD2/3 and CSV to capture CTCs. Once captured, CTCs on the device are enumerated and the capture efficiency for each marker is measured. Patient samples were captured using the LFAM chip. Results We report the evaluation of GD2, GD3, and CSV as markers for OS cell capture in cell lines and in patient samples. The results of our capture studies correlate with our flow cytometry data and have shown a low capture efficiency of OS cells using EpCAM antibodies, while showing a moderate capture efficiency of OS cells using the GD2, GD3, and CSV antibodies independently. The combination of biomarkers demonstrate a high capture efficiency of approximately 80%. This is further supported by the detection of 1-1.5 CTCs per mL of blood using GD2 + CSV in OS patient samples. Conclusions The combination of GD2 + CSV significantly increased the capture efficacy of OS cells. The detection of CTCs through routine blood sampling may be used clinically for earlier detection of metastases and monitoring the therapeutic effect of treatments in metastatic osteosarcomas.
Collapse
Key Words
- CK, Cytokeratin
- CSV, Cell Surface Vimentin
- CTC, Circulating Tumor Cell
- Circulating tumor cells
- DAPI, 4′,6-diamidino-2-phenylindole
- EpCAM, Epithelial Cell Adhesion Molecule
- GD2, Ganglioside 2
- GD3, Ganglioside 3
- Ganglioside GD2
- Ganglioside GD3
- IHC, Immunohistochemistry
- OS, Osteosarcoma
- Osteosarcoma
- PET, Positron Emission Tomography
- Vimentin
- mL, Milliliter
Collapse
Affiliation(s)
- Henrietta O. Fasanya
- Department of Radiation Oncology, University of Florida, Gainesville, FL, USA
- College of Medicine MD-PhD Program, University of Florida, Gainesville, FL, USA
- Corresponding authors at: Interdisciplinary Microsystems Group, Department of Mechanical and Aerospace Engineering, Gainesville, FL, USA (Z.H. Fan) Department of Radiation Oncology, University of Florida, Gainesville, FL, USA (H.O. Fasanya).
| | - Pablo J. Dopico
- Interdisciplinary Microsystems Group, Department of Mechanical and Aerospace Engineering, Gainesville, FL, USA
| | - Zachary Yeager
- Interdisciplinary Microsystems Group, Department of Mechanical and Aerospace Engineering, Gainesville, FL, USA
| | - Z. Hugh Fan
- J. Crayton Pruitt Family Department of Biomedical Engineering, Gainesville, FL, USA
- Corresponding authors at: Interdisciplinary Microsystems Group, Department of Mechanical and Aerospace Engineering, Gainesville, FL, USA (Z.H. Fan) Department of Radiation Oncology, University of Florida, Gainesville, FL, USA (H.O. Fasanya).
| | - Dietmar W. Siemann
- Department of Radiation Oncology, University of Florida, Gainesville, FL, USA
- Corresponding authors at: Interdisciplinary Microsystems Group, Department of Mechanical and Aerospace Engineering, Gainesville, FL, USA (Z.H. Fan) Department of Radiation Oncology, University of Florida, Gainesville, FL, USA (H.O. Fasanya).
| |
Collapse
|
35
|
Ly S, Anand V, El-Dana F, Nguyen K, Cai Y, Cai S, Piwnica-Worms H, Tripathy D, Sahin AA, Andreeff M, Battula VL. Anti-GD2 antibody dinutuximab inhibits triple-negative breast tumor growth by targeting GD2 + breast cancer stem-like cells. J Immunother Cancer 2021; 9:e001197. [PMID: 33722905 PMCID: PMC7970220 DOI: 10.1136/jitc-2020-001197] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/26/2021] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND Triple-negative breast cancer (TNBC) is the most aggressive breast cancer subtype with no effective standard therapy. Breast cancer stem-like cells (BCSCs) in primary TNBCs are reported to be responsible for metastatic spread of the disease and resistance to chemotherapy, but no available therapeutic tools target BCSCs. We previously reported that the ganglioside GD2 is highly expressed on BCSCs and that inhibition of its expression hampers TNBC growth. We therefore hypothesized that the anti-GD2 antibody dinutuximab (ch14.18) targets GD2+ BCSCs and inhibits TNBC growth. METHOD To test our hypothesis, we first determined GD2 expression via immunohistochemistry in frozen primary tumor samples from patients with TNBC (n=89). Then, we examined the effects of dinutuximab on TNBC cell adhesion, migration, and mammosphere formation in vitro and on tumor growth in vivo using TNBC cell-line and patient-derived xenograft (PDX) models. RESULTS We found that GD2 was expressed in around 60% of primary TNBC tumors at variable levels and was associated with worse overall survival of patients with TNBC (p=0.002). GD2 was found to be expressed in tumors and stroma, but normal ducts and lobules in adjacent tissues have shown low or no GD2 staining, indicating that GD2 is potentially a novel biomarker for tumor and its microenvironment. Treatment with dinutuximab significantly decreased adhesion and migration of MDA-MB-231 and SUM159 TNBC cells. Moreover, dinutuximab treatment inhibited mTOR signaling, which has been shown to be regulated by GD2 in BCSCs. Dinutuximab also reduced tumor growth in nude mice bearing TNBC cell-line xenografts. Finally, dinutuximab in combination with activated natural killer cells inhibited tumor growth in a TNBC PDX model and improved overall survival of tumor-bearing mice. CONCLUSIONS Dinutuximab successfully eliminated GD2+ cells and reduced tumor growth in both in vivo models. Our data provide proof-of-concept for the criticality of GD2 in BCSCs and demonstrate the potential of dinutuximab as a novel therapeutic approach for TNBC.
Collapse
Affiliation(s)
- Stanley Ly
- Leukemia, MD Anderson Cancer Center, The University of Texas, Houston, Texas, USA
| | - Vivek Anand
- Leukemia, MD Anderson Cancer Center, The University of Texas, Houston, Texas, USA
| | - Fouad El-Dana
- Leukemia, MD Anderson Cancer Center, The University of Texas, Houston, Texas, USA
| | - Khoa Nguyen
- Leukemia, MD Anderson Cancer Center, The University of Texas, Houston, Texas, USA
| | - Yiming Cai
- Leukemia, MD Anderson Cancer Center, The University of Texas, Houston, Texas, USA
| | - Shirong Cai
- Experimental Radiation Oncology, MD Anderson Cancer Center, The University of Texas, Houston, Texas, USA
| | - Helen Piwnica-Worms
- Experimental Radiation Oncology, MD Anderson Cancer Center, The University of Texas, Houston, Texas, USA
| | - Debasish Tripathy
- Breast Medical Oncology, Division of Cancer Medicine, MD Anderson Cancer Center, The University of Texas, Houston, Texas, USA
| | - Aysegul A Sahin
- Pathology, MD Anderson Cancer Center, The University of Texas, Houston, Texas, USA
| | - Michael Andreeff
- Leukemia, MD Anderson Cancer Center, The University of Texas, Houston, Texas, USA
| | - Venkata Lokesh Battula
- Leukemia, MD Anderson Cancer Center, The University of Texas, Houston, Texas, USA
- Breast Medical Oncology, Division of Cancer Medicine, MD Anderson Cancer Center, The University of Texas, Houston, Texas, USA
| |
Collapse
|
36
|
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: 13] [Impact Index Per Article: 4.3] [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
|
37
|
Park JA, Cheung NKV. GD2 or HER2 targeting T cell engaging bispecific antibodies to treat osteosarcoma. J Hematol Oncol 2020; 13:172. [PMID: 33303017 PMCID: PMC7731630 DOI: 10.1186/s13045-020-01012-y] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Accepted: 11/25/2020] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND The cure rate for metastatic osteosarcoma has not substantially improved over the past decades. Clinical trials of anti-HER2 trastuzumab or anti-GD2 dinutuximab for metastatic or refractory osteosarcoma were not successful, and neither was immune checkpoint inhibitors (ICIs). METHODS We tested various target antigen expressions on osteosarcoma cell lines using flow cytometry and analyzed in vitro T cell engaging BsAb (T-BsAb)-dependent T cell-mediated cytotoxicity using 4-h 51Cr release assay. We tested in vivo anti-tumor activities of T-BsAb targeting GD2 or HER2 in established osteosarcoma cell line or patient-derived xenograft (PDX) mouse models carried out in BALB-Rag2-/-IL-2R-γc-KO (BRG) mice. We also generated ex vivo BsAb-armed T cells (EATs) and studied their tumor-suppressive effect against osteosarcoma xenografts. In order to improve the anti-tumor response, ICIs, anti-human PD-1 (pembrolizumab) or anti-human PD-L1 (atezolizumab) antibodies were tested their synergy with GD2- or HER2-BsAb against osteosarcoma. RESULTS GD2 and HER2 were chosen from a panel of surface markers on osteosarcoma cell lines and PDXs. Anti-GD2 BsAb or anti-HER2 BsAb exerted potent anti-tumor effect against osteosarcoma tumors in vitro and in vivo. T cells armed with anti-GD2-BsAb (GD2-EATs) or anti-HER2-BsAb (HER2-EATs) showed significant anti-tumor activities as well. Anti-PD-L1 combination treatment enhanced BsAb-armed T cell function in vivo and improved tumor control and survival of the mice, when given sequentially and continuously. CONCLUSION Anti-GD2 and anti-HER2 BsAbs were effective in controlling osteosarcoma. These data support the clinical investigation of GD2 and HER2 targeted T-BsAb treatment in combination with immune checkpoint inhibitors, particularly anti-PD-L1, in patients with osteosarcoma to improve their treatment outcome.
Collapse
Affiliation(s)
- Jeong A Park
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, 1275 York Ave, Box 170, New York, NY, USA
| | - Nai-Kong V Cheung
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, 1275 York Ave, Box 170, New York, NY, USA.
| |
Collapse
|
38
|
Bartish M, Del Rincón SV, Rudd CE, Saragovi HU. Aiming for the Sweet Spot: Glyco-Immune Checkpoints and γδ T Cells in Targeted Immunotherapy. Front Immunol 2020; 11:564499. [PMID: 33133075 PMCID: PMC7550643 DOI: 10.3389/fimmu.2020.564499] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2020] [Accepted: 08/31/2020] [Indexed: 11/23/2022] Open
Abstract
Though a healthy immune system is capable of recognizing and eliminating emergent cancerous cells, an established tumor is adept at escaping immune surveillance. Altered and tumor-specific expression of immunosuppressive cell surface carbohydrates, also termed the “tumor glycocode,” is a prominent mechanism by which tumors can escape anti-tumor immunity. Given their persistent and homogeneous expression, tumor-associated glycans are promising targets to be exploited as biomarkers and therapeutic targets. However, the exploitation of these glycans has been a challenge due to their low immunogenicity, immunosuppressive properties, and the inefficient presentation of glycolipids in a conventional major histocompatibility complex (MHC)-restricted manner. Despite this, a subset of T-cells expressing the gamma and delta chains of the T-cell receptor (γδ T cells) exist with a capacity for MHC-unrestricted antigen recognition and potent inherent anti-tumor properties. In this review, we discuss the role of tumor-associated glycans in anti-tumor immunity, with an emphasis on the potential of γδ T cells to target the tumor glycocode. Understanding the many facets of this interaction holds the potential to unlock new ways to use both tumor-associated glycans and γδ T cells in novel therapeutic interventions.
Collapse
Affiliation(s)
- Margarita Bartish
- Lady Davis Institute, Jewish General Hospital, Translational Center for Research in Cancer, McGill University, Montreal, QC, Canada
| | - Sonia V Del Rincón
- Lady Davis Institute, Jewish General Hospital, Translational Center for Research in Cancer, McGill University, Montreal, QC, Canada.,Oncology and Experimental Medicine, McGill University, Montreal, QC, Canada
| | - Christopher E Rudd
- Division of Immuno-Oncology, Research Center Maisonneuve-Rosemont Hospital, Montreal, QC, Canada.,Département de Médecine, Université de Montréal, Montreal, QC, Canada
| | - H Uri Saragovi
- Lady Davis Institute, Jewish General Hospital, Translational Center for Research in Cancer, McGill University, Montreal, QC, Canada.,Oncology and Experimental Medicine, McGill University, Montreal, QC, Canada.,Pharmacology and Therapeutics, and Ophthalmology and Vision Sciences, McGill University, Montreal, QC, Canada
| |
Collapse
|
39
|
Schengrund CL. Gangliosides and Neuroblastomas. Int J Mol Sci 2020; 21:E5313. [PMID: 32726962 PMCID: PMC7432824 DOI: 10.3390/ijms21155313] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Revised: 07/09/2020] [Accepted: 07/18/2020] [Indexed: 12/19/2022] Open
Abstract
The focus of this review is the ganglio-series of glycosphingolipids found in neuroblastoma (NB) and the myriad of unanswered questions associated with their possible role(s) in this cancer. NB is one of the more common solid malignancies of children. Five-year survival for those diagnosed with low risk NB is 90-95%, while that for children with high-risk NB is around 40-50%. Much of the survival rate reflects age of diagnosis with children under a year having a much better prognosis than those over two. Identification of expression of GD2 on the surface of most NB cells led to studies of the effectiveness and subsequent approval of anti-GD2 antibodies as a treatment modality. Despite much success, a subset of patients, possibly those whose tumors fail to express concentrations of gangliosides such as GD1b and GT1b found in tumors from patients with a good prognosis, have tumors refractory to treatment. These observations support discussion of what is known about control of ganglioside synthesis, and their actual functions in NB, as well as their possible relationship to treatment response.
Collapse
Affiliation(s)
- Cara-Lynne Schengrund
- Department of Biochemistry and Molecular Biology, College of Medicine, Pennsylvania State University, Hershey, PA 17033, USA
| |
Collapse
|
40
|
Impact of Eukaryotic Translation Initiation Factors on Breast Cancer: Still Much to Investigate. Cancers (Basel) 2020; 12:cancers12071984. [PMID: 32708122 PMCID: PMC7409344 DOI: 10.3390/cancers12071984] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Revised: 07/04/2020] [Accepted: 07/08/2020] [Indexed: 12/11/2022] Open
Abstract
Breast carcinoma (BC) remains one of the most serious health problems. It is a heterogeneous entity, and mainly classified according to receptor status for estrogen (ER), progesterone (PR) and egf (HER2/Neu), as well as the proliferation marker ki67. Gene expression in eukaryotes is regulated at the level of both gene transcription and translation, where eukaryotic initiation factors (eIFs) are key regulators of protein biosynthesis. Aberrant translation results in an altered cellular proteome, and this clearly effects cell growth supporting tumorigenesis. The relationship between various eIFs and BC entities, as well as the related regulatory mechanisms, has meanwhile become a focus of scientific interest. Here, we give an overview on the current research state of eIF function, focusing on BC.
Collapse
|
41
|
Hamamura K, Hotta H, Murakumo Y, Shibuya H, Kondo Y, Furukawa K. SSEA-3 and 4 are not essential for the induction or properties of mouse iPS cells. J Oral Sci 2020; 62:393-396. [PMID: 32684574 DOI: 10.2334/josnusd.19-0513] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/01/2022]
Abstract
Stage-specific embryonic antigens (SSEA-1, 3, and 4) are carbohydrate antigens that have been used as markers of embryonic stem (ES) cells. However, the roles of these antigens in the establishment and maintenance of stemness of ES and induced pluripotent stem (iPS) cells are still poorly understood. This study investigated the biological and functional significance of globo-series glycolipids such as SSEA-3 and 4 in mouse iPS cells induced from tail-tip fibroblasts (TTFs) of α1,4Gal-T-knockout mice (lacking SSEA-3 and 4). These iPS cells were induced by retroviral transduction of four factors (Oct3/4, Sox2, Klf4, and c-Myc) into TTFs, and colonies were picked up. Morphologically, the colonies resembled ES cells and were positive for alkaline phosphatase and ES cell markers. Furthermore, in vitro-differentiated induction experiments after embryoid body formation revealed that some colonies derived from α1, 4Gal-T knockout mice were able to differentiate into three germ layers. Three germ layers were also observed in teratomas from iPS cells derived from α1,4Gal-T-knockout mice. These results suggest that SSEA-3 and 4 are not essential, at least for the establishment and maintenance of stemness of mouse iPS cells.
Collapse
Affiliation(s)
- Kazunori Hamamura
- Department of Pharmacology, School of Dentistry, Aichi Gakuin University
| | - Hiroshi Hotta
- Department of Biochemistry II, Nagoya University Graduate School of Medicine
| | | | - Hidenobu Shibuya
- Department of Biochemistry II, Nagoya University Graduate School of Medicine
| | - Yuji Kondo
- Department of Biochemistry II, Nagoya University Graduate School of Medicine
| | - Koichi Furukawa
- Department of Biochemistry II, Nagoya University Graduate School of Medicine.,Department of Biomedical Sciences, Chubu University College of Life and Health Sciences
| |
Collapse
|
42
|
Cavdarli S, Delannoy P, Groux-Degroote S. O-acetylated Gangliosides as Targets for Cancer Immunotherapy. Cells 2020; 9:cells9030741. [PMID: 32192217 PMCID: PMC7140702 DOI: 10.3390/cells9030741] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2020] [Revised: 03/16/2020] [Accepted: 03/16/2020] [Indexed: 12/25/2022] Open
Abstract
O-acetylation of sialic acid residues is one of the main modifications of gangliosides, and modulates ganglioside functions. O-acetylation of gangliosides is dependent on sialyl-O-acetyltransferases and sialyl-O-acetyl-esterase activities. CAS1 Domain-Containing Protein 1 (CASD1) is the only human sialyl-O-acetyltransferases (SOAT) described until now. O-acetylated ganglioside species are mainly expressed during embryonic development and in the central nervous system in healthy adults, but are re-expressed during cancer development and are considered as markers of cancers of neuroectodermal origin. However, the specific biological roles of O-acetylated gangliosides in developing and malignant tissues have not been extensively studied, mostly because of the requirement of specific approaches and tools for sample preparation and analysis. In this review, we summarize our current knowledge of ganglioside biosynthesis and expression in normal and pathological conditions, of ganglioside O-acetylation analysis and expression in cancers, and of the possible use of O-acetylated gangliosides as targets for cancer immunotherapy.
Collapse
Affiliation(s)
- Sumeyye Cavdarli
- UMR 8576 - UGSF - Unité de Glycobiologie Structurale et Fonctionnelle, CNRS, Université de Lille, F-59000 Lille, France; (S.C.); (P.D.)
- OGD2 Pharma, Institut de Recherche en Santé de l’Université de Nantes, 44007 Nantes, France
| | - Philippe Delannoy
- UMR 8576 - UGSF - Unité de Glycobiologie Structurale et Fonctionnelle, CNRS, Université de Lille, F-59000 Lille, France; (S.C.); (P.D.)
- Institut pour la Recherche sur le Cancer de Lille – IRCL – Place de Verdun, F-59000 Lille, France
| | - Sophie Groux-Degroote
- UMR 8576 - UGSF - Unité de Glycobiologie Structurale et Fonctionnelle, CNRS, Université de Lille, F-59000 Lille, France; (S.C.); (P.D.)
- Correspondence:
| |
Collapse
|
43
|
Yoshida H, Koodie L, Jacobsen K, Hanzawa K, Miyamoto Y, Yamamoto M. B4GALNT1 induces angiogenesis, anchorage independence growth and motility, and promotes tumorigenesis in melanoma by induction of ganglioside GM2/GD2. Sci Rep 2020; 10:1199. [PMID: 31988291 PMCID: PMC6985110 DOI: 10.1038/s41598-019-57130-2] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2019] [Accepted: 11/22/2019] [Indexed: 12/11/2022] Open
Abstract
β-1,4-N-Acetyl-Galactosaminyltransferase 1 (B4GALNT1) encodes the key enzyme B4GALNT1 to generate gangliosides GM2/GD2. GM2/GD2 gangliosides are surface glycolipids mainly found on brain neurons as well as peripheral nerves and skin melanocytes and are reported to exacerbate the malignant potential of melanomas. In order to elucidate the mechanism, we performed functional analyses of B4GALNT1-overexpressing cells. We analyzed ganglioside pattern on four melanoma and two neuroblastoma cell lines by high performance liquid chromatography (HPLC). We overexpressed B4GALNT1 in GM2/GD2-negative human melanoma cell line (SH4) and confirmed production of GM2/GD2 by HPLC. They showed higher anchorage independence growth (AIG) in colony formation assay, and exhibited augmented motility. In vitro, cell proliferation was not affected by GM2/GD2 expression. In vivo, GM2/GD2-positive SH4 clones showed significantly higher tumorigenesis in NOD/Scid/IL2Rγ-null mice, and immunostaining of mouse CD31 revealed that GM2/GD2 induced remarkable angiogenesis. No differences were seen in melanoma stem cell and Epithelial-Mesenchymal Transition markers between GM2/GD2-positive and -negative SH4 cells. We therefore concluded that B4GALNT1, and consequently GM2/GD2, enhanced tumorigenesis via induction of angiogenesis, AIG, and cell motility. RNA-Seq suggested periostin as a potential key factor for angiogenesis and AIG. These findings may lead to development of novel therapy for refractory melanoma.
Collapse
Affiliation(s)
- Hideki Yoshida
- Department of Surgery, University of Minnesota, Minneapolis, Minnesota, USA
| | - Lisa Koodie
- Department of Surgery, University of Minnesota, Minneapolis, Minnesota, USA
| | - Kari Jacobsen
- Department of Surgery, University of Minnesota, Minneapolis, Minnesota, USA
| | - Ken Hanzawa
- Department of Molecular Biology, Osaka International Cancer Institute, Osaka, Japan
| | - Yasuhide Miyamoto
- Department of Molecular Biology, Osaka International Cancer Institute, Osaka, Japan
| | - Masato Yamamoto
- Department of Surgery, University of Minnesota, Minneapolis, Minnesota, USA.
- Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota, USA.
- Stem Cell Institute, University of Minnesota, Minneapolis, Minnesota, USA.
| |
Collapse
|
44
|
Seitz CM, Schroeder S, Knopf P, Krahl AC, Hau J, Schleicher S, Martella M, Quintanilla-Martinez L, Kneilling M, Pichler B, Lang P, Atar D, Schilbach K, Handgretinger R, Schlegel P. GD2-targeted chimeric antigen receptor T cells prevent metastasis formation by elimination of breast cancer stem-like cells. Oncoimmunology 2019; 9:1683345. [PMID: 32002293 PMCID: PMC6959445 DOI: 10.1080/2162402x.2019.1683345] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2019] [Revised: 10/16/2019] [Accepted: 10/16/2019] [Indexed: 02/07/2023] Open
Abstract
Expression of the disialoganglioside GD2 has been identified as a marker antigen associated with a breast cancer stem-like cell (BCSC) phenotype. Here, we report on the evaluation of GD2 as a BCSC-specific target antigen for immunotherapy. GD2 expression was confirmed at variable degree in a set of breast cancer cell lines, predominantly in triple-negative breast cancer (TNBC). To target GD2, we have generated novel anti-GD2 chimeric antigen receptors (GD2-CAR), based on single-chain variable fragments (scFv) derived from the monoclonal antibody (mAb) ch14.18, also known as dinutuximab beta. Expressed on T cells, GD2-CARs mediated specific GD2-dependent T-cell activation and target cell lysis. In contrast to previously described GD2-CARs, no signs of exhaustion by tonic signaling were found. Importantly, application of GD2-CAR expressing T cells (GD2-CAR-T) in an orthotopic xenograft model of TNBC (MDA-MB-231) halted local tumor progression and completely prevented lung metastasis formation. In line with the BCSC model, GD2 expression was only found in a subpopulation (4-6%) of MDA-MB-231 cells before injection. Significant expansion of GD2-CAR-T in tumor-bearing mice as well as T-cell infiltrates in the primary tumor and the lungs were found, indicating site-specific activation of GD2-CAR-T. Our data strongly support previous findings of GD2 as a BCSC-associated antigen. GD2-targeted immunotherapies have been extensively studied in human. In conclusion, GD2-CAR-T should be considered a promising novel approach for GD2-positive breast cancer, especially to eliminate disseminated tumor cells and prevent metastasis formation.
Collapse
Affiliation(s)
- Christian M Seitz
- Department of Pediatric Hematology and Oncology, University Children's Hospital Tuebingen, Tuebingen, Germany
| | - Sarah Schroeder
- Department of Pediatric Hematology and Oncology, University Children's Hospital Tuebingen, Tuebingen, Germany
| | - Philipp Knopf
- Werner Siemens Imaging Center, Department of Preclinical Imaging and Radiopharmacy, Eberhard Karls University Tuebingen, Tuebingen, Germany
| | - Ann-Christin Krahl
- Department of Pediatric Hematology and Oncology, University Children's Hospital Tuebingen, Tuebingen, Germany
| | - Jana Hau
- Department of Pediatric Hematology and Oncology, University Children's Hospital Tuebingen, Tuebingen, Germany
| | - Sabine Schleicher
- Department of Pediatric Hematology and Oncology, University Children's Hospital Tuebingen, Tuebingen, Germany
| | - Manuela Martella
- Department of Pathology, Eberhard Karls University Tuebingen, Tuebingen, Germany
| | | | - Manfred Kneilling
- Werner Siemens Imaging Center, Department of Preclinical Imaging and Radiopharmacy, Eberhard Karls University Tuebingen, Tuebingen, Germany
| | - Bernd Pichler
- Werner Siemens Imaging Center, Department of Preclinical Imaging and Radiopharmacy, Eberhard Karls University Tuebingen, Tuebingen, Germany.,Cluster of Excellence iFIT (EXC 2180) "Image-Guided and Functionally Instructed Tumor Therapies", University of Tuebingen, Tuebingen, Germany
| | - Peter Lang
- Department of Pediatric Hematology and Oncology, University Children's Hospital Tuebingen, Tuebingen, Germany
| | - Daniel Atar
- Department of Pediatric Hematology and Oncology, University Children's Hospital Tuebingen, Tuebingen, Germany
| | - Karin Schilbach
- Department of Pediatric Hematology and Oncology, University Children's Hospital Tuebingen, Tuebingen, Germany
| | - Rupert Handgretinger
- Department of Pediatric Hematology and Oncology, University Children's Hospital Tuebingen, Tuebingen, Germany.,Cluster of Excellence iFIT (EXC 2180) "Image-Guided and Functionally Instructed Tumor Therapies", University of Tuebingen, Tuebingen, Germany
| | - Patrick Schlegel
- Department of Pediatric Hematology and Oncology, University Children's Hospital Tuebingen, Tuebingen, Germany.,Cluster of Excellence iFIT (EXC 2180) "Image-Guided and Functionally Instructed Tumor Therapies", University of Tuebingen, Tuebingen, Germany
| |
Collapse
|
45
|
Takeuchi R, Kambe M, Miyata M, Jeyadevan U, Tajima O, Furukawa K, Furukawa K. TNFα-signal and cAMP-mediated signals oppositely regulate melanoma- associated ganglioside GD3 synthase gene in human melanocytes. Sci Rep 2019; 9:14740. [PMID: 31611597 PMCID: PMC6791844 DOI: 10.1038/s41598-019-51333-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2018] [Accepted: 09/26/2019] [Indexed: 12/17/2022] Open
Abstract
Analyses of expression and regulation of ganglioside synthases in melanocytes are important to understand roles of gangliosides in melanomagenesis. In this study, we analyzed the expression and regulatory mechanisms of glycosyltransferase genes responsible for ganglioside synthesis in normal melanocytes. We reported previously that culture supernatants of UVB-irradiated keratinocytes induced upregulation of ganglioside GD3 synthase gene in melanocytes, and mainly TNFα was responsible for it. Then, we found that elimination of dibutyryl cyclic AMP and IBMX from the medium also resulted in upregulation of the GD3 synthase gene. The addition of α-melanocyte-stimulating hormone which increases cAMP, to the medium led to a significant reduction in the GD3 synthase gene expression level, and a PKA inhibitor enhanced the GD3 synthase gene level. These results suggest that signals mediated via TNFα and cAMP oppositely regulate GD3 synthase gene expression in melanocytes. The results of an IKK inhibitor indicate the possibility that TNFα induces GD3 synthase gene expression via NF-κB signaling in melanocytes. When melanoma cells were treated by these factors, no fluctuation in the GD3 synthase gene expression level was observed, although an IKK inhibitor significantly suppressed it, suggesting that ganglioside synthase genes are regulated in distinct manners between melanocytes and melanomas.
Collapse
Affiliation(s)
- Rika Takeuchi
- Department of Biomedical Sciences, Chubu University College of Life and Health Sciences, Matsumoto 1200, Kasugai, Aichi, 487-8501, Japan
| | - Mariko Kambe
- Department of Biomedical Sciences, Chubu University College of Life and Health Sciences, Matsumoto 1200, Kasugai, Aichi, 487-8501, Japan
| | - Maiko Miyata
- Department of Biomedical Sciences, Chubu University College of Life and Health Sciences, Matsumoto 1200, Kasugai, Aichi, 487-8501, Japan
| | - Upul Jeyadevan
- Department of Biomedical Sciences, Chubu University College of Life and Health Sciences, Matsumoto 1200, Kasugai, Aichi, 487-8501, Japan
| | - Orie Tajima
- Department of Biomedical Sciences, Chubu University College of Life and Health Sciences, Matsumoto 1200, Kasugai, Aichi, 487-8501, Japan
| | - Koichi Furukawa
- Department of Biomedical Sciences, Chubu University College of Life and Health Sciences, Matsumoto 1200, Kasugai, Aichi, 487-8501, Japan
| | - Keiko Furukawa
- Department of Biomedical Sciences, Chubu University College of Life and Health Sciences, Matsumoto 1200, Kasugai, Aichi, 487-8501, Japan.
| |
Collapse
|
46
|
Folkert IW, Devalaraja S, Linette GP, Weber K, Haldar M. Primary Bone Tumors: Challenges and Opportunities for CAR-T Therapies. J Bone Miner Res 2019; 34:1780-1788. [PMID: 31441962 DOI: 10.1002/jbmr.3852] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/31/2019] [Revised: 07/10/2019] [Accepted: 07/27/2019] [Indexed: 12/22/2022]
Abstract
Primary malignant bone tumors are rare, occur in all age groups, and include distinct entities such as osteosarcoma, Ewing sarcoma, and chondrosarcoma. Traditional treatment with some combination of chemotherapy, surgery, and radiation has reached the limit of efficacy, with substantial room for improvement in patient outcome. Furthermore, genomic characterization of these tumors reveals a paucity of actionable molecular targets. Against this backdrop, recent advances in cancer immunotherapy represent a silver lining in the treatment of primary bone cancer. Major strategies in cancer immunotherapy include stimulating naturally occurring anti-tumor T cells and adoptive transfer of tumor-specific cytotoxic T cells. Chimeric antigen receptor T cells (CAR-T cells) belong to the latter strategy and are an impressive application of both insights into T cell biology and advances in genetic engineering. In this review, we briefly describe the CAR-T approach and discuss its applications in primary bone tumors. © 2019 American Society for Bone and Mineral Research.
Collapse
Affiliation(s)
- Ian W Folkert
- Department of Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Samir Devalaraja
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Gerald P Linette
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Kristy Weber
- Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.,Penn Sarcoma Program, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Malay Haldar
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.,Penn Sarcoma Program, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.,Abramson Family Cancer Research Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
47
|
Cavdarli S, Groux-Degroote S, Delannoy P. Gangliosides: The Double-Edge Sword of Neuro-Ectodermal Derived Tumors. Biomolecules 2019; 9:E311. [PMID: 31357634 PMCID: PMC6723632 DOI: 10.3390/biom9080311] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2019] [Revised: 07/25/2019] [Accepted: 07/26/2019] [Indexed: 12/12/2022] Open
Abstract
Gangliosides, the glycosphingolipids carrying one or several sialic acid residues, are mostly localized at the plasma membrane in lipid raft domains and implicated in many cellular signaling pathways mostly by interacting with tyrosine kinase receptors. Gangliosides are divided into four series according to the number of sialic acid residues, which can be also modified by O-acetylation. Both ganglioside expression and sialic acid modifications can be modified in pathological conditions such as cancer, which can induce either pro-cancerous or anti-cancerous effects. In this review, we summarize the specific functions of gangliosides in neuro-ectodermal derived tumors, and their roles in reprogramming the lipidomic profile of cell membrane occurring with the induction of epithelial-mesenchymal transition.
Collapse
Affiliation(s)
- Sumeyye Cavdarli
- Université de Lille, CNRS, UMR8576-UGSF-Unité de Glycobiologie Structurale et Fonctionnelle, F59000 Lille, France
| | - Sophie Groux-Degroote
- Université de Lille, CNRS, UMR8576-UGSF-Unité de Glycobiologie Structurale et Fonctionnelle, F59000 Lille, France
| | - Philippe Delannoy
- Université de Lille, CNRS, UMR8576-UGSF-Unité de Glycobiologie Structurale et Fonctionnelle, F59000 Lille, France.
| |
Collapse
|
48
|
Tong W, Maira M, Roychoudhury R, Galan A, Brahimi F, Gilbert M, Cunningham AM, Josephy S, Pirvulescu I, Moffett S, Saragovi HU. Vaccination with Tumor-Ganglioside Glycomimetics Activates a Selective Immunity that Affords Cancer Therapy. Cell Chem Biol 2019; 26:1013-1026.e4. [DOI: 10.1016/j.chembiol.2019.03.018] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2018] [Revised: 09/19/2018] [Accepted: 03/27/2019] [Indexed: 02/06/2023]
|
49
|
Deficiency of GD3 Synthase in Mice Resulting in the Attenuation of Bone Loss with Aging. Int J Mol Sci 2019; 20:ijms20112825. [PMID: 31185614 PMCID: PMC6600367 DOI: 10.3390/ijms20112825] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2019] [Revised: 05/28/2019] [Accepted: 06/07/2019] [Indexed: 12/14/2022] Open
Abstract
Gangliosides are widely expressed in almost all tissues and cells and are also considered to be essential in the development and maintenance of various organs and tissues. However, little is known about their roles in bone metabolism. In this study, we investigated the effects of genetic deletion of ganglioside D3 (GD3) synthase, which is responsible for the generation of all b-series gangliosides, on bone metabolism. Although b-series gangliosides were not expressed in osteoblasts, these gangliosides were expressed in pre-osteoclasts. However, the expression of these gangliosides was decreased after induction of osteoclastogenesis by receptor activator of nuclear factor kappa-B ligand (RANKL). Three-dimensional micro-computed tomography (3D-μCT) analysis revealed that femoral cancellous bone mass in GD3 synthase-knockout (GD3S KO) mice was higher than that in wild type (WT) mice at the age of 40 weeks, although there were no differences in that between GD3S KO and WT mice at 15 weeks old. Whereas bone formation parameters (osteoblast numbers/bone surface and osteoblast surface/bone surface) in GD3S KO mice did not differ from WT mice, bone resorption parameters (osteoclast numbers/bone surface and osteoclast surface/bone surface) in GD3S KO mice became significantly lower than those in WT mice at 40 weeks of age. Collectively, this study demonstrates that deletion of GD3 synthase attenuates bone loss that emerges with aging.
Collapse
|
50
|
Furukawa K, Ohmi Y, Ohkawa Y, Bhuiyan RH, Zhang P, Tajima O, Hashimoto N, Hamamura K, Furukawa K. New era of research on cancer-associated glycosphingolipids. Cancer Sci 2019; 110:1544-1551. [PMID: 30895683 PMCID: PMC6501054 DOI: 10.1111/cas.14005] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2018] [Revised: 03/13/2019] [Accepted: 03/19/2019] [Indexed: 12/31/2022] Open
Abstract
Cancer‐associated glycosphingolipids have been used as markers for diagnosis and targets for immunotherapy of malignant tumors. Recent progress in the analysis of their implications in the malignant properties of cancer cells revealed that cancer‐associated glycosphingolipids are not only tumor markers, but also functional molecules regulating various signals introduced by membrane microdomains, lipid rafts. In particular, a novel approach, enzyme‐mediated activation of radical sources combined with mass spectrometry, has enabled us to clarify the mechanisms by which cancer‐associated glycosphingolipids regulate cell signals based on the interaction with membrane molecules and formation of molecular complexes on the cell surface. Novel findings obtained from these approaches are now providing us with insights into the development of new anticancer therapies targeting membrane molecular complexes consisting of cancer‐associated glycolipids and their associated membrane molecules. Thus, a new era of cancer‐associated glycosphingolipids has now begun.
Collapse
Affiliation(s)
- Koichi Furukawa
- Department of Biomedical Sciences, Chubu University College of Life and Health Sciences, Kasugai, Japan.,Department of Molecular Biochemistry, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Yuhsuke Ohmi
- Department of Medical Technology, Chubu University College of Life and Health Sciences, Kasugai, Japan
| | - Yuki Ohkawa
- Department of Biomedical Sciences, Chubu University College of Life and Health Sciences, Kasugai, Japan
| | - Robiul H Bhuiyan
- Department of Biomedical Sciences, Chubu University College of Life and Health Sciences, Kasugai, Japan
| | - Pu Zhang
- Department of Biomedical Sciences, Chubu University College of Life and Health Sciences, Kasugai, Japan.,Department of Molecular Biochemistry, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Orie Tajima
- Department of Biomedical Sciences, Chubu University College of Life and Health Sciences, Kasugai, Japan
| | - Noboru Hashimoto
- Department of Molecular Biochemistry, Nagoya University Graduate School of Medicine, Nagoya, Japan.,Department of Anatomy, Faculty of Medical and Dental Sciences, Tokushima University, Tokushima, Japan
| | - Kazunori Hamamura
- Department of Pharmacology, School of Dentistry, Aichi-Gakuin University, Nagoya, Japan
| | - Keiko Furukawa
- Department of Biomedical Sciences, Chubu University College of Life and Health Sciences, Kasugai, Japan
| |
Collapse
|