Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 31
Filter
Add more filters










Publication year range
1.
Oncogenesis ; 6(5): e335, 2017 May 15.
Article in English | MEDLINE | ID: mdl-28504690

ABSTRACT

Despite advances in early diagnosis and the development of molecularly targeted therapy, curative treatment of colon cancer once it has metastasized is yet to be accomplished. This is closely associated with deregulated CRC cell proliferation and resistance to apoptosis. Here we reveal that upregulation of microRNA-645 (miR-645) through DNA copy number gain is responsible for enhanced proliferation and resistance to apoptosis in colon cancer. MiR-645 was upregulated in most colon cancer tissues related to adjacent normal mucosa. This appeared to be associated with amplification of a section of chromosome 20q13.13, where miR-645 is located. Inhibition of miR-645 reduced proliferation and enhanced sensitivity to apoptosis triggered by the chemotherapeutic drugs 5-fluorouracil and cisplatin in CRC cells, and retarded colon cancer xenograft growth. Conversely, overexpression of miR-645 in normal colon epithelial cells enhanced proliferation and triggered anchorage-independent cell growth. Although SRY-related HMG-box 30 (SOX30) was identified as a miR-645 target, its expression was only partially affected by miR-645, suggesting that miR-645 is a fine-tuning mechanism of SOX30 expression. Moreover, overexpression of SOX30 only moderately inhibited promotion of CRC cell proliferation by miR-645, indicating that miR-645 may have more targets that contribute to its pro-proliferation effect in colon cancer. Together, this study reveals that miR-645 can regulate oncogenesis in colon cancer with SOX30 being one of its targets.

2.
Nutr Metab Cardiovasc Dis ; 26(3): 254-60, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26803595

ABSTRACT

BACKGROUND AND AIMS: Circulating microparticles (MP) are the source of a plasma derived form of the scavenger receptor CD36, termed soluble (s)CD36, the levels of which correlate with markers of atherosclerosis and risk of cardiovascular disease. Long chain n-3 polyunsaturated fatty acids have cardioprotective effects that we have previously reported to be gender specific. The aim of this study was to determine if dietary docosahexaenoic acid (DHA) and/or eicosapentaenoic acid (EPA) supplementation affect circulating CD36 + MP levels, and if this occurs differentially in healthy men and women. METHODS AND RESULTS: Participants (43M, 51F) aged 39.6 ± 1.7 years received 4 weeks of daily supplementation with DHA rich (200 mg EPA; 1000 mg DHA), EPA rich (1000 mg EPA; 200 mg DHA), or placebo (sunola) oil in a double-blinded, randomised, placebo controlled trial. Plasma CD36 + MP were enumerated by flow cytometry and differences between genders and treatments were evaluated by Student's or paired t-test and one way ANOVA. Males and females had similar levels of CD36 + MP at baseline (mean = 1018 ± 325 vs 980 ± 318; p = 0.577) and these were not significantly changed after DHA (M, p = 0.571; F, p = 0.444) or EPA (M, p = 0.361; F, p = 0.901) supplementation. Likewise, the overall percent change in these levels were not different between supplemented cohorts compared to placebo when all participants were combined (% change in CD36 + MP: DHA = 5.7 ± 37.5, EPA = -3.4 ± 35.4, placebo = -11.5 ± 32.9; p = 0.158) or stratified by gender (M, DHA = -2.6 ± 30.6, EPA = -15.1 ± 20.1, placebo = -21.4 ± 28.7, p = 0.187; F, DHA = 11.7 ± 41.5, EPA = 6.8 ± 42.9, placebo = -2.8 ± 34.7, p = 0.552). CONCLUSION: The cardioprotective effects of DHA and EPA do not act through a CD36 + MP mechanism.


Subject(s)
CD36 Antigens/blood , Cell-Derived Microparticles/drug effects , Dietary Supplements , Docosahexaenoic Acids/administration & dosage , Eicosapentaenoic Acid/administration & dosage , Adult , Biomarkers/blood , Body Mass Index , Cardiovascular Diseases/blood , Cardiovascular Diseases/drug therapy , Docosahexaenoic Acids/blood , Double-Blind Method , Eicosapentaenoic Acid/blood , Female , Fish Oils/administration & dosage , Humans , Male , Middle Aged , Particle Size , Treatment Outcome , Young Adult
3.
Oncogene ; 35(23): 3049-61, 2016 06 09.
Article in English | MEDLINE | ID: mdl-26411369

ABSTRACT

Inositol polyphosphate 4-phosphatase type II (INPP4B) negatively regulates phosphatidylinositol 3-kinase signaling and is a tumor suppressor in some types of cancers. However, we have found that it is frequently upregulated in human colon cancer cells. Here we show that silencing of INPP4B blocks activation of Akt and serum- and glucocorticoid-regulated kinase 3 (SGK3), inhibits colon cancer cell proliferation and retards colon cancer xenograft growth. Conversely, overexpression of INPP4B increases proliferation and triggers anchorage-independent growth of normal colon epithelial cells. Moreover, we demonstrate that the effect of INPP4B on Akt and SGK3 is associated with inactivation of phosphate and tensin homolog through its protein phosphatase activity and that the increase in INPP4B is due to Ets-1-mediated transcriptional upregulation in colon cancer cells. Collectively, these results suggest that INPP4B may function as an oncogenic driver in colon cancer, with potential implications for targeting INPP4B as a novel approach to treat this disease.


Subject(s)
Colonic Neoplasms/genetics , Phosphoric Monoester Hydrolases/genetics , Cell Line, Tumor , Cell Proliferation/genetics , Colonic Neoplasms/metabolism , Colonic Neoplasms/pathology , Humans , Immunohistochemistry , Phosphoric Monoester Hydrolases/metabolism
4.
Nutr Diabetes ; 3: e59, 2013 Feb 04.
Article in English | MEDLINE | ID: mdl-23381664

ABSTRACT

OBJECTIVE: Elevated plasma levels of the fatty acid transporter, CD36, have been shown to constitute a novel biomarker for type 2 diabetes mellitus (T2DM). We recently reported such circulating CD36 to be entirely associated with cellular microparticles (MPs) and aim here to determine the absolute levels and cellular origin(s) of these CD36+MPs in persons with T2DM. DESIGN: An ex vivo case-control study was conducted using plasma samples from 33 obese individuals with T2DM (body mass index (BMI)=39.9±6.4 kg m(-2); age=57±9 years; 18 male:15 female) and age- and gender-matched lean and obese non-T2DM controls (BMI=23.6±1.8 kg m(-2) and 33.5±5.9 kg m(-2), respectively). Flow cytometry was used to analyse surface expression of CD36 together with tissue-specific markers: CD41, CD235a, CD14, CD105 and phosphatidyl serine on plasma MPs. An enzyme-linked immunosorbent assay was used to quantify absolute CD36 protein concentrations. RESULTS: CD36+MP levels were significantly higher in obese people with T2DM (P<0.00001) and were primarily derived from erythrocytes (CD235a+=35.8±14.6%); although this did not correlate with haemoglobin A(1c). By contrast, the main source of CD36+MPs in non-T2DM individuals was endothelial cells (CD105+=40.9±8.3% and 33.9±8.3% for lean and obese controls, respectively). Across the entire cohort, plasma CD36 protein concentration varied from undetectable to 22.9 µg ml(-1) and was positively correlated with CD36+MPs measured by flow cytometry (P=0.0006) but only weakly associated with the distribution of controls and T2DM (P=0.021). Multivariate analysis confirmed that plasma CD36+MP levels were a much better biomarker for diabetes than CD36 protein concentration (P=0.009 vs P=0.398, respectively). CONCLUSIONS: Both the levels and cellular profile of CD36+MPs differ in T2DM compared with controls, suggesting that these specific vesicles could represent distinct biological vectors contributing to the pathology of the disease.

5.
Oncogene ; 32(15): 1910-20, 2013 Apr 11.
Article in English | MEDLINE | ID: mdl-22710713

ABSTRACT

Past studies have shown that amplified insulin-like growth factor 1 (IGF1)/IGF1 receptor (IGF1-R) signalling has an important role in colorectal cancer (CRC) development, progression and resistance to treatment. In this report, we demonstrate that downregulation of microRNA-497 (miR-497) as a result of DNA copy number reduction is involved in upregulation of IGF1-R in CRC cells. MiR-497 and miR-195 of the miR-15/16/195/424/497 family that share the same 3' untranslated region (3'UTR) binding seed sequence and are predicted to target IGF1-R were concurrently downregulated in the majority of CRC tissues relative to paired adjacent normal mucosa. However, only overexpression of miR-497 led to suppression of the IGF1-R 3'UTR activity and downregulation of the endogenous IGF1-R protein in CRC cells. This was associated with inhibition of cell survival, proliferation and invasion, and increased sensitivity to apoptosis induced by various stimuli including the chemotherapeutic drugs cisplatin and 5-fluorouracil, and the death ligand tumour necrosis factor-related apoptosis-inducing ligand. The biological effect of miR-497 on CRC cells was largely mediated by inhibition of phosphatidylinositol 3-kinase/Akt signalling, as overexpression of an active form of Akt reversed its impact on cell survival and proliferation, recapitulating the effect of overexpression of IGF1-R. Downregulation of miR-497 and miR-195 appeared to associate with copy number loss of a segment of chromosome 17p13.1, where these miRs are located at proximity. Similarly to miR-195, the members of the same miR family, miR-424 that was upregulated, and miR-15a, miR-15b and miR-16 that were unaltered in expression in CRC tissues compared with paired adjacent normal mucosa, did not appear to have a role in regulating the expression of IGF1-R. Taken together, these results identify downregulation of miR-497 as an important mechanism of upregulation of IGF1-R in CRC cells that contributes to malignancy of CRC.


Subject(s)
Colorectal Neoplasms/genetics , Colorectal Neoplasms/metabolism , DNA Copy Number Variations , MicroRNAs/metabolism , Receptor, IGF Type 1/metabolism , 3' Untranslated Regions , Apoptosis/genetics , Cell Line, Tumor , Cell Proliferation , Cell Survival/genetics , Cisplatin/pharmacology , Disease Progression , Down-Regulation , Drug Resistance, Neoplasm/genetics , Fluorouracil/pharmacology , Gene Expression Regulation, Neoplastic , Humans , Insulin-Like Growth Factor I/metabolism , MicroRNAs/genetics , Neoplasm Invasiveness/genetics , Phosphatidylinositol 3-Kinase/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction/genetics
6.
Cell Death Dis ; 3: e337, 2012 Jun 28.
Article in English | MEDLINE | ID: mdl-22739989

ABSTRACT

Endoplasmic reticulum (ER) stress triggers apoptosis by activating Bim in diverse types of cells, which involves dephosphorylation of Bim(EL) by protein phosphatase 2A (PP2A). However, melanoma cells are largely resistant to ER stress-induced apoptosis, suggesting that Bim activation is suppressed in melanoma cells undergoing ER stress. We show here that ER stress reduces PP2A activity leading to increased ERK activation and subsequent phosphorylation and proteasomal degradation of Bim(EL). Despite sustained upregulation of Bim at the transcriptional level, the Bim(EL) protein expression was downregulated after an initial increase in melanoma cells subjected to pharmacological ER stress. This was mediated by increased activity of ERK, whereas the phosphatase activity of PP2A was reduced by ER stress in melanoma cells. The increase in ERK activation was, at least in part, due to reduced dephosphorylation by PP2A, which was associated with downregulation of the PP2A catalytic C subunit. Notably, instead of direct dephosphorylation of Bim(EL), PP2A inhibited its phosphorylation indirectly through dephosphorylation of ERK in melanoma cells. Taken together, these results identify downregualtion of PP2A activity as an important protective mechanism of melanoma cells against ER stress-induced apoptosis.


Subject(s)
Endoplasmic Reticulum Stress , Melanoma/metabolism , Protein Phosphatase 2/genetics , Apoptosis , Apoptosis Regulatory Proteins/genetics , Apoptosis Regulatory Proteins/metabolism , Bcl-2-Like Protein 11 , Cell Line, Tumor , Down-Regulation , Humans , Melanoma/genetics , Melanoma/pathology , Membrane Proteins/genetics , Membrane Proteins/metabolism , Phosphorylation , Protein Phosphatase 2/metabolism , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , Signal Transduction , Transcription, Genetic , Up-Regulation
7.
Leukemia ; 26(5): 918-26, 2012 May.
Article in English | MEDLINE | ID: mdl-22116550

ABSTRACT

Improved survival of patients with acute lymphoblastic leukemia (ALL) has emerged from identifying new prognostic markers; however, 20% of children still suffer recurrence. Previously, the altered expression of Fat1 cadherin has been implicated in a number of solid tumors. In this report, in vitro analysis shows that Fat1 protein is expressed by a range of leukemia cell lines, but not by normal peripheral blood (PB) and bone marrow (BM) cells from healthy donors. In silico analysis of expression of array data from clinical leukemias found significant levels of Fat1 transcript in 11% of acute myeloid leukemia, 29% and 63% of ALL of B and T lineages, respectively, and little or no transcript present in normal PB or BM. Furthermore, in two independent studies of matched diagnosis-relapse of precursor B-cell (preB) ALL pediatric samples (n=32 and n=27), the level of Fat1 mRNA expression was prognostic at the time of diagnosis. High Fat1 mRNA expression was predictive of shorter relapse-free and overall survival, independent of other traditional prognostic markers, including white blood cell count, sex and age. The data presented demonstrate that Fat1 expression in preB-ALL has a role in the emergence of relapse and could provide a suitable therapeutic target in high-risk preB-ALL.


Subject(s)
Cadherins/metabolism , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/metabolism , Cadherins/genetics , Child , Genes, Tumor Suppressor , Humans , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/diagnosis , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/pathology , Prognosis , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Recurrence , Survival Analysis
8.
Oncogene ; 30(34): 3716-26, 2011 Aug 25.
Article in English | MEDLINE | ID: mdl-21423203

ABSTRACT

Past studies have shown that upregulation of the anti-apoptotic Bcl-2 family protein Mcl-1 is a major adaptive mechanism of melanoma cells to endoplasmic reticulum (ER) stress, and has an important role in resistance of the cells to apoptosis. In this study, we show that the increase in transcription of Mcl-1 in melanoma cells triggered by pharmacological ER stress inducers is mediated by the transcription factor Ets-1. By incremental deletion analysis of the Mcl-1 promoter, we identified a DNA fragment containing an Ets-1 binding site that is transcriptionally responsive to ER stress. Mutations in the Ets-1 binding site or knockdown of Ets-1 inhibited the increase in Mcl-1, indicating that Ets-1 has a critical role in transcriptional upregulation of Mcl-1. Similar to Mcl-1, Ets-1 was transcriptionally upregulated by ER stress. This was mediated by the IRE1α/XBP-1 branch of the unfolded protein response, as upregulation of Ets-1 was inhibited in melanoma cell lines deficient in IRE1α or XBP-1 established by short hairpin RNA knockdown. Activation of the PI3k/Akt pathway downstream of XBP-1 was also involved, in that inhibition of the pathway blocked upregulation of Ets-1. Inhibition of Ets-1 enhanced ER stress-induced apoptosis in melanoma cell lines and in fresh melanoma isolates, recapitulating the effect of inhibition of Mcl-1. These results reveal a key mechanism by which Mcl-1 is transcriptionally upregulated in melanoma cells by ER stress, and identify Ets-1 as a potential target for inhibition to sensitize melanoma cells to apoptosis.


Subject(s)
DNA-Binding Proteins/physiology , Endoplasmic Reticulum/metabolism , Melanoma/metabolism , Proto-Oncogene Protein c-ets-1/physiology , Proto-Oncogene Proteins c-bcl-2/physiology , Transcription Factors/physiology , Up-Regulation/physiology , Apoptosis , Base Sequence , Blotting, Western , Cell Line, Tumor , Chromatin Immunoprecipitation , DNA Primers , Electrophoretic Mobility Shift Assay , Humans , Melanoma/pathology , Myeloid Cell Leukemia Sequence 1 Protein , Polymerase Chain Reaction , Promoter Regions, Genetic , Proto-Oncogene Proteins c-bcl-2/genetics , Regulatory Factor X Transcription Factors , Transcription, Genetic , X-Box Binding Protein 1
9.
J Thromb Haemost ; 9(4): 844-51, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21276198

ABSTRACT

BACKGROUND: CD36 is a widely expressed cell surface receptor that binds lipoproteins, and its function has been implicated in many complications of the metabolic syndrome. A cell-free form of CD36, soluble CD36 (sCD36), has been reported in human plasma, found to be elevated in obesity and diabetes, and claimed as a marker of insulin resistance. OBJECTIVE: To determine the nature of sCD36; in particular, whether sCD36 is truly soluble or, as hypothesized, is found as a component of circulating microparticles (MPs). METHODS: Lipoproteins were fractionated by density gradient centrifugation, and plasma MPs were isolated by ultracentrifugation, size exclusion, and immunoprecipitation with CD36 detected by immunoblotting. MPs from plasma and activated platelets were analyzed by multicolor flow cytometry, with a DyLight-488 anti-CD36 conjugate in combination with antibodies against different cellular markers. RESULTS: Cell-free plasma CD36 was not observed associated with lipoproteins and was not a proteolytic fragment; rather, it was associated with the plasma MP fraction, suggesting that sCD36 in the plasma of normal subjects is a product of circulating MPs. Cytometric and immunoblotting analyses of plasma from normal donors showed that these MPs were derived mainly from platelets. Analysis of in vitro activated platelets also showed that CD36 to be secreted in the form of MPs. CONCLUSIONS: sCD36 is not a proteolytic product, but rather is associated with a specific subset of circulating MPs that can readily be analysed. This finding will enable more specific investigations into the cellular source of the increased levels of plasma CD36 found in subjects with diabetes.


Subject(s)
Biomarkers/blood , CD36 Antigens/blood , Diabetes Mellitus/blood , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Humans , Particle Size , Ultracentrifugation
10.
Cell Death Differ ; 17(8): 1354-67, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20300110

ABSTRACT

Past studies have identified a number of distinct mechanisms that contribute to the resistance of melanoma cells against apoptosis induced by TNF-related apoptosis-inducing ligand (TRAIL). In this report we show that cystatin B is another endogenous inhibitor of TRAIL-induced apoptosis. Cystatin B-deficient melanoma cell lines established by shRNA knockdown displayed increased apoptosis that was associated with enhanced activation of caspase-8 induced by TRAIL. This was not related to the inhibitory effect of cystatin B on the lysosomal cysteine proteases, cathepsin B and L, as they did not have a role in TRAIL-induced apoptosis in most melanoma cell lines even when cystatin B was inhibited. Instead, sensitization of melanoma cells to TRAIL-induced apoptosis by inhibition of cystatin B appeared associated with decreased stability of FLIP(L) as the levels of FLIP(L) were reduced because of shortened half-life time in melanoma cells deficient in cystatin B. In contrast, over-expression of cystatin B increased the levels of FLIP(L), decreased the amount of the E3 ligase Itch associated with FLIP(L), and reduced FLIP(L) ubiquitination. Inhibition of Itch by siRNA restored the levels of FLIP(L) and blocked sensitization to TRAIL-induced apoptosis associated with deficiency in cystatin B. Taken together, these results indicate that cystatin B regulates Itch-mediated degradation of FLIP(L) and thereby TRAIL-induced apoptosis in melanoma cells.


Subject(s)
Apoptosis , CASP8 and FADD-Like Apoptosis Regulating Protein/metabolism , Cystatin B/metabolism , Melanoma/metabolism , Repressor Proteins/metabolism , TNF-Related Apoptosis-Inducing Ligand/pharmacology , Ubiquitin-Protein Ligases/metabolism , Caspase 8/metabolism , Cell Line, Tumor , Cystatin B/genetics , Gene Knockdown Techniques , Humans , Melanoma/enzymology , RNA Interference , RNA, Small Interfering/metabolism
11.
Cell Death Dis ; 1: e69, 2010 Sep 02.
Article in English | MEDLINE | ID: mdl-21364673

ABSTRACT

Bim is known to be critical in killing of melanoma cells by inhibition of the RAF/MEK/ERK pathway. However, the potential role of the most potent apoptosis-inducing isoform of Bim, Bim(S), remains largely unappreciated. Here, we show that inhibition of the mutant B-RAF(V600E) triggers preferential splicing to produce Bim(S), which is particularly important in induction of apoptosis in B-RAF(V600E) melanoma cells. Although the specific B-RAF(V600E) inhibitor PLX4720 upregulates all three major isoforms of Bim, Bim(EL), Bim(L), and Bim(S), at the protein and mRNA levels in B-RAF(V600E) melanoma cells, the increase in the ratios of Bim(S) mRNA to Bim(EL) and Bim(L) mRNA indicates that it favours Bim(S) splicing. Consistently, enforced expression of B-RAF(V600E) in wild-type B-RAF melanoma cells and melanocytes inhibits Bim(S) expression. The splicing factor SRp55 appears necessary for the increase in Bim(S) splicing, as SRp55 is upregulated, and its inhibition by small interfering RNA blocks induction of Bim(S) and apoptosis induced by PLX4720. The PLX4720-induced, SRp55-mediated increase in Bim(S) splicing is also mirrored in freshly isolated B-RAF(V600E) melanoma cells. These results identify a key mechanism for induction of apoptosis by PLX4720, and are instructive for sensitizing melanoma cells to B-RAF(V600E) inhibitors.


Subject(s)
Apoptosis Regulatory Proteins/metabolism , Apoptosis , Melanoma/metabolism , Membrane Proteins/metabolism , Proto-Oncogene Proteins B-raf/metabolism , Proto-Oncogene Proteins/metabolism , Amino Acid Substitution , Apoptosis Regulatory Proteins/genetics , Bcl-2-Like Protein 11 , Cell Line, Tumor , Humans , Indoles/pharmacology , Membrane Proteins/genetics , Mutation , Nuclear Proteins/metabolism , Phosphoproteins/metabolism , Protein Isoforms/genetics , Protein Isoforms/metabolism , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins B-raf/antagonists & inhibitors , Proto-Oncogene Proteins B-raf/genetics , RNA Interference , RNA Splicing , RNA, Messenger/metabolism , RNA, Small Interfering/metabolism , RNA-Binding Proteins , Serine-Arginine Splicing Factors , Sulfonamides/pharmacology
12.
Oncogene ; 25(56): 7401-10, 2006 Nov 30.
Article in English | MEDLINE | ID: mdl-16785995

ABSTRACT

Cancer progression is associated with enhanced directional cell migration, both of the tumour cells invading into the stroma and stromal cells infiltrating the tumour site. In cell-based assays to study directional cell migration, phorbol esters are frequently used as a chemotactic agent. However, the molecular mechanism by which these activators of protein kinase C (PKC) result in the establishment of a polarized migratory phenotype is not known. Here we show that CD44 expression is essential for chemotaxis towards a phorbol ester gradient. In an investigation of CD44 phosphorylation kinetics in resting and stimulated cells, Ser316 was identified as a novel site of phosphorylation following activation of PKC. PKC does not phosphorylate Ser316 directly, but rather mediates the activation of downstream Ser316 kinase(s). In transfection studies, a phosphorylation-deficient Ser316 mutant was shown to act in a dominant-negative fashion to impair chemotaxis mediated by endogenous CD44 in response to a phorbol ester gradient. Importantly, this mutation had no effect on random cell motility or the ability of cells to migrate directionally towards a cocktail of chemoattractants. These studies demonstrate that CD44 functions to provide directional cues to migrating cells without affecting the motility apparatus.


Subject(s)
Hyaluronan Receptors/metabolism , Phorbol 12,13-Dibutyrate/pharmacology , Tetradecanoylphorbol Acetate/pharmacology , Amino Acid Sequence , Cell Line, Tumor , Chemotaxis , Cyclic AMP-Dependent Protein Kinases/metabolism , Humans , Hyaluronan Receptors/chemistry , Phosphorylation , Protein Kinase C/metabolism , Serine/metabolism , Signal Transduction
13.
J Biol Chem ; 275(45): 35264-75, 2000 Nov 10.
Article in English | MEDLINE | ID: mdl-10956645

ABSTRACT

Lateral association between different transmembrane glycoproteins can serve to modulate integrin function. Here we characterize a physical association between the integrins alpha(3)beta(1) and alpha(6)beta(1) and CD36 on the surface of melanoma cells and show that ectopic expression of CD36 by CD36-negative MV3 melanoma cells increases their haptotactic migration on extracellular matrix components. The association was demonstrated by co-immunoprecipitation, reimmunoprecipitation, and immunoblotting of surface-labeled cells lysed in Brij 96 detergent. Confocal microscopy illustrated the co-association of alpha(3) and CD36 in cell membrane projections and ruffles. A requirement for the extracellular domain of CD36 in this association was shown by co-immunoprecipitation experiments using surface-labeled MV3 melanoma or COS-7 cells that had been transiently transfected with chimeric constructs between CD36 and intercellular adhesion molecule 1 (ICAM-1) or with a truncation mutant of CD36. CD36 is known to engage in signal transduction and to localize to membrane microdomains or rafts in several cell types. Toward a mechanistic explanation for the functional effects of CD36 expression, we demonstrate that in fractionated Triton X-100 lysates of the MV3 cells stably transfected with CD36, CD36 was greatly enriched with the detergent-insoluble fractions that represent plasma membrane rafts. Significantly, when these fractionated lysates were reprobed for endogenous beta(1) integrin, it was found that a 4-fold increase in the proportion of the mature protein was contained within the detergent-insoluble fractions when extracted from the CD36-transfected cells compared with MV3 cells transfected with vector only. These results suggest that in melanoma cells CD36 expression may induce the sequestration of certain integrins into membrane microdomains and promote cell migration.


Subject(s)
CD36 Antigens/metabolism , Integrins/metabolism , Melanoma/metabolism , Membrane Glycoproteins , Plant Oils , Animals , Antigens, CD/metabolism , COS Cells , Calcium-Binding Proteins/metabolism , Calreticulin , Caveolin 1 , Caveolins/metabolism , Cell Adhesion , Cell Membrane , Cell Movement , Cell Separation , DNA, Complementary/metabolism , Detergents/pharmacology , Down-Regulation , Flow Cytometry , Humans , Immunoblotting , Integrin alpha3beta1 , Integrin alpha6beta1 , Intercellular Adhesion Molecule-1/metabolism , Membrane Microdomains/metabolism , Microscopy, Confocal , Mutagenesis , Octoxynol/pharmacology , Polyethylene Glycols/pharmacology , Precipitin Tests , Protein Binding , Protein Structure, Tertiary , Ribonucleoproteins/metabolism , Signal Transduction , Tetraspanin 29 , Transfection , Tumor Cells, Cultured
14.
Biochem Biophys Res Commun ; 275(2): 446-54, 2000 Aug 28.
Article in English | MEDLINE | ID: mdl-10964685

ABSTRACT

The CD36 receptor sequence predicts two hydrophobic domains located at the N- and C-termini of the protein, but there are conflicting reports as to whether the N-terminal uncleaved leader sequence functions as a transmembrane domain. To investigate the topology of CD36, we generated a panel of mutants lacking either one or both hydrophobic regions and analyzed their folding and transport in COS-7 cells. The N- and the C-terminal hydrophobic regions were both sufficient to anchor CD36 in the membrane, and a FLAG epitope inserted at the N-terminus was located intracellularly. These results indicate that CD36 adopts a ditopic configuration. Accordingly, neither N- nor C-terminal truncation mutants were secreted. Analysis with conformation-specific monoclonal antibodies showed that the N-terminal transmembrane domain truncated molecule was slowly transported through the exocytic pathway and largely accumulated intracellularly. Thus, dual membrane insertion dictates the correct topogenesis and seems to be necessary for efficient folding and intracellular transport.


Subject(s)
CD36 Antigens/metabolism , Amino Acid Sequence , Animals , Base Sequence , Biological Transport , CD36 Antigens/chemistry , CD36 Antigens/genetics , COS Cells , Cell Membrane/metabolism , DNA Primers , Molecular Sequence Data , Mutagenesis, Site-Directed , Protein Folding , Sequence Homology, Amino Acid
15.
J Gen Virol ; 81(Pt 4): 889-94, 2000 Apr.
Article in English | MEDLINE | ID: mdl-10725413

ABSTRACT

Coxsackievirus A21 (CAV-21) employs a cell receptor complex of decay-accelerating factor (DAF) and intercellular adhesion molecule-1 (ICAM-1) for cell infectivity. In this study, the nature of potential extra- and/or intracellular interactions between DAF and ICAM-1 involved in picornaviral cell entry was investigated. Firstly, it was shown that intracellular interplay between DAF and ICAM-1 is not required for CAV-21 infection, as CAV-21 lytic infection mediated via the DAF/ICAM-1 receptor complex is not inhibited by replacement of the transmembrane and cytoplasmic domains of ICAM-1 with those from an unrelated cell surface molecule, CD36. By immunoprecipitation, chemical cross-linking and picornaviral binding assays, the existence of a close spatial association between DAF and ICAM-1 on the surface of ICAM-1-transfected RD cells was confirmed. Furthermore, it was shown that potential extracellular DAF/ICAM-1 interactions are likely to occur in an area on or proximal to DAF SCR3 and may influence the route of CAV-21 cell entry.


Subject(s)
CD55 Antigens/physiology , Coxsackievirus Infections/virology , Enterovirus/physiology , Intercellular Adhesion Molecule-1/physiology , Animals , Cell Line , Coxsackievirus Infections/metabolism , Cross-Linking Reagents , Protein Binding , Signal Transduction
16.
Mol Cell Biochem ; 214(1-2): 89-95, 2000 Nov.
Article in English | MEDLINE | ID: mdl-11195795

ABSTRACT

CD36 is a membrane glycoprotein expressed by several cell types, and play a role as a receptor for different physiological and pathological ligands. An immunodominant domain of CD36 has been described in the amino acidic region 155-183, where many ligands and monoclonal antibodies (MoAbs) react. MoAbs against CD36 have proved useful in structural as well as functional studies. One of these antibodies, MoAb NL07, recognizes a conformational epitope that is acquired in the late steps of the CD36 maturation. The NL07 epitope appears to be functionally relevant and blocks CD36-mediated binding to red blood cells infected with the malaria parasite Plasmodium falciparum (IRBC). In this work a mutant COS-7 clone expressing NL07-negative CD36 molecules on the cell surface was investigated. In the mutant, the methionine in position 156 of the wild type CD36 sequence was replaced by a valine. It was determined that methionine 156 was essential for NL07 reactivity, mapping the NL07 epitope to the vicinity of the functionally important immunodominant domain (aa 155-183) of CD36. Although methionine 156 is located in this region, the CD36V156 mutated molecule was apparently functional and able to bind IRBC and oxidized LDL.


Subject(s)
CD36 Antigens/chemistry , Amino Acid Sequence , Amino Acid Substitution , Animals , Antibodies, Monoclonal , Base Sequence , CD36 Antigens/genetics , COS Cells , DNA Primers/genetics , Humans , Immunodominant Epitopes/chemistry , Immunodominant Epitopes/genetics , In Vitro Techniques , Malaria, Falciparum/immunology , Methionine/chemistry , Mice , Point Mutation , Protein Structure, Tertiary , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/immunology , Transfection
17.
Cell Adhes Commun ; 6(1): 21-38, 1998 Jun.
Article in English | MEDLINE | ID: mdl-9759519

ABSTRACT

The LIM 1863 colon carcinoma cell line grows as structured organoids around a central lumen, and we have previously demonstrated that the three-dimensional arrangement protects the individual cells from apoptosis induced by an anti-alpha v integrin antibody, 23C6 (Bates et al., 1994). Here we show that the intercellular forces which drive spheroid formation can be overcome by exposure of the cells to a collagen substrate, or more specifically through ligation of the CD44 receptor by a monoclonal antibody. Binding to immobilized anti-CD44 antibody induced a monolayer morphology which is accompanied by fibronectin production and secretion, and expression of the integrin alpha v beta 6. Significantly, the cells of the monolayer acquired resistance to 23C6 antibody-mediated apoptosis over time and this property was sustained even after removal from the monolayer. We provide data to show that this resistance is not dependent on monolayer morphology, constant engagement of the CD44 receptor, loss of the 23C6 antigen, or elevation of Bcl-2 or Bcl-XL protein. The CD44 expressed by LIM 1863 is shown to be the metastatic variant of the molecule therefore these results provide a possible explanation for the selective advantages conferred by expression of this variant for metastasizing colon cancer cells. Overall, the findings of this study support a model for the development of malignancy through the production of specific survival and growth signals as a direct consequence of a signaling event induced by stimulation of an epithelial variant of CD44.


Subject(s)
Apoptosis/physiology , Hyaluronan Receptors/physiology , Animals , Antibodies, Monoclonal/immunology , Cell Adhesion/physiology , Colonic Neoplasms/immunology , Colonic Neoplasms/pathology , Extracellular Matrix/physiology , Fibronectins/metabolism , Humans , Hyaluronan Receptors/immunology , Integrins/metabolism , Mice , Neoplasm Metastasis , Proto-Oncogene Proteins c-bcl-2/analysis , RNA, Messenger/analysis , Rabbits , Time Factors , Tumor Cells, Cultured
18.
J Immunol ; 160(5): 2318-23, 1998 Mar 01.
Article in English | MEDLINE | ID: mdl-9498772

ABSTRACT

Decay-accelerating factor (DAF), a widely expressed membrane complement-regulatory protein, is utilized as a cellular receptor by many human enteric pathogens. We show here that the binding of two enteroviruses to individual short consensus repeats (SCR) of DAF on the cell surface is greatly augmented by mAb binding to an alternate SCR: Coxsackievirus A21 binding to the SCR1 of DAF is increased by Ab binding to SCR3 and, conversely, Echovirus 7 binding to SCR3 is enhanced severalfold by Ab binding to SCR1. These Ab-induced increases in viral binding also resulted in increased viral infectivity. Using purified soluble DAF in a solid phase assay it was found that Ab binding to SCR1 is increased greatly in the presence of an Ab against SCR3 and, reciprocally, Ab against SCR1 greatly increases Ab binding to SCR3. In contrast to the results obtained with the larger viral particles, however, this reciprocal Ab-induced enhancement of binding is not seen when measuring Ab binding to membrane-bound DAF SCR on the cell surface. These findings provide a possible explanation for functional differences between membrane-bound and soluble DAF with implications for a potential role for DAF-binding molecules in regulating DAF function. This is the first demonstration of enhancement of viral infectivity mediated by Ab against the viral receptor.


Subject(s)
Binding Sites, Antibody , CD55 Antigens/immunology , CD55 Antigens/metabolism , Enterovirus/immunology , Enterovirus/pathogenicity , Repetitive Sequences, Nucleic Acid/immunology , Animals , CD55 Antigens/chemistry , CHO Cells , Cell Adhesion , Cell Membrane/immunology , Cell Membrane/virology , Consensus Sequence/immunology , Cricetinae , Enterovirus/physiology , Enterovirus B, Human/immunology , Enterovirus B, Human/pathogenicity , Enterovirus B, Human/physiology , Humans , Receptors, Fc/physiology , Rhabdomyosarcoma , Solubility , Tumor Cells, Cultured , Tumor Virus Infections/immunology , Tumor Virus Infections/virology
19.
Biochem Biophys Res Commun ; 240(3): 812-8, 1997 Nov 26.
Article in English | MEDLINE | ID: mdl-9398651

ABSTRACT

CD36 is a transmembrane glycoprotein expressed on the surface of a number of cell types. The analysis of CD36 from platelets using immunoblotting, gel filtration, and native PAGE indicated the presence of high molecular complexes exceeding the Mr of monomeric CD36. Experiments using transfected COS-7 cells revealed these complexes were homodimers and -multimers of CD36. The multimers could be dissociated by treatment with a reducing agent, indicating they were formed by intermolecular cysteine-bridging. Mutagenesis of the cDNA for CD36 implicated the cysteines in the extracellular domain of the molecule. The potential physiological roles of CD36 multimerisation are discussed.


Subject(s)
Blood Platelets/chemistry , CD36 Antigens/chemistry , Protein Conformation , Animals , Biopolymers , CD36 Antigens/blood , CD36 Antigens/isolation & purification , CD36 Antigens/metabolism , COS Cells , Chromatography, Gel , Cysteine/chemistry , Cysteine/metabolism , Dimerization , Electrophoresis, Gel, Two-Dimensional , Electrophoresis, Polyacrylamide Gel , Fluorescent Antibody Technique , Immunoblotting , Microscopy, Fluorescence , Mutagenesis , Sulfhydryl Compounds/pharmacology , Sulfur Radioisotopes , Transfection
20.
J Immunol ; 158(7): 3353-8, 1997 Apr 01.
Article in English | MEDLINE | ID: mdl-9120293

ABSTRACT

CD63 belongs to the transmembrane 4 superfamily of membrane proteins and is expressed in several normal tissues as well as in melanoma cells. Previous reports have suggested that CD63 may play an important role in inhibiting melanoma progression, and this was supported by our studies showing that CD63 was associated with suppression of the growth of melanoma in nude mice. Recently, we and others have shown that CD63 may form noncovalent associations with beta1 integrins, which suggests that the function of CD63 may be related to that of integrins. To further explore the role of CD63 in melanoma, we transfected CD63 into a highly motile, CD63-negative melanoma cell line, KM3, which was shown to express alpha(v)beta5 as the predominant integrin with only trace amounts of beta1 integrins. Following transfection, CD63 was shown to associate with beta1 integrins, and beta1 expression appeared to be up-regulated. Cell motility in serum-containing media was markedly suppressed following transfection of CD63. This inhibition was potentiated by mAbs to CD63, and correlated with the level of CD63 expression. The CD63-transfected, but not the untransfected, melanoma cells showed increased adhesion and migration on the beta1 substrates, fibronectin, laminin, and collagen, whereas rates of migration were similar on the beta5 substrate, vitronectin. These results show that CD63 is involved in regulation of the motility of melanoma cells and their adhesion and migration on substrates associated with beta1 integrins. We suggest they provide further insights into the role of CD63 in tumor progression.


Subject(s)
Antigens, CD/physiology , Cell Movement/immunology , Melanoma/immunology , Platelet Membrane Glycoproteins/physiology , Antigens, CD/biosynthesis , Cell Adhesion/immunology , Cell Adhesion Molecules/biosynthesis , Cell Adhesion Molecules/pharmacology , Collagen/immunology , Fibronectins/immunology , Humans , Integrins/biosynthesis , Laminin/immunology , Melanoma/metabolism , Platelet Membrane Glycoproteins/biosynthesis , Skin Neoplasms/immunology , Skin Neoplasms/metabolism , Substrate Specificity/immunology , Tetraspanin 30 , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL
...