Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
Biochim Biophys Acta Gen Subj ; 1868(4): 130565, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38244702

ABSTRACT

N-glycosylation and proper processing of N-glycans are required for the function of membrane proteins including cell surface receptors. Fibroblast growth factor receptor (FGFR) is involved in a wide variety of biological processes including embryonic development, osteogenesis, angiogenesis, and cell proliferation. Human FGFR3 contains six potential N-glycosylation sites, however, the roles of glycosylation have not been elucidated. The site-specific profiles of N-glycans of the FGFR3 extracellular domain expressed and secreted by CHO-K1 cells were examined, and glycan occupancies and structures of four sites were determined. The results indicated that most sites were fully occupied by glycans, and the dominant populations were the complex type. By examining single N-glycan deletion mutants of FGFR3, it was found that N262Q mutation significantly increased the population with oligomannose-type N-glycans, which was localized in the endoplasmic reticulum. Protein stability assay suggested that fraction with oligomannose-type N-glycans in the N262Q mutant is more stable than those in the wild type and other mutants. Furthermore, it was found that ligand-independent phosphorylation was significantly upregulated in N262Q mutants with complex type N-glycans. The findings suggest that N-glycans on N262 of FGFR3 affect the intracellular localization and phosphorylation status of the receptor.


Subject(s)
Biological Phenomena , Polysaccharides , Cricetinae , Animals , Humans , Phosphorylation , Glycosylation , CHO Cells , Cricetulus , Polysaccharides/metabolism , Receptor, Fibroblast Growth Factor, Type 3/genetics , Receptor, Fibroblast Growth Factor, Type 3/metabolism
2.
Glycobiology ; 34(3)2024 Apr 01.
Article in English | MEDLINE | ID: mdl-38109791

ABSTRACT

Glycans found on receptor tyrosine kinases (RTKs) have emerged as promising targets for cancer chemotherapy, aiming to address issues such as drug resistance. However, to effectively select the target glycans, it is crucial to define the structure and function of candidate glycans in advance. Through mass spectrometric analysis, this study presents a "glycoform atlas" of epidermal growth factor receptor 2 (ErbB2), an RTK targeted for the treatment of ErbB2-positive cancers. Our analysis provides an in-depth and site-specific glycosylation profile, including both asparagine- and serine/threonine-linked glycosylation. Molecular dynamics simulations of N-glycosylated ErbB2 incorporating the identified glycan structures suggested that the N-glycan at N124 on the long flexible loop in the N-terminal region plays a role in stabilizing the ErbB2 structure. Based on the model structures obtained from the simulations, analysis employing an ErbB2 mutant deficient in N-glycosylation at N124 exhibited a significantly shorter intracellular half-life and suppressed autophosphorylation compared to wild-type ErbB2. Moreover, a structural comparison between the N-glycosylated forms of ErbB2 and its structurally homologous receptor, epidermal growth factor receptor (EGFR), demonstrated distinct variations in the distribution and density of N-glycans across these two molecules. These findings provide valuable insights into the structural and functional implications of ErbB2 glycosylation and will contribute to facilitating the establishment of glycan-targeted therapeutic strategies for ErbB2-positive cancers.


Subject(s)
Neoplasms , Humans , Glycosylation , Phosphorylation , Polysaccharides/metabolism
3.
Nat Commun ; 14(1): 1205, 2023 03 02.
Article in English | MEDLINE | ID: mdl-36864068

ABSTRACT

Pulmonary alveolar microlithiasis is an autosomal recessive lung disease caused by a deficiency in the pulmonary epithelial Npt2b sodium-phosphate co-transporter that results in accumulation of phosphate and formation of hydroxyapatite microliths in the alveolar space. The single cell transcriptomic analysis of a pulmonary alveolar microlithiasis lung explant showing a robust osteoclast gene signature in alveolar monocytes and the finding that calcium phosphate microliths contain a rich protein and lipid matrix that includes bone resorbing osteoclast enzymes and other proteins suggested a role for osteoclast-like cells in the host response to microliths. While investigating the mechanisms of microlith clearance, we found that Npt2b modulates pulmonary phosphate homeostasis through effects on alternative phosphate transporter activity and alveolar osteoprotegerin, and that microliths induce osteoclast formation and activation in a receptor activator of nuclear factor-κB ligand and dietary phosphate dependent manner. This work reveals that Npt2b and pulmonary osteoclast-like cells play key roles in pulmonary homeostasis and suggest potential new therapeutic targets for the treatment of lung disease.


Subject(s)
Lung Diseases , Osteogenesis , Humans , Homeostasis , Lung
4.
bioRxiv ; 2023 Feb 18.
Article in English | MEDLINE | ID: mdl-36824953

ABSTRACT

The pathophysiology of silicosis is poorly understood, limiting development of therapies for those who have been exposed to the respirable particle. We explored the mechanisms of silica-induced pulmonary fibrosis in a mouse model using multiple modalities including whole-lung single-nucleus RNA sequencing. These analyses revealed that in addition to pulmonary inflammation and fibrosis, intratracheal silica challenge induced osteoclast-like differentiation of alveolar macrophages and recruited monocytes, driven by induction of the osteoclastogenic cytokine, receptor activator of nuclear factor-κB ligand (RANKL) in pulmonary lymphocytes and alveolar type II cells. Furthermore, anti-RANKL monoclonal antibody treatment suppressed silica-induced osteoclast-like differentiation in the lung and attenuated silica-induced pulmonary fibrosis. We conclude that silica induces osteoclast-like differentiation of distinct recruited and tissue resident monocyte populations, leading to progressive lung injury, likely due to sustained elaboration of bone resorbing proteases and hydrochloric acid. Interrupting osteoclast-like differentiation may therefore constitute a promising avenue for moderating lung damage in silicosis.

5.
Cancer Sci ; 113(4): 1292-1304, 2022 Apr.
Article in English | MEDLINE | ID: mdl-35092134

ABSTRACT

MET, the receptor for the hepatocyte growth factor (HGF), is strongly associated with resistance to tyrosine kinase inhibitors, key drugs that are used in the therapy of non-small cell lung cancer. MET contains 11 potential N-glycosylation sites, but the site-specific roles of these N-glycans have not been elucidated. We report herein that these N-glycans regulate the proteolytic processing of MET and HGF-induced MET signaling, and that this regulation is site specific. Inhibitors of N-glycosylation were found to suppress the processing and trafficking of endogenous MET in H1975 and EBC-1 lung cancer cells and exogenous MET in CHO-K1 cells. We purified the recombinant extracellular domain of human MET and determined the site-specific N-glycan structures and occupancy using mass spectrometry. The results indicated that most sites were fully glycosylated and that the dominant population was the complex type. To examine the effects of the deletion of N-glycans of MET, we prepared endogenous MET knockout Flp-In CHO cells and transfected them with a series of N-glycan-deletion mutants of MET. The results showed that several N-glycans are implicated in the processing of MET. The findings also suggested that the N-glycans of the SEMA domain of MET positively regulate HGF signaling, and the N-glycans of the region other than the SEMA domain negatively regulate HGF signaling. Processing, cell surface expression, and signaling were significantly suppressed in the case of the all-N-glycan-deletion mutant. The overall findings suggest that N-glycans of MET affect the status and the function of the receptor in a site-specific manner.


Subject(s)
Carcinoma, Non-Small-Cell Lung , Lung Neoplasms , Animals , Cricetinae , Cricetulus , Glycosylation , Hepatocyte Growth Factor/metabolism , Humans , Lung Neoplasms/drug therapy , Proto-Oncogene Proteins c-met
6.
Biochemistry ; 60(21): 1708-1721, 2021 06 01.
Article in English | MEDLINE | ID: mdl-33983715

ABSTRACT

Endoplasmic reticulum (ER) stress has been reported in a variety of diseases. Although ER stress can be detected using specific markers, it is still difficult to quantitatively evaluate the degree of stress and to identify the cause of the stress. The ER is the primary site for folding of secretory or transmembrane proteins as well as the site where glycosylation is initiated. This study therefore postulates that tracing the biosynthetic pathway of asparagine-linked glycans (N-glycans) would be a reporter for reflecting the state of the ER and serve as a quantitative descriptor of ER stress. Glycoblotting-assisted mass spectrometric analysis of the HeLa cell line enabled quantitative determination of the changes in the structures of N-glycans and degraded free oligosaccharides (fOSs) in response to tunicamycin- or thapsigargin-induced ER stress. The integrated analysis of neutral and sialylated N-glycans and fOSs showed the potential to elucidate the cause of ER stress, which cannot be readily done by protein markers alone. Changes in the total amount of glycans, increase in the ratio of high-mannose type N-glycans, increase in fOSs, and changes in the ratio of sialylated N-glycans in response to ER stress were shown to be potential descriptors of ER stress. Additionally, drastic clearance of accumulated N-glycans was observed in thapsigargin-treated cells, which may suggest the observation of ER stress-mediated autophagy or ER-phagy in terms of glycomics. Quantitative analysis of N-glycoforms composed of N-glycans and fOSs provides the dynamic indicators reflecting the ER status and the promising strategies for quantitative evaluation of ER stress.


Subject(s)
Endoplasmic Reticulum Stress/physiology , Endoplasmic Reticulum/pathology , Asparagine/metabolism , Biomarkers , Glycosylation , HeLa Cells , Humans , Mannose/metabolism , Mass Spectrometry/methods , Membrane Proteins/metabolism , Oligosaccharides/chemistry , Oligosaccharides/metabolism , Polysaccharides/chemistry , Polysaccharides/metabolism , Structure-Activity Relationship , Tunicamycin/pharmacology
7.
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
8.
Proc Natl Acad Sci U S A ; 114(32): E6613-E6622, 2017 08 08.
Article in English | MEDLINE | ID: mdl-28739896

ABSTRACT

Development of pneumonia is the most lethal consequence of influenza, increasing mortality more than 50-fold compared with uncomplicated infection. The spread of viral infection from conducting airways to the alveolar epithelium is therefore a pivotal event in influenza pathogenesis. We found that mitogenic stimulation with keratinocyte growth factor (KGF) markedly accelerated mortality after infectious challenge with influenza A virus (IAV). Coadministration of KGF with IAV markedly accelerated the spread of viral infection from the airways to alveoli compared with challenge with IAV alone, based on spatial and temporal analyses of viral nucleoprotein staining of lung tissue sections and dissociated lung cells. To better define the temporal relationship between KGF administration and susceptibility to IAV infection in vivo, we administered KGF 120, 48, 24, and 0 h before intrapulmonary IAV challenge and assessed the percentages of proliferating and IAV-infected, alveolar type II (AECII) cells in dispersed lung cell populations. Peak AECII infectivity coincided with the timing of KGF administration that also induced peak AECII proliferation. AECII from mice that were given intrapulmonary KGF before isolation and then infected with IAV ex vivo exhibited the same temporal pattern of proliferation and infectious susceptibility. KGF-induced increases in mortality, AECII proliferation, and enhanced IAV susceptibility were all reversed by pretreatment of the animals with the mTOR inhibitor rapamycin before mitogenic stimulation. Taken together, these data suggest mTOR signaling-dependent, mitogenic conditioning of AECII is a determinant of host susceptibility to infection with IAV.


Subject(s)
Alveolar Epithelial Cells/metabolism , Cell Proliferation/drug effects , Fibroblast Growth Factor 7/pharmacology , Influenza A virus/metabolism , Mitogens/pharmacology , Orthomyxoviridae Infections/metabolism , Alveolar Epithelial Cells/pathology , Animals , Disease Susceptibility/chemically induced , Female , Mice , Mice, Inbred DBA , Orthomyxoviridae Infections/pathology
9.
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
10.
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
11.
Sci Transl Med ; 7(313): 313ra181, 2015 Nov 11.
Article in English | MEDLINE | ID: mdl-26560359

ABSTRACT

Pulmonary alveolar microlithiasis (PAM) is a rare, autosomal recessive lung disorder associated with progressive accumulation of calcium phosphate microliths. Inactivating mutations in SLC34A2, which encodes the NPT2b sodium-dependent phosphate cotransporter, has been proposed as a cause of PAM. We show that epithelial deletion of Npt2b in mice results in a progressive pulmonary process characterized by diffuse alveolar microlith accumulation, radiographic opacification, restrictive physiology, inflammation, fibrosis, and an unexpected alveolar phospholipidosis. Cytokine and surfactant protein elevations in the alveolar lavage and serum of PAM mice and confirmed in serum from PAM patients identify serum MCP-1 (monocyte chemotactic protein 1) and SP-D (surfactant protein D) as potential biomarkers. Microliths introduced by adoptive transfer into the lungs of wild-type mice produce marked macrophage-rich inflammation and elevation of serum MCP-1 that peaks at 1 week and resolves at 1 month, concomitant with clearance of stones. Microliths isolated by bronchoalveolar lavage readily dissolve in EDTA, and therapeutic whole-lung EDTA lavage reduces the burden of stones in the lungs. A low-phosphate diet prevents microlith formation in young animals and reduces lung injury on the basis of reduction in serum SP-D. The burden of pulmonary calcium deposits in established PAM is also diminished within 4 weeks by a low-phosphate diet challenge. These data support a causative role for Npt2b in the pathogenesis of PAM and the use of the PAM mouse model as a preclinical platform for the development of biomarkers and therapeutic strategies.


Subject(s)
Biomarkers/blood , Calcinosis/etiology , Calcinosis/physiopathology , Calcinosis/therapy , Genetic Diseases, Inborn/etiology , Genetic Diseases, Inborn/physiopathology , Genetic Diseases, Inborn/therapy , Lung Diseases/etiology , Lung Diseases/physiopathology , Lung Diseases/therapy , Sodium-Phosphate Cotransporter Proteins, Type IIb/deficiency , Sodium-Phosphate Cotransporter Proteins, Type IIb/genetics , Animals , Diet , Disease Models, Animal , Epithelium/metabolism , Epithelium/pathology , Lung/metabolism , Lung/pathology , Mice , Mutation , Phosphates/metabolism , Pulmonary Alveoli/metabolism
12.
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
13.
Nihon Kokyuki Gakkai Zasshi ; 49(2): 87-92, 2011 Feb.
Article in Japanese | MEDLINE | ID: mdl-21400903

ABSTRACT

A 15-year-old asymptomatic girl had an abnormal shadow pointed out on a chest roentgenogram during a school medical health check. Her chest X-ray films showed a mass in the left pulmonary hilum that had increased in size during the past 2 years. Chest computed tomography (CT) showed a mass, which showed slight accumulation on FDG-PET in the left pulmonary hilum. Left lower lobectomy was performed for diagnosis, and the histopathological diagnosis was hyaline vascular-type Castleman disease. We report a case of hyaline vascular-type Castleman disease in the right pulmonary hilum which increased quite rapidly.


Subject(s)
Castleman Disease/pathology , Lung/pathology , Adolescent , Castleman Disease/diagnostic imaging , Female , Fluorodeoxyglucose F18 , Humans , Hyalin , Lung/diagnostic imaging , Positron-Emission Tomography , Tomography, X-Ray Computed
SELECTION OF CITATIONS
SEARCH DETAIL
...