Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 44
Filter
1.
Int J Mol Sci ; 25(13)2024 Jul 02.
Article in English | MEDLINE | ID: mdl-39000374

ABSTRACT

Cell adhesion is a dynamic process that plays a fundamental role in cell proliferation, maintenance, differentiation, and migration. Basal cell adhesion molecule (BCAM), also known as Lutheran (Lu), belongs to the immunoglobulin superfamily of cell adhesion molecules. Lu/BCAM, which is widely expressed in red blood cells, endothelial cells, smooth muscle cells and epithelial cells across various tissues, playing a crucial role in many cellular processes, including cell adhesion, cell motility and cell migration. Moreover, Lu/BCAM, dysregulated in many diseases, such as blood diseases and various types of cancer, may act as a biomarker and target for the treatment of these diseases. This review explores the significance of Lu/BCAM in cell adhesion and its potential as a novel target for treating hematological diseases and tumors.


Subject(s)
Hematologic Diseases , Neoplasms , Humans , Neoplasms/metabolism , Neoplasms/pathology , Hematologic Diseases/metabolism , Lutheran Blood-Group System/metabolism , Cell Adhesion , Animals , Cell Adhesion Molecules/metabolism , Cell Movement
2.
Hypertension ; 79(12): 2830-2842, 2022 12.
Article in English | MEDLINE | ID: mdl-36252138

ABSTRACT

BACKGROUND: Preeclampsia is a pregnancy syndrome that may utilize multiple pathogenic mechanisms. Insufficient trophoblast invasion and impaired uterine spiral artery remodeling are believed to be the pathological basis; yet the underlying mechanisms remain largely unclear. METHODS: The placental BCAM (basal cell adhesion molecule) expression and important clinical indicators were detected and correlation analysis was performed. MiRNAs directly targeting BCAM were predicted and further verified by dual-luciferase reporter gene, and the downstream molecular mechanisms of BCAM were investigated in both HTR-8/SVneo and JAR cells. In addition, pregnant/nonpregnant rats were treated with adenoviruses containing BCAM shRNA genes (Ad-shBCAM) on gestational 9.5 days to detect the preeclamptic features. RESULTS: The BCAM is highly expressed on the trophoblast membrane and decreased in the preeclamptic placentae. In HTR-8/SVneo and JAR cells, BCAM knockdown inhibited trophoblast proliferation, migration, and invasion, and suppressed phosphorylation on Y705 of STAT3 dependent on the downregulation of PIK3R6. Moreover, miR-199a-5p mediated the degradation of BCAM and also inhibited trophoblast proliferation, migration, and invasion. In vivo, BCAM deficiency induced a preeclampsia-like phenotype included elevated systolic blood pressure, proteinuria, impaired morphology and function of multiple organs (placenta, liver, and kidney), and fetal growth restriction. The expression of placenta BCAM/PIK3R6/p-STAT3 signaling was also downregulated in this preeclampsia rat model. CONCLUSIONS: MiR-199a-5p mediated-BCAM deficiency contributes to the suppression of trophoblast proliferation, migration, and invasion by inhibiting PIK3R6/p-STAT3 signaling, which may lead to poor placentation and result in preeclampsia-like phenotypes. Our study provides a new academic perspective on the pathogenesis of preeclampsia.


Subject(s)
MicroRNAs , Pre-Eclampsia , Animals , Female , Pregnancy , Rats , Cell Adhesion Molecules/metabolism , Cell Line , Cell Movement , Lutheran Blood-Group System/metabolism , MicroRNAs/genetics , MicroRNAs/metabolism , Placenta/metabolism , STAT3 Transcription Factor/genetics , STAT3 Transcription Factor/metabolism , Trophoblasts/metabolism
3.
Clin Epigenetics ; 14(1): 99, 2022 08 08.
Article in English | MEDLINE | ID: mdl-35941663

ABSTRACT

BACKGROUND: Clear cell renal cell carcinoma (ccRCC) is the most common and highly heterogeneous subtype of renal cell carcinoma. Dysregulated basal cell adhesion molecule (BCAM) gene is associated with poor prognosis in various cancers. However, the dysregulated functions and related multi-omics features of BCAM in ccRCC stay unclear. RESULTS: BCAM expression was aberrantly downregulated in ccRCC and correlated with adverse pathological parameters and poor prognosis. Low mRNA expression of BCAM was remarkably associated with its CpG methylation levels and BAP1 mutation status. Patients with lower-expressed BCAM concomitant with BAP1 mutation had a worse prognosis. Using RNA-seq data from The cancer genome atlas, we found that compared to the BCAM-high expression subgroup, ccRCC patients in the BCAM-low expression subgroup had significantly higher levels of immune infiltration, higher immune checkpoint expression levels and lower TIDE (tumor immune dysfunction and exclusion) score, indicating potential better response to immunotherapy. Data from the Clinical Proteomic Tumor Analysis Consortium further validated the association between low BCAM expression and CD8 + inflamed phenotype at protein level. Meanwhile, our results suggested that the angiogenesis-related pathways were enriched in the BCAM-high expression subgroup. More importantly, according to the data from the GDSC database, we revealed that the BCAM-high expression subgroup should be more sensitive to anti-angiogenetic therapies, including sorafenib, pazopanib and axitinib. CONCLUSIONS: These results suggest that BCAM could serve as a biomarker distinguishing different tumor microenvironment phenotypes, predicting prognosis and helping therapeutic decision-making for patients with ccRCC.


Subject(s)
Carcinoma, Renal Cell , Carcinoma , Kidney Neoplasms , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Carcinoma/genetics , Carcinoma, Renal Cell/metabolism , Cell Adhesion Molecules/genetics , DNA Methylation , Epigenesis, Genetic , Humans , Kidney Neoplasms/pathology , Lutheran Blood-Group System/metabolism , Prognosis , Proteomics , Tumor Microenvironment/genetics
4.
Blood Adv ; 6(21): 5798-5810, 2022 11 08.
Article in English | MEDLINE | ID: mdl-35349634

ABSTRACT

Severe malarial anemia (SMA) is the main cause of malaria-associated infant mortality in malaria endemic countries. One major factor that contributes to SMA is the accumulation of uninfected red blood cells (uRBCs) in the spleen. We report the activation of adhesion molecules Lutheran/basal cell adhesion molecule (Lu/BCAM) and CD44 on uRBCs from Plasmodium falciparum in vitro cultures and patients with malaria that mediates adherence to the splenic extracellular matrix (ECM) components laminin-α5 and hyaluronic acid (HA), respectively. This tight ECM-adhesion molecule interaction was associated with elevated intracellular Ca2+ levels, increased shedding of microvesicles, and Lu/BCAM clustering on altered uRBCs. Moreover, we observed that a soluble parasite-derived factor promoted the adhesive phenotype of uRBCs, as the incubation of RBCs with filtered malaria-conditioned medium reproduced the same adhesive effect in malaria culture-derived uRBCs. Eventually, Lu/BCAM and CD44 activation facilitate the adherence to ECM components of the red pulp, resulting in the enhanced splenic retention of uRBCs. Our results suggest a novel adhesion molecule-dependent mechanism that augments malaria-induced anemia.


Subject(s)
Anemia, Sickle Cell , Malaria , Humans , Lutheran Blood-Group System/metabolism , Cell Adhesion Molecules/genetics , Erythrocytes/metabolism
5.
Development ; 149(1)2022 01 01.
Article in English | MEDLINE | ID: mdl-35020896

ABSTRACT

In early placental development, progenitor cytotrophoblasts (CTB) differentiate along one of two cellular trajectories: the villous or extravillous pathways. CTB committed to the villous pathway fuse with neighboring CTB to form the outer multinucleated syncytiotrophoblast (SCT), whereas CTB committed to the extravillous pathway differentiate into invasive extravillous trophoblasts (EVT). Unfortunately, little is known about the processes controlling human CTB progenitor maintenance and differentiation. To address this, we established a single cell RNA sequencing (scRNA-seq) dataset from first trimester placentas to identify cell states important in trophoblast progenitor establishment, renewal and differentiation. Multiple distinct trophoblast states were identified, representing progenitor CTB, column CTB, SCT precursors and EVT. Lineage trajectory analysis identified a progenitor origin that was reproduced in human trophoblast stem cell organoids. Heightened expression of basal cell adhesion molecule (BCAM) defined this primitive state, where BCAM enrichment or gene silencing resulted in enhanced or diminished organoid growth, respectively. Together, this work describes at high-resolution trophoblast heterogeneity within the first trimester, resolves gene networks within human CTB progenitors and identifies BCAM as a primitive progenitor marker and possible regulator.


Subject(s)
Cell Adhesion Molecules/metabolism , Cell Lineage , Lutheran Blood-Group System/metabolism , Trophoblasts/metabolism , Adult , Cell Adhesion Molecules/genetics , Cell Differentiation , Cells, Cultured , Female , Gene Expression Regulation, Developmental , Gene Regulatory Networks , Human Embryonic Stem Cells/cytology , Human Embryonic Stem Cells/metabolism , Humans , Lutheran Blood-Group System/genetics , Organoids/cytology , Organoids/metabolism , Trophoblasts/cytology
6.
Haematologica ; 106(9): 2478-2488, 2021 09 01.
Article in English | MEDLINE | ID: mdl-32855277

ABSTRACT

Vaso-occlusive crises are the hallmark of sickle cell disease (SCD). They are believed to occur in two steps, starting with adhesion of deformable low-dense red blood cells (RBCs), or other blood cells such as neutrophils, to the wall of post-capillary venules, followed by trapping of the denser RBCs or leukocytes in the areas of adhesion because of reduced effective lumen-diameter. In SCD, RBCs are heterogeneous in terms of density, shape, deformability and surface proteins, which accounts for the differences observed in their adhesion and resistance to shear stress. Sickle RBCs exhibit abnormal adhesion to laminin mediated by Lu/BCAM protein at their surface. This adhesion is triggered by Lu/BCAM phosphorylation in reticulocytes but such phosphorylation does not occur in mature dense RBCs despite firm adhesion to laminin. In this study, we investigated the adhesive properties of sickle RBC subpopulations and addressed the molecular mechanism responsible for the increased adhesion of dense RBCs to laminin in the absence of Lu/BCAM phosphorylation. We provide evidence for the implication of oxidative stress in post-translational modifications of Lu/BCAM that impact its distribution and cis-interaction with glycophorin C at the cell surface activating its adhesive function in sickle dense RBCs.


Subject(s)
Anemia, Sickle Cell , Laminin , Cell Adhesion , Cell Adhesion Molecules/metabolism , Erythrocytes/metabolism , Humans , Laminin/metabolism , Lutheran Blood-Group System/metabolism , Oxidative Stress
7.
J Biol Chem ; 294(41): 14911-14921, 2019 10 11.
Article in English | MEDLINE | ID: mdl-31413112

ABSTRACT

Tumor cell migration depends on the interactions of adhesion proteins with the extracellular matrix. Lutheran/basal cell adhesion molecule (Lu/BCAM) promotes tumor cell migration by binding to laminin α5 chain, a subunit of laminins 511 and 521. Lu/BCAM is a type I transmembrane protein with a cytoplasmic domain of 59 (Lu) or 19 (Lu(v13)) amino acids. Here, using an array of techniques, including site-directed mutagenesis, immunoblotting, FRET, and proximity-ligation assays, we show that both Lu and Lu(v13) form homodimers at the cell surface of epithelial cancer cells. We mapped two small-XXX-small motifs in the transmembrane domain as potential sites for monomers docking and identified three cysteines in the cytoplasmic domain as being critical for covalently stabilizing dimers. We further found that Lu dimerization and phosphorylation of its cytoplasmic domain were concomitantly needed to promote cell migration. We conclude that Lu is the critical isoform supporting tumor cell migration on laminin 521 and that the Lu:Lu(v13) ratio at the cell surface may control the balance between cellular firm adhesion and migration.


Subject(s)
Cell Adhesion Molecules/chemistry , Cell Adhesion Molecules/metabolism , Cell Movement/drug effects , Laminin/pharmacology , Lutheran Blood-Group System/chemistry , Lutheran Blood-Group System/metabolism , Protein Multimerization/drug effects , Amino Acid Sequence , Animals , Caco-2 Cells , Dogs , Humans , Madin Darby Canine Kidney Cells , Models, Molecular , Phosphorylation/drug effects , Protein Domains , Protein Structure, Quaternary
8.
Elife ; 72018 07 30.
Article in English | MEDLINE | ID: mdl-30059007

ABSTRACT

Under chronic or severe liver injury, liver progenitor cells (LPCs) of biliary origin are known to expand and contribute to the regeneration of hepatocytes and cholangiocytes. This regeneration process is called ductular reaction (DR), which is accompanied by dynamic remodeling of biliary tissue. Although the DR shows apparently distinct mode of biliary extension depending on the type of liver injury, the key regulatory mechanism remains poorly understood. Here, we show that Lutheran (Lu)/Basal cell adhesion molecule (BCAM) regulates the morphogenesis of DR depending on liver disease models. Lu+ and Lu- biliary cells isolated from injured liver exhibit opposite phenotypes in cell motility and duct formation capacities in vitro. By overexpression of Lu, Lu- biliary cells acquire the phenotype of Lu+ biliary cells. Lu-deficient mice showed severe defects in DR. Our findings reveal a critical role of Lu in the control of phenotypic heterogeneity of DR in distinct liver disease models.


Subject(s)
Bile Ducts/metabolism , Bile Ducts/physiology , Cell Adhesion Molecules/metabolism , Liver Regeneration , Lutheran Blood-Group System/metabolism , Membrane Glycoproteins/metabolism , Animals , Antibody Specificity/immunology , Cell Movement/genetics , Cell Separation , Choline , Diet , Disease Models, Animal , Epithelial Cell Adhesion Molecule/metabolism , Gene Expression Regulation , Humans , Integrin beta1/genetics , Integrin beta1/metabolism , Laminin/metabolism , Liver/metabolism , Liver Regeneration/genetics , Membrane Glycoproteins/genetics , Mice, Inbred C57BL , Mice, Knockout , Models, Biological , Phenotype , RNA, Messenger/genetics , RNA, Messenger/metabolism , Reproducibility of Results
9.
Sci Rep ; 8(1): 6612, 2018 04 26.
Article in English | MEDLINE | ID: mdl-29700410

ABSTRACT

Antibody-drug conjugates (ADCs) are attractive in cancer therapy because they can directly bind to cancer cells and provide anticancer activity. To kill cancer cells with ADCs, the target antigens are required not only to be highly and/or selectively expressed on cancer cells but also internalized by the cells. CD239, also known as the Lutheran blood group glycoprotein (Lu) or basal cell adhesion molecule (B-CAM), is a specific receptor for laminin α5, a major component of basement membranes. Here, we show that CD239 is strongly expressed in a subset of breast cancer cells and internalized into the cells. We also produced a human single-chain variable fragment (scFv) specific to CD239 fused with human IgG1 Fc, called C7-Fc. The binding affinity of the C7-Fc antibody is similar to that of mouse monoclonal antibodies. Although the C7-Fc antibody alone does not influence cellular functions, when conjugated with a fragment of diphtheria toxin lacking the receptor-binding domain (fDT), it can selectively kill breast cancer cells. Interestingly, fDT-bound C7-Fc shows anticancer activity in CD239-highly positive SKBR3 cells, but not in weakly positive cells. Our results show that CD239 is a promising antigen for ADC-based breast cancer therapy.


Subject(s)
Breast Neoplasms/metabolism , Cell Adhesion Molecules/metabolism , Endocytosis , Lutheran Blood-Group System/metabolism , Animals , Biomarkers, Tumor , Breast Neoplasms/drug therapy , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cell Adhesion Molecules/antagonists & inhibitors , Cell Adhesion Molecules/genetics , Cell Line, Tumor , Cytotoxicity Tests, Immunologic , Female , Humans , Immunoconjugates/pharmacology , Immunoglobulin Fc Fragments/immunology , Immunohistochemistry , Lutheran Blood-Group System/genetics , Mice , Protein Binding , Protein Transport , Single-Chain Antibodies/pharmacology
10.
Toxins (Basel) ; 10(3)2018 03 13.
Article in English | MEDLINE | ID: mdl-29533999

ABSTRACT

Cytotoxic Necrotizing Factor 1 (CNF1) was identified in 1983 as a protein toxin produced by certain pathogenic strains of Escherichia coli. Since then, numerous studies have investigated its particularities. For instance, it is associated with the single chain AB-toxin family, and can be divided into different functional and structural domains, e.g., catalytic and transmembrane domain and interaction sites. A few years ago, the identification of the Lutheran (Lu) adhesion glycoprotein/basal cell adhesion molecule (BCAM) as a cellular receptor for CNF1 provided new insights into the adhesion process of CNF1. Very recently, the Ig-like domain 2 of Lu/BCAM was confirmed as the main interaction site using protein-protein interaction and competition studies with various different mutants. Here, I present in silico approaches that precisely explain the impact of these mutations, leading to a better explanation of these experimental studies. These results can be used in the development of future antitoxin strategies.


Subject(s)
Bacterial Toxins/genetics , Bacterial Toxins/metabolism , Cell Adhesion Molecules/metabolism , Escherichia coli Proteins/genetics , Escherichia coli Proteins/metabolism , Lutheran Blood-Group System/metabolism , Bacterial Toxins/chemistry , Catalytic Domain , Computer Simulation , Escherichia coli Proteins/chemistry , Point Mutation
11.
Blood Adv ; 2(1): 14-24, 2018 01 09.
Article in English | MEDLINE | ID: mdl-29344581

ABSTRACT

Lutheran/basal cell adhesion molecule (Lu/BCAM) is a transmembrane adhesion molecule expressed by erythrocytes and endothelial cells that can interact with the extracellular matrix protein laminin-α5. In sickle cell disease, Lu/BCAM is thought to contribute to adhesion of sickle erythrocytes to the vascular wall, especially during vaso-occlusive crises. On healthy erythrocytes however, its function is unclear. Here we report that Lu/BCAM is activated during erythrocyte aging. We show that Lu/BCAM-mediated binding to laminin-α5 is restricted by interacting, in cis, with glycophorin-C-derived sialic acid residues. Following loss of sialic acid during erythrocyte aging, Lu/BCAM is released from glycophorin-C and allowed to interact with sialic acid residues on laminin-α5. Decreased glycophorin-C sialylation, as observed in individuals lacking exon 3 of glycophorin-C, the so-called Gerbich phenotype, was found to correlate with increased Lu/BCAM-dependent binding to laminin-α5. In addition, we identified the sialic acid-binding site within the third immunoglobulin-like domain within Lu/BCAM that accounts for the interaction with glycophorin-C and laminin-α5. Last, we present evidence that neuraminidase-expressing pathogens, such as Streptococcus pneumoniae, can similarly induce Lu/BCAM-mediated binding to laminin-α5, by cleaving terminal sialic acid residues from the erythrocyte membrane. These results shed new light on the mechanisms contributing to increased adhesiveness of erythrocytes at the end of their lifespan, possibly facilitating their clearance. Furthermore, this work may contribute to understanding the pathology induced by neuraminidase-positive bacteria, because they are especially harmful to patients suffering from sickle cell disease and are associated with the occurrence of vaso-occlusive crises.


Subject(s)
Cell Adhesion Molecules/metabolism , Cell Adhesion , Erythrocyte Aging , Glycophorins/metabolism , Lutheran Blood-Group System/metabolism , N-Acetylneuraminic Acid/metabolism , Anemia, Sickle Cell/blood , Binding Sites , Humans , Laminin/metabolism , Neuraminidase
12.
Curr Comput Aided Drug Des ; 14(3): 253-268, 2018.
Article in English | MEDLINE | ID: mdl-27439722

ABSTRACT

BACKGROUND: Drepanocytosis is a genetic blood disorder characterized by red blood cells that assume an abnormal, rigid, sickle shape. In the pathogenesis of vaso-occlusive crises of sickle cell disease, red blood cells bind to the endothelium and promote vaso-occlusion. At the surface of these sickle red blood cells, the overexpressed protein Lutheran strongly interacts with the Laminin 511/521. The aim of this study is to identify a PPI inhibitor with a high probability of binding to Lutheran for the inhibition of the Lutheran-Laminin 511/521 interaction. METHODS: A virtual screening was performed with 395 601 compounds that target Lutheran. Prior validation of a robust docking and scoring protocol was considered on the protein CD80 because this protein has a binding site with similar topological and physico-chemical characteristics and it also has a series of ligands with known affinity constants. This protocol consisted of multiple filtering steps based on docked scores, molecular dynamics simulations, post-screening scores, and molecular properties. RESULTS: A robust docking and scoring protocol was validated on the protein CD80 with the docking program DOCK6 and the secondary scoring function XSCORE. We identified four molecules for Lutheran that have good structural and physico-chemical properties. CONCLUSION: We took advantage of the similarities between the binding site of Lutheran and that of the protein CD80 to set up a robust docking and scoring protocol. Our protocol for primary scoring filtering, molecular dynamics simulation filtering, secondary scoring filtering, and molecular property filtering allows discarding most of the ligands with four compounds that are promising candidates for inhibiting the Lutheran-Laminin 511/521 interaction.


Subject(s)
Cell Adhesion Molecules/metabolism , Drug Discovery , Laminin/metabolism , Lutheran Blood-Group System/metabolism , Protein Interaction Maps/drug effects , Cell Adhesion Molecules/antagonists & inhibitors , Computer-Aided Design , Drug Discovery/methods , Humans , Laminin/antagonists & inhibitors , Ligands , Molecular Docking Simulation , Molecular Dynamics Simulation , Small Molecule Libraries/chemistry , Small Molecule Libraries/pharmacology
13.
Toxins (Basel) ; 10(1)2017 12 21.
Article in English | MEDLINE | ID: mdl-29267242

ABSTRACT

The protein toxin Cytotoxic Necrotizing Factor 1 (CNF1) is a major virulence factor of pathogenic Escherichia coli strains. It belongs to a family of single chain AB-toxins, which enter mammalian cells by receptor-mediated endocytosis. Recently, we identified the Lutheran (Lu) adhesion glycoprotein/basal cell adhesion molecule (BCAM) as a cellular receptor for CNF1. Here, we identified the Ig-like domain 2 of Lu/BCAM as main interaction site of the toxin by direct protein-protein interaction and competition studies. Using surface plasmon resonance, we showed a high affinity CNF-Lu/BCAM interaction with a KD of 2.8 nM. Furthermore, we performed small-angle X-ray scattering to define the molecular envelope of the Lu/BCAM-CNF1 complex, suggesting a 6:1 ratio of Lu/BCAM to CNF1 in the receptor-toxin complex. This study leads to a deeper understanding of the interaction between CNF1 and Lu/BCAM, and presents novel opportunities for the development of future anti-toxin strategies.


Subject(s)
Bacterial Toxins/metabolism , Escherichia coli Proteins/metabolism , Lutheran Blood-Group System/metabolism , Membrane Glycoproteins/metabolism , Animals , Cell Line , Chlorocebus aethiops , Humans , Protein Binding
14.
J Biomed Sci ; 24(1): 61, 2017 Aug 26.
Article in English | MEDLINE | ID: mdl-28841878

ABSTRACT

BACKGROUND: Lutheran/basal cell adhesion molecule (Lu/BCAM) is a membrane bound glycoprotein. This study was performed to investigate the role and downstream signaling pathway of Lu/BCAM in human bladder tumorigenesis. METHODS: Five human bladder cancer (E6, RT4, TSGH8301, TCCSUP and J82), one stable mouse fibroblast cell line (NIH-Lu) expressing Lu/BCAM transgene and sixty human uroepithelial carcinoma specimens were analyzed by real-time PCR, immunohistochemistry (IHC), immunofluorescence (IFA) staining, Western blotting and promoter luciferase assay for Lu/BCAM, respectively. The tumorigenicity of Lu/BCAM was demonstrated by focus formation, colony-forming ability, tumour formation, cell adhesion and migration. RESULTS: H-ras V12 was revealed to up-regulate Lu/BCAM at both transcriptional and translation levels. Lu/BCAM expression was detected on the membrane of primary human bladder cancer cells. Over-expression of Lu/BCAM in NIH-Lu stable cells increased focus number, colony formation and cell adhesion accompanied with F-actin rearrangement and decreased cell migration compared with parental NIH3T3 fibroblasts. In the presence of laminin ligand, Lu/BCAM overexpression further suppressed cell migration accompanied with increased cell adhesion. We further revealed that laminin-Lu/BCAM-induced cell adhesion and F-actin rearrangement were through increased Erk phosphorylation with an increase of RhoA and a decrease of Rac1 activity. Similarly, high Lu/BCAM expression was detected in the tumors of human renal pelvis, ureter and bladder, and was significantly associated with advanced tumor stage (p = 0.02). Patients with high Lu/BCAM expression showed a trend toward larger tumor size (p = 0.07) and lower disease-specific survival (p = 0.08), although not reaching statistical significance. CONCLUSION: This is the first report showing that Lu/BCAM, in the presence of its ligand laminin, is oncogenic in human urothelial cancers and may have potential as a novel therapeutic target.


Subject(s)
Carcinogenesis/genetics , Cell Adhesion Molecules/genetics , Lutheran Blood-Group System/genetics , Signal Transduction , Urinary Bladder Neoplasms/genetics , Animals , Carcinogenicity Tests , Cell Adhesion Molecules/metabolism , Cell Line, Tumor , Fibroblasts , Humans , Laminin/genetics , Ligands , Lutheran Blood-Group System/metabolism , Mice , NIH 3T3 Cells , Transcriptome
15.
Clin Cancer Res ; 22(19): 4923-4933, 2016 Oct 01.
Article in English | MEDLINE | ID: mdl-27143691

ABSTRACT

PURPOSE: KRAS mutations confer adverse prognosis to colorectal cancer, and no targeted therapies have shown efficacy in this patient subset. Paracrine, nongenetic events induced by KRAS-mutant tumor cells are expected to result in specific deregulation and/or relocation of tumor microenvironment (TME) proteins, which in principle can be exploited as alternative therapeutic targets. EXPERIMENTAL DESIGN: A multimodal strategy combining ex vivo/in vitro phage display screens with deep-sequencing and bioinformatics was applied to uncover TME-specific targets in KRAS-mutant hepatic metastasis from colorectal cancer. Expression and localization of BCAM and LAMA5 were validated by immunohistochemistry in preclinical models of human hepatic metastasis and in a panel of human specimens (n = 71). The antimetastatic efficacy of two BCAM-mimic peptides was evaluated in mouse models. The role of BCAM in the interaction of KRAS-mutant colorectal cancer cells with TME cells was investigated by adhesion assays. RESULTS: BCAM and LAMA5 were identified as molecular targets within both tumor cells and TME of KRAS-mutant hepatic metastasis from colorectal cancer, where they were specifically overexpressed. Two BCAM-mimic peptides inhibited KRAS-mutant hepatic metastasis in preclinical models. Genetic suppression and biochemical inhibition of either BCAM or LAMA5 impaired adhesion of KRAS-mutant colorectal cancer cells specifically to endothelial cells, whereas adhesion to pericytes and hepatocytes was unaffected. CONCLUSIONS: These data show that the BCAM/LAMA5 system plays a functional role in the metastatic spreading of KRAS-mutant colorectal cancer by mediating tumor-TME interactions and as such represents a valuable therapeutic candidate for this large, currently untreatable patient group. Clin Cancer Res; 22(19); 4923-33. ©2016 AACR.


Subject(s)
Cell Adhesion Molecules/metabolism , Colorectal Neoplasms/pathology , Laminin/metabolism , Liver Neoplasms/secondary , Lutheran Blood-Group System/metabolism , Neoplasm Invasiveness/genetics , Animals , Cell Adhesion/physiology , Cell Line, Tumor , Colorectal Neoplasms/genetics , Colorectal Neoplasms/metabolism , Endothelium, Vascular/pathology , Heterografts , Humans , Liver Neoplasms/genetics , Liver Neoplasms/metabolism , Mice , Mice, Nude , Paracrine Communication/physiology , Proto-Oncogene Proteins p21(ras)/genetics , Tumor Microenvironment/physiology
16.
J Mal Vasc ; 40(6): 350-8, 2015 Dec.
Article in French | MEDLINE | ID: mdl-26362408

ABSTRACT

UNLABELLED: Negative BCR ABL myeloproliferative neoplasm (MPN) such as polycythemia vera (PV), essential thrombocythemia (ET) and primary myelofibrosis (MFP) are clonal hematological malignancies and may lead to a high risk of venous, arterial or microcirculatory thrombosis. Atypical sites of thrombosis can sometimes reveal the neoplasm disorder. Their diagnoses are a major issue because of the propensity to develop acute myeloid leukemia and/or myelofibrosis. The acquired JAK2V617F variant (Janus kinase 2; 9p24) is a prevalent MPN and also a sensitive marker for PV diagnosis (95% positive mutation), but not specific since found in approximately 50% of patients with ET and MFP. PATIENT AND METHODS: We present a diagnostic and a therapeutic approach based on one patient with microcirculatory ischemic manifestations in the toes, and who had strictly normal cell blood counts and was positive for JAK2V617F mutation: thrombotic risk factor evaluation; bone marrow biopsy; red cell adhesion assays. These experimental assays are promising for the development of new therapeutics in MPN; they assess red cell adherence to the vascular endothelium after the phosphorylation of Lu/BCAM subsequent to a positive JAK2V617F mutation. RESULTS: Compared with controls, our patient exhibited increased Lu/BCAM receptor phosphorylation and red blood cell adhesion. CONCLUSION: This development may lead to improved care for patients with thrombotic manifestations, normal blood cell counts, and a positive JAK2V617F mutation: multidisciplinary management, including regular hematological monitoring, could lead to the introduction of a cytoreductive treatment.


Subject(s)
Blood Cell Count , Cell Adhesion Molecules/metabolism , Lutheran Blood-Group System/metabolism , Myeloproliferative Disorders/diagnosis , Aged , Bone Marrow/pathology , Cell Adhesion , Comorbidity , Diagnosis, Differential , Embolism, Cholesterol/diagnosis , Endothelium, Vascular/pathology , Erythrocytes/pathology , Female , France/epidemiology , Humans , Hydroxyurea/therapeutic use , Ischemia/etiology , Janus Kinase 2/genetics , Janus Kinase 2/metabolism , Laminin/metabolism , Male , Microcirculation , Mutation, Missense , Myeloproliferative Disorders/complications , Myeloproliferative Disorders/drug therapy , Myeloproliferative Disorders/epidemiology , Myeloproliferative Disorders/genetics , Phosphorylation , Point Mutation , Protein Processing, Post-Translational , Risk Factors , Smoking/adverse effects , Thrombophilia/etiology , Toes/blood supply
17.
Exp Cell Res ; 328(1): 197-206, 2014 Oct 15.
Article in English | MEDLINE | ID: mdl-25051049

ABSTRACT

Lutheran (Lu), an immunoglobulin superfamily transmembrane receptor, is also known as basal cell adhesion molecule (B-CAM). Lu/B-CAM is a specific receptor for laminin α5, a subunit of laminin-511 (LM-511) that is a major component of basement membranes in various tissues. Our previous study showed that Lu/B-CAM was cleaved by MT1-MMP and released from cell surfaces. In this study we examined the soluble Lu/B-CAM in culture media and in plasma of mice bearing HuH-7 hepatocellular carcinoma (HCC) cells and patients with HCC. Two HCC cell lines, HepG2 and HuH-7, released Lu/B-CAM into the culture media. Although Lu/B-CAM was cleaved by MT1-MMP in HuH-7 cells, HepG2 cells released Lu/B-CAM in a MMP-independent manner. The concentration of Lu/B-CAM released into mouse plasma correlated with tumor size. Moreover the soluble Lu/B-CAM in plasma of HCC patients was significantly decreased after resection of the tumor. Immunohistochemical studies showed that although the expression of Lu/B-CAM was observed in most HCCs, MT1-MMP was not always expressed in tumor tissues, suggesting that a part of Lu/B-CAM in plasma of HCC patients was also released in a MMP-independent manner. In vitro studies showed that the soluble Lu/B-CAM released from HCC cells bound to LM-511. Moreover the soluble Lu/B-CAM influenced cell migration on LM-511. These results suggest that soluble Lu/B-CAM serves as not only a novel marker for HCC but also a modulator in tumor progression.


Subject(s)
Biomarkers, Tumor/blood , Carcinoma, Hepatocellular/metabolism , Cell Adhesion Molecules/metabolism , Cell Movement , Cell Proliferation , Laminin/metabolism , Liver Neoplasms/metabolism , Lutheran Blood-Group System/metabolism , Adult , Aged , Animals , Blotting, Western , Carcinoma, Hepatocellular/pathology , Carcinoma, Hepatocellular/surgery , Female , Flow Cytometry , Glycosylation , Humans , Immunoenzyme Techniques , Immunoprecipitation , In Vitro Techniques , Liver Neoplasms/pathology , Liver Neoplasms/surgery , Male , Matrix Metalloproteinase 14/chemistry , Matrix Metalloproteinase 14/genetics , Matrix Metalloproteinase 14/metabolism , Mice , Mice, Nude , Middle Aged , Protein Interaction Domains and Motifs , RNA, Small Interfering/genetics , Tumor Cells, Cultured
18.
J Biol Chem ; 289(16): 11512-11521, 2014 Apr 18.
Article in English | MEDLINE | ID: mdl-24616094

ABSTRACT

Vaso-occlusive crises are the main acute complication in sickle cell disease. They are initiated by abnormal adhesion of circulating blood cells to vascular endothelium of the microcirculation. Several interactions involving an intricate network of adhesion molecules have been described between sickle red blood cells and the endothelial vascular wall. We have shown previously that young sickle reticulocytes adhere to resting endothelial cells through the interaction of α4ß1 integrin with endothelial Lutheran/basal cell adhesion molecule (Lu/BCAM). In the present work, we investigated the functional impact of endothelial exposure to hydroxycarbamide (HC) on this interaction using transformed human bone marrow endothelial cells and primary human pulmonary microvascular endothelial cells. Adhesion of sickle reticulocytes to HC-treated endothelial cells was decreased despite the HC-derived increase of Lu/BCAM expression. This was associated with decreased phosphorylation of Lu/BCAM and up-regulation of the cAMP-specific phosphodiesterase 4A expression. Our study reveals a novel mechanism for HC in endothelial cells where it could modulate the function of membrane proteins through the regulation of phosphodiesterase expression and cAMP-dependent signaling pathways.


Subject(s)
Anemia, Sickle Cell/metabolism , Antisickling Agents/pharmacology , Cell Adhesion Molecules/metabolism , Cyclic Nucleotide Phosphodiesterases, Type 4/biosynthesis , Endothelial Cells/metabolism , Hydroxyurea/pharmacology , Lutheran Blood-Group System/metabolism , Reticulocytes/metabolism , Anemia, Sickle Cell/drug therapy , Anemia, Sickle Cell/genetics , Anemia, Sickle Cell/pathology , Cell Adhesion/drug effects , Cell Adhesion/genetics , Cell Adhesion Molecules/genetics , Cyclic AMP/genetics , Cyclic AMP/metabolism , Cyclic Nucleotide Phosphodiesterases, Type 4/genetics , Endothelial Cells/pathology , Gene Expression Regulation, Enzymologic/drug effects , Gene Expression Regulation, Enzymologic/genetics , Humans , K562 Cells , Lutheran Blood-Group System/genetics , Phosphorylation/drug effects , Phosphorylation/genetics , Reticulocytes/pathology , Second Messenger Systems/drug effects , Second Messenger Systems/genetics , Up-Regulation/drug effects , Up-Regulation/genetics
19.
Biophys J ; 106(6): 1258-67, 2014 Mar 18.
Article in English | MEDLINE | ID: mdl-24655501

ABSTRACT

Human normal and sickle red blood cells (RBCs) adhere with high affinity to the alpha5 chain of laminin (LAMA5) via the basal cell adhesion molecule/Lutheran (BCAM/Lu) receptor, which is implicated in vasoocclusive episodes in sickle cell disease and activated through the cyclic adenosine monophosphate (cAMP) signaling pathway. However, the effect of the cAMP pathway on the expression of active BCAM/Lu receptors at the single-molecule level is unknown. We established an in vitro technique, based on atomic force microscopy, which enables detection of single BCAM/Lu proteins on the RBC surface and measures the unbinding force between BCAM/Lu and LAMA5. We showed that the expression of active BCAM/Lu receptors is higher in homozygous sickle RBCs (SS-RBCs) than normal RBCs and that it is critically dependent on the cAMP signaling pathway on both normal and SS-RBCs. Of importance, we illustrated that A-kinase anchoring proteins are crucial for BCAM/Lu receptor activation. Furthermore, we found that SS-RBCs from hydroxyurea-treated patients show a lower expression of active BCAM/Lu receptors, a lower unbinding force to LAMA5, and insignificant stimulation by epinephrine as compared to SS-RBCs from untreated patients. To our knowledge, these findings may lead to novel antiadhesive targets for vasoocclusive episodes in sickle cell disease.


Subject(s)
A Kinase Anchor Proteins/metabolism , Anemia, Sickle Cell/metabolism , Cell Adhesion Molecules/metabolism , Erythrocytes/metabolism , Lutheran Blood-Group System/metabolism , Adult , Case-Control Studies , Cyclic AMP/metabolism , Erythrocytes/ultrastructure , Humans , Laminin/metabolism , Microscopy, Atomic Force/methods , Protein Binding
20.
PLoS Pathog ; 10(1): e1003884, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24453976

ABSTRACT

The Cytotoxic Necrotizing Factor 1 (CNF1) is a protein toxin which is a major virulence factor of pathogenic Escherichia coli strains. Here, we identified the Lutheran (Lu) adhesion glycoprotein/basal cell adhesion molecule (BCAM) as cellular receptor for CNF1 by co-precipitation of cell surface molecules with tagged toxin. The CNF1-Lu/BCAM interaction was verified by direct protein-protein interaction analysis and competition studies. These studies revealed amino acids 720 to 1014 of CNF1 as the binding site for Lu/BCAM. We suggest two cell interaction sites in CNF1: first the N-terminus, which binds to p37LRP as postulated before. Binding of CNF1 to p37LRP seems to be crucial for the toxin's action. However, it is not sufficient for the binding of CNF1 to the cell surface. A region directly adjacent to the catalytic domain is a high affinity interaction site for Lu/BCAM. We found Lu/BCAM to be essential for the binding of CNF1 to cells. Cells deficient in Lu/BCAM but expressing p37LRP could not bind labeled CNF1. Therefore, we conclude that LRP and Lu/BCAM are both required for toxin action but with different functions.


Subject(s)
Bacterial Toxins/metabolism , Cell Adhesion Molecules/metabolism , Escherichia coli Proteins/metabolism , Escherichia coli/metabolism , Escherichia coli/pathogenicity , Lutheran Blood-Group System/metabolism , Virulence Factors/metabolism , Bacterial Toxins/genetics , Cell Adhesion Molecules/immunology , Escherichia coli/genetics , Escherichia coli Proteins/genetics , HEK293 Cells , HeLa Cells , Humans , Lutheran Blood-Group System/immunology , Protein Binding , Protein Structure, Tertiary , Virulence Factors/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...