Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 71
Filter
1.
Nat Biotechnol ; 42(4): 572-573, 2024 Apr.
Article in English | MEDLINE | ID: mdl-37813982
2.
Dev Cell ; 58(17): 1548-1561.e10, 2023 09 11.
Article in English | MEDLINE | ID: mdl-37442140

ABSTRACT

Tumor-associated macrophages (TAMs) are a heterogeneous population of cells that facilitate cancer progression. However, our knowledge of the niches of individual TAM subsets and their development and function remain incomplete. Here, we describe a population of lymphatic vessel endothelial hyaluronan receptor-1 (LYVE-1)-expressing TAMs, which form coordinated multi-cellular "nest" structures that are heterogeneously distributed proximal to vasculature in tumors of a spontaneous murine model of breast cancer. We demonstrate that LYVE-1+ TAMs develop in response to IL-6, which induces their expression of the immune-suppressive enzyme heme oxygenase-1 and promotes a CCR5-dependent signaling axis, which guides their nest formation. Blocking the development of LYVE-1+ TAMs or their nest structures, using gene-targeted mice, results in an increase in CD8+ T cell recruitment to the tumor and enhanced response to chemotherapy. This study highlights an unappreciated collaboration of a TAM subset to form a coordinated niche linked to immune exclusion and resistance to anti-cancer therapy.


Subject(s)
Neoplasms , Mice , Animals , Neoplasms/pathology , Macrophages/metabolism
3.
Cells ; 11(7)2022 03 25.
Article in English | MEDLINE | ID: mdl-35406676

ABSTRACT

Epigenetic regulation of gene expression is crucial to the determination of cell fate in development and differentiation, and the Polycomb (PcG) and Trithorax (TrxG) groups of proteins, acting antagonistically as complexes, play a major role in this regulation. Although originally identified in Drosophila, these complexes are conserved in evolution and the components are well defined in mammals. Each complex contains a protein with methylase activity (KMT), which can add methyl groups to a specific lysine in histone tails, histone 3 lysine 27 (H3K27), by PcG complexes, and H3K4 and H3K36 by TrxG complexes, creating transcriptionally repressive or active marks, respectively. Histone demethylases (KDMs), identified later, added a new dimension to histone methylation, and mutations or changes in levels of expression are seen in both methylases and demethylases and in components of the PcG and TrX complexes across a range of cancers. In this review, we focus on both methylases and demethylases governing the methylation state of the suppressive and active marks and consider their action and interaction in normal tissues and in cancer. A picture is emerging which indicates that the changes which occur in cancer during methylation of histone lysines can lead to repression of genes, including tumour suppressor genes, or to the activation of oncogenes. Methylases or demethylases, which are themselves tumour suppressors, are highly mutated. Novel targets for cancer therapy have been identified and a methylase (KMT6A/EZH2), which produces the repressive H3K27me3 mark, and a demethylase (KDM1A/LSD1), which demethylates the active H3K4me2 mark, are now under clinical evaluation.


Subject(s)
Histone Demethylases , Histone Methyltransferases , Histones , Neoplasms , Animals , Epigenesis, Genetic , Histone Demethylases/metabolism , Histone Methyltransferases/metabolism , Histones/metabolism , Humans , Lysine/metabolism , Neoplasms/genetics , Polycomb-Group Proteins/metabolism
4.
Int J Oncol ; 59(5)2021 11.
Article in English | MEDLINE | ID: mdl-34713299

ABSTRACT

Post­translational modification of histones serve a crucial role in the control of gene transcription. Trimethylation of lysine 4 on histone 3 is associated with transcription activation. There are currently six known methylases and six known demethylases that can control the methylation status of this site. Lysine demethylase 5B (KDM5B) is one such demethylase, which can repress gene expression. In particular KDM5B has been found to be overexpressed in a number of cancer types, and small­molecular weight inhibitors of its demethylase activity have been identified. Previous characterisation of Kdm5b knock­out mice has revealed that this genotype leads to either embryonic or neonatal lethality. However, the ΔA­T rich interaction domain (ΔARID)­KDM5B strain of mice, which have the ARID domain and five amino acids within the Jumonji (Jmj)N domain spliced out from KDM5B, remain viable and fertile. In the present study, ΔARID­KDM5B was found to have no demethylase activity as determined by in vitro demethylase assays and by immunofluorescence in transfected Cos­1 cells. Furthermore, molecular dynamic simulations revealed conformational changes within the ΔARID­KDM5B structure compared with that in WT­KDM5B, particularly in the JmjC domain, which is responsible for the catalytic activity of WT­KDM5B. This supports the experimental data that shows the loss of demethylase activity. Since Kdm5b knock­out mice show varying degrees of lethality, these data suggest that KDM5B serves a crucial function in development in a manner that is independent of its demethylase activity.


Subject(s)
DNA-Binding Proteins/metabolism , Epigenesis, Genetic , Jumonji Domain-Containing Histone Demethylases/metabolism , Protein Domains/genetics , Animals , DNA Demethylation , DNA-Binding Proteins/genetics , DNA-Binding Proteins/ultrastructure , Enzyme Assays , Female , Fertility/genetics , Gene Expression Regulation, Developmental , Jumonji Domain-Containing Histone Demethylases/genetics , Jumonji Domain-Containing Histone Demethylases/ultrastructure , Male , Mice , Mice, Transgenic , Molecular Dynamics Simulation , Protein Processing, Post-Translational/genetics
5.
Cells ; 10(7)2021 07 01.
Article in English | MEDLINE | ID: mdl-34359823

ABSTRACT

Pancreatic ductal adenocarcinoma (PDAC) is associated with poor prognosis. This is attributed to the disease already being advanced at presentation and having a particularly aggressive tumor biology. The PDAC tumor microenvironment (TME) is characterized by a dense desmoplastic stroma, dominated by cancer-associated fibroblasts (CAF), extracellular matrix (ECM) and immune cells displaying immunosuppressive phenotypes. Due to the advanced stage at diagnosis, the depletion of immune effector cells and lack of actionable genomic targets, the standard treatment is still apoptosis-inducing regimens such as chemotherapy. Paradoxically, it has emerged that the direct induction of apoptosis of cancer cells may fuel oncogenic processes in the TME, including education of CAF and immune cells towards pro-tumorigenic phenotypes. The direct effect of cytotoxic therapies on CAF may also enhance tumorigenesis. With the awareness that CAF are the predominant cell type in PDAC driving tumorigenesis with various tumor supportive functions, efforts have been made to try to target them. However, efforts to target CAF have, to date, shown disappointing results in clinical trials. With the help of sophisticated single cell analyses it is now appreciated that CAF in PDAC are a heterogenous population with both tumor supportive and tumor suppressive functions. Hence, there remains a debate whether targeting CAF in PDAC is a valid therapeutic strategy. In this review we discuss how cytotoxic therapies and the induction of apoptosis in PDAC fuels oncogenesis by the education of surrounding stromal cells, with a particular focus on the potential pro-tumorigenic outcomes arising from targeting CAF. In addition, we explore therapeutic avenues to potentially avoid the oncogenic effects of apoptosis in PDAC CAF.


Subject(s)
Apoptosis , Cancer-Associated Fibroblasts/pathology , Pancreatic Neoplasms/pathology , Tumor Microenvironment , Carcinogenesis/pathology , Humans , Models, Biological
6.
Glycobiology ; 31(3): 200-210, 2021 04 01.
Article in English | MEDLINE | ID: mdl-32776095

ABSTRACT

Aberrant mucin-type O-linked glycosylation is a common occurrence in cancer where the upregulation of sialyltransferases is often seen leading to the early termination of O-glycan chains. Mucin-type O-linked glycosylation is not limited to mucins and occurs on many cell surface glycoproteins including EGFR, where the number of sites can be limited. Upon EGF ligation, EGFR induces a signaling cascade and may also translocate to the nucleus where it directly regulates gene transcription, a process modulated by Galectin-3 and MUC1 in some cancers. Here, we show that upon EGF binding, breast cancer cells carrying different O-glycans respond by transcribing different gene expression signatures. MMP10, the principal gene upregulated when cells carrying sialylated core 1 glycans were stimulated with EGF, is also upregulated in ER-positive breast carcinoma reported to express high levels of ST3Gal1 and hence mainly core 1 sialylated O-glycans. In contrast, isogenic cells engineered to carry core 2 glycans upregulate CX3CL1 and FGFBP1 and these genes are upregulated in ER-negative breast carcinomas, also known to express longer core 2 O-glycans. Changes in O-glycosylation did not significantly alter signal transduction downstream of EGFR in core 1 or core 2 O-glycan expressing cells. However, striking changes were observed in the formation of an EGFR/galectin-3/MUC1/ß-catenin complex at the cell surface that is present in cells carrying short core 1-based O-glycans but absent in core 2 carrying cells.


Subject(s)
Breast Neoplasms/metabolism , Mucin-1/metabolism , Breast Neoplasms/pathology , ErbB Receptors/metabolism , Female , Glycosylation , Humans , Receptors, Estrogen/metabolism
7.
Commun Biol ; 3(1): 644, 2020 11 04.
Article in English | MEDLINE | ID: mdl-33149188

ABSTRACT

The tumour microenvironment plays a crucial role in the growth and progression of cancer, and the presence of tumour-associated macrophages (TAMs) is associated with poor prognosis. Recent studies have demonstrated that TAMs display transcriptomic, phenotypic, functional and geographical diversity. Here we show that a sialylated tumour-associated glycoform of the mucin MUC1, MUC1-ST, through the engagement of Siglec-9 can specifically and independently induce the differentiation of monocytes into TAMs with a unique phenotype that to the best of our knowledge has not previously been described. These TAMs can recruit and prolong the lifespan of neutrophils, inhibit the function of T cells, degrade basement membrane allowing for invasion, are inefficient at phagocytosis, and can induce plasma clotting. This macrophage phenotype is enriched in the stroma at the edge of breast cancer nests and their presence is associated with poor prognosis in breast cancer patients.


Subject(s)
Macrophages/physiology , Monocytes/physiology , Mucin-1/metabolism , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Cell Differentiation , Cell Line, Tumor , Female , Gene Expression Regulation, Neoplastic , Humans , Mucin-1/genetics
8.
Clin Transl Immunology ; 9(3): e01120, 2020.
Article in English | MEDLINE | ID: mdl-32194962

ABSTRACT

There is growing recognition that mucus and mucin biology have a considerable impact on respiratory health, and subsequent global morbidity and mortality. Mucins play a critical role in chronic lung disease, not only by providing a physical barrier and clearing pathogens, but also in immune homeostasis. The aim of this review is to familiarise the reader with the role of mucins in both lung health and disease, with particular focus on function in immunity, infection and inflammation. We will also discuss their receptors, termed glycan-binding proteins, and how they provide an attractive prospect for therapeutic intervention.

9.
Biochem Soc Trans ; 46(4): 779-788, 2018 08 20.
Article in English | MEDLINE | ID: mdl-29903935

ABSTRACT

Changes in mucin-type O-linked glycosylation are seen in over 90% of breast cancers where increased sialylation is often observed and a change from branched glycans to linear glycans is often seen. There are many mechanisms involved including increased/altered expression of glycosyltransferases and relocalisation to the endoplasmic reticulum of the enzymes responsible for the addition of the first sugar, N-acetyl-d-galactosamine. It is now becoming clear that these changes can contribute to tumour growth and progression by modulating the micro-environment through glycan-sensing lectins expressed on immune cells, by modulating interactions with tumour surface receptors and by binding to selectins. The understanding of how changes in mucin-type O-linked glycosylation influence tumour growth and progression reveals new potential targets for therapeutic intervention in the treatment of breast cancer.


Subject(s)
Breast Neoplasms/metabolism , Mucin-1/metabolism , Acetylgalactosamine/metabolism , Biological Transport , Breast Neoplasms/immunology , Breast Neoplasms/pathology , Disease Progression , Female , Gene Expression Regulation, Enzymologic , Glycoside Hydrolases/genetics , Glycoside Hydrolases/metabolism , Glycosylation , Glycosyltransferases/genetics , Glycosyltransferases/metabolism , Golgi Apparatus/enzymology , Golgi Apparatus/metabolism , Humans , Hydrogen-Ion Concentration , Neoplasm Metastasis , Tumor Microenvironment
10.
Biochem Soc Trans ; 46(3): 659-668, 2018 06 19.
Article in English | MEDLINE | ID: mdl-29784646

ABSTRACT

Currently, there is renewed interest in attempting to recruit the host immune system to eliminate cancers, and within this renewed activity, MUC1 continues to arouse interest. MUC1 has been considered a possible therapeutic target for the past 30 years as it is up-regulated, aberrantly glycosylated and its polarization is lost in many adenocarcinomas. Moreover, MUC1 is expressed by some haematopoietic cancers, including acute myeloid leukaemia and myeloma. Although multiple clinical trials have been initiated and immune responses have been documented, effective clinical benefit worthy of approval for general application has not as yet been achieved. However, this does not appear to have quelled the interest in MUC1 as a therapeutic target, as shown by the increase in the number of MUC1-based clinical trials initiated in 2017 ( Figure 1). As with all translational studies, incorporating new relevant research findings into therapeutic strategy is difficult. Decisions are made to commit to a specific strategy based on the information and data available when the trial is initiated. However, the time required for preclinical studies and early trials can render the founding concept not always appropriate for proceeding to a larger definitive trial. Here, we summarize the attempts made, to date, to bring MUC1 into the world of cancer immunotherapy and discuss how research findings regarding MUC1 structure and function together with expanded knowledge of its interactions with the tumour environment and immune effector cells could lead to improved therapeutic approaches. ppbiost;46/3/659/BST20170400CF1F1BST-2017-0400CF1Figure 1.Number of MUC1-targeted trials initiated each year.


Subject(s)
Immunotherapy , Mucin-1/immunology , Neoplasms/therapy , Antimetabolites, Antineoplastic/therapeutic use , Clinical Trials as Topic , Combined Modality Therapy , Deoxycytidine/analogs & derivatives , Deoxycytidine/therapeutic use , Glycosylation , Humans , Mucin-1/chemistry , Mucin-1/physiology , Neoplasms/drug therapy , Tumor Microenvironment , Gemcitabine
11.
Glycobiology ; 28(7): 437-442, 2018 07 01.
Article in English | MEDLINE | ID: mdl-29618060

ABSTRACT

The mucin-type O-glycome in cancer aberrantly expresses the truncated glycans Tn (GalNAcα1-Ser/Thr) and STn (Neu5Acα2,6GalNAcα1-Ser/Thr). However, the role of Tn and STn in cancer and other diseases is not well understood. Our recent discovery of the self-binding properties (carbohydrate-carbohydrate interactions, CCIs) of Tn (Tn-Tn) and STn (STn-STn) provides a model for their possible roles in cellular transformation. We also review evidence that Tn and STn are members of a larger family of glycan tumor antigens that possess CCIs, which may participate in oncogenesis.


Subject(s)
Antigens, Neoplasm/metabolism , Carcinogenesis , Polysaccharides/metabolism , Animals , Antigens, Neoplasm/chemistry , Humans , Polysaccharides/chemistry
13.
Clin Cancer Res ; 24(7): 1617-1628, 2018 04 01.
Article in English | MEDLINE | ID: mdl-29339440

ABSTRACT

Purpose: Unprecedented clinical outcomes have been achieved in a variety of cancers by targeting immune checkpoint molecules. This preclinical study investigates heme oxygenase-1 (HO-1), an immunosuppressive enzyme that is expressed in a wide variety of cancers, as a potential immune checkpoint target in the context of a chemotherapy-elicited antitumor immune response. We evaluate repurposing tin mesoporphyrin (SnMP), which has demonstrated safety and efficacy targeting hepatic HO in the clinic for the treatment of hyperbilirubinemia, as an immune checkpoint blockade therapy for the treatment of cancer.Experimental Design: SnMP and genetic inactivation of myeloid HO-1 were evaluated alongside 5-fluorouracil in an aggressive spontaneous murine model of breast cancer (MMTV-PyMT). Single-cell RNA sequencing analysis, tumor microarray, and clinical survival data from breast cancer patients were used to support the clinical relevance of our observations.Results: We demonstrate that SnMP inhibits immune suppression of chemotherapy-elicited CD8+ T cells by targeting myeloid HO-1 activity in the tumor microenvironment. Microarray and survival data from breast cancer patients reveal that HO-1 is a poor prognostic factor in patients receiving chemotherapy. Single-cell RNA-sequencing analysis suggests that the myeloid lineage is a significant source of HO-1 expression, and is co-expressed with the immune checkpoints PD-L1/2 in human breast tumors. In vivo, we therapeutically compare the efficacy of targeting these two pathways alongside immune-stimulating chemotherapy, and demonstrate that the efficacy of SnMP compares favorably with PD-1 blockade in preclinical models.Conclusions: SnMP could represent a novel immune checkpoint therapy, which may improve the immunological response to chemotherapy. Clin Cancer Res; 24(7); 1617-28. ©2018 AACR.


Subject(s)
Breast Neoplasms/drug therapy , Breast Neoplasms/immunology , Metalloporphyrins/pharmacology , Animals , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Breast Neoplasms/metabolism , CD8-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Disease Models, Animal , Female , Fluorouracil/pharmacology , Heme Oxygenase-1/metabolism , Humans , Mice , Mice, Inbred C57BL , Mice, Transgenic , Tumor Microenvironment/drug effects , Tumor Microenvironment/immunology
14.
PLoS One ; 12(4): e0175323, 2017.
Article in English | MEDLINE | ID: mdl-28414807

ABSTRACT

Carbohydrate-protein interactions govern many crucial processes in biological systems including cell recognition events. We have used the sensitive force probe optical tweezers to quantify the interactions occurring between MGL lectins and MUC1 carrying the cancer-associated glycan antigens mucins Tn and STn. Unbinding forces of 7.6 pN and 7.1 pN were determined for the MUC1(Tn)-MGL and MUC1(STn)-MGL interactions, at a force loading rate of ~40 pN/s. The interaction strength increased with increasing force loading rate, to 27 and 37 pN at a force loading rate of ~ 310 pN/s. No interactions were detected between MGL and MUC1(ST), a glycoform of MUC1 also expressed by breast carcinoma cells. Interestingly, this glycan (ST) can be found on proteins expressed by normal cells, although in this case not on MUC1. Additionally, GalNAc decorated polyethylene glycol displayed similar rupture forces as observed for MUC1(Tn) and MUC1(STn) when forced to unbind from MGL, indicating that GalNAc is an essential group in these interactions. Since the STn glycan decoration is more frequently found on the surface of carcinomas than the Tn glycan, the binding of MUC1 carrying STn to MGL may be more physiologically relevant and may be in part responsible for some of the characteristics of STn expressing tumours.


Subject(s)
Breast Neoplasms/metabolism , Lectins, C-Type/metabolism , Mucin-1/metabolism , Animals , Antigens, Tumor-Associated, Carbohydrate/metabolism , CHO Cells , Carbohydrates , Cell Line , Cricetulus , Female , Humans , Optical Tweezers , Polysaccharides/metabolism
16.
Immunity ; 45(5): 945-946, 2016 11 15.
Article in English | MEDLINE | ID: mdl-27851917

ABSTRACT

We read with interest the manuscript by June and colleagues published recently in Immunity in which they describe targeting of aberrantly glycosylated tumor-associated cell membrane mucin MUC1 using chimeric antigen receptor-engineered human T cells (Posey et al., 2016). In that study, the authors used a second generation 4-1BB costimulatory-molecule-based chimeric antigen receptor (CAR) (Imai et al., 2004) in which targeting was achieved using a single-chain variable fragment (scFv) derived from the 5E5 antibody. This CAR selectively binds MUC1 that carries the Tn or sialyl (S)Tn glycan. Both of these truncated glycans are aberrantly expressed on the MUC1 glycoprotein in a spectrum of malignancies and consequently represent attractive targets for immunotherapeutic exploitation.


Subject(s)
Mucin-1/immunology , T-Lymphocytes/immunology , Antigens, Neoplasm , Glycosylation , Humans , Neoplasms/immunology
17.
Nat Immunol ; 17(11): 1273-1281, 2016 Nov.
Article in English | MEDLINE | ID: mdl-27595232

ABSTRACT

Siglec-9 is a sialic-acid-binding lectin expressed predominantly on myeloid cells. Aberrant glycosylation occurs in essentially all types of cancers and results in increased sialylation. Thus, when the mucin MUC1 is expressed on cancer cells, it is decorated by multiple short, sialylated O-linked glycans (MUC1-ST). Here we found that this cancer-specific MUC1 glycoform, through engagement of Siglec-9, 'educated' myeloid cells to release factors associated with determination of the tumor microenvironment and disease progression. Moreover, MUC1-ST induced macrophages to display a tumor-associated macrophage (TAM)-like phenotype, with increased expression of the checkpoint ligand PD-L1. Binding of MUC1-ST to Siglec-9 did not activate the phosphatases SHP-1 or SHP-2 but, unexpectedly, induced calcium flux that led to activation of the kinases MEK-ERK. This work defines a critical role for aberrantly glycosylated MUC1 and identifies an activating pathway that follows engagement of Siglec-9.


Subject(s)
Antigens, CD/metabolism , Mucin-1/metabolism , Neoplasms/immunology , Neoplasms/metabolism , Sialic Acid Binding Immunoglobulin-like Lectins/metabolism , Tumor Microenvironment/immunology , Antigens, CD/genetics , Biomarkers , Cell Differentiation , Cell Line , Dendritic Cells/immunology , Dendritic Cells/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , Gene Expression , Glycosylation , Humans , Macrophages/immunology , Macrophages/metabolism , Mitogen-Activated Protein Kinases/metabolism , Myeloid Cells/cytology , Myeloid Cells/immunology , Myeloid Cells/metabolism , Neoplasms/genetics , Neoplasms/pathology , Phenotype , Protein Binding , Sialic Acid Binding Immunoglobulin-like Lectins/genetics
18.
Glycobiology ; 26(12): 1338-1350, 2016 12.
Article in English | MEDLINE | ID: mdl-27282157

ABSTRACT

The molecular mechanism(s) underlying the enhanced self-interactions of mucins possessing the Tn (GalNAcα1-Ser/Thr) or STn (NeuNAcα2-6GalNAcα1-Ser/Thr) cancer markers were investigated using optical tweezers (OT). The mucins examined included modified porcine submaxillary mucin containing the Tn epitope (Tn-PSM), ovine submaxillary mucin with the STn epitope (STn-OSM), and recombinant MUC1 analogs with either the Tn and STn epitope. OT experiments in which the mucins were immobilized onto polystyrene beads revealed identical self-interaction characteristics for all mucins. Identical binding strength and energy landscape characteristics were also observed for synthetic polymers displaying multiple GalNAc decorations. Polystyrene beads without immobilized mucins showed no self-interactions and also no interactions with mucin-decorated polystyrene beads. Taken together, the experimental data suggest that in these molecules, the GalNAc residue mediates interactions independent of the anchoring polymer backbone. Furthermore, GalNAc-GalNAc interactions appear to be responsible for self-interactions of mucins decorated with the STn epitope. Hence, Tn-MUC1 and STn-MUC1 undergo self-interactions mediated by the GalNAc residue in both epitopes, suggesting a possible molecular role in cancer. MUC1 possessing the T (Galß1-3GalNAcα1-Ser/Thr) or ST antigen (NeuNAcα2-3Galß1-3GalNAcα1-Ser/Thr) failed to show self-interactions. However, in the case of ST-MUC1, self-interactions were observed after subsequent treatment with neuraminidase and ß-galactosidase. This enzymatic treatment is expected to introduce Tn-epitopes and these observations thus further strengthen the conclusion that the observed interactions are mediated by the GalNAc groups.


Subject(s)
Acetylgalactosamine/metabolism , Antigens, Tumor-Associated, Carbohydrate/metabolism , Mucin-1/metabolism , Mucins/metabolism , Animals , Cattle , Humans , Swine
19.
Int J Oncol ; 49(1): 265-75, 2016 Jul.
Article in English | MEDLINE | ID: mdl-27176937

ABSTRACT

Distant metastases account for the majority of cancer-related deaths in breast cancer. The rate and site of metastasis differ between estrogen receptor (ER)-negative and ER-positive tumours, and metastatic fate can be very diverse even within the ER-negative group. Characterisation of new pro-metastatic markers may help to identify patients with higher risk and improve their care accordingly. Selectin ligands aberrantly expressed by cancer cells promote metastasis by enabling interaction between circulating tumour cells and endothelial cells in distant organs. These ligands consist in carbohydrate molecules, such as sialyl-Lewis x antigen (sLex), borne by glycoproteins or glycolipids on the cancer cell surface. We have previously demonstrated that the molecular scaffold presenting sLex to selectins (e.g. glycolipid vs. glycoproteins) was crucial for these interactions to occur. Moreover, we reported that detection of sLex alone in breast carcinomas was only of limited prognostic value. However, since sLex was found to be carried by several glycoproteins in cancer cells, we hypothesized that the combination of the carbohydrate with its carriers could be more relevant than each marker independently. In this study, we addressed this question by analysing sLex expression together with two glycoproteins (BST-2 and LGALS3BP), shown to interact with E-selectin in a carbohydrate-dependent manner, in a cohort of 249 invasive breast cancers. We found both glycoproteins to be associated with distant metastasis risk and poorer survival. Importantly, concomitant high expression of BST-2 with sLex defined a sub-group of patients with ER-negative tumours displaying higher risks of liver and brain metastasis and a 3-fold decreased survival rate.


Subject(s)
Antigens, CD/biosynthesis , Antigens, Neoplasm/biosynthesis , Biomarkers, Tumor/biosynthesis , Breast Neoplasms/genetics , Carrier Proteins/biosynthesis , E-Selectin/biosynthesis , Glycoproteins/biosynthesis , Adult , Aged , Aged, 80 and over , Antigens, CD/genetics , Antigens, Neoplasm/genetics , Biomarkers, Tumor/genetics , Breast Neoplasms/pathology , Carrier Proteins/genetics , Disease-Free Survival , E-Selectin/genetics , Estrogen Receptor alpha/genetics , Female , GPI-Linked Proteins/biosynthesis , GPI-Linked Proteins/genetics , Gene Expression Regulation, Neoplastic , Glycoproteins/genetics , Humans , Lewis X Antigen/genetics , Ligands , Middle Aged , Neoplasm Metastasis , Prognosis , Sialyl Lewis X Antigen
20.
PLoS One ; 11(3): e0151155, 2016.
Article in English | MEDLINE | ID: mdl-26958843

ABSTRACT

Around 7-17% of metastatic breast cancer patients will develop brain metastases, associated with a poor prognosis. To reach the brain parenchyma, cancer cells need to cross the highly restrictive endothelium of the Blood-Brain Barrier (BBB). As treatments for brain metastases are mostly inefficient, preventing cancer cells to reach the brain could provide a relevant and important strategy. For that purpose an in vitro approach is required to identify cellular and molecular interaction mechanisms between breast cancer cells and BBB endothelium, notably at the early steps of the interaction. However, while numerous studies are performed with in vitro models, the heterogeneity and the quality of BBB models used is a limitation to the extrapolation of the obtained results to in vivo context, showing that the choice of a model that fulfills the biological BBB characteristics is essential. Therefore, we compared pre-established and currently used in vitro models from different origins (bovine, mice, human) in order to define the most appropriate tool to study interactions between breast cancer cells and the BBB. On each model, the BBB properties and the adhesion capacities of breast cancer cell lines were evaluated. As endothelial cells represent the physical restriction site of the BBB, all the models consisted of endothelial cells from animal or human origins. Among these models, only the in vitro BBB model derived from human stem cells both displayed BBB properties and allowed measurement of meaningful different interaction capacities of the cancer cell lines. Importantly, the measured adhesion and transmigration were found to be in accordance with the cancer cell lines molecular subtypes. In addition, at a molecular level, the inhibition of ganglioside biosynthesis highlights the potential role of glycosylation in breast cancer cells adhesion capacities.


Subject(s)
Blood-Brain Barrier/cytology , Breast Neoplasms/pathology , Animals , Antigens, CD34/metabolism , Cattle , Cell Differentiation/physiology , Cell Line, Tumor , Cells, Cultured , Female , Fetal Blood/cytology , Humans , Mice , Neuroglia/cytology , Neuroglia/metabolism , Pericytes/cytology , Pericytes/metabolism , Rats
SELECTION OF CITATIONS
SEARCH DETAIL
...