1
|
Sánchez-Martínez H, Jiménez-Castillo V, Illescas-Barbosa D, Ávila-Curiel BX, Hernández-Huerta MT, Díaz-Castillejos R, Torres-Rosas R, Zenteno E, Pereyra-Morales MA, Solórzano-Mata CJ. Expression of 9- O-Acetylated Sialic Acid in HPV+ Oral Squamous Cell Carcinoma Cells. Life (Basel) 2025; 15:663. [PMID: 40283217 PMCID: PMC12028955 DOI: 10.3390/life15040663] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2025] [Revised: 04/09/2025] [Accepted: 04/14/2025] [Indexed: 04/29/2025] Open
Abstract
Oral squamous cell carcinoma (OSCC) is a common type of head and neck malignancy that represents a significant global health issue. Sialylations are common events in tumor transformation, proliferation, metastasis, and immune evasion. Modifications in sialylation can be detected by lectins, whose changes in OSCC have been related to grade, invasion, and metastasis. The presence of 9-O-acetylated sialic acid (Neu5,9Ac2) in OSCC cells and its potential expression, modification, and role are unknown. This study aimed to analyze the expression of Neu5,9Ac2 using the Macrobrachium rosenbergii lectin (MrL) that recognizes this sialic acid (Neu5Ac) residue and also compare its effect on the SCC-152 cell line (CRL-3240, ATCC) and immortalized keratinocytes (HaCaT) as a control. We observed by immunocytochemistry that SCC-152 cells expressed more Neu5,9Ac2 compared to HaCaT cells; the specificity of MrL was confirmed after the sialidase treatment of cells in which the loss of lectin's recognition of Neu5,9Ac2 was observed. The electrophoretic profile was similar between both cell line types; however, the Western blot showed differences in the glycoprotein patterns recognized by lectin for each cell type. MrL increased the proliferation of SCC-152 cells, as well as the integrity and morphology of the colonies. Therefore, our results suggest that Neu5,9Ac2 glycosylated receptors could be involved in the survival and proliferation of OSCC cells, which offers a promising avenue for developing diagnostic and prognostic tools (tumor markers) against oral squamous cell carcinoma in the future.
Collapse
Affiliation(s)
- Hugo Sánchez-Martínez
- Faculty of Dentistry, Universidad Autónoma Benito Juárez de Oaxaca, Oaxaca 68120, Mexico; (H.S.-M.); (D.I.-B.); (B.X.Á.-C.); (R.D.-C.); (R.T.-R.)
| | - Victoria Jiménez-Castillo
- Faculty of Medicine and Surgery, Universidad Autónoma Benito Juárez de Oaxaca, Oaxaca 68120, Mexico;
| | - Daniela Illescas-Barbosa
- Faculty of Dentistry, Universidad Autónoma Benito Juárez de Oaxaca, Oaxaca 68120, Mexico; (H.S.-M.); (D.I.-B.); (B.X.Á.-C.); (R.D.-C.); (R.T.-R.)
| | - Beatriz Xochitl Ávila-Curiel
- Faculty of Dentistry, Universidad Autónoma Benito Juárez de Oaxaca, Oaxaca 68120, Mexico; (H.S.-M.); (D.I.-B.); (B.X.Á.-C.); (R.D.-C.); (R.T.-R.)
| | | | - Risk Díaz-Castillejos
- Faculty of Dentistry, Universidad Autónoma Benito Juárez de Oaxaca, Oaxaca 68120, Mexico; (H.S.-M.); (D.I.-B.); (B.X.Á.-C.); (R.D.-C.); (R.T.-R.)
| | - Rafael Torres-Rosas
- Faculty of Dentistry, Universidad Autónoma Benito Juárez de Oaxaca, Oaxaca 68120, Mexico; (H.S.-M.); (D.I.-B.); (B.X.Á.-C.); (R.D.-C.); (R.T.-R.)
| | - Edgar Zenteno
- Departament of Biochemistry, Faculty of Medicine, Universidad Nacional Autónoma de México, Ciudad de México 04510, Mexico; (E.Z.); (M.A.P.-M.)
| | - Mohamed Alí Pereyra-Morales
- Departament of Biochemistry, Faculty of Medicine, Universidad Nacional Autónoma de México, Ciudad de México 04510, Mexico; (E.Z.); (M.A.P.-M.)
| | - Carlos Josué Solórzano-Mata
- Faculty of Dentistry, Universidad Autónoma Benito Juárez de Oaxaca, Oaxaca 68120, Mexico; (H.S.-M.); (D.I.-B.); (B.X.Á.-C.); (R.D.-C.); (R.T.-R.)
- Faculty of Medicine and Surgery, Universidad Autónoma Benito Juárez de Oaxaca, Oaxaca 68120, Mexico;
| |
Collapse
|
2
|
Zhang N, Liu Q, Wang D, Wang X, Pan Z, Han B, He G. Multifaceted roles of Galectins: from carbohydrate binding to targeted cancer therapy. Biomark Res 2025; 13:49. [PMID: 40134029 PMCID: PMC11934519 DOI: 10.1186/s40364-025-00759-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2024] [Accepted: 03/05/2025] [Indexed: 03/27/2025] Open
Abstract
Galectins play pivotal roles in cellular recognition and signaling processes by interacting with glycoconjugates. Extensive research has highlighted the significance of Galectins in the context of cancer, aiding in the identification of biomarkers for early detection, personalized therapy, and predicting treatment responses. This review offers a comprehensive overview of the structural characteristics, ligand-binding properties, and interacting proteins of Galectins. We delve into their biological functions and examine their roles across various cancer types. Galectins, characterized by a conserved carbohydrate recognition domain (CRD), are divided into prototype, tandem-repeat, and chimera types based on their structural configurations. Prototype Galectins contain a single CRD, tandem-repeat Galectins contain two distinct CRDs linked by a peptide, and the chimera-type Galectin-3 features a unique structural arrangement. The capacity of Galectins to engage in multivalent interactions allows them to regulate a variety of signaling pathways, thereby affecting cell fate and function. In cancer, Galectins contribute to tumor cell transformation, angiogenesis, immune evasion, and metastasis, making them critical targets for therapeutic intervention. This review discusses the multifaceted roles of Galectins in cancer progression and explores current advancements in the development of Galectin-targeted therapies. We also address the challenges and future directions for integrating Galectin research into clinical practice to enhance cancer treatment outcomes. In brief, understanding the complex functions of Galectins in cancer biology opens new avenues for therapeutic strategies. Continued research on Galectin interactions and their pathological roles is essential for developing effective carbohydrate-based treatments and improving clinical interventions for cancer patients.
Collapse
Affiliation(s)
- Nan Zhang
- State Key Laboratory of Southwestern Chinese Medicine Resources, College of Medical Technology and School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
- Institute of Precision Drug Innovation and Cancer Center, the Second Hospital of Dalian Medical University, Dalian, 116023, China
| | - Qiao Liu
- Institute of Precision Drug Innovation and Cancer Center, the Second Hospital of Dalian Medical University, Dalian, 116023, China
| | - Daihan Wang
- Institute of Precision Drug Innovation and Cancer Center, the Second Hospital of Dalian Medical University, Dalian, 116023, China
| | - Xiaoyun Wang
- Institute of Precision Drug Innovation and Cancer Center, the Second Hospital of Dalian Medical University, Dalian, 116023, China
| | - Zhaoping Pan
- Institute of Precision Drug Innovation and Cancer Center, the Second Hospital of Dalian Medical University, Dalian, 116023, China
| | - Bo Han
- State Key Laboratory of Southwestern Chinese Medicine Resources, College of Medical Technology and School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China.
| | - Gu He
- Institute of Precision Drug Innovation and Cancer Center, the Second Hospital of Dalian Medical University, Dalian, 116023, China.
| |
Collapse
|
3
|
Toomajian V, Tundo A, Ural EE, Greeson EM, Contag CH, Makela AV. Magnetic Particle Imaging Reveals that Iron-Labeled Extracellular Vesicles Accumulate in Brains of Mice with Metastases. ACS APPLIED MATERIALS & INTERFACES 2024; 16:30860-30873. [PMID: 38860682 PMCID: PMC11194773 DOI: 10.1021/acsami.4c04920] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Revised: 05/27/2024] [Accepted: 05/29/2024] [Indexed: 06/12/2024]
Abstract
The incidence of breast cancer remains high worldwide and is associated with a significant risk of metastasis to the brain that can be fatal; this is due, in part, to the inability of therapeutics to cross the blood-brain barrier (BBB). Extracellular vesicles (EVs) have been found to cross the BBB and further have been used to deliver drugs to tumors. EVs from different cell types appear to have different patterns of accumulation and retention as well as the efficiency of bioactive cargo delivery to recipient cells in the body. Engineering EVs as delivery tools to treat brain metastases, therefore, will require an understanding of the timing of EV accumulation and their localization relative to metastatic sites. Magnetic particle imaging (MPI) is a sensitive and quantitative imaging method that directly detects superparamagnetic iron. Here, we demonstrate MPI as a novel tool to characterize EV biodistribution in metastatic disease after labeling EVs with superparamagnetic iron oxide (SPIO) nanoparticles. Iron-labeled EVs (FeEVs) were collected from iron-labeled parental primary 4T1 tumor cells and brain-seeking 4T1BR5 cells, followed by injection into the mice with orthotopic tumors or brain metastases. MPI quantification revealed that FeEVs were retained for longer in orthotopic mammary carcinomas compared to SPIOs. MPI signal due to iron could only be detected in brains of mice bearing brain metastases after injection of FeEVs, but not SPIOs, or FeEVs when mice did not have brain metastases. These findings indicate the potential use of EVs as a therapeutic delivery tool in primary and metastatic tumors.
Collapse
Affiliation(s)
- Victoria
A. Toomajian
- Institute
for Quantitative Health Science and Engineering, Michigan State University, East Lansing, Michigan 48824, United States
- Department
of Biomedical Engineering, Michigan State
University, East Lansing, Michigan 48824, United States
| | - Anthony Tundo
- Institute
for Quantitative Health Science and Engineering, Michigan State University, East Lansing, Michigan 48824, United States
| | - Evran E. Ural
- Institute
for Quantitative Health Science and Engineering, Michigan State University, East Lansing, Michigan 48824, United States
- Department
of Biomedical Engineering, Michigan State
University, East Lansing, Michigan 48824, United States
| | - Emily M. Greeson
- Institute
for Quantitative Health Science and Engineering, Michigan State University, East Lansing, Michigan 48824, United States
- Department
of Microbiology, Genetics & Immunology, Michigan State University, East
Lansing, Michigan 48824, United States
| | - Christopher H. Contag
- Institute
for Quantitative Health Science and Engineering, Michigan State University, East Lansing, Michigan 48824, United States
- Department
of Biomedical Engineering, Michigan State
University, East Lansing, Michigan 48824, United States
- Department
of Microbiology, Genetics & Immunology, Michigan State University, East
Lansing, Michigan 48824, United States
| | - Ashley V. Makela
- Institute
for Quantitative Health Science and Engineering, Michigan State University, East Lansing, Michigan 48824, United States
| |
Collapse
|
4
|
dos Santos SN, Junior DSG, Pereira JPM, Iadocicco NM, Silva AH, do Nascimento T, Dias LAP, de Oliveira Silva FR, Ricci-Junior E, Santos-Oliveira R, Bernardes ES. Development of glycan-targeted nanoparticles as a novel therapeutic opportunity for gastric cancer treatment. Cancer Nanotechnol 2023. [DOI: 10.1186/s12645-023-00161-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/30/2023] Open
Abstract
AbstractChemotherapy resistance remains a major cause of therapeutic failure in gastric cancer. The combination of genetic material such as interference RNAs (iRNAs) to silence cancer-associated genes with chemotherapeutics has become a novel approach for cancer treatment. However, finding the right target genes and developing non-toxic, highly selective nanocarrier systems remains a challenge. Here we developed a novel sialyl-Tn-targeted polylactic acid—didodecyldimethylammonium bromide nanoparticle (PLA-DDAB) nanoparticles (NPs) loaded with dsRNA targeting ST6GalNac-I and/or galectin-3 genes. Using single photon emission computed tomography (SPECT), we have demonstrated that 99mtechnetium radiolabeled sialyl-Tn-targeted nanoparticles can reach the tumor site and downregulate ST6GalNAc-I and galectin-3 RNA expression levels when injected intravenously. Furthermore, using an in vivo gastric tumor model, these nanoparticles increased the effectiveness of 5-FU in reducing tumor growth. Our findings indicate that cancer-associated glycan-targeted NPs loaded with dsRNA targeting ST6GalNAc-I and/or galectin-3 in combination with standard chemotherapy, have the potential to become a novel therapeutic tool for gastric cancer.
Collapse
|
5
|
Gupta R, Ponangi R, Indresh KG. Role of glycosylation in breast cancer progression and metastasis: implications for miRNA, EMT and multidrug resistance. Glycobiology 2023; 33:545-555. [PMID: 37283470 DOI: 10.1093/glycob/cwad046] [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: 05/13/2022] [Revised: 04/18/2023] [Accepted: 06/02/2023] [Indexed: 06/08/2023] Open
Abstract
Breast cancer (BC) is one of the leading causes of death in women, globally. A variety of biological processes results in metastasis, a poorly understood pathological phenomenon, causing a high relapse rate. Glycosylation, microribonucleic acids (miRNAs) and epithelial to mesenchymal transition (EMT), have been shown to regulate this cascade where tumor cells detach from their primary site, enter the circulatory system and colonize distant sites. Integrated proteomics and glycomics approaches have been developed to probe the molecular mechanism regulating such metastasis. In this review, we describe specific aspects of glycosylation and its interrelation with miRNAs, EMT and multidrug resistance during BC progression and metastasis. We explore various approaches that determine the role of proteomes and glycosylation in BC diagnosis, therapy and drug discovery.
Collapse
Affiliation(s)
- Rohitesh Gupta
- Cancer Biology, CSIR-Centre for Cellular and Molecular Biology, Uppal Road, Hyderabad, 500007 Telangana, India
| | - Rohan Ponangi
- Cancer Biology, CSIR-Centre for Cellular and Molecular Biology, Uppal Road, Hyderabad, 500007 Telangana, India
| | - Kuppanur G Indresh
- Cancer Biology, CSIR-Centre for Cellular and Molecular Biology, Uppal Road, Hyderabad, 500007 Telangana, India
| |
Collapse
|
6
|
Funkhouser A, Shuster H, Martin JC, Edenfield WJ, Blenda AV. Pattern Analysis of Serum Galectins-1, -3, and -9 in Breast Cancer. Cancers (Basel) 2023; 15:3809. [PMID: 37568625 PMCID: PMC10417135 DOI: 10.3390/cancers15153809] [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: 06/22/2023] [Revised: 07/24/2023] [Accepted: 07/25/2023] [Indexed: 08/13/2023] Open
Abstract
Galectins have been shown to have roles in cancer progression via their contributions to angiogenesis, metastasis, cell division, and the evasion of immune destruction. This study analyzes galectin-1, -3, and -9 serum concentrations in breast cancer patients through enzyme-linked immunosorbent assay (ELISA) against the characteristics of the patient and the tumor such as stage, molecular subtype, and receptor expression. Galectin-9 was found to be statistically significantly increased in HER2-enriched tumors and reduced in patients with hormone-receptor-positive tumors. Galectin-1 was found to be statistically significantly increased in the serum of patients who had undergone hormonal, immunotherapy, or chemotherapy. These findings provide insight into the changes in galectin levels during the progress of cancer, the response to treatment, and the molecular phenotype. These findings are valuable in the further understanding of the relationships between galectin and tumor biology and can inform future research on therapeutic targets for galectin inhibitors and the utility of galectin biomarkers.
Collapse
Affiliation(s)
- Avery Funkhouser
- Department of Biomedical Sciences, University of South Carolina School of Medicine Greenville, Greenville, SC 29605, USA
| | - Hayden Shuster
- Department of Biomedical Sciences, University of South Carolina School of Medicine Greenville, Greenville, SC 29605, USA
| | - Julie C. Martin
- Prisma Health Cancer Institute, Prisma Health, Greenville, SC 29605, USA
| | - W. Jeffery Edenfield
- Department of Biomedical Sciences, University of South Carolina School of Medicine Greenville, Greenville, SC 29605, USA
- Prisma Health Cancer Institute, Prisma Health, Greenville, SC 29605, USA
| | - Anna V. Blenda
- Department of Biomedical Sciences, University of South Carolina School of Medicine Greenville, Greenville, SC 29605, USA
- Prisma Health Cancer Institute, Prisma Health, Greenville, SC 29605, USA
| |
Collapse
|
7
|
Morishita A, Oura K, Tadokoro T, Shi T, Fujita K, Tani J, Atsukawa M, Masaki T. Galectin-9 in Gastroenterological Cancer. Int J Mol Sci 2023; 24:ijms24076174. [PMID: 37047155 PMCID: PMC10094448 DOI: 10.3390/ijms24076174] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Revised: 03/07/2023] [Accepted: 03/20/2023] [Indexed: 03/29/2023] Open
Abstract
Immunochemotherapy has become popular in recent years. The detailed mechanisms of cancer immunity are being elucidated, and new developments are expected in the future. Apoptosis allows tissues to maintain their form, quantity, and function by eliminating excess or abnormal cells. When apoptosis is inhibited, the balance between cell division and death is disrupted and tissue homeostasis is impaired. This leads to dysfunction and the accumulation of genetically abnormal cells, which can contribute to carcinogenesis. Lectins are neither enzymes nor antibodies but proteins that bind sugar chains. Among soluble endogenous lectins, galectins interact with cell surface sugar chains outside the cell to regulate signal transduction and cell growth. On the other hand, intracellular lectins are present at the plasma membrane and regulate signal transduction by regulating receptor–ligand interactions. Galectin-9 expressed on the surface of thymocytes induces apoptosis of T lymphocytes and plays an essential role in immune self-tolerance by negative selection in the thymus. Furthermore, the administration of extracellular galectin-9 induces apoptosis of human cancer and immunodeficient cells. However, the detailed pharmacokinetics of galectin-9 in vivo have not been elucidated. In addition, the cell surface receptors involved in galectin-9-induced apoptosis of cancer cells have not been identified, and the intracellular pathways involved in apoptosis have not been fully investigated. We have previously reported that galectin-9 induces apoptosis in various gastrointestinal cancers and suppresses tumor growth. However, the mechanism of galectin-9 and apoptosis induction in gastrointestinal cancers and the detailed mechanisms involved in tumor growth inhibition remain unknown. In this article, we review the effects of galectin-9 on gastrointestinal cancers and its mechanisms.
Collapse
|
8
|
Valdivia AO, He Y, Ren X, Wen D, Dong L, Nazari H, Li X. Probable Treatment Targets for Diabetic Retinopathy Based on an Integrated Proteomic and Genomic Analysis. Transl Vis Sci Technol 2023; 12:8. [PMID: 36745438 PMCID: PMC9910385 DOI: 10.1167/tvst.12.2.8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Purpose Using previously approved medications for new indications can expedite the lengthy and expensive drug development process. We describe a bioinformatics pipeline that integrates genomics and proteomics platforms to identify already-approved drugs that might be useful to treat diabetic retinopathy (DR). Methods Proteomics analysis of vitreous humor samples from 12 patients undergoing pars plana vitrectomy for DR and a whole genome dataset (UKBiobank TOPMed-imputed) from 1330 individuals with DR and 395,155 controls were analyzed independently to identify biological pathways associated with DR. Common biological pathways shared between both datasets were further analyzed (STRING and REACTOME analyses) to identify target proteins for probable drug modulation. Curated target proteins were subsequently analyzed by the BindingDB database to identify chemical compounds they interact with. Identified chemical compounds were further curated through the Expasy SwissSimilarity database for already-approved drugs that interact with target proteins. Results The pathways in each dataset (proteomics and genomics) converged in the upregulation of a previously unknown pathway involved in DR (RUNX2 signaling; constituents MMP-13 and LGALS3), with an emphasis on its role in angiogenesis and blood-retina barrier. Bioinformatics analysis identified U.S. Food and Drug Administration (FDA)-approved medications (raltitrexed, pemetrexed, glyburide, probenecid, clindamycin hydrochloride, and ticagrelor) that, in theory, may modulate this pathway. Conclusions The bioinformatics pipeline described here identifies FDA-approved drugs that can be used for new alternative indications. These theoretical candidate drugs should be validated with experimental studies. Translational Relevance Our study suggests possible drugs for DR treatment based on an integrated proteomics and genomics pipeline. This approach can potentially expedite the drug discovery process by identifying already-approved drugs that might be used for new indications.
Collapse
Affiliation(s)
- Anddre Osmar Valdivia
- Department of Ophthalmology and Visual Neuroscience, University of Minnesota Medical School, Minneapolis, MN, USA
| | - Ye He
- Tianjin Key Laboratory of Retinal Functions and Diseases, Tianjin Branch of National Clinical Research Center for Ocular Disease, Eye Institute and School of Optometry, Tianjin Medical University Eye Hospital, Tianjin, China
| | - Xinjun Ren
- Tianjin Key Laboratory of Retinal Functions and Diseases, Tianjin Branch of National Clinical Research Center for Ocular Disease, Eye Institute and School of Optometry, Tianjin Medical University Eye Hospital, Tianjin, China
| | - Dejia Wen
- Tianjin Key Laboratory of Retinal Functions and Diseases, Tianjin Branch of National Clinical Research Center for Ocular Disease, Eye Institute and School of Optometry, Tianjin Medical University Eye Hospital, Tianjin, China
| | - Lijie Dong
- Tianjin Key Laboratory of Retinal Functions and Diseases, Tianjin Branch of National Clinical Research Center for Ocular Disease, Eye Institute and School of Optometry, Tianjin Medical University Eye Hospital, Tianjin, China
| | - Hossein Nazari
- Department of Ophthalmology and Visual Neuroscience, University of Minnesota Medical School, Minneapolis, MN, USA
| | - Xiaorong Li
- Tianjin Key Laboratory of Retinal Functions and Diseases, Tianjin Branch of National Clinical Research Center for Ocular Disease, Eye Institute and School of Optometry, Tianjin Medical University Eye Hospital, Tianjin, China
| |
Collapse
|
9
|
Li J, Li X, Guan F. What are the diagnostic capabilities of glycans for breast cancer? Expert Rev Mol Diagn 2023; 23:1-7. [PMID: 36705933 DOI: 10.1080/14737159.2023.2173577] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Affiliation(s)
- Jing Li
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, College of Life Sciences, Northwest University, Xi'an, Shaanxi, China
| | - Xiang Li
- Provincial Key Laboratory of Biotechnology, School of Medicine, Northwest University, Xi'an, Shaanxi, China
| | - Feng Guan
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, College of Life Sciences, Northwest University, Xi'an, Shaanxi, China
| |
Collapse
|
10
|
Mohammed NBB, Antonopoulos A, Dell A, Haslam SM, Dimitroff CJ. The pleiotropic role of galectin-3 in melanoma progression: Unraveling the enigma. Adv Cancer Res 2022; 157:157-193. [PMID: 36725108 PMCID: PMC9895887 DOI: 10.1016/bs.acr.2022.06.001] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Melanoma is a highly aggressive skin cancer with poor outcomes associated with distant metastasis. Intrinsic properties of melanoma cells alongside the crosstalk between melanoma cells and surrounding microenvironment determine the tumor behavior. Galectin-3 (Gal-3), a ß-galactoside-binding lectin, has emerged as a major effector in cancer progression, including melanoma behavior. Data from melanoma models and patient studies reveal that Gal-3 expression is dysregulated, both intracellularly and extracellularly, throughout the stages of melanoma progression. This review summarizes the most recent data and hypotheses on Gal-3 and its tumor-modulating functions, highlighting its role in driving melanoma growth, invasion, and metastatic colonization. It also provides insight into potential Gal-3-targeted strategies for melanoma diagnosis and treatment.
Collapse
Affiliation(s)
- Norhan B B Mohammed
- Department of Translational Medicine, Translational Glycobiology Institute at FIU (TGIF), Herbert Wertheim College of Medicine, Florida International University, Miami, FL, United States; Department of Medical Biochemistry, Faculty of Medicine, South Valley University, Qena, Egypt
| | | | - Anne Dell
- Department of Life Sciences, Imperial College London, London, United Kingdom
| | - Stuart M Haslam
- Department of Life Sciences, Imperial College London, London, United Kingdom
| | - Charles J Dimitroff
- Department of Translational Medicine, Translational Glycobiology Institute at FIU (TGIF), Herbert Wertheim College of Medicine, Florida International University, Miami, FL, United States.
| |
Collapse
|
11
|
Funkhouser AT, Strigenz AM, Blair BB, Miller AP, Shealy JC, Ewing JA, Martin JC, Funk CR, Edenfield WJ, Blenda AV. KIT Mutations Correlate with Higher Galectin Levels and Brain Metastasis in Breast and Non-Small Cell Lung Cancer. Cancers (Basel) 2022; 14:cancers14112781. [PMID: 35681762 PMCID: PMC9179545 DOI: 10.3390/cancers14112781] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Revised: 05/26/2022] [Accepted: 06/02/2022] [Indexed: 11/16/2022] Open
Abstract
To investigate a potential role for galectins as biomarkers that enable diagnosis or prognostication of breast or non-small cell lung cancer, the serum levels of galectins -1, -3, -7, -8, and -9 of cancer patients determined by ELISA assays were compared to the mutation status of 50 known cancer-critical genes, which were determined using multiplex PCR in tumors of the same patients. Mutations in the KIT proto-oncogene, which codes for the c-Kit protein, a receptor tyrosine kinase, correlated with higher levels of galectins -1, -3, -8, and -9 in breast cancer patients and galectin-1 in non-small cell lung cancer patients. Mutations in the KIT gene were more likely found in brain metastases from both of these primary cancers. The most common KIT mutation in our panel was p.M541L, a missense mutation in the transmembrane domain of the c-Kit protein. These results demonstrate an association between KIT oncogenic signaling and elevated serum galectins in patients with metastatic disease. Changes in protein trafficking and the glycocalyx composition of cancer cells may explain the observed alterations in galectin expression. This study can be useful for the targeted selection of receptor tyrosine kinase and galectin inhibitor anti-cancer treatments.
Collapse
Affiliation(s)
- Avery T Funkhouser
- Department of Biomedical Sciences, University of South Carolina School of Medicine Greenville, Greenville, SC 29605, USA
| | - Alexander M Strigenz
- Department of Biomedical Sciences, University of South Carolina School of Medicine Greenville, Greenville, SC 29605, USA
| | - Bailey B Blair
- Department of Biomedical Sciences, University of South Carolina School of Medicine Greenville, Greenville, SC 29605, USA
| | - Andrew P Miller
- Department of Biomedical Sciences, University of South Carolina School of Medicine Greenville, Greenville, SC 29605, USA
| | - Jonah C Shealy
- Department of Biomedical Sciences, University of South Carolina School of Medicine Greenville, Greenville, SC 29605, USA
| | - Joseph A Ewing
- Data Support Core, Prisma Health, Greenville, SC 29605, USA
| | - Julie C Martin
- Prisma Health Cancer Institute, Prisma Health, Greenville, SC 29605, USA
| | - Christopher R Funk
- Department of Hematology and Medical Oncology, Emory University School of Medicine, Atlanta, GA 30322, USA
| | | | - Anna V Blenda
- Department of Biomedical Sciences, University of South Carolina School of Medicine Greenville, Greenville, SC 29605, USA
- Prisma Health Cancer Institute, Prisma Health, Greenville, SC 29605, USA
| |
Collapse
|
12
|
Hozhabri H, Ghasemi Dehkohneh RS, Razavi SM, Razavi SM, Salarian F, Rasouli A, Azami J, Ghasemi Shiran M, Kardan Z, Farrokhzad N, Mikaeili Namini A, Salari A. Comparative analysis of protein-protein interaction networks in metastatic breast cancer. PLoS One 2022; 17:e0260584. [PMID: 35045088 PMCID: PMC8769308 DOI: 10.1371/journal.pone.0260584] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Accepted: 11/12/2021] [Indexed: 12/27/2022] Open
Abstract
Metastatic lesions leading causes of the majority of deaths in patients with the breast cancer. The present study aimed to provide a comprehensive analysis of the differentially expressed genes (DEGs) in the brain (MDA-MB-231 BrM2) and lung (MDA-MB-231 LM2) metastatic cell lines obtained from breast cancer patients compared with those who have primary breast cancer. We identified 981 and 662 DEGs for brain and lung metastasis, respectively. Protein-protein interaction (PPI) analysis revealed seven shared (PLCB1, FPR1, FPR2, CX3CL1, GABBR2, GPR37, and CXCR4) hub genes between brain and lung metastasis in breast cancer. Moreover, GNG2 and CXCL8, C3, and PTPN6 in the brain and SAA1 and CCR5 in lung metastasis were found as unique hub genes. Besides, five co-regulation of clusters via seven important co-expression genes (COL1A2, LUM, SPARC, THBS2, IL1B, CXCL8, THY1) were identified in the brain PPI network. Clusters screening followed by biological process (BP) function and pathway enrichment analysis for both metastatic cell lines showed that complement receptor signalling, acetylcholine receptor signalling, and gastric acid secretion pathways were common between these metastases, whereas other pathways were site-specific. According to our findings, there are a set of genes and functional pathways that mark and mediate breast cancer metastasis to the brain and lungs, which may enable us understand the molecular basis of breast cancer development in a deeper levele to the brain and lungs, which may help us gain a more complete understanding of the molecular underpinnings of breast cancer development.
Collapse
Affiliation(s)
- Hossein Hozhabri
- Institute of Biochemistry and Biophysics, University of Tehran, Tehran, Iran
- Salari Institute of Cognitive and Behavioral Disorders (SICBD), Karaj, Alborz, Iran
- Systems Biology Research Lab, Bioinformatics Group, Systems Biology of the Next Generation Company (SBNGC), Qom, Iran
| | - Roxana Sadat Ghasemi Dehkohneh
- Salari Institute of Cognitive and Behavioral Disorders (SICBD), Karaj, Alborz, Iran
- Systems Biology Research Lab, Bioinformatics Group, Systems Biology of the Next Generation Company (SBNGC), Qom, Iran
- Department of Cell and Molecular Biology and Microbiology, Faculty of Biological Science and Technology, University of Isfahan, Isfahan, Iran
| | - Seyed Morteza Razavi
- Salari Institute of Cognitive and Behavioral Disorders (SICBD), Karaj, Alborz, Iran
- Systems Biology Research Lab, Bioinformatics Group, Systems Biology of the Next Generation Company (SBNGC), Qom, Iran
- Department of Cell and Molecular Biology, Faculty of Biological Sciences, Kharazmi University, Tehran, Iran
| | - S. Mostafa Razavi
- Department of Chemical, Biomolecular and Corrosion Engineering, The University of Akron, Akron, Ohio, United States of America
| | - Fatemeh Salarian
- Systems Biology Research Lab, Bioinformatics Group, Systems Biology of the Next Generation Company (SBNGC), Qom, Iran
- Department of Cellular Biotechnology, Cell Science Research Center, Royan Institute for Biotechnology, ACECR, Isfahan, Iran
| | - Azade Rasouli
- Systems Biology Research Lab, Bioinformatics Group, Systems Biology of the Next Generation Company (SBNGC), Qom, Iran
- Department of Toxicology and Pharmacology, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Jalil Azami
- Systems Biology Research Lab, Bioinformatics Group, Systems Biology of the Next Generation Company (SBNGC), Qom, Iran
- Faculty of Veterinary Medicine, Urmia University, Urmia, Iran
| | - Melika Ghasemi Shiran
- Salari Institute of Cognitive and Behavioral Disorders (SICBD), Karaj, Alborz, Iran
- Systems Biology Research Lab, Bioinformatics Group, Systems Biology of the Next Generation Company (SBNGC), Qom, Iran
- Department of Biology, Faculty of Sciences, Central Tehran Branch, Islamic Azad University, Tehran, Iran
| | - Zahra Kardan
- Systems Biology Research Lab, Bioinformatics Group, Systems Biology of the Next Generation Company (SBNGC), Qom, Iran
- Department of Cellular Molecular Biology, Faculty of Life Science and Biotechnology, Shahid Beheshti University, Tehran, Iran
| | - Negar Farrokhzad
- Systems Biology Research Lab, Bioinformatics Group, Systems Biology of the Next Generation Company (SBNGC), Qom, Iran
- Department of Biology, Faculty of Sciences, University of Guilan, Rasht, Iran
| | - Arsham Mikaeili Namini
- Systems Biology Research Lab, Bioinformatics Group, Systems Biology of the Next Generation Company (SBNGC), Qom, Iran
- Department of Animal Biology, Faculty of Biological Sciences, Kharazmi University, Tehran, Iran
| | - Ali Salari
- Salari Institute of Cognitive and Behavioral Disorders (SICBD), Karaj, Alborz, Iran
- Systems Biology Research Lab, Bioinformatics Group, Systems Biology of the Next Generation Company (SBNGC), Qom, Iran
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| |
Collapse
|
13
|
A Novel Hypoxic-Angiogenesis-Immune-Related Gene Model for Prognostic and Therapeutic Effect Prediction in Hepatocellular Carcinoma Patients. DISEASE MARKERS 2022; 2022:9428660. [PMID: 35069936 PMCID: PMC8769836 DOI: 10.1155/2022/9428660] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Accepted: 12/07/2021] [Indexed: 12/04/2022]
Abstract
Background Hepatocellular carcinoma (HCC) is one of the most heterogeneous malignant tumors that have been discovered so far, which makes the prognostic prediction difficult. The hypoxia, angiogenesis, and immunity-related genes (HAIRGs) are closely related to the development of liver cancer. However, the prognostic and treatment effect of hypoxia, angiogenesis, and immunity-related genes in HCC continues to be further clarified. Methods The gene expression quantification data and clinical information in patients with liver cancer were downloaded from the TCGA database, and HAIRG signature was built by using the least absolute shrinkage and selection operator (LASSO) technique. Patient from the ICGC database validated the model. Then, tumor immune dysfunction and exclusion (TIDE) algorithm was applied to estimate the clinical response to immunotherapy and the sensitivity of drugs was evaluated by the half-maximal inhibitory concentration (IC50). Result The HAIRGs were identified between the HCC patients and normal patients in the TCGA database. In univariate Cox regression analysis, seventeen differentially expressed genes (DEGs) were associated with overall survival (OS). An eight HAIRG signature model was constructed and was used to divide the patients into two groups according to the median value of the risk score base on the TCGA dataset. Patients in the high-risk group had a significant reduction in OS compared to those in the low-risk group (P < 0.001 in the TCGA, P < 0.001 in the ICGC). For TCGA and ICGC databases of univariate Cox regression analyses, the risk score was used as an independent predictor of OS (HR > 1, P < 0.001). Functional analysis showed that the relevant immune pathways and immune responses were enriched, cellular component analysis showed that the immunoglobulin complex and other related substances were enriched, and immune status existed a difference in the high- and low-risk groups. Then, the tumor immune dysfunction and exclusion (TIDE) algorithm presented differences in immune response in the high- and low-risk groups (P < 0.05), and based on drug sensitivity prediction, patients in the high-risk group were more sensitive to cisplatin compared to those in the low-risk group in both the TCGA and ICGC cohorts (P < 0.05). Conclusions HAIRG signature can be utilized for prognostic prediction in HCC, while it can be considered a prediction model for clinical evaluation of immunotherapy response and chemotherapy sensitivity in HCC.
Collapse
|
14
|
Lu X. Structure and functions of T-cell immunoglobulin-domain and mucin- domain protein 3 in cancer. Curr Med Chem 2021; 29:1851-1865. [PMID: 34365943 DOI: 10.2174/0929867328666210806120904] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Revised: 06/17/2021] [Accepted: 06/21/2021] [Indexed: 11/22/2022]
Abstract
BACKGROUND T-cell immunoglobulin (Ig)-domain and mucin-domain (TIM) proteins represent a family of receptors expressed on T-cells that play essential cellular immunity roles. The TIM proteins span across the membrane belonging to type I transmembrane proteins. The N terminus contains an Ig-like V-type domain and a Ser/Thr-rich mucin stalk as a co-inhibitory receptor. The C-terminal tail oriented toward the cytosol predominantly mediates intracellular signaling. METHODS This review discusses the structural features and functions of TIM-3, specifically on its role in mediating immune responses in different cell types, and the rationale for TIM-3-targeted cancer immunotherapy. RESULTS TIM-3 has gained significant importance to be a potential biomarker in cancer immunotherapy. It has been shown that blockade with checkpoint inhibitors promotes anti-tumor immunity and inhibits tumor growth in several preclinical tumor models. CONCLUSION TIM-3 is an immune regulating molecule expressed on several cell types, including IFNγ-producing T-cells, FoxP3+ Treg cells, and innate immune cells. The roles of TIM-3 in immunosuppression support its merit as a target for cancer immunotherapy.
Collapse
Affiliation(s)
- Xinjie Lu
- The Mary and Garry Weston Molecular Immunology Laboratory, Thrombosis Research Institute, London, SW3 6LR. United Kingdom
| |
Collapse
|
15
|
Boutas I, Potiris A, Makrakis E, Messaropoulos P, Papaioannou GK, Kalantaridou SΝ. The expression of Galectin-3 in breast cancer and its association with metastatic disease: a systematic review of the literature. Mol Biol Rep 2021; 48:807-815. [PMID: 33398681 DOI: 10.1007/s11033-020-06122-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Accepted: 12/22/2020] [Indexed: 12/24/2022]
Abstract
Breast cancer is the most common form of cancer and the second highest cause of cancer mortality in female patients. The significance of the expression of Galectin-3 has been correlated with various malignancy types and in data from several research papers, the expression of Galectin-3 has been associated with the progression and metastasis of breast cancer. In the present study, the authors' goal is to identify whether the expression of Galectin-3 in breast cancer can be associated with the presence and/or recurrence of a metastatic disease. Both Scopus and PubMed databases were utilized, by inputting the following combination of keywords: (((Breast) AND Metastasis)) AND ((Galectin 3) OR Galectin-3). The time of publication and text availability were not considered when searching the databases and all relevant articles in English were initially accepted. We included one case-control study, three retrospective case studies and one retrospective cohort study. In two of the included studies, the levels in concentration of Galectin-3 were not correlated with a significant difference in prognosis. In two studies, the lacking in expression of Galectin-3 was associated with a worse prognosis and in one of the studies selected, the elevated levels of Galectin-3 were correlated with recurrence of disease in triple negative breast cancer cases. For most of the studies selected for this review, the results were contradictory in regard to the role of Galectin-3 for prognosis and metastatic potential in female breast cancer patients. It is still unclear, whether Galectin-3 can be used as a prognostic marker for advanced breast cancer disease.
Collapse
Affiliation(s)
- Ioannis Boutas
- Third Department of Obstetrics and Gynecology, Attikon University Hospital, Medical School of the National and Kapodistrian University of Athens, Athens, Greece
| | - Anastasios Potiris
- Third Department of Obstetrics and Gynecology, Attikon University Hospital, Medical School of the National and Kapodistrian University of Athens, Athens, Greece.
| | - Evangelos Makrakis
- Third Department of Obstetrics and Gynecology, Attikon University Hospital, Medical School of the National and Kapodistrian University of Athens, Athens, Greece
| | - Pantelis Messaropoulos
- Third Department of Obstetrics and Gynecology, Attikon University Hospital, Medical School of the National and Kapodistrian University of Athens, Athens, Greece
| | - Georgios-Konstantinos Papaioannou
- Third Department of Obstetrics and Gynecology, Attikon University Hospital, Medical School of the National and Kapodistrian University of Athens, Athens, Greece
| | - Sophia Ν Kalantaridou
- Third Department of Obstetrics and Gynecology, Attikon University Hospital, Medical School of the National and Kapodistrian University of Athens, Athens, Greece
| |
Collapse
|
16
|
Nishida-Aoki N, Tominaga N, Kosaka N, Ochiya T. Altered biodistribution of deglycosylated extracellular vesicles through enhanced cellular uptake. J Extracell Vesicles 2020; 9:1713527. [PMID: 32082512 PMCID: PMC7006786 DOI: 10.1080/20013078.2020.1713527] [Citation(s) in RCA: 63] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2019] [Revised: 12/30/2019] [Accepted: 01/02/2020] [Indexed: 01/15/2023] Open
Abstract
Extracellular vesicles (EVs) from cancer are delivered both proximal and distal organs. EVs are highly glycosylated at the surface where EVs interact with cells and therefore has an impact on their properties and biological functions. Aberrant glycosylation in cancer is associated with cancer progression and metastasis. However, the biological function of glycosylation on the surface of EV is uncovered. We first demonstrated differential glycosylation profiles of EVs and their originated cells, and distinct glycosylation profiles in a brain-metastatic subline BMD2a from its parental human breast cancer cell line, MDA-MB-231-luc-D3H2LN by lectin blot. We then investigated the roles of surface glycoconjugates on EV uptake. N- and/or O-glycosylation removal of fluorescent-labelled BMD2a EVs enhanced cellular uptake to endothelial cells, suggesting that surface glycosylation has inhibitory effects on cellular uptake. Biodistribution of glycosylation-deprived BMD2a EVs administrated intravenously into mice was further analysed ex vivo using near-infrared lipophilic dye. EVs treated with O-deglycosylation enzymes enhanced the accumulation of EVs to the lungs after 24 h from the injection, while N-deglycosylation did not markedly alter biodistribution. As the lungs are first organs in which intravenous blood flows, we suggest that surface glycosylation of cancer-derived EVs avoid promiscuous adhesion to proximal tissues to be delivered to distant organs.
Collapse
Affiliation(s)
- Nao Nishida-Aoki
- Division of Molecular and Cellular Medicine, National Cancer Center Research Institute, Tokyo, Japan.,Human Biology Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Naoomi Tominaga
- Division of Molecular and Cellular Medicine, National Cancer Center Research Institute, Tokyo, Japan.,Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Nobuyoshi Kosaka
- Division of Molecular and Cellular Medicine, National Cancer Center Research Institute, Tokyo, Japan.,Department of Molecular and Cellular Medicine, Institute of Medical Science, Tokyo Medical University, Tokyo, Japan
| | - Takahiro Ochiya
- Division of Molecular and Cellular Medicine, National Cancer Center Research Institute, Tokyo, Japan.,Department of Molecular and Cellular Medicine, Institute of Medical Science, Tokyo Medical University, Tokyo, Japan
| |
Collapse
|
17
|
Integrated Transcriptomics, Proteomics, and Glycomics Reveals the Association between Up-regulation of Sialylated N-glycans/Integrin and Breast Cancer Brain Metastasis. Sci Rep 2019; 9:17361. [PMID: 31758065 PMCID: PMC6874669 DOI: 10.1038/s41598-019-53984-8] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2019] [Accepted: 11/07/2019] [Indexed: 12/21/2022] Open
Abstract
Breast cancer brain metastasis has been recognized as one of the central issues in breast cancer research. The elucidation of the processes and pathways that mediate this step will provide important clues for a better understanding of breast cancer metastasis. Increasing evidence suggests that aberrant glycosylation patterns greatly contribute to cell invasion and cancer metastasis. Herein, we combined next-generation RNA sequencing with liquid chromatography-tandem mass spectrometry-based proteomic and N-glycomic analysis from five breast cancer cell lines and one brain cancer cell line to investigate the possible mechanisms of breast cancer brain metastasis. The genes/proteins associated with cell movement were highlighted in breast cancer brain metastasis. The integrin signaling pathway and the up-regulation of α-integrin (ITGA2, ITGA3) were associated with the brain metastatic process. 12 glycogenes showed unique expression in 231BR, which could result in an increase of sialylation during brain metastasis. In agreement with the changes of glycogenes, 60 out of 63 N-glycans that were identified exhibited differential expression among cell lines. The correlation between glycogenes and glycans revealed the importance of sialylation and sialylated glycans in breast cancer brain metastasis. Highly sialylated N-glycans, which were up-regulated in brain-seeking cell line 231BR, likely play a role in brain metastasis.
Collapse
|
18
|
Kopec M, Imiela A, Abramczyk H. Monitoring glycosylation metabolism in brain and breast cancer by Raman imaging. Sci Rep 2019; 9:166. [PMID: 30655566 PMCID: PMC6336853 DOI: 10.1038/s41598-018-36622-7] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2018] [Accepted: 11/20/2018] [Indexed: 12/11/2022] Open
Abstract
We have shown that Raman microspectroscopy is a powerful method for visualization of glycocalyx offering cellular interrogation without staining, unprecedented spatial and spectral resolution, and biochemical information. We showed for the first time that Raman imaging can be used to distinguish successfully between glycosylated and nonglycosylated proteins in normal and cancer tissue. Thousands of protein, lipid and glycan species exist in cells and tissues and their metabolism is monitored via numerous pathways, networks and methods. The metabolism can change in response to cellular environment alterations, such as development of a disease. Measuring such alterations and understanding the pathways involved are crucial to fully understand cellular metabolism in cancer development. In this paper Raman markers of glycogen, glycosaminoglycan, chondroitin sulfate, heparan sulfate proteoglycan were identified based on their vibrational signatures. High spatial resolution of Raman imaging combined with chemometrics allows separation of individual species from many chemical components present in each cell. We have found that metabolism of proteins, lipids and glycans is markedly deregulated in breast (adenocarcinoma) and brain (medulloblastoma) tumors. We have identified two glycoforms in the normal breast tissue and the malignant brain tissue in contrast to the breast cancer tissue where only one glycoform has been identified.
Collapse
Affiliation(s)
- M Kopec
- Lodz University of Technology, Faculty of Chemistry, Institute of Applied Radiation Chemistry, Laboratory of Laser Molecular Spectroscopy, Wroblewskiego 15, 93-590, Lodz, Poland
| | - A Imiela
- Lodz University of Technology, Faculty of Chemistry, Institute of Applied Radiation Chemistry, Laboratory of Laser Molecular Spectroscopy, Wroblewskiego 15, 93-590, Lodz, Poland
| | - H Abramczyk
- Lodz University of Technology, Faculty of Chemistry, Institute of Applied Radiation Chemistry, Laboratory of Laser Molecular Spectroscopy, Wroblewskiego 15, 93-590, Lodz, Poland.
| |
Collapse
|
19
|
Ghapanchi J, Andisheh-Tadbir A, Torkaman P, Malekzadeh M, Mardani M. Evaluation of the serum levels of galectin-3 in patients with oral lichen planus disease. Oral Dis 2019; 25:466-470. [DOI: 10.1111/odi.13012] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2018] [Revised: 10/31/2018] [Accepted: 11/20/2018] [Indexed: 11/29/2022]
Affiliation(s)
- Janan Ghapanchi
- Department of Oral and Maxillofacial Medicine, School of Dentistry; Shiraz University of Medical Sciences; Shiraz Iran
| | - Azadeh Andisheh-Tadbir
- Oral and Dental Disease Research Center, School of Dentistry; Shiraz University of Medical Sciences; Shiraz Iran
| | - Pooriya Torkaman
- Department of Oral and Maxillofacial Medicine, School of Dentistry; Shiraz University of Medical Sciences; Shiraz Iran
| | - Mahyar Malekzadeh
- Shiraz Institute for Cancer Research, School of Medicine; Shiraz University of Medical Sciences; Shiraz Iran
| | - Maryam Mardani
- Oral and Dental Disease Research Center, School of Dentistry; Shiraz University of Medical Sciences; Shiraz Iran
| |
Collapse
|
20
|
Wang H, Song X, Huang Q, Xu T, Yun D, Wang Y, Hu L, Yan Y, Chen H, Lu D, Chen J. LGALS3 Promotes Treatment Resistance in Glioblastoma and Is Associated with Tumor Risk and Prognosis. Cancer Epidemiol Biomarkers Prev 2018; 28:760-769. [PMID: 30341098 DOI: 10.1158/1055-9965.epi-18-0638] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2018] [Revised: 07/28/2018] [Accepted: 10/15/2018] [Indexed: 11/16/2022] Open
Abstract
BACKGROUND LGALS3 promotes tumor progression in diverse cancers. However, the involvement of LGALS3 in glioblastoma has not yet been broadly illuminated. METHODS Microarray was performed to detect the gene expression profiles of radioresistance in T98G cells and identified a universally upregulated gene, LGALS3. The impact of LGALS3 on the survival of glioblastoma cells facing ionizing irradiation or temozolomide was investigated by the Cell Counting Kit-8 (CCK-8). A total of 120 glioblastoma cases were collected to analyze the relationship between LGALS3 expression and patient prognosis. Another 961 patients with glioma and 1,351 healthy controls were recruited to study the association of SNPs across the LGALS3 gene with glioblastoma susceptibility. The functional SNP sites were also studied in cellular experiments. RESULTS An effective protection of LGALS3 from ionizing irradiation or temozolomide-induced cell death in T98G and U251 cells was found. In addition, high expression of LGALS3 could work as an independent risk factor for survival of patients with glioblastoma. Two SNP sites (rs4644 and rs4652) across the LGALS3 gene were associated with increased risk for glioblastoma, and the C allele of rs4652 and the A allele of rs4644 could enhance glioblastoma resistance to radio-chemotherapy, but not cell proliferation. CONCLUSIONS Our results suggest that LGALS3 is an important biomarker influencing glioblastoma risk and prognosis and a potential target for treating the malignancy, especially ones with resistance against the standard therapy. IMPACT LGALS3 promotes glioblastoma cells' resistance to ionizing irradiation and temozolomide and predicts poor prognosis. Targeting LGALS3 may limit the therapeutic resistance in glioblastoma and increase patient survival.
Collapse
Affiliation(s)
- Hongxiang Wang
- Department of Neurosurgery, Shanghai Institute of Neurosurgery, Changzheng Hospital, Second Military Medical University, Shanghai, China
| | - Xiao Song
- Department of Thoracic Surgery, Lung Cancer Diagnosis and Treatment Center, Shanghai Pulmonary Hospital, Tongji University, Shanghai, China
| | - Qilin Huang
- Department of Neurosurgery, Shanghai Institute of Neurosurgery, Changzheng Hospital, Second Military Medical University, Shanghai, China
| | - Tao Xu
- Department of Neurosurgery, Shanghai Institute of Neurosurgery, Changzheng Hospital, Second Military Medical University, Shanghai, China
| | - Dapeng Yun
- State Key Laboratory of Genetic Engineering and MOE Key Laboratory of Contemporary Anthropology, School of Life Sciences, Fudan University, Shanghai, China
| | - Yuqi Wang
- State Key Laboratory of Genetic Engineering and MOE Key Laboratory of Contemporary Anthropology, School of Life Sciences, Fudan University, Shanghai, China
| | - Lingna Hu
- State Key Laboratory of Genetic Engineering and MOE Key Laboratory of Contemporary Anthropology, School of Life Sciences, Fudan University, Shanghai, China
| | - Yong Yan
- Department of Neurosurgery, Shanghai Institute of Neurosurgery, Changzheng Hospital, Second Military Medical University, Shanghai, China
| | - Hongyan Chen
- State Key Laboratory of Genetic Engineering and MOE Key Laboratory of Contemporary Anthropology, School of Life Sciences, Fudan University, Shanghai, China
| | - Daru Lu
- State Key Laboratory of Genetic Engineering and MOE Key Laboratory of Contemporary Anthropology, School of Life Sciences, Fudan University, Shanghai, China.
| | - Juxiang Chen
- Department of Neurosurgery, Shanghai Institute of Neurosurgery, Changzheng Hospital, Second Military Medical University, Shanghai, China.
| |
Collapse
|
21
|
Cancer Therapy Due to Apoptosis: Galectin-9. Int J Mol Sci 2017; 18:ijms18010074. [PMID: 28045432 PMCID: PMC5297709 DOI: 10.3390/ijms18010074] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2016] [Revised: 11/25/2016] [Accepted: 12/27/2016] [Indexed: 12/22/2022] Open
Abstract
Dysregulation of apoptosis is a major hallmark in cancer biology that might equip tumors with a higher malignant potential and chemoresistance. The anti-cancer activities of lectin, defined as a carbohydrate-binding protein that is not an enzyme or antibody, have been investigated for over a century. Recently, galectin-9, which has two distinct carbohydrate recognition domains connected by a linker peptide, was noted to induce apoptosis in thymocytes and immune cells. The apoptosis of these cells contributes to the development and regulation of acquired immunity. Furthermore, human recombinant galectin-9, hG9NC (null), which lacks an entire region of the linker peptide, was designed to resist proteolysis. The hG9NC (null) has demonstrated anti-cancer activities, including inducing apoptosis in hematological, dermatological and gastrointestinal malignancies. In this review, the molecular characteristics, history and apoptosis-inducing potential of galectin-9 are described.
Collapse
|
22
|
Santos SN, Junqueira MS, Francisco G, Vilanova M, Magalhães A, Baruffi MD, Chammas R, Harris AL, Reis CA, Bernardes ES. O-glycan sialylation alters galectin-3 subcellular localization and decreases chemotherapy sensitivity in gastric cancer. Oncotarget 2016; 7:83570-83587. [PMID: 27835877 PMCID: PMC5347789 DOI: 10.18632/oncotarget.13192] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2015] [Accepted: 10/21/2016] [Indexed: 12/12/2022] Open
Abstract
ST6GalNAc-I, the sialyltransferase responsible for sialyl-Tn (sTn) synthesis, has been previously reported to be positively associated with cancer aggressiveness. Here we describe a novel sTn-dependent mechanism for chemotherapeutic resistance. We show that sTn protects cancer cells against chemotherapeutic-induced cell death by decreasing the interaction of cell surface glycan receptors with galectin-3 and increasing its intracellular accumulation. Moreover, exogenously added galectin-3 potentiated the chemotherapeutics-induced cytotoxicity in sTn non-expressing cells, while sTn overexpressing cells were protected. We also found that the expression of sTn was associated with a reduction in galectin-3-binding sites in human gastric samples tumors. ST6GalNAc-I knockdown restored galectin-3-binding sites on the cell surface and chemotherapeutics sensibility. Our results clearly demonstrate that an interruption of O-glycans extension caused by ST6GalNAc-I enzymatic activity leads to tumor cells resistance to chemotherapeutic drugs, highlighting the need for the development of novel strategies to target galectin-3 and/or ST6GalNAc-I.
Collapse
MESH Headings
- Animals
- Antigens, Tumor-Associated, Carbohydrate/genetics
- Antigens, Tumor-Associated, Carbohydrate/metabolism
- Antineoplastic Agents/pharmacology
- Blood Proteins
- Cell Line, Tumor
- Cell Proliferation
- Cisplatin/pharmacology
- Dose-Response Relationship, Drug
- Drug Resistance, Neoplasm
- Galectin 3/metabolism
- Galectins
- Glycosylation
- Humans
- Mice, Inbred BALB C
- Mice, Nude
- Protein Processing, Post-Translational
- Protein Transport
- RNA Interference
- Sialyltransferases/genetics
- Sialyltransferases/metabolism
- Stomach Neoplasms/drug therapy
- Stomach Neoplasms/genetics
- Stomach Neoplasms/metabolism
- Stomach Neoplasms/pathology
- Time Factors
- Transfection
- Tumor Burden
Collapse
Affiliation(s)
- Sofia N. Santos
- Department of Radiopharmacy, Nuclear Energy Research Institute, Radiopharmacy Center, São Paulo, Brazil
| | - Mara S. Junqueira
- Department of Center for Translational Oncology Cellular, Biology Group, Center for Translational Oncology, Cancer Institute of the State of Sao Paulo-ICESP, Brazil
| | - Guilherme Francisco
- Department of Center for Translational Oncology Cellular, Biology Group, Center for Translational Oncology, Cancer Institute of the State of Sao Paulo-ICESP, Brazil
| | - Manuel Vilanova
- I3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Portugal
- IBMC Instituto de Biologia Molecular e Celular, Universidade do Porto, Portugal
- ICBAS-UP – Instituto de Ciências Biomédicas Abel Salazar, University of Porto, Porto, Portugal
| | - Ana Magalhães
- I3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Portugal
- Department of Glycobiology in Cancer, IPATIMUP - Institute of Molecular Pathology and Immunology from the University of Porto, Porto, Portugal
| | - Marcelo Dias Baruffi
- Department of Clinical, Toxicological and Bromatological Analysis, Faculdade de Ciências Farmaceuticas de Ribeirão Preto, Universidade de São Paulo, Brazil
| | - Roger Chammas
- Department of Center for Translational Oncology Cellular, Biology Group, Center for Translational Oncology, Cancer Institute of the State of Sao Paulo-ICESP, Brazil
| | - Adrian L. Harris
- Department of Medical Oncology, Molecular Oncology Laboratories, Weatherall Institute of Molecular Medicine, University of Oxford, Oxford OX3 9DS, UK
| | - Celso A. Reis
- I3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Portugal
- ICBAS-UP – Instituto de Ciências Biomédicas Abel Salazar, University of Porto, Porto, Portugal
- Department of Glycobiology in Cancer, IPATIMUP - Institute of Molecular Pathology and Immunology from the University of Porto, Porto, Portugal
- Department of Pathology and Oncology, Medical Faculty, University of Porto, Portugal
| | - Emerson S. Bernardes
- Department of Radiopharmacy, Nuclear Energy Research Institute, Radiopharmacy Center, São Paulo, Brazil
| |
Collapse
|
23
|
Kölbl AC, Andergassen U, Jeschke U. The Role of Glycosylation in Breast Cancer Metastasis and Cancer Control. Front Oncol 2015; 5:219. [PMID: 26528431 PMCID: PMC4602128 DOI: 10.3389/fonc.2015.00219] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2015] [Accepted: 09/24/2015] [Indexed: 01/08/2023] Open
Abstract
Glycosylation and its correlation to the formation of remote metastasis in breast cancer had been an important scientific topic in the last 25 years. With the development of new analytical techniques, new insights were gained on the mechanisms underlying metastasis formation and the role of aberrant glycosylation within. Mucin-1 and Galectin were recognized as key players in glycosylation. Interestingly, aberrant carbohydrate structures seem to support the development of brain metastasis in breast cancer patients, as changes in glycosylation structures facilitate an overcoming of blood–brain barrier. Changes in the gene expression of glycosyltransferases are the leading cause for a modification of carbohydrate chains, so that also altered gene expression plays a role for glycosylation. In consequence, glycosylation and changes within can be useful for cancer diagnosis, determination of tumor stage, and prognosis, but can as well be targets for therapeutic strategies. Thus, further research on this topic would worthwhile for cancer combating.
Collapse
Affiliation(s)
- Alexandra C Kölbl
- Department of Obstetrics and Gynaecology, Ludwig-Maximilians-University of Munich , Munich , Germany
| | - Ulrich Andergassen
- Department of Obstetrics and Gynaecology, Ludwig-Maximilians-University of Munich , Munich , Germany
| | - Udo Jeschke
- Department of Obstetrics and Gynaecology, Ludwig-Maximilians-University of Munich , Munich , Germany
| |
Collapse
|
24
|
He K, Lv W, Zheng D, Cheng F, Zhou T, Ye S, Ban Q, Ying Q, Huang B, Chen L, Wu G, Liu D. The stromal genome heterogeneity between breast and prostate tumors revealed by a comparative transcriptomic analysis. Oncotarget 2015; 6:8687-97. [PMID: 25826086 PMCID: PMC4496176 DOI: 10.18632/oncotarget.3478] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2015] [Accepted: 02/12/2015] [Indexed: 11/25/2022] Open
Abstract
Stromal microenvironment increases tumor cell survival, proliferation and migration, and promotes angiogenesis. In order to provide comprehensive information on the stromal heterogeneity of diverse tumors, here we employed the microarray datasets of human invasive breast and prostate cancer-associated stromals and applied Gene Set Enrichment Analysis (GSEA) to compare the gene expression profiles between them. As a result, 8 up-regulated pathways and 73 down-regulated pathways were identified in the breast tumor stroma, while 32 up-regulated pathways and 18 down-regulated pathways were identified in the prostate tumor stroma. Only 9 pathways such as tryptophan metabolism were commonly up or down regulated, but most of them (including ABC transporters) were specific for these two tumors. Several essential tumors stromal marker genes were also significantly identified. For example, CDH3 was significantly up-regulated in the stromals of both breast and prostate tumors, however EGFR was only significantly down-regulated in the stromal of breast tumor. Our study would be helpful for future therapeutic and predictive applications in breast and prostate cancers.
Collapse
Affiliation(s)
- Kan He
- Center for Stem Cell and Translational Medicine, School of Life Sciences, Anhui University, Hefei City, Anhui, China
| | - Wenwen Lv
- Center for Stem Cell and Translational Medicine, School of Life Sciences, Anhui University, Hefei City, Anhui, China
| | - Dongni Zheng
- Center for Stem Cell and Translational Medicine, School of Life Sciences, Anhui University, Hefei City, Anhui, China
| | - Fei Cheng
- Center for Stem Cell and Translational Medicine, School of Life Sciences, Anhui University, Hefei City, Anhui, China
| | - Tao Zhou
- Center for Stem Cell and Translational Medicine, School of Life Sciences, Anhui University, Hefei City, Anhui, China
| | - Shoudong Ye
- Center for Stem Cell and Translational Medicine, School of Life Sciences, Anhui University, Hefei City, Anhui, China
- Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research, Department of Cell and Neurobiology, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
- Department of Molecular Genetics, Shanghai Medical School, Fudan University, Shanghai, China
| | - Qian Ban
- Center for Stem Cell and Translational Medicine, School of Life Sciences, Anhui University, Hefei City, Anhui, China
| | - Qilong Ying
- Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research, Department of Cell and Neurobiology, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - Bei Huang
- Center for Stem Cell and Translational Medicine, School of Life Sciences, Anhui University, Hefei City, Anhui, China
| | - Lei Chen
- Center for Stem Cell and Translational Medicine, School of Life Sciences, Anhui University, Hefei City, Anhui, China
| | - Guohua Wu
- Laboratory of Quality & Safety Risk Assessment for Sericultural Products and Edible Insects, Ministry of Agriculture, College of Biotechnology and Sericultural Research Institute, Jiangsu University of Science and Technology, Zhenjiang, China
| | - Dahai Liu
- Center for Stem Cell and Translational Medicine, School of Life Sciences, Anhui University, Hefei City, Anhui, China
| |
Collapse
|
25
|
Thijssen VL, Heusschen R, Caers J, Griffioen AW. Galectin expression in cancer diagnosis and prognosis: A systematic review. Biochim Biophys Acta Rev Cancer 2015; 1855:235-47. [PMID: 25819524 DOI: 10.1016/j.bbcan.2015.03.003] [Citation(s) in RCA: 159] [Impact Index Per Article: 15.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2015] [Revised: 03/14/2015] [Accepted: 03/16/2015] [Indexed: 02/07/2023]
Abstract
Galectins are a family of proteins that bind to specific glycans thereby deciphering the information captured within the glycome. In the last two decades, several galectin family members have emerged as versatile modulators of tumor progression. This has initiated the development and preclinical assessment of galectin-targeting compounds. With the first compounds now entering clinical trials it is pivotal to gain insight in the diagnostic and prognostic value of galectins in cancer as this will allow a more rational selection of the patients that might benefit most from galectin-targeted therapies. Here, we present a systematic review of galectin expression in human cancer patients. Malignant transformation is frequently associated with altered galectin expression, most notably of galectin-1 and galectin-3. In most cancers, increased galectin-1 expression is associated with poor prognosis while elevated galectin-9 expression is emerging as a marker of favorable disease outcome. The prognostic value of galectin-3 appears to be tumor type dependent and the other galectins require further investigation. Regarding the latter, additional studies using larger patient cohorts are essential to fully unravel the diagnostic and prognostic value of galectin expression. Furthermore, to better compare different findings, consensus should be reached on how to assess galectin expression, not only with regard to localization within the tissue and within cellular compartments but also regarding alternative splicing and genomic variations. Finally, linking galectin expression and function to aberrant glycosylation in cancer cells will improve our understanding of how these versatile proteins can be exploited for diagnostic, prognostic and even therapeutic purposes in cancer patients.
Collapse
Affiliation(s)
- Victor L Thijssen
- Angiogenesis Laboratory, Department Medical Oncology, VU University Medical Center, Amsterdam, the Netherlands; Angiogenesis Laboratory, Department of Radiation Oncology, VU University Medical Center, Amsterdam, the Netherlands.
| | - Roy Heusschen
- Laboratory of Hematology, GIGA-Research, University of Liege, Liege, Belgium
| | - Jo Caers
- Laboratory of Hematology, GIGA-Research, University of Liege, Liege, Belgium
| | - Arjan W Griffioen
- Angiogenesis Laboratory, Department Medical Oncology, VU University Medical Center, Amsterdam, the Netherlands
| |
Collapse
|
26
|
Jiang SS, Weng DS, Wang QJ, Pan K, Zhang YJ, Li YQ, Li JJ, Zhao JJ, He J, Lv L, Pan QZ, Xia JC. Galectin-3 is associated with a poor prognosis in primary hepatocellular carcinoma. J Transl Med 2014; 12:273. [PMID: 25260879 PMCID: PMC4179848 DOI: 10.1186/s12967-014-0273-3] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2014] [Accepted: 09/19/2014] [Indexed: 02/05/2023] Open
Abstract
BACKGROUND Galectin-3, a member of the beta-galactoside-binding lectin family, is a multifunctional protein with various biological functions, including the proliferation and differentiation of tumor cells, angiogenesis, cancer progression, and metastasis. We aimed to clarify if expression of galectin-3 is related to the clinicopathological characteristics and prognosis of hepatocellular carcinoma (HCC) patients, and to explore the possible mechanisms of galectin-3 in hepatocellular carcinoma. METHODS First, we investigated galectin-3 mRNA and protein expression by using RT-PCR and Western blotting. Second, tissues from 165 HCC patients were used to evaluate clinicopathological characteristics and prognosis through immunohistochemical analyses. Furthermore, the functions of galectin-3 were analyzed with respect to the proliferation, cell cycle,apoptosis, migration, and invasion of HCC cell lines. Finally, we analyzed galectin-3 expression and micro-vessel density (MVD) by immunohistochemistry (IHC) to find its correlation with angiogenesis in Hepatocellular Carcinoma. Flow cytometer was used to explore apoptosis and Western-blot was used to detect the pathway proteins of apoptosis. RESULTS Galectin-3 showed high expression at the mRNA and protein levels in HCC cancer tissues and cell lines. Clinicopathological analyses revealed that increased expression of galectin-3 in tumors was closely associated with a poor prognosis. Galectin-3 knockdown by siRNA significantly inhibited cell growth, migration, and invasion, and induced apoptosis in HCC cells in vitro, whereas galectin-3 overexpression promoted cell growth, migration, and invasion. Correlation analysis of galectin-3 expression and micro-vessel density (MVD) showed that galectin-3 expression in tumor cells stimulates angiogenesis. The observed regulation of cell apoptosis was accompanied by the galectin-3-mediated modulation of caspase3 signaling pathways in HCC cells. CONCLUSIONS These data suggest that galectin-3 plays an important part in HCC progression and may serve as a prognostic factor for HCC.
Collapse
|
27
|
Galectin-3 in cancer. Clin Chim Acta 2014; 431:185-91. [PMID: 24530298 DOI: 10.1016/j.cca.2014.01.019] [Citation(s) in RCA: 126] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2013] [Revised: 01/04/2014] [Accepted: 01/09/2014] [Indexed: 11/21/2022]
Abstract
Galectin-3 (Gal-3) plays important roles in cell proliferation, adhesion, differentiation, angiogenesis and apoptosis in normal and pathologic tissues. Accumulated evidences indicate that Gal-3 is closely involved in tumor cell transformation, migration, invasion and metastasis. In this review, the associations of the expression and localization of Gal-3 as well as its potential action mechanism in tumorigenesis in a variety of cancers were summarized and concluded. Gal-3 is gaining its attraction as a potential new biomarker for the diagnosis, treatment and prognosis of certain tumors.
Collapse
|
28
|
Expression of galectin-7 is induced in breast cancer cells by mutant p53. PLoS One 2013; 8:e72468. [PMID: 23967302 PMCID: PMC3743813 DOI: 10.1371/journal.pone.0072468] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2013] [Accepted: 07/17/2013] [Indexed: 12/16/2022] Open
Abstract
Galectin-7 was initially described as a marker of epithelial differentiation expressed in the stratified epithelium of various tissues. Like other members of the galectin family, its expression level is often significantly altered in cancer cells. In breast cancer, its expression is significantly augmented in aggressive molecular subtypes, most notably in estrogen receptor-negative tumors and in cell lines with a basal-like phenotype. Studies using experimental mouse models have further shown high expression of galectin-7 was sufficient to increase the metastatic behavior of poorly metastatic breast cancer cells, rendering them more resistant to apoptosis. This expression pattern in breast cancer cells is unexpected because galectin-7 was originally identified as a p53-induced gene. To address this paradox, we have examined the molecular mechanisms regulating galectin-7 in breast cancer cells. Our results showed that transfection of breast cancer cells with expression vectors encoding mutant p53 was sufficient to induce galectin-7 at both mRNA and protein levels. Doxorubicin treatment of breast cancer cells harboring a mutant p53 also induced galectin-7. This induction was specific since knockdown of endogenous mutant p53 inhibited doxorubicin-induced galectin-7 expression. The p53-induced galectin-7 expression in breast cancer cells correlated with increased NF-κB activity and was inhibited by NF-κB inhibitors, indicating that the ability of mutant p53 to induce galectin-7 was dependent on NF-κB activity. The implication of NF-κB was further supported by data showing that NF-κB bound to the endogenous galectin-7 promoter and that TNFα-induced galectin-7 expression was abolished by NF-κB inhibitors. Taken together, our data provide an explanation to the observed high galectin-7 expression levels in cancer cells and suggest that galectin-7 could be part of a common pathway used by mutant p53 to promote cancer progression.
Collapse
|
29
|
Galectin-3 gene silencing inhibits migration and invasion of human tongue cancer cells in vitro via downregulating β-catenin. Acta Pharmacol Sin 2013; 34:176-84. [PMID: 23103626 DOI: 10.1038/aps.2012.150] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
AIM Galectin-3 (Gal-3) is a member of the carbohydrate-binding protein family that contributes to neoplastic transformation, tumor survival, angiogenesis, and metastasis. The aim of this study is to investigate the role of Gal-3 in human tongue cancer progression. METHODS Human tongue cancer cell lines (SCC-4 and CAL27) were transfected with a small-interfering RNA against Gal-3 (Gal-3-siRNA). The migration and invasion of the cells were examined using a scratch assay and BD BioCoat Matrigel Invasion Chamber, respectively. The mRNA and protein levels of β-catenin, Akt/pAkt, GSK-3β/pGSK-3β, MMP-9 in the cells were measured using RT-PCR and Western blotting, respectively. RESULTS Transient silencing of Gal-3 gene for 48 h significantly suppressed the migration and invasion of both SCC-4 and CAL27 cells. Silencing of Gal-3 gene significantly decreased the protein level of β-catenin, leaving the mRNA level of β-catenin unaffected. Furthermore, silencing Gal-3 gene significantly decreased the levels of phosphorylated Akt and GSK-3β, and suppressed the mRNA and protein levels of MMP-9 in the cells. CONCLUSION Our data suggest that Gal-3 mediates the migration and invasion of tongue cancer cells in vitro via regulating the Wnt/β-catenin signaling pathway and Akt phosphorylation.
Collapse
|
30
|
Taniuchi K, Cerny RL, Tanouchi A, Kohno K, Kotani N, Honke K, Saibara T, Hollingsworth MA. Overexpression of GalNAc-transferase GalNAc-T3 promotes pancreatic cancer cell growth. Oncogene 2011; 30:4843-54. [PMID: 21625220 PMCID: PMC3373266 DOI: 10.1038/onc.2011.194] [Citation(s) in RCA: 63] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2010] [Revised: 04/15/2011] [Accepted: 04/19/2011] [Indexed: 01/08/2023]
Abstract
O-linked glycans of secreted and membrane-bound proteins have an important role in the pathogenesis of pancreatic cancer by modulating immune responses, inflammation and tumorigenesis. A critical aspect of O-glycosylation, the position at which proteins are glycosylated with N-acetyl-galactosamine on serine and threonine residues, is regulated by the substrate specificity of UDP-GalNAc:polypeptide N-acetylgalactosaminyl-transferases (GalNAc-Ts). Thus, GalNAc-Ts regulate the first committed step in O-glycosylated protein biosynthesis, determine sites of O-glycosylation on proteins and are important for understanding normal and carcinoma-associated O-glycosylation. We have found that one of these enzymes, GalNAc-T3, is overexpressed in human pancreatic cancer tissues and suppression of GalNAc-T3 significantly attenuates the growth of pancreatic cancer cells in vitro and in vivo. In addition, suppression of GalNAc-T3 induces apoptosis of pancreatic cancer cells. Our results indicate that GalNAc-T3 is likely involved in pancreatic carcinogenesis. Modification of cellular glycosylation occurs in nearly all types of cancer as a result of alterations in the expression levels of glycosyltransferases. We report guanine the nucleotide-binding protein, α-transducing activity polypeptide-1 (GNAT1) as a possible substrate protein of GalNAc-T3. GalNAc-T3 is associated with O-glycosylation of GNAT1 and affects the subcellular distribution of GNAT1. Knocking down endogenous GNAT1 significantly suppresses the growth/survival of PDAC cells. Our results imply that GalNAc-T3 contributes to the function of O-glycosylated proteins and thereby affects the growth and survival of pancreatic cancer cells. Thus, substrate proteins of GalNAc-T3 should serve as important therapeutic targets for pancreatic cancers.
Collapse
Affiliation(s)
- K Taniuchi
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE, USA.
| | | | | | | | | | | | | | | |
Collapse
|
31
|
Li Y, Tao SC, Zhu H, Schneck JP. High-throughput lectin microarray-based analysis of live cell surface glycosylation. CURRENT PROTOCOLS IN PROTEIN SCIENCE 2011; Chapter 12:12.9.1-12.9.7. [PMID: 21400689 PMCID: PMC3090205 DOI: 10.1002/0471140864.ps1209s63] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Lectins, plant-derived glycan-binding proteins, have long been used to detect glycans on cell surfaces. However, the techniques used to characterize serum or cells have largely been limited to mass spectrometry, blots, flow cytometry, and immunohistochemistry. While these lectin-based approaches are well established and they can discriminate a limited number of sugar isomers by concurrently using a limited number of lectins, they are not amenable for adaptation to a high-throughput platform. Fortunately, given the commercial availability of lectins with a variety of glycan specificities, lectins can be printed on a glass substrate in a microarray format to profile accessible cell-surface glycans. This method is an inviting alternative for analysis of a broad range of glycans in a high-throughput fashion and has been demonstrated to be a feasible method of identifying binding-accessible cell surface glycosylation on living cells. The current unit presents a lectin-based microarray approach for analyzing cell surface glycosylation in a high-throughput fashion.
Collapse
Affiliation(s)
- Yu Li
- Department of Pathology, Oncology & Medicine and Institute for Cell Engineering, Johns Hopkins University, Baltimore, Maryland
| | - Sheng-ce Tao
- Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore, Maryland
,The High-Throughput Biology Center, Johns Hopkins University School of Medicine, Baltimore, Maryland
,Shanghai Center for Systems Biomedicine, Shanghai Jiaotong University, Shanghai, China
| | - Heng Zhu
- Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore, Maryland
,The High-Throughput Biology Center, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Jonathan P. Schneck
- Department of Pathology, Oncology & Medicine and Institute for Cell Engineering, Johns Hopkins University, Baltimore, Maryland
| |
Collapse
|
32
|
Lopez G, Jmenez S, Martinez R, Pina MDS, Gallegos B, Pérez-Campos E, Zenteno E, Hernández P. Glycosylation pattern in the appendix testis in children with cryptorchidism. Prep Biochem Biotechnol 2011; 41:22-29. [PMID: 21229461 DOI: 10.1080/10826068.2010.525398] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
In humans, at about week 6, sex cords develop within the forming testes. Testes normally descend to the scrotum; cryptorchidism occurs when one or two testes do not descend to scrotum and in some case are accompanied by the appendix testis. The appendix testis is a small sessile or polypoid structure located at the antero superior pole of the testis, adjacent to the head of the epididymis. Glycans can be involved in development of the appendix testis and cryptorchidism. In this work, lectin histochemistry was used to evaluate glycans expression in appendix testis in children with cryptorchidism. Our results showed that lectin from Lens culinaris, Ulex europaeus I., Canavalia ensiformis, Artocarpus integrifolia, Glycine max, and Griffonia simplicifolia recognizes epithelial and estromal cells. Not interaction was observed with lectin from Amaranthus leucocarpus, while lectin from Dolichus biflorus lectin only recognizes epithelial cells. Our results suggest that O-glycans linked in some glycoproteins represent important elements in appendix testis development.
Collapse
Affiliation(s)
- Gerardo Lopez
- Centro de Investigaciones en Ciencias Medicas y Biológicas Facultad de Medicina, Universidad Autónoma Benito Juárez de Oaxaca, Oaxaca, México
| | | | | | | | | | | | | | | |
Collapse
|
33
|
Li M, Song L, Qin X. Glycan changes: cancer metastasis and anti-cancer vaccines. J Biosci 2010; 35:665-73. [PMID: 21289447 DOI: 10.1007/s12038-010-0073-8] [Citation(s) in RCA: 63] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2009] [Accepted: 10/11/2010] [Indexed: 01/01/2023]
Abstract
Complex carbohydrates, which are major components of the cell membrane, perform important functions in cell-cell and cell-extracellular matrix interactions, as well as in signal transduction. They comprise three kinds of biomolecules: glycoproteins, proteoglycans and glycosphingolipids. Recent studies have also shown that glycan changes in malignant cells take a variety of forms and mediate key pathophysiological events during the various stages of tumour progression. Glycosylation changes are universal hallmarks of malignant transformation and tumour progression in human cancer, which take place on the whole cells or some specific molecules. Accordingly, those changes make them prominent candidates for cancer biomarkers in the meantime. This review mainly focuses on the correlation between glycosylation and the metastasis potential of tumour cells from comprehensive aspects to further address the vital roles of glycans in oncogenesising. Moreover, utilizing these glycosylation changes to ward off tumour metastasis by means of anti-adhesion approach or devising anti-cancer vaccine is one of promising targets of future study.
Collapse
Affiliation(s)
- Min Li
- Department of General Surgery, Zhongshan Hospital, Shanghai Medical School, Fudan University, 180, Fenglin Road, Shanghai, 200032, China
| | | | | |
Collapse
|
34
|
Bogoeva VP, Varriale A, John CM, D'Auria S. Human galectin-3 interacts with two anticancer drugs. Proteomics 2010; 10:1946-53. [PMID: 20209510 DOI: 10.1002/pmic.200900581] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Human galectin-3 (hGal-3) is a mammalian lectin involved in regulation of RNA splicing, apoptosis, cell differentiation, and proliferation. Multimerized extracellular hGal-3 is thought to crosslink cells by binding to glycoproteins and glycosylated cancer antigens on the cell surface or extracellular matrix. Fluorescence spectroscopy and circular dichroism were used to study the interaction of hGal-3 with two anticancer agents: bohemine and Zn porphyrin (ZnTPPS(4)). The dissociation constant (k(D)) for binding of bohemine with hGal-3 was k(D) 0.23+/-0.05 microM. The hyperbolic titration curve indicated the presence of a single bohemine binding site. The binding of ZnTPPS(4) to hGal-3 (with and without lactose) is of high affinity having k(D)=0.18-0.20 microM and is not inhibited by lactose, indicating that ZnTPPS(4) and carbohydrate bind different sites. Circular dichroism spectra of the hGal-3 complexes suggested that the binding of the hydrophobic compounds changed the hGal-3 secondary structure. In summary, we show that two compounds with anticancer activity, bohemine and ZnTPPS(4), have high affinity for hGal-3 at a site that is distinct from its carbohydrate site. Since hGal-3 binds to several carbohydrate cancer antigens, the results suggest that it may have utility in the targeted delivery of drugs for cancer.
Collapse
Affiliation(s)
- Vanya P Bogoeva
- Institute of Molecular Biology, Bulgarian Academy of Sciences, Sofia, Bulgaria.
| | | | | | | |
Collapse
|
35
|
Gallegos B, Pérez-Campos E, Martinez R, Leyva P, Martinez M, Hernández R, Pina S, Hernández C, Zenteno E, Hernández P. O-glycosylation expression in fibroadenoma. Prep Biochem Biotechnol 2010; 40:1-12. [PMID: 20024790 DOI: 10.1080/10826060903386071] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
Fibroadenomas are human benign breast tumors characterized by proliferation of epithelial and stroma cells of the terminal ductal unit. Expression of O-glycans seems to contribute to the proliferation and transformation events. With this in mind, we evaluated the expression of glycans in fibroadenoma tissue through immunohistochemistry with antibodies against mucin epitopes (Anti CA15-3 and MUC1), as well as with lectins specific for glycans linked to proteins or lipids, and we compared findings with healthy breast specimens. Our results show positive expression of CA15-3 and MUC1 in fibroadenoma tissue, mainly in duct and stroma cells, whereas, in normal samples, staining was observed in duct cells. The lectin from Glycine max recognized equally well duct and stroma cells; this was the only lectin showing co-localization with anti-CA15-3 in healthy and tumor tissues. Dolichos biflorus, Artocarpus integrifolia, and Griffonia simplicifolia lectins recognized duct cells in control healthy tissues as well as in fibroadenoma tissue. The lectin from Amaranthus leucocarpus recognized only duct cells in control samples, whereas, in fibroadenoma tissue, it recognized duct and some stromal cells, suggesting that O-glycans-type mucin linked to proteins and mucin participate in the development of fibroadenomas.
Collapse
Affiliation(s)
- Belem Gallegos
- Centro de Investigaciones en Ciencias Medicas y Biológicas, Facultad de Medicina, Universidad Autónoma Benito Juárez de Oaxaca, Oaxaca, México
| | | | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Abstract
The O-glycosylation of Ser and Thr by N-acetylgalactosamine-linked (mucin-type) oligosaccharides is often overlooked in protein analysis. Three characteristics make O-linked glycosylation more difficult to analyse than N-linked glycosylation, namely: (a) no amino acid consensus sequence is known; (b) there is no universal enzyme for the release of O-glycans from the protein backbone; and (c) the density and number of occupied sites may be very high. For significant biological conclusions to be drawn, the complete picture of O-linked glycosylation on a protein needs to be determined. This review specifically addresses the analytical approaches that have been used, and the challenges remaining, in the characterization of both the composition and structure of mucin-type O-glycans, and the determination of the occupancy and heterogeneity at each amino acid attachment site.
Collapse
Affiliation(s)
- Pia H Jensen
- Department of Chemistry and Biomolecular Sciences, Faculty of Science, Biomolecular Frontiers Research Centre, Macquarie University, Sydney, Australia
| | | | | |
Collapse
|
37
|
Kovacheva VP, Davison JM, Mellott TJ, Rogers AE, Yang S, O'Brien MJ, Blusztajn JK. Raising gestational choline intake alters gene expression in DMBA-evoked mammary tumors and prolongs survival. FASEB J 2008; 23:1054-63. [PMID: 19047067 DOI: 10.1096/fj.08-122168] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
Choline is an essential nutrient that serves as a donor of metabolic methyl groups used during gestation to establish the epigenetic DNA methylation patterns that modulate tissue-specific gene expression. Because the mammary gland begins its development prenatally, we hypothesized that choline availability in utero may affect the gland's susceptibility to cancer. During gestational days 11-17, pregnant rats were fed a control, choline-supplemented, or choline-deficient diet (8, 36, and 0 mmol/kg of choline, respectively). On postnatal day 65, the female offspring received 25 mg/kg of a carcinogen 7,12-dimethylbenz[alpha]anthracene. Approximately 70% of the rats developed mammary adenocarcinomas; prenatal diet did not affect tumor latency, incidence, size, and multiplicity. Tumor growth rate was inversely related to choline content in the prenatal diet, resulting in 50% longer survival until euthanasia, determined by tumor size, of the prenatally choline-supplemented rats compared with the prenatally choline-deficient rats. This was accompanied by distinct expression patterns of approximately 70 genes in tumors derived from the three dietary groups. Tumors from the prenatally choline-supplemented rats overexpressed genes that confer favorable prognosis in human cancers (Klf6, Klf9, Nid2, Ntn4, Per1, and Txnip) and underexpressed those associated with aggressive disease (Bcar3, Cldn12, Csf1, Jag1, Lgals3, Lypd3, Nme1, Ptges2, Ptgs1, and Smarcb1). DNA methylation within the tumor suppressor gene, stratifin (Sfn, 14-3-3sigma), was proportional to the prenatal choline supply and correlated inversely with the expression of its mRNA and protein in tumors, suggesting that an epigenetic mechanism may underlie the altered molecular phenotype and tumor growth. Our results suggest a role for adequate maternal choline nutrition during pregnancy in prevention/alleviation of breast cancer in daughters.
Collapse
Affiliation(s)
- Vesela P Kovacheva
- Department of Pathology and Laboratory Medicine, Boston University School of Medicine, Boston, MA 02118, USA
| | | | | | | | | | | | | |
Collapse
|