Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 47
Filter
Add more filters










Publication year range
1.
Int J Mol Sci ; 25(6)2024 Mar 19.
Article in English | MEDLINE | ID: mdl-38542421

ABSTRACT

Extracellular vesicles produced by tumor cells (TEVs) influence all stages of cancer development and spread, including tumorigenesis, cancer progression, and metastasis. TEVs can trigger profound phenotypic and functional changes in target cells through three main general mechanisms: (i) docking of TEVs on target cells and triggering of intra-cellular signaling; (ii) fusion of TEVs and target cell membranes with release of TEVs molecular cargo in the cytoplasm of recipient cell; and (iii) uptake of TEVs by recipient cells. Though the overall tumor-promoting effects of TEVs as well as the general mechanisms involved in TEVs interactions with, and uptake by, recipient cells are relatively well established, current knowledge about the molecular determinants that mediate the docking and uptake of tumor-derived EVs by specific target cells is still rather deficient. These molecular determinants dictate the cell and organ tropism of TEVs and ultimately control the specificity of TEVs-promoted metastases. Here, we will review current knowledge on selected specific molecules that mediate the tropism of TEVs towards specific target cells and organs, including the integrins, ICAM-1 Inter-Cellular Adhesion Molecule), ALCAM (Activated Leukocyte Cell Adhesion Molecule), CD44, the metalloproteinases ADAM17 (A Disintegrin And Metalloproteinase member 17) and ADAM10 (A Disintegrin And Metalloproteinase member 10), and the tetraspanin CD9.


Subject(s)
Disintegrins , Extracellular Vesicles , Humans , Cell Communication , Tetraspanins/metabolism , Carcinogenesis/metabolism , Extracellular Vesicles/metabolism
2.
J Exp Clin Cancer Res ; 43(1): 27, 2024 Jan 23.
Article in English | MEDLINE | ID: mdl-38254102

ABSTRACT

BACKGROUND: Peritoneal metastasis, which accounts for 85% of all epithelial ovarian carcinoma (EOC) metastases, is a multistep process that requires the establishment of adhesive interactions between cancer cells and the peritoneal membrane. Interrelations between EOC and the mesothelial stroma are critical to facilitate the metastatic process. No data is available so far on the impact of histone acetylation/deacetylation, a potentially relevant mechanism governing EOC metastasis, on mesothelial cells (MCs)-mediated adhesion. METHODS: Static adhesion and peritoneal clearance experiments were performed pretreating mesenchymal-like MCs and platinum-sensitive/resistant EOC cell lines with MS-275-a Histone deacetylase (HDAC)1-3 pharmacological inhibitor currently used in combination trials. Results were acquired by confocal microscopy and were analyzed with an automated Opera software. The role of HDAC1/2 was validated by genetic silencing. The role of α4-, α5-α1 Integrins and Fibronectin-1 was validated using specific monoclonal antibodies. Quantitative proteomic analysis was performed on primary MCs pretreated with MS-275. Decellularized matrices were generated from either MS-275-exposed or untreated cells to study Fibronectin-1 extracellular secretion. The effect of MS-275 on ß1 integrin activity was assessed using specific monoclonal antibodies. The role of Talin-1 in MCs/EOC adhesion was analyzed by genetic silencing. Talin-1 ectopic expression was validated as a rescue tool from MS-275-induced phenotype. The in vivo effect of MS-275-induced MC remodeling was validated in a mouse model of peritoneal EOC dissemination. RESULTS: Treatment of MCs with non-cytotoxic concentrations of MS-275 caused a consistent reduction of EOC adhesion. Proteomic analysis revealed several pathways altered upon MC treatment with MS-275, including ECM deposition/remodeling, adhesion receptors and actin cytoskeleton regulators. HDAC1/2 inhibition hampered actin cytoskeleton polymerization by downregulating actin regulators including Talin-1, impairing ß1 integrin activation, and leading to abnormal extracellular secretion and distribution of Fibronectin-1. Talin-1 ectopic expression rescued EOC adhesion to MS-275-treated MCs. In an experimental mouse model of metastatic EOC, MS-275 limited tumor invasion, Fibronectin-1 secretion and the sub-mesothelial accumulation of MC-derived carcinoma-associated fibroblasts. CONCLUSION: Our study unveils a direct impact of HDAC-1/2 in the regulation of MC/EOC adhesion and highlights the regulation of MC plasticity by epigenetic inhibition as a potential target for therapeutic intervention in EOC peritoneal metastasis.


Subject(s)
Benzamides , Carcinoma, Ovarian Epithelial , Cell Adhesion , Histone Deacetylase 1 , Histone Deacetylase 2 , Ovarian Neoplasms , Peritoneal Neoplasms , Animals , Female , Humans , Mice , Actin Cytoskeleton/metabolism , Antibodies, Monoclonal , Carcinoma, Ovarian Epithelial/metabolism , Epithelium , Extracellular Matrix Proteins/metabolism , Fibronectins , Histone Deacetylase 1/metabolism , Integrin alpha5 , Integrin beta1/genetics , Ovarian Neoplasms/genetics , Ovarian Neoplasms/metabolism , Peritoneal Neoplasms/genetics , Peritoneal Neoplasms/metabolism , Proteomics , Pyridines , Talin/genetics , Talin/metabolism , Histone Deacetylase 2/metabolism , Cell Adhesion/genetics
3.
Front Immunol ; 13: 951280, 2022.
Article in English | MEDLINE | ID: mdl-36238292

ABSTRACT

Activation of the integrin phagocytic receptors CR3 (αMß2, CD11b/CD18) and CR4 (αXß2, CD11c/CD18) requires Rap1 activation and RIAM function. RIAM controls integrin activation by recruiting Talin to ß2 subunits, enabling the Talin-Vinculin interaction, which in term bridges integrins to the actin-cytoskeleton. RIAM also recruits VASP to phagocytic cups and facilitates VASP phosphorylation and function promoting particle internalization. Using a CRISPR-Cas9 knockout approach, we have analyzed the requirement for RIAM, VASP and Vinculin expression in neutrophilic-HL-60 cells. All knockout cells displayed abolished phagocytosis that was accompanied by a significant and specific reduction in ITGAM (αM), ITGAX (αX) and ITGB2 (ß2) mRNA, as revealed by RT-qPCR. RIAM, VASP and Vinculin KOs presented reduced cellular F-actin content that correlated with αM expression, as treatment with the actin filament polymerizing and stabilizing drug jasplakinolide, partially restored αM expression. In general, the expression of αX was less responsive to jasplakinolide treatment than αM, indicating that regulatory mechanisms independent of F-actin content may be involved. The Serum Response Factor (SRF) was investigated as the potential transcription factor controlling αMß2 expression, since its coactivator MRTF-A requires actin polymerization to induce transcription. Immunofluorescent MRTF-A localization in parental cells was primarily nuclear, while in knockouts it exhibited a diffuse cytoplasmic pattern. Localization of FHL-2 (SRF corepressor) was mainly sub-membranous in parental HL-60 cells, but in knockouts the localization was disperse in the cytoplasm and the nucleus, suggesting RIAM, VASP and Vinculin are required to maintain FHL-2 close to cytoplasmic membranes, reducing its nuclear localization and inhibiting its corepressor activity. Finally, reexpression of VASP in the VASP knockout resulted in a complete reversion of the phenotype, as knock-ins restored αM expression. Taken together, our results suggest that RIAM, VASP and Vinculin, are necessary for the correct expression of αMß2 and αXß2 during neutrophilic differentiation in the human promyelocytic HL-60 cell line, and strongly point to an involvement of these proteins in the acquisition of a phagocytic phenotype.


Subject(s)
Actins , Talin , Adaptor Proteins, Signal Transducing , Cell Adhesion Molecules , Co-Repressor Proteins , HL-60 Cells , Humans , Integrin alphaXbeta2 , Integrins/metabolism , Macrophage-1 Antigen , Membrane Proteins , Microfilament Proteins , Neutrophils/metabolism , Phosphoproteins , RNA, Messenger , Serum Response Factor , Talin/genetics , Talin/metabolism , Vinculin/genetics , Vinculin/metabolism
4.
Int J Mol Sci ; 23(10)2022 May 20.
Article in English | MEDLINE | ID: mdl-35628559

ABSTRACT

Colorectal cancer (CRC) and ovarian cancer (OvC) patients frequently develop peritoneal metastasis, a condition associated with a very poor prognosis. In these cancers, tumor-derived extracellular vesicles (EVs) cause immunosuppression, facilitate the direct attachment and invasion of cancer cells through the mesothelium, induce the conversion of peritoneal mesothelial cells (PMCs) into cancer-associated fibroblasts (CAFs) and transfer a more aggressive phenotype amongst cancer cells. Although the promoting role of EVs in CRC and OvC peritoneal metastasis is well established, the specific molecules that mediate the interactions between tumor-derived EVs and immune and non-immune target cells remain elusive. Here, we employed the SKOV-3 (ovarian adenocarcinoma) and Colo-320 (colorectal adenocarcinoma) human cell lines as model systems to study the interactions and uptake of EVs produced by ovarian carcinoma and colorectal carcinoma cells, respectively. We established that the adhesion molecule ALCAM/CD166 is involved in the interaction of cancer-derived EVs with recipient cancer cells (a process termed "EV binding" or "EV docking") and in their subsequent uptake by these cells. The identification of ALCAM/CD166 as a molecule mediating the docking and uptake of CRC and OvC-derived EVs may be potentially exploited to block the peritoneal metastasis cascade promoted by EVs in CRC and OvC patients.


Subject(s)
Adenocarcinoma , Antigens, CD , Cell Adhesion Molecules, Neuronal , Extracellular Vesicles , Fetal Proteins , Ovarian Neoplasms , Peritoneal Neoplasms , Activated-Leukocyte Cell Adhesion Molecule/metabolism , Adenocarcinoma/pathology , Antigens, CD/metabolism , Carcinoma, Ovarian Epithelial/pathology , Cell Adhesion Molecules, Neuronal/metabolism , Extracellular Vesicles/metabolism , Female , Fetal Proteins/metabolism , Humans , Ovarian Neoplasms/pathology , Peritoneal Neoplasms/metabolism
5.
Int J Mol Sci ; 22(21)2021 Oct 25.
Article in English | MEDLINE | ID: mdl-34768926

ABSTRACT

Most patients with ovarian cancer (OvCA) present peritoneal disseminated disease at the time of diagnosis. During peritoneal metastasis, cancer cells detach from the primary tumor and disseminate through the intraperitoneal fluid. The peritoneal mesothelial cell (PMC) monolayer that lines the abdominal cavity is the first barrier encountered by OvCA cells. Subsequent progression of tumors through the peritoneum leads to the accumulation into the peritoneal stroma of a sizeable population of carcinoma-associated fibroblasts (CAFs), which is mainly originated from a mesothelial-to-mesenchymal transition (MMT) process. A common characteristic of OvCA patients is the intraperitoneal accumulation of ascitic fluid, which is composed of cytokines, chemokines, growth factors, miRNAs, and proteins contained in exosomes, as well as tumor and mesothelial suspended cells, among other components that vary in proportion between patients. Exosomes are small extracellular vesicles that have been shown to mediate peritoneal metastasis by educating a pre-metastatic niche, promoting the accumulation of CAFs via MMT, and inducing tumor growth and chemoresistance. This review summarizes and discusses the pivotal role of exosomes and MMT as mediators of OvCA peritoneal colonization and as emerging diagnostic and therapeutic targets.


Subject(s)
Carcinoma, Ovarian Epithelial/pathology , Epithelial-Mesenchymal Transition/physiology , Exosomes/metabolism , Ovarian Neoplasms/pathology , Peritoneal Neoplasms/secondary , Ascitic Fluid/chemistry , Ascitic Fluid/cytology , Cell Line, Tumor , Cytokines/analysis , Epithelium/pathology , Female , Humans , Intercellular Signaling Peptides and Proteins/analysis , Peritoneum/pathology
6.
Int J Mol Sci ; 22(18)2021 Sep 14.
Article in English | MEDLINE | ID: mdl-34576100

ABSTRACT

Approximately 25% of colorectal cancer (CRC) patients develop peritoneal metastasis, a condition associated with a bleak prognosis. The CRC peritoneal dissemination cascade involves the shedding of cancer cells from the primary tumor, their transport through the peritoneal cavity, their adhesion to the peritoneal mesothelial cells (PMCs) that line all peritoneal organs, and invasion of cancer cells through this mesothelial cell barrier and underlying stroma to establish new metastatic foci. Exosomes produced by cancer cells have been shown to influence many processes related to cancer progression and metastasis. In epithelial ovarian cancer these extracellular vesicles (EVs) have been shown to favor different steps of the peritoneal dissemination cascade by changing the functional phenotype of cancer cells and PMCs. Little is currently known, however, about the roles played by exosomes in the pathogenesis and peritoneal metastasis cascade of CRC and especially about the molecules that mediate their interaction and uptake by target PMCs and tumor cells. We isolated exosomes by size-exclusion chromatography from CRC cells and performed cell-adhesion assays to immobilized exosomes in the presence of blocking antibodies against surface proteins and measured the uptake of fluorescently-labelled exosomes. We report here that the interaction between integrin α5ß1 on CRC cells (and PMCs) and its ligand ADAM17 on exosomes mediated the binding and uptake of CRC-derived exosomes. Furthermore, this process was negatively regulated by the expression of tetraspanin CD9 on exosomes.


Subject(s)
ADAM17 Protein/metabolism , Colorectal Neoplasms/metabolism , Exosomes/metabolism , Integrin alpha5beta1/metabolism , Adenocarcinoma/metabolism , Cell Adhesion , Cell Line, Tumor , Epithelium/pathology , Exosomes/ultrastructure , Fibronectins/metabolism , Humans , Peritoneum/pathology , Tetraspanin 29/metabolism
7.
Nat Commun ; 12(1): 4012, 2021 06 29.
Article in English | MEDLINE | ID: mdl-34188035

ABSTRACT

Recognition of laminin by integrin receptors is central to the epithelial cell adhesion to basement membrane, but the structural background of this molecular interaction remained elusive. Here, we report the structures of the prototypic laminin receptor α6ß1 integrin alone and in complex with three-chain laminin-511 fragment determined via crystallography and cryo-electron microscopy, respectively. The laminin-integrin interface is made up of several binding sites located on all five subunits, with the laminin γ1 chain C-terminal portion providing focal interaction using two carboxylate anchor points to bridge metal-ion dependent adhesion site of integrin ß1 subunit and Asn189 of integrin α6 subunit. Laminin α5 chain also contributes to the affinity and specificity by making electrostatic interactions with large surface on the ß-propeller domain of α6, part of which comprises an alternatively spliced X1 region. The propeller sheet corresponding to this region shows unusually high mobility, suggesting its unique role in ligand capture.


Subject(s)
Epithelial Cells/metabolism , Integrin alpha6/metabolism , Integrin alpha6beta1/metabolism , Integrin beta1/metabolism , Laminin/metabolism , Amino Acid Sequence , Basement Membrane/metabolism , Binding Sites/physiology , Cell Adhesion/physiology , Cryoelectron Microscopy , Crystallography, X-Ray , Humans , Protein Conformation , Protein Domains/physiology , Static Electricity
8.
Methods Mol Biol ; 2217: 47-56, 2021.
Article in English | MEDLINE | ID: mdl-33215376

ABSTRACT

Integrins are adhesion receptors that mediate many intercellular and cell-extracellular matrix interactions with relevance in physiology and pathology. Unlike other cellular receptors, integrins critically require activation for ligand binding. Through interaction in cis with other molecules and the formation of tetraspanin-enriched membrane microdomains (TEMs), the tetraspanin CD9 regulates integrin activity and avidity. Here we present three techniques used to study CD9-integrin interactions and integrin activation.


Subject(s)
Cell Adhesion/drug effects , Immunoprecipitation/methods , Lymphocyte Function-Associated Antigen-1/metabolism , Tetraspanin 28/metabolism , Tetraspanin 29/metabolism , Tetraspanin 30/metabolism , Animals , B-Lymphocytes/cytology , B-Lymphocytes/drug effects , B-Lymphocytes/metabolism , Cell Line , Cross-Linking Reagents/chemistry , Gene Expression , Humans , Jurkat Cells , Leukocytes, Mononuclear/cytology , Leukocytes, Mononuclear/drug effects , Leukocytes, Mononuclear/metabolism , Lymphocyte Function-Associated Antigen-1/genetics , Primary Cell Culture , Protein Binding , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Succinimides/chemistry , THP-1 Cells , Tetradecanoylphorbol Acetate/pharmacology , Tetraspanin 28/genetics , Tetraspanin 29/genetics , Tetraspanin 30/genetics , U937 Cells
9.
Front Immunol ; 11: 738, 2020.
Article in English | MEDLINE | ID: mdl-32425937

ABSTRACT

Phagocytic integrins are endowed with the ability to engulf and dispose of particles of different natures. Evolutionarily conserved from worms to humans, they are involved in pathogen elimination and apoptotic and tumoral cell clearance. Research in the field of integrin-mediated phagocytosis has shed light on the molecular events controlling integrin activation and their effector functions. However, there are still some aspects of the regulation of the phagocytic process that need to be clarified. Here, we have revised the molecular events controlling phagocytic integrin activation and the downstream signaling driving particle engulfment, and we have focused particularly on αMß2/CR3, αXß2/CR4, and a brief mention of αVß5/αVß3integrins.


Subject(s)
Integrins/physiology , Phagocytosis/physiology , Adaptor Proteins, Signal Transducing/physiology , Animals , Apoptosis , Humans , Integrin alphaXbeta2/physiology , Integrins/chemistry , Macrophage-1 Antigen/physiology , Membrane Proteins/physiology , Protein-Tyrosine Kinases/physiology , Signal Transduction/physiology , Talin/physiology , rap1 GTP-Binding Proteins/physiology
10.
Cells ; 9(5)2020 05 08.
Article in English | MEDLINE | ID: mdl-32397169

ABSTRACT

The phagocytic integrins and complement receptors αMß2/CR3 and αXß2/CR4 are classically associated with the phagocytosis of iC3b-opsonized particles. The activation of this receptor is dependent on signals derived from other receptors (inside-out signaling) with the crucial involvement of the Rap1-RIAM-Talin-1 pathway. Here, we analyze the implication of RIAM and its binding partner VASP in the signaling events occurring downstream of ß2 integrins (outside-in) during complement-mediated phagocytosis. To this end, we used HL-60 promyelocytic cell lines deficient in RIAM or VASP or overexpressing EGFP-tagged VASP to determine VASP dynamics at phagocytic cups. Our results indicate that RIAM-deficient HL-60 cells presented impaired particle internalization and altered integrin downstream signaling during complement-dependent phagocytosis. Similarly, VASP deficiency completely blocked phagocytosis, while VASP overexpression increased the random movement of phagocytic particles at the cell surface, with reduced internalization. Moreover, the recruitment of VASP to particle contact sites, amount of pSer157-VASP and formation of actin-rich phagocytic cups were dependent on RIAM expression. Our results suggested that RIAM worked as a relay for integrin complement receptors in outside-in signaling, coordinating integrin activation and cytoskeletal rearrangements via its interaction with VASP.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Cell Adhesion Molecules/metabolism , Integrins/metabolism , Membrane Proteins/metabolism , Microfilament Proteins/metabolism , Phagocytosis , Phosphoproteins/metabolism , Receptors, Complement/metabolism , Signal Transduction , Actins/metabolism , Cell Membrane/drug effects , Cell Membrane/metabolism , Complement System Proteins/metabolism , Gene Knockdown Techniques , HL-60 Cells , Humans , Manganese/pharmacology , Phagocytosis/drug effects , Phosphorylation/drug effects , Signal Transduction/drug effects
11.
Med Microbiol Immunol ; 209(4): 461-471, 2020 Aug.
Article in English | MEDLINE | ID: mdl-32385608

ABSTRACT

Human papillomaviruses (HPV) are causative agents of various tumours such as cervical cancer. HPV binding to the cell surface of keratinocytes leads to virus endocytosis at tetraspanin enriched microdomains. Complex interactions of the capsid proteins with host proteins as well as ADAM17-dependent ERK1/2 signal transduction enable the entry platform assembly of the oncogenic HPV type 16. Here, we studied the importance of tetraspanin CD9, also known as TSPAN29, in HPV16 infection of different epithelial cells. We found that both overexpression and loss of the tetraspanin decreased infection rates in cells with low endogenous CD9 levels, while reduction of CD9 expression in keratinocytes that exhibit high-CD9 protein amounts, led to an increase of infection. Therefore, we concluded that low-CD9 supports infection. Moreover, we found that changes in CD9 amounts affect the shedding of the ADAM17 substrate transforming growth factor alpha (TGFα) and the downstream phosphorylation of ERK. These effects correlate with those on infection rates suggesting that a specific CD9 optimum promotes ADAM17 activity, ERK signalling and virus infection. Together, our findings implicate that CD9 regulates HPV16 infection through the modulation of ADAM17 sheddase activity.


Subject(s)
ADAM17 Protein/metabolism , MAP Kinase Signaling System , Papillomavirus Infections/metabolism , Tetraspanin 29/metabolism , ADAM17 Protein/genetics , Endocytosis , Gene Expression Regulation , Gene Knockdown Techniques , HaCaT Cells , HeLa Cells , Human papillomavirus 16 , Humans , Keratinocytes/virology , Papillomavirus Infections/virology , Tetraspanin 29/genetics , Transforming Growth Factor alpha/metabolism , Virus Internalization
13.
Sci Rep ; 9(1): 10522, 2019 07 19.
Article in English | MEDLINE | ID: mdl-31324885

ABSTRACT

The outstanding potential of Extracellular Vesicles (EVs) in medicine, deserves a detailed study of the molecular aspects regulating their incorporation into target cells. However, because EV size lies below the limit of resolution of optical techniques, quantification together with discrimination between EV binding to the target cell and uptake is usually not completely achieved with current techniques. Human tetraspanins CD9 and CD63 were fused to a dual EGFP-Renilla-split tag. Subcellular localization and incorporation of these fusion proteins into EVs was assessed by western-blot and fluorescence microscopy. EV binding and uptake was measured using either a classical Renilla substrate or a cytopermeable one. Incubation of target cells expressing DSP2 with EVs containing the complementary DSP1 portion could not recover fluorescence or luciferase activity. However, using EVs carrying the fully reconstituted Dual-EGFP-Renilla protein and the cytopermeable Renilla luciferase substrate, we could distinguish EV binding from uptake. We provide proof of concept of the system by analysing the effect of different chemical inhibitors, demonstrating that this method is highly sensitive and quantitative, allowing a dynamic follow-up in a high-throughput scheme to unravel the molecular mechanisms of EV uptake in different biological systems.


Subject(s)
Extracellular Vesicles/metabolism , High-Throughput Screening Assays/methods , Tetraspanin 29/metabolism , Tetraspanin 30/metabolism , Biological Transport , Breast Neoplasms/pathology , Cell Line, Tumor , Female , Genes, Reporter , Green Fluorescent Proteins/analysis , Green Fluorescent Proteins/genetics , Humans , Imidazoles/metabolism , Luciferases, Renilla/analysis , Luciferases, Renilla/genetics , Luminescent Measurements , Nanoparticles , Pyrazines/metabolism , Recombinant Fusion Proteins/analysis , Recombinant Fusion Proteins/metabolism , Sensitivity and Specificity , Subcellular Fractions/chemistry , Tetraspanin 29/genetics , Tetraspanin 30/genetics
14.
Front Immunol ; 9: 2655, 2018.
Article in English | MEDLINE | ID: mdl-30515156

ABSTRACT

The regulatory role of most dual specific phosphatases during T cell activation remains unknown. Here, we have studied the expression and function of phosphatases of regenerating liver (PRLs: PRL-1, PRL-2, and PRL-3) during T cell activation, as well as, the dynamic delivery of PRL-1 to the Immunological Synapse (IS). We found that T cell activation downregulates the expression of PRL-2, resulting in an increased PRL-1/PRL-2 ratio. PRL-1 redistributed at the IS in two stages: Initially, it was transiently accumulated at scanning membranes enriched in CD3 and actin, and at later times, it was delivered at the contact site from pericentriolar, CD3ζ-containing, vesicles. Once at the established IS, PRL-1 distributed to LFA-1 and CD3ε sites. Remarkably, PRL-1 was found to regulate actin dynamics during IS assembly and the secretion of IL-2. Moreover, pharmacological inhibition of the catalytic activity of the three PRLs reduced the secretion of IL-2. These results provide evidence indicating a regulatory role of PRL-1 during IS assembly and highlight the involvement of PRLs in immune responses by mature T cells.


Subject(s)
Actins/immunology , Cell Cycle Proteins/immunology , Immunological Synapses/immunology , Lymphocyte Activation , Membrane Proteins/immunology , Protein Tyrosine Phosphatases/immunology , T-Lymphocytes/immunology , CD3 Complex/immunology , Female , Humans , Interleukin-2/immunology , Lymphocyte Function-Associated Antigen-1/immunology , Male
15.
Front Immunol ; 9: 2474, 2018.
Article in English | MEDLINE | ID: mdl-30455686

ABSTRACT

Integrin α5ß1 is a crucial adhesion molecule that mediates the adherence of many cell types to the extracellular matrix through recognition of its classic ligand fibronectin as well as to other cells through binding to an alternative counter-receptor, the metalloproteinase ADAM17/TACE. Interactions between integrin α5ß1 and ADAM17 may take place both in trans (between molecules expressed on different cells) or in cis (between molecules expressed on the same cell) configurations. It has been recently reported that the cis association between α5ß1 and ADAM17 keeps both molecules inactive, whereas their dissociation results in activation of their adhesive and metalloproteinase activities. Here we show that the tetraspanin CD9 negatively regulates integrin α5ß1-mediated cell adhesion by enhancing the cis interaction of this integrin with ADAM17 on the cell surface. Additionally we show that, similarly to CD9, the monoclonal antibody 2A10 directed to the disintegrin domain of ADAM17 specifically inhibits integrin α5ß1-mediated cell adhesion to its ligands fibronectin and ADAM17.


Subject(s)
ADAM17 Protein/metabolism , Leukocytes/immunology , Neoplastic Cells, Circulating/immunology , Tetraspanin 29/metabolism , ADAM17 Protein/genetics , ADAM17 Protein/immunology , Antibodies, Monoclonal/metabolism , CRISPR-Cas Systems , Cell Adhesion , Fibronectins/metabolism , Gene Knockdown Techniques , Humans , Integrin alpha5beta1/metabolism , K562 Cells , Protein Binding
16.
Front Immunol ; 9: 738, 2018.
Article in English | MEDLINE | ID: mdl-29760691

ABSTRACT

Cells release membrane enclosed nano-sized vesicles termed extracellular vesicles (EVs) that function as mediators of intercellular communication by transferring biological information between cells. Tumor-derived EVs have emerged as important mediators in cancer development and progression, mainly through transfer of their bioactive content which can include oncoproteins, oncogenes, chemokine receptors, as well as soluble factors, transcripts of proteins and miRNAs involved in angiogenesis or inflammation. This transfer has been shown to influence the metastatic behavior of primary tumors. Moreover, tumor-derived EVs have been shown to influence distant cellular niches, establishing favorable microenvironments that support growth of disseminated cancer cells upon their arrival at these pre-metastatic niches. It is generally accepted that cells release a number of major EV populations with distinct biophysical properties and biological functions. Exosomes, microvesicles, and apoptotic bodies are EV populations most widely studied and characterized. They are discriminated based primarily on their intracellular origin. However, increasing evidence suggests that even within these EV populations various subpopulations may exist. This heterogeneity introduces an extra level of complexity in the study of EV biology and function. For example, EV subpopulations could have unique roles in the intricate biological processes underlying cancer biology. Here, we discuss current knowledge regarding the role of subpopulations of EVs in cancer development and progression and highlight the relevance of EV heterogeneity. The position of tetraspanins and integrins therein will be highlighted. Since addressing EV heterogeneity has become essential for the EV field, current and novel techniques for isolating EV subpopulations will also be discussed. Further dissection of EV heterogeneity will advance our understanding of the critical roles of EVs in health and disease.


Subject(s)
Extracellular Vesicles/physiology , Neoplasms/pathology , Animals , Disease Progression , Humans
17.
Front Immunol ; 9: 863, 2018.
Article in English | MEDLINE | ID: mdl-29760699

ABSTRACT

The tetraspanin CD9 is expressed by all the major subsets of leukocytes (B cells, CD4+ T cells, CD8+ T cells, natural killer cells, granulocytes, monocytes and macrophages, and immature and mature dendritic cells) and also at a high level by endothelial cells. As a typical member of the tetraspanin superfamily, a prominent feature of CD9 is its propensity to engage in a multitude of interactions with other tetraspanins as well as with different transmembrane and intracellular proteins within the context of defined membranal domains termed tetraspanin-enriched microdomains (TEMs). Through these associations, CD9 influences many cellular activities in the different subtypes of leukocytes and in endothelial cells, including intracellular signaling, proliferation, activation, survival, migration, invasion, adhesion, and diapedesis. Several excellent reviews have already covered the topic of how tetraspanins, including CD9, regulate these cellular processes in the different cells of the immune system. In this mini-review, however, we will focus particularly on describing and discussing the regulatory effects exerted by CD9 on different adhesion molecules that play pivotal roles in the physiology of leukocytes and endothelial cells, with a particular emphasis in the regulation of adhesion molecules of the integrin and immunoglobulin superfamilies.


Subject(s)
Cell Adhesion/immunology , Endothelial Cells/immunology , Leukocytes/immunology , Tetraspanin 29/immunology , Animals , Humans
18.
Sci Rep ; 7(1): 11271, 2017 09 12.
Article in English | MEDLINE | ID: mdl-28900146

ABSTRACT

Most experimental approaches commonly employed for the characterization and quantitation of EVs are time consuming, require of specialized instrumentation and often are rather inaccurate. To circumvent the caveats imposed by EV small size, we used general and specific membrane markers in bead assisted flow cytometry, to provide a semi-quantitative measure of EV content in a given sample. EVs were isolated from in vitro cultured cells-conditioned medium and biological fluids by size exclusion chromatography and coupled to latex beads to allow their detection by standard flow cytometers. Our analyses demonstrate a linear correlation between EV concentration and Mean Fluorescence Intensity values in samples cleared of protein contaminants. Comparison with one of the most widespread method such as NTA, suggests a similar linear range and reliable accuracy to detect saturation. However, although detection of the different biomarkers is feasible when tested on ultracentrifugation-enriched samples, protein contamination impairs quantitation of this type of samples by bead-based flow cytometry. Thus, we provide evidence that bead-assisted flow cytometry method is an accurate and reliable method for the semiquantitative bulk analysis of EVs, which could be easily implemented in most laboratories.


Subject(s)
Extracellular Vesicles/metabolism , Flow Cytometry , Biomarkers , Cell Line , Chromatography, Gel , Flow Cytometry/methods , Humans , Nanoparticles
19.
EMBO J ; 36(5): 629-645, 2017 03 01.
Article in English | MEDLINE | ID: mdl-28122868

ABSTRACT

We show that the three conformational states of integrin α5ß1 have discrete free energies and define activation by measuring intrinsic affinities for ligand of each state and the equilibria linking them. The 5,000-fold higher affinity of the extended-open state than the bent-closed and extended-closed states demonstrates profound regulation of affinity. Free energy requirements for activation are defined with protein fragments and intact α5ß1 On the surface of K562 cells, α5ß1 is 99.8% bent-closed. Stabilization of the bent conformation by integrin transmembrane and cytoplasmic domains must be overcome by cellular energy input to stabilize extension. Following extension, headpiece opening is energetically favored. N-glycans and leg domains in each subunit that connect the ligand-binding head to the membrane repel or crowd one another and regulate conformational equilibria in favor of headpiece opening. The results suggest new principles for regulating signaling in the large class of receptors built from extracellular domains in tandem with single-span transmembrane domains.


Subject(s)
Integrin alpha5beta1/chemistry , Integrin alpha5beta1/metabolism , Cell Line , Humans , Models, Molecular , Protein Binding , Protein Conformation , Thermodynamics
20.
Proc Natl Acad Sci U S A ; 113(27): E3872-81, 2016 07 05.
Article in English | MEDLINE | ID: mdl-27317747

ABSTRACT

Whether ß1 integrin ectodomains visit conformational states similarly to ß2 and ß3 integrins has not been characterized. Furthermore, despite a wealth of activating and inhibitory antibodies to ß1 integrins, the conformational states that these antibodies stabilize, and the relation of these conformations to function, remain incompletely characterized. Using negative-stain electron microscopy, we show that the integrin α5ß1 ectodomain adopts extended-closed and extended-open conformations as well as a bent conformation. Antibodies SNAKA51, 8E3, N29, and 9EG7 bind to different domains in the α5 or ß1 legs, activate, and stabilize extended ectodomain conformations. Antibodies 12G10 and HUTS-4 bind to the ß1 ßI domain and hybrid domains, respectively, activate, and stabilize the open headpiece conformation. Antibody TS2/16 binds a similar epitope as 12G10, activates, and appears to stabilize an open ßI domain conformation without requiring extension or hybrid domain swing-out. mAb13 and SG/19 bind to the ßI domain and ßI-hybrid domain interface, respectively, inhibit, and stabilize the closed conformation of the headpiece. The effects of the antibodies on cell adhesion to fibronectin substrates suggest that the extended-open conformation of α5ß1 is adhesive and that the extended-closed and bent-closed conformations are nonadhesive. The functional effects and binding sites of antibodies and fibronectin were consistent with their ability in binding to α5ß1 on cell surfaces to cross-enhance or inhibit one another by competitive or noncompetitive (allosteric) mechanisms.


Subject(s)
Integrin alpha5beta1/chemistry , Escherichia coli , Fibronectins/metabolism , Humans , Integrin alpha5beta1/metabolism , Protein Conformation , Structure-Activity Relationship
SELECTION OF CITATIONS
SEARCH DETAIL
...