Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
J Biol Chem ; 298(8): 102206, 2022 08.
Article in English | MEDLINE | ID: mdl-35772493

ABSTRACT

Mutations in the SLC35C1 gene encoding the Golgi GDP-fucose transporter are known to cause leukocyte adhesion deficiency II. However, improvement of fucosylation in leukocyte adhesion deficiency II patients treated with exogenous fucose suggests the existence of an SLC35C1-independent route of GDP-fucose transport, which remains a mystery. To investigate this phenomenon, we developed and characterized a human cell-based model deficient in SLC35C1 activity. The resulting cells were cultured in the presence/absence of exogenous fucose and mannose, followed by examination of fucosylation potential and nucleotide sugar levels. We found that cells displayed low but detectable levels of fucosylation in the absence of SLC35C1. Strikingly, we show that defects in fucosylation were almost completely reversed upon treatment with millimolar concentrations of fucose. Furthermore, we show that even if fucose was supplemented at nanomolar concentrations, it was still incorporated into glycans by these knockout cells. We also found that the SLC35C1-independent transport preferentially utilized GDP-fucose from the salvage pathway over the de novo biogenesis pathway as a source of this substrate. Taken together, our results imply that the Golgi systems of GDP-fucose transport discriminate between substrate pools obtained from different metabolic pathways, which suggests a functional connection between nucleotide sugar transporters and nucleotide sugar synthases.


Subject(s)
Fucose , Guanosine Diphosphate Fucose , Leukocyte-Adhesion Deficiency Syndrome/therapy , Fucose/metabolism , Golgi Apparatus/metabolism , Guanosine Diphosphate Fucose/metabolism , Humans , Membrane Transport Proteins/metabolism , Monosaccharide Transport Proteins/genetics , Monosaccharide Transport Proteins/metabolism , Polysaccharides/metabolism
2.
Cells ; 10(8)2021 07 21.
Article in English | MEDLINE | ID: mdl-34440617

ABSTRACT

Skin melanocytes reside on the basement membrane (BM), which is mainly composed of laminin, collagen type IV, and proteoglycans. For melanoma cells, in order to invade into the skin, melanocytes must cross the BM. It has been reported that changes in the composition of the BM accompany melanocytes tumorigenesis. Previously, we reported high gelsolin (GSN)-an actin-binding protein-levels in melanoma cell lines and GSN's importance for migration of A375 cells. Here we investigate whether melanoma cells migrate differently depending on the type of fibrous extracellular matrix protein. We obtained A375 melanoma cells deprived of GSN synthesis and tested their migratory properties on laminin, collagens type I and IV, fibronectin, and Matrigel, which resembles the skin's BM. We applied confocal and structured illuminated microscopy (SIM), gelatin degradation, and diverse motility assays to assess GSN's influence on parameters associated with cells' ability to protrude. We show that GSN is important for melanoma cell migration, predominantly on laminin, which is one of the main components of the skin's BM.


Subject(s)
Basement Membrane/metabolism , Cell Movement , Extracellular Matrix/metabolism , Gelsolin/metabolism , Melanoma/metabolism , Skin Neoplasms/metabolism , Tumor Microenvironment , Basement Membrane/pathology , Collagen Type I/metabolism , Collagen Type IV/metabolism , Extracellular Matrix/pathology , Fibronectins/metabolism , Gelsolin/genetics , Humans , Laminin/metabolism , Melanoma/genetics , Melanoma/pathology , Neoplasm Invasiveness , Podosomes/metabolism , Podosomes/pathology , Signal Transduction , Skin Neoplasms/genetics , Skin Neoplasms/pathology
3.
Cells ; 10(4)2021 03 31.
Article in English | MEDLINE | ID: mdl-33807338

ABSTRACT

Thymosin ß4 (Tß4) is a small, 44-amino acid polypeptide. It has been implicated in multiple processes, including cell movement, angiogenesis, and stemness. Previously, we reported that melanoma cell lines differ in Tß4 levels. Studies on stable clones with silenced TMSB4X expression showed that Tß4 impacted adhesion and epithelial-mesenchymal transition progression. Here, we show that the cells with silenced TMSB4X expression exhibited altered actin cytoskeleton's organization and subcellular relocalization of two intermediate filament proteins: Nestin and Vimentin. The rearrangement of the cell cytoskeleton resulted in changes in the cells' topology, height, and stiffness defined by Young's modulus. Simultaneously, only for some A375 clones with a lowered Tß4 level, we observed a decreased ability to initiate colony formation in soft agar, tumor formation in vivo, and alterations in Nanog's expression level transcription factor regulating stemness. Thus, we show for the first time that in A375 cells, biomechanical properties are not directly coupled to stemness features, and this cell line is phenotypically heterogeneous.


Subject(s)
Gene Silencing , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Thymosin/metabolism , Actin Cytoskeleton/metabolism , Biomarkers, Tumor/metabolism , Biomechanical Phenomena , Carcinogenesis/pathology , Cell Cycle , Cell Line, Tumor , Cell Proliferation , Cell Shape , Humans , Intermediate Filaments/metabolism , Melanoma/pathology , Models, Biological , Nestin/metabolism , Vimentin/metabolism
4.
Front Cell Dev Biol ; 7: 304, 2019.
Article in English | MEDLINE | ID: mdl-31921836

ABSTRACT

Thymosin ß4 (Tß4), a multifunctional 44-amino acid polypeptide and a member of actin-binding proteins (ABPs), plays an important role in developmental processes and wound healing. In recent years an increasing number of data has been published suggesting Tß4's involvement in tumorigenesis. However, Tß4's role in melanoma tumor development still remains to be elucidated. In our study we demonstrate that Tß4 is crucial for melanoma adhesion and invasion. For the purpose of our research we tested melanoma cell lines differing in invasive potential. Moreover, we applied shRNAs to silence TMSB4X (gene encoding Tß4) expression in a cell line with high TMSB4X expression. We found out that Tß4 is not only a component of focal adhesions (FAs) and interacts with several FAs components but also regulates FAs formation. We demonstrate that Tß4 level has an impact on FAs' number and morphology. Moreover, manipulation with TMSB4X expression resulted in changes in cells' motility on non-coated and MatrigelTM (resembling basement membrane composition)-coated surfaces and drastically decreased invasion abilities of the cells. Additionally, a correlation between Tß4 expression level and exhibition of mesenchymal-like [epithelial-mesenchymal transition (EMT)] features was discovered. Cells with lowered TMSB4X expression were less EMT-progressed than control cells. Summarizing, obtained results show that Tß4 by regulating melanoma cells' adhesion has an impact on motility features and EMT. Our study not only contributes to a better understanding of the processes underlying melanoma cells' capacity to create metastases but also highlights Tß4 as a potential target for melanoma management therapy.

5.
Eur J Cell Biol ; 95(1): 26-41, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26598132

ABSTRACT

Gelsolin, a multifunctional actin binding protein, plays a not yet fully understood role in tumorigenesis. Therefore the goal of this study was to identify additional molecular partners of gelsolin in human melanoma cells, separately in the cytoplasmic compartment and cell nuclei. For this purpose we performed immunoprecipitation experiments based on a modified protocol followed by mass spectrometry. The obtained results were confirmed by Western blot analysis, proximity ligation assays and confocal microscopy. As expected gelsolin interacted with actin, in particular we demonstrate its interaction with cytoplasmic ß and γ actins, and a newly discovered actin isoform, actbl2. As new gelsolin-interacting partners we identified the ribosomal protein Rpsa, also known as a non-integrin laminin receptor (LamR), and the heterogeneous nuclear ribonucleoprotein hnRNP U. Our data furthermore indicate that gelsolin interacts with particular components of the three cytoskeleton systems: nestin (intermediate filaments), Arp3 (actin cytoskeleton) and ß-tubulin (microtubules). We also report for the first time that gelsolin is a constituent of midbodies, a tubulin containing structure formed at the end of cytokinesis.


Subject(s)
Actin Cytoskeleton/metabolism , Cytoskeletal Proteins/metabolism , Gelsolin/metabolism , Melanoma/metabolism , Actin-Related Protein 3/metabolism , Blotting, Western , Cell Line, Tumor , Heterogeneous-Nuclear Ribonucleoprotein U/metabolism , Humans , Immunoprecipitation , Mass Spectrometry , Microscopy, Confocal , Nestin/metabolism , Receptors, Laminin/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Ribosomal Proteins , Transfection , Tubulin/metabolism
6.
Brain Struct Funct ; 221(1): 515-34, 2016 Jan.
Article in English | MEDLINE | ID: mdl-25352156

ABSTRACT

Gelsolin is one of the most intensively studied actin-binding proteins. However, in the literature comprehensive studies of GSN expression during development have not been performed yet in all model organisms. In zebrafish, gelsolin is a dorsalizing factor that modulates bone morphogenetic proteins signaling pathways, whereas knockout of the gelsolin coding gene, GSN is not lethal in murine model. To study the role of gelsolin in development of higher vertebrates, it is crucial to estimate GSN expression pattern during development. Here, we examined GSN expression in the developing chicken embryo. We applied numerous methods to track GSN expression in developing embryos at mRNA and protein level. We noted a characteristic GSN expression pattern. Although GSN transcripts were present in several cell types starting from early developmental stages, a relatively high GSN expression was observed in eye, brain vesicles, midbrain, neural tube, heart tube, and splanchnic mesoderm. In older embryos, we observed a high GSN expression in the cranial ganglia and dorsal root ganglia. A detailed analysis of 10-day-old chicken embryos revealed high amounts of gelsolin especially within the head region: in the olfactory and optic systems, meninges, nerves, muscles, presumptive pituitary gland, and pericytes, but not oligodendrocytes in the brain. Obtained results suggest that GSN is expressed at high levels in some tissues of ectodermal origin including all neural crest derivatives. Additionally, we describe that silencing of GSN expression in brain vesicles leads to altered morphology of the mesencephalon. This implies gelsolin is crucial for chicken brain development.


Subject(s)
Gelsolin/metabolism , Neural Crest/metabolism , Amino Acid Sequence , Animals , Blotting, Western , Brain/abnormalities , Brain/metabolism , Chick Embryo , Electroporation , Embryo, Nonmammalian/metabolism , Gelsolin/genetics , Gene Expression Regulation, Developmental , Gene Transfer Techniques , Molecular Sequence Data , RNA Interference , RNA, Messenger/genetics , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Time Factors
7.
Histochem Cell Biol ; 144(5): 417-28, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26239425

ABSTRACT

Actin is a highly conserved protein that is expressed in all eukaryotic cells and has essential functions in the cytoplasm and the nucleus. Nuclear actin is involved in transcription by all three RNA polymerases, chromatin remodelling, RNA processing, intranuclear transport, nuclear export and in maintenance of the nuclear architecture. The nuclear actin level and polymerization state are important factors regulating nuclear processes such as transcription. Our study shows that, in contrast to the cytoplasm, the majority of endogenous nuclear actin is unpolymerized in human melanoma A375 cells. Most mammalian cells express the two non-muscle ß- and γ-actin isoforms that differ in only four amino acids. Despite their sequence similarity, studies analysing the cytoplasmic functions of these isoforms demonstrated that ß- and γ-actins show differences in localization and function. However, little is known about the involvement of the individual actin isoforms in nuclear processes. Here, we used the human melanoma A375 cell line to analyse actin isoforms in regard to their nuclear localization. We show that both ß- and γ-non-muscle actin isoforms are present in nuclei of these cells. Immunolocalization studies demonstrate that both isoforms co-localize with RNA polymerase II and hnRNP U. However, we observe differences in the ratio of cytoplasmic to nuclear actin distribution between the isoforms. We show that ß-actin has a significantly higher nucleus-to-cytoplasm ratio than γ-actin.


Subject(s)
Actins/metabolism , Cell Nucleus/metabolism , Melanoma/pathology , Actins/analysis , Cell Nucleus/chemistry , Cell Nucleus/pathology , Humans , Melanoma/metabolism , Protein Isoforms/analysis , Protein Isoforms/metabolism , Tumor Cells, Cultured
8.
Eur J Cell Biol ; 94(2): 101-13, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25555464

ABSTRACT

The formin homology domain-containing protein1 (FHOD1) suppresses actin polymerization by inhibiting nucleation, but bundles actin filaments and caps filament barbed ends. Two polyclonal antibodies against FHOD1 were generated against (i) its N-terminal sequence (residues 1-339) and (ii) a peptide corresponding the sequence from position 358-371, which is unique for FHOD1 and does not occur in its close relative FHOD3. After affinity purification both antibodies specifically stain purified full length FHOD1 and a band of similar molecular mass in homogenates of cardiac muscle. The antibody against the N-terminus of FHOD1 was used for immunostaining cells of established lines, primary neonatal (NRC) and adult (ARC) rat cardiomyocytes and demonstrated the presence of FHOD1 in HeLa and fibroblastic cells along stress fibers and within presumed lamellipodia and actin arcs. In NRCs and ARCs we observed a prominent staining of presumed intercalated discs (ICD). Immunostaining of sections of hearts with both anti-FHOD1 antibodies confirmed the presence of FHOD1 in ICDs and double immunostaining demonstrated its colocalisation with cadherin, plakoglobin and a probably slightly shifted localization to connexin43. Similarly, immunostaining of isolated mouse or pig ICDs corroborated the presence of FHOD1 and its colocalisation with the mentioned cell junctional components. Anti-FHOD1 immunoblots of isolated ICDs demonstrated the presence of an immunoreactive band comigrating with purified FHOD1. Conversely, an anti-peptide antibody specific for FHOD3 with no cross-reactivity against FHOD1 immunostained on sections of cardiac muscle and ARCs the myofibrils in a cross-striated pattern but not the ICDs. In addition, the anti-peptide-FHOD1 antibody stained the lateral sarcolemma of ARCs in a banded pattern. Double immunostaining with anti-cadherin and -integrin-ß1 indicated the additional localization of FHOD1 in costameres. Immunostaining of cardiac muscle sections or ARCs with antibodies against mDia3-FH2-domain showed colocalisation with cadherin along the lateral border of cardiomyocytes suggesting also its presence in costameres.


Subject(s)
Costameres/metabolism , Fetal Proteins/metabolism , Myocardium/metabolism , Nuclear Proteins/metabolism , Animals , Animals, Newborn , Antibodies/metabolism , Cadherins/metabolism , Cell Line , Connexin 43/metabolism , Formins , Humans , Mice , Microfilament Proteins/metabolism , Myocardium/cytology , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/ultrastructure , Rats , Stress Fibers/metabolism , Swine , gamma Catenin/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...