Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 104
Filter
1.
Cell Physiol Biochem ; 49(2): 512-529, 2018.
Article in English | MEDLINE | ID: mdl-30157491

ABSTRACT

BACKGROUND/AIMS: Pigment epithelium-derived factor (PEDF) is a potent endogenous inhibitor of angiogenesis, and a promising anticancer agent. We have previously shown that PEDF can be phosphorylated, and that distinct phosphorylations differentially regulate its physiological functions. We also demonstrated that triple phosphomimetic mutant (EEE-PEDF), has significantly increased antiangiogenic activity, and is much more efficient than WT-PEDF in inhibiting neovascularization and tumor growth. The enhanced antiangiogenic effect was associated with a direct ability to facilitate apoptosis of tumor-residing endothelial cells (EC), and subsequently, disruption of intratumoral vascularization. In the present report, we elucidated the molecular mechanism by which EEE-PEDF exerts more profound effects at the cellular level. METHODS: Here we used Western blotting, as well as in vitro binding, proliferation, apoptosis and migration assays to follow the signaling components responsible for the PEDF and EEE-PEDF effects. RESULTS: We found that EEE-PEDF suppresses EC proliferation due to caspase-3-dependent apoptosis, and also inhibits migration of the EC much better than WT-PEDF. Although WT-PEDF and EEE-PEDF did not affect proliferation and did not induce apoptosis of cancer cells, these agents efficiently inhibited cancer cell motility, with EEE-PEDF showing stronger effect. The stronger activity of EEE-PEDF was correlated to a better binding to laminin receptors. Furthermore, the proapoptotic and antimigratory activities of WT-PEDF and EEE-PEDF were found respectively regulated by differential activation of two distinct MAPK pathways, namely JNK and p38. We show that JNK and p38 phosphorylation is much higher in cells treated with EEE-PEDF. JNK leads to apoptosis of ECs, while p38 leads to antimigratory effect in both EC and cancer cells. CONCLUSION: These results reveal the molecular signaling mechanism by which the phosphorylated PEDF exerts its stronger antiangiogenic, antitumor activities.


Subject(s)
Eye Proteins/pharmacology , Nerve Growth Factors/pharmacology , Serpins/pharmacology , Animals , Apoptosis/drug effects , Cattle , Cell Line , Cell Movement/drug effects , Cell Proliferation/drug effects , Eye Proteins/genetics , Eye Proteins/metabolism , Fibroblast Growth Factor 2/pharmacology , Humans , JNK Mitogen-Activated Protein Kinases/antagonists & inhibitors , JNK Mitogen-Activated Protein Kinases/metabolism , Lipase/chemistry , Lipase/genetics , Lipase/metabolism , Mutagenesis , Nerve Growth Factors/genetics , Nerve Growth Factors/metabolism , Phosphorylation/drug effects , Protein Binding , Protein Kinase Inhibitors/pharmacology , Receptors, Laminin/chemistry , Receptors, Laminin/genetics , Receptors, Laminin/metabolism , Recombinant Proteins/biosynthesis , Recombinant Proteins/isolation & purification , Recombinant Proteins/pharmacology , Serpins/genetics , Serpins/metabolism , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors , p38 Mitogen-Activated Protein Kinases/metabolism
2.
Methods ; 113: 56-63, 2017 01 15.
Article in English | MEDLINE | ID: mdl-27789335

ABSTRACT

Lysyl-tRNA synthetase (KRS) is an enzyme that conjugates lysine to its cognate tRNAs in the process of protein synthesis. In addition to its catalytic function, KRS binds to the 67-kDa laminin receptor (LR) on the cell membrane and facilitates cell migration and metastasis. Modulation of this interaction by small-molecule inhibitors can be exploited to suppress cancer metastasis. In this study, we present fragment-based methods for the identification of inhibitors and monitoring protein-protein interactions between KRS and LR. First, we identified the amino acid residues, located on the KRS anticodon-binding domain, which interact with the C-terminal extension of the LR. One-dimensional (1D) relaxation-edited nuclear magnetic resonance spectroscopy (NMR) and competition experiments were designed and optimized to screen the fragment library. For screening using two-dimensional (2D) NMR, we identified the indicative signals in the KRS anticodon-binding domain and selected inhibitors that bind to KRS and compete with LR at the KRS-LR binding interface. These methods may offer an efficient approach for the discovery of anti-metastatic drugs.


Subject(s)
Enzyme Inhibitors/pharmacology , High-Throughput Screening Assays , Lysine-tRNA Ligase/antagonists & inhibitors , Lysine/metabolism , Receptors, Laminin/antagonists & inhibitors , Small Molecule Libraries/pharmacology , Amino Acid Motifs , Anticodon/chemistry , Anticodon/metabolism , Antineoplastic Agents/pharmacology , Binding Sites , Drug Discovery/methods , Escherichia coli/enzymology , Escherichia coli/genetics , Gene Expression , Humans , Lysine-tRNA Ligase/chemistry , Lysine-tRNA Ligase/genetics , Lysine-tRNA Ligase/metabolism , Nuclear Magnetic Resonance, Biomolecular , Protein Binding , Protein Interaction Domains and Motifs , RNA, Transfer, Lys/genetics , RNA, Transfer, Lys/metabolism , Receptors, Laminin/chemistry , Receptors, Laminin/genetics , Receptors, Laminin/metabolism , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Transfer RNA Aminoacylation
3.
Biochim Biophys Acta ; 1864(12): 1707-1713, 2016 12.
Article in English | MEDLINE | ID: mdl-27663887

ABSTRACT

Aminoacyl-tRNA synthetases (ARSs) are essential enzymes that conjugate specific amino acids to their cognate tRNAs for protein synthesis. Besides their catalytic activity, recent studies have uncovered many additional functions of these enzymes through their interactions with diverse cellular factors. Among human ARSs, cytosolic lysyl-tRNA synthetase (KRS) is often highly expressed in cancer cells and tissues, and facilitates cancer cell migration and invasion through the interaction with the 67kDa laminin receptor on the plasma membrane. Specific modulation of this interaction by small molecule inhibitors has revealed a new way to control metastasis. Here, we summarize the pro-metastatic functions of KRS and their patho-physiological implications.


Subject(s)
Carcinogenesis/metabolism , Lysine-tRNA Ligase/chemistry , Lysine-tRNA Ligase/metabolism , Active Transport, Cell Nucleus , Amino Acyl-tRNA Synthetases/chemistry , Amino Acyl-tRNA Synthetases/metabolism , Biocatalysis , Cell Membrane/enzymology , Cell Movement , Epithelial-Mesenchymal Transition , Humans , Models, Biological , Models, Molecular , Neoplasm Invasiveness , Neoplasm Metastasis , Protein Interaction Domains and Motifs , Receptors, Laminin/chemistry , Receptors, Laminin/metabolism
4.
Cell Mol Biol Lett ; 20(4): 571-85, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26146125

ABSTRACT

The 37-kDa laminin receptor (37LRP or RPSA) is a remarkable, multifaceted protein that functions in processes ranging from matrix adhesion to ribosome biogenesis. Its ability to engage extracellular laminin is further thought to contribute to cellular migration and invasion. Most commonly associated with metastatic cancer, RPSA is also increasingly found to be important in other pathologies, including microbial infection, neurodegenerative disease and developmental malformations. Importantly, it is thought to have higher molecular weight forms, including a 67-kDa species (67LR), the expression of which is linked to strong laminin binding and metastatic behavior. The composition of these larger forms has remained elusive and controversial. Homo- and heterodimerization have been proposed as events capable of building the larger species from the monomeric 37-kDa precursor, but solid evidence is lacking. Here, we present data suggesting that higher molecular weight species require SUMOylation to form. We also comment on the difficulty of isolating larger RPSA species for unambiguous identification and demonstrate that cell lines stably expressing tagged RPSA for long periods of time fail to produce tagged higher molecular weight RPSA. It is possible that higher molecular weight species like 67LR are not derived from RPSA.


Subject(s)
Receptors, Laminin/chemistry , Receptors, Laminin/metabolism , Ribosomal Proteins/chemistry , Ribosomal Proteins/metabolism , Animals , Gene Knockdown Techniques , HeLa Cells , Humans , Immunoprecipitation , Mice , Molecular Weight , NIH 3T3 Cells , Receptors, Laminin/genetics , Ribosomal Proteins/genetics , Sumoylation , Ubiquitin-Conjugating Enzymes/genetics , Ubiquitin-Conjugating Enzymes/metabolism
5.
Clin Exp Pharmacol Physiol ; 42(5): 549-58, 2015 May.
Article in English | MEDLINE | ID: mdl-25800042

ABSTRACT

Insufficient trophoblast invasion often occurs in patients experiencing preeclampsia. The 67-kDa laminin receptor (LR1) is a multifunctional protein that binds to laminin and interacts with the extracellular matrix. We recently demonstrated that LR1 is implicated in trophoblast migration and invasion. However, whether LR1 is involved in hypoxia-mediated trophoblastic invasion remains unclear and requires further investigation. This study demonstrates that two trophoblast-like cell lines (JEG3 and BeWo cells) cultured at 3% oxygen exerted enhanced migratory and invasive capabilities as compared with their counterparts exposed to 20% oxygen. LR1 expression was increased in hypoxic JEG3 cells but decreased after transfection with hypoxia-inducible factor 1 alpha (HIF-1α) specific siRNA. Moreover, shRNA targeting LR1 mRNA significantly inhibited hypoxia-induced increase in matrix metalloproteinase (MMP)-9 activity in JEG3 cells. Forced overexpression of LR1 augmented JEG3 cell migration and invasion, and enhanced MMP-9 expression and activity. Additionally, the blockade of the MMP-9 effect with its neutralizing antibody reduced LR1 elevation-promoted trophoblastic invasion. In summary, this study demonstrates that LR1 contributes to hypoxia-induced migration and invasion of trophoblast cells at least partly by mediating MMP-9 in vitro.


Subject(s)
Cell Movement , Matrix Metalloproteinase 9/metabolism , Trophoblasts/cytology , Cell Hypoxia , Cell Line, Tumor , Gene Expression Regulation, Enzymologic , Gene Knockdown Techniques , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/deficiency , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Matrix Metalloproteinase 9/genetics , Molecular Weight , Receptors, Laminin/chemistry , Receptors, Laminin/deficiency , Receptors, Laminin/genetics , Receptors, Laminin/metabolism , Ribosomal Proteins
6.
Biochem Biophys Res Commun ; 445(1): 218-24, 2014 Feb 28.
Article in English | MEDLINE | ID: mdl-24508265

ABSTRACT

Delivery of optimal amounts of brain-derived neurotrophic factor (BDNF) to regions of the brain affected by neurodegenerative diseases is a daunting task. Using natural products with neuroprotective properties, such as green tea polyphenols, would be a highly useful complementary approach for inexpensive long-term treatment of these diseases. In this study, we used PC12(TrkB) cells which ectopically express TrkB, a high affinity receptor for BDNF. They differentiate and induce neurite outgrowth in response to BDNF. Using this model, we show for the first time that treatment with extremely low concentrations (<0.1 µg/ml) of unfractionated green tea polyphenols (GTPP) and low concentrations (<0.5 µM) of their active ingredient, epigallocatechin-3-gallate (EGCG), potentiated the neuritogenic ability of a low concentration (2 ng/ml) of BDNF. A synergistic interaction was observed between GTPP constituents, where epigallocatechin and epicatechin, both individually lacking this activity, promoted the action of EGCG. GTPP-induced potentiation of BDNF action required the cell-surface associated 67 kDa laminin receptor (67LR) to which EGCG binds with high affinity. A cell-permeable catalase abolished GTPP/EGCG-induced potentiation of BDNF action, suggesting the possible involvement of H2O2 in the potentiation. Consistently, exogenous sublethal concentrations of H2O2, added as a bolus dose (5 µM) or more effectively through a steady-state generation (1 µM), potentiated BDNF action. Collectively, these results suggest that EGCG, dependent on 67 LR and H2O2, potentiates the neuritogenic action of BDNF. Intriguingly, this effect requires only submicromolar concentrations of EGCG. This is significant as extremely low concentrations of polyphenols are believed to reach the brain after drinking green tea.


Subject(s)
Brain-Derived Neurotrophic Factor/pharmacology , Catechin/pharmacology , Neurites/drug effects , Tea/chemistry , Animals , Antioxidants/pharmacology , Catechin/analogs & derivatives , Dose-Response Relationship, Drug , Drug Synergism , Hydrogen Peroxide/metabolism , Hydrogen Peroxide/pharmacology , Molecular Weight , Neurites/physiology , Oxidants/metabolism , Oxidants/pharmacology , PC12 Cells , Polyphenols/pharmacology , Rats , Receptor, trkB/genetics , Receptor, trkB/metabolism , Receptors, Laminin/chemistry , Receptors, Laminin/metabolism , Receptors, Laminin/physiology
7.
Phytomedicine ; 118: 154970, 2023 Sep.
Article in English | MEDLINE | ID: mdl-37516056

ABSTRACT

BACKGROUND: Oolonghomobisflavans are unique polyphenols found in oolong teas. Oolonghomobisflavan B (OHBFB), a dimer of (-)-epigallocatechin-3-O-gallate (EGCG), is an active compound found in green tea. PURPOSE: OHBFB has been reported to exert an inhibitory effect on lipase enzyme activity. However, little is known regarding its intercellular signaling induction effect. Further, there are no reports describing the anti-cancer effects of OHBFB. METHODS: The effect of OFBFB on B16 melanoma cells was evaluated by cell counting, and its mechanisms were determined by western blot analysis with or without protein phosphatase 2A (PP2A) inhibitor treatment. Intracellular cyclic adenosine monophosphate (cAMP) levels were evaluated by time-resolved fluorescence resonance energy transfer analysis. Quartz crystal microbalance (QCM) analysis was performed to assess the binding of OHBFB to 67LR. RESULTS: Cell growth assay and western blot analyses showed that OHBFB inhibited melanoma cell growth, followed by myosin phosphatase target subunit 1 (MYPT1) and myosin regulatory light chain (MRLC) dephosphorylation via protein phosphatase 2A (PP2A)-dependent mechanisms. These effects are mediated by intracellular cAMP- and protein kinase A (PKA) A-dependent mechanisms. QCM analysis identified the 67-kDa laminin receptor (67LR) as an OHBFB receptor with a Kd of 3.7 µM. We also demonstrated for the first time that OHBFB intake suppresses tumor growth in vivo. CONCLUSIONS: Taken together, these results indicate that the cAMP/PKA/PP2A/MYPT1/MRLC pathway is a key mediator of melanoma cell growth inhibition following OHBFB binding to 67LR and that OHBFB suppresses tumor growth in vivo.


Subject(s)
Catechin , Melanoma, Experimental , Animals , Humans , Protein Phosphatase 2/metabolism , Polyphenols/pharmacology , Catechin/pharmacology , Cell Cycle , Melanoma, Experimental/drug therapy , Receptors, Laminin/chemistry , Receptors, Laminin/metabolism
8.
Biochemistry ; 51(24): 4807-21, 2012 Jun 19.
Article in English | MEDLINE | ID: mdl-22640394

ABSTRACT

The human ribosomal protein SA (RPSA) is a multilocus protein, present in most cellular compartments. It is a multifunctional protein, which belongs to the ribosome but is also a membrane receptor for laminin, growth factors, prion, pathogenic microorganisms, toxins, and the anticarcinogen epigallocatechin gallate. It contributes to the crossing of the blood-brain barrier by neurotropic viruses and bacteria and is used as a biomarker of metastasis. RPSA includes an N-terminal domain, which is homologous to the prokaryotic ribosomal proteins S2, and a C-terminal extension, which is conserved in vertebrates. The structure of its N-domain has been determined from crystals grown at 17 °C. The structure of its C-domain remains unknown. We produced in Escherichia coli and purified the full-length RPSA and its N- and C-domains. We characterized the folding states of these recombinant proteins mainly by methods of fluorescence and circular dichroism spectrometry, in association with quantitative analyses of their unfolding equilibria, induced with heat or urea. The necessary equations were derived from first principles. The results showed that the N-domain unfolded according to a three-state equilibrium. The monomeric intermediate was predominant at the body temperature of 37 °C. It also existed in the full-length RPSA and bound ANS, a small fluorescent molecule. The C-domain was in an intrinsically disordered state. The recombinant N- and C-domains weakly interacted together. These results indicated a high plasticity of RPSA, which could be important for its multiple cellular localizations and functional interactions.


Subject(s)
Anticarcinogenic Agents/metabolism , Laminin/metabolism , Microbiology , Protein Folding , Receptors, Laminin/chemistry , Receptors, Laminin/metabolism , Ribosomal Proteins/chemistry , Ribosomal Proteins/metabolism , Amino Acid Sequence , Animals , Humans , Mice , Models, Molecular , Molecular Sequence Data , Protein Structure, Tertiary , Protein Unfolding/drug effects , Receptors, Laminin/isolation & purification , Ribosomal Proteins/isolation & purification , Spectrometry, Fluorescence , Urea/pharmacology
9.
J Proteome Res ; 11(10): 4863-72, 2012 Oct 05.
Article in English | MEDLINE | ID: mdl-22909348

ABSTRACT

Human nonintegrin laminin receptor is a multifunctional protein acting as an integral component of the ribosome and a cell surface receptor for laminin-1. The laminin receptor is overexpressed in several human cancers and is also the cell surface receptor for several viruses and pathogenic prion proteins, making it a pathologically significant protein. This study focused on the proteomic characterization of laminin receptor interacting proteins from Mus musculus. The use of affinity chromatography with immobilized recombinant laminin receptor coupled with mass spectrometry analysis identified 45 proteins with high confidence. Following validation through coimmunoprecipitation, the proteins were classified based on predicted function into ribosomal, RNA processing, signal transduction/metabolism, protein processing, cytoskeleton/cell anchorage, DNA/chromatin, and unknown functions. A significant portion of the identified proteins is related to functions or localizations previously described for laminin receptor. This work represents a comprehensive proteomic approach to studying laminin receptor and provides an essential stepping stone to a better mechanistic understanding of this protein's diverse functions.


Subject(s)
Proteome/chemistry , Animals , Chaperonin Containing TCP-1/chemistry , Chaperonin Containing TCP-1/isolation & purification , Chromatography, Affinity , Cytoskeletal Proteins/chemistry , Cytoskeletal Proteins/isolation & purification , Hexosyltransferases , Histones/chemistry , Histones/isolation & purification , Humans , Intracellular Signaling Peptides and Proteins/chemistry , Intracellular Signaling Peptides and Proteins/isolation & purification , Mice , NIH 3T3 Cells , Proteasome Endopeptidase Complex/chemistry , Proteasome Endopeptidase Complex/isolation & purification , Protein Binding , Proteome/isolation & purification , Proteomics , RNA-Binding Proteins/chemistry , RNA-Binding Proteins/isolation & purification , Receptors, Laminin/chemistry , Ribosomal Proteins/chemistry , Ribosomal Proteins/isolation & purification , Transcription Factors/chemistry , Transcription Factors/isolation & purification
10.
J Med Virol ; 84(4): 615-23, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22337301

ABSTRACT

Japanese encephalitis virus (JEV) a mosquito-borne flavivirus is a major cause of viral encephalitis in Asia. While the principle target cells for JEV in the central nervous system are believed to be neurons, microglia are activated in response to JEV and have been proposed to act as a long lasting virus reservoir. Viral attachment to a host cell is the first step of the viral entry process and is a critical mediator of tissue tropism. This study sought to identify molecules associated with JEV entry to microglial cells. Virus overlay protein-binding assay (VOPBA) and liquid chromatography-mass spectrometry (LC/MS/MS) identified the 37/67 kDa high-affinity laminin receptor protein and nucleolin as a potential JEV-binding proteins. These proteins were subsequently investigated for a contribution to JEV entry to mouse microglial BV-2 cells together with other possible candidate receptor molecules including Hsp70, Hsp90, GRP78, CD14, and CD4. In antibody mediated inhibition of infection experiments, both anti-laminin receptor and anti-CD4 antibodies significantly reduced virus entry while anti-Hsp70 and 90 antibodies produced a slight reduction. Significant inhibition of virus entry (up to 80%) was observed in the presence of lipopolysaccharide (LPS) which resulted in a complete down-regulation of CD4 and moderate down-regulation of CD14. These results suggest that multiple receptor proteins may mediate the entry of JEV to microglial cells, with CD4 playing a major role.


Subject(s)
Encephalitis Virus, Japanese/physiology , Neuroglia/virology , Phosphoproteins/metabolism , RNA-Binding Proteins/metabolism , Receptors, Laminin/metabolism , Receptors, Virus/metabolism , Virus Attachment , Animals , CD4 Antigens/metabolism , Chromatography, Liquid , Endoplasmic Reticulum Chaperone BiP , Mass Spectrometry , Mice , Molecular Weight , Phosphoproteins/chemistry , Protein Binding , RNA-Binding Proteins/chemistry , Receptors, Laminin/chemistry , Receptors, Virus/chemistry , Nucleolin
11.
Eur Biophys J ; 41(3): 353-8, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22290616

ABSTRACT

The 37/67-kDa human laminin receptor(LamR) is a cell surface protein that interacts with molecules located in the extra-cellular matrix. In particular, interactions between LamR and laminins play a major role in mediating changes in the cellular environment that affect cell adhesion, neurite outgrowth, tumor growth and metastasis. The exact interaction mode of laminin-1 and LamR is not fully understood. Laminin-1 is thought to bind to LamR through interaction with the so-called peptide G (residues 161­180) and the C-terminal helix (residues 205­229). Here we performed 100-ns atomistic force field based molecular dynamics simulations to explore the structure and dynamics of LamR related to laminin-1 interactions. Our main finding is that loop 188­197 in the C-terminal region is highly flexible. It undergoes a major change resulting in a conformational switch that partially solvent exposes the R180 residue in the final part of the G peptide. So, R180 could contribute to laminin-1 binding. Projection of the simulations along the first two principal components also confirms the importance of this conformational switch in the LamR. This may be a basic prerequisite to clarify the key structural determinants of the interaction of LamR with laminin-1.


Subject(s)
Laminin/metabolism , Molecular Dynamics Simulation , Receptors, Laminin/chemistry , Receptors, Laminin/metabolism , Amino Acid Sequence , Humans , Laminin/chemistry , Molecular Sequence Data , Protein Binding , Protein Conformation
12.
Proteomics ; 11(8): 1429-48, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21365757

ABSTRACT

One of the major questions in the field of obesity is why some humans become obese (obesity prone, OP) and others resist the development of obesity (obesity resistant, OR) when exposed to a high-calorie diet, which has not been completely studied. Therefore, in the present study, in order to gain insight into the molecular mechanisms underlying this propensity, we have performed a comparative analysis of protein expression profiles in white adipose tissue (WAT) and brown adipose tissue (BAT) of rats fed a high-fat diet by 2-DE and MALDI-TOF-MS. Protein mapping of homogenates revealed significant alterations to a number of proteins; 60 and 70 proteins were differentially regulated in BAT and WAT, respectively. For careful interpretation of proteomic results, we categorized the identified proteins into two groups by analysis of both average spot density of pooled six rat adipose tissues and individual spot density of each adipose tissue of six rats as a function of body weight. One of the most striking findings of this study was that significant changes of Ehd1 and laminin receptor in BAT as well as antiquitin, DJ-1 protein, and paraoxonase 2 in WAT were found for the first time in obese rats. In addition, we confirmed the increased expression of some thermogenic enzymes and decreased lipogenic enzymes in adipose tissues of OR rats by immunoblot analysis. To our knowledge, this is the first proteomic study of profiling of protein modulation in OP and OR rats, thereby providing the first global evidence for different propensities to obesity between OP and OR rats.


Subject(s)
Adipose Tissue, Brown/chemistry , Adipose Tissue, White/chemistry , Dietary Fats/administration & dosage , Obesity/metabolism , Adipose Tissue, Brown/metabolism , Adipose Tissue, White/metabolism , Animals , Aryldialkylphosphatase/biosynthesis , Aryldialkylphosphatase/isolation & purification , Dietary Fats/metabolism , Disease Susceptibility/metabolism , Immunoblotting , L-Aminoadipate-Semialdehyde Dehydrogenase , Male , Microtubule-Associated Proteins/biosynthesis , Microtubule-Associated Proteins/isolation & purification , Protein Array Analysis , Protein Deglycase DJ-1 , Proteins/isolation & purification , Rats , Rats, Sprague-Dawley , Receptors, Laminin/chemistry , Vesicular Transport Proteins/chemistry
13.
Curr Opin Cell Biol ; 8(5): 625-31, 1996 Oct.
Article in English | MEDLINE | ID: mdl-8939660

ABSTRACT

Dystroglycan provides a crucial linkage between the cytoskeleton and the basement membrane for skeletal muscle cells. Disruption of this linkage leads to various forms of muscular dystrophy. Significant recent advances in understanding the structure and function of dystroglycan include detailed in vitro and in vivo analyses of its binding partners in muscle, an examination of its function at the neuromuscular junction, and emerging evidence of its roles in nonmuscle tissues.


Subject(s)
Cytoskeletal Proteins/metabolism , Membrane Glycoproteins/metabolism , Receptors, Laminin/metabolism , Amino Acid Sequence , Animals , Cytoskeletal Proteins/chemistry , Cytoskeleton/metabolism , Dystroglycans , Extracellular Matrix/metabolism , Humans , Membrane Glycoproteins/chemistry , Molecular Sequence Data , Neuromuscular Junction/metabolism , Receptors, Laminin/chemistry , Sarcolemma/metabolism
14.
Nat Med ; 3(12): 1383-8, 1997 Dec.
Article in English | MEDLINE | ID: mdl-9396609

ABSTRACT

Prions are thought to consist of infectious proteins that cause transmissible spongiform encephalopathies. According to overwhelming evidence, the pathogenic prion protein PrPSc converts its host encoded isoform PrPC into insoluble aggregates of PrPSc, concomitant with pathological modifications (for review, see refs. 1-3). Although the physiological role of PrPC is poorly understood, studies with PrP knockout mice demonstrated that PrPC is required for the development of prion diseases. Using the yeast two-hybrid technology in Saccharomyces cerevisiae, we identified the 37-kDa laminin receptor precursor (LRP) as interacting with the cellular prion protein PrPC. Mapping analysis of the LRP-PrP interaction site in S. cerevisiae revealed that PrP and laminin share the same binding domain (amino acids 161 to 180) on LRP. The LRP-PrP interaction was confirmed in vivo in insect (Sf9) and mammalian cells (COS-7). The LRP level was increased in scrapie-infected murine N2a cells and in brain and spleen of scrapie-infected mice. In contrast, the LRP concentration was not significantly altered in these organs from mice infected with the bovine spongiform encephalopathic agent (BSE), which have a lower PrPSc accumulation. LRP levels, however, were dramatically increased in brain and pancreas, slightly increased in the spleen and not altered in the liver of crapie-infected hamsters. These data show that enhanced LRP concentrations are correlated with PrPSc accumulation in organs from mice and hamsters. The laminin receptor precursor, which is highly conserved among mammals and is located on the cell surface, may act as a receptor or co-receptor for the prion protein on mammalian cells.


Subject(s)
PrPSc Proteins/metabolism , Protein Precursors/metabolism , Receptors, Laminin/metabolism , Actins/metabolism , Animals , Binding Sites , COS Cells , Cell Line , Cricetinae , Eukaryotic Cells , Humans , Mice , Mice, Inbred C57BL , Protein Precursors/chemistry , Protein Precursors/genetics , Rabbits , Receptors, Laminin/chemistry , Receptors, Laminin/genetics , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Saccharomyces cerevisiae/metabolism , Spodoptera/cytology
15.
Article in English | MEDLINE | ID: mdl-21422740

ABSTRACT

Green tea polyphenols have emerged over the past two decades as an important dietary factor for health promotion. There is considerable evidence that tea polyphenols, in particular (-)-epigallocatechin-3-gallate (EGCG) inhibit carcinogenesis. However, the mechanisms for the cancer-preventive activity of EGCG are not completely characterized and many features remain to be elucidated. Recently we have identified a cell-surface EGCG receptor and the relating molecules that confer EGCG responsiveness to many cancer cells at physiological concentrations. Here, we review some of the reported mechanisms for the cancer chemopreventive action of EGCG and provide an overview of several molecules that sense and manage the physiological functions of EGCG.


Subject(s)
Catechin/analogs & derivatives , Tea/chemistry , Animals , Catechin/metabolism , Catechin/pharmacokinetics , Catechin/pharmacology , Catechin/therapeutic use , Humans , Hypersensitivity/drug therapy , Inflammation/drug therapy , Neoplasms/metabolism , Neoplasms/pathology , Neoplasms/prevention & control , Receptors, Laminin/chemistry , Receptors, Laminin/metabolism
16.
Cancer Sci ; 101(7): 1653-60, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20491781

ABSTRACT

It has been reported that the 67-kDa laminin receptor (67LR) is implicated in cancer metastasis. We recently showed that 37LRP, the 67LR precursor, is a hypoxia-inducible factor 1 (HIF-1) target gene exposed to hypoxia in gastric cancer. Here, we investigated the role of 67LR in hypoxic metastasis and invasion in gastric cancer. Immunohistochemical analysis, western blotting, and RT-PCR assays revealed that 67LR was highly expressed in metastatic gastric cancers in vivo. Knockdown of the 67LR protein by RNA interference significantly decreased the adhesive, invasive, and in vivo metastatic abilities of the gastric cancer cell lines SGC7901 and MKN-45. Western blot analysis showed that 67LR increased the expression of urokinase-type plasminogen activator (uPA) and matrix metalloproteinase (MMP)-9, and decreased tissue inhibitor of matrix metalloproteinase (TIMP)-1 protein. We further showed that hypoxia induced 67LR expression in a time-dependent manner and this induction was inhibited by HIF-1 small-interfering (si) RNA. Both ERK and JNK inhibitors significantly inhibited hypoxia-induced expression of 67LR and the subsequent expression of uPA and MMP 9. SiRNA against 67LR or antibody against MMP9 and uPA significantly inhibited hypoxia-induced in vitro invasive ability. Taken together, these results reveal that 67LR promotes the invasive and metastatic ability of the gastric cancer cells through increasing uPA and MMP 9 expression, with involvement of the ERK and JNK signal pathway in hypoxia-induced 67 LR expressions and subsequent uPA and MMP9 expression.


Subject(s)
Gene Expression Regulation, Neoplastic , Receptors, Laminin/genetics , Stomach Neoplasms/genetics , Actins/chemistry , Actins/genetics , Base Sequence , Cell Adhesion , Cell Hypoxia , Humans , Immunohistochemistry , Lymph Nodes/pathology , Matrix Metalloproteinase 9/genetics , Molecular Sequence Data , Neoplasm Invasiveness , Neoplasm Metastasis , RNA, Small Interfering/genetics , Receptors, Laminin/chemistry , Reverse Transcriptase Polymerase Chain Reaction , Stomach Neoplasms/pathology , Stomach Neoplasms/physiopathology , Urokinase-Type Plasminogen Activator/genetics
17.
Biochem Genet ; 48(11-12): 962-9, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20839046

ABSTRACT

Scrapie is a prion disease in sheep and goats. Ribosomal protein SA (RPSA), also called 37 kDa laminin receptor precursor/67 kDa laminin receptor has been demonstrated to be a putative cell surface receptor for prion. To investigate the caprine RPSA, we cloned the full-length coding sequence of the gene of goat and submitted it to GenBank. The length of the open reading frame is 888 bp, encoding 295 amino acids. The putative amino acid sequence is highly similar to that of other mammals. The caprine amino acid sequence of RPSA is shown to be identical to the sequence of species susceptible to scrapie at positions 241, 272, and 291. The phylogenetic tree analysis revealed that the genetic distance between sheep and goat is the smallest. Moreover, RT-PCR results of 11 tissues indicated that RPSA mRNA is expressed in all selected caprine tissues.


Subject(s)
Goats , Open Reading Frames , Polymorphism, Genetic , Protein Precursors/chemistry , Protein Precursors/genetics , Protein Precursors/metabolism , Receptors, Laminin/chemistry , Receptors, Laminin/genetics , Receptors, Laminin/metabolism , Amino Acid Sequence , Animals , Base Sequence , Goat Diseases/genetics , Goat Diseases/virology , Goats/genetics , Goats/virology , Humans , Molecular Sequence Data , Open Reading Frames/genetics , Phylogeny , Prions/genetics , Prions/metabolism , Protein Interaction Domains and Motifs/genetics , Ribosomal Proteins/chemistry , Ribosomal Proteins/genetics , Ribosomal Proteins/metabolism , Scrapie/genetics , Scrapie/metabolism , Scrapie/virology , Sequence Alignment , Sheep/genetics , Sheep/virology
19.
J Bacteriol ; 191(18): 5814-23, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19617361

ABSTRACT

The common pathogen Streptococcus pyogenes colonizes the human skin and tonsils and can invade underlying tissues. This requires the adhesion of S. pyogenes to host surface receptors mediated through adhesins. The laminin-binding protein Lbp has been suggested as an adhesin, specific for the human extracellular matrix protein laminin. Sequence alignments, however, indicate a relationship between Lbp and a family of bacterial metal-binding receptors. To further analyze the role of Lbp in S. pyogenes and its potential role in pathogenicity, Lbp has been crystallized, and its structure has been solved at a resolution of 2.45 A (R = 0.186; R(free) = 0.251). Lbp has the typical metal-binding receptor fold, comprising two globular (beta/alpha)(4) domains connected by a helical backbone. The two domains enclose the metal-binding site, which contains a zinc ion. The interaction of Lbp with laminin was further investigated and shown to be specific in vitro. Localization studies with antibodies specific for Lbp show that the protein is attached to the membrane. The data suggest that Lbp is primarily a zinc-binding protein, and we suggest that its interaction with laminin in vivo may be mediated via zinc bound to laminin.


Subject(s)
Bacterial Proteins/metabolism , Carrier Proteins/metabolism , Laminin/metabolism , Receptors, Laminin/metabolism , Streptococcus pyogenes/metabolism , Animals , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Biotinylation , Enzyme-Linked Immunosorbent Assay , Humans , Immunoblotting , Immunoprecipitation , Molecular Sequence Data , Rabbits , Receptors, Laminin/chemistry , Receptors, Laminin/genetics , Streptococcus pyogenes/genetics , Zinc/metabolism
20.
Proteins ; 76(2): 461-76, 2009 Aug 01.
Article in English | MEDLINE | ID: mdl-19241470

ABSTRACT

Elastin-derived peptides (EDPs) have been intensively studied in view of their widely diverse biological activities. These are triggered both in normal and tumor cells, through peptide anchoring at the surface of the elastin-binding protein (EBP), a subunit of the elastin/laminin receptor. In this study, we investigated both the structure of the Sgal peptide, representing the elastin-binding domain of EBP, and its interaction with EDPs, through a combination of experimental and theoretical methods. Although the conformation of the Sgal peptide is highly flexible, we detected a type I beta-turn at the QDEA sequence. This represents the best structured motif in the entire Sgal peptide, which might therefore contribute to its binding activity. We further propose a novel three-dimensional model for the interaction between the Sgal peptide and EDPs; folding of the EDPs at the GXXP motif, in a conformation close to a type VIII beta-turn, provides the efficient contact of the protein with the Q residue of the Sgal peptide. This residue is exposed to the peptide surface, because of the beta-turn structure of the QDEA residues in the peptide sequence. We further show that this complex is stabilized by three hydrogen bonds involving EDPs backbone atoms.


Subject(s)
Elastin/chemistry , Peptide Fragments/chemistry , Receptors, Laminin/chemistry , Receptors, Laminin/metabolism , Amino Acid Sequence , Binding Sites , Circular Dichroism , Elastin/metabolism , Hydrogen Bonding , Models, Molecular , Molecular Sequence Data , Peptide Fragments/metabolism , Protein Conformation
SELECTION OF CITATIONS
SEARCH DETAIL