Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 80
Filter
1.
J Biol Chem ; 300(3): 105755, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38364890

ABSTRACT

XK-related 8 (XKR8), in complex with the transmembrane glycoprotein basigin, functions as a phospholipid scramblase activated by the caspase-mediated cleavage or phosphorylation of its C-terminal tail. It carries a putative phospholipid translocation path of multiple hydrophobic and charged residues in the transmembrane region. It also has a crucial tryptophan at the exoplasmic end of the path that regulates its scrambling activity. We herein investigated the tertiary structure of the human XKR8-basigin complex embedded in lipid nanodiscs at an overall resolution of 3.66 Å. We found that the C-terminal tail engaged in intricate polar and van der Waals interactions with a groove at the cytoplasmic surface of XKR8. These interactions maintained the inactive state of XKR8. Point mutations to disrupt these interactions strongly enhanced the scrambling activity of XKR8, suggesting that the activation of XKR8 is mediated by releasing the C-terminal tail from the cytoplasmic groove. We speculate that the cytoplasmic tail region of XKR8 functions as a plug to prevent the scrambling of phospholipids.


Subject(s)
Apoptosis Regulatory Proteins , Basigin , Membrane Proteins , Phospholipid Transfer Proteins , Humans , Apoptosis Regulatory Proteins/chemistry , Apoptosis Regulatory Proteins/genetics , Basigin/chemistry , Cell Membrane/metabolism , Liposomes/chemistry , Membrane Proteins/chemistry , Membrane Proteins/genetics , Nanoparticles/chemistry , Phospholipid Transfer Proteins/chemistry , Phospholipid Transfer Proteins/genetics , Phospholipids , Protein Conformation, alpha-Helical , Single Molecule Imaging
2.
Mol Vis ; 29: 13-24, 2023.
Article in English | MEDLINE | ID: mdl-37287642

ABSTRACT

Purpose: Basigin gene products are positioned on adjacent cell types in the neural retina and are thought to compose a lactate metabolon important for photoreceptor cell function. The Ig0 domain of basigin isoform 1 (basigin-1) is highly conserved throughout evolution, which suggests a conserved function. It has been suggested that the Ig0 domain has proinflammatory properties, and it is hypothesized to interact with basigin isoform 2 (basigin-2) for cell adhesion and lactate metabolon formation. Therefore, the purpose of the present study was to determine whether the Ig0 domain of basigin-1 binds to basigin-2 and whether the region of the domain used for binding is also used to stimulate interleukin-6 (IL-6) expression. Methods: Binding was assessed using recombinant proteins corresponding to the Ig0 domain of basigin-1 and endogenously expressed basigin-2 from mouse neural retina and brain protein lysates. The proinflammatory properties of the Ig0 domain were analyzed with exposure of the recombinant proteins to the mouse monocyte RAW 264.7 cell line and subsequent measurement of the IL-6 concentration in the culture medium via enzyme-linked immunosorbent assay (ELISA). Results: The data indicate that the Ig0 domain interacts with basigin-2 through a region within the amino half of the domain and that the Ig0 domain does not stimulate the expression of IL-6 in mouse cells in vitro. Conclusions: The Ig0 domain of basigin-1 binds to basigin-2 in vitro. In addition, contrary to previous reports, there was no evidence that the Ig0 domain potentiates IL-6 expression in a mouse monocyte cell line in vitro. However, it is possible that the Ig0 domain stimulates the expression of proinflammatory cytokines other than IL-6, or that the potential involvement of the Ig0 domain of basigin-1 in the acute inflammatory response is dependent on species.


Subject(s)
Basigin , Interleukin-6 , Mice , Animals , Basigin/chemistry , Basigin/genetics , Basigin/metabolism , Interleukin-6/genetics , Interleukin-6/metabolism , Monocytes , Retina/metabolism , Protein Isoforms/genetics , Protein Isoforms/metabolism , Recombinant Proteins/metabolism , Lactates/metabolism
3.
Nat Struct Mol Biol ; 28(10): 825-834, 2021 10.
Article in English | MEDLINE | ID: mdl-34625749

ABSTRACT

Xkr8-Basigin is a plasma membrane phospholipid scramblase activated by kinases or caspases. We combined cryo-EM and X-ray crystallography to investigate its structure at an overall resolution of 3.8 Å. Its membrane-spanning region carrying 22 charged amino acids adopts a cuboid-like structure stabilized by salt bridges between hydrophilic residues in transmembrane helices. Phosphatidylcholine binding was observed in a hydrophobic cleft on the surface exposed to the outer leaflet of the plasma membrane. Six charged residues placed from top to bottom inside the molecule were essential for scrambling phospholipids in inward and outward directions, apparently providing a pathway for their translocation. A tryptophan residue was present between the head group of phosphatidylcholine and the extracellular end of the path. Its mutation to alanine made the Xkr8-Basigin complex constitutively active, indicating that it plays a vital role in regulating its scramblase activity. The structure of Xkr8-Basigin provides insights into the molecular mechanisms underlying phospholipid scrambling.


Subject(s)
Apoptosis Regulatory Proteins/chemistry , Basigin/chemistry , Cell Membrane/metabolism , Membrane Proteins/chemistry , Phospholipids/metabolism , Apoptosis Regulatory Proteins/metabolism , Basigin/metabolism , Cryoelectron Microscopy , Humans , Hydrophobic and Hydrophilic Interactions , Membrane Proteins/metabolism , Models, Molecular , Multiprotein Complexes/chemistry , Multiprotein Complexes/isolation & purification , Multiprotein Complexes/metabolism , Phosphatidylcholines/chemistry , Phosphatidylcholines/metabolism , Phospholipid Transfer Proteins/chemistry , Phospholipid Transfer Proteins/metabolism , Phospholipids/chemistry , Protein Structure, Tertiary , Tryptophan/chemistry
4.
FEBS J ; 288(20): 6052-6062, 2021 10.
Article in English | MEDLINE | ID: mdl-33999492

ABSTRACT

Monocarboxylate transporter isoforms 1-4, MCT, of the solute carrier SLC16A family facilitate proton-coupled transport of l-lactate. Growth of tumors that exhibit the Warburg effect, that is, high rates of anaerobic glycolysis despite availability of oxygen, relies on swift l-lactate export, whereas oxygenic cancer cells import circulating l-lactate as a fuel. Currently, MCTs are viewed as promising anticancer targets. Small-molecule inhibitors have been found, and, recently, high-resolution protein structures have been obtained. Key questions, however, regarding the exact binding sites of cysteine-modifying inhibitors and the substrate translocation cycle lack a conclusive experimental basis. Here, we report Cys159 of the ubiquitous human MCT1 to reside in a critical hinge region of the alternating access-type transporter. We identified Cys159 as the binding site of the organomercurial pCMBS. The inhibitory effect of pCMBS was proposed to be indirect via modification of the chaperone basigin. We provide evidence that pCMBS locks MCT1 in its outward open conformation in a wedge-like fashion. We corroborated this finding using smaller cysteine-modifying reagents that size-dependently inhibited l-lactate transport. The smallest modifiers targeted additional cysteines as shown by a C159S mutant. We found a Cys399/Cys400 pair to constitute the second hinge of the transporter that tolerated only individual replacement by serine. The hinge cysteines, in particular the selectively addressable Cys159, provide natural anchors for placing probes into MCTs to report, for instance, on the electrostatics or hydration upon binding of the transported l-lactate substrate and the proton cosubstrate.


Subject(s)
4-Chloromercuribenzenesulfonate/pharmacology , Basigin/chemistry , Cysteine/chemistry , Enzyme Inhibitors/pharmacology , Monocarboxylic Acid Transporters/antagonists & inhibitors , Small Molecule Libraries/pharmacology , Symporters/antagonists & inhibitors , Basigin/genetics , Basigin/metabolism , Humans , Monocarboxylic Acid Transporters/metabolism , Protein Conformation , Symporters/metabolism
5.
PLoS One ; 16(3): e0249110, 2021.
Article in English | MEDLINE | ID: mdl-33770122

ABSTRACT

Transmembrane transport of l-lactate by members of the monocarboxylate transporter family, MCT, is vital in human physiology and a malignancy factor in cancer. Interaction with an accessory protein, typically basigin, is required to deliver the MCT to the plasma membrane. It is unknown whether basigin additionally exerts direct effects on the transmembrane l-lactate transport of MCT1. Here, we show that the presence of basigin leads to an intracellular accumulation of l-lactate 4.5-fold above the substrate/proton concentrations provided by the external buffer. Using basigin truncations we localized the effect to arise from the extracellular Ig-I domain. Identification of surface patches of condensed opposite electrostatic potential, and experimental analysis of charge-affecting Ig-I mutants indicated a bivalent harvesting antenna functionality for both, protons and substrate anions. From these data, and determinations of the cytosolic pH with a fluorescent probe, we conclude that the basigin Ig-I domain drives lactate uptake by locally increasing the proton and substrate concentration at the extracellular MCT entry site. The biophysical properties are physiologically relevant as cell growth on lactate media was strongly promoted in the presence of the Ig-I domain. Lack of the domain due to shedding, or misfolding due to breakage of a stabilizing disulfide bridge reversed the effect. Tumor progression according to classical or reverse Warburg effects depends on the transmembrane l-lactate distribution, and this study shows that the basigin Ig-I domain is a pivotal determinant.


Subject(s)
Basigin/metabolism , Intracellular Space/metabolism , Lactic Acid/metabolism , Basigin/chemistry , Cell Line, Tumor , Humans , Protein Domains , Protein Transport
6.
ACS Appl Bio Mater ; 4(5): 4422-4431, 2021 05 17.
Article in English | MEDLINE | ID: mdl-35006854

ABSTRACT

In the treatment of tumor-targeted small-molecule anti-cancer drugs, antibody-mediated therapies, especially for antibody-drug conjugates (ADCs), have revealed great latent force. However, the therapeutic drugs provided by ADCs possess limitation. Considering that the combination of antibodies and nano-drugs can broaden their applicability in the field of tumor treatment, herein, we developed an antibody conjugated polymeric prodrug nanoparticles SAE-PEG-b-PBYP-ss-CPT for targeted camptothecin (CPT) delivery to liver tumor cells. The diblock copolymer was composed of PEG and biodegradable polyphosphoester (PBYP) containing alkynyl groups in the side chain. A derivative of CPT (CPT-ss-N3) was bonded to the PBYP via "click" reaction. The diethyl squarate (SAE) in the terminal of PEG chain was used as a functional group to bond with CD147 monoclonal antibody (CD147 mAb). The particle size and size distribution of the both nanoparticles, with antibody binding (namely CD147-CPT NPs) and without antibody (abbreviated as CPT-loaded NPs), were measured by dynamic light scattering (DLS). The morphologies of both two kinds of nanoparticles were observed by transmission electron microscope (TEM). The results of X-ray photoelectron spectroscopy (XPS) showed that CD147 mAb had been coupled to the surface of CPT-loaded NPs. Endocytosis test indicated that CD147-CPT NPs had higher uptake rate and accumulation in HepG2 cells than those of CPT-loaded NPs without antibodies, due to CD147 mAb can specifically bind to CD147 protein overexpressed in HepG2 cells. We establish a method to bond monoclonal antibodies to anti-cancer polymeric prodrugs, and endow biodegradable polymeric prodrugs with precise targeting functions to liver cancer cells.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Basigin/chemistry , Biocompatible Materials/pharmacology , Camptothecin/pharmacology , Carcinoma, Hepatocellular/drug therapy , Immunoconjugates/pharmacology , Liver Neoplasms/drug therapy , Antineoplastic Agents, Phytogenic/chemistry , Basigin/genetics , Basigin/metabolism , Biocompatible Materials/chemistry , Camptothecin/chemistry , Carcinoma, Hepatocellular/pathology , Cell Line , Cell Survival/drug effects , Drug Delivery Systems , Drug Screening Assays, Antitumor , Humans , Immunoconjugates/chemistry , Liver Neoplasms/pathology , Materials Testing , Molecular Structure , Nanomedicine , Particle Size , Prodrugs/chemistry , Prodrugs/pharmacology
7.
Cell ; 184(2): 370-383.e13, 2021 01 21.
Article in English | MEDLINE | ID: mdl-33333023

ABSTRACT

Proton-coupled monocarboxylate transporters MCT1-4 catalyze the transmembrane movement of metabolically essential monocarboxylates and have been targeted for cancer treatment because of their enhanced expression in various tumors. Here, we report five cryo-EM structures, at resolutions of 3.0-3.3 Å, of human MCT1 bound to lactate or inhibitors in the presence of Basigin-2, a single transmembrane segment (TM)-containing chaperon. MCT1 exhibits similar outward-open conformations when complexed with lactate or the inhibitors BAY-8002 and AZD3965. In the presence of the inhibitor 7ACC2 or with the neutralization of the proton-coupling residue Asp309 by Asn, similar inward-open structures were captured. Complemented by structural-guided biochemical analyses, our studies reveal the substrate binding and transport mechanism of MCTs, elucidate the mode of action of three anti-cancer drug candidates, and identify the determinants for subtype-specific sensitivities to AZD3965 by MCT1 and MCT4. These findings lay out an important framework for structure-guided drug discovery targeting MCTs.


Subject(s)
Antineoplastic Agents/pharmacology , Monocarboxylic Acid Transporters/antagonists & inhibitors , Monocarboxylic Acid Transporters/chemistry , Symporters/antagonists & inhibitors , Symporters/chemistry , Amino Acid Sequence , Animals , Basigin/chemistry , Binding Sites , Cryoelectron Microscopy , Humans , Ligands , Models, Molecular , Monocarboxylic Acid Transporters/ultrastructure , Mutant Proteins/chemistry , Mutant Proteins/metabolism , Protons , Pyrimidinones/chemistry , Pyrimidinones/pharmacology , Rats , Structural Homology, Protein , Substrate Specificity , Symporters/ultrastructure , Thiophenes/chemistry , Thiophenes/pharmacology
8.
Chembiochem ; 22(7): 1201-1204, 2021 04 06.
Article in English | MEDLINE | ID: mdl-33174659

ABSTRACT

Fluorescent fusion proteins are powerful tools for studying biological processes in living cells, but universal application is limited due to the voluminous size of those tags, which might have an impact on the folding, localization or even the biological function of the target protein. The designed biocatalyst trypsiligase enables site-directed linkage of small-sized fluorescence dyes on the N terminus of integral target proteins located in the outer membrane of living cells through a stable native peptide bond. The function of the approach was tested by using the examples of covalent derivatization of the transmembrane proteins CD147 as well as the EGF receptor, both presented on human HeLa cells. Specific trypsiligase recognition of the site of linkage was mediated by the dipeptide sequence Arg-His added to the proteins' native N termini, pointing outside the cell membrane. The labeling procedure takes only about 5 minutes, as demonstrated for couplings of the fluorescence dye tetramethyl rhodamine and the affinity label biotin as well.


Subject(s)
Basigin/metabolism , ErbB Receptors/metabolism , Fluorescent Dyes/metabolism , Trypsin/metabolism , Basigin/chemistry , Biocatalysis , Dipeptides/metabolism , ErbB Receptors/chemistry , Fluorescent Dyes/chemistry , Green Fluorescent Proteins/chemistry , Green Fluorescent Proteins/metabolism , HeLa Cells , Humans , Microscopy, Confocal , Substrate Specificity , Trypsin/genetics
9.
Med Hypotheses ; 144: 110155, 2020 Nov.
Article in English | MEDLINE | ID: mdl-33254482

ABSTRACT

The world is experiencing one of the most difficult moments in history with the COVID-19 pandemic, a disease caused by SARS-CoV-2, a new type of coronavirus. Virus infectivity is mediated by the binding of Spike transmembrane glycoprotein to specific protein receptors present on cell host surface. Spike is a homotrimer that emerges from the virion, each monomer containing two subunits named S1 and S2, which are related to cell recognition and membrane fusion, respectively. S1 is subdivided in domains S1A (or NTD) and S1B (or RBD), with experimental and in silico studies suggesting that the former binds to sialic acid-containing glycoproteins, such as CD147, whereas the latter binds to ACE2 receptor. Recent findings indicate that the ABO blood system modulates susceptibility and progression of infection, with type-A individuals being more susceptible to infection and/or manifestation of a severe condition. Seeking to understand the molecular mechanisms underlying this susceptibility, we carried out an extensive bibliographic survey on the subject. Based on this survey, we hypothesize that the correlation between the ABO blood system and susceptibility to SARS-CoV-2 infection can be presumably explained by the modulation of sialic acid-containing receptors distribution on host cell surface induced by ABO antigens through carbohydrate-carbohydrate interactions, which could maximize or minimize the virus Spike protein binding to the host cell. This model could explain previous sparse observations on the molecular mechanism of infection and can direct future research to better understand of COVID-19 pathophysiology.


Subject(s)
ABO Blood-Group System , COVID-19/blood , COVID-19/diagnosis , Carbohydrates/chemistry , Disease Susceptibility , Spike Glycoprotein, Coronavirus/chemistry , Angiotensin-Converting Enzyme 2/chemistry , Animals , Basigin/chemistry , Binding Sites , COVID-19/epidemiology , Humans , Models, Theoretical , Molecular Dynamics Simulation , Protein Binding , Protein Domains , SARS-CoV-2 , Virus Internalization
11.
Oncogene ; 39(8): 1710-1723, 2020 02.
Article in English | MEDLINE | ID: mdl-31723238

ABSTRACT

Tumor cells rely on glycolysis to meet their elevated demand for energy. Thereby they produce significant amounts of lactate and protons, which are exported via monocarboxylate transporters (MCTs), supporting the formation of an acidic microenvironment. The present study demonstrates that carbonic anhydrase IX (CAIX), one of the major acid/base regulators in cancer cells, forms a protein complex with MCT1 and MCT4 in tissue samples from human breast cancer patients, but not healthy breast tissue. Formation of this transport metabolon requires binding of CAIX to the Ig1 domain of the MCT1/4 chaperon CD147 and is required for CAIX-mediated facilitation of MCT1/4 activity. Application of an antibody, directed against the CD147-Ig1 domain, displaces CAIX from the transporter and suppresses CAIX-mediated facilitation of proton-coupled lactate transport. In cancer cells, this "metabolon disruption" results in a decrease in lactate transport, reduced glycolysis, and ultimately reduced cell proliferation. Taken together, the study shows that carbonic anhydrases form transport metabolons with acid/base transporters in human tumor tissue and that these interactions can be exploited to interfere with tumor metabolism and proliferation.


Subject(s)
Breast Neoplasms/pathology , Carbonic Anhydrase IX/metabolism , Monocarboxylic Acid Transporters/metabolism , Muscle Proteins/metabolism , Symporters/metabolism , Basigin/chemistry , Basigin/metabolism , Humans , MCF-7 Cells , Models, Molecular , Protein Domains
12.
PLoS Biol ; 17(10): e3000490, 2019 10.
Article in English | MEDLINE | ID: mdl-31613878

ABSTRACT

Many important infectious diseases are the result of zoonoses, in which pathogens that normally infect animals acquire mutations that enable the breaching of species barriers to permit the infection of humans. Our understanding of the molecular events that enable host switching are often limited, and yet this is a fundamentally important question. Plasmodium falciparum, the etiological agent of severe human malaria, evolved following a zoonotic transfer of parasites from gorillas. One gene-rh5-which encodes an essential ligand for the invasion of host erythrocytes, is suspected to have played a critical role in this host switch. Genome comparisons revealed an introgressed sequence in the ancestor of P. falciparum containing rh5, which likely allowed the ancestral parasites to infect both gorilla and human erythrocytes. To test this hypothesis, we resurrected the ancestral introgressed reticulocyte-binding protein homologue 5 (RH5) sequence and used quantitative protein interaction assays to demonstrate that this ancestral protein could bind the basigin receptor from both humans and gorillas. We also showed that this promiscuous receptor binding phenotype of RH5 was shared with the parasite clade that transferred its genome segment to the ancestor of P. falciparum, while the other lineages exhibit host-specific receptor binding, confirming the central importance of this introgression event for Plasmodium host switching. Finally, since its transfer to humans, P. falciparum, and also the RH5 ligand, have evolved a strong human specificity. We show that this subsequent restriction to humans can be attributed to a single amino acid mutation in the RH5 sequence. Our findings reveal a molecular pathway for the origin and evolution of human P. falciparum malaria and may inform molecular surveillance to predict future zoonoses.


Subject(s)
Basigin/genetics , Carrier Proteins/genetics , Genome, Protozoan , Malaria, Falciparum/transmission , Malaria, Falciparum/veterinary , Plasmodium falciparum/genetics , Amino Acid Substitution , Animals , Basigin/chemistry , Basigin/metabolism , Binding Sites , Carrier Proteins/chemistry , Carrier Proteins/metabolism , Erythrocytes/parasitology , Gene Expression , Genetic Introgression , Gorilla gorilla/parasitology , History, Ancient , Host Specificity , Humans , Malaria, Falciparum/epidemiology , Malaria, Falciparum/history , Models, Molecular , Mutation , Phylogeny , Plasmodium falciparum/classification , Plasmodium falciparum/metabolism , Plasmodium falciparum/pathogenicity , Protein Binding , Protein Structure, Secondary , Zoonoses
13.
Cell Death Dis ; 10(6): 462, 2019 06 12.
Article in English | MEDLINE | ID: mdl-31189879

ABSTRACT

An increased surface level of CIE (clathrin-independent endocytosis) proteins is a new feature of malignant neoplasms. CD147 is a CIE glycoprotein highly up-regulated in hepatocellular carcinoma (HCC). The ability to sort out the early endosome and directly target the recycling pathway confers on CD147 a prolonged surface half-life. However, current knowledge on CD147 trafficking to and from the cell-surface is limited. In this study, an MSP (membrane and secreted protein)-cDNA library was screened against EpoR/LR-F3/CD147EP-expressed cells by MAPPIT (mammalian protein-protein interaction trap). CD147 co-expressing with the new binder was investigated by GEPIA (gene expression profiling interactive analysis). The endocytosis, ER-Golgi trafficking and recycling of CD147 were measured by confocal imaging, flow cytometry, and biotin-labeled chase assays, respectively. Rab GTPase activation was checked by GST-RBD pull-down and MMP activity was measured by gelatin zymography. HCC malignant phenotypes were determined by cell adhesion, proliferation, migration, Transwell motility, and invasion assays. An ER-Golgi-resident transmembrane protein YIPF2 was identified as an intracellular binder to CD147. YIPF2 correlated and co-expressed with CD147, which is a survival predictor for HCC patients. YIPF2 is critical for CD147 glycosylation and trafficking functions in HCC cells. YIPF2 acts as a Rab-GDF (GDI-displacement factor) regulating three independent trafficking steps. First, YIPF2 recruits and activates Rab5 and Rab22a GTPases to the endomembrane structures. Second, YIPF2 modulates the endocytic recycling of CD147 through distinctive regulation on Rab5 and Rab22a. Third, YIPF2 mediates the mature processing of CD147 via the ER-Golgi trafficking route. Decreased YIPF2 expression induced a CD147 efficient delivery to the cell-surface, promoted MMP secretion, and enhanced the adhesion, motility, migration, and invasion behaviors of HCC cells. Thus, YIPF2 is a new trafficking determinant essential for CD147 glycosylation and transport. Our findings revealed a novel YIPF2-controlled ER-Golgi trafficking signature that promotes CD147-medated malignant phenotypes in HCC.


Subject(s)
Basigin/metabolism , Carcinoma, Hepatocellular/metabolism , Endocytosis , Liver Neoplasms/metabolism , Membrane Proteins/metabolism , rab GTP-Binding Proteins/metabolism , 3T3 Cells , Animals , Basigin/chemistry , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/mortality , Cell Adhesion , Cell Line, Tumor , Cell Movement , Endocytosis/genetics , Endoplasmic Reticulum/metabolism , Glycosylation , Golgi Apparatus/metabolism , Humans , Liver Neoplasms/genetics , Liver Neoplasms/mortality , Matrix Metalloproteinases/metabolism , Membrane Proteins/genetics , Mice , Protein Transport/genetics , rab5 GTP-Binding Proteins/metabolism
14.
Cell Oncol (Dordr) ; 42(4): 537-554, 2019 Aug.
Article in English | MEDLINE | ID: mdl-31016558

ABSTRACT

PURPOSE: CD147 is a tumor-associated antigen that plays a key regulatory role in tumor invasion and distant metastasis. However, the exact role of CD147 phosphorylation, which is deregulated during cancer progression, is unknown. Here, the effects of CD147 phosphorylation on the malignant behavior of hepatocellular carcinoma (HCC) cells and its possible underlying mechanisms are explored. METHODS: An in situ Duolink-proximity ligation assay (PLA) was used to detect CD147 phosphorylation. Tandem mass spectrometry was employed to identify the phosphorylation sites of CD147. The effects of CD147 phosphorylation on the malignant behavior of HCC cells were evaluated using scratch wound healing assays, transwell invasion assays and cell cycle assays. The genes regulated by CD147 phosphorylation were detected by RNA sequencing. RESULTS: We identified phosphorylated serine-246 in the C terminus of CD147 in primary HCC tissues, whereas serine to alanine substitution mutation analysis suggested that CD147 is phosphorylated mainly at serine-252 in HCC-derived Huh-7 cells. Recovery expression of S246A/S252A mutants in CD147 knockout cells revealed significantly increased migration and invasion capacities compared to wildtype CD147 expressing cells. Cyclophilin A (CyPA) treatment decreased the phosphorylation level of CD147, whereas NIMA-related kinase 6 (NEK6) increased the CD147 phosphorylation level. Moreover, the CD147 phosphorylation level was found to be dramatically decreased in HCC tissues in patients with distant metastases, and a low phosphorylation level of CD147 was found to be associated with a high serum AFP level, recurrence and a poor overall survival. CONCLUSIONS: From our data we conclude that hypo-phosphorylated CD147 promotes the migration and invasion of HCC cells and correlates with an unfavorable prognosis in HCC patients, indicating that targeting the aberrantly hypo-phosphorylated form of CD147 may be instrumental for the development of novel therapeutic modalities directed against HCC metastasis.


Subject(s)
Basigin/metabolism , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/pathology , Cell Movement , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Amino Acid Sequence , Basigin/chemistry , Carcinoma, Hepatocellular/genetics , Cell Line, Tumor , Cell Movement/drug effects , Cyclophilin A/metabolism , Extracellular Matrix/drug effects , Extracellular Matrix/metabolism , Female , Gene Expression Regulation, Neoplastic/drug effects , Humans , Liver Neoplasms/genetics , Male , NIMA-Related Kinases/metabolism , Neoplasm Invasiveness , Neoplasm Metastasis , Phosphorylation/drug effects , Phosphoserine/metabolism , Prognosis , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-akt/metabolism , STAT3 Transcription Factor/metabolism , Signal Transduction/drug effects
15.
Int J Mol Sci ; 20(8)2019 04 22.
Article in English | MEDLINE | ID: mdl-31013576

ABSTRACT

The transmembrane glycoprotein basigin, a member of the immunoglobulin superfamily, stimulates matrix metalloproteinase (MMP)-mediated extracellular matrix (ECM) degradation and thereby drives cancer cell invasion. Basigin is proteolytically shed from the cell surface and high concentrations of soluble basigin in the blood dictates poor prognosis in cancer patients. A positive correlation between basigin and a disintegrin and metalloproteinase (ADAM)-12 in serum from prostate cancer patients has been reported. Yet, the functional relevance of this correlation is unknown. Here, we show that ADAM12 interacts with basigin and cleaves it in the juxtamembrane region. Specifically, overexpression of ADAM12 increases ectodomain shedding of an alkaline phosphatase-tagged basigin reporter protein from the cell surface. Moreover, CRISPR/Cas9-mediated knockout of ADAM12 in human HeLa carcinoma cells results in reduced shedding of the basigin reporter, which can be rescued by ADAM12 re-expression. We detected endogenous basigin fragments, corresponding to the expected size of the ADAM12-generated ectodomain, in conditioned media from ADAM12 expressing cancer cell-lines, as well as serum samples from a healthy pregnant donor and five bladder cancer patients, known to contain high ADAM12 levels. Supporting the cancer relevance of our findings, we identified several cancer-associated mutations in the basigin membrane proximal region. Subsequent in vitro expression showed that some of these mutants are more prone to ADAM12-mediated shedding and that the shed ectodomain can enhance gelatin degradation by cancer cells. In conclusion, we identified ADAM12 as a novel basigin sheddase with a potential implication in cancer.


Subject(s)
ADAM12 Protein/metabolism , Basigin/metabolism , ADAM12 Protein/chemistry , ADAM12 Protein/genetics , Amino Acid Sequence , Basigin/chemistry , Basigin/genetics , CRISPR-Cas Systems , Cell Line , Gene Expression , Gene Knockdown Techniques , Genes, Reporter , Humans , Mutation , Substrate Specificity
16.
Life Sci ; 224: 76-87, 2019 May 01.
Article in English | MEDLINE | ID: mdl-30904494

ABSTRACT

CD147/Basigin/EMMPRIN (Extracellular Matrix Metalloproteinase inducer) is a single pass type1 transmembrane protein playing a central role in developmental process, wound healing, nutrient transport, inflammation, arthritis and also in microbial pathologies. It is also found to be a potent stimulator of MMP (matrix metalloproteinases) and has been considered as a prognostic marker in cancer. Dysregulation of CD147 is reported in several types of cancer. It activates cell proliferation, invasion, metastasis and inhibits tumor cell apoptosis under hypoxic condition. Thus, CD147 serves as a hub protein in cancer, as it is involved in several homophilic and heterophilic cellular interactions spanning the major hallmarks of cancer. Targeting these interactions is considered to be an efficient therapeutic modality in cancerous conditions. Hence, by this review we intend to collate the structure-function relationships of CD147, with an exclusive thrust on potential druggable hotspots based on its intra and inter molecular interactions.


Subject(s)
Antineoplastic Agents/pharmacology , Basigin/chemistry , Basigin/metabolism , Neoplasms/drug therapy , Animals , Drug Design , Humans , Neoplasms/metabolism , Neoplasms/pathology
17.
Int J Cancer ; 144(12): 3138-3145, 2019 06 15.
Article in English | MEDLINE | ID: mdl-30365872

ABSTRACT

Within the "seed and soil" theory of organ tropic cancer metastasis is a growing compilation of evidence that S100A8/A9 functions as a soil signal that attracts cancer cells to certain organs, which prove beneficial to their growth. S100A8/A9-sensing receptors including Toll-like receptor 4 (TLR4), advanced glycation end products (RAGE), and also important receptors we recently succeeded in identifying (EMMPRIN, NPTNß, MCAM, and ALCAM) have the potential to become promising therapeutic targets. In our study, we prepared extracellular regions of these novel molecules and fused them to human IgG2-Fc to extend half-life expectancy, and we evaluated the anti-metastatic effects of the purified decoy proteins on metastatic cancer cells. The purified proteins markedly suppressed S100A8/A9-mediated lung tropic cancer metastasis. We hence expect that our novel biologics may become a prominent medicine to prevent cancer metastasis in clinical settings through cutting the linkage between "seed and soil".


Subject(s)
Calgranulin A/metabolism , Calgranulin B/metabolism , Melanoma, Experimental/prevention & control , Melanoma, Experimental/secondary , Recombinant Fusion Proteins/pharmacology , Animals , Basigin/chemistry , Basigin/pharmacology , Breast Neoplasms/drug therapy , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Adhesion Molecules/chemistry , Cell Adhesion Molecules/pharmacology , Humans , Immunoglobulin Fc Fragments/chemistry , Immunoglobulin Fc Fragments/pharmacology , Immunoglobulin G/immunology , Lung Neoplasms/drug therapy , Lung Neoplasms/metabolism , Lung Neoplasms/secondary , Melanoma, Experimental/metabolism , Melanoma, Experimental/pathology , Mice , Mice, Inbred BALB C , Mice, Nude , Protein Domains , Receptor for Advanced Glycation End Products/chemistry , Recombinant Fusion Proteins/isolation & purification , Recombinant Fusion Proteins/metabolism
18.
J Biol Chem ; 294(2): 593-607, 2019 01 11.
Article in English | MEDLINE | ID: mdl-30446621

ABSTRACT

Monocarboxylate transporters (MCTs) mediate the proton-coupled exchange of high-energy metabolites, including lactate and pyruvate, between cells and tissues. The transport activity of MCT1, MCT2, and MCT4 can be facilitated by the extracellular carbonic anhydrase IV (CAIV) via a noncatalytic mechanism. Combining physiological measurements in HEK-293 cells and Xenopus oocytes with pulldown experiments, we analyzed the direct interaction between CAIV and the two MCT chaperones basigin (CD147) and embigin (GP70). Our results show that facilitation of MCT transport activity requires direct binding of CAIV to the transporters chaperones. We found that this binding is mediated by the highly conserved His-88 residue in CAIV, which is also the central residue of the enzyme's intramolecular proton shuttle, and a charged amino acid residue in the Ig1 domain of the chaperone. Although the position of the CAIV-binding site in the chaperone was conserved, the amino acid residue itself varied among different species. In human CD147, binding of CAIV was mediated by the negatively charged Glu-73 and in rat CD147 by the positively charged Lys-73. In rat GP70, we identified the positively charged Arg-130 as the binding site. Further analysis of the CAIV-binding site revealed that the His-88 in CAIV can either act as H donor or H acceptor for the hydrogen bond, depending on the charge of the binding residue in the chaperone. Our results suggest that the CAIV-mediated increase in MCT transport activity requires direct binding between CAIV-His-88 and a charged amino acid in the extracellular domain of the transporter's chaperone.


Subject(s)
Basigin/metabolism , Carbonic Anhydrase IV/metabolism , Glycoproteins/metabolism , Membrane Glycoproteins/metabolism , Molecular Chaperones/metabolism , Monocarboxylic Acid Transporters/metabolism , Protein Interaction Maps , Amino Acid Sequence , Animals , Basigin/chemistry , HEK293 Cells , Humans , Membrane Proteins , Models, Molecular , Protein Domains , Rats , Sequence Alignment , Symporters/metabolism , Xenopus
19.
J Mol Graph Model ; 85: 281-293, 2018 10.
Article in English | MEDLINE | ID: mdl-30253283

ABSTRACT

The interaction of the proteins, tumor necrosis factor receptor-associated factor6 (TRAF6) and Basigin (CD147), is known to be associated with the over-expression of matrix metalloproteinases (MMPs) in melanoma cells. MMPs are known to be responsible for melanoma metastasis. Hence, the TRAF6-Basigin complex can act as a potential therapeutic target. In previous studies, amino acid residues Lys340, Lys 384, Glu417 and Glu511 of TRAF6 were identified as the most vital residues on the basis of their contributions to interaction energy, relative solvent accessibility and electrostatic interactions in the TRAF6-Basigin protein-protein interaction (PPI) scheme. In our current work, we performed structure-based virtual screenings of some natural compounds obtained from ZINC database (n = 14509) to search for molecules which can act as inhibitors against the formation of TRAF6-Basigin complex. Three potential inhibitors were identified which were observed to make intermolecular interactions with Lys384 and Glu511 of TRAF6. Molecular dynamics simulation results suggested the substantial pharmacological importance of the ligand molecules as it was observed that there was total destabilization of TRAF6-Basigin complex upon binding of the molecule ZINC02578057. From our studies, we could conclude that the ligands termed as ZINC49048033, ZINC02578057 and ZINC72320240 could have great potentials to act as inhibitors to prevent melanoma metastasis.


Subject(s)
Basigin/chemistry , Biological Products/chemistry , Molecular Docking Simulation , Molecular Dynamics Simulation , TNF Receptor-Associated Factor 6/chemistry , Basigin/metabolism , Biological Products/pharmacology , Drug Discovery , Humans , Hydrogen Bonding , Ligands , Melanoma/drug therapy , Melanoma/metabolism , Protein Binding/drug effects , Quantitative Structure-Activity Relationship , Solvents/chemistry , TNF Receptor-Associated Factor 6/metabolism
20.
Int J Mol Sci ; 19(2)2018 Feb 08.
Article in English | MEDLINE | ID: mdl-29419744

ABSTRACT

The receptor EMMPRIN is involved in the development and progression of cardiovascular diseases and in the pathogenesis of myocardial infarction. There are several binding partners of EMMPRIN mediating the effects of EMMPRIN in cardiovascular diseases. EMMPRIN interaction with most binding partners leads to disease progression by mediating cytokine or chemokine release, the activation of platelets and monocytes, as well as the formation of monocyte-platelet aggregates (MPAs). EMMPRIN is also involved in atherosclerosis by mediating the infiltration of pro-inflammatory cells. There is also evidence that EMMPRIN controls energy metabolism of cells and that EMMPRIN binding partners modulate intracellular glycosylation and trafficking of EMMPRIN towards the cell membrane. In this review, we systematically discuss these multifaceted roles of EMMPRIN and its interaction partners, such as Cyclophilins, in cardiovascular disease.


Subject(s)
Basigin/genetics , Basigin/metabolism , Cardiovascular Diseases/etiology , Cardiovascular Diseases/metabolism , Animals , Basigin/chemistry , Blood Platelets/metabolism , Carrier Proteins/metabolism , Cell Adhesion , Cell Communication , Cell Movement , Extracellular Matrix/metabolism , Humans , Inflammation/etiology , Inflammation/metabolism , Monocytes/metabolism , Platelet Aggregation , Protein Binding , Protein Interaction Domains and Motifs
SELECTION OF CITATIONS
SEARCH DETAIL
...