The publications below were released in the framework of the GLYCANC project.  

The immunomodulatory role of tumor Syndecan-1 (CD138) on ex vivo tumor microenvironmental CD4+ T cell polarization in inflammatory and non-inflammatory breast cancer patients

Abstract

Herein, we aimed to identify the immunomodulatory role of tumor Syndecan-1 (CD138) in the polarization of CD4+ T helper (Th) subsets isolated from the tumor microenvironment of inflammatory breast cancer (IBC) and non-IBC patients. Lymphocytes and mononuclear cells isolated from the axillary tributaries of non-IBC and IBC patients during modified radical mastectomy were either stimulated with the secretome as indirect co-culture or directly co-cultured with control and Syndecan-1-silenced SUM-149 IBC cells. In addition, peripheral blood mononuclear cells (PBMCs) of normal subjects were used for the direct co-culture. Employing flow cytometry, we analyzed the expression of the intracellular IFN-γ, IL-4, IL-17, and Foxp3 markers as readout for basal and co-cultured Th1, Th2, Th17, and Treg CD4+ subsets, respectively. Our data revealed that IBC displayed a lower basal frequency of Th1 and Th2 subsets than non-IBC. Syndecan-1-silenced SUM-149 cells significantly upregulated only Treg subset polarization of normal subjects relative to controls. However, Syndecan-1 silencing significantly enhanced the polarization of Th17 and Treg subsets of non-IBC under both direct and indirect conditions and induced only Th1 subset polarization under indirect conditions compared to control. Interestingly, qPCR revealed that there was a negative correlation between Syndecan-1 and each of IL-4, IL-17, and Foxp3 mRNA expression in carcinoma tissues of IBC and that the correlation was reversed in non-IBC. Mechanistically, Syndecan-1 knockdown in SUM-149 cells promoted Th17 cell expansion via upregulation of IL-23 and the Notch ligand DLL4. Overall, this study indicates a low frequency of the circulating antitumor Th1 subset in IBC and suggests that tumor Syndecan-1 silencing enhances ex vivo polarization of CD4+ Th17 and Treg cells of non-IBC, whereby Th17 polarization is possibly mediated via upregulation of IL-23 and DLL4. These findings suggest the immunoregulatory role of tumor Syndecan-1 expression in Th cell polarization that may have therapeutic implications for breast cancer.

Moshira Ezzat Saleh, Ramy Gadalla, Hebatallah Hassan, Ahmed Afifi, Martin Götte, Mohamed El-Shinawi, Mona Mostafa Mohamed, Sherif Abdelaziz Ibrahim

PLOS, Published: May 30, 2019 doi.org/10.1371/journal.pone.0217550

Tumor-specific inhibitory action of decorin on different hepatoma cell lines

Authors: Zsolt Horváth, Andrea Reszegi, László Szilák, Titanilla Dankó, Ilona Kovalszky, Kornélia Baghy Cellular Signalling, Volume 62, October 2019, 109354 doi.org/10.1016/j.cellsig.2019.109354

A pre-print version of this article will be made available soon.

Proteoglycans and glycosaminoglycans as regulators of cancer stem cell function and therapeutic resistance

Abstract

In contrast to the bulk of the tumor, a subset of cancer cells called cancer stem cells (CSC; or tumor‐initiating cells) is characterized by self‐renewal, unlimited proliferative potential, expression of multidrug resistance proteins, active DNA repair capacity, apoptosis resistance, and a considerable developmental plasticity. Due to these properties, CSCs display increased resistance to chemo‐ and radiotherapy. Recent findings indicate that aberrant functions of proteoglycans (PGs) and glycosaminoglycans (GAGs) contribute substantially to the CSC phenotype and therapeutic resistance. In this review, we summarize how the diverse functions of the glycoproteins and carbohydrates facilitate acquisition and maintenance of the CSC phenotype, and how this knowledge can be exploited to develop novel anticancer therapies. For example, the large transmembrane chondroitin sulfate PG NG2/CSPG4 marks stem cell (SC) populations in brain tumors. Cell surface heparan sulfate PGs of the syndecan and glypican families modulate the stemness‐associated Wnt, hedgehog, and notch signaling pathways, whereas the interplay of hyaluronan in the SC niche with CSC CD44 determines the maintenance of stemness and promotes therapeutic resistance. A better understanding of the molecular mechanisms by which PGs and GAGs regulate CSC function will aid the development of targeted therapeutic approaches which could avoid relapse after an otherwise successful conventional therapy. Chimeric antigen receptor T cells, PG‐primed dendritic cells, PG‐targeted antibody–drug conjugates, and inhibitory peptides and glycans have already shown highly promising results in preclinical models.

Daiana Vitale, Sampath Kumar Katakam, Burkhard Greve, Bohee Jang, Eok‐Soo Oh, Laura Alaniz, Martin Götte

FEBS Journal, 23 June 2019 doi.org/10.1111/febs.14967

Hyaluronan preconditioning of monocytes/macrophages affects their angiogenic behavior and regulation of TSG‐6 expression in a tumor type‐specific manner

Abstract

Hyaluronan is a glycosaminoglycan normally present in the extracellular matrix in most tissues. Hyaluronan is a crucial player in many processes associated with cancer, such as angiogenesis, invasion, and metastasis. However, little has been reported regarding the action of hyaluronan on monocytes/macrophages (Mo/MØ) in tumor angiogenesis and its consequences on tumor development. In the present study, we investigated the effects of hyaluronan of different sizes on human Mo/MØ angiogenic behavior in colorectal and breast carcinoma. In vitro, the treatment of Mo/MØ with lysates and conditioned media from a breast but not from colorectal carcinoma cell line plus high‐molecular weight hyaluronan induced: (a) an increased expression of angiogenic factors VEGF, IL‐8, FGF‐2, and MMP‐2, (b) an increased endothelial cell migration, and (c) a differential expression of hyaluronan‐binding protein TSG‐6. Similar results were observed in Mo/MØ derived from breast cancer patients treated with tumor lysates. Besides, macrophages primed with high‐molecular weight hyaluronan and inoculated in human breast cancer xenograft tumor increased blood vessel formation and diminished TSG‐6 levels. In contrast, the effects triggered by high‐molecular weight hyaluronan on Mo/MØ in breast cancer context were not observed in the context of colorectal carcinoma. Taken together, these results indicate that the effect of high‐molecular weight hyaluronan as an inductor of the angiogenic behavior of macrophages in breast tumor context is in part consequence of the presence of TSG‐6.

Fiorella M. Spinelli, Daiana L. Vitale, Antonella Icardi, Ilaria Caon, Alejandra Brandone, Paula Giannoni, Virginia Saturno, Alberto Passi, Mariana García, Ina Sevic, Laura Alaniz.

FEBS Journal, 01 May 2019 doi.org/10.1111/febs.14871

Syndecan-1 in Liver Diseases

Abstract

Liver diseases such as liver cirrhosis, primary and metastatic liver cancers are still a major medical challenge. Syndecan-1 is one of the most important proteoglycans in the liver. Syndecan-1 is normally expressed on the surfaces of hepatocytes and cholangiocytes. Due to liver diseases the amount of syndecan-1 increases in the liver. Despite the emerging data of the biological function of syndecan-1, the clinical usefulness of this proteoglycan is still unknown. In our study we correlated syndecan-1 expression to clinico-pathological data. We found that syndecan-1 proved to be a good marker for hepatitis C virus based hepatocellular carcinoma and increased with liver dysfunction.

Eszter Regős, Katalin Karászi, Andrea Reszegi, András Kiss, Zsuzsa Schaff, Kornélia Baghy, Ilona Kovalszky.

Pathol. Oncol. Res. (2019). doi.org/10.1007/s12253-019-00617-0

Please click here for a full text document of the article.

Tumor-suppressive functions of 4-MU on breast cancer cells of different ER status: Regulation of hyaluronan/HAS2/CD44 and specific matrix effectors.

Authors: Theodoros T.Karalis, Paraskevi Heldin, Demitrios H.Vynios, Thomas Neill, Simone Buraschi, Renato V.Iozzo, Nikos K.Karamanos, Spyros S. Skandalis

Matrix Biology, Volumes 78–79, May 2019, Pages 118-138 doi.org/10.1016/j.matbio.2018.04.007

This article was not published with open access.

Co-treatment of tumor cells with hyaluronan plus doxorubicin affects endothelial cell behavior independently of VEGF expression

Abstract

Hyaluronan, the main glycosaminoglycan of extracellular matrices, is concentrated in tissues with high cell proliferation and migration rates. In cancer, hyaluronan expression is altered and it becomes fragmented into low-molecular-weight forms, affecting mechanisms associated with cell proliferation, invasion, angiogenesis and multidrug resistance. Here, we analyzed the effect of low-molecular-weight hyaluronan on the response of T lymphoma, osteosarcoma, and mammary adenocarcinoma cell lines to the antineoplastic drug doxorubicin, and whether co-treatment with hyaluronan and doxorubicin modified the behavior of endothelial cells. Our aim was to associate the hyaluronan-doxorubicin response with angiogenic alterations in these tumors. After hyaluronan and doxorubicin co-treatment, hyaluronan altered drug accumulation and modulated the expression of ATP-binding cassette transporters in T-cell lymphoma cells. In contrast, no changes in drug accumulation were observed in cells from solid tumors, indicating that hyaluronan might not affect drug efflux. However, when we evaluated the effect on angiogenic mechanisms, the supernatant from tumor cells treated with doxorubicin exhibited a pro-angiogenic effect on endothelial cells. Hyaluronan-doxorubicin co-treatment increased migration and vessel formation in endothelial cells. This effect was independent of vascular endothelial growth factor but related to fibroblast growth factor-2 expression. Besides, we observed a pro-angiogenic effect on endothelial cells during hyaluronan and doxorubicin co-treatment in the in vivo murine model of T-cell lymphoma. Our results demonstrate for the first time that hyaluronan is a potential modulator of doxorubicin response by mechanisms that involve not only drug efflux but also angiogenic processes, providing an adverse tumor stroma during chemotherapy.

Daiana L. Vitale, Fiorella M. Spinelli, Daiana Del Dago, Antonella Icardi, Gianina Demarchi, Ilaria Caon, Mariana García, Marcela F. Bolontrade, Alberto Passi, Carolina Cristina, and Laura Alaniz. Oncotarget. 2018; 9:36585-36602. doi.org/10.18632/oncotarget.26379

Advances in targeting epidermal growth factor receptor signaling pathway in mammary cancer

This publication is an overview article and was not published as open access. Please find more details under the link to PubMed.

Authors: Konstantina Kyriakopoulou, Elena Kefali, Zoi Piperigkou, Heba Bassiony, Nikos K. Karamanos

Cellular Signalling, Volume 51, November 2018, Pages 99-109

Extracellular matrix functions in lung cancer

Abstract Lung cancer is one of the most devastating types of malignant tumors, and a prime example of a chemical carcinogenesis-driven tumor entity. Lung cancer progression does not only depend on the presence of driver mutations within the cancer cells, but also on its interactions with the cellular and extracellular matrix environment. Genetic and epigenetic changes such as aberrant promoter methylation or altered expression of microRNAs cause the misexpression of collagens, laminins, proteoglycans, proteases and integrins in the tumor microenvironment. Associated signaling processes modulate the conversion of fibroblasts to contractile cancer-associated fibroblasts, tumor angiogenesis, escape from the immune system, and create a permissive niche for cancer stem cells. Protease-mediated digestion and posttranslational crosslinking of large matrix glycoproteins promote cancer cell motility by facilitating basement membrane penetration and by altering the stiffness of interstitial matrices. New therapeutic approaches utilizing specific matrix proteins as targets and tools for drug delivery have shown promising results in preclinical models of lung cancer. Martin Götte, Ilona Kovalszky (2018). Extracellular matrix functions in lung cancer. Matrix Biology, published online 27 February 2018, DOI: doi.org/10.1016/j.matbio.2018.02.018 ;

Syndecan-1 inhibits early stages of liver fibrogenesis by interfering with TGFβ1 action and upregulating MMP14

AbstractIncreased expression of syndecan-1 is a characteristic feature of human liver cirrhosis. However, no data are available on the significance of this alteration. To address this question we designed a transgenic mouse strain that driven by albumin promoter, expresses human syndecan-1 in the hepatocytes. Liver cirrhosis was induced by thioacetamide in wild type and hSDC1+/+ mice of the identical strain. The process of fibrogenesis, changes in signal transduction and proteoglycan expression were followed. In an in vitro experiment, the effect of syndecan-1 overexpression on the action of TGFβ1 was determined. Human syndecan-1 and TGFβ1 levels were measured by ELISA in the circulation. Without challenge, no morphological differences were observed between wild type and transgenic mice livers, although significant upregulation of phospho-Akt and FAK was observed in the latter. Fibrogenesis in the transgenic livers, characterized by picrosirius staining, collagen type I, and SMA levels, lagged behind that of control in the first and second months. Changes in signal transduction involved in the process of fibrogenesis, as SMAD, MAPK, Akt and GSK, pointed to the decreased effect of TGFβ1, and this was corroborated by the decreased mRNA expression of TIEG and the growth factor itself. In vitro experiments exposing the LX2 hepatic stellate cell line to conditioned media of wild type and syndecan-1 transfected Hep3B cell lines proved that medium with high syndecan-1 content inhibits TGFβ1-induced upregulation of SMA, TIEG, collagen type I and thrombospondin-1 expression. Detection of liver proteoglycans and heparan sulfate level revealed that their amounts are much higher in control transgenic liver, than that in the wild type. However, it decreases dramatically as a result of shedding after hepatic injury. Shedding is likely promoted by the upregulation of MMP14. As syndecan-1 can bind thrombospondin-1, and as our result demonstrated that the same is true for TGFβ1, shed syndecan-1 can remove the growth factor and its activator together into the systemic circulation.Taking together, our results indicate that the effect of syndecan-1 is accomplished on two levels: a, the shedded syndecan can bind, inhibit and remove TGFβ1; b, interferes with the activation of TGFβ1 by downregulation and binding thrombospondin-1, the activator of the growth factor. However, by the end of the fourth month the protective effect was lost, which is explained by the considerable decrease of syndecan-1 and the almost complete loss of heparan sulfate from the surface of hepatocytes. Eszter Regős, Hadeer Hesham Abdelfattah, Andrea Reszegi, László Szilák, Klára Werling, Gábor Szabó, András Kiss, Zsuzsa Schaff, Ilona Kovalszky, Kornélia Baghy  (2018). Syndecan-1 inhibits early stages of liver fibrogenesis by interfering with TGFβ1 action and upregulating MMP14. Matrix Biology, Published online on 16th February 2018, DOI: doi.org/10.1016/j.matbio.2018.02.008

Changes in hyaluronan deposition in the rat myenteric plexus after experimentally-induced colitis

AbstractMyenteric plexus alterations hamper gastrointestinal motor function during intestinal inflammation. Hyaluronan (HA), an extracellular matrix glycosaminoglycan involved in inflammatory responses, may play a role in this process. In the colon of control rats, HA-binding protein (HABP), was detected in myenteric neuron soma, perineuronal space and ganglia surfaces. Prominent hyaluronan synthase 2 (HAS2) staining was found in myenteric neuron cytoplasm, suggesting that myenteric neurons produce HA. In the myenteric plexus of rats with 2, 4-dinitrobenzene sulfonic (DNBS)-induced colitis HABP staining was altered in the perineuronal space, while both HABP staining and HA levels increased in the muscularis propria. HAS2 immunopositive myenteric neurons and HAS2 mRNA and protein levels also increased. Overall, these observations suggest that inflammation alters HA distribution and levels in the gut neuromuscular compartment. Such changes may contribute to alterations in the myenteric plexus. Authors: Viviana Filpa, Michela Bistoletti, Ilaria Caon, Elisabetta Moro, Annalisa Grimaldi, Paola Moretto, Andreina Baj, Maria Cecilia Giron, Evgenia Karousou, Manuela Viola, Francesca Crema, Gianmario Frigo, Alberto Passi, Cristina Giaroni & Davide Vigetti Scientific REPorts | 7: 17644 | DOI:10.1038/s41598-017-18020-7 Please click here for the article.

Insights into the key roles of epigenetics in matrix macromolecules-associated wound healing

Abstract Extracellularmatrix (ECM) is a dynamic network ofmacromolecules, playing a regulatory role in cell functions, tissue regeneration and remodeling. Wound healing is a tissue repair process necessary for the maintenance of the functionality of tissues and organs. This highly orchestrated process is divided into four temporally overlapping phases, including hemostasis, inflammation, proliferation and tissue remodeling. The dynamic interplay between ECMand resident cells exerts its critical role in many aspects of wound healing, including cell proliferation, migration, differentiation, survival, matrix degradation and biosynthesis. Several epigenetic regulatory factors, such as the endogenous non-coding microRNAs (miRNAs), are the drivers of the wound healing response. microRNAs have pivotal roles in regulating ECMcomposition duringwound healing and dermal regeneration. Their expression is associated with the distinct phases ofwound healing and they serve as target biomarkers and targets for systematic regulation of wound repair. In this article we critically present the importance of epigenetics with particular emphasis on miRNAs regulating ECM components (i.e. glycoproteins, proteoglycans and matrix proteases) that are key players in wound healing. The clinical relevance of miRNA targeting as well as the delivery strategies designed for clinical applications are also presented and discussed. Please find a link to the article here.Z. Piperigkou, et al., Insights into the key roles of epigenetics inmatrixmacromolecules-associated wound healing, Adv. Drug Deliv. Rev. (2017), doi.org/10.1016/j.addr.2017.10.008

Extracellular matrix alterations in senescent cells and their significance in tissue homeostasis

Abstract Normal cells after a defined number of successive divisions or after exposure to genotoxic stresses are becoming senescent, characterized by a permanent growth arrest. In addition, they secrete increased levels of pro-inflammatory and catabolic mediators, collectively termed “senescence-associated secretory phenotype”. Furthermore, senescent cells exhibit an altered expression and organization of many extracellular matrix components, leading to specific remodeling of their microenvironment. In this review we present the current knowledge on extracellular matrix alterations associated with cellular senescence and critically discuss certain characteristic examples, highlighting the ambiguous role of senescent cells in the homeostasis of various tissues under both normal and pathologic conditions. Please find more information here. E. Mavrogonatou, et al., Extracellular matrix alterations in senescent cells and their significance in tissue homeostasis, Matrix Biol (2017), doi.org/10.1016/j.matbio.2017.10.004

Estrogen receptor beta as epigenetic mediator of miR-10b and miR-145 in mammary cancer

Abstract Even though the role of estrogen receptor alpha (ERα) in the modulation of breast cancer cells’ behavior is thoroughly studied, the biological functions of its isoform, ERβ, are less elucidated. The suppression of ERβ in the aggressive ERα-negative MDA-MB- 231 breast cancer cells resulted in the inhibition of epithelial to mesenchymal transition (EMT) through and major changes in the basic functional properties expression levels of certain matrix components of breast cancer cells. This arrest in metastatic potential of breast cancer cells suggests the contribution of ERβ in the induction of a more aggressive phenotype in MDA-MB-231 breast cancer cells. The epigenetic alterations are responsible for the ability of the tumor cells to metastasize. Here, we report for the first time that the suppression of ERβ in MDA-MB-231 breast cancer cells leads to significant changes in the expression profiles of specific microRNAs, including miR-10b, miR-200b and miR-145. Growth of MCF-7 and MDA-MB-231 cells with estrogen-free medium has a diverse impact on miRNA expression and the behaviour of these cells suggesting the diverse effect of estradiol on the miRNA expression profile depending on the ER status of breast cancer cells. Transfections of miR-10b precursor and miR-145 inhibitor clearly revealed Enhanced miR-10b expression or silencing of miR-145 clearly revealed that these microRNAs can regulate the functional properties, EMT program and the expression of major matrix components known to be implicated in breast cancer aggressiveness. Our data revealed that miR-10b is strongly implicated in the regulation of functional properties, EMT program and Erk1/2 signaling in shERβ MDA-MB-231 cells, thus affecting the extracellular matrix (ECM) composition, including syndecan-1, proteolytic behaviour, especially MMP2, MMP7 and MMP9 expression and subsequently the aggressiveness of these cells. Accordingly, the inhibition of miR-145 expression significantly increased the aggressiveness of shERβ MDA-MB-231 cells and induced EMT. Moreover, miR-145 inhibition resulted in important changes in the gene and protein levels of ECM mediators, such as HER2 and several MMPs, whereas it significantly increased the phosphorylated levels of Erk1/2 kinases in these cells, suggesting the crucial role of miR-145 in this signaling pathway. These novel results suggest that the alterations in cell behaviour and in ECM composition caused by the suppression of ERβ in MDA-MB-231 cells are closely related to certain epigenetic miRNA-induced alterations. Targeting the ERβ-regulated miR-10b and miR-145 is a promising tool for diagnosis and pharmaceutical targeting in breast cancer.Please find a pre-print version of the article here.Piperigkou, Zoi, Franchi, Marco, G¨otte, Martin, Karamanos, Nikos K., Estrogen receptor beta as epigenetic mediator of miR-10b and miR-145 in mammary cancer, Matrix Biology (2017), doi:10.1016/j.matbio.2017.08.002

Inhibition of Heparanase in Pediatric Brain Tumor Cells Attenuates their Proliferation, Invasive Capacity, and In Vivo Tumor Growth

AbstractCurative therapy for medulloblastoma and other pediatric embryonal brain tumors has improved, but the outcome still remains poor and current treatment causes long-term complications. Malignant brain tumors infiltrate the healthy brain tissue and, thus despite resection, cells that have already migrated cause rapid tumor regrowth. Heparan sulfate proteoglycans (HSPG), major components of the extracellular matrix (ECM), modulate the activities of a variety of proteins. The major enzyme that degrades HS, heparanase (HPSE), is an important regulator of the ECM. Here, we report that the levels of HPSE in pediatric brain tumors are higher than in healthy brain tissue and that treatment of pediatric brain tumor cells with HPSE stimulated their growth. In addition, the latent, 65 kDa form of HPSE (that requires intracellular enzymatic processing for activation) enhanced cell viability and rapidly activated the ERK and AKT signaling pathways, before enzymatically active HPSE was detected. The HPSE inhibitor PG545 efficiently killed pediatric brain tumor cells, but not normal human astrocytes, and this compound also reduced tumor cell invasion in vitro and potently reduced the size of flank tumors in vivo. Our findings indicate that HPSE in malignant brain tumors affects both the tumor cells themselves and their ECM. In conclusion, HPSE plays a substantial role in childhood brain tumors, by contributing to tumor aggressiveness and thereby represents a potential therapeutic target. Please click here for the pdf.Inhibition of Heparanase in Pediatric Brain Tumor Cells Attenuates their Proliferation, Invasive Capacity, and In Vivo Tumor Growth. Argyris Spyrou, Soumi Kundu, Lulu Haseeb, Di Yu , Tommie Olofsson, Keith Dredge, Edward Hammond, Uri Barash, Israel Vlodavsky, and Karin Forsberg-Nilsson. Molecular Cancer Therapeutics Online © 2017 American Association for Cancer Research. doi: 10.1158/1535-7163.MCT-16-0900. Published Online First July 17, 2017.

Probing glycosaminoglycan spectral signatures in live cells and their conditioned media by Raman microspectroscopy

Abstract Spectroscopic markers characteristic of reference glycosaminoglycan molecules were identified previously based on their vibrational signatures. Infrared spectral signatures of glycosaminoglycans in fixed cells were also recently demonstrated but probing live cells still remains challenging. Raman microspectroscopy is potentially interesting to perform studies in physiological conditions. The aim of the present work was to identify the Raman spectral signatures of GAGs in fixed and live cells and in their conditioned media. Biochemical and Raman analyses were performed from five cell types: chondrocytes, dermal fibroblasts, melanoma (SK-MEL-28), wild type CHO, and glycosaminoglycan-defective mutant CHO-745 cells. Biochemical assay of sulfated GAGs in conditioned media was only possible for chondrocytes, dermal fibroblasts, wild type CHO due to the detection limit of the test. In contrast, Raman microspectroscopy allowed probing total glycosaminoglycan content in conditioned media, fixed and live cells and the data were analysed by principal component analysis. Our results showed that the Raman technique is sensitive enough to identify spectral markers of glycosaminoglycans that were useful to characterise the conditioned media of the five cell types. The results were confirmed at the single cell level on both live and fixed cells with a good differentiation between the cell types. Further, the principal component loadings revealed prominent glycosaminoglycanrelated spectral information. Raman microspectroscopy allows to monitor glycosaminoglycan profiles of single live cell and could therefore be developed for cell screening purposes and holds promises for identifying glycosaminoglycan signatures as a marker of cancer progression in tissues.The full text is available in a pre-print version under:  pubs.rsc.org/en/content/articlelanding/2017/an/c6an01951j S. Brézillon, V. Untereiner, H. T. Mohamed, J. Hodin, A. Chatron-Colliet, F. X. Maquart and G. D. Sockalingum. Probing glycosaminoglycan spectral signatures in live cells and their conditioned media by Raman microspectroscopy. Analyst, 2017, DOI: 10.1039/C6AN01951J.

Syndecan-1 is a novel molecular marker for triple negative inflammatory breast cancer and modulates the cancer stem cell phenotype via the IL-6/STAT3, Notch and EGFR signaling pathways

Abstract Background: Inflammatory breast cancer (IBC), a particularly aggressive form of breast cancer, is characterized by cancer stem cell (CSC) phenotype. Due to a lack of targeted therapies, the identification of molecular markers of IBC is of major importance. The heparan sulfate proteoglycan Syndecan-1 acts as a coreceptor for growth factors and chemokines, modulating inflammation, tumor progression, and cancer stemness, thus it may emerge as a molecular marker for IBC. Methods: We characterized expression of Syndecan-1 and the CSC marker CD44, Notch-1 & -3 and EGFR in carcinoma tissues of triple negative IBC (n = 13) and non-IBC (n = 17) patients using qPCR and immunohistochemistry. Impact of siRNA-mediated Syndecan-1 knockdown on the CSC phenotype of the human triple negative IBC cell line SUM-149 and HER-2-overexpressing non-IBC SKBR3 cells employing qPCR, flow cytometry, Western blotting, secretome profiling and Notch pharmacological inhibition experiments. Data were statistically analyzed using Student’s t-test/Mann-Whitney U-test or one-way ANOVA followed by Tukey’s multiple comparison tests. Results: Our data indicate upregulation and a significant positive correlation of Syndecan-1 with CD44 protein, and Notch-1 & -3 and EGFR mRNA in IBC vs non-IBC. ALDH1 activity and the CD44(+)CD24(-/low) subset as readout of a CSC phenotype were reduced upon Syndecan-1 knockdown. Functionally, Syndecan-1 silencing significantly reduced 3D spheroid and colony formation. Intriguingly, qPCR results indicate downregulation of the IL-6, IL-8, CCL20, gp130 and EGFR mRNA upon Syndecan-1 suppression in both cell lines. Moreover, Syndecan-1 silencing significantly downregulated Notch-1, -3, -4 and Hey-1 in SUM-149 cells, and downregulated only Notch-3 and Gli-1 mRNA in SKBR3 cells. Secretome profiling unveiled reduced IL-6, IL-8, GRO-alpha and GRO a/b/g cytokines in conditioned media of Syndecan-1 knockdown SUM-149 cells compared to controls. The constitutively activated STAT3 and NFκB, and expression of gp130, Notch-1 & -2, and EGFR proteins were suppressed upon Syndecan-1 ablation. Mechanistically, gamma-secretase inhibition experiments suggested that Syndecan-1 may regulate the expression of IL-6, IL-8, gp130, Hey-1, EGFR and p-Akt via Notch signaling.The full text is avalable here.Sherif Abdelaziz Ibrahim, Ramy Gadalla, Eslam A. El-Ghonaimy, Omnia Samir, Hossam Taha Mohamed, Hebatallah Hassan, Burkhard Greve, Mohamed El-Shinawi, Mona Mostafa Mohamed, and Martin Götte (2017). Syndecan-1 is a novel molecular marker for triple negative inflammatory breast cancer and modulates the cancer stem cell phenotype via the IL-6/STAT3, Notch and EGFR signaling pathways. Molecular Cancer. DOI 10.1186/s12943-017-0621-z. Published online 07.03.2017.

MDA-MB-231 breast cancer cell viability, motility and matrix adhesion are regulated by a complex interplay of heparan sulfate, chondroitin−/dermatan sulfate and hyaluronan biosynthesis

Abstract Proteoglycans and glycosaminoglycans modulate numerous cellular processes relevant to tumour progression, including cell proliferation, cell-matrix interactions, cell motility and invasive growth. Among the glycosaminoglycans with a well-documented role in tumour progression are heparan sulphate, chondroitin/dermatan sulphate and hyaluronic acid/hyaluronan. While the mode of biosynthesis differs for sulphated glycosaminoglycans, which are synthesised in the ER and Golgi compartments, and hyaluronan, which is synthesized at the plasma membrane, these polysaccharides partially compete for common substrates. In this study, we employed a siRNA knockdown approach for heparan sulphate (EXT1) and heparan/chondroitin/dermatan sulphate-biosynthetic enzymes (β4GalT7) in the aggressive human breast cancer cell line MDA-MB-231 to study the impact on cell behaviour and hyaluronan biosynthesis. Knockdown of β4GalT7 expression resulted in a decrease in cell viability, motility and adhesion to fibronectin, while these parameters were unchanged in EXT1-silenced cells. Importantly, these changes were associated with a decreased expression of syndecan-1, decreased signalling response to HGF and an increase in the synthesis of hyaluronan, due to an upregulation of the hyaluronan synthases HAS2 and HAS3. Interestingly, EXT1-depleted cells showed a downregulation of the UDP-sugar transporter SLC35D1, whereas SLC35D2 was downregulated in β4GalT7-depleted cells, indicating an intricate regulatory network that connects all glycosaminoglycans synthesis. The results of our in vitro study suggest that a modulation of breast cancer cell behaviour via interference with heparan sulphate biosynthesis may result in a compensatory upregulation of hyaluronan biosynthesis. These findings have important implications for the development of glycosaminoglycan-targeted therapeutic approaches for malignant diseases. Plesae find the full text here.Viola M, Brüggemann K, Karousou E, Caon I, Caravà E, Vigetti D, Greve B, Stock C, De Luca G, Passi A, Götte M. MDA-MB-231 breast cancer cell viability, motility and matrix adhesion are regulated by a complex interplay of heparan sulfate, chondroitin−/dermatan sulfate and hyaluronan biosynthesis. Glycoconj J. DOI 10.1007/s10719-016-9735-6. Copyright Springer Science+Business Media New York, Published online 15 October 2016

microRNA miR-142-3p Inhibits Breast Cancer Cell Invasiveness by Synchronous Targeting of WASL, Integrin Alpha V, and Additional Cytoskeletal Elements

Abstract MicroRNAs (miRNAs, micro ribonucleic acids) are pivotal post-transcriptional regulators of gene expression. These endogenous small non-coding RNAs play significant roles in tumorigenesis and tumor progression. miR-142-3p expression is dysregulated in several breast cancer subtypes. We aimed at investigating the role of miR-142-3p in breast cancer cell invasiveness. Supported by transcriptomic Affymetrix array analysis and confirmatory investigations at the mRNA and protein level, we demonstrate that overexpression of miR- 142-3p in MDA-MB-231, MDA-MB-468 and MCF-7 breast cancer cells leads to downregulation of WASL (Wiskott-Aldrich syndrome-like, protein: N-WASP), Integrin-αV, RAC1, and CFL2, molecules implicated in cytoskeletal regulation and cell motility. ROCK2, IL6ST, KLF4, PGRMC2 and ADCY9 were identified as additional targets in a subset of cell lines. Decreased Matrigel invasiveness was associated with the miR-142-3p-induced expression changes. Confocal immunofluorescence microscopy, nanoscale atomic force microscopy and digital holographic microscopy revealed a change in cell morphology as well as a reduced cell volume and size. A more cortical actin distribution and a loss of membrane protrusions were observed in cells overexpressing miR-142-3p. Luciferase activation assays confirmed direct miR-142-3p-dependent regulation of the 3’-untranslated region of ITGAV and WASL. siRNA-mediated depletion of ITGAV and WASL resulted in a significant reduction of cellular invasiveness, highlighting the contribution of these factors to the miRNAdependent invasion phenotype. While knockdown of WASL significantly reduced the number of membrane protrusions compared to controls, knockdown of ITGAV resulted in a decreased cell volume, indicating differential contributions of these factors to the miR-142-3p-induced phenotype. Our data identify WASL, ITGAV and several additional cytoskeleton- associated molecules as novel invasion-promoting targets of miR-142-3p in breast cancer. Please click here for the full text article. Schwickert A, Weghake E, Brüggemann K, Engbers A, Brinkmann BF, Kemper B, Seggewiß J, Stock C, Ebnet K, Kiesel L, Riethmüller C, Götte M. microRNA miR-142-3p Inhibits Breast Cancer Cell Invasiveness by Synchronous Targeting of WASL, Integrin Alpha V, and Additional Cytoskeletal Elements. PLoS ONE 10(12): e0143993. doi:10.1371/journal.pone.0143993

Shed proteoglycans in the tumor stroma

Abstract Cancer cell behavior is not only governed by tumor cell-autonomous properties, but also by the surrounding tumor stroma. Cancer-associated fibroblasts, blood vessels, immune cells and the extracellular matrix of the tumor microenvironment have a profound influence on tumor progression. Proteoglycans control various normal and pathological processes, modulating cell proliferation and motility, cell-matrix interactions, immune cell recruitment and angiogenesis. They are major mediators of cancer cell behavior though a dynamic interplay with extracellular matrix components. During cancer progression, their altered expression can promote the activation of several signaling cascades regulating crucial functional properties of cancer cells. Notably, the function of cell surface proteoglycans can be altered by ectodomain shedding, which converts membrane-bound coreceptors into soluble paracrine effector molecules. In this review, we highlight the importance of proteoglycans and their soluble counterparts in cancer progression and the consequences if their interactions with the adjacent stroma. The dynamic interplay among shed proteoglycans and proteolytic enzymes has a significant impact both on tumor cells and their surrounding stroma, with important implications for the diagnosis of this disease and novel therapeutic approaches. Please click here for the full text (pre-print version).Piperigkou Z, Mohr B, Karamanos N, Götte M. Shed proteoglycans in the tumor stroma. DOI 10.1007/s00441-016-2452-4, Received: 4 May 2016 /Accepted: 8 June 2016, Published online July 01, 2016 The copyright of the final version is owned by Springer-Verlag Berlin Heidelberg 2016. The final version can be found at link.springer.com/article/10.1007%2Fs00441-016-2452-4

Roles and Targeting of the HAS/Hyaluronan/CD44 Molecular System in Cancer

Abstract Synthesis, deposition, and interactions of hyaluronan (HA) with its cellular receptor CD44 are crucial events that regulate the onset and progression of tumors. The intracellular signaling pathways initiated by HA interactions with CD44 leading to tumorigenic responses are complex. Moreover, HA molecules may perform dual functions depending on their concentration and size. Overexpression of variant isoforms of CD44 (CD44v) is most commonly linked to cancer progression, whereas their loss is associated with inhibition of tumor growth. In this review, we highlight that the regulation of HA synthases (HASes) by post-translational modifications, such as O-GlcNAcylation and ubiquitination, environmental factors and the action of microRNAs is important for HA synthesis and secretion in the tumor microenvironment. Moreover, we focus on the roles and interactions of CD44 with various proteins that reside extra- and intracellularly, as well as on cellular membranes with particular reference to the CD44-HA axis in cancer stem cell functions, and the importance of CD44/CD44v6 targeting to inhibit tumorigenesis. Please find here the full text.Karousou, Evgenia; Misra, Suniti, Ghatak, Shibnath; Dobra, Katalin; Götte, Martin; Vigetti, Davide; Passi, Alberto; Karamanos, Nikos K.; Skandalis, Spyros S. Roles and targeting of the HAS/hyaluronan/CD44 molecular system in cancer, Matrix Biology (2016), doi:10.1016/j.matbio.2016.10.001

Biological function of unique sulfated glycosaminoglycans in primitive chordates

Abstract Glycosaminoglycans with unique sulfation patterns have been identified in different species of ascidians (sea squirts), a group of marine invertebrates of the Phylum Chordata, sub-phylum Tunicata (or Urochordata). Oversulfated dermatan sulfate composed of [4-α-L-IdoA-(2-O-SO3)−1 → 3-β-D-GalNAc(4-OSO3)−1]n repeating disaccharide units is found in the extracellular matrix of several organs, where it seems to interact with collagen fibers. This dermatan sulfate co-localizes with a decorin-like protein, as indicated by immunohistochemical analysis. Low sulfated heparin/heparan sulfate-like glycans composed mainly of [4-α-L-IdoA-(2-OSO3)−1 → 4-α-D-GlcN(SO3)−1 (6-O-SO3)−1]n and [4-α-L-IdoA-(2-O-SO3)−1 → 4-α-D-GlcN(SO3)−1]n have also been described in ascidians. These heparin-like glycans occur in intracellular granules of oocyte assessory cells, named test cells, in circulating basophil-like cells in the hemolymph, and at the basement membrane of different ascidian organs. In this review, we present an overview of the structure, distribution, extracellular and intracellular localization of the sulfated glycosaminoglycans in different species and tissues of ascidians. Considering the phylogenetic position of the subphylum Tunicata in the phylum Chordata, a careful analysis of these data can reveal important information about how these glycans evolved from invertebrate to vertebrate animals. Please click here for the full text.Karamanou K, Restrepo Espinosa DC, Fortuna-Costa A, & Pavão MSG. Biological function of unique sulfated glycosaminoglycans in primitive chordates. DOI 10.1007/s10719-016-9728-5. Glycoconj J. Copyright Springer Science+Business Media New York 2016. Published online 10 September 2016

Implementation of infrared and Raman modalities for glycosaminoglycan characterization in complex systems

AbstractGlycosaminoglycans (GAGs) are natural, linear and negatively charged heteropolysaccharides which are incident in every mammalian tissue. They consist of repeating disaccharide units, which are composed of either sulfated or non-sulfated monosaccharides. Depending on tissue types, GAGs exhibit structural heterogeneity such as the position and degree of sulfation or within their disaccharide units composition being heparin, heparan sulfate, chondroitine sulfate, dermatan sulfate, keratan sulfate, and hyaluronic acid. They are covalently linked to a core protein (proteoglycans) or as free chains (hyaluronan). GAGs affect cell properties and functions either by direct interaction with cell receptors or by sequestration of growth factors. These evidences of divert biological roles of GAGs make their characterization at cell and tissue levels of importance. Thus, non-invasive techniques are interesting to investigate, to qualitatively and quantitatively characterize GAGs in vitro in order to use them as diagnostic biomarkers and/or as therapeutic targets in several human diseases including cancer. Infrared and Raman microspectroscopies and imaging are sensitive enough to differentiate and classify GAG types and subtypes in spite of their close molecular structures. Spectroscopic markers characteristic of reference GAG molecules were identified. Beyond these investigations of the standard GAG spectral signature, infrared and Raman spectral signatures of GAG were searched in complex biological systems like cells. The aim of the present review is to describe the implementation of these complementary vibrational spectroscopy techniques, and to discuss their potentials, advantages and disadvantages for GAG analysis. In addition, this review presents new data as we show for the first time GAG infrared and Raman spectral signatures from conditioned media and live cells, respectively.Please click here for the full text.Mohamed HT, Untereiner V, Sockalingum GD, Brézillon S. Implementation of infrared and Raman modalities for glycosaminoglycan characterization in complex systems. PMID: 27928742. DOI: 10.1007/s10719-016-9743-6 [PubMed - as supplied by publisher] Glycoconj J. 2016 Dec 7. [Epub ahead of print]KEYWORDS: CHO-745; CHO-WT; Chondrocytes; Conditioned media; Data analysis; Glycosaminoglycans; Infrared spectroscopy; Raman spectroscopy

IGF-IR cooperates with ERα to inhibit breast cancer cell aggressiveness by regulating the expression and localisation of ECM molecules

In their recent publication "IGF-IR cooperates with ERα to inhibit breast cancer cell aggressiveness by regulating the expression and localisation of ECM molecules" in Scientific Reports (from Nature Publishers), the research teams from UPAT and BRIC highlight IGF-IR as a major molecular target for novel therapeutic strategies in breast cancer. In particular, they report a nodal role of IGF-IR in the regulation of ERα-positive breast cancer cell aggressiveness and the regulation of expression levels of several extracellular matrix molecules. Activation of IGF-IR, but not EGFR, in MCF-7 breast cancer cells results in the reduction of specific matrix metalloproteinases. In contrast, IGF-IR inhibition leads to the depletion by endocytosis of syndecan-4. Global important changes in cell adhesion receptors, which include integrins and syndecan-4 triggered by IGF-IR inhibition, regulate adhesion and invasion. Notably, the protective ability of IGF-IR is lost in ERα-knockdown cells highlighting the importance of E2-ERα signaling pathway in IGF-IR actions.Please click here for the full-text access. Afratis NA, Bouris P, Skandalis SS, Multhaupt HA, Couchman JR, Theocharis AD, Karamanos NK. IGF-IR cooperates with ERα to inhibit breast cancer cell aggressiveness by regulating the expression and localisation of ECM molecules. Sci Rep. (2017) 7:40138. doi: 10.1038/srep40138. www.ncbi.nlm.nih.gov/pubmed/28079144