Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
Respir Res ; 16: 86, 2015 Jul 15.
Article in English | MEDLINE | ID: mdl-26173671

ABSTRACT

BACKGROUND: Human lung mast cells (HLMCs) infiltrate the airway epithelium and airway smooth muscle (ASM) in asthmatic airways. The mechanism of HLMC adhesion to both cell types is only partly defined, and adhesion is not inhibited by function-blocking anti-Kit and anti-stem cell factor (SCF) antibodies. Our aim was to identify adhesion molecules expressed by human mast cells that mediate adhesion to human ASM cells (HASMCs) and human airway epithelial cells. METHODS: We used phage-display to isolate single chain Fv (scFv) antibodies with adhesion-blocking properties from rabbits immunised with HLMC and HMC-1 membrane proteins. RESULTS: Post-immune rabbit serum labelled HLMCs in flow cytometry and inhibited their adhesion to human BEAS-2B epithelial cells. Mast cell-specific scFvs were identified which labelled mast cells but not Jurkat cells by flow cytometry. Of these, one scFv (A1) consistently inhibited mast cell adhesion to HASMCs and BEAS-2B epithelial cells by about 30 %. A1 immunoprecipitated Kit (CD117) from HMC-1 lysates and bound to a human Kit-expressing mouse mast cell line, but did not interfere with SCF-dependent Kit signalling. CONCLUSION: Kit contributes to human mast cell adhesion to human airway epithelial cells and HASMCs, but may utilise a previously unidentified adhesion domain that lies outside the SCF binding site. Targeting this adhesion pathway might offer a novel approach for the inhibition of mast cell interactions with structural airway cells, without detrimental effects on Kit signalling in other tissues.


Subject(s)
Mast Cells/metabolism , Myocytes, Smooth Muscle/metabolism , Proto-Oncogene Proteins c-kit/metabolism , Respiratory Mucosa/metabolism , Amino Acid Sequence , Animals , Cell Adhesion/physiology , Cells, Cultured , Female , Humans , Mice , Molecular Sequence Data , Proto-Oncogene Proteins c-kit/genetics , Rabbits
2.
PLoS One ; 9(1): e85980, 2014.
Article in English | MEDLINE | ID: mdl-24465823

ABSTRACT

CADM1 is a major receptor for the adhesion of mast cells (MCs) to fibroblasts, human airway smooth muscle cells (HASMCs) and neurons. It also regulates E-cadherin and alpha6beta4 integrin in other cell types. Here we investigated a role for CADM1 in MC adhesion to both cells and extracellular matrix (ECM). Downregulation of CADM1 in the human MC line HMC-1 resulted not only in reduced adhesion to HASMCs, but also reduced adhesion to their ECM. Time-course studies in the presence of EDTA to inhibit integrins demonstrated that CADM1 provided fast initial adhesion to HASMCs and assisted with slower adhesion to ECM. CADM1 downregulation, but not antibody-dependent CADM1 inhibition, reduced MC adhesion to ECM, suggesting indirect regulation of ECM adhesion. To investigate potential mechanisms, phosphotyrosine signalling and polymerisation of actin filaments, essential for integrin-mediated adhesion, were examined. Modulation of CADM1 expression positively correlated with surface KIT levels and polymerisation of cortical F-actin in HMC-1 cells. It also influenced phosphotyrosine signalling and KIT tyrosine autophosphorylation. CADM1 accounted for 46% of surface KIT levels and 31% of F-actin in HMC-1 cells. CADM1 downregulation resulted in elongation of cortical actin filaments in both HMC-1 cells and human lung MCs and increased cell rigidity of HMC-1 cells. Collectively these data suggest that CADM1 is a key adhesion receptor, which regulates MC net adhesion, both directly through CADM1-dependent adhesion, and indirectly through the regulation of other adhesion receptors. The latter is likely to occur via docking of KIT and polymerisation of cortical F-actin. Here we propose a stepwise model of adhesion with CADM1 as a driving force for net MC adhesion.


Subject(s)
Actin Cytoskeleton/metabolism , Cell Adhesion Molecules/metabolism , Extracellular Matrix/metabolism , Immunoglobulins/metabolism , Mast Cells/metabolism , Actins/metabolism , Blotting, Western , Cell Adhesion/drug effects , Cell Adhesion Molecule-1 , Cell Adhesion Molecules/genetics , Cell Line , Cells, Cultured , Chelating Agents/pharmacology , Edetic Acid/pharmacology , Humans , Immunoglobulins/genetics , Mast Cells/cytology , Microscopy, Confocal , Microscopy, Fluorescence , Myocytes, Smooth Muscle/cytology , Myocytes, Smooth Muscle/metabolism , Phosphorylation , Polymerization , Proto-Oncogene Proteins c-kit/metabolism , RNA Interference , Respiratory System/cytology , Time Factors , Tyrosine/metabolism
3.
PLoS One ; 8(4): e61579, 2013.
Article in English | MEDLINE | ID: mdl-23620770

ABSTRACT

BACKGROUND: Mast cells (MCs) play a central role in the development of many diseases including asthma and pulmonary fibrosis. Interactions of human lung mast cells (HLMCs) with human airway smooth muscle cells (HASMCs) are partially dependent on adhesion mediated by cell adhesion molecule-1 (CADM1), but the adhesion mechanism through which HLMCs interact with human lung fibroblasts (HLFs) is not known. CADM1 is expressed as several isoforms (SP4, SP1, SP6) in HLMCs, with SP4 dominant. These isoforms differentially regulate HLMC homotypic adhesion and survival. OBJECTIVE: In this study we have investigated the role of CADM1 isoforms in the adhesion of HLMCs and HMC-1 cells to primary HASMCs and HLFs. METHODS: CADM1 overexpression or downregulation was achieved using adenoviral delivery of CADM1 short hairpin RNAs or isoform-specific cDNAs respectively. RESULTS: Downregulation of CADM1 attenuated both HLMC and HMC-1 adhesion to both primary HASMCs and HLFs. Overexpression of either SP1 or SP4 isoforms did not alter MC adhesion to HASMCs, whereas overexpression of SP4, but not SP1, significantly increased both HMC-1 cell and HLMC adhesion to HLFs. The expression level of CADM1 SP4 strongly predicted the extent of MC adhesion; linear regression indicated that CADM1 accounts for up to 67% and 32% of adhesion to HLFs for HMC-1 cells and HLMCs, respectively. HLFs supported HLMC proliferation and survival through a CADM1-dependent mechanism. With respect to CADM1 counter-receptor expression, HLFs expressed both CADM1 and nectin-3, whereas HASMCs expressed only nectin-3. CONCLUSION AND CLINICAL RELEVANCE: Collectively these data indicate that the CADM1 SP4 isoform is a key receptor mediating human MC adhesion to HASMCs and HLFs. The differential expression of CADM1 counter-receptors on HLFs compared to HASMCs may allow the specific targeting of either HLMC-HLF or HLMC-HASMC interactions in the lung parenchyma and airways.


Subject(s)
Cell Adhesion Molecules/metabolism , Fibroblasts/cytology , Fibroblasts/metabolism , Immunoglobulins/metabolism , Lung/cytology , Mast Cells/cytology , Myocytes, Smooth Muscle/cytology , Receptors, Cell Surface/metabolism , Adenoviridae/metabolism , Cell Adhesion , Cell Adhesion Molecule-1 , Cell Line , Cell Membrane/metabolism , Cell Proliferation , Cell Survival , Cells, Cultured , Down-Regulation , Humans , Myocytes, Smooth Muscle/metabolism , Protein Isoforms/metabolism , Regression Analysis , Transduction, Genetic
4.
Mol Immunol ; 53(4): 345-54, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23063768

ABSTRACT

Cell adhesion molecule 1 (CADM1) is implicated in the pathogenesis of several diseases and is responsible for adhesion and survival of mast cells (MCs). Differential expression of CADM1 isoforms was found in different species. We previously cloned SP4, SP1, SP6 and a dysfunctional isoform from human lung MCs (HLMCs) and the MC line HMC-1. The aim of this study was to identify all isoforms expressed in human MCs. The functional isoforms SP4, SP1, SP6 and SP3, with alternative splicing between exons 7/11, were detected in human MCs by RT-PCR. Two dysfunctional isoforms with alternative splicing of cryptic exons A and B between exons 1/2, leading to premature termination of translation, were found in ∼40% of MC specimens. Sequencing of genomic DNA showed that splicing of cryptic exon B did not result from specific SNPs within this exon or its putative splice branch point. Highly glycosylated CADM1 (∼105 kDa) was detected by western blotting, but an extracellular domain (∼95 kDa) was found only in the culture medium from HLMCs, but not HMC-1 cells, indicating differential protein expression. Transfection of SP1 and SP6, but not SP4, reduced adhesion of HMC-1 cells to human lung fibroblasts but not airway smooth muscle cells. Hence, dysfunctional and functional CADM1 isoforms are found in human MCs. The longer SP1 and SP6 were most evident in differentiated HLMCs and displayed differential adhesion compared to SP4. These multiple isoforms are likely to contribute to MC function in both health and disease.


Subject(s)
Alternative Splicing , Cell Adhesion Molecules/genetics , Gene Expression Regulation , Immunoglobulins/genetics , Lung/metabolism , Mast Cells/metabolism , Base Sequence , Cell Adhesion , Cell Adhesion Molecule-1 , Cell Adhesion Molecules/metabolism , Cell Differentiation , Cells, Cultured , Exons , Fibroblasts/cytology , Fibroblasts/metabolism , Glycosylation , Humans , Immunoglobulins/metabolism , Lung/cytology , Mast Cells/cytology , Molecular Sequence Data , Peptide Chain Termination, Translational , Protein Isoforms/genetics , Protein Isoforms/metabolism , Species Specificity , Transfection
5.
Cell Mol Life Sci ; 69(16): 2751-64, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22438059

ABSTRACT

Cell adhesion molecule 1 (CADM1), expressed by human lung mast cells (HLMCs), mediates their adhesion to airway smooth muscle (ASM), and contributes to ASM-dependent HLMC proliferation and survival. CADM1 is expressed in alternatively spliced isoforms, but those present in HLMCs and their function are not known. We cloned three functional and one cryptic non-functional isoform with alternative splicing between exons 7/11 and 1/2, respectively, from HLMCs and human MC lines (HMC-1 and LAD2). Differentiated HLMCs and LAD2 cells expressed the functional isoform SP4 containing exons 7/8/11 (~80% of clones), as well as SP1 (exons 7/8/9/11) and a novel SP6 (exons 7/8/9/10/11). In contrast, immature HMC-1 cells expressed only functional SP4. SP4 overexpression in HMC-1 cells and HLMCs augmented homotypic adhesion to a greater extent than SP1 in various conditions. In contrast, CADM1 downregulation abolished homotypic adhesion, indicating that CADM1 is the sole receptor mediating mast cell aggregation. CADM1-mediated adhesion was enhanced by the presence of cell survival factors. SP1 overexpression in HMC-1 cells compromised survival compared to SP4 overexpression or control. CADM1 downregulation resulted in reduced viability and decreased expression of the pro-survival protein Mcl-1(L), but not Blc-2 or Bcl-X(L), and increased caspase-3/7 activity in both HMC-1 cells and HLMCs. This coincided with decreased basal Kit levels in HLMCs. In summary, human MCs express multiple CADM1 isoforms which exhibit differential regulation of survival and homotypic adhesion. The most highly expressed SP4 isoform is likely to contribute to MC aggregation and longevity in mastocytosis, and augment the pathophysiology of allergic diseases.


Subject(s)
Cell Adhesion Molecules/metabolism , Cell Adhesion , Immunoglobulins/metabolism , Leukemia, Mast-Cell/pathology , Lung/cytology , Mast Cells/cytology , Mast-Cell Sarcoma/pathology , Blotting, Western , Cell Adhesion Molecule-1 , Cell Aggregation , Cell Survival , Cells, Cultured , Humans , Leukemia, Mast-Cell/metabolism , Lung/metabolism , Mast Cells/metabolism , Mast-Cell Sarcoma/metabolism , Protein Isoforms , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction
6.
Adv Exp Med Biol ; 716: 235-69, 2011.
Article in English | MEDLINE | ID: mdl-21713660

ABSTRACT

Mast cells play an important role in the lung in both health and disease. Their primary role is to initiate an appropriate program of inflammation and repair in response to tissue damage initiated by a variety of diverse stimuli. They are important for host immunity against bacterial infection and potentially in the host immune response to non small cell lung cancer. In situations of ongoing tissue damage, the sustained release of numerous pro-inflammatory mediators, proteases and cytokines, contributes to the pathophysiology of lung diseases such as asthma and interstitial lung disease. A key goal is the development of treatments which attenuate adverse mast cell function when administered chronically to humans in vivo. Such therapies may offer a novel approach to the treatment of many life-threatening diseases.


Subject(s)
Lung Diseases/immunology , Mast Cells/immunology , Animals , Humans
7.
Cancer Prev Res (Phila) ; 2(7): 611-6, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19584074

ABSTRACT

There is a large body of evidence that the consumption of fruit and vegetables can decrease the risk of cancer. However, the link between diet and health is extremely complex. Some dietary phytochemicals seem to offer protection in an exposure-related manner and many molecular targets and signaling pathways affected by phytochemicals have been discovered. Although in vitro studies have contributed significantly to our understanding, quite a number use concentrations orders of magnitude greater than those achievable in humans or toxic to normal tissues (exemplified by toxic concentrations of indole-3-carbinol, epigallocatechin-3-gallate, curcumin, and genistein for breast cells). Such studies may produce results that are physiologically irrelevant, thus hindering predictions of efficacy. Here, we argue for careful consideration to be given to the in vitro experimental conditions under which dietary phytochemicals are investigated. Design features, such as the use of appropriate nontoxic concentrations, extended treatment times, three-dimensional cultures, primary tumor cultures, and comparison of susceptibility of various cancer subtypes, should improve our understanding of their molecular targets. This in turn would facilitate predictions as to their potential usefulness in the clinic.


Subject(s)
Diet , Food , Neoplasms/prevention & control , Animals , Catechin/analogs & derivatives , Catechin/pharmacology , Cell Transformation, Neoplastic , Curcumin/pharmacology , Fruit , Genistein/pharmacology , Humans , Indoles/chemistry , Mice , Models, Biological , Phytotherapy/methods , Vegetables
8.
Breast Cancer Res Treat ; 109(3): 451-62, 2008 Jun.
Article in English | MEDLINE | ID: mdl-17653853

ABSTRACT

Indole-3-carbinol (I3C), a dietary chemopreventive compound, induces cell death in human breast cancer cells by modulating activities of Src and epidermal growth factor receptor (EGFR). The effect of I3C on NF-kappaB, constitutively activated in breast cancer cells, was investigated. Nuclear extracts of MDA-MB-468, MDA-MB-231 and HBL100 cells contained all of the Rel proteins with similar expression patterns in the latter two. The level of NF-kappaB-regulated reporter gene expression was in the order HBL100 << MDA-MB-468 << MDA-MB-231. Upstream inhibition, using PI3K, EGFR or IKKbeta inhibitors, resulted in cell-specific effects on expression of the NF-kappaB-regulated reporter gene and endogenous genes Bcl-xL, IkappaBalpha and IL-6, as well as on cell viability. The expression patterns of Rel and several NF-kappaB-regulated genes and the response to LY249002 in MDA-MB-468 cells contrasted with those in other cells. I3C induced NF-kappaB-regulated reporter gene expression at 12 h in MDA-MB-468 cells. Conversely, it was reduced at 24 h in HBL100 cells. I3C treatment for 6 h alone or in combination with TNFalpha induced NF-kappaB-regulated reporter gene expression, detected 5 h later, in MDA-MB-468, but not HBL100 cells. I3C induced NF-kappaB p65/p50 DNA binding at 6.5 h, preceded by association of IKKbeta with the Src/EGFR complex and increased phospho-IkappaBalpha in MDA-MB468 cells. TNFalpha increased I3C-induced apoptosis in MDA-MB-468 and MDA-MB-231 cells. It also induced apoptosis, enhanced by I3C, in HBL100 cells. Hence, regulation of constitutive NF-kappaB was cell-specific. I3C influenced the NF-kappaB pathway in a cell-specific manner, which was not related to apoptosis. However, the combination of I3C and TNFalpha increased apoptosis in all cell lines.


Subject(s)
Anticarcinogenic Agents/pharmacology , Apoptosis/drug effects , Breast Neoplasms/drug therapy , Indoles/pharmacology , NF-kappa B/metabolism , Signal Transduction/drug effects , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Survival/drug effects , Chromones/pharmacology , DNA/metabolism , Female , Humans , Morpholines/pharmacology , Tumor Necrosis Factor-alpha/pharmacology
9.
Mol Cancer Ther ; 6(11): 3071-9, 2007 Nov.
Article in English | MEDLINE | ID: mdl-18025290

ABSTRACT

Dietary phytochemicals exhibit chemopreventive potential in vivo through persistent low-dose exposures, whereas mechanistic in vitro studies with these agents generally use a high-dose single treatment. Because the latter approach is not representative of an in vivo steady state, we investigated antitumor activity of curcumin, 3,3'-diindolylmethane (DIM), epigallocatechin gallate (EGCG), genistein, or indole-3-carbinol (I3C) in breast cancer MDA-MB-231 cells, exposed in long-term culture to low concentrations, achievable in vivo. Curcumin and EGCG increased cell doubling time. Curcumin, EGCG, and I3C inhibited clonogenic growth by 55% to 60% and induced 1.5- to 2-fold higher levels of the basal caspase-3/7 activity. No changes in expression of cell cycle-related proteins or survivin were found; however, I3C reduced epidermal growth factor receptor expression, contributing to apoptosis. Because some phytochemicals are shown to inhibit DNA and histone modification, modulation of expression by the agents in a set of genes (cadherin-11, p21Cip1, urokinase-type plasminogen activator, and interleukin-6) was compared with changes induced by inhibitors of DNA methylation or histone deacetylation. The phytochemicals modified protein and/or RNA expression of these genes, with EGCG eliciting the least and DIM the most changes in gene expression. DIM and curcumin decreased cadherin-11 and increased urokinase-type plasminogen activator levels correlated with increased cell motility. Curcumin, DIM, EGCG, and genistein reduced cell sensitivity to radiation-induced DNA damage without affecting DNA repair. This model has revealed that apoptosis and not arrest is likely to be responsible for growth inhibition. It also implicated new molecular targets and activities of the agents under conditions relevant to human exposure.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Apoptosis/drug effects , Diet , Gene Expression Regulation, Neoplastic/drug effects , Neoplasms/pathology , Biomarkers, Tumor/metabolism , Catechin/analogs & derivatives , Catechin/pharmacology , Cell Cycle/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Curcumin/pharmacology , DNA Damage , Genistein/pharmacology , Humans , Indoles/pharmacology , Neoplasm Proteins/metabolism , RNA, Neoplasm/metabolism , Time Factors
10.
Acta Pharmacol Sin ; 28(9): 1274-304, 2007 Sep.
Article in English | MEDLINE | ID: mdl-17723163

ABSTRACT

There is growing interest in the ability of phytochemicals to prevent chronic diseases, such as cancer and heart disease. However, some of these agents have poor bioavailability and many of the in-depth studies into their mechanisms of action have been carried out in vitro using doses which are unachievable in humans. In order to optimize the design of chemopreventive treatment, it is important to determine which of the many reported mechanisms of action are clinically relevant. In this review we consider the physiologically achievable doses for a few of the best studied agents (indole-3-carbinol, diindolylmethane, curcumin, epigallocatechin-3-gallate and resveratrol) and summarize the data derived from studies using these low concentrations in cell culture. We then cite examples of in vitro effects which have been observed in vivo. Finally, the ability of agent combinations to act synergistically or antagonistically is considered. We conclude that each of the compounds shows an encouraging range of activities in vitro at concentrations which are likely to be physiologically relevant. There are also many examples of in vivo studies which validate in vitro observations. An important consideration is that combinations of agents can result in significant activity at concentrations where any single agent is inactive. Thus, for each of the compounds reviewed here, in vitro studies have provided useful insights into their mechanisms of action in humans. However, data are lacking on the full range of activities at low doses in vitro and the benefits or otherwise of combinations in vivo.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacokinetics , Catechin/analogs & derivatives , Curcumin/pharmacokinetics , Indoles/pharmacokinetics , Neoplasms/prevention & control , Stilbenes/pharmacokinetics , Animals , Antineoplastic Agents, Phytogenic/administration & dosage , Antineoplastic Agents, Phytogenic/therapeutic use , Antineoplastic Combined Chemotherapy Protocols , Catechin/administration & dosage , Catechin/pharmacokinetics , Catechin/therapeutic use , Curcumin/administration & dosage , Curcumin/therapeutic use , Humans , Indoles/administration & dosage , Indoles/therapeutic use , Resveratrol , Stilbenes/administration & dosage , Stilbenes/therapeutic use
11.
Carcinogenesis ; 28(2): 435-45, 2007 Feb.
Article in English | MEDLINE | ID: mdl-16956907

ABSTRACT

Indole-3-carbinol (I3C), a dietary chemopreventive compound, induced marked reduction in epidermal growth factor receptor (EGFR) prior to cell death in cells representing three breast cancer subtypes. Signalling pathways, linking these events were investigated in detail. I3C modulated tyrosine phosphorylation from 30 min in four cell lines. In MDA-MB-468 and HBL100 cells, it induced Src activation after 5 h. In MDA-MB-468 cells, I3C induced signalling between 4.5 and 7 h, which involved sequential activation of Src, EGFR, STAT-1 and STAT-3, followed by EGFR degradation. It also induced physical association between activated Src and EGFR. In MCF7 and MDA-MB-231 cells, I3C modulated expression of cell cycle-related proteins, p21Cip1, p27Kip1, cyclin E, cyclin D1 and CDK6, with upregulation of p21Cip1 and cyclin E being dependent on Src. Inhibition of EGFR by specific inhibitors PD153035 or ZD1839 increased susceptibility to I3C-induced apoptosis of MCF7, MDA-MB-468 and MDA-MB-231 cells. Inhibition of Src sensitized MDA-MB-468 and MDA-MB-231 cells to I3C, whereas overexpression of c-Src increased resistance to I3C in MDA-MB-468 and HBL100 cells. Modulation of Src in MDA-MB-468 cells influenced the basal level of EGFR expression and cell viability; the latter being positively correlated with EGFR activation levels. Therefore, EGFR and Src activities are essential for I3C-induced cell cycle arrest and death; however, I3C-induced pathways depend on specific features of breast cancer cells. The cancer types, which rely on 'EGFR addiction' or Src deregulation, are likely to be susceptible to I3C.


Subject(s)
Apoptosis/physiology , Breast Neoplasms/pathology , Cell Cycle/physiology , ErbB Receptors/physiology , Indoles/pharmacology , Proto-Oncogene Proteins pp60(c-src)/physiology , Apoptosis/drug effects , Cell Cycle/drug effects , Cell Cycle Proteins/physiology , Cell Line, Tumor , Humans , Phosphorylation , Tyrosine/metabolism
12.
Biochem Biophys Res Commun ; 310(3): 1010-6, 2003 Oct 24.
Article in English | MEDLINE | ID: mdl-14550305

ABSTRACT

Galectin-1, a beta-galactoside-binding dimeric lectin, is involved in adhesion, migration, and proliferation of vascular smooth muscle cells (SMC), the key steps in the development of atherosclerosis and restenosis. Here we investigated the molecular basis of the interactions between galectin-1 and SMCs. Galectin-1 modulated SMC attachment in a dose- and beta-galactoside-dependent manner. Direct binding of galectin-1 to beta1 integrin was detected by the immune precipitation of beta1 integrin after chemical cross-linking of 125I-labelled galectin-1 to the cell surface proteins. Galectin-1 transiently increased availability of beta1 integrins on the cell surface to antibodies against beta1 integrin. Incubation of SMCs with galectin-1 transiently increased the amount of the active form of beta1 integrin and tyrosine phosphorylation of two cytoskeleton-associated proteins; one of them coincided with focal adhesion kinase (FAK). Galectin-1 is likely to affect SMC adhesion by interacting with beta1 integrin on the cell surface of SMCs and inducing outside-in signalling.


Subject(s)
Galectin 1/metabolism , Integrin beta1/metabolism , Animals , Blotting, Western , Cell Adhesion , Cross-Linking Reagents/pharmacology , Cytoskeleton/metabolism , Flow Cytometry , Focal Adhesion Kinase 1 , Focal Adhesion Protein-Tyrosine Kinases , Galectin 1/chemistry , Humans , Integrin beta1/chemistry , Lectins/metabolism , Phosphorylation , Precipitin Tests , Protein Binding , Protein Structure, Tertiary , Protein-Tyrosine Kinases/metabolism , Rats , Signal Transduction , Time Factors , Tyrosine/metabolism
13.
J Bone Miner Res ; 18(2): 195-203, 2003 Feb.
Article in English | MEDLINE | ID: mdl-12568396

ABSTRACT

Prostate cancer cells metastasize to bone causing a predominantly osteosclerotic response. It has been shown that cells from the human prostate cancer cell line PC3 secrete factors that influence the behavior of osteoblast-like cells. Some of these factors with mitogenic activity have been found to be proteins with molecular weights between 20 and 30 kDa, but the identity of the osteoblastic mitogenic factor or factors produced by prostate cancer cells is still unknown. Therefore, the aim of this study was to characterize the protein profile of conditioned medium (CM) from PC3 cells in the molecular weight range from 5 to 30 kDa using proteome analysis. A protein profile of the CM from PC3 cells was performed by two-dimensional polyacrylamide gel electrophoresis (2D-PAGE). Thirty protein spots with molecular weights ranging from 5 to 30 kDa were analyzed by matrix assisted laser desorption/ionization time of flight mass spectrometry (MALDI-TOF MS). One of these spots was identified as galectin-1. We examined whether PC3 CM, recombinant galectin-1 alone, or combined with insulin-like growth factor-I (IGF-I) had any effects on the proliferation or differentiation of human bone marrow stromal (hBMS) cells. Furthermore, we tested whether adhesion of PC3 cells to plastic, laminin, fibronectin, and collagen type I was influenced by lactose, which inhibits galectin-1. Galectin-1 (1000 ng/ml) inhibited the proliferation of hBMS cells up to 70 +/- 12% (treated/control) of control in contrast to PC3 CM, which induced hBMS cell proliferation by 3-fold. This effect was abolished by IGF-I. PC3 CM and galectin-1 in concentrations of 10 and 1000 ng/ml increased the alkaline phosphatase (ALP) activity of hBMS cells up to 175 +/- 27%, 137 +/- 8%, and 131 +/- 11%, respectively, compared with ALP activity of untreated cells, and inhibited the secretion of osteocalcin (OC) up to 81 +/- 3%, 93 +/- 1%, and 58 +/- 2%, respectively, compared with OC secretion of untreated cells. These effects were affected by IGF-I. Lactose inhibited adhesion of PC3 cells to plastic, fibronectin, laminin, and collagen type I up to 58 +/- 4%, 30 +/- 12, 72 +/- 9%, and 86 +/- 4%. In conclusion, galectin-1 modulated osteoblastic proliferation and differentiation. These effects were affected by IGF-I. Thus, galectin-1 is likely be involved in the osteoblastic response, caused by prostate cancer cells metastasizing into bone, by affecting the matrix mineralization.


Subject(s)
Bone Marrow Cells/cytology , Bone Neoplasms/secondary , Galectin 1/physiology , Osteoblasts/metabolism , Prostatic Neoplasms/pathology , Proteome , Cell Adhesion , Cell Differentiation , Cell Division , Collagen/metabolism , Collagen Type I/metabolism , Electrophoresis, Gel, Two-Dimensional , Electrophoresis, Polyacrylamide Gel , Fibronectins/metabolism , Galectin 1/metabolism , Humans , Lactose/metabolism , Laminin/metabolism , Male , Neoplasm Metastasis , Peptide Mapping , Recombinant Proteins/metabolism , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Tumor Cells, Cultured
14.
Biochim Biophys Acta ; 1619(2): 125-32, 2003 Jan 20.
Article in English | MEDLINE | ID: mdl-12527107

ABSTRACT

Galectin-1, a beta-galactoside-binding dimeric lectin, interacts with the extracellular matrix (ECM) of smooth muscle cells (SMCs) and with particular ECM proteins. Enrichment of the ECM with galectin-1 affects adhesion and proliferation of cultured SMCs. Here we investigated whether galectin-1 (1) interacts with glycosaminoglycan (GAG) chains, (2) cross-links between ligands and facilitates the incorporation of GAGs, vitronectin and plasma fibronectin in the ECM of vascular SMCs. A recombinant galectin-1 fusion protein GalH, used in this study, formed dimers and interacted with ECM proteins. GAG chains inhibited these interactions. Among the studied GAG chains, only chondroitin sulfate B interacted with GalH in beta-galactoside-dependent manner. GalH did not bridge between ECM proteins on solid phase and [125I]-labelled ECM proteins or GAGs in solution. The ECM incorporated less vitronectin in the presence of soluble GalH. GalH-enriched ECM incorporated less vitronectin and chondroitin sulfate B. The ECM partially depleted of endogenous galectins incorporated more chondroitin sulfate B compared to untreated ECM. These results suggest that galectin-1 is likely to be involved in the ECM assembly affecting incorporation of some ECM components important for SMC behaviour.


Subject(s)
Dermatan Sulfate/metabolism , Extracellular Matrix/metabolism , Galectin 1/analogs & derivatives , Galectin 1/physiology , Muscle, Smooth, Vascular/metabolism , Vitronectin/metabolism , Cells, Cultured , Cross-Linking Reagents/chemistry , Dimerization , Galectin 1/chemistry , Galectin 1/pharmacology , Humans , Recombinant Fusion Proteins/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL
...