Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 71
Filter
1.
J Biol Chem ; 292(45): 18565-18576, 2017 11 10.
Article in English | MEDLINE | ID: mdl-28972165

ABSTRACT

We recently reported that the lectin surfactant protein D (SP-D) suppresses epidermal growth factor receptor (EGFR) signaling by interfering with ligand binding to EGFR through an interaction between the carbohydrate-recognition domain (CRD) of SP-D and N-glycans of EGFR. Here, we report that surfactant protein A (SP-A) also suppresses EGF signaling in A549 human lung adenocarcinoma cells and in CHOK1 cells stably expressing human EGFR and that SP-A inhibits the proliferation and motility of the A549 cells. Results with 125I-EGF indicated that SP-A interferes with EGF binding to EGFR, and a ligand blot analysis suggested that SP-A binds EGFR in A549 cells. We also found that SP-A directly binds the recombinant extracellular domain of EGFR (soluble EGFR or sEGFR), and this binding, unlike that of SP-D, was not blocked by EDTA, excess mannose, or peptide:N-glycosidase F treatment. We prepared a collagenase-resistant fragment (CRF) of SP-A, consisting of CRD plus the neck domain of SP-A, and observed that CRF directly binds sEGFR but does not suppress EGF-induced phosphorylation of EGFR in or proliferation of A549 cells. These results indicated that SP-A binds EGFR and down-regulates EGF signaling by inhibiting ligand binding to EGFR as well as SP-D. However, unlike for SP-D, SP-A lectin activity and EGFR N-glycans were not involved in the interaction between SP-A and EGFR. Furthermore, our results suggested that oligomerization of SP-A is necessary to suppress the effects of SP-A on EGF signaling.


Subject(s)
Epidermal Growth Factor/antagonists & inhibitors , ErbB Receptors/antagonists & inhibitors , Pulmonary Alveoli/metabolism , Pulmonary Surfactant-Associated Protein A/metabolism , Pulmonary Surfactant-Associated Protein D/metabolism , Signal Transduction , A549 Cells , Animals , CHO Cells , Cell Line, Tumor , Cell Movement , Cell Proliferation , Cricetulus , Epidermal Growth Factor/genetics , Epidermal Growth Factor/metabolism , ErbB Receptors/agonists , ErbB Receptors/genetics , ErbB Receptors/metabolism , Humans , Ligands , Peptide Fragments/chemistry , Peptide Fragments/genetics , Peptide Fragments/metabolism , Phosphorylation , Protein Interaction Domains and Motifs , Protein Interaction Mapping , Protein Processing, Post-Translational , Pulmonary Surfactant-Associated Protein A/genetics , Pulmonary Surfactant-Associated Protein D/genetics , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/metabolism , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism
2.
Sci Rep ; 7(1): 8304, 2017 08 16.
Article in English | MEDLINE | ID: mdl-28814727

ABSTRACT

The extent to which defective innate immune responses contribute to chronic obstructive pulmonary disease (COPD) is not fully understood. Pulmonary surfactant protein A (SP-A) plays an important role in regulating innate immunity in the lungs. In this study, we hypothesised that cigarette smoke (CS) and its component acrolein might influence pulmonary innate immunity by affecting the function of SP-A. Indeed, acrolein-modified SP-A was detected in the lungs of mice exposed to CS for 1 week. To further confirm this finding, recombinant human SP-A (hSP-A) was incubated with CS extract (CSE) or acrolein and then analysed by western blotting and nanoscale liquid chromatography-matrix-assisted laser desorption/ionisation time-of-flight tandem mass spectrometry. These analyses revealed that CSE and acrolein induced hSP-A oligomerisation and that acrolein induced the modification of six residues in hSP-A: His39, His116, Cys155, Lys180, Lys221, and Cys224. These modifications had significant effects on the innate immune functions of hSP-A. CSE- or acrolein-induced modification of hSP-A significantly decreased hSP-A's ability to inhibit bacterial growth and to enhance macrophage phagocytosis. These findings suggest that CS-induced structural and functional defects in SP-A contribute to the dysfunctional innate immune responses observed in the lung during cigarette smoking.


Subject(s)
Acrolein/chemistry , Nicotiana/chemistry , Pulmonary Surfactant-Associated Protein A/chemistry , Pulmonary Surfactant-Associated Protein A/metabolism , Aldehydes/chemistry , Animals , CHO Cells , Cigarette Smoking/adverse effects , Cricetulus , Female , Macrophages/immunology , Macrophages/metabolism , Mice , Models, Biological , Molecular Structure , Phagocytosis , Protein Conformation , RAW 264.7 Cells , Sulfhydryl Compounds/chemistry
3.
Biochem Biophys Res Commun ; 485(1): 107-112, 2017 03 25.
Article in English | MEDLINE | ID: mdl-28188794

ABSTRACT

Human ß-defensin 3 (hBD3) is known to be involved in mast cell activation. However, molecular mechanisms underlying the regulation of hBD3-induced mast cell activation have been poorly understood. We previously reported that SP-A and SP-A-derived peptide 01 (SAP01) regulate the function of hBD3. In this study, we focused on the effects of SP-A and SAP01 on the activation of mast cells induced by hBD3. SAP01 directly bound to hBD3. Mast cell-mediated vascular permeability and edema in hBD3 administered rat ears were decreased when injected with SP-A or SAP01. Compatible with the results in rat ear model, both SP-A and SAP01 inhibited hBD3-induced chemotaxis of mast cells in vitro. Direct interaction between SP-A or SAP01 and hBD3 seemed to be responsible for the inhibitory effects on chemotaxis. Furthermore, SAP01 attenuated hBD3-induced accumulation of mast cells and eosinophils in tracheas of the OVA-sensitized inflammatory model. SP-A might contribute to the regulation of inflammatory responses mediated by mast cells during infection.


Subject(s)
Chemotaxis/drug effects , Inflammation/immunology , Mast Cells/immunology , Pulmonary Surfactant-Associated Protein A/immunology , beta-Defensins/immunology , Animals , Capillary Permeability/drug effects , Edema/drug therapy , Edema/immunology , Humans , Inflammation/drug therapy , Male , Mast Cells/cytology , Mast Cells/drug effects , Peptides/chemistry , Peptides/pharmacology , Pulmonary Surfactant-Associated Protein A/chemistry , Pulmonary Surfactant-Associated Protein A/pharmacology , Rats, Sprague-Dawley
4.
J Immunol ; 198(7): 2898-2905, 2017 04 01.
Article in English | MEDLINE | ID: mdl-28228557

ABSTRACT

Surfactant protein A (SP-A) is a multifunctional host defense collectin that was first identified as a component of pulmonary surfactant. Although SP-A is also expressed in various tissues, including the urinary tract, its innate immune functions in nonpulmonary tissues are poorly understood. In this study, we demonstrated that adherence of uropathogenic Escherichia coli (UPEC) to the bladder was enhanced in SP-A-deficient mice, which suggests that SP-A plays an important role in innate immunity against UPEC. To understand the innate immune functions of SP-A in detail, we performed in vitro experiments. SP-A directly bound to UPEC in a Ca2+-dependent manner, but it did not agglutinate UPEC. Our results suggest that a bouquet-like arrangement seems unsuitable to agglutinate UPEC. Meanwhile, SP-A inhibited growth of UPEC in human urine. Furthermore, the binding of SP-A to UPEC decreased the adherence of bacteria to urothelial cells. These results indicate that direct action of SP-A on UPEC is important in host defense against UPEC. Additionally, adhesion of UPEC to urothelial cells was decreased when the cells were preincubated with SP-A. Adhesion of UPEC to urothelial cells is achieved via interaction between FimH, an adhesin located at bacterial pili, and uroplakin Ia, a glycoprotein expressed on the urothelium. SP-A directly bound to uroplakin Ia and competed with FimH for uroplakin Ia binding. These results lead us to conclude that SP-A plays important roles in host defense against UPEC.


Subject(s)
Escherichia coli Infections/immunology , Pulmonary Surfactant-Associated Protein A/immunology , Urinary Tract Infections/immunology , Animals , Cell Proliferation , Humans , Immunity, Innate/immunology , Immunohistochemistry , Mice , Mice, Inbred C57BL , Mice, Knockout , Urinary Tract Infections/microbiology , Uropathogenic Escherichia coli/immunology
5.
Clin Sci (Lond) ; 130(20): 1781-92, 2016 10 01.
Article in English | MEDLINE | ID: mdl-27612953

ABSTRACT

Numerous signal-transduction-related molecules are secreted proteins or membrane proteins, and the mechanism by which these molecules are regulated by glycan chains is a very important issue for developing an understanding of the cellular events that transpire. This review covers the functional regulation of epidermal growth factor receptor (EGFR), ErbB3 and the transforming growth factor ß (TGF-ß) receptor by N-glycans. This review shows that the N-glycans play important roles in regulating protein conformation and interactions with carbohydrate recognition molecules. These results point to the possibility of a novel strategy for controlling cell signalling and developing novel glycan-based therapeutics.


Subject(s)
Polysaccharides/metabolism , Receptors, Growth Factor/metabolism , Animals , Gene Expression Regulation , Glycosylation , Humans , Intercellular Signaling Peptides and Proteins/metabolism , Receptors, Growth Factor/genetics , Signal Transduction
6.
J Proteomics ; 127(Pt B): 386-94, 2015 Sep 08.
Article in English | MEDLINE | ID: mdl-26206179

ABSTRACT

We previously reported that knockout mice for α1,6-fucosyltransferase (Fut8), which catalyzes the biosynthesis of core-fucose in N-glycans, develop emphysema and that Fut8 heterozygous knockout mice are more sensitive to cigarette smoke-induced emphysema than wild-type mice. Moreover, a lower FUT8 activity was found to be associated with a faster decline in lung function among chronic obstructive pulmonary disease (COPD) patients. These results led us to hypothesize that core-fucosylation levels in a glycoprotein could be used as a biomarker for COPD. We focused on a lung-specific glycoprotein, surfactant protein D (SP-D), which plays a role in immune responses and is present in the distal airways, alveoli, and blood circulation. The results of a glycomic analysis reported herein demonstrate the presence of a core-fucose in an N-glycan on enriched SP-D from pooled human sera. We developed an antibody-lectin enzyme immunoassay (EIA) for assessing fucosylation (core-fucose and α1,3/4 fucose) in COPD patients. The results indicate that fucosylation levels in serum SP-D are significantly higher in COPD patients than in non-COPD smokers. The severity of emphysema was positively associated with fucosylation levels in serum SP-D in smokers. Our findings suggest that increased fucosylation levels in serum SP-D are associated with the development of COPD. BIOLOGICAL SIGNIFICANCE: It has been proposed that serum SP-D concentrations are predictive of COPD pathogenesis, but distinguishing between COPD patients and healthy individuals to establish a clear cut-off value is difficult because smoking status highly affects circulating SP-D levels. Herein, we focused on N-glycosylation in SP-D and examined whether or not N-glycosylation patterns in SP-D are associated with the pathogenesis of COPD. We performed an N-glycomic analysis of human serum SP-D and the results show that a core-fucose is present in its N-glycan. We also found that the N-glycosylation in serum SP-D was indeed altered in COPD, that is, fucosylation levels including core-fucosylation are significantly increased in COPD patients compared with non-COPD smokers. The severity of emphysema was positively associated with fucosylation levels in serum SP-D in smokers. Our findings shed new light on the discovery and/or development of a useful biomarker based on glycosylation changes for diagnosing COPD. This article is part of a Special Issue entitled: HUPO 2014.


Subject(s)
Fucose , Pulmonary Disease, Chronic Obstructive/blood , Pulmonary Surfactant-Associated Protein D/blood , Aged , Aged, 80 and over , Animals , Biomarkers/blood , Female , Glycosylation , Humans , Male , Mice , Mice, Knockout , Middle Aged
7.
Data Brief ; 5: 707-11, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26958622

ABSTRACT

In order to verify the protein enriched from pooled human sera to be a lung-specific protein surfactant protein-D (SP-D), we performed peptide mass fingerprinting (PMF)-based protein identification. MASCOT search results of the obtained PMF unequivocally demonstrated that it is identical to human SP-D. Meanwhile, we performed MALDI-QIT-TOF mass spectrometry-based N-glycomic analysis of the recombinant human SP-D produced in murine myeloma cells. The obtained mass spectra of N-glycans from the recombinant SP-D demonstrated that the recombinant protein is almost exclusively modified with core-fucosylated N-glycans [1].

8.
BMC Pulm Med ; 14: 196, 2014 Dec 08.
Article in English | MEDLINE | ID: mdl-25488319

ABSTRACT

BACKGROUND: Surfactant proteins SP-A and SP-D are useful biomarkers in diagnosis, monitoring, and prognosis of idiopathic pulmonary fibrosis (IPF). Despite their high structural homology, their serum concentrations often vary in IPF patients. This retrospective study aimed to investigate distinct compartmentalization of SP-A and SP-D in the vasculature and lungs by bronchoalveolar lavage fluid (BALF)/serum analysis, hydrophilicity and immunohistochemistry. METHODS: We included 36 IPF patients, 18 sarcoidosis (SAR) patients and 20 healthy subjects. Low-speed centrifugal supernatants of BALF (Sup-1) were obtained from each subject. Sera were also collected from each patient. Furthermore, we separated Sup-1 of IPF patients into hydrophilic supernatant (Sup-2) and hydrophobic precipitate (Ppt) by high-speed centrifugation. We measured SP-A and SP-D levels of each sample with the sandwich ELISA technique. We analyzed the change of the BALF/serum level ratios of the two proteins in IPF patients and their hydrophilicity in BALF. The distribution in the IPF lungs was also examined by immunohistochemical staining. RESULTS: In BALF, SP-A levels were comparable between the groups; however, SP-D levels were significantly lower in IPF patients than in others. Although IPF reduced the BALF/serum level ratios of the two proteins, the change in concentration of SP-D was more evident than SP-A. This suggests a higher disease impact for SP-D. Regarding hydrophilicity, although more than half of the SP-D remained in hydrophilic fractions (Sup-2), almost all of the SP-A sedimented in the Ppt with phospholipids. Hydrophilicity suggests that SP-D migrates into the blood more easily than SP-A in IPF lungs. Immunohistochemistry revealed that SP-A was confined to thick mucus-filling alveolar space, whereas SP-D was often intravascular. This data also suggests that SP-D easily leaks into the bloodstream, whereas SP-A remains bound to surfactant lipids in the alveolar space. CONCLUSIONS: The current study investigated distinct compartmentalization of SP-A and SP-D in the vasculature and lungs. Our results suggest that serum levels of SP-D could reflect pathological changes of the IPF lungs more incisively than those of SP-A.


Subject(s)
Bronchoalveolar Lavage Fluid/chemistry , Endothelium, Vascular/chemistry , Hydrophobic and Hydrophilic Interactions , Idiopathic Pulmonary Fibrosis/metabolism , Pulmonary Alveoli/chemistry , Pulmonary Surfactant-Associated Protein A/analysis , Pulmonary Surfactant-Associated Protein D/analysis , Aged , Biomarkers/analysis , Female , Humans , Male , Middle Aged , Pulmonary Surfactant-Associated Protein A/blood , Pulmonary Surfactant-Associated Protein D/blood , Retrospective Studies , Sarcoidosis/metabolism
9.
Biochem Biophys Res Commun ; 454(3): 364-8, 2014 11 21.
Article in English | MEDLINE | ID: mdl-25451255

ABSTRACT

It has been well documented that activation of the ErbB3-PI3K-Akt pathway is implicated in tumor survival and progression. We previously demonstrated that the single N-glycan deletion mutant of soluble ErbB3 protein (sErbB3 N418Q) attenuates heregulin ß1-induced ErbB3 signaling. The active PI3K-Akt pathway augments the nuclear accumulation of hypoxia inducible factor (HIF)-1α, which activates the transcription of many target genes and drives cancer progression. In this study, we focused on the effects of sErbB3 N418Q mutant on nuclear accumulation of HIF-1α. Pretreatment with the sErbB3 N418Q mutant suppressed heregulin ß1-induced HIF-1α activation in MCF7 cells. Similar results were also obtained in other breast cancer cell lines, T47D and BT474. Interestingly, these suppressive effects were not observed with the sErbB3 wild type. In addition, pretreatment with the sErbB3 N418Q mutant suppressed the cell migration of MCF7 cells induced by heregulin ß1. Furthermore, incubation with heregulin ß1 also induced the nuclear accumulation of Nrf2, and this effect was also reduced by the sErbB3 N418Q mutant, but not the sErbB3 wild type. These findings indicated that the sErbB3 N418Q mutant suppressed malignant formation of cancer cells by blocking of the HIF-1α and Nrf2 pathways.


Subject(s)
Breast Neoplasms/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , NF-E2-Related Factor 2/metabolism , Neuregulin-1/metabolism , Point Mutation , Receptor, ErbB-3/genetics , Signal Transduction , Breast/metabolism , Breast/pathology , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Movement , Disease Progression , Female , Gene Deletion , Humans , MCF-7 Cells , Receptor, ErbB-3/chemistry , Receptor, ErbB-3/metabolism , Solubility
10.
Protein Pept Lett ; 21(3): 292-305, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24164306

ABSTRACT

Leucine rich repeats (LRRs) are present in over 20,000 proteins from viruses to eukaryotes. Two to sixty-two LRRs occur in tandem. Each repeat is typically 20-30 residues long and can be divided into an HCS (Highly conserved segment) and a VS (Variable segment). The HCS part consists of an eleven or a twelve residue stretch, LxxLxLxxNx(x/-)L, in which "L" is Leu, Ile, Val, or Phe, "N" is Asn, Thr, Ser, or Cys, "x" is a non-conserved residue, and "-" is a possible deletion site. Eight classes have been recognized. However, there are many unclassified or unrecognized LRRs. Here we performed to search novel LRRs using protein sequence database. The novel LRR domains are present over three hundred proteins, which include fungal ECM33 protein and Monosiga brevicollis LRR receptor kinase, from unicellular eukaryotes and bacteria. The HCS part is clearly different from that of the known LRRs and consists of a twelve or a thirteen residue stretch, VxGx(L/F)x(L/C)xxNx(x/-)L, that is characterized by the addition of Gly between the first conserved Val and the second conserved Leu. The novel LRRs identified here form a new family. The novel LRR domains were classified into four classes. The VS parts of the two classes are consistent with those of known, normal "SDS22-like" and "IRREKO" classes, while the other two classes have unique VS parts. The structures, functions, and evolution of the novel LRR domains and their proteins are described. The present results should stimulate various experimental studies.


Subject(s)
Bacteria/chemistry , Bacterial Proteins/chemistry , Choanoflagellata/chemistry , Fungal Proteins/chemistry , Fungi/chemistry , Leucine/chemistry , Protozoan Proteins/chemistry , Amino Acid Sequence , Databases, Protein , Membrane Proteins/chemistry , Molecular Sequence Data , Protein Structure, Tertiary , Saccharomyces cerevisiae/chemistry , Saccharomyces cerevisiae Proteins/chemistry , Vibrio cholerae/chemistry
11.
J Biol Chem ; 288(46): 32910-21, 2013 Nov 15.
Article in English | MEDLINE | ID: mdl-24097984

ABSTRACT

Heregulin signaling is involved in various tumor proliferations and invasions; thus, receptors of heregulin are targets for the cancer therapy. In this study we examined the suppressing effects of extracellular domains of ErbB2, ErbB3, and ErbB4 (soluble ErbB (sErbB)) on heregulin ß signaling in human breast cancer cell line MCF7. It was found that sErbB3 suppresses ligand-induced activation of ErbB receptors, PI3K/Akt and Ras/Erk pathways most effectively; sErbB2 scarcely suppresses ligand-induced signaling, and sErbB4 suppresses receptor activation at ∼10% efficiency of sErbB3. It was revealed that sErbB3 does not decrease the effective ligands but decreases the effective receptors. By using small interfering RNA (siRNA) for ErbB receptors, we determined that sErbB3 suppresses the heregulin ß signaling by interfering ErbB3-containing heterodimers including ErbB2/ErbB3. By introducing the mutation of N418Q to sErbB3, the signaling-inhibitory effects were increased by 2-3-fold. Moreover, the sErbB3 N418Q mutant enhanced anticancer effects of lapatinib more effectively than the wild type. We also determined the structures of N-glycan on Asn-418. Results suggested that the N-glycan-deleted mutant of sErbB3 suppresses heregulin signaling via ErbB3-containing heterodimers more effectively than the wild type. Thus, we demonstrated that the sErbB3 N418Q mutant is a potent inhibitor for heregulin ß signaling.


Subject(s)
MAP Kinase Signaling System , Mutation, Missense , Neuregulin-1/metabolism , Protein Multimerization , Receptor, ErbB-3/metabolism , Amino Acid Substitution , Antineoplastic Agents/pharmacology , Cell Line, Tumor , ErbB Receptors/genetics , ErbB Receptors/metabolism , Humans , Lapatinib , Neuregulin-1/genetics , Protein Structure, Tertiary , Quinazolines/pharmacology , Receptor, ErbB-2/genetics , Receptor, ErbB-2/metabolism , Receptor, ErbB-3/genetics , Receptor, ErbB-4
12.
Proc Natl Acad Sci U S A ; 110(12): 4714-9, 2013 Mar 19.
Article in English | MEDLINE | ID: mdl-23471986

ABSTRACT

Although endogenous ligands for Toll-like receptor (TLR)4-myeloid differentiation factor 2 (MD2) have not been well-understood, we here report that a globo-series glycosphingolipid, globotetraosylceramide (Gb4), attenuates the toxicity of lipopolysaccharides (LPSs) by binding to TLR4-MD-2. Because α1,4-galactosyltransferase (A4galt)-deficient mice lacking globo-series glycosphingolipids showed higher sensitivity to LPS than wild-type mice, we examined mechanisms by which globo-series glycosphingolipids attenuate LPS toxicity. Cultured endothelial cells lacking A4galt showed higher expression of LPS-inducible genes upon LPS treatment. In turn, introduction of A4galt cDNA resulted in the neo expression of Gb4, leading to the reduced expression of LPS-inducible genes. Exogenous Gb4 induced similar effects. As a mechanism for the suppressive effects of Gb4 on LPS signals, specific binding of Gb4 to the LPS receptor TLR4-MD-2 was demonstrated by coprecipitation of Gb4 with recombinant MD-2 and by native PAGE. A docking model also supported these data. Taken together with colocalization of TLR4-MD-2 with Gb4 in lipid rafts after LPS stimulation, it was suggested that Gb4 competes with LPS for binding to TLR4-MD-2. Finally, administration of Gb4 significantly protected mice from LPS-elicited mortality. These results suggest that Gb4 is an endogenous ligand for TLR4-MD-2 and is capable of attenuating LPS toxicity, indicating the possibility for its therapeutic application in endotoxin shock.


Subject(s)
Globosides/immunology , Lymphocyte Antigen 96/immunology , Multiprotein Complexes/immunology , Toll-Like Receptor 4/immunology , Animals , Galactosyltransferases/genetics , Galactosyltransferases/immunology , Galactosyltransferases/metabolism , Gene Expression Regulation/drug effects , Gene Expression Regulation/genetics , Gene Expression Regulation/immunology , Globosides/genetics , Globosides/metabolism , Lipopolysaccharides/toxicity , Lymphocyte Antigen 96/genetics , Lymphocyte Antigen 96/metabolism , Membrane Microdomains/genetics , Membrane Microdomains/immunology , Membrane Microdomains/metabolism , Mice , Mice, Mutant Strains , Multiprotein Complexes/genetics , Multiprotein Complexes/metabolism , Protein Binding , Shock, Septic/chemically induced , Shock, Septic/genetics , Shock, Septic/immunology , Shock, Septic/metabolism , Toll-Like Receptor 4/genetics , Toll-Like Receptor 4/metabolism
13.
Cancer Sci ; 104(1): 43-7, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23004020

ABSTRACT

Gangliosides are glycosphingolipids found on the cell surface. They act as recognition molecules or signal modulators and regulate cell proliferation and differentiation. N-glycolylneuraminic acid (NeuGc)-containing gangliosides have been detected in some neoplasms in humans, although they are usually absent in normal human tissues. Our aim was to evaluate the presence of NeuGc-containing gangliosides including GM3 (NeuGc) and assess their relationship with the prognosis of non-small-cell lung cancer (NSCLC). NeuGc-containing ganglioside expression in NSCLC tissues was analyzed immunohistochemically using the mouse monoclonal antibody GMR8, which is specific for gangliosides with NeuGc alpha 2,3Gal-terminal structures. On the basis of NeuGc-containing ganglioside expression, we performed survival analysis. We also investigated the differences in the effects of GM3 (N-acetylneuraminic acid [NeuAc]) and GM3 (NeuGc) on inhibition of epidermal growth factor receptor (EGFR) tyrosine kinase in A431 cells. As a result, the presence of NeuGc-containing gangliosides was evident in 86 of 93 (93.5%) NSCLC samples. The NSCLC patients with high NeuGc-containing ganglioside expression had a low overall survival rate and a significantly low progression-free survival rate. In the in vitro study, the inhibitory effect of GM3 on EGFR tyrosine kinase in A431 cells after exposure to GM3 (NeuGc) was lower than that after exposure to GM3 (NeuAc). In conclusion, NeuGc-containing gangliosides including GM3 (NeuGc) are widely expressed in NSCLC, and NeuGc-containing ganglioside expression is associated with patient survival. The difference in the effects of GM3 (NeuGc) and GM3 (NeuAc) on the inhibition of EGFR tyrosine kinase might contribute to improvement in the prognosis of NSCLC patients.


Subject(s)
Carcinoma, Non-Small-Cell Lung/chemistry , Gangliosides/analysis , Lung Neoplasms/chemistry , N-Acetylneuraminic Acid/analysis , Neuraminic Acids/analysis , Adult , Aged , Aged, 80 and over , Antibodies, Monoclonal/immunology , Cell Line, Tumor , ErbB Receptors/antagonists & inhibitors , Female , G(M3) Ganglioside/analysis , Gangliosides/chemistry , Gangliosides/immunology , Glycosphingolipids/analysis , Humans , Immunohistochemistry , Male , Middle Aged , Phosphorylation , Prognosis , Survival Rate
14.
J Biol Chem ; 287(47): 39578-88, 2012 Nov 16.
Article in English | MEDLINE | ID: mdl-23012359

ABSTRACT

The adherence of uropathogenic Escherichia coli (UPEC) to the host urothelial surface is the first step for establishing UPEC infection. Uroplakin Ia (UPIa), a glycoprotein expressed on bladder urothelium, serves as a receptor for FimH, a lectin located at bacterial pili, and their interaction initiates UPEC infection. Surfactant protein D (SP-D) is known to be expressed on mucosal surfaces in various tissues besides the lung. However, the functions of SP-D in the non-pulmonary tissues are poorly understood. The purposes of this study were to investigate the possible function of SP-D expressed in the bladder urothelium and the mechanisms by which SP-D functions. SP-D was expressed in human bladder mucosa, and its mRNA was increased in the bladder of the UPEC infection model in mice. SP-D directly bound to UPEC and strongly agglutinated them in a Ca(2+)-dependent manner. Co-incubation of SP-D with UPEC decreased the bacterial adherence to 5637 cells, the human bladder cell line, and the UPEC-induced cytotoxicity. In addition, preincubation of SP-D with 5637 cells resulted in the decreased adherence of UPEC to the cells and in a reduced number of cells injured by UPEC. SP-D directly bound to UPIa and competed with FimH for UPIa binding. Consistent with the in vitro data, the exogenous administration of SP-D inhibited UPEC adherence to the bladder and dampened UPEC-induced inflammation in mice. These results support the conclusion that SP-D can protect the bladder urothelium against UPEC infection and suggest a possible function of SP-D in urinary tract.


Subject(s)
Bacterial Adhesion , Escherichia coli Infections/metabolism , Pulmonary Surfactant-Associated Protein D/metabolism , Urinary Bladder/metabolism , Urinary Tract Infections/metabolism , Uropathogenic Escherichia coli/metabolism , Urothelium/metabolism , Adhesins, Escherichia coli/genetics , Adhesins, Escherichia coli/metabolism , Animals , Escherichia coli Infections/pathology , Female , Fimbriae Proteins/genetics , Fimbriae Proteins/metabolism , Humans , Male , Mice , Pulmonary Surfactant-Associated Protein D/genetics , Rabbits , Tetraspanins/biosynthesis , Tetraspanins/genetics , Urinary Bladder/microbiology , Urinary Bladder/pathology , Urinary Tract Infections/pathology , Uroplakin Ia/biosynthesis , Uroplakin Ia/genetics , Urothelium/microbiology , Urothelium/pathology
15.
Biochim Biophys Acta ; 1820(11): 1787-96, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22820017

ABSTRACT

BACKGROUND: Aldehyde reductase (AKR1A; EC 1.1.1.2) catalyzes the reduction of various types of aldehydes. To ascertain the physiological role of AKR1A, we examined AKR1A knockout mice. METHODS: Ascorbic acid concentrations in AKR1A knockout mice tissues were examined, and the effects of human AKR1A transgene were analyzed. We purified AKR1A and studied the activities of glucuronate reductase and glucuronolactone reductase, which are involved in ascorbic acid biosynthesis. Metabolomic analysis and DNA microarray analysis were performed for a comprehensive study of AKR1A knockout mice. RESULTS: The levels of ascorbic acid in tissues of AKR1A knockout mice were significantly decreased which were completely restored by human AKR1A transgene. The activities of glucuronate reductase and glucuronolactone reductase, which are involved in ascorbic acid biosynthesis, were suppressed in AKR1A knockout mice. The accumulation of d-glucuronic acid and saccharate in knockout mice tissue and the expression of acute-phase proteins such as serum amyloid A2 are significantly increased in knockout mice liver. CONCLUSIONS: AKR1A plays a predominant role in the reduction of both d-glucuronic acid and d-glucurono-γ-lactone in vivo. The knockout of AKR1A in mice results in accumulation of d-glucuronic acid and saccharate as well as a deficiency of ascorbic acid, and also leads to upregulation of acute phase proteins. GENERAL SIGNIFICANCE: AKR1A is a major enzyme that catalyzes the reduction of d-glucuronic acid and d-glucurono-γ-lactone in vivo, besides acting as an aldehyde-detoxification enzyme. Suppression of AKR1A by inhibitors, which are used to prevent diabetic complications, may lead to the accumulation of d-glucuronic acid and saccharate.


Subject(s)
Aldehyde Reductase/physiology , Aldehyde Reductase/genetics , Animals , Ascorbic Acid/analysis , Calcium-Binding Proteins/analysis , Female , Glucuronates/metabolism , Glucuronic Acid/metabolism , Humans , Intracellular Signaling Peptides and Proteins/analysis , Liver/chemistry , Male , Metabolomics , Mice , Mice, Inbred C57BL , Mice, Knockout , Oligonucleotide Array Sequence Analysis
16.
J Biomed Biotechnol ; 2012: 532071, 2012.
Article in English | MEDLINE | ID: mdl-22675254

ABSTRACT

Pulmonary surfactant is a mixture of lipids and proteins that covers alveolar surfaces and keeps alveoli from collapsing. Four specific proteins have been identified in surfactant. Among them, two C-type lectins, surfactant proteins A and D (SP-A and SP-D), are known to be implicated in host defense and regulation of inflammatory responses of the lung. These host defense lectins are structurally characterized by N-terminal collagen-like domains and lectin domains and are called pulmonary collectins. They prevent dissemination of infectious microbes by their biological activities including agglutination and growth inhibition. They also promote clearance of microbes by enhancing phagocytosis in macrophages. In addition, they interact with the other pattern-recognition molecules, including Toll-like receptors (TLRs) and TLR-associated molecules, CD14 and MD-2, and regulate inflammatory responses. Furthermore, recent studies have demonstrated that these collectins modulate functions of neutrophil-derived innate immune molecules by interacting with them. These findings indicate that pulmonary collectins play critical roles in host defense of the lung.


Subject(s)
Collectins/immunology , Pneumonia/immunology , Pulmonary Surfactants/immunology , Animals , Humans
17.
Infect Immun ; 80(8): 2956-62, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22615243

ABSTRACT

We propose two antigenic types of Helicobacter pylori lipopolysaccharides (LPS): highly antigenic epitope-carrying LPS (HA-LPS) and weakly antigenic epitope-carrying LPS (WA-LPS) based on human serum reactivity. Strains carrying WA-LPS are highly prevalent in isolates from gastric cancer patients. WA-LPS exhibits more potent biological activities compared to HA-LPS, namely, upregulation of Toll-like receptor 4 (TLR4) expression and induction of enhanced epithelial cell proliferation. The results of competitive binding assays using monosaccharides and methylglycosides, as well as binding assays using glycosidase-treated LPS, suggested that ß-linked N-acetyl-D-glucosamine and ß-linked D-galactose residues largely contributed to the highly antigenic epitope and the weakly antigenic epitope, respectively. WA-LPS exhibited greater binding activity to surfactant protein D (SP-D) in a Ca(2+)-dependent manner, and this interaction was inhibited by methyl-ß-D-galactoside. The biological activities of WA-LPS were markedly enhanced by the addition of SP-D. Lines of evidence suggested that removal of ß-N-acetyl-D-glucosamine residue, which comprises the highly antigenic epitope, results in exposure of the weakly antigenic epitope. The weakly antigenic epitope interacted preferentially with SP-D, and SP-D enhanced the biological activity of WA-LPS.


Subject(s)
Helicobacter pylori/metabolism , Lipopolysaccharides/metabolism , Pulmonary Surfactant-Associated Protein D/metabolism , Antigens, Bacterial/metabolism , Blotting, Western , Cell Line , Cell Proliferation , Enzyme-Linked Immunosorbent Assay , Epithelial Cells/metabolism , Epithelial Cells/microbiology , Epitopes/chemistry , Epitopes/immunology , Epitopes/metabolism , Glycoside Hydrolases/metabolism , Helicobacter pylori/cytology , Helicobacter pylori/genetics , Humans , Interleukin-8/genetics , Interleukin-8/metabolism , Lipopolysaccharides/immunology , Protein Binding , Pulmonary Surfactant-Associated Protein D/genetics , Stomach/cytology , Toll-Like Receptor 4/genetics , Toll-Like Receptor 4/metabolism
18.
Gene ; 503(2): 235-43, 2012 Jul 25.
Article in English | MEDLINE | ID: mdl-22587897

ABSTRACT

Toll-like receptors (TLRs) that initiate an innate immune response contain an extracellular leucine rich repeat (LRR) domain and an intracellular Toll IL-receptor (TIR) domain. There are fifteen different TLRs in vertebrates. The LRR domains, which adopt a solenoid structure, usually have higher rates of evolution than do the TIR globular domains. It is important to understand the molecular evolution and functional roles of TLRs from this standpoint. Both pairwise genetic distances and Ka/Ks's (the ratios between non synonymous and synonymous substitution rates) were compared between the LRR domain and the TIR domain of 366 vertebrate TLRs from 96 species (from fish to primates). In fourteen members (TLRs 1, 2, 3, 4, 5, 6, 7, 8, 9, 11/12, 13, 14, 21, and 22/23) the LRR domains evolved significantly more rapidly than did the corresponding TIR domains. The evolutionary rates of the LRR domains are significantly different among these members; LRR domains from TLR3 and TLR7 from primates to fishes have the lowest rate of evolution. In contrast, the fifteenth member, TLR10, shows no significant differences; its TIR domain is not highly conserved. The present results suggest that TLR10 may have a different function in signaling from those other members and that a higher conservation of TLR3 and TLR7 may reflect a more ancient mechanism and/or structure in the innate immune response system. Gene conversions are suggested to have occurred in platypus TLR6 and TLR10. This study provides new insight about structural and functional diversification of vertebrate TLRs.


Subject(s)
Evolution, Molecular , Toll-Like Receptors/chemistry , Toll-Like Receptors/genetics , Vertebrates/genetics , Amino Acid Motifs , Amino Acid Sequence , Animals , Biological Evolution , Protein Structure, Tertiary , Sequence Alignment , Signal Transduction , Vertebrates/immunology
19.
J Biol Chem ; 287(18): 15034-43, 2012 Apr 27.
Article in English | MEDLINE | ID: mdl-22418431

ABSTRACT

Defensins are important molecules in the innate immune system that eliminate infectious microbes. They also exhibit cytotoxicity against host cells in higher concentrations. The mechanisms by which hosts protect their own cells from cytotoxicity of defensins have been poorly understood. We found that the cytotoxicity of human ß-defensin 3 (hBD3) against lung epithelial cells was dose-dependently attenuated by pulmonary surfactant protein A (SP-A), a collectin implicated in host defense and regulation of inflammatory responses in the lung. The direct interaction between SP-A and hBD3 may be an important factor in decreasing this cytotoxicity because preincubation of epithelial cells with SP-A did not affect the cytotoxicity. Consistent with in vitro analysis, intratracheal administration of hBD3 to SP-A(-/-) mice resulted in more severe tissue damage compared with that in WT mice. These data indicate that SP-A protects lung epithelium from tissue injury caused by hBD3. Furthermore, we found that the functional region of SP-A lies within Tyr(161)-Lys(201). Synthetic peptide corresponding to this region, tentatively called SP-A Y161-G200, also inhibited cytotoxicity of hBD3 in a dose-dependent manner. The SP-A Y161-G200 is a candidate as a therapeutic reagent that prevents tissue injury during inflammation.


Subject(s)
Cytotoxins/pharmacology , Lung/metabolism , Peptides/pharmacology , Pulmonary Surfactant-Associated Protein A/metabolism , Respiratory Mucosa/metabolism , beta-Defensins/pharmacology , Animals , Cell Line , Cytotoxins/adverse effects , Cytotoxins/metabolism , Humans , Lung/pathology , Mice , Mice, Knockout , Pneumonia/drug therapy , Pneumonia/metabolism , Pneumonia/pathology , Protein Binding , Pulmonary Surfactant-Associated Protein A/genetics , Respiratory Mucosa/pathology , beta-Defensins/adverse effects , beta-Defensins/metabolism
20.
Int Immunol ; 24(2): 97-106, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22190574

ABSTRACT

Hepatitis C virus (HCV) chronic infection is characterized by low-level or undetectable cellular immune response against HCV antigens. HCV proteins affect various intracellular events and modulate immune responses, although the mechanisms that mediate these effects are not fully understood. In this study, we examined the effect of HCV proteins on the differentiation of human peripheral blood monocytes to dendritic cells (DCs). The HCV core (HCVc) and non-structural 3 (NS3) proteins inhibited the expression of CD1a, CD1b and DC-SIGN during monocyte differentiation to DCs, while increasing some markers characteristic of macrophages (CD14 and HLA-DR) and also PD-L1 expression. Meanwhile, HCVc and NS3 could induce differentiating monocytes to secrete IL-10. However, anti-IL-10 mAb could not reverse HCVc and NS3 inhibition of monocyte differentiation into DCs. The HCVc and NS3 proteins increased IL-6 secretion both in immature and in fully differentiated DCs and also promoted CD4+ T-cell IL-17 production. Since T(h) 17 cells are active in many examples of immunopathology, these effects may contribute to HCV autoimmune responses in chronically infected patients.


Subject(s)
Dendritic Cells/immunology , Hepacivirus/immunology , Hepatitis C Antigens/immunology , Hepatitis C, Chronic/immunology , Th17 Cells/immunology , Viral Core Proteins/immunology , Viral Nonstructural Proteins/immunology , Antigens, CD/immunology , Cell Differentiation/immunology , Cells, Cultured , Gene Expression Regulation/immunology , Humans , Immunomodulation , Interleukin-10/immunology , Interleukin-10/metabolism , Interleukin-6/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL