1
|
Zhang C, Wang Y, Yu Y, Pang Y, Xiao X, Hao L. Overexpression of ST8Sia1 inhibits tumor progression by TGF-β1 signaling in rectal adenocarcinoma and promotes the tumoricidal effects of CD8 + T cells by granzyme B and perforin. Ann Med 2025; 57:2439539. [PMID: 39656552 PMCID: PMC11633436 DOI: 10.1080/07853890.2024.2439539] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/09/2024] [Revised: 05/23/2024] [Accepted: 10/29/2024] [Indexed: 12/12/2024] Open
Abstract
BACKGROUND Rectal adenocarcinoma (READ) involves the dysregulated expression of alpha 2,8-Sialyltransferase1 (ST8Sia1) although its role during READ's progression is unclear. METHODS The mRNA level of ST8Sia1 was analyzed based on The Cancer Genome Atlas (TCGA), Gene Expression Omnibus (GEO), and Tumor Immune Estimation Resource (TIMER) 2.0. Furthermore, the prognostic and significance of ST8Sia1 in READ was assessed through Kaplan-Meier curve, univariate, multivariate Cox regression, and receiver operating characteristic (ROC) methods. The role of ST8Sia1 in the READ immune microenvironment was explored using ESTIMATE analysis and TIMER databases. Furthermore, the expression of ST8Sia1 in tissues was analyzed using real-time quantitative polymerase chain reaction (RT-qPCR), western blotting (WB), and immunohistochemistry (IHC). Perforin and Granzyme B secretion by CD8+ T cells, as well as tumor cell apoptosis, were detected after co-culturing CD8+ T cells with READ tumor cells and ST8Sia1-overexpression (ST8Sia1-OE) tumor cells. Furthermore, we examined the interaction between ST8Sia1 and TGF-β1 in READ cells. RESULTS ST8Sia1 exhibited excellent diagnostic capability for READ, with positive correlations to immune response and negative correlations to tumor purity. Increased levels of perforin and Granzyme B from CD8+ T cells were observed in vitro, enhancing tumor cell apoptosis. ST8Sia1 interacts with TGF-β1, mediating its inhibitory effects on READ development. CONCLUSIONS ST8Sia1 is a potential diagnostic biomarker and therapeutic target for READ, enhancing CD8+ T cell function and possibly improving patient outcomes through cellular immunotherapy.
Collapse
Affiliation(s)
- Chang Zhang
- Department of Anorectal, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai City, Shandong Province, China
| | - Yeli Wang
- Department of Anorectal, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai City, Shandong Province, China
| | - Yao Yu
- Department of General Pediatric Surgery, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai City, Shandong Province, China
| | - Yanchao Pang
- Department of Anorectal, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai City, Shandong Province, China
| | - Xiao Xiao
- Department of Neurology, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai City, Shandong Province, China
| | - Leilei Hao
- Department of Anorectal, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai City, Shandong Province, China
| |
Collapse
|
2
|
Habeeb IF, Alao TE, Delgado D, Buffone A. When a negative (charge) is not a positive: sialylation and its role in cancer mechanics and progression. Front Oncol 2024; 14:1487306. [PMID: 39628991 PMCID: PMC11611868 DOI: 10.3389/fonc.2024.1487306] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2024] [Accepted: 10/10/2024] [Indexed: 12/06/2024] Open
Abstract
Sialic acids and sialoglycans are critical actors in cancer progression and metastasis. These terminal sugar residues on glycoproteins and glycolipids modulate key cellular processes such as immune evasion, cell adhesion, and migration. Aberrant sialylation is driven by overexpression of sialyltransferases, resulting in hypersialylation on cancer cell surfaces as well as enhancing tumor aggressiveness. Sialylated glycans alter the structure of the glycocalyx, a protective barrier that fosters cancer cell detachment, migration, and invasion. This bulky glycocalyx also increases membrane tension, promoting integrin clustering and downstream signaling pathways that drive cell proliferation and metastasis. They play a critical role in immune evasion by binding to Siglecs, inhibitory receptors on immune cells, which transmit signals that protect cancer cells from immune-mediated destruction. Targeting sialylation pathways presents a promising therapeutic opportunity to understand the complex roles of sialic acids and sialoglycans in cancer mechanics and progression, which is crucial for developing novel diagnostic and therapeutic strategies that can disrupt these processes and improve cancer treatment outcomes.
Collapse
Affiliation(s)
- Issa Funsho Habeeb
- Department of Biomedical Engineering, New Jersey Institute of Technlogy, Newark, NJ, United States
| | - Toheeb Eniola Alao
- Department of Biomedical Engineering, New Jersey Institute of Technlogy, Newark, NJ, United States
| | - Daniella Delgado
- Department of Biomedical Engineering, New Jersey Institute of Technlogy, Newark, NJ, United States
| | - Alexander Buffone
- Department of Biomedical Engineering, New Jersey Institute of Technlogy, Newark, NJ, United States
- Chemical and Materials Engineering, New Jersey Institute of Technlogy, Newark, NJ, United States
| |
Collapse
|
3
|
Cumin C, Gee L, Litfin T, Muchabaiwa R, Martin G, Cooper O, Heinzelmann-Schwarz V, Lange T, von Itzstein M, Jacob F, Everest-Dass A. Highly Sensitive Spatial Glycomics at Near-Cellular Resolution by On-Slide Derivatization and Mass Spectrometry Imaging. Anal Chem 2024; 96:11163-11171. [PMID: 38953530 PMCID: PMC11256013 DOI: 10.1021/acs.analchem.3c05984] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2023] [Revised: 06/17/2024] [Accepted: 06/18/2024] [Indexed: 07/04/2024]
Abstract
Glycans on proteins and lipids play important roles in maturation and cellular interactions, contributing to a variety of biological processes. Aberrant glycosylation has been associated with various human diseases including cancer; however, elucidating the distribution and heterogeneity of glycans in complex tissue samples remains a major challenge. Matrix-assisted laser desorption/ionization (MALDI) mass spectrometry imaging (MSI) is routinely used to analyze the spatial distribution of a variety of molecules including N-glycans directly from tissue surfaces. Sialic acids are nine carbon acidic sugars that often exist as the terminal sugars of glycans and are inherently difficult to analyze using MALDI-MSI due to their instability prone to in- and postsource decay. Here, we report on a rapid and robust method for stabilizing sialic acid on N-glycans in FFPE tissue sections. The established method derivatizes and identifies the spatial distribution of α2,3- and α2,6-linked sialic acids through complete methylamidation using methylamine and PyAOP ((7-azabenzotriazol-1-yloxy)tripyrrolidinophosphonium hexafluorophosphate). Our in situ approach increases the glycans detected and enhances the coverage of sialylated species. Using this streamlined, sensitive, and robust workflow, we rapidly characterize and spatially localize N-glycans in human tumor tissue sections. Additionally, we demonstrate this method's applicability in imaging mammalian cell suspensions directly on slides, achieving cellular resolution with minimal sample processing and cell numbers. This workflow reveals the cellular locations of distinct N-glycan species, shedding light on the biological and clinical significance of these biomolecules in human diseases.
Collapse
Affiliation(s)
- Cécile Cumin
- Institute
for Glycomics, Griffith University, Gold Coast, Queensland 4222, Australia
- Ovarian
Cancer Research, University Hospital Basel,
University of Basel, Basel 4001, Switzerland
| | - Lindsay Gee
- Institute
for Glycomics, Griffith University, Gold Coast, Queensland 4222, Australia
| | - Thomas Litfin
- Institute
for Glycomics, Griffith University, Gold Coast, Queensland 4222, Australia
| | - Ropafadzo Muchabaiwa
- Institute
for Glycomics, Griffith University, Gold Coast, Queensland 4222, Australia
| | - Gael Martin
- Institute
for Glycomics, Griffith University, Gold Coast, Queensland 4222, Australia
| | - Oren Cooper
- Institute
for Glycomics, Griffith University, Gold Coast, Queensland 4222, Australia
| | - Viola Heinzelmann-Schwarz
- Ovarian
Cancer Research, University Hospital Basel,
University of Basel, Basel 4001, Switzerland
- Hospital
for Women, Department of Gynaecology and Gynaecological Oncology, University Hospital Basel and University of Basel, Basel 4001, Switzerland
| | - Tobias Lange
- Institute
of Anatomy and Experimental Morphology, University Cancer Center Hamburg
(UCCH), University Medical Center Hamburg-Eppendorf, Hamburg 20251, Germany
- Institute
of Anatomy I, Comprehensive Cancer Center Central Germany (CCCG), Jena University Hospital, Jena 07740, Germany
| | - Mark von Itzstein
- Institute
for Glycomics, Griffith University, Gold Coast, Queensland 4222, Australia
| | - Francis Jacob
- Ovarian
Cancer Research, University Hospital Basel,
University of Basel, Basel 4001, Switzerland
| | - Arun Everest-Dass
- Institute
for Glycomics, Griffith University, Gold Coast, Queensland 4222, Australia
| |
Collapse
|
4
|
Ma Y, Guo W, Mou Q, Shao X, Lyu M, Garcia V, Kong L, Lewis W, Ward C, Yang Z, Pan X, Yi SS, Lu Y. Spatial imaging of glycoRNA in single cells with ARPLA. Nat Biotechnol 2024; 42:608-616. [PMID: 37217750 PMCID: PMC10663388 DOI: 10.1038/s41587-023-01801-z] [Citation(s) in RCA: 29] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Accepted: 04/24/2023] [Indexed: 05/24/2023]
Abstract
Little is known about the biological roles of glycosylated RNAs (glycoRNAs), a recently discovered class of glycosylated molecules, because of a lack of visualization methods. We report sialic acid aptamer and RNA in situ hybridization-mediated proximity ligation assay (ARPLA) to visualize glycoRNAs in single cells with high sensitivity and selectivity. The signal output of ARPLA occurs only when dual recognition of a glycan and an RNA triggers in situ ligation, followed by rolling circle amplification of a complementary DNA, which generates a fluorescent signal by binding fluorophore-labeled oligonucleotides. Using ARPLA, we detect spatial distributions of glycoRNAs on the cell surface and their colocalization with lipid rafts as well as the intracellular trafficking of glycoRNAs through SNARE protein-mediated secretory exocytosis. Studies in breast cell lines suggest that surface glycoRNA is inversely associated with tumor malignancy and metastasis. Investigation of the relationship between glycoRNAs and monocyte-endothelial cell interactions suggests that glycoRNAs may mediate cell-cell interactions during the immune response.
Collapse
Affiliation(s)
- Yuan Ma
- Department of Chemistry, The University of Texas at Austin, Austin, TX, USA
| | - Weijie Guo
- Department of Molecular Biosciences, The University of Texas at Austin, Austin, TX, USA
- Interdisciplinary Life Sciences Graduate Programs, The University of Texas at Austin, Austin, TX, USA
| | - Quanbing Mou
- Department of Chemistry, The University of Texas at Austin, Austin, TX, USA
| | - Xiangli Shao
- Department of Chemistry, The University of Texas at Austin, Austin, TX, USA
| | - Mingkuan Lyu
- Department of Chemistry, The University of Texas at Austin, Austin, TX, USA
| | - Valeria Garcia
- Department of Molecular Biosciences, The University of Texas at Austin, Austin, TX, USA
- Interdisciplinary Life Sciences Graduate Programs, The University of Texas at Austin, Austin, TX, USA
| | - Linggen Kong
- Department of Molecular Biosciences, The University of Texas at Austin, Austin, TX, USA
- Interdisciplinary Life Sciences Graduate Programs, The University of Texas at Austin, Austin, TX, USA
| | - Whitney Lewis
- Department of Chemistry, The University of Texas at Austin, Austin, TX, USA
- Interdisciplinary Life Sciences Graduate Programs, The University of Texas at Austin, Austin, TX, USA
| | - Carson Ward
- Department of Molecular Biosciences, The University of Texas at Austin, Austin, TX, USA
| | - Zhenglin Yang
- Department of Chemistry, The University of Texas at Austin, Austin, TX, USA
| | - Xingxin Pan
- Department of Oncology, Livestrong Cancer Institutes, Dell Medical School, The University of Texas at Austin, Austin, TX, USA
| | - S Stephen Yi
- Interdisciplinary Life Sciences Graduate Programs, The University of Texas at Austin, Austin, TX, USA
- Department of Oncology, Livestrong Cancer Institutes, Dell Medical School, The University of Texas at Austin, Austin, TX, USA
- Oden Institute for Computational Engineering and Sciences (ICES), The University of Texas at Austin, Austin, TX, USA
| | - Yi Lu
- Department of Chemistry, The University of Texas at Austin, Austin, TX, USA.
- Interdisciplinary Life Sciences Graduate Programs, The University of Texas at Austin, Austin, TX, USA.
| |
Collapse
|
5
|
Jangid AK, Kim S, Park HW, Kim HJ, Kim K. Ex Vivo Surface Decoration of Phenylboronic Acid onto Natural Killer Cells for Sialic Acid-Mediated Versatile Cancer Cell Targeting. Biomacromolecules 2024; 25:222-237. [PMID: 38130077 DOI: 10.1021/acs.biomac.3c00916] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2023]
Abstract
Phenylboronic acid (PBA) has been highly acknowledged as a significant cancer recognition moiety in sialic acid-overexpressing cancer cells. In this investigation, lipid-mediated biomaterial integrated PBA molecules onto the surface of natural killer (NK) cells to make a receptor-mediated immune cell therapeutic module. Therefore, a 1,2-distearoyl-sn-glycero-3-phosphorylethanolamine (DSPE) lipid-conjugated di-PEG-PBA (DSPEPEG-di(PEG-PBA) biomaterial was synthesized. The DSPEPEG-di(PEG-PBA) biomaterial exhibited a high affinity for sialic acid (SA), confirmed by fluorescence spectroscopy at pH 6.5 and 7.4. DSPEPEG-di(PEG-PBA) was successfully anchored onto NK cell surfaces (PBA-NK), and this biomaterial maintains intrinsic properties such as viability, ligand availability (FasL & TRAIL), and cytokine secretion response to LPS. The anticancer efficacy of PBA-NK cells was evaluated against 2D cancer cells (MDA-MB-231, HepG2, and HCT-116) and 3D tumor spheroids of MDA-MB-231 cells. PBA-NK cells exhibited greatly enhanced anticancer effects against SA-overexpressing cancer cells. Thus, PBA-NK cells represent a new anticancer strategy for cancer immunotherapy.
Collapse
Affiliation(s)
- Ashok Kumar Jangid
- Department of Chemical & Biochemical Engineering, College of Engineering, Dongguk University, Seoul 04620, South Korea
| | - Sungjun Kim
- Department of Chemical & Biochemical Engineering, College of Engineering, Dongguk University, Seoul 04620, South Korea
| | - Hee Won Park
- Department of Chemical & Biochemical Engineering, College of Engineering, Dongguk University, Seoul 04620, South Korea
| | - Hyun Jin Kim
- Department of Biological Engineering, College of Engineering, Inha University, Incheon 22212, South Korea
| | - Kyobum Kim
- Department of Chemical & Biochemical Engineering, College of Engineering, Dongguk University, Seoul 04620, South Korea
| |
Collapse
|
6
|
Hussen BM, Abdullah KH, Abdullah SR, Majeed NM, Mohamadtahr S, Rasul MF, Dong P, Taheri M, Samsami M. New insights of miRNA molecular mechanisms in breast cancer brain metastasis and therapeutic targets. Noncoding RNA Res 2023; 8:645-660. [PMID: 37818447 PMCID: PMC10560790 DOI: 10.1016/j.ncrna.2023.09.003] [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: 06/15/2023] [Revised: 09/17/2023] [Accepted: 09/17/2023] [Indexed: 10/12/2023] Open
Abstract
Brain metastases in breast cancer (BC) patients are often associated with a poor prognosis. Recent studies have uncovered the critical roles of miRNAs in the initiation and progression of BC brain metastasis, highlighting the disease's underlying molecular pathways. miRNA-181c, miRNA-10b, and miRNA-21, for example, are all overexpressed in BC patients. It has been shown that these three miRNAs help tumors grow and metastasize by targeting genes that control how cells work. On the other hand, miRNA-26b5p, miRNA-7, and miRNA-1013p are all downregulated in BC brain metastasis patients. They act as tumor suppressors by controlling the expression of genes related to cell adhesion, angiogenesis, and invasion. Therapeutic miRNA targeting has considerable promise in treating BC brain metastases. Several strategies have been proposed to modulate miRNA expression, including miRNA-Mimics, antagomirs, and small molecule inhibitors of miRNA biogenesis. This review discusses the aberrant expression of miRNAs and metastatic pathways that lead to the spread of BC cells to the brain. It also explores miRNA therapeutic target molecular mechanisms and BC brain metastasis challenges with advanced strategies. The targeting of certain miRNAs opens a new door for the development of novel therapeutic approaches for this devastating disease.
Collapse
Affiliation(s)
- Bashdar Mahmud Hussen
- Department of Biomedical Sciences, College of Science, Cihan University-Erbil, Kurdistan Region, 44001, Iraq
- Department of Clinical Analysis, College of Pharmacy, Hawler Medical University, Kurdistan Region, Erbil, Iraq
| | - Khozga Hazhar Abdullah
- Medical Laboratory Science, College of Health Sciences, Lebanese French University, Kurdistan Region, Erbil, Iraq
| | - Snur Rasool Abdullah
- Medical Laboratory Science, College of Health Sciences, Lebanese French University, Kurdistan Region, Erbil, Iraq
| | | | - Sayran Mohamadtahr
- Department of Pharmacology and Toxicology, College of Pharmacy, Hawler Medical University, Erbil, Iraq
| | - Mohammed Fatih Rasul
- Department of Pharmaceutical Basic Science, Tishk International University, Erbil, Kurdistan Region, Iraq
| | - Peixin Dong
- Department of Obstetrics and Gynecology, Hokkaido University School of Medicine, Hokkaido University, Sapporo, Japan
| | - Mohammad Taheri
- Institute of Human Genetics, Jena University Hospital, Jena, Germany
- Urology and Nephrology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Majid Samsami
- Cancer Research Center, Loghman Hakim Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
7
|
Alsharabasy AM, Aljaabary A, Bohara R, Farràs P, Glynn SA, Pandit A. Nitric Oxide-Scavenging, Anti-Migration Effects, and Glycosylation Changes after Hemin Treatment of Human Triple-Negative Breast Cancer Cells: A Mechanistic Study. ACS Pharmacol Transl Sci 2023; 6:1416-1432. [PMID: 37854626 PMCID: PMC10580390 DOI: 10.1021/acsptsci.3c00115] [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: 06/09/2023] [Indexed: 10/20/2023]
Abstract
The enhanced expression of nitric oxide (•NO) synthase predicts triple-negative breast cancer outcome and its resistance to different therapeutics. Our earlier work demonstrated the efficiency of hemin to scavenge the intra- and extracellular •NO, proposing its potency as a therapeutic agent for inhibiting cancer cell migration. In continuation, the present work evaluates the effects of •NO on the migration of MDA-MB-231 cells and how hemin modulates the accompanied cellular behavior, focusing on the corresponding expression of cellular glycoproteins, migration-associated markers, and mitochondrial functions. We demonstrated for the first time that while •NO induced cell migration, hemin contradicted that by •NO-scavenging. This was in combination with modulation of the •NO-enhanced glycosylation patterns of cellular proteins with inhibition of the expression of specific proteins involved in the epithelial-mesenchymal transition. These effects were in conjunction with changes in the mitochondrial functions related to both •NO, hemin, and its nitrosylated product. Together, these results suggest that hemin can be employed as a potential anti-migrating agent targeting •NO-scavenging and regulating the expression of migration-associated proteins.
Collapse
Affiliation(s)
- Amir M. Alsharabasy
- CÚRAM,
SFI Research Centre for Medical Devices, University of Galway, Galway H91 W2TY, Ireland
| | - Amal Aljaabary
- CÚRAM,
SFI Research Centre for Medical Devices, University of Galway, Galway H91 W2TY, Ireland
| | - Raghvendra Bohara
- CÚRAM,
SFI Research Centre for Medical Devices, University of Galway, Galway H91 W2TY, Ireland
| | - Pau Farràs
- CÚRAM,
SFI Research Centre for Medical Devices, University of Galway, Galway H91 W2TY, Ireland
- School
of Biological and Chemical Sciences, Ryan Institute, University of Galway, Galway H91 TK33, Ireland
| | - Sharon A. Glynn
- CÚRAM,
SFI Research Centre for Medical Devices, University of Galway, Galway H91 W2TY, Ireland
- Discipline
of Pathology, Lambe Institute for Translational Research, School of
Medicine, University of Galway, Galway H91 YR71, Ireland
| | - Abhay Pandit
- CÚRAM,
SFI Research Centre for Medical Devices, University of Galway, Galway H91 W2TY, Ireland
| |
Collapse
|
8
|
Al Saoud R, Hamrouni A, Idris A, Mousa WK, Abu Izneid T. Recent advances in the development of sialyltransferase inhibitors to control cancer metastasis: A comprehensive review. Biomed Pharmacother 2023; 165:115091. [PMID: 37421784 DOI: 10.1016/j.biopha.2023.115091] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Revised: 06/23/2023] [Accepted: 06/26/2023] [Indexed: 07/10/2023] Open
Abstract
Metastasis accounts for the majority of cancer-associated mortalities, representing a huge health and economic burden. One of the mechanisms that enables metastasis is hypersialylation, characterized by an overabundance of sialylated glycans on the tumor surface, which leads to repulsion and detachment of cells from the original tumor. Once the tumor cells are mobilized, sialylated glycans hijack the natural killer T-cells through self-molecular mimicry and activatea downstream cascade of molecular events that result in inhibition of cytotoxicity and inflammatory responses against cancer cells, ultimately leading to immune evasion. Sialylation is mediated by a family of enzymes known as sialyltransferases (STs), which catalyse the transfer of sialic acid residue from the donor, CMP-sialic acid, onto the terminal end of an acceptor such as N-acetylgalactosamine on the cell-surface. Upregulation of STs increases tumor hypersialylation by up to 60% which is considered a distinctive hallmark of several types of cancers such as pancreatic, breast, and ovarian cancer. Therefore, inhibiting STs has emerged as a potential strategy to prevent metastasis. In this comprehensive review, we discuss the recent advances in designing novel sialyltransferase inhibitors using ligand-based drug design and high-throughput screening of natural and synthetic entities, emphasizing the most successful approaches. We analyse the limitations and challenges of designing selective, potent, and cell-permeable ST inhibitors that hindered further development of ST inhibitors into clinical trials. We conclude by analysing emerging opportunities, including advanced delivery methods which further increase the potential of these inhibitors to enrich the clinics with novel therapeutics to combat metastasis.
Collapse
Affiliation(s)
- Ranim Al Saoud
- Pharmaceutical Sciences Program, College of Pharmacy, Al Ain University, P.O. Box 112612, Al Ain, Abu Dhabi, United Arab Emirates; AAU Health and Biomedical Research Center, Al Ain University, P.O. Box 112612, Abu Dhabi, United Arab Emirates
| | - Amar Hamrouni
- Pharmaceutical Sciences Program, College of Pharmacy, Al Ain University, P.O. Box 112612, Al Ain, Abu Dhabi, United Arab Emirates; AAU Health and Biomedical Research Center, Al Ain University, P.O. Box 112612, Abu Dhabi, United Arab Emirates
| | - Adi Idris
- School of Biomedical Sciences, Queensland University of Technology, Gardens Point, QLD, Australia; School of Pharmacy and Medical Science, Menzies Health Institute Queensland, Griffith University, Gold Coast, QLD, Australia
| | - Walaa K Mousa
- Pharmaceutical Sciences Program, College of Pharmacy, Al Ain University, P.O. Box 112612, Al Ain, Abu Dhabi, United Arab Emirates; AAU Health and Biomedical Research Center, Al Ain University, P.O. Box 112612, Abu Dhabi, United Arab Emirates
| | - Tareq Abu Izneid
- Pharmaceutical Sciences Program, College of Pharmacy, Al Ain University, P.O. Box 112612, Al Ain, Abu Dhabi, United Arab Emirates; AAU Health and Biomedical Research Center, Al Ain University, P.O. Box 112612, Abu Dhabi, United Arab Emirates.
| |
Collapse
|
9
|
Tan Z, Chen X, Zuo J, Fu S, Wang J, Wang H. Integrating Bulk and Single-Cell RNA Sequencing Reveals Heterogeneity, Tumor Microenvironment, and Immunotherapeutic Efficacy Based on Sialylation-Related Genes in Bladder Cancer. J Inflamm Res 2023; 16:3399-3417. [PMID: 37600224 PMCID: PMC10438438 DOI: 10.2147/jir.s418433] [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: 05/11/2023] [Accepted: 07/24/2023] [Indexed: 08/22/2023] Open
Abstract
Background As known abnormal sialylation exerts crucial roles in the growth, metastasis, and immune evasion of cancers, but the molecular characteristics and roles in bladder cancer (BLCA) remain unclear. This study intends to establish BLCA risk stratification based on sialylation-related genes and elucidate its role in prognosis, tumor microenvironment, and immunotherapy of BLCA. Methods Bulk RNA-seq and scRNA-seq data were downloaded from open-access databases. The scRNA-seq data were processed using the R package "Seurat" to identify the core cell types. The tumor sub-typing of BLCA samples was performed by the R package "ConsensusClusterPlus" in the bulk RNA-seq data. Signature genes were identified by the R package "limma" and univariate regression analysis to calculate risk scores using the R package "GSVA" and establish risk stratification of BLCA patients. Finally, the differences in clinicopathological characteristics, tumor microenvironment, and immunotherapy efficacy between the different groups were investigated. Results 5 core cell types were identified in the scRNA-seq dataset, with monocytes and macrophages presenting the greatest percentage, sialylation-related gene expression, and sialylation scores. The bulk RNA-seq samples were classified into 3 tumor subtypes based on 19 prognosis-related sialylation genes. The 10 differential expressed genes (DEGs) with the smallest p-values were collected as signature genes, and the risk score was calculated, with the samples divided into high and low-risk score groups. The results showed that patients in the high-risk score group exhibited worse survival outcomes, higher tumor grade, more advanced stage, more frequency of gene mutations, higher expression levels of immune checkpoints, and lower immunotherapy response. Conclusion We established a novel risk stratification of BLCA from a glycomics perspective, which demonstrated good accuracy in determining the prognostic outcome, clinicopathological characteristics, immune microenvironment, and immunotherapy efficacy of patients, and we are proposing to apply it to direct the choice of clinical treatment options for patients.
Collapse
Affiliation(s)
- Zhiyong Tan
- Department of Urology, the Second Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, People’s Republic of China
- Urological Disease Clinical Medical Center of Yunnan Province, the Second Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, People’s Republic of China
- Scientific and Technological Innovation Team of Basic and Clinical Research of Bladder Cancer in Yunnan Universities, the Second Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, People’s Republic of China
| | - Xiaorong Chen
- Department of Kidney Transplantation, the Third Hospital of Sun Yat-Sen University, Guangzhou, People’s Republic of China
| | - Jieming Zuo
- Department of Urology, the Second Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, People’s Republic of China
- Urological Disease Clinical Medical Center of Yunnan Province, the Second Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, People’s Republic of China
- Scientific and Technological Innovation Team of Basic and Clinical Research of Bladder Cancer in Yunnan Universities, the Second Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, People’s Republic of China
| | - Shi Fu
- Department of Urology, the Second Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, People’s Republic of China
- Urological Disease Clinical Medical Center of Yunnan Province, the Second Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, People’s Republic of China
- Scientific and Technological Innovation Team of Basic and Clinical Research of Bladder Cancer in Yunnan Universities, the Second Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, People’s Republic of China
| | - Jiansong Wang
- Department of Urology, the Second Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, People’s Republic of China
- Urological Disease Clinical Medical Center of Yunnan Province, the Second Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, People’s Republic of China
- Scientific and Technological Innovation Team of Basic and Clinical Research of Bladder Cancer in Yunnan Universities, the Second Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, People’s Republic of China
| | - Haifeng Wang
- Department of Urology, the Second Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, People’s Republic of China
- Urological Disease Clinical Medical Center of Yunnan Province, the Second Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, People’s Republic of China
- Scientific and Technological Innovation Team of Basic and Clinical Research of Bladder Cancer in Yunnan Universities, the Second Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, People’s Republic of China
| |
Collapse
|
10
|
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
|
11
|
Zhao S, Yang Y, Wang Y, Liu H, Ju H, Chen Y. In situ evaluation of in vivo sialylation with a dual-color imaging strategy. Chem Commun (Camb) 2023; 59:7815-7818. [PMID: 37272281 DOI: 10.1039/d3cc01949g] [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: 06/06/2023]
Abstract
This work designs a functional dendrimer probe to conveniently identify newly generated sialic acid groups in vivo with a dual-color imaging strategy, which achieves in situ semiquantitative evaluation of the sialylation difference between tumor and normal tissues to reveal sialylation-related biological events and promote clinical tumor diagnosis.
Collapse
Affiliation(s)
- Shiya Zhao
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210023, China.
| | - Yuanjiao Yang
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210023, China.
| | - Yuru Wang
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210023, China.
| | - Huipu Liu
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210023, China.
| | - Huangxian Ju
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210023, China.
| | - Yunlong Chen
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210023, China.
| |
Collapse
|
12
|
Zhou X, Chi K, Zhang C, Liu Q, Yang G. Sialylation: A Cloak for Tumors to Trick the Immune System in the Microenvironment. BIOLOGY 2023; 12:832. [PMID: 37372117 DOI: 10.3390/biology12060832] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Revised: 06/03/2023] [Accepted: 06/06/2023] [Indexed: 06/29/2023]
Abstract
The tumor microenvironment (TME), where the tumor cells incite the surrounding normal cells to create an immune suppressive environment, reduces the effectiveness of immune responses during cancer development. Sialylation, a type of glycosylation that occurs on cell surface proteins, lipids, and glycoRNAs, is known to accumulate in tumors and acts as a "cloak" to help tumor cells evade immunological surveillance. In the last few years, the role of sialylation in tumor proliferation and metastasis has become increasingly evident. With the advent of single-cell and spatial sequencing technologies, more research is being conducted to understand the effects of sialylation on immunity regulation. This review provides updated insights into recent research on the function of sialylation in tumor biology and summarizes the latest developments in sialylation-targeted tumor therapeutics, including antibody-mediated and metabolic-based sialylation inhibition, as well as interference with sialic acid-Siglec interaction.
Collapse
Affiliation(s)
- Xiaoman Zhou
- The Key Laboratory of Carbohydrate Chemistry and Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi 214122, China
| | - Kaijun Chi
- The Key Laboratory of Carbohydrate Chemistry and Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi 214122, China
| | - Chairui Zhang
- The Key Laboratory of Carbohydrate Chemistry and Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi 214122, China
| | - Quan Liu
- Department of Medical Oncology, Affiliated Hospital of Jiangnan University, Wuxi 214122, China
| | - Ganglong Yang
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing 100190, China
| |
Collapse
|
13
|
Tamoxifen Modulates the Immune Landscape of the Tumour Microenvironment: The Paired Siglec-5/14 Checkpoint in Anti-Tumour Immunity in an In Vitro Model of Breast Cancer. Int J Mol Sci 2023; 24:ijms24065512. [PMID: 36982588 PMCID: PMC10057974 DOI: 10.3390/ijms24065512] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Revised: 03/07/2023] [Accepted: 03/13/2023] [Indexed: 03/17/2023] Open
Abstract
Since the role of sialome–Siglec axis has been described as a regulatory checkpoint of immune homeostasis, the promotion of stimulatory or inhibitory Siglec-related mechanisms is crucial in cancer progression and therapy. Here, we investigated the effect of tamoxifen on the sialic acid–Siglec interplay and its significance in immune conversion in breast cancer. To mimic the tumour microenvironment, we used oestrogen-dependent or oestrogen-independent breast cancer cells/THP-1 monocytes transwell co-cultures exposed to tamoxifen and/or β-estradiol. We found changes in the cytokine profiles accompanied by immune phenotype switching, as measured by the expression of arginase-1. The immunomodulatory effects of tamoxifen in THP-1 cells occurred with the altered SIGLEC5 and SIGLEC14 genes and the expression of their products, as confirmed by RT-PCR and flow cytometry. Additionally, exposure to tamoxifen increased the binding of Siglec-5 and Siglec-14 fusion proteins to breast cancer cells; however, these effects appeared to be unassociated with oestrogen dependency. Our results suggest that tamoxifen-induced alterations in the immune activity of breast cancer reflect a crosstalk between the Siglec-expressing cells and the tumour’s sialome. Given the distribution of Siglec-5/14, the expression profile of inhibitory and activatory Siglecs in breast cancer patients may be useful in the verification of therapeutic strategies and predicting the tumour’s behaviour and the patient’s overall survival.
Collapse
|
14
|
Wang Y, Yang Y, Guo J, Ju H, Chen Y. Tumor identification via in vivo portable Raman detection of sialic acid with a dual gold nanoprobe system. Chem Sci 2023; 14:923-927. [PMID: 36755728 PMCID: PMC9890552 DOI: 10.1039/d2sc05163j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2022] [Accepted: 12/21/2022] [Indexed: 12/24/2022] Open
Abstract
A dual gold nanoprobe system was designed for in vivo portable Raman detection of sialic acid (SA) for tumor identification. The dual gold nanoprobe system contained two gold nanoprobes, Au10-DTTC/PEG-PBA and Au40-PEG-SA. Au10-DTTC/PEG-PBA was constructed on a 10 nm gold nanoparticle modified with 3,3'-diethylthia tricarbocyanine iodide (DTTCI) as the Raman reporter and 3-aminophenylboronic acid (APBA) through a thiol PEG succinimidyl carboxymethyl ester (HS-PEG-NHS) linker for specific recognition of SA. Au40-PEG-SA was constructed on a 40 nm gold nanoparticle modified with SA through HS-PEG-NHS. For in vivo detection of SA, Au10-DTTC/PEG-PBA and Au40-PEG-SA were subsequently injected into tumor xenografted mice with optimal interval and retention times. Through the specific recognition between PBA and SA, the conjugates of Au10-DTTC/PEG-PBA and Au40-PEG-SA formed in the tumor region emitted strong SERS signals of DTTC, which could be detected by a portable Raman detector. This work provides a convenient and portable method to detect SA in tumor xenografted mice, which is useful for family-stay identification and clinical cleavage of tumors.
Collapse
Affiliation(s)
- Yuru Wang
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University Nanjing 210023 China
| | - Yuanjiao Yang
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University Nanjing 210023 China
| | - Jingxing Guo
- School of Chemistry, Chemical Engineering and Life Sciences, Wuhan University of TechnologyWuhan 430070P. R. China
| | - Huangxian Ju
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University Nanjing 210023 China
| | - Yunlong Chen
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University Nanjing 210023 China
| |
Collapse
|
15
|
Modeling N-Glycosylation: A Systems Biology Approach for Evaluating Changes in the Steady-State Organization of Golgi-Resident Proteins. Methods Mol Biol 2022; 2557:663-690. [PMID: 36512244 DOI: 10.1007/978-1-0716-2639-9_40] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The organization of Golgi-resident proteins is crucial for sorting molecules within the secretory pathway and regulating posttranslational modifications. However, evaluating changes to Golgi organization can be challenging, often requiring extensive experimental investigations. Here, we propose a systems biology approach in which changes to Golgi-resident protein sorting and localization can be deduced using cellular N-glycan profiles as the only experimental input.The approach detailed here utilizes the influence of Golgi organization on N-glycan biosynthesis to investigate the mechanisms involved in establishing and maintaining Golgi organization. While N-glycosylation is carried out in a non-template-driven manner, the distribution of N-glycan biosynthetic enzymes within the Golgi ensures this process is not completely random. Therefore, changes to N-glycan profiles provide clues into how altered cell phenotypes affect the sorting and localization of Golgi-resident proteins. Here, we generate a stochastic simulation of N-glycan biosynthesis to produce a simulated glycan profile similar to that obtained experimentally and then combine this with Bayesian fitting to enable inference of changes in enzyme amounts and localizations. Alterations to Golgi organization are evaluated by calculating how the fitted enzyme parameters shift when moving from simulating the glycan profile of one cellular state (e.g., a wild type) to an altered cellular state (e.g., a mutant). Our approach illustrates how an iterative combination of mathematical systems biology and minimal experimental cell biology can be utilized to maximally integrate biological knowledge to gain insightful knowledge of the underlying mechanisms in a manner inaccessible to either alone.
Collapse
|
16
|
Duca M, Malagolini N, Dall’Olio F. The Mutual Relationship between Glycosylation and Non-Coding RNAs in Cancer and Other Physio-Pathological Conditions. Int J Mol Sci 2022; 23:ijms232415804. [PMID: 36555445 PMCID: PMC9781064 DOI: 10.3390/ijms232415804] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Revised: 12/08/2022] [Accepted: 12/09/2022] [Indexed: 12/15/2022] Open
Abstract
Glycosylation, which consists of the enzymatic addition of sugars to proteins and lipids, is one of the most important post-co-synthetic modifications of these molecules, profoundly affecting their activity. Although the presence of carbohydrate chains is crucial for fine-tuning the interactions between cells and molecules, glycosylation is an intrinsically stochastic process regulated by the relative abundance of biosynthetic (glycosyltransferases) and catabolic (glycosidases) enzymes, as well as sugar carriers and other molecules. Non-coding RNAs, which include microRNAs, long non-coding RNAs and circRNAs, establish a complex network of reciprocally interacting molecules whose final goal is the regulation of mRNA expression. Likewise, these interactions are stochastically regulated by ncRNA abundance. Thus, while protein sequence is deterministically dictated by the DNA/RNA/protein axis, protein abundance and activity are regulated by two stochastic processes acting, respectively, before and after the biosynthesis of the protein axis. Consequently, the worlds of glycosylation and ncRNA are closely interconnected and mutually interacting. In this paper, we will extensively review the many faces of the ncRNA-glycosylation interplay in cancer and other physio-pathological conditions.
Collapse
|
17
|
Nag S, Mandal A, Joshi A, Jain N, Srivastava RS, Singh S, Khattri A. Sialyltransferases and Neuraminidases: Potential Targets for Cancer Treatment. Diseases 2022; 10:diseases10040114. [PMID: 36547200 PMCID: PMC9777960 DOI: 10.3390/diseases10040114] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2022] [Revised: 11/06/2022] [Accepted: 11/16/2022] [Indexed: 11/29/2022] Open
Abstract
Cancers are the leading cause of death, causing around 10 million deaths annually by 2020. The most common cancers are those affecting the breast, lungs, colon, and rectum. However, it has been noted that cancer metastasis is more lethal than just cancer incidence and accounts for more than 90% of cancer deaths. Thus, early detection and prevention of cancer metastasis have the capability to save millions of lives. Finding novel biomarkers and targets for screening, determination of prognosis, targeted therapies, etc., are ways of doing so. In this review, we propose various sialyltransferases and neuraminidases as potential therapeutic targets for the treatment of the most common cancers, along with a few rare ones, on the basis of existing experimental and in silico data. This compilation of available cancer studies aiming at sialyltransferases and neuraminidases will serve as a guide for scientists and researchers working on possible targets for various cancers and will also provide data about the existing drugs which inhibit the action of these enzymes.
Collapse
Affiliation(s)
- Sagorika Nag
- Department of Pharmaceutical Engineering and Technology, Indian Institute of Technology, Banaras Hindu University, Varanasi 221005, India
| | - Abhimanyu Mandal
- Department of Pharmaceutical Engineering and Technology, Indian Institute of Technology, Banaras Hindu University, Varanasi 221005, India
| | - Aryaman Joshi
- Department of Chemical Engineering and Technology, Indian Institute of Technology, Banaras Hindu University, Varanasi 221005, India
| | - Neeraj Jain
- Division of Cancer Biology, CSIR-Central Drug Research Institute, Lucknow 226031, India
| | - Ravi Shanker Srivastava
- Department of Pharmacology, Career Institute of Medical Sciences & Hospital, Lucknow 226020, India
| | - Sanjay Singh
- Department of Pharmaceutical Engineering and Technology, Indian Institute of Technology, Banaras Hindu University, Varanasi 221005, India
| | - Arun Khattri
- Department of Pharmaceutical Engineering and Technology, Indian Institute of Technology, Banaras Hindu University, Varanasi 221005, India
- Correspondence: ; Tel.: +91-70-6811-1755
| |
Collapse
|
18
|
Ahmed R, Samanta S, Banerjee J, Kar SS, Dash SK. Modulatory role of miRNAs in thyroid and breast cancer progression and insights into their therapeutic manipulation. CURRENT RESEARCH IN PHARMACOLOGY AND DRUG DISCOVERY 2022; 3:100131. [PMID: 36568259 PMCID: PMC9780070 DOI: 10.1016/j.crphar.2022.100131] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Revised: 08/22/2022] [Accepted: 09/25/2022] [Indexed: 11/07/2022] Open
Abstract
Over the past few decades, thyroid cancer has become one of the most common types of endocrine cancer, contributing to an increase in prevalence. In the year 2020, there were 586,202 newly diagnosed cases of thyroid cancer around the world. This constituted approximately 3.0% of all patients diagnosed with cancer. The World Health Organization reported that there will be 2.3 million women receiving treatment for breast cancer in 2020, with 685,000. Despite the fact that carcinoma is one of the world's leading causes of death, there is still a paucity of information about its biology. MicroRNAs (miRNAs; miRs) are non-coding RNAs that can reduce gene expression by cleaving the 3' untranslated regions of mRNA. These factors make them a potential protein translation inhibitor. Diverse biological mechanisms implicated in the genesis of cancer are modulated by miRNA. The investigation of global miRNA expression in cancer showed regulatory activity through up regulation and down-regulation in several cancers, including thyroid cancer and breast cancer. In thyroid cancer, miRNA influences several cancers related signaling pathways through modulating MAPK, PI3K, and the RAS pathway. In breast cancer, the regulatory activity of miRNA was played through the cyclin protein family, protein kinases and their inhibitors, and other growth promoters or suppressors, which modulated cell proliferation and cell cycle progression. This article's goal is to discuss key miRNA expressions that are involved in the development of thyroid and breast cancer as well as their therapeutic manipulation for these two specific cancer types.
Collapse
Affiliation(s)
- Rubai Ahmed
- Department of Physiology, University of Gour Banga, Malda, 732103, West Bengal, India
| | - Sovan Samanta
- Department of Physiology, University of Gour Banga, Malda, 732103, West Bengal, India
| | - Jhimli Banerjee
- Department of Physiology, University of Gour Banga, Malda, 732103, West Bengal, India
| | - Suvrendu Sankar Kar
- Department of Medicine, R.G.Kar Medical College and Hospital, Kolkata, 700004, West Bengal, India
| | - Sandeep Kumar Dash
- Department of Physiology, University of Gour Banga, Malda, 732103, West Bengal, India,Corresponding author.
| |
Collapse
|
19
|
Aberrant Sialylation in Cancer: Therapeutic Opportunities. Cancers (Basel) 2022; 14:cancers14174248. [PMID: 36077781 PMCID: PMC9454432 DOI: 10.3390/cancers14174248] [Citation(s) in RCA: 49] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Revised: 08/15/2022] [Accepted: 08/23/2022] [Indexed: 11/16/2022] Open
Abstract
The surface of every eukaryotic cell is coated in a thick layer of glycans that acts as a key interface with the extracellular environment. Cancer cells have a different ‘glycan coat’ to healthy cells and aberrant glycosylation is a universal feature of cancer cells linked to all of the cancer hallmarks. This means glycans hold huge potential for the development of new diagnostic and therapeutic strategies. One key change in tumour glycosylation is increased sialylation, both on N-glycans and O-glycans, which leads to a dense forest of sialylated structures covering the cell surface. This hypersialylation has far-reaching consequences for cancer cells, and sialylated glycans are fundamental in tumour growth, metastasis, immune evasion and drug resistance. The development of strategies to inhibit aberrant sialylation in cancer represents an important opportunity to develop new therapeutics. Here, I summarise recent advances to target aberrant sialylation in cancer, including the development of sialyltransferase inhibitors and strategies to inhibit Siglecs and Selectins, and discuss opportunities for the future.
Collapse
|
20
|
Wang S, Wang R, Gao F, Huang J, Zhao X, Li D. Pan-cancer analysis of the DNA methylation patterns of long non-coding RNA. Genomics 2022; 114:110377. [PMID: 35513292 DOI: 10.1016/j.ygeno.2022.110377] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2022] [Revised: 03/23/2022] [Accepted: 04/27/2022] [Indexed: 11/04/2022]
Abstract
Long non-coding RNA (lncRNA) regulated by abnormal DNA methylation (ADM-lncRNA) emerges as a biomarker for cancer diagnosis and treatment. This study comprehensively described the methylation patterns of lncRNA in pan-cancer using the cancer data set in The Cancer Genome Atlas (TCGA). Based on the cancer heterogeneity of ADM-lncRNA in pan-cancer, we constructed a co-expression network of pan-cancer ADM-lncRNA (pADM-lncRNA) in 10 cancers, highlighting the combined action mode of abnormal DNA methylation, and indicating the internal connection among different cancers. Functional analysis revealed the pan-carcinogenic pathway of pADM-lncRNA and suggested potential factors for cancer heterogeneity and tumor immune microenvironment changes. Survival analysis showed the potential of pADM-lncRNA-mRNA co-expression pair as cancer biomarkers. Revealing the action mode of lncRNA and DNA methylation in cancer may help understand the key molecular mechanisms of cell carcinogenesis.
Collapse
Affiliation(s)
- Shijia Wang
- School of Biomedical Engineering, Capital Medical University, 10 You An Men Wai, Xi Tou Tiao, Beijing 100069, China; Beijing Key Laboratory of Fundamental Research on Biomechanics in Clinical, Capital Medical University, Beijing 100069, China
| | - Rendong Wang
- School of Biomedical Engineering, Capital Medical University, 10 You An Men Wai, Xi Tou Tiao, Beijing 100069, China; Beijing Key Laboratory of Fundamental Research on Biomechanics in Clinical, Capital Medical University, Beijing 100069, China
| | - Fang Gao
- Health Management Center, Binzhou People's Hospital, Shandong Province, China
| | - Jun Huang
- School of Biomedical Engineering, Capital Medical University, 10 You An Men Wai, Xi Tou Tiao, Beijing 100069, China; Beijing Key Laboratory of Fundamental Research on Biomechanics in Clinical, Capital Medical University, Beijing 100069, China
| | - Xiaoxiao Zhao
- School of Biomedical Engineering, Capital Medical University, 10 You An Men Wai, Xi Tou Tiao, Beijing 100069, China; Beijing Key Laboratory of Fundamental Research on Biomechanics in Clinical, Capital Medical University, Beijing 100069, China
| | - Dongguo Li
- School of Biomedical Engineering, Capital Medical University, 10 You An Men Wai, Xi Tou Tiao, Beijing 100069, China; Beijing Key Laboratory of Fundamental Research on Biomechanics in Clinical, Capital Medical University, Beijing 100069, China.
| |
Collapse
|
21
|
Vilen Z, Reeves AE, O’Leary TR, Joeh E, Kamasawa N, Huang ML. Cell Surface Engineering Enables Surfaceome Profiling. ACS Chem Biol 2022; 18:701-710. [PMID: 35443134 PMCID: PMC9901301 DOI: 10.1021/acschembio.1c00865] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Cell surface proteins (CSPs) are vital molecular mediators for cells and their extracellular environment. Thus, understanding which CSPs are displayed on cells, especially in different cell states, remains an important endeavor in cell biology. Here, we describe the integration of cell surface engineering with radical-mediated protein biotinylation to profile CSPs. This method relies on the prefunctionalization of cells with cholesterol lipid groups, followed by sortase-catalyzed conjugation with an APEX2 ascorbate peroxidase enzyme. In the presence of biotin-phenol and H2O2, APEX2 catalyzes the formation of highly reactive biotinyl radicals that covalently tag electron-rich residues within CSPs for subsequent streptavidin-based enrichment and analysis by quantitative mass spectrometry. While APEX2 is traditionally used to capture proximity-based interactomes, we envisioned using it in a "baitless" manner on cell surfaces to capture CSPs. We evaluate this strategy in light of another CSP labeling method that relies on the presence of cell surface sialic acid. Using the APEX2 strategy, we describe the CSPs found in three mammalian cell lines and compare CSPs in adherent versus three-dimensional pancreatic adenocarcinoma cells.
Collapse
Affiliation(s)
- Zak Vilen
- Department of Molecular Medicine, Scripps Research, 120 Scripps Way, Jupiter, FL 33458-5284,Skaggs Graduate School of Chemical and Biological Sciences, Scripps Research, 10550 N Torrey Pines Rd, La Jolla, CA 92037,Department of Molecular Medicine, Scripps Research, 10550 N Torrey Pines Rd, La Jolla, CA 92037
| | - Abigail E. Reeves
- Department of Molecular Medicine, Scripps Research, 120 Scripps Way, Jupiter, FL 33458-5284,Skaggs Graduate School of Chemical and Biological Sciences, Scripps Research, 10550 N Torrey Pines Rd, La Jolla, CA 92037,Department of Molecular Medicine, Scripps Research, 10550 N Torrey Pines Rd, La Jolla, CA 92037
| | - Timothy R. O’Leary
- Department of Molecular Medicine, Scripps Research, 120 Scripps Way, Jupiter, FL 33458-5284
| | - Eugene Joeh
- Department of Molecular Medicine, Scripps Research, 120 Scripps Way, Jupiter, FL 33458-5284,Skaggs Graduate School of Chemical and Biological Sciences, Scripps Research, 10550 N Torrey Pines Rd, La Jolla, CA 92037,Department of Molecular Medicine, Scripps Research, 10550 N Torrey Pines Rd, La Jolla, CA 92037
| | - Naomi Kamasawa
- The Imaging Center and Electron Microscopy Core Facility, Max Planck Florida Institute for Neuroscience, 1 Max Planck Way, Jupiter, FL, 33458
| | - Mia L. Huang
- Department of Molecular Medicine, Scripps Research, 120 Scripps Way, Jupiter, FL 33458-5284,Skaggs Graduate School of Chemical and Biological Sciences, Scripps Research, 10550 N Torrey Pines Rd, La Jolla, CA 92037,Department of Molecular Medicine, Scripps Research, 10550 N Torrey Pines Rd, La Jolla, CA 92037,Corresponding author:
| |
Collapse
|
22
|
Zarrineh M, Ashrafian S, Jensen P, Nawrocki A, Ansari AM, Rezadoost H, Ghassempour A, Larsen MR. Comprehensive proteomics and sialiomics of the anti-proliferative activity of safranal on triple negative MDA-MB-231 breast cancer cell lines. J Proteomics 2022; 259:104539. [PMID: 35240313 DOI: 10.1016/j.jprot.2022.104539] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Revised: 12/27/2021] [Accepted: 02/04/2022] [Indexed: 12/17/2022]
Abstract
Triple-negative breast cancer (TNBC) is an aggressive subtype of breast cancer with no efficient treatment. Researchers have indicated the importance of quantitative approaches on proteome and different post-translation modifications studies both in diagnosis and treatment purposes. Sialic acid-containing glycopeptides (the sialiome) is one of these modifications which can be used as a tool in cancer diagnosis or therapeutic strategies since the sialylation is strongly associated with cancer migration and metastasis. Based on our study, safranal, which is a non-toxic compound in orally intakes, exhibits a significant cytotoxic effect on MDA-MB-231 in comparison to normal cells. We conducted a comprehensive proteomics and sialiomics analysis of safranal treated MDA-MB-231 cells by using a combination of TMT labeling and titanium dioxide enrichment of sialylated N-linked glycopeptides to investigate the underlying molecular mechanism behind safranal-induced apoptosis. Safranal has main effect on the inhibition of metabolism and mitochondrial dysfunction. It regulates proteins considered as activator of DNA fragmentation and apoptosis mediators. Moreover, safranal regulates sialylation of glycoproteins involving in cellular adhesion, migration and survival. It suppresses cell survival and metastasis through the alteration of the sialylation level on important signaling receptors. These results highlight the impact of safranal as a potent anticancer compound on TNBCs which also can be strongly used in daily diets. SIGNIFICANCE: In first step, we evaluated the cell viability of MDA-MB-231 cell lines against the purified saffron components (total crocin, picrocrocin, crocin I and safranal). Safranal was the only compound demonstrated the anti-proliferation effect. In order to obtain an understanding of safranal cytotoxic effect on MDA-MB-231, we designed the three set of treated cell lines in 30 min, 12 h and 24 h time-points in three replicates and a combination of TMT-based labeling quantitative proteomics and titanium dioxide (TiO2)-based enrichment of sialylated N-linked glycopeptides for sialiomics analysis as a strategy to follow the more detailed mechanisms of safranal effect. The results of bioinformatics analysis revealed the multifunction role of safranal on MDA-MB-231 cell lines. Safranal mainly dysregulates mitochondrial function, inhibits metabolism and starts initial signaling of apoptosis which lead to DNA fragmentation. Moreover, safranal caused the majority of down-regulation in sialylation profile in all time-points. Safranal also declines the cell survival, adhesion and migration by dysregulation of the sialylation level in important proteins including integrins, tumor necrosis factor receptor and cell adhesion molecules (CAMs). The results provide a set of therapeutic targets for triple negative breast cancer which can help designing of effective anticancer drugs specially in targeted therapies.
Collapse
Affiliation(s)
- Mahshid Zarrineh
- Medicinal Plants and Drugs Research Institute, Shahid Beheshti University, Tehran 1983963113, Iran
| | - Shahrbanou Ashrafian
- Medicinal Plants and Drugs Research Institute, Shahid Beheshti University, Tehran 1983963113, Iran
| | - Pia Jensen
- Protein research group, Department of Biochemistry and Molecular Biology, University of Southern Denmark, DK-5230 Odense M, Denmark
| | - Arkadiusz Nawrocki
- Protein research group, Department of Biochemistry and Molecular Biology, University of Southern Denmark, DK-5230 Odense M, Denmark
| | - Alireza Madjid Ansari
- Integrative Oncology Department, Breast Cancer Research Center, Motamed Cancer Institute, ACECR, Tehran, Iran
| | - Hassan Rezadoost
- Medicinal Plants and Drugs Research Institute, Shahid Beheshti University, Tehran 1983963113, Iran
| | - Alireza Ghassempour
- Medicinal Plants and Drugs Research Institute, Shahid Beheshti University, Tehran 1983963113, Iran.
| | - Martin R Larsen
- Protein research group, Department of Biochemistry and Molecular Biology, University of Southern Denmark, DK-5230 Odense M, Denmark.
| |
Collapse
|
23
|
Rosa-Fernandes L, Oba-Shinjo SM, Macedo-da-Silva J, Marie SKN, Palmisano G. Aberrant Protein Glycosylation in Brain Cancers, with Emphasis on Glioblastoma. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2022; 1382:39-70. [DOI: 10.1007/978-3-031-05460-0_4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/09/2022]
|
24
|
Hugonnet M, Singh P, Haas Q, von Gunten S. The Distinct Roles of Sialyltransferases in Cancer Biology and Onco-Immunology. Front Immunol 2021; 12:799861. [PMID: 34975914 PMCID: PMC8718907 DOI: 10.3389/fimmu.2021.799861] [Citation(s) in RCA: 53] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2021] [Accepted: 12/02/2021] [Indexed: 12/24/2022] Open
Abstract
Aberrant glycosylation is a key feature of malignant transformation. Hypersialylation, the enhanced expression of sialic acid-terminated glycoconjugates on the cell surface, has been linked to immune evasion and metastatic spread, eventually by interaction with sialoglycan-binding lectins, including Siglecs and selectins. The biosynthesis of tumor-associated sialoglycans involves sialyltransferases, which are differentially expressed in cancer cells. In this review article, we provide an overview of the twenty human sialyltransferases and their roles in cancer biology and immunity. A better understanding of the individual contribution of select sialyltransferases to the tumor sialome may lead to more personalized strategies for the treatment of cancer.
Collapse
Affiliation(s)
- Marjolaine Hugonnet
- Institute of Pharmacology, University of Bern, Bern, Switzerland
- Bern Center for Precision Medicine (BCPM), University of Bern, Bern, Switzerland
| | - Pushpita Singh
- Institute of Pharmacology, University of Bern, Bern, Switzerland
| | - Quentin Haas
- Institute of Pharmacology, University of Bern, Bern, Switzerland
| | - Stephan von Gunten
- Institute of Pharmacology, University of Bern, Bern, Switzerland
- Bern Center for Precision Medicine (BCPM), University of Bern, Bern, Switzerland
| |
Collapse
|
25
|
Tang Y, Jia C, Wang Y, Wan W, Li H, Huang G, Zhang X. Lactate Consumption via Cascaded Enzymes Combined VEGF siRNA for Synergistic Anti-Proliferation and Anti-Angiogenesis Therapy of Tumors. Adv Healthc Mater 2021; 10:e2100799. [PMID: 34310079 DOI: 10.1002/adhm.202100799] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2021] [Revised: 06/08/2021] [Indexed: 02/07/2023]
Abstract
Lactate, as the most abundant component with concentrations of 4-40 mm in tumors, contributes to the regulation of metabolic pathways, angiogenesis, and immunosuppression, exhibiting remarkable potential in cancer treatment. Therefore, a codelivery strategy that combined the cascaded enzymes Lactate oxidase/Catalase (LOx/CAT) and vascular endothelial growth factor (VEGF) siRNA (siVEGF) to suppress tumor proliferation and angiogenesis synergistically is creatively proposed. In brief, the cationic liposomes (LIP) encapsulated with LOx/CAT and siVEGF via hydrophilic interaction and electrostatic adsorption followed by coating with PEGylated phenylboronic acid (PP) is established (PPL@[LOX+CAT]). Moreover, a simple 3-aminophenylboronic acid (PBA)-shielded strategy via fructose (Fru) is applied to further enhance the targeting efficiency in the tumor site. The obtained co-encapsulated nanoparticles (NPs) can simultaneous intracellular release of LOx/CAT and siVEGF, and the collaborative use of LOx and CAT can promote lactate consumption even under a hypoxic tumor microenvironment (TME) without producing systemic toxicity. The combined application of lactate depletion and VEGF silencing demonstrated the efficient migration suppression of 4T1 cells in vitro and superior antitumor and antimetastatic properties in vivo. This work offers a promising tumor treatment strategy via integrating cascaded enzymes and gene therapy, and explores a promising therapy regimen for 4T1 triple-negative breast cancer.
Collapse
Affiliation(s)
- Yan Tang
- Department of Pharmaceutics College of Pharmaceutical Sciences Soochow University Suzhou 215123 China
| | - Changhao Jia
- Department of Pharmaceutics College of Pharmaceutical Sciences Soochow University Suzhou 215123 China
| | - Yu Wang
- Department of Pharmaceutics College of Pharmaceutical Sciences Soochow University Suzhou 215123 China
| | - Wenjun Wan
- Department of Pharmaceutics College of Pharmaceutical Sciences Soochow University Suzhou 215123 China
| | - Hui Li
- Department of Pharmaceutics College of Pharmaceutical Sciences Soochow University Suzhou 215123 China
| | - Gui Huang
- Department of Pharmaceutics College of Pharmaceutical Sciences Soochow University Suzhou 215123 China
| | - Xuenong Zhang
- Department of Pharmaceutics College of Pharmaceutical Sciences Soochow University Suzhou 215123 China
| |
Collapse
|
26
|
Wang N, Cao S, Wang X, Zhang L, Yuan H, Ma X. lncRNA MALAT1/miR‑26a/26b/ST8SIA4 axis mediates cell invasion and migration in breast cancer cell lines. Oncol Rep 2021; 46:181. [PMID: 34278507 PMCID: PMC8273684 DOI: 10.3892/or.2021.8132] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2021] [Accepted: 06/03/2021] [Indexed: 12/25/2022] Open
Abstract
Metastasis-associated lung adenocarcinoma transcript 1 (MALAT1) is a long non-coding RNA that is overexpressed in various human cancers, including breast cancer. Evidence has associated the function of the α-2,8-sialyltransferase (ST8SIA) family with breast cancer. The present study aimed to investigate the potential roles of MALAT1 in breast cancer development and progression using analyses of both breast cancer tissues and cell lines. The mRNA levels of MALAT1, microRNA (miR)-26a/26b and ST8SIA4 were detected by reverse transcription-quantitative PCR (RT-qPCR) and the protein level of ST8SIA4 was assessed by western blot analysis. Cell proliferation, invasion and migration were detected by CCK-8, wound healing and Transwell assays, respectively. Interactions between MALAT1 and miR-26a/26b were assessed using fluorescence in situ hybridization, RNA immunoprecipitation and luciferase reporter assays. Herein, different levels of MALAT1 were primarily observed in human breast cancer samples and cells. Upregulated MALAT1 was a crucial predictor of poor breast cancer prognosis. Altered MALAT1 modulated cell progression in breast cancer. Moreover, miR-26a/26b was confirmed as a direct regulator of MALAT1, and ST8SIA4 was predicted as a target of miR-26a/26b. Functional analysis in human breast cancer cell lines demonstrated that MALAT1 modulated breast cancer cell tumorigenicity by acting as a competing endogenous lncRNA (ceRNA) to regulate ST8SIA4 levels by sponging miR-26a/26b. The identification of the MALAT1/miR-26a/26b/ST8SIA4 axis which contributes to breast cancer progression may constitute a potential new therapeutic target.
Collapse
Affiliation(s)
- Nan Wang
- Department of Clinical Laboratory Medicine, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning 116011, P.R. China
| | - Shengji Cao
- Department of Clinical Laboratory Medicine, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning 116011, P.R. China
| | - Xiaoxi Wang
- Department of Clinical Laboratory Medicine, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning 116011, P.R. China
| | - Lina Zhang
- Department of Radiology Medicine, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning 116011, P.R. China
| | - Hong Yuan
- Department of Clinical Laboratory Medicine, Dalian Municipal Central Hospital, Dalian, Liaoning 116033, P.R. China
| | - Xiaolu Ma
- Department of Clinical Laboratory Medicine, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning 116011, P.R. China
| |
Collapse
|
27
|
Harvey DJ. ANALYSIS OF CARBOHYDRATES AND GLYCOCONJUGATES BY MATRIX-ASSISTED LASER DESORPTION/IONIZATION MASS SPECTROMETRY: AN UPDATE FOR 2015-2016. MASS SPECTROMETRY REVIEWS 2021; 40:408-565. [PMID: 33725404 DOI: 10.1002/mas.21651] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/06/2020] [Accepted: 07/24/2020] [Indexed: 06/12/2023]
Abstract
This review is the ninth update of the original article published in 1999 on the application of matrix-assisted laser desorption/ionization (MALDI) mass spectrometry to the analysis of carbohydrates and glycoconjugates and brings coverage of the literature to the end of 2016. Also included are papers that describe methods appropriate to analysis by MALDI, such as sample preparation techniques, even though the ionization method is not MALDI. Topics covered in the first part of the review include general aspects such as theory of the MALDI process, matrices, derivatization, MALDI imaging, fragmentation and arrays. The second part of the review is devoted to applications to various structural types such as oligo- and poly-saccharides, glycoproteins, glycolipids, glycosides and biopharmaceuticals. Much of this material is presented in tabular form. The third part of the review covers medical and industrial applications of the technique, studies of enzyme reactions and applications to chemical synthesis. The reported work shows increasing use of combined new techniques such as ion mobility and the enormous impact that MALDI imaging is having. MALDI, although invented over 30 years ago is still an ideal technique for carbohydrate analysis and advancements in the technique and range of applications show no sign of deminishing. © 2020 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- David J Harvey
- Nuffield Department of Medicine, Target Discovery Institute, University of Oxford, Roosevelt Drive, Oxford, OX3 7FZ, United Kingdom
| |
Collapse
|
28
|
Singh P, Kumari M, Bal A, Srinivasan R, Ghosh S. Heat shock protein 60 is a disease-associated sialoglycoprotein in human non-small cell lung cancer. Biol Chem 2021; 401:969-983. [PMID: 32049642 DOI: 10.1515/hsz-2019-0352] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2019] [Accepted: 02/03/2020] [Indexed: 01/23/2023]
Abstract
The diagnostic and therapeutic potential of Maackia amurensis agglutinin (MAA) have been reported in various malignancies. Earlier, we have found that MAA specifically interacted with human non-small cell lung-cancer (NSCLC) cells and induced apoptosis in these cells. The present study was designed to identify M. amurensis leukoagglutinin (MAL-I, one of the components of MAA, having the same carbohydrate specificity as MAA) interacting membrane sialoglycoprotein(s) of two subtypes of human NSCLC cell lines. Nine proteins were identified using two-dimensional (2D)-polyacrylamide gel electrophoresis (PAGE) followed by MAL-I-overlay transblotting and matrix assisted laser desorption ionization-time of flight mass spectrometry (MALDI-TOF-MS). Among these proteins, HSP60 was selected for further characterization. The sialoglycoprotein nature of membrane-HSP60 of NSCLC cell lines was confirmed by its reduced reactivity with MAL-I in Western blots in the presence of GM2 and by dual staining of the cell lines with MAL-I and HSP60-antibody. These findings were further substantiated by enzymatic analysis of membrane-HSP60 as well as in-silico evidence regarding this protein. Our observations were validated by immunohistochemical analysis of both subtypes of NSCLC tissue sections. Membrane-HSP60 was found to be involved in the inhibition of MAL-I-induced morphological alteration of NSCLC cells and also in the proliferation and migration of these cells, indicating the probable role of sialylated membrane-HSP60 in this disease.
Collapse
Affiliation(s)
- Praveen Singh
- Department of Experimental Medicine and Biotechnology, PGIMER, Chandigarh 160012, India
| | - Munmun Kumari
- Department of Experimental Medicine and Biotechnology, PGIMER, Chandigarh 160012, India
| | - Amanjit Bal
- Department of Histopathology, PGIMER, Chandigarh 160012, India
| | - Radhika Srinivasan
- Department of Cytology and Gynecological Pathology, PGIMER, Chandigarh 160012, India
| | - Sujata Ghosh
- Department of Experimental Medicine and Biotechnology, PGIMER, Chandigarh 160012, India
| |
Collapse
|
29
|
Aberrant Sialylation in Cancer: Biomarker and Potential Target for Therapeutic Intervention? Cancers (Basel) 2021; 13:cancers13092014. [PMID: 33921986 PMCID: PMC8122436 DOI: 10.3390/cancers13092014] [Citation(s) in RCA: 60] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Revised: 04/19/2021] [Accepted: 04/20/2021] [Indexed: 12/14/2022] Open
Abstract
Simple Summary Sialylation is a post-translational modification that consists in the addition of sialic acid to growing glycan chains on glycoproteins and glycolipids. Aberrant sialylation is an established hallmark of several types of cancer, including breast, ovarian, pancreatic, prostate, colorectal and lung cancers, melanoma and hepatocellular carcinoma. Hypersialylation can be the effect of increased activity of sialyltransferases and results in an excess of negatively charged sialic acid on the surface of cancer cells. Sialic acid accumulation contributes to tumor progression by several paths, including stimulation of tumor invasion and migration, and enhancing immune evasion and tumor cell survival. In this review we explore the mechanisms by which sialyltransferases promote cancer progression. In addition, we provide insights into the possible use of sialyltransferases as biomarkers for cancer and summarize findings on the development of sialyltransferase inhibitors as potential anti-cancer treatments. Abstract Sialylation is an integral part of cellular function, governing many biological processes including cellular recognition, adhesion, molecular trafficking, signal transduction and endocytosis. Sialylation is controlled by the levels and the activities of sialyltransferases on glycoproteins and lipids. Altered gene expression of these enzymes in cancer yields to cancer-specific alterations of glycoprotein sialylation. Mounting evidence indicate that hypersialylation is closely associated with cancer progression and metastatic spread, and can be of prognostic significance in human cancer. Aberrant sialylation is not only a result of cancer, but also a driver of malignant phenotype, directly impacting key processes such as tumor cell dissociation and invasion, cell-cell and cell-matrix interactions, angiogenesis, resistance to apoptosis, and evasion of immune destruction. In this review we provide insights on the impact of sialylation in tumor progression, and outline the possible application of sialyltransferases as cancer biomarkers. We also summarize the most promising findings on the development of sialyltransferase inhibitors as potential anti-cancer treatments.
Collapse
|
30
|
Suleiman RB, Muhammad A, Umara IA, Ibrahima MA, Erukainure OL, Forcados GE, Katsayal SB. Kolaviron Ameliorates 7, 12-Dimethylbenzanthracene - Induced Mammary Damage in Female Wistar Rats. Anticancer Agents Med Chem 2021; 22:181-192. [PMID: 34225638 DOI: 10.2174/1871520621666210322101232] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2020] [Revised: 12/27/2020] [Accepted: 01/25/2021] [Indexed: 11/22/2022]
Abstract
BACKGROUND Kolaviron (KV) is a flavonoid rich portion obtained from Garcinia kola seeds with a number of reported pharmacological effects. However, its ameliorative effects on 7,12-Dimethylbenzanthracene (DMBA)-induced mammary damage has not been fully investigated, despite the reported use of the seeds in the treatment of inflammatory related disorders. OBJECTIVE To evaluate the ameliorative effects of KV on DMBA-induced mammary damage in female Wistar rats. METHODS Forty-nine (49) female Wistar rats were randomly assigned into seven groups of seven rats each. DMBA was administered orally to rats in five of the groups as a single dose of 80 mg/kg body wt while the remaining two groups received the vehicle. The rats were palpated weekly for 3 months to monitor tumor formation. After 3 months of DMBA administration, 1 ml of blood was collected to assay for estrogen receptor- α (ER-α) level. Thereafter, the vehicle (dimethyl sulfoxide) was daily administered to the negative control and positive control groups for the 14 days duration of the experiment while three groups were each given a daily oral dose of 50, 100 and 200 mg/kg body wt of KV for the duration of the experiment. The last DMBA-induced group received 10 mg/kg body wt of the standard drug tamoxifen twice in a week and the remaining DMBA-free group received 200 mg/kg body wt KV. Subsequently, the animals were humanly sacrificed and ER-α, sialic acids, sialidase, sialyltransferase levels were assay for in blood and mammary tissues followed by histopathological examinations. RESULTS Significantly higher levels of estrogen receptor-α (ER-α), formation of lobular neoplastic cells, epithelial hyperplasia, lymphocyte infiltration and increased sialylation were detected in DMBA-induced rats. Treatment with KV at 50, 100 and 200 mg/kg body weight resulted in a significant (p<0.05) decrease in ER-α level, significantly (p<0.05) lower free serum sialic acid (21.1%), total sialic acid level of the mammary tissue (21.57%), sialyltransferase activity (30.83%) as well as mRNA level of the sialyltransferase gene (ST3Gal1) were observed after KV interventions. CONCLUSION The findings suggest that KV could be further explored in targeting DMBA-induced mammary damage implicated in mammary carcinogenesis.
Collapse
Affiliation(s)
- Rabiatu B Suleiman
- Department of Biochemistry, Faculty of Life Sciences, Ahmadu Bello University, Zaria, Kaduna State, Nigeria
| | - Aliyu Muhammad
- Department of Biochemistry, Faculty of Life Sciences, Ahmadu Bello University, Zaria, Kaduna State, Nigeria
| | - Ismaila A Umara
- Department of Biochemistry, Faculty of Life Sciences, Ahmadu Bello University, Zaria, Kaduna State, Nigeria
| | - Mohammed A Ibrahima
- Department of Biochemistry, Faculty of Life Sciences, Ahmadu Bello University, Zaria, Kaduna State, Nigeria
| | - Ochuko L Erukainure
- Department of Pharmacology, University of the Free State, Bloemfontein 9300. South Africa
| | - Gilead E Forcados
- Department of Biochemistry, Faculty of Life Sciences, Ahmadu Bello University, Zaria, Kaduna State, Nigeria
| | - Sanusi B Katsayal
- Department of Biochemistry, Faculty of Life Sciences, Ahmadu Bello University, Zaria, Kaduna State, Nigeria
| |
Collapse
|
31
|
Rezaei Z, Sadri F. MicroRNAs Involved in Inflammatory Breast Cancer: Oncogene and Tumor Suppressors with Possible Targets. DNA Cell Biol 2021; 40:499-512. [PMID: 33493414 DOI: 10.1089/dna.2020.6320] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Inflammatory breast cancer (IBC) as a rare and highly aggressive type of breast cancer displays phenotypic characteristics. To date, the IBC-associated molecular mechanisms are entirely unknown. In addition, there is an urgent need to identify the new biomarkers involved in the diagnosis and therapeutic purposes of IBC. MicroRNAs, a category of short noncoding RNAs, are capable of controlling the post-transcriptional expression of genes and thus can act as diagnostic predictive tools. In this review, we addressed the status of oncogenic and tumor suppressor miRNA-mediated IBC in current studies. Furthermore, based on their targets, their involvement in cancer progression, angiogenesis, metastasis, and apoptosis were determined.
Collapse
Affiliation(s)
- Zohreh Rezaei
- Department of Biology, University of Sistan and Baluchestan, Zahedan, Iran.,Cellular and Molecular Research Center, Birjand University of Medical Sciences, Birjand, Iran
| | - Farzad Sadri
- Student Research Committee, Birjand University of Medical Sciences, Birjand, Iran.,Young Researchers and Elite Club, Yasooj Branch, Islamic Azad University, Yasooj, Iran
| |
Collapse
|
32
|
Fu W, Yu G, Liang J, Fan P, Dong K, Zhang B, Chen X, Zhu H, Chu L. miR-144-5p and miR-451a Inhibit the Growth of Cholangiocarcinoma Cells Through Decreasing the Expression of ST8SIA4. Front Oncol 2021; 10:563486. [PMID: 33520692 PMCID: PMC7841262 DOI: 10.3389/fonc.2020.563486] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Accepted: 12/02/2020] [Indexed: 12/15/2022] Open
Abstract
Accumulating evidences indicate that non-coding RNAs play crucial roles in the progression of an extensive range of carcinomas. This study aimed to investigate the action mechanism of miR-144-5p and miR-451a in cholangiocarcinoma. We found that miR-144-5p and miR-451a were significantly decreased in cholangiocarcinoma patient samples compared to the adjacent normal bile duct samples. The downregulation of these two miRNAs was correlated with a more advanced disease state of cholangiocarcinoma patients. Overexpression of miR-144-5p and miR-451a suppressed the proliferation, invasion and migration of cholangiocarcinoma cells in vitro and inhibited xenograft tumor growth. Knockdown of these two miRNAs had the opposite effects. miR-144-5p and miR-451a regulated the expression of ST8 alpha-N-acetyl-neuraminide alpha-2,8-sialyltransferase 4 (ST8SIA4), and presented a correlation with ST8SIA4 in patient samples. Overexpression of ST8SIA4 promoted the proliferation, invasion and migration of cholangiocarcinoma cells, and the changes were reversed by upregulating the expression of miR-144-5p and miR-451a. Our findings indicated that miR-144-5p and miR-451a displayed a tumor suppressor role through decreasing the expression of ST8SIA4 in cholangiocarcinoma.
Collapse
Affiliation(s)
- Wan Fu
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Hubei Provincial Key Laboratory of Hepatobiliary and Pancreatic Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Guangcai Yu
- Institute of Organ Transplantation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Junnan Liang
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Hubei Provincial Key Laboratory of Hepatobiliary and Pancreatic Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Pan Fan
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Hubei Provincial Key Laboratory of Hepatobiliary and Pancreatic Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Keshuai Dong
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Hubei Provincial Key Laboratory of Hepatobiliary and Pancreatic Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Bixiang Zhang
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Hubei Provincial Key Laboratory of Hepatobiliary and Pancreatic Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaoping Chen
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Hubei Provincial Key Laboratory of Hepatobiliary and Pancreatic Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Hong Zhu
- Department of Medical Oncology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Liang Chu
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Hubei Provincial Key Laboratory of Hepatobiliary and Pancreatic Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
33
|
Wu HJ, Chu PY. Recent Discoveries of Macromolecule- and Cell-Based Biomarkers and Therapeutic Implications in Breast Cancer. Int J Mol Sci 2021; 22:ijms22020636. [PMID: 33435254 PMCID: PMC7827149 DOI: 10.3390/ijms22020636] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Revised: 12/31/2020] [Accepted: 01/08/2021] [Indexed: 12/13/2022] Open
Abstract
Breast cancer is the most commonly diagnosed cancer type and the leading cause of cancer-related mortality in women worldwide. Breast cancer is fairly heterogeneous and reveals six molecular subtypes: luminal A, luminal B, HER2+, basal-like subtype (ER−, PR−, and HER2−), normal breast-like, and claudin-low. Breast cancer screening and early diagnosis play critical roles in improving therapeutic outcomes and prognosis. Mammography is currently the main commercially available detection method for breast cancer; however, it has numerous limitations. Therefore, reliable noninvasive diagnostic and prognostic biomarkers are required. Biomarkers used in cancer range from macromolecules, such as DNA, RNA, and proteins, to whole cells. Biomarkers for cancer risk, diagnosis, proliferation, metastasis, drug resistance, and prognosis have been identified in breast cancer. In addition, there is currently a greater demand for personalized or precise treatments; moreover, the identification of novel biomarkers to further the development of new drugs is urgently needed. In this review, we summarize and focus on the recent discoveries of promising macromolecules and cell-based biomarkers for the diagnosis and prognosis of breast cancer and provide implications for therapeutic strategies.
Collapse
Affiliation(s)
- Hsing-Ju Wu
- Department of Biology, National Changhua University of Education, Changhua 500, Taiwan;
- Research Assistant Center, Show Chwan Memorial Hospital, Changhua 500, Taiwan
- Department of Medical Research, Chang Bing Show Chwan Memorial Hospital, Lukang Town, Changhua County 505, Taiwan
| | - Pei-Yi Chu
- School of Medicine, College of Medicine, Fu Jen Catholic University, New Taipei City 231, Taiwan
- Department of Pathology, Show Chwan Memorial Hospital, No. 542, Sec. 1 Chung-Shan Rd., Changhua 500, Taiwan
- Department of Health Food, Chung Chou University of Science and Technology, Changhua 510, Taiwan
- National Institute of Cancer Research, National Health Research Institutes, Tainan 704, Taiwan
- Correspondence: ; Tel.: +886-975-611-855; Fax: +886-4-7227-116
| |
Collapse
|
34
|
Läubli H, Kawanishi K, George Vazhappilly C, Matar R, Merheb M, Sarwar Siddiqui S. Tools to study and target the Siglec-sialic acid axis in cancer. FEBS J 2020; 288:6206-6225. [PMID: 33251699 DOI: 10.1111/febs.15647] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2020] [Revised: 11/18/2020] [Accepted: 11/23/2020] [Indexed: 12/16/2022]
Abstract
Siglecs are widely expressed on leucocytes and bind to ubiquitously presented glycans containing sialic acids (sialoglycans). Most Siglecs carry an immunoreceptor tyrosine-based inhibition motif (ITIM) and elicit an inhibitory intracellular signal upon ligand binding. A few Siglec receptors can, however, recruit immunoreceptor tyrosine-based activation motif (ITAM)-containing factors, which activate cells. The role of hypersialylation (the enhanced expression of sialoglycans) has recently been explored in cancer progression. Mechanistic studies have shown that hypersialylation on cancer cells can engage inhibitory Siglecs on the surface of immune cells and induce immunosuppression. These recent studies strongly suggest that the Siglec-sialic acid axis can act as a potential target for cancer immunotherapy. Moreover, the use of new tools and techniques is facilitating these studies. In this review, we summarise techniques used to study Siglecs, including different mouse models, monoclonal antibodies, Siglec fusion proteins, and sialoglycan arrays. Furthermore, we discuss the recent major developments in the study of Siglecs in cancer immunosuppression, tools, and techniques used in targeting the Siglec-sialic acid axis and the possibility of clinical intervention.
Collapse
Affiliation(s)
- Heinz Läubli
- Laboratory for Cancer Immunotherapy, Department of Biomedicine, and Medical Oncology, Department of Internal Medicine, University Hospital Basel, Switzerland
| | - Kunio Kawanishi
- Kidney and Vascular Pathology, University of Tsukuba, Ibaraki, Japan
| | | | - Rachel Matar
- Department of Biotechnology, American University of Ras Al Khaimah (AURAK), UAE
| | - Maxime Merheb
- Department of Biotechnology, American University of Ras Al Khaimah (AURAK), UAE
| | | |
Collapse
|
35
|
Abstract
Glycosylation is a sophisticated informational system that controls specific biological functions at the cellular and organismal level. Dysregulation of glycosylation may underlie some of the most complex and common diseases of the modern era. In the past 5 years, microRNAs have come to the forefront as a critical regulator of the glycome. Herein, we review the current literature on miRNA regulation of glycosylation and how this work may point to a new way to identify the biological importance of glycosylation enzymes.
Collapse
Affiliation(s)
- Chu T Thu
- Biomedical Chemistry Institute, Department of Chemistry, New York University, New York, New York 10003, United States
| | - Lara K Mahal
- Biomedical Chemistry Institute, Department of Chemistry, New York University, New York, New York 10003, United States
| |
Collapse
|
36
|
The Emerging Role of MicroRNAs in Breast Cancer. JOURNAL OF ONCOLOGY 2020; 2020:9160905. [PMID: 32714393 PMCID: PMC7354667 DOI: 10.1155/2020/9160905] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 04/04/2020] [Accepted: 06/10/2020] [Indexed: 12/24/2022]
Abstract
Breast cancer (BC) is the most common malignancy in women. Due to BC heterogeneity, complexity, and metastasis, many BC patients do not successfully respond to therapies. The effective management of BC depends on early diagnosis and monitoring of drug response. Therefore, identifying new biomarkers for the diagnosis, prognosis, and development of new drugs is urgently required. Dysregulation of microRNAs (miRNAs) participates in the tumorigenesis and progression of cancers, especially breast cancer (BC). Several studies demonstrated that miRNAs could perform their function as oncogenes or tumor suppressors. This review describes recent progress on the role of microRNAs in the diagnosis, prognosis, hallmark, and treatment of BC. According to a recent literature survey, miRNAs play a pivotal role in the regulation of hallmarks of cancer, such as proliferation, apoptosis, invasion, metastasis, and tumor stemness. Many miRNAs are potential biomarkers for BC for diagnosis, and some are indicators of prognosis. Moreover, circulating miRNA profiles, as minimally invasive, diagnostic, and prognostic markers, are broadly used in BC therapy, and some miRNAs are good predictors of therapeutic outcomes. Other miRNAs are involved in overcoming chemoresistance and in increasing BC drug sensitivity.
Collapse
|
37
|
SS Pindiprolu SK, Krishnamurthy PT, Ghanta VR, Chintamaneni PK. Phenyl boronic acid-modified lipid nanocarriers of niclosamide for targeting triple-negative breast cancer. Nanomedicine (Lond) 2020; 15:1551-1565. [DOI: 10.2217/nnm-2020-0003] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Aim: To study the active targeting efficacy of phenylboronic acid-modified niclosamide solid lipid nanoparticles (PBA-Niclo-SLN) in triple-negative breast cancer (TNBC). Materials & methods: PBA-Niclo-SLNs were formulated by an emulsification-solvent evaporation method using PBA-associated stearylamine (PBSA) as lipid. The drug uptake and the anticancer propensity of PBA-Niclo-SLN were studied in TNBC (MDA-MB231) cells and tumor-bearing mice. Results: PBA-Niclo-SLN formulation resulted in greater antitumor efficacy by inducing G0/G1 cell cycle arrest and apoptosis. Besides, PBA-Niclo-SLN effectively inhibited STAT3, CD44+/CD24- TNBC stem cell subpopulation, epithelial–mesenchymal transition markers. Besides, PBA-Niclo-SLN selectively accumulated at the tumor site with more significant tumor regression and improved the survivability in TNBC tumor-bearing mice. Conclusion: PBA-Niclo-SLN formulation would be an effective strategy to eradicate TNBC cells (breast cancer stem cells and nonbreast cancer stem cells) efficiently.
Collapse
Affiliation(s)
- Sai Kiran SS Pindiprolu
- Department of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Ooty, The Nilgiris, 643001, Tamil Nadu, India
| | - Praveen T Krishnamurthy
- Department of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Ooty, The Nilgiris, 643001, Tamil Nadu, India
| | - Venkata Rao Ghanta
- Synthetic Organic Chemistry Division, GVK Biosciences Private Limited, IDA Nacharam, Hyderabad, 500076, Telangana, India
| | - Pavan Kumar Chintamaneni
- Department of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Ooty, The Nilgiris, 643001, Tamil Nadu, India
| |
Collapse
|
38
|
Hachim IY, López-Ozuna VM, Hachim MY, Lebrun JJ, Ali S. Prolactin hormone exerts anti-tumorigenic effects in HER-2 overexpressing breast cancer cells through regulation of stemness. Stem Cell Res 2019; 40:101538. [PMID: 31450192 DOI: 10.1016/j.scr.2019.101538] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/08/2019] [Revised: 07/18/2019] [Accepted: 08/15/2019] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Breast cancers characterized by HER2 overexpression, belong to HER-2 enriched or luminal B subtypes, are frequently associated with higher incidence of tumor recurrence and therapeutic failure. These aggressive features have been attributed to the presence of cancer stem-like cell subpopulations known to have high tumor initiation, self -renewal capacities and high metastatic potential. Depleting these stem-like cells in these tumors therefore might help in improving therapeutic response and patient outcome. METHODS Here we used human breast cancer cells representative of HER2- enriched and luminal B subtypes as well as purified ALDH-positive stem-like cell subpopulation for in vitro cell viability, proliferation, tumorshpere formation analyses and gene expression studies. In addition, we used a pre-clinical xenograft HER2 mouse model (NOD/SCID mice) for in vivo tumorigenesis assessment. Furthermore, patient survival outcomes were evaluated using in silico bioinformatics analyses of publicly available datasets. RESULTS Our results indicate that prolactin (PRL) exerts anti-tumorigenic effects in HER-2 positive breast cancer cells. Importantly, PRL caused a significant reduction in ALDHhi stem-like subpopulation, as well as their viability and tumorsphere formation capacity. Molecularly we found PRL to suppress gene expression of markers involved in stemness, tumor initiation, drug resistance and poor patient outcome found to be enriched in the ALDHhi stem-like subpopulation. Furthermore, we show PRL to impede tumor growth of HER-2 xenografts and to suppress expression of Ki67 proliferative marker. Finally, we found PRL pathway gene signature to correlate with favorable patient outcomes in HER-2 and luminal B breast cancer patients. CONCLUSION Together these results emphasize an anti-tumorigenic role with a potential therapeutic value for PRL in HER-2 and luminal B breast cancer subtypes targeting the cancer stem-like cells.
Collapse
Affiliation(s)
- Ibrahim Y Hachim
- Department of Medicine, Cancer Research Program, McGill University Health Centre, Montreal, QC H4A 3J1, Canada.
| | - Vanessa M López-Ozuna
- Department of Medicine, Cancer Research Program, McGill University Health Centre, Montreal, QC H4A 3J1, Canada.
| | - Mahmood Y Hachim
- Sharjah Institute for Medical Research, University of Sharjah, United Arab Emirates.
| | - Jean-Jacques Lebrun
- Department of Medicine, Cancer Research Program, McGill University Health Centre, Montreal, QC H4A 3J1, Canada.
| | - Suhad Ali
- Department of Medicine, Cancer Research Program, McGill University Health Centre, Montreal, QC H4A 3J1, Canada.
| |
Collapse
|
39
|
Scott DA, Drake RR. Glycosylation and its implications in breast cancer. Expert Rev Proteomics 2019; 16:665-680. [PMID: 31314995 PMCID: PMC6702063 DOI: 10.1080/14789450.2019.1645604] [Citation(s) in RCA: 54] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2019] [Accepted: 07/15/2019] [Indexed: 02/07/2023]
Abstract
Introduction: For decades, the role of glycans and glycoproteins in the progression of breast cancer and other cancers have been evaluated. Through extensive studies focused on elucidating the biological functions of glycosylation, researchers have been able to implicate alterations in these functions to tumor formation and metastasis. Areas covered: In this review, we summarize how changes in glycosylation are associated with tumorigenesis, with emphasis on breast cancers. An overview of the changes in N-linked and O-linked glycans associated with breast cancer tumors and biofluids are described. Recent advances in glycomics are emphasized in the context of continuing to decipher the glycosylation changes associated with breast cancer progression. Expert opinion: While changes in glycosylation have been studied in breast cancer for many years, the clinical relevance of these studies has been limited. This reflects the inherent biological and clinical heterogeneity of breast cancers. Glycomics analysis lags behind the advances in genomics and proteomics, but new approaches are emerging. A summary of known glycosylation changes associated with breast cancer is necessary to implement new findings in the context of clinical outcomes and therapeutic strategies. A better understanding of the dynamics of tumor and immune glycosylation is critical to improving emerging immunotherapeutic treatments.
Collapse
Affiliation(s)
- Danielle A Scott
- Department of Cell and Molecular Pharmacology and Experimental Therapeutics and MUSC, Proteomics Center, Medical University of South Carolina , Charleston , SC , USA
| | - Richard R Drake
- Department of Cell and Molecular Pharmacology and Experimental Therapeutics and MUSC, Proteomics Center, Medical University of South Carolina , Charleston , SC , USA
| |
Collapse
|
40
|
Garnham R, Scott E, Livermore KE, Munkley J. ST6GAL1: A key player in cancer. Oncol Lett 2019; 18:983-989. [PMID: 31423157 PMCID: PMC6607188 DOI: 10.3892/ol.2019.10458] [Citation(s) in RCA: 64] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2019] [Accepted: 06/04/2019] [Indexed: 02/06/2023] Open
Abstract
Aberrant glycosylation is a universal feature of cancer cells and there is now overwhelming evidence that glycans can modulate pathways intrinsic to tumour cell biology. Glycans are important in all of the cancer hallmarks and there is a renewed interest in the glycomic profiling of tumours to improve early diagnosis, determine patient prognosis and identify targets for therapeutic intervention. One of the most widely occurring cancer associated changes in glycosylation is abnormal sialylation which is often accompanied by changes in sialyltransferase activity. Several sialyltransferases are implicated in cancer, but in recent years ST6 β-galactoside α-2,6-sialyltransferase 1 (ST6GAL1) has become increasingly dominant in the literature. ST6GAL1 catalyses the addition of α2,6-linked sialic acids to terminal N-glycans and can modify glycoproteins and/or glycolipids. ST6GAL1 is upregulated in numerous types of cancer (including pancreatic, prostate, breast and ovarian cancer) and can promote growth, survival and metastasis. The present review discusses ST6GAL in relation to the hallmarks of cancer, and highlights its key role in multiple mechanisms intrinsic to tumour cell biology.
Collapse
Affiliation(s)
- Rebecca Garnham
- Institute of Genetic Medicine, Newcastle University, International Centre for Life, Newcastle Upon Tyne NE1 3BZ, UK
| | - Emma Scott
- Institute of Genetic Medicine, Newcastle University, International Centre for Life, Newcastle Upon Tyne NE1 3BZ, UK
| | - Karen E Livermore
- Institute of Genetic Medicine, Newcastle University, International Centre for Life, Newcastle Upon Tyne NE1 3BZ, UK
| | - Jennifer Munkley
- Institute of Genetic Medicine, Newcastle University, International Centre for Life, Newcastle Upon Tyne NE1 3BZ, UK
| |
Collapse
|
41
|
Hussain S, Saxena S, Shrivastava S, Mohanty AK, Kumar S, Singh RJ, Kumar A, Wani SA, Gandham RK, Kumar N, Sharma AK, Tiwari AK, Singh RK. Gene expression profiling of spontaneously occurring canine mammary tumours: Insight into gene networks and pathways linked to cancer pathogenesis. PLoS One 2018; 13:e0208656. [PMID: 30517191 PMCID: PMC6281268 DOI: 10.1371/journal.pone.0208656] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2018] [Accepted: 11/20/2018] [Indexed: 11/24/2022] Open
Abstract
Spontaneously occurring canine mammary tumours (CMTs) are the most common neoplasms of unspayed female dogs leading to thrice higher mortality rates than human breast cancer. These are also attractive models for human breast cancer studies owing to clinical and molecular similarities. Thus, they are important candidates for biomarker studies and understanding cancer pathobiology. The study was designed to explore underlying molecular networks and pathways in CMTs for deciphering new prognostic factors and therapeutic targets. To gain an insight into various pathways and networks associated with the development and pathogenesis of CMTs, comparative cDNA microarray expression profiling was performed using CMT tissues and healthy mammary gland tissues. Upon analysis, 1700 and 1287 differentially expressed genes (DEGs, P ≤ 0.05) were identified in malignant and benign tissues, respectively. DEGs identified from microarray analysis were further annotated using the Ingenuity Systems Pathway Analysis (IPA) tool for detection of deregulated canonical pathways, upstream regulators, and networks associated with malignant, as well as, benign disease. Top scoring key networks in benign and malignant mammary tumours were having central nodes of VEGF and BUB1B, respectively. Cyclins & cell cycle regulation and TREM1 signalling were amongst the top activated canonical pathways in CMTs. Other cancer related significant pathways like apoptosis signalling, dendritic cell maturation, DNA recombination and repair, Wnt/β-catenin signalling, etc. were also found to be altered. Furthermore, seven proteins (ANXA2, APOCII, CDK6, GATC, GDI2, GNAQ and MYH9) highly up-regulated in malignant tissues were identified by two-dimensional gel electrophoresis (2DE) and MALDI-TOF PMF studies which were in concordance with microarray data. Thus, the study has uncovered ample number of candidate genes associated with CMTs which need to be further validated as therapeutic targets and prognostic markers.
Collapse
Affiliation(s)
- Shahid Hussain
- Division of Veterinary Biotechnology, ICAR-Indian Veterinary Research Institute [Deemed University], Izatnagar, Bareilly, UP, India
| | - Sonal Saxena
- Division of Veterinary Biotechnology, ICAR-Indian Veterinary Research Institute [Deemed University], Izatnagar, Bareilly, UP, India
- * E-mail: (SON); (SAM); (RKS)
| | - Sameer Shrivastava
- Division of Veterinary Biotechnology, ICAR-Indian Veterinary Research Institute [Deemed University], Izatnagar, Bareilly, UP, India
- * E-mail: (SON); (SAM); (RKS)
| | - Ashok Kumar Mohanty
- Animal Biotechnology Division, ICAR-National Dairy Research Institute [Deemed University], Karnal, Haryana, India
| | - Sudarshan Kumar
- Animal Biotechnology Division, ICAR-National Dairy Research Institute [Deemed University], Karnal, Haryana, India
| | - Rajkumar James Singh
- Division of Veterinary Biotechnology, ICAR-Indian Veterinary Research Institute [Deemed University], Izatnagar, Bareilly, UP, India
| | - Abhinav Kumar
- Department of Computer Science and Engineering, Indian Institute of Technology (IIT) BHU, Varanasi, India
| | | | - Ravi Kumar Gandham
- National Institute of Animal Biotechnology, Miyapur, Hyderabad, Telangana, India
| | - Naveen Kumar
- Division of Veterinary Surgery, ICAR-Indian Veterinary Research Institute [Deemed University], Izatnagar, Bareilly, UP, India
| | - Anil Kumar Sharma
- Division of Veterinary Pathology, ICAR-Indian Veterinary Research Institute [Deemed University], Izatnagar, Bareilly, UP, India
| | - Ashok Kumar Tiwari
- Division of Veterinary Biotechnology, ICAR-Indian Veterinary Research Institute [Deemed University], Izatnagar, Bareilly, UP, India
| | - Raj Kumar Singh
- Division of Veterinary Biotechnology, ICAR-Indian Veterinary Research Institute [Deemed University], Izatnagar, Bareilly, UP, India
- * E-mail: (SON); (SAM); (RKS)
| |
Collapse
|
42
|
Pan Y, Wu Y, Hu J, Shan Y, Ma J, Ma H, Qi X, Jia L. Long noncoding RNA HOTAIR promotes renal cell carcinoma malignancy through alpha-2, 8-sialyltransferase 4 by sponging microRNA-124. Cell Prolif 2018; 51:e12507. [PMID: 30105850 DOI: 10.1111/cpr.12507] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2018] [Accepted: 07/03/2018] [Indexed: 12/17/2022] Open
Abstract
OBJECTIVES Accumulating evidence demonstrated that the long noncoding RNA (lncRNA) HOTAIR (Hox transcript antisense intergenic RNA) plays key role in renal cell carcinoma (RCC) malignancy, while microRNA-124 (miR-124) is a tumour suppressor in RCC. The aim of this work was to assess the biological function of HOTAIR and to explore underlying mechanism involved in HOTAIR/miR-124/alpha-2, 8-sialyltransferase 4 (ST8SIA4) axis-regulated progression in RCC. MATERIALS AND METHODS Real-time PCR analyses and western blots were performed to the levels of HOTAIR, miR-124 and ST8SIA4 expression in human RCC tissues and RCC cell lines (ACHN and 786-O). Bioinformatics analysis and dual-luciferase reporter assay were used to illustrate relationship between HOTAIR and miR-124 in RCC. Colony formation assays, EdU assays, Ki67 assays and apoptosis assays were taken to evaluate cell proliferation. Tumour xenograft was created to explore the functions of HOTAIR and ST8SIA4 in tumorigenesis in vivo. Migration assays, invasion assays and cell adhesion assays and were also taken to analyse the carcinoma progression. RESULTS In this study, HOTAIR level was confirmed to be significantly upregulated in RCC samples and RCC cell lines compared with those in the paired adjacent tissues and normal renal cell line. Overexpression of HOTAIR promoted the capability of proliferation, migration and invasion in RCC cell lines. HOTAIR directly bound to miR-124, while miR-124 mediated the expression of ST8SIA4 in RCC cell lines. ST8SIA4 was upregulated in RCC tissues and RCC cell lines. Ectopic expression of ST8SIA4 modulated the proliferation, migration and invasion of RCC cells. Further results indicated that HOTAIR promoted the proliferation and metastasis as a competing endogenous RNA to regulate ST8SIA4 expression by sponging miR-124 in RCC. CONCLUSIONS Our results demonstrated that HOTAIR mediated RCC progression in part through miR-124/ST8SIA4 axis, which functioned as a new prognostic biomarker in RCC.
Collapse
Affiliation(s)
- Yue Pan
- College of Laboratory Medicine, Dalian Medical University, Dalian, Liaoning Province, China
| | - Yongjin Wu
- Benxi Jinshan Hospital, Benxi, Liaoning Province, China
| | - Jialei Hu
- College of Laboratory Medicine, Dalian Medical University, Dalian, Liaoning Province, China
| | - Yujia Shan
- College of Laboratory Medicine, Dalian Medical University, Dalian, Liaoning Province, China
| | - Jia Ma
- College of Laboratory Medicine, Dalian Medical University, Dalian, Liaoning Province, China
| | - Huipeng Ma
- College of Laboratory Medicine, Dalian Medical University, Dalian, Liaoning Province, China
| | - Xia Qi
- College of Laboratory Medicine, Dalian Medical University, Dalian, Liaoning Province, China
| | - Li Jia
- College of Laboratory Medicine, Dalian Medical University, Dalian, Liaoning Province, China
| |
Collapse
|
43
|
Ding Q, Wang Y, Zuo Z, Gong Y, Krishnamurthy S, Li CW, Lai YJ, Wei W, Wang J, Manyam GC, Diao L, Zhang X, Lin F, Symmans WF, Sun L, Liu CG, Liu X, Debeb BG, Ueno NT, Harano K, Alvarez RH, Wu Y, Cristofanilli M, Huo L. Decreased expression of microRNA-26b in locally advanced and inflammatory breast cancer. Hum Pathol 2018; 77:121-129. [PMID: 29689244 DOI: 10.1016/j.humpath.2018.04.002] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/22/2018] [Revised: 04/04/2018] [Accepted: 04/13/2018] [Indexed: 01/23/2023]
Abstract
Advanced-stage breast cancer patients comprise a smaller proportion of breast cancer patients than do early stage patients and are more likely to experience a poor outcome. Understanding the underlying molecular mechanisms and identifying new biomarkers for treatment in this subgroup of patients is paramount. With the aim of identifying microRNAs that are regulated in advanced-stage breast cancer, we found lower expression of miR-26b, a member of the miR-26 family, in inflammatory breast cancer and noninflammatory locally advanced breast cancer tissue than in normal breast tissue, by quantitative real-time polymerase chain reaction and in situ hybridization. Quantitative real-time polymerase chain reaction (but not in situ hybridization) also revealed lower miR-26b expression in inflammatory breast cancer than in noninflammatory locally advanced breast cancer. Furthermore, lower expression of miR-26b was correlated with shorter distant metastasis-free survival and overall survival in univariate analysis, and with shorter overall survival in multivariate analysis. The expression of miRNA-26b was inversely associated with EZH2 protein expression in several breast cancer cell lines, and overexpression and knockdown of miR-26b caused corresponding changes in EZH2 expression. Our study shows that miR-26b may regulate EZH2 expression in breast cancer and may be useful as a therapeutic target for inflammatory breast cancer and noninflammatory locally advanced breast cancer.
Collapse
Affiliation(s)
- Qingqing Ding
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Yan Wang
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Zhuang Zuo
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Yun Gong
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX; Morgan Welch Inflammatory Breast Cancer Research Program and Clinic, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Savitri Krishnamurthy
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX; Morgan Welch Inflammatory Breast Cancer Research Program and Clinic, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Chia-Wei Li
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Yun-Ju Lai
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Wei Wei
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Jing Wang
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Ganiraju C Manyam
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Lixia Diao
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Xinna Zhang
- Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Feng Lin
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - William F Symmans
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Li Sun
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Chang-Gong Liu
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Xiuping Liu
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Bisrat G Debeb
- Morgan Welch Inflammatory Breast Cancer Research Program and Clinic, The University of Texas MD Anderson Cancer Center, Houston, TX; Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Naoto T Ueno
- Morgan Welch Inflammatory Breast Cancer Research Program and Clinic, The University of Texas MD Anderson Cancer Center, Houston, TX; Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Kenichi Harano
- Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Ricardo H Alvarez
- Morgan Welch Inflammatory Breast Cancer Research Program and Clinic, The University of Texas MD Anderson Cancer Center, Houston, TX; Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Yun Wu
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX
| | | | - Lei Huo
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX; Morgan Welch Inflammatory Breast Cancer Research Program and Clinic, The University of Texas MD Anderson Cancer Center, Houston, TX.
| |
Collapse
|
44
|
Rodrigues E, Macauley MS. Hypersialylation in Cancer: Modulation of Inflammation and Therapeutic Opportunities. Cancers (Basel) 2018; 10:cancers10060207. [PMID: 29912148 PMCID: PMC6025361 DOI: 10.3390/cancers10060207] [Citation(s) in RCA: 139] [Impact Index Per Article: 19.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2018] [Revised: 06/13/2018] [Accepted: 06/14/2018] [Indexed: 02/06/2023] Open
Abstract
Cell surface glycosylation is dynamic and often changes in response to cellular differentiation under physiological or pathophysiological conditions. Altered glycosylation on cancers cells is gaining attention due its wide-spread occurrence across a variety of cancer types and recent studies that have documented functional roles for aberrant glycosylation in driving cancer progression at various stages. One change in glycosylation that can correlate with cancer stage and disease prognosis is hypersialylation. Increased levels of sialic acid are pervasive in cancer and a growing body of evidence demonstrates how hypersialylation is advantageous to cancer cells, particularly from the perspective of modulating immune cell responses. Sialic acid-binding receptors, such as Siglecs and Selectins, are well-positioned to be exploited by cancer hypersialylation. Evidence is also mounting that Siglecs modulate key immune cell types in the tumor microenvironment, particularly those responsible for maintaining the appropriate inflammatory environment. From these studies have come new and innovative ways to block the effects of hypersialylation by directly reducing sialic acid on cancer cells or blocking interactions between sialic acid and Siglecs or Selectins. Here we review recent works examining how cancer cells become hypersialylated, how hypersialylation benefits cancer cells and tumors, and proposed therapies to abrogate hypersialylation of cancer.
Collapse
Affiliation(s)
- Emily Rodrigues
- Department of Chemistry, University of Alberta, Edmonton, AB T6G 2G2, Canada.
| | - Matthew S Macauley
- Department of Chemistry, University of Alberta, Edmonton, AB T6G 2G2, Canada.
- Department of Medical Microbiology and Immunology, University of Alberta, Edmonton, AB T6G 2G2, Canada.
| |
Collapse
|
45
|
Saeui CT, Nairn AV, Galizzi M, Douville C, Gowda P, Park M, Dharmarha V, Shah SR, Clarke A, Austin M, Moremen KW, Yarema KJ. Integration of genetic and metabolic features related to sialic acid metabolism distinguishes human breast cell subtypes. PLoS One 2018; 13:e0195812. [PMID: 29847599 PMCID: PMC5976204 DOI: 10.1371/journal.pone.0195812] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2018] [Accepted: 03/29/2018] [Indexed: 11/18/2022] Open
Abstract
In this report we use 'high-flux' tributanoyl-modified N-acetylmannosamine (ManNAc) analogs with natural N-acetyl as well as non-natural azido- and alkyne N-acyl groups (specifically, 1,3,4-O-Bu3ManNAc, 1,3,4-O-Bu3ManNAz, and 1,3,4-O-Bu3ManNAl respectively) to probe intracellular sialic acid metabolism in the near-normal MCF10A human breast cell line in comparison with earlier stage T-47D and more advanced stage MDA-MB-231 breast cancer lines. An integrated view of sialic acid metabolism was gained by measuring intracellular sialic acid production in tandem with transcriptional profiling of genes linked to sialic acid metabolism. The transcriptional profiling showed several differences between the three lines in the absence of ManNAc analog supplementation that helps explain the different sialoglycan profiles naturally associated with cancer. Only minor changes in mRNA transcript levels occurred upon exposure to the compounds confirming that metabolic flux alone can be a key determinant of sialoglycoconjugate display in breast cancer cells; this result complements the well-established role of genetic control (e.g., the transcription of STs) of sialylation abnormalities ubiquitously associated with cancer. A notable result was that the different cell lines produced significantly different levels of sialic acid upon exogenous ManNAc supplementation, indicating that feedback inhibition of UDP-GlcNAc 2-epimerase/ManNAc kinase (GNE)-generally regarded as the 'gatekeeper' enzyme for titering flux into sialic acid biosynthesis-is not the only regulatory mechanism that limits production of this sugar. A notable aspect of our metabolic glycoengineering approach is its ability to discriminate cell subtype based on intracellular metabolism by illuminating otherwise hidden cell type-specific features. We believe that this strategy combined with multi-dimensional analysis of sialic acid metabolism will ultimately provide novel insights into breast cancer subtypes and provide a foundation for new methods of diagnosis.
Collapse
Affiliation(s)
- Christopher T. Saeui
- Department of Biomedical Engineering and the Translational Tissue Engineering Center, The Johns Hopkins University, Baltimore, Maryland, United States of America
| | - Alison V. Nairn
- Complex Carbohydrate Research Center, University of Georgia, Athens, Georgia, United States of America
| | - Melina Galizzi
- Complex Carbohydrate Research Center, University of Georgia, Athens, Georgia, United States of America
| | - Christopher Douville
- Department of Biomedical Engineering and the Translational Tissue Engineering Center, The Johns Hopkins University, Baltimore, Maryland, United States of America
| | - Prateek Gowda
- Department of Biomedical Engineering and the Translational Tissue Engineering Center, The Johns Hopkins University, Baltimore, Maryland, United States of America
| | - Marian Park
- Department of Biomedical Engineering and the Translational Tissue Engineering Center, The Johns Hopkins University, Baltimore, Maryland, United States of America
| | - Vrinda Dharmarha
- Department of Biomedical Engineering and the Translational Tissue Engineering Center, The Johns Hopkins University, Baltimore, Maryland, United States of America
| | - Sagar R. Shah
- Department of Biomedical Engineering and the Translational Tissue Engineering Center, The Johns Hopkins University, Baltimore, Maryland, United States of America
| | - Amelia Clarke
- Department of Biomedical Engineering and the Translational Tissue Engineering Center, The Johns Hopkins University, Baltimore, Maryland, United States of America
| | - Melissa Austin
- Department of Biomedical Engineering and the Translational Tissue Engineering Center, The Johns Hopkins University, Baltimore, Maryland, United States of America
| | - Kelley W. Moremen
- Complex Carbohydrate Research Center, University of Georgia, Athens, Georgia, United States of America
- Department of Biochemistry and Molecular Biology, University of Georgia, Athens, Georgia, United States of America
| | - Kevin J. Yarema
- Department of Biomedical Engineering and the Translational Tissue Engineering Center, The Johns Hopkins University, Baltimore, Maryland, United States of America
| |
Collapse
|
46
|
Teoh ST, Ogrodzinski MP, Ross C, Hunter KW, Lunt SY. Sialic Acid Metabolism: A Key Player in Breast Cancer Metastasis Revealed by Metabolomics. Front Oncol 2018; 8:174. [PMID: 29892572 PMCID: PMC5985449 DOI: 10.3389/fonc.2018.00174] [Citation(s) in RCA: 60] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2018] [Accepted: 05/04/2018] [Indexed: 11/13/2022] Open
Abstract
Metastatic breast cancer is currently incurable. It has recently emerged that different metabolic pathways support metastatic breast cancer. To further uncover metabolic pathways enabling breast cancer metastasis, we investigated metabolic differences in mouse tumors of differing metastatic propensities using mass spectrometry-based metabolomics. We found that sialic acid metabolism is upregulated in highly metastatic breast tumors. Knocking out a key gene in sialic acid metabolism, Cmas, inhibits synthesis of the activated form of sialic acid, cytidine monophosphate-sialic acid and decreases the formation of lung metastases in vivo. Thus, the sialic acid pathway may be a new target against metastatic breast cancer.
Collapse
Affiliation(s)
- Shao Thing Teoh
- Department of Biochemistry and Molecular Biology, Michigan State University, East Lansing, MI, United States
| | - Martin P Ogrodzinski
- Department of Biochemistry and Molecular Biology, Michigan State University, East Lansing, MI, United States.,Department of Physiology, Michigan State University, East Lansing, MI, United States
| | - Christina Ross
- Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| | - Kent W Hunter
- Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| | - Sophia Y Lunt
- Department of Biochemistry and Molecular Biology, Michigan State University, East Lansing, MI, United States.,Department of Chemical Engineering and Materials Science, Michigan State University, East Lansing, MI, United States
| |
Collapse
|
47
|
Kong X, Zhang J, Li J, Shao J, Fang L. MiR-130a-3p inhibits migration and invasion by regulating RAB5B in human breast cancer stem cell-like cells. Biochem Biophys Res Commun 2018; 501:486-493. [PMID: 29746865 DOI: 10.1016/j.bbrc.2018.05.018] [Citation(s) in RCA: 81] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2018] [Accepted: 05/03/2018] [Indexed: 02/07/2023]
Abstract
Breast cancer stem cells (BCSCs) constitute a subpopulation of tumor cells that express stem cell-associated markers and have a high capacity for tumor generation in vivo. MicroRNAs (miRNAs) are involved in tumorigenesis by regulating specific oncogenes and tumor suppressor genes, and their roles in BCSCs are becoming more apparent. We try to reveal the mechanism by which specific miRNA plays its function in BCSCs. Herein, we show that miR-130a-3p is down-regulated in human breast cancer tissues and exosomes from circulating blood. Overexpression of miR-130a-3p in BCSCs inhibited cellular proliferation, migration, and invasion, and silencing of miR-130a-3p had the opposite effects. We also confirmed that RAB5B is directly down-regulated by miR-130a-3p. Knockdown of RAB5B also inhibited cell proliferation, migration and invasion. Furthermore, we found that lower levels of exosome-derived miR-130a-3p are associated with lymph node metastasis and advanced TNM stage. Taken together, our results demonstrate that miR-130a-3p may act as a disease progression monitoring indicator and therapeutic target in breast cancer.
Collapse
Affiliation(s)
- Xiangjie Kong
- Department of Urology, Wuxi People's Hospital, Nanjing Medical University, Wuxi, Jiangsu, China
| | - Junfeng Zhang
- Department of Breast and Thyroid, Shanghai Tenth People's Hospital, Tongji University, Shanghai, China
| | - Jia Li
- Department of Breast and Thyroid, Shanghai Tenth People's Hospital, Tongji University, Shanghai, China
| | - Jianfeng Shao
- Department of Urology, Wuxi People's Hospital, Nanjing Medical University, Wuxi, Jiangsu, China.
| | - Lin Fang
- Department of Breast and Thyroid, Shanghai Tenth People's Hospital, Tongji University, Shanghai, China.
| |
Collapse
|
48
|
Garbar C, Mascaux C, Merrouche Y, Bensussan A. Triple-negative and HER2-overexpressing breast cancer cell sialylation impacts tumor microenvironment T-lymphocyte subset recruitment: a possible mechanism of tumor escape. Cancer Manag Res 2018; 10:1051-1059. [PMID: 29765252 PMCID: PMC5942397 DOI: 10.2147/cmar.s162932] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
Introduction Breast cancers develop different patterns of sialylation to modulate their tumor-infiltrating lymphocyte (TIL) environment. We studied the relationship between α-2,6 sialyltransferases and the TIL in different breast cancer molecular subgroups. Materials and methods Immunohistochemical preparations were made from 39 luminal (LUM), 13 human epidermal growth factor receptor 2-overexpressing (HER2) and 47 triple-negative (TN) breast carcinomas. Targeted proteins included ST6Gal-I, ST6Gal-II, ST6GalNac-I, CD8, CD4 and granzyme-B in both cytotoxic T lymphocytes and NK lymphocytes (CTL/NK). Results CTL/NK populations were significantly more frequent in TN than LUM (P <0.001). TN showed a lower level of ST6Gal-I expression than LUM or HER2 (both P > 0.001). ST6GalNac-I expression was lower in LUM than in TN or HER2 (P = 0.002 and P = 0.02, respectively). In HER2, a significant association was found between a low level of ST6Gal-I expression and a high TIL level. In TN, a significant association was observed between a high level of ST6Gal-II expression and a high TIL level. Conclusion An increase in infiltrating lymphocytes could be influenced by low expression of ST6Gal-I in HER2 and by high expression of ST6Gal-II in TN breast cancers. Thus, targeting these sialylation pathways could modulate the levels of TIL.
Collapse
Affiliation(s)
- Christian Garbar
- Biopathology Department, Institut Jean Godinot - Unicancer, Reims, France.,DERM-I-C EA7319, Université de Reims Champagne - Ardenne, Reims, France
| | - Corinne Mascaux
- Biopathology Department, Institut Jean Godinot - Unicancer, Reims, France.,DERM-I-C EA7319, Université de Reims Champagne - Ardenne, Reims, France
| | - Yacine Merrouche
- Biopathology Department, Institut Jean Godinot - Unicancer, Reims, France.,DERM-I-C EA7319, Université de Reims Champagne - Ardenne, Reims, France
| | - Armand Bensussan
- INSERM U976; Université Paris Diderot, Sorbonne Paris Cité, Laboratory of Immunology, Dermatology & Oncology, Paris, France
| |
Collapse
|
49
|
Liu J, Mi B, Wang Y, Shi C, Mi X, Lu Y, Yu P. miR-26a suppresses osteosarcoma migration and invasion by directly targeting HMGA1. Oncol Lett 2018; 15:8303-8310. [PMID: 29928320 PMCID: PMC6004719 DOI: 10.3892/ol.2018.8359] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2017] [Accepted: 02/28/2018] [Indexed: 12/17/2022] Open
Abstract
Osteosarcoma (OS) is identified as the most commonly diagnosed malignant cancer of bone, and has approximately three million new cases annually. miR-26a plays an important role in the development of various types of cancer. We investigated whether miR-26a can regulate the migration and invasion of OS by targeting high-mobility group A1 HMGA1. Western blot analysis was used to identify the changes of protein levels. Reverse transcription-quantitative PCR was used to test expression levels of genes and miR-26a. Luciferase reporter assay was used to test the specific target gene of miR-26a. Transwell assay was employed to determine the migration and invasion of OS cell lines. In the present study, miRNA-26a was frequently downregulated in OS tissues and cells. Overexpression of miR-26a inhibited cell migration and invasion in vitro. In addition, miR-26a downregulated HMGA1 by targeting its 3′-UTR and knockdown of HMGA1 significantly suppressed the migration and invasion of two osteosarcoma cell lines in vitro. miR-26a suppressed the migration and invasion of OS cells by targeting HMGA1, suggesting that miR-26a/HMGA1 axis provides a new prospective therapeutic strategy for OS.
Collapse
Affiliation(s)
- Jianyong Liu
- Department of Orthopedics, People's Hospital of Weifang, Weifang, Shandong 261000, P.R. China
| | - Bo Mi
- Department of Medicine, The First People's Hospital of Jinan, Jinan, Shandong 250000, P.R. China
| | - Yi Wang
- Department of Respiratory Medicine, The People's Hospital of Zhangqiu Area, Jinan, Shandong 250200, P.R. China
| | - Chunling Shi
- Department of Neurology, People's Hospital of Rizhao, Rizhao, Shandong 276800, P.R. China
| | - Xiufang Mi
- Department of Medicine, The People's Hospital of Zhangqiu Area, Jinan, Shandong 250200, P.R. China
| | - Yingying Lu
- Department of Clinical Laboratory, People's Hospital of Rizhao, Rizhao, Shandong 276800, P.R. China
| | - Peilin Yu
- Department of Orthopedics, Qingdao Hiser Medical Center, Qingdao, Shandong 266033, P.R. China
| |
Collapse
|
50
|
Zhou J, Zhang WW, Peng F, Sun JY, He ZY, Wu SG. Downregulation of hsa_circ_0011946 suppresses the migration and invasion of the breast cancer cell line MCF-7 by targeting RFC3. Cancer Manag Res 2018; 10:535-544. [PMID: 29593432 PMCID: PMC5865555 DOI: 10.2147/cmar.s155923] [Citation(s) in RCA: 67] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
Introduction Although some circRNAs have been found to regulate the progression of malignancies, their functions and coupled molecular mechanisms are still unclear. In our study, we sought to assess the underlying molecular mechanisms of circRNAs in breast cancer and therefore explored the differentially expressed circRNAs and co-expression networks, followed by in vitro experiments. Materials and methods High-throughput RNA sequencing was performed to obtain an unbiased profile of circRNA expression. CircRNA-miRNA-mRNA co-expression networks were predicted, and sequence analyses were carried out. The MTT, transwell migration and invasion assay was conducted in Michigan Cancer Foundation-7 cells that had been transfected with si-circRNA and si-negative control (si-NC). Results A total of 152 circRNAs were differentially expressed in breast cancer tissues, among which 85 were upregulated and 67 downregulated. Out of these, hsa_circ_0011946 was selected and the subsequent bioinformatics analysis predicted that hsa_circ_0011946 sponging miR-26a/b directly targeted replication factor C subunit 3 (RFC3) and that its knockdown could inhibit RFC3 mRNA and protein expression. Furthermore, hsa_circ_0011946 loss-of-function significantly suppressed the migration and invasion of Michigan Cancer Foundation-7 cells. Conclusion Together, these results indicate that hsa_circ_0011946 and RFC3 comprise a novel pathway involved in the progression of breast cancer.
Collapse
Affiliation(s)
- Juan Zhou
- Department of Obstetrics and Gynecology, the First Affiliated Hospital of Xiamen University, Xiamen 361003, People's Republic of China
| | - Wen-Wen Zhang
- Department of Radiation Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center of Cancer Medicine, Guangzhou 510060, People's Republic of China
| | - Fang Peng
- Department of Radiation Oncology, the First Affiliated Hospital of Sun Yat-sen University, Guangzhou 510080, People's Republic of China
| | - Jia-Yuan Sun
- Department of Radiation Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center of Cancer Medicine, Guangzhou 510060, People's Republic of China
| | - Zhen-Yu He
- Department of Radiation Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center of Cancer Medicine, Guangzhou 510060, People's Republic of China
| | - San-Gang Wu
- Department of Radiation Oncology, Xiamen Cancer Hospital, the First Affiliated Hospital of Xiamen University, Xiamen 361003, People's Republic of China
| |
Collapse
|