Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
Add more filters










Publication year range
1.
IUBMB Life ; 74(10): 969-981, 2022 10.
Article in English | MEDLINE | ID: mdl-35833571

ABSTRACT

Radiotherapy is widely used for the treatment of breast cancer. However, we have shown that ionizing radiation can provoke premature senescence in breast stromal cells. In particular, breast stromal fibroblasts can become senescent after irradiation both in vitro and in vivo and they express an inflammatory phenotype and an altered profile of extracellular matrix components, thus facilitating tumor progression. Adipose-derived stem cells (ASCs) represent another major component of the breast tissue stroma. They are multipotent cells and due to their ability to differentiate in multiple cell lineages they play an important role in tissue maintenance and repair in normal and pathologic conditions. Here, we investigated the characteristics of human breast ASCs that became senescent prematurely after their exposure to ionizing radiation. We found decreased expression levels of the specific mesenchymal cell surface markers CD105, CD73, CD44, and CD90. In parallel, we demonstrated a significantly reduced expression of transcription factors regulating osteogenic (i.e., RUNX2), adipogenic (i.e., PPARγ), and chondrogenic (i.e., SOX9) differentiation; this was followed by an analogous reduction in their differentiation capacity. Furthermore, they overexpress inflammatory markers, that is, IL-6, IL-8, and ICAM-1, and a catabolic phenotype, marked by the reduction of collagen type I and the increase of MMP-1 and MMP-13 expression. Finally, we detected changes in proteoglycan expression, for example, the upregulation of syndecan 1 and syndecan 4 and the downregulation of decorin. Notably, all these alterations, when observed in the breast stroma, represent poor prognostic factors for tumor development. In conclusion, we showed that ionizing radiation-mediated prematurely senescent human breast ASCs have a decreased differentiation potential and express specific changes adding to the formation of a permissive environment for tumor growth.


Subject(s)
Core Binding Factor Alpha 1 Subunit , Syndecan-1 , Adipose Tissue/metabolism , Cell Differentiation/physiology , Cells, Cultured , Collagen Type I , Core Binding Factor Alpha 1 Subunit/metabolism , Decorin/metabolism , Extracellular Matrix/genetics , Humans , Intercellular Adhesion Molecule-1/metabolism , Interleukin-6/metabolism , Interleukin-8/metabolism , Matrix Metalloproteinase 1/metabolism , Matrix Metalloproteinase 13/metabolism , PPAR gamma/metabolism , Stem Cells/metabolism , Syndecan-1/metabolism , Syndecan-4/metabolism
2.
Cancers (Basel) ; 13(19)2021 Sep 30.
Article in English | MEDLINE | ID: mdl-34638415

ABSTRACT

Lumican, a small leucine-rich proteoglycan (SLRP) of the extracellular matrix (ECM), displays anti-tumor properties through its direct interaction with MMP-14. Lumican-derived peptides, such as lumcorin (17 amino acids) or L9M (10 amino acids), are able to inhibit the proteolytic activity of MMP-14 and melanoma progression. This work aimed to visualize the interactions of lumican-derived peptides and MMP-14. Molecular modeling was used to characterize the interactions between lumican-derived peptides, such as lumcorin, L9M, and cyclic L9M (L9Mc, 12 amino acids), and MMP-14. The interaction of L9Mc with MMP-14 was preferential with the MT-Loop domain while lumcorin interacted more with the catalytic site. Key residues in the MMP-14 amino acid sequence were highlighted for the interaction between the inhibitory SLRP-derived peptides and MMP-14. In order to validate the in silico data, MMP-14 activity and migration assays were performed using murine B16F1 and human HT-144 melanoma cells. In contrast to the HT-144 melanoma cell line, L9Mc significantly inhibited the migration of B16F1 cells and the activity of MMP-14 but with less efficacy than lumican and lumcorin. L9Mc significantly inhibited the proliferation of B16F1 but not of HT-144 cells in vitro and primary melanoma tumor growth in vivo. Thus, the site of interaction between the domains of MMP-14 and lumcorin or L9Mc were different, which might explain the differences in the inhibitory effect of MMP-14 activity. Altogether, the biological assays validated the prediction of the in silico study. Possible and feasible improvements include molecular dynamics results.

3.
Int J Mol Sci ; 22(16)2021 Aug 05.
Article in English | MEDLINE | ID: mdl-34445148

ABSTRACT

The gram-positive pathogenic bacterium Clavibacter michiganensis subsp. michiganensis (Cmm) causes bacterial canker disease in tomato, affecting crop yield and fruit quality. To understand how tomato plants respond, the dynamic expression profile of host genes was analyzed upon Cmm infection. Symptoms of bacterial canker became evident from the third day. As the disease progressed, the bacterial population increased in planta, reaching the highest level at six days and remained constant till the twelfth day post inoculation. These two time points were selected for transcriptomics. A progressive down-regulation of key genes encoding for components of the photosynthetic apparatus was observed. Two temporally separated defense responses were observed, which were to an extent interdependent. During the primary response, genes of the phenylpropanoid pathway were diverted towards the synthesis of monolignols away from S-lignin. In dicots, lignin polymers mainly consist of G- and S-units, playing an important role in defense. The twist towards G-lignin enrichment is consistent with previous findings, highlighting a response to generate an early protective barrier and to achieve a tight interplay between lignin recomposition and the primary defense response mechanism. Upon progression of Cmm infection, the temporal deactivation of phenylpropanoids coincided with the upregulation of genes that belong in a secondary response mechanism, supporting an elegant reprogramming of the host transcriptome to establish a robust defense apparatus and suppress pathogen invasion. This high-throughput analysis reveals a dynamic reorganization of plant defense mechanisms upon bacterial infection to implement an array of barriers preventing pathogen invasion and spread.


Subject(s)
Down-Regulation/genetics , Photosynthesis/genetics , Plant Immunity/genetics , Plant Immunity/immunology , Solanum lycopersicum/genetics , Solanum lycopersicum/microbiology , Clavibacter/genetics , Gram-Positive Bacterial Infections/genetics , Gram-Positive Bacterial Infections/immunology , Gram-Positive Bacterial Infections/microbiology , Host-Pathogen Interactions/genetics , Host-Pathogen Interactions/immunology , Photosynthesis/immunology , Plant Diseases/genetics , Plant Diseases/immunology , Plant Diseases/microbiology , Transcriptome/genetics , Up-Regulation/genetics
4.
Molecules ; 26(14)2021 Jul 17.
Article in English | MEDLINE | ID: mdl-34299608

ABSTRACT

The wall is the last frontier of a plant cell involved in modulating growth, development and defense against biotic stresses. Cellulose and additional polysaccharides of plant cell walls are the most abundant biopolymers on earth, having increased in economic value and thereby attracted significant interest in biotechnology. Cellulose biosynthesis constitutes a highly complicated process relying on the formation of cellulose synthase complexes. Cellulose synthase (CesA) and Cellulose synthase-like (Csl) genes encode enzymes that synthesize cellulose and most hemicellulosic polysaccharides. Arabidopsis and rice are invaluable genetic models and reliable representatives of land plants to comprehend cell wall synthesis. During the past two decades, enormous research progress has been made to understand the mechanisms of cellulose synthesis and construction of the plant cell wall. A plethora of cesa and csl mutants have been characterized, providing functional insights into individual protein isoforms. Recent structural studies have uncovered the mode of CesA assembly and the dynamics of cellulose production. Genetics and structural biology have generated new knowledge and have accelerated the pace of discovery in this field, ultimately opening perspectives towards cellulose synthesis manipulation. This review provides an overview of the major breakthroughs gathering previous and recent genetic and structural advancements, focusing on the function of CesA and Csl catalytic domain in plants.


Subject(s)
Glucosyltransferases/metabolism , Plant Proteins/metabolism , Plants/metabolism , Catalytic Domain , Glucosyltransferases/chemistry , Glucosyltransferases/genetics , Models, Molecular , Mutation , Phylogeny , Plant Proteins/chemistry , Plant Proteins/genetics , Plants/chemistry , Plants/genetics
5.
Cells ; 10(4)2021 04 08.
Article in English | MEDLINE | ID: mdl-33917849

ABSTRACT

It was reported that lumican inhibits the activity of metalloproteinase MMP-14 and melanoma cell migration in vitro and in vivo. Moreover, Snail triggers epithelial-to-mesenchymal transition and the metastatic potential of cancer cells. Therefore, the aim of this study was to examine the effect of lumican on Mock and Snail overexpressing melanoma B16F1 cells in vivo. Lung metastasis was analyzed after intravenous injections of Mock-B16F1 and Snail-B16F1 cells in Lum+/+ and Lum-/- mice. At day 14, mice were sacrificed, and lungs were collected. The number of lung metastatic nodules was significantly higher in mice injected with Snail-B16F1 cells as compared to mice injected with Mock-B16F1 cells confirming the pro-metastatic effect of Snail. This effect was stronger in Lum-/- mice as compared to Lum+/+, suggesting that endogenous lumican of wild-type mice significantly inhibits metastasis to lungs. Scanning electron and confocal microscopy investigations demonstrated that lumican inhibits the development of elongated cancer cell phenotypes which are known to develop invadopodia releasing MMPs. Moreover, lumican was shown to affect the expression of cyclin D1, cortactin, vinculin, hyaluronan synthase 2, heparanase, MMP-14 and the phosphorylation of FAK, AKT, p130 Cas and GSK3α/ß. Altogether, these data demonstrated that lumican significantly inhibits lung metastasis in vivo, as well as cell invasion in vitro, suggesting that a lumican-based strategy targeting Snail-induced metastasis could be useful for melanoma treatment.


Subject(s)
Biomarkers, Tumor/metabolism , Lumican/metabolism , Melanoma/pathology , Podosomes/pathology , Skin Neoplasms/pathology , Animals , Cell Cycle , Cell Line, Tumor , Cell Movement , Cell Proliferation , Cell Shape , Cortactin/metabolism , Cyclin D1/metabolism , Extracellular Matrix/metabolism , Focal Adhesions/metabolism , Humans , Hyaluronic Acid/metabolism , Lung Neoplasms/secondary , Melanoma/metabolism , Melanoma/ultrastructure , Mice, Inbred C57BL , Neoplasm Metastasis , Phosphorylation , Signal Transduction , Skin Neoplasms/metabolism , Skin Neoplasms/ultrastructure , Snail Family Transcription Factors/metabolism , Vinculin/metabolism
6.
FEBS J ; 287(22): 4862-4880, 2020 11.
Article in English | MEDLINE | ID: mdl-32160387

ABSTRACT

The small leucine-rich proteoglycan lumican regulates estrogen receptors (ERs)-associated functional properties of breast cancer cells, expression of matrix macromolecules, and epithelial-to-mesenchymal transition. However, it is not known whether the ER-dependent lumican effects on breast cancer cells are related to the expression of integrins and their intracellular signaling pathways. Here, we analyzed the effects of lumican in three breast cancer cell lines: the highly metastatic ERß-positive MDA-MB-231, cells with the respective ERß-suppressed (shERßMDA-MB-231), and lowly invasive ERα-positive MCF-7/c breast cancer cells. Scanning electron microscopy, confocal microscopy, real-time PCR, western blot, and cell adhesion assays were performed. Lumican effects on breast cancer cell morphology were also investigated in 3-dimensional collagen cultures. Lumican treatment induced cell-cell contacts and cell grouping and inhibited microvesicles and microvilli formation. The expression of the cell surface adhesion receptor CD44, its isoform and variants, hyaluronan (HA), and HA synthases was also investigated. Lumican inhibited the expression of CD44 and HA synthases, and its effect on cell adhesion revealed a major role of α1, α2, α3, αVß3, and αVß5 integrins in MDA-MB-231 cells, but not in MCF-7/c cells. Lumican upregulated the expression of α2 and ß1 integrin subunits both in MDA-MB-231 and in shERßMDA-MB-231 as compared to MCF-7/c cells. Downstream signaling pathways for integrins, such as FAK, ERK 1/2 MAPK 42/44, and Akt, were found to be downregulated by lumican. Our data shed light to the molecular mechanisms responsible for the anticancer activity of lumican in invasive breast cancer.


Subject(s)
Breast Neoplasms/metabolism , Cell Shape/drug effects , Cortactin/metabolism , Integrins/metabolism , Lumican/pharmacology , Signal Transduction/drug effects , Breast Neoplasms/pathology , Breast Neoplasms/ultrastructure , Cell Adhesion/drug effects , Cell Line, Tumor , Cell Movement/drug effects , Focal Adhesion Kinase 1/metabolism , Humans , Hyaluronan Receptors/metabolism , Hyaluronan Synthases/metabolism , MCF-7 Cells , Microscopy, Electron, Scanning , Phosphorylation/drug effects
7.
Semin Cancer Biol ; 62: 125-133, 2020 05.
Article in English | MEDLINE | ID: mdl-31401293

ABSTRACT

A great hallmark of breast cancer is the absence or presence of estrogen receptors ERα and ERß, with a dominant role in cell proliferation, differentiation and cancer progression. Both receptors are related with Epithelial-to-Mesenchymal Transition (EMT) since there is a relation between ERs and extracellular matrix (ECM) macromolecules expression, and therefore, cell-cell and cell-ECM interactions. The endocrine resistance of ERα endows epithelial cells with increased aggressiveness and induces cell proliferation, resulting into a mesenchymal phenotype and an EMT status. ERα signaling may affect the transcriptional factors which govern EMT. Knockdown or silencing of ERα and ERß in MCF-7 and MDA-MB-231 breast cancer cells respectively, provoked pivotal changes in phenotype, cellular functions, mRNA and protein levels of EMT markers, and consequently the EMT status. Mesenchymal cells owe their migratory and invasive properties to invadopodia, while in epithelial cells, lamellipodia and filopodia are mostly observed. Invadopodia, are actin-rich protrusions of plasma membrane, promoting proteolytic degradation of ECM and tumor invasion. Cortactin and MMP-14 govern the formation and principal functions of invadopodia. In vitro experiments proved that lumican inhibits cortactin and MMP-14 expression, alters the formation of lamellipodia and transforms mesenchymal cells into epithelial-like. Conclusively, lumican may inhibit or even reverse the several metastatic features that EMT endows in breast cancer cells. Therefore, a lumican-based anti-cancer therapy which will pharmacologically target and inhibit EMT might be interesting to be developed.


Subject(s)
Biomarkers, Tumor , Breast Neoplasms/etiology , Breast Neoplasms/metabolism , Epithelial-Mesenchymal Transition , Podosomes/metabolism , Animals , Breast Neoplasms/pathology , Breast Neoplasms/therapy , Epithelial-Mesenchymal Transition/genetics , Extracellular Matrix/metabolism , Female , Gene Expression Regulation, Neoplastic , Humans , Lumican/genetics , Lumican/metabolism , Molecular Targeted Therapy , Signal Transduction
8.
Matrix Biol ; 75-76: 271-285, 2019 01.
Article in English | MEDLINE | ID: mdl-29253518

ABSTRACT

Small leucine-rich proteoglycans (SLRPs) are important regulators of extracellular matrix assembly and cell signaling. They are a family of proteoglycans that are present in extracellular matrix and that share in common multiple repeats of a leucine-rich structural motif. SLRPs have been identified as inhibitors of cancer progression by affecting MMPs, especially MMP-14 activity. Lumican, a member of the SLRPs family, and its derived peptides were shown to possess anti-tumor activity. Interestingly, it was demonstrated recently that lumican interacts directly with the catalytic domain of MMP-14 and inhibits its activity. The aim of this review was to summarize the interactions between SLRPs and MMPs with a special interest to lumican.


Subject(s)
Lumican/genetics , Matrix Metalloproteinase 14/genetics , Neoplasms/genetics , Small Leucine-Rich Proteoglycans/genetics , Catalytic Domain/genetics , Disease Progression , Extracellular Matrix/genetics , Extracellular Matrix/metabolism , Humans , Matrix Metalloproteinase 14/metabolism , Neoplasms/pathology , Signal Transduction/genetics
9.
Adv Drug Deliv Rev ; 129: 344-351, 2018 04.
Article in English | MEDLINE | ID: mdl-29501701

ABSTRACT

Wound healing, a complex physiological process, is responsible for tissue repair after exposure to destructive stimuli, without resulting in complete functional regeneration. Injuries can be stromal or epithelial, and most cases of wound repair have been studied in the skin and cornea. Lumican, a small leucine-rich proteoglycan, is expressed in the extracellular matrices of several tissues, such as the cornea, cartilage, and skin. This molecule has been shown to regulate collagen fibrillogenesis, keratinocyte phenotypes, and corneal transparency modulation. Lumican is also involved in the extravasation of inflammatory cells and angiogenesis, which are both critical in stromal wound healing. Lumican is the only member of the small leucine-rich proteoglycan family expressed by the epithelia during wound healing. This review summarizes the importance of lumican in wound healing and potential methods of lumican drug delivery to target wound repair are discussed. The involvement of lumican in corneal wound healing is described based on in vitro and in vivo models, with critical emphasis on its underlying mechanisms of action. Similarly, the expression and role of lumican in the healing of other tissues are presented, with emphasis on skin wound healing. Overall, lumican promotes normal wound repair and broadens new therapeutic perspectives for impaired wound healing.


Subject(s)
Lumican/pharmacology , Wound Healing/drug effects , Animals , Drug Delivery Systems , Humans
10.
Methods Mol Biol ; 1731: 325-348, 2018.
Article in English | MEDLINE | ID: mdl-29318564

ABSTRACT

Matrix metalloproteinases (MMPs) are a family of zinc-dependent endopeptidases that are capable of degrading numerous extracellular matrix (ECM) components thus participating in physiological and pathological processes. Apart from the remodeling of ECM, they affect cell-cell and cell-matrix interactions and are implicated in the development and progression of various diseases such as cancer. Numerous studies have demonstrated that MMPs evoke epithelial to mesenchymal transition (EMT) of cancer cells and affect their signaling, adhesion, migration and invasion to promote cancer cell aggressiveness. Various studies have suggested MMPs as suitable targets for treatment of malignancies, and several MMP inhibitors (MMPIs) have been developed. Although initial trials have failed to establish MMPIs as anticancer agents due to lack of specificity and side effects, new MMPIs have been developed with improved action that are currently being investigated. Furthermore, novel strategies that target MMPs for improving drug delivery and regulating their activity in tumors are presented. This review summarizes the implication of MMPs in cancer progression and discusses the advancements in their targeting.


Subject(s)
Antineoplastic Agents/pharmacology , Epithelial-Mesenchymal Transition/drug effects , Matrix Metalloproteinase Inhibitors/pharmacology , Matrix Metalloproteinases/metabolism , Neoplasms/drug therapy , Animals , Clinical Trials as Topic , Disease Progression , Drug Delivery Systems/methods , Enzyme Activation/drug effects , Extracellular Matrix/drug effects , Extracellular Matrix/pathology , Gene Expression Regulation, Neoplastic/drug effects , Humans , Matrix Metalloproteinases/genetics , MicroRNAs/metabolism , Molecular Targeted Therapy/methods , Neoplasms/genetics , Neoplasms/pathology , Protein Domains/drug effects
11.
Sci Rep ; 7: 45138, 2017 03 23.
Article in English | MEDLINE | ID: mdl-28332606

ABSTRACT

Lumican is a small leucine-rich proteoglycan that has been shown to contribute in several physiological processes, but also to exert anticancer activity. On the other hand, it has been recently shown that knockdown of the estrogen receptor α (ERα) in low invasive MCF-7 (ERα+) breast cancer cells and the suppression of ERß in highly aggressive MDA-MB-231 (ERß+) cells significantly alter the functional properties of breast cancer cells and the gene expression profile of matrix macromolecules related to cancer progression and cell morphology. In this report, we evaluated the effects of lumican in respect to the ERs-associated breast cancer cell behaviour, before and after suppression of ERs, using scanning electron and confocal microscopies, qPCR and functional assays. Our data pinpointed that lumican significantly attenuated cell functional properties, including proliferation, migration and invasion. Furthermore, it modified cell morphology, inducing cell-cell junctions, evoked EMT/MET reprogramming and suppressed the expression of major matrix effectors (matrix metalloproteinases and EGFR) implicated in breast cancer progression. The effects of lumican were found to be related to the type of breast cancer cells and the ERα/ß type. These data support the anticancer activity of lumican and open a new area for the pharmacological targeting of the invasive breast cancer.


Subject(s)
Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Epithelial-Mesenchymal Transition/drug effects , Epithelial-Mesenchymal Transition/genetics , Extracellular Matrix/metabolism , Lumican/pharmacology , Receptors, Estrogen/metabolism , Cell Line, Tumor , Cellular Reprogramming/genetics , Female , Gene Expression Profiling , Gene Knockout Techniques , Humans , MCF-7 Cells , Proto-Oncogene Proteins c-met/genetics , Proto-Oncogene Proteins c-met/metabolism , RNA, Small Interfering/genetics
12.
Glycoconj J ; 34(3): 277-283, 2017 06.
Article in English | MEDLINE | ID: mdl-27614617

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 â†’ 3-ß-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 â†’ 4-α-D-GlcN(SO3)-1 (6-O-SO3)-1]n and [4-α-L-IdoA-(2-O-SO3)-1 â†’ 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.


Subject(s)
Animal Structures/physiology , Dermatan Sulfate/chemistry , Disaccharides/chemistry , Phylogeny , Urochordata/physiology , Animal Structures/anatomy & histology , Animal Structures/chemistry , Animals , Carbohydrate Conformation , Carbohydrate Sequence , Collagen/chemistry , Decorin/chemistry , Dermatan Sulfate/isolation & purification , Disaccharides/isolation & purification , Extracellular Matrix/chemistry , Extracellular Matrix/physiology , Hemolymph/chemistry , Hemolymph/physiology , Urochordata/anatomy & histology , Urochordata/chemistry , Urochordata/classification
13.
Glycoconj J ; 34(3): 299-307, 2017 06.
Article in English | MEDLINE | ID: mdl-27778131

ABSTRACT

Glycosaminoglycans are integral part of the dynamic extracellular matrix (ECM) network that control crucial biochemical and biomechanical signals required for tissue morphogenesis, differentiation, homeostasis and cancer development. Breast cancer cells communicate with stromal ones to modulate ECM mainly through release of soluble effectors during cancer progression. The intracellular cross-talk between cell surface receptors and estrogen receptors is important for the regulation of breast cancer cell properties and production of ECM molecules. In turn, reorganized ECM-cell surface interface modulates signaling cascades, which regulate almost all aspects of breast cell behavior. Heparan sulfate chains present on cell surface and matrix proteoglycans are involved in regulation of breast cancer functions since they are capable of binding numerous matrix molecules, growth factors and inflammatory mediators thus modulating their signaling. In addition to its anticoagulant activity, there is accumulating evidence highlighting various anticancer activities of heparin and nano-heparin derivatives in numerous types of cancer. Importantly, heparin derivatives significantly reduce breast cancer cell proliferation and metastasis in vitro and in vivo models as well as regulates the expression profile of major ECM macromolecules, providing strong evidence for therapeutic targeting. Nano-formulations of the glycosaminoglycan heparin are possibly novel tools for targeting tumor microenvironment. In this review, the role of heparan sulfate/heparin and its nano-formulations in breast cancer biology are presented and discussed in terms of future pharmacological targeting.


Subject(s)
Antineoplastic Agents/therapeutic use , Breast Neoplasms/drug therapy , Gene Expression Regulation, Neoplastic , Heparin/chemistry , Heparitin Sulfate/therapeutic use , Animals , Antineoplastic Agents/chemistry , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Communication/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Cytokines/genetics , Cytokines/metabolism , Extracellular Matrix/chemistry , Extracellular Matrix/metabolism , Female , Heparin/therapeutic use , Heparitin Sulfate/chemistry , Humans , Intercellular Signaling Peptides and Proteins/genetics , Intercellular Signaling Peptides and Proteins/metabolism , Protein Binding/drug effects , Signal Transduction , Stromal Cells/drug effects , Stromal Cells/metabolism , Stromal Cells/pathology
14.
Arch Toxicol ; 91(3): 1031-1048, 2017 Mar.
Article in English | MEDLINE | ID: mdl-27438349

ABSTRACT

With the expansion of the nanomedicine field, the knowledge focusing on the behavior of nanoparticles in the biological milieu has rapidly escalated. Upon introduction to a complex biological system, nanomaterials dynamically interact with all the encountered biomolecules and form the protein "bio-corona." The decoration with these surface biomolecules endows nanoparticles with new properties. The present review will address updates of the protein bio-corona characteristics as influenced by nanoparticle's physicochemical properties and by the particularities of the encountered biological milieu. Undeniably, bio-corona generation influences the efficacy of the nanodrug and guides the actions of innate and adaptive immunity. Exploiting the dynamic process of protein bio-corona development in combination with the new engineered horizons of drugs linked to nanoparticles could lead to innovative functional nanotherapies. Therefore, bio-medical nanotechnologies should focus on the interactions of nanoparticles with the immune system for both safety and efficacy reasons.


Subject(s)
Nanomedicine/methods , Nanostructures/chemistry , Nanostructures/toxicity , Protein Corona/immunology , Adaptive Immunity/drug effects , Animals , Humans , Hydrophobic and Hydrophilic Interactions , Immunity, Innate/drug effects , Particle Size , Protein Corona/chemistry , Protein Corona/metabolism
15.
PLoS One ; 11(3): e0150226, 2016.
Article in English | MEDLINE | ID: mdl-26930497

ABSTRACT

Lumican, a small leucine rich proteoglycan, inhibits MMP-14 activity and melanoma cell migration in vitro and in vivo. Snail triggers epithelial-mesenchymal transitions endowing epithelial cells with migratory and invasive properties during tumor progression. The aim of this work was to investigate lumican effects on MMP-14 activity and migration of Snail overexpressing B16F1 (Snail-B16F1) melanoma cells and HT-29 colon adenocarcinoma cells. Lumican inhibits the Snail induced MMP-14 activity in B16F1 but not in HT-29 cells. In Snail-B16F1 cells, lumican inhibits migration, growth, and melanoma primary tumor development. A lumican-based strategy targeting Snail-induced MMP-14 activity might be useful for melanoma treatment.


Subject(s)
Cell Movement/drug effects , Chondroitin Sulfate Proteoglycans/pharmacology , Keratan Sulfate/pharmacology , Matrix Metalloproteinase 14/metabolism , Melanoma/metabolism , Transcription Factors/metabolism , Cell Line, Tumor , Cell Movement/physiology , HT29 Cells , Humans , Lumican , Melanoma/pathology , Snail Family Transcription Factors
16.
Food Chem Toxicol ; 91: 42-57, 2016 May.
Article in English | MEDLINE | ID: mdl-26969113

ABSTRACT

Nanotechnology is an evolving scientific field that has allowed the manufacturing of materials with novel physicochemical and biological properties, offering a wide spectrum of potential applications. Properties of nanoparticles that contribute to their usefulness include their markedly increased surface area in relation to mass, surface reactivity and insolubility, ability to agglomerate or change size in different media and enhanced endurance over conventional-scale substance. Here, we review nanoparticle classification and their emerging applications in several fields; from active food packaging to drug delivery and cancer research. Nanotechnology has exciting therapeutic applications, including novel drug delivery for the treatment of cancer. Additionally, we discuss that exposure to nanostructures incorporated to polymer composites, may result in potential human health risks. Therefore, the knowledge of processes, including absorption, distribution, metabolism and excretion, as well as careful toxicological assessment is critical in order to determine the effects of nanomaterials in humans and other biological systems. Expanding the knowledge of nanoparticle toxicity will facilitate designing of safer nanocomposites and their application in a beneficial manner.


Subject(s)
Agriculture , Food Industry , Nanotechnology , Neoplasms/therapy , Toxicology , Humans
17.
Toxicol Lett ; 240(1): 32-42, 2016 Jan 05.
Article in English | MEDLINE | ID: mdl-26476401

ABSTRACT

The glycosaminoglycan heparin and its derivatives act strongly on blood coagulation, controlling the activity of serine protease inhibitors in plasma. Nonetheless, there is accumulating evidence highlighting different anticancer activities of these molecules in numerous types of cancer. Nano-heparins may have great biological significance since they can inhibit cell proliferation and invasion as well as inhibiting proteasome activation. Moreover, they can cause alterations in the expression of major modulators of the tumor microenvironment, regulating cancer cell behavior. In the present study, we evaluated the effects of two nano-heparin formulations: one isolated from porcine intestine and the other from the sea squirt Styela plicata, on a breast cancer cell model. We determined whether these nano-heparins are able to affect cell proliferation, apoptosis and invasion, as well as proteasome activity and the expression of extracellular matrix molecules. Specifically, we observed that nano-Styela compared to nano-Mammalian analogue has higher inhibitory role on cell proliferation, invasion and proteasome activity. Moreover, nano-Styela regulates cell apoptosis, expression of inflammatory molecules, such as IL-6 and IL-8 and reduces the expression levels of extracellular matrix macromolecules, such as the proteolytic enzymes MT1-MMP, uPA and the cell surface proteoglycans syndecan-1 and -2, but not on syndecan-4. The observations reported in the present article indicate that nano-heparins and especially ascidian heparin are effective agents for heparin-induced effects in critical cancer cell functions, providing an important possibility in pharmacological targeting.


Subject(s)
Heparin/toxicity , Nanostructures/chemistry , Proteasome Inhibitors/toxicity , Apoptosis/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Extracellular Matrix/chemistry , Humans , Interleukin-6/metabolism , Interleukin-8/metabolism , Proteasome Endopeptidase Complex/metabolism
18.
Matrix Biol ; 43: 42-60, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25728938

ABSTRACT

The 17ß-estradiol (E2)/estrogen receptor alpha (ERα) signaling pathway is one of the most important pathways in hormone-dependent breast cancer. E2 plays pivotal roles in cancer cell growth, survival, and architecture as well as in gene expression regulatory mechanisms. In this study, we established stably transfected MCF-7 cells by knocking down the ERα gene (designated as MCF-7/SP10+ cells), using specific shRNA lentiviral particles, and compared them with the control cells (MCF-7/c). Interestingly, ERα silencing in MCF-7 cells strongly induced cellular phenotypic changes accompanied by significant changes in gene and protein expression of several markers typical of epithelial to mesenchymal transition (EMT). Notably, these cells exhibited enhanced cell proliferation, migration and invasion. Moreover, ERα suppression strongly affected the gene and protein expression of EGFR and HER2 receptor tyrosine kinases, and various extracellular matrix (ECM) effectors, including matrix metalloproteinases and their endogenous inhibitors (MMPs/TIMPs) and components of the plasminogen activation system. The action caused by E2 in MCF-7/c cells in the expression of HER2, MT1-MMP, MMP1, MMP9, uPA, tPA, and PAI-1 was abolished in MCF-7/SP10+ cells lacking ERα. These data suggested a regulatory role for the E2/ERα pathway in respect to the composition and activity of the extracellular proteolytic molecular network. Notably, loss of ERα promoted breast cancer cell migration and invasion by inducing changes in the expression levels of certain matrix macromolecules (especially uPA, tPA, PAI-1) through the EGFR-ERK signaling pathway. In conclusion, loss of ERα in breast cancer cells results in a potent EMT characterized by striking changes in the expression profile of specific matrix macromolecules highlighting the potential nodal role of matrix effectors in breast cancer endocrine resistance.


Subject(s)
Breast Neoplasms/pathology , Epithelial-Mesenchymal Transition , Estrogen Receptor alpha/genetics , Extracellular Matrix/metabolism , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Cell Movement , Cell Proliferation , Estrogen Receptor alpha/metabolism , Extracellular Matrix/genetics , Female , Gene Expression Regulation, Neoplastic , Gene Knockdown Techniques , Humans , MCF-7 Cells
19.
FEBS J ; 281(22): 4980-92, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25040101

ABSTRACT

Cell behavior is determined by both genetic and environmental factors. The cell microenvironment is not only a scene in which various actors play a role, but is itself an active participant, able to influence many cellular responses by binding signaling molecules or by modulating intracellular signaling cascades. Further, extracellular matrix remodeling is a critical step to allow physiological as well as pathological processes. As environmental factors are able to modulate gene expression by epigenetic modifications, this review focuses on new aspects of the regulation of extracellular matrix remodeling enzymes. Moreover, as one of the main components of cell microenvironment is the glycosaminoglycan hyaluronan, novel findings regarding the control of hyaluronan synthesis are discussed in terms of epigenetics and from the post-translational point of view.


Subject(s)
Epigenesis, Genetic , Extracellular Matrix/metabolism , Hyaluronic Acid/metabolism , Animals , Diabetes Mellitus/metabolism , Diabetes Mellitus/pathology , Extracellular Matrix/genetics , Glucuronosyltransferase/metabolism , Histones/metabolism , Humans , Hyaluronan Synthases , Hyaluronic Acid/genetics , MicroRNAs/physiology , Protein Processing, Post-Translational
SELECTION OF CITATIONS
SEARCH DETAIL
...