Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
1.
PLoS One ; 19(5): e0302628, 2024.
Article in English | MEDLINE | ID: mdl-38723000

ABSTRACT

Blood vessels permit the selective passage of molecules and immune cells between tissues and circulation. Uncontrolled inflammatory responses from an infection can increase vascular permeability and edema, which can occasionally lead to fatal organ failure. We identified mexenone as a vascular permeability blocker by testing 2,910 compounds in the Clinically Applied Compound Library using the lipopolysaccharide (LPS)-induced vascular permeability assay. Mexenone suppressed the LPS-induced downregulation of junctional proteins and phosphorylation of VE-cadherin in Bovine Aortic Endothelial Cells (BAECs). The injection of mexenone 1 hr before LPS administration completely blocked LPS-induced lung vascular permeability and acute lung injury in mice after 18hr. Our results suggest that mexenone-induced endothelial cell (EC) barrier stabilization could be effective in treating sepsis patients.


Subject(s)
Endothelial Cells , Lipopolysaccharides , Sepsis , Animals , Sepsis/drug therapy , Sepsis/chemically induced , Sepsis/metabolism , Mice , Cattle , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Capillary Permeability/drug effects , Acute Lung Injury/chemically induced , Acute Lung Injury/drug therapy , Acute Lung Injury/metabolism , Acute Lung Injury/pathology , Acute Lung Injury/prevention & control , Male , Cadherins/metabolism , Mice, Inbred C57BL , Antigens, CD/metabolism
2.
J Periodontal Res ; 56(3): 492-500, 2021 Jun.
Article in English | MEDLINE | ID: mdl-33641180

ABSTRACT

BACKGROUND AND OBJECTIVES: Periodontal pathogens initiate various diseases and induce inflammatory host responses. The activation of inflammasomes triggers caspase-1 and interleukin (IL)-1ß-mediated pyroptosis via gasdermin D (GSDMD). Differentiated embryo chondrocyte 2 (Dec2) is a transcription repressor that controls the expression of genes involved in innate immune and inflammatory responses. However, the effects of Dec2 on inflammasome-induced pyroptosis in periodontal tissues remain elusive. This study aimed to characterize the activation of Dec2 inflammasomes that contribute to P. gingivalis lipopolysaccharide (LPS)-induced pyroptosis and its functional and regulatory importance in periodontal inflammation. MATERIALS AND METHODS: Human gingival fibroblasts (HGFs) and human periodontal ligament fibroblasts (HPDLFs) were stimulated with P. gingivalis LPS in vitro. An experimental periodontitis mouse model (wild-type (WT) and Dec2KO) was established to profile periodontal pyroptosis. RESULTS: The results demonstrate that P. gingivalis LPS activates caspase-1, caspase-11, and NF-κB in HGFs and in HPDLFs. siRNA knockdown of Dec2 stimulated the induction and further upregulated LPS-induced pyroptosis in HGFs and HPDLFs, resulting in the release of IL-1ß. Further, a deficiency of Dec2 alleviated periodontal pyroptosis via the transcriptional induction of GSDMD. In addition, P. gingivalis-induced IL-1ß expression and Dec2-deficient mice subsequently increased the inflammatory effect of P. gingivalis in HGFs and in HPDLFs, confirming the importance of Dec2 in the activation of inflammasomes and the regulation of pyroptosis. CONCLUSION: Our results demonstrate that Dec2 alleviates periodontal pyroptosis by regulating the expression of NF-κB, caspase-1 and GSDMD, suggesting that Dec2 is a crucial component of inflammasome activation and subsequent pyroptosis.


Subject(s)
Inflammasomes , Pyroptosis , Animals , Caspase 1 , Cells, Cultured , Inflammation , Interleukin-1beta , Intracellular Signaling Peptides and Proteins , Mice , Phosphate-Binding Proteins
3.
Mol Biol Rep ; 48(2): 1423-1431, 2021 Feb.
Article in English | MEDLINE | ID: mdl-33507476

ABSTRACT

Periodontal ligament fibroblasts (PDLFs) are integral to the homeostasis of periodontal tissue. The transcription factor Dec1 functions to modulate Porphyromonas gingivalis-induced periodontal inflammation. Here, we aimed to characterize the Dec1-mediated autophagy in PDLFs under inflammatory conditions. Human PDLFs were subjected to an inflammatory environment using P. gingivalis Lipopolysaccaride (LPS) along with Dec1 siRNA in vitro. Quantitative real-time polymerase chain reaction and Western blot analyses were used to evaluate the expression levels of autophagy-related genes and their upstream AKT/mTOR signaling pathways. An experimental P. gingivalis-treated Dec1 knockout (Dec1KO) mouse model was used to confirm the expression of autophagy in PDLFs in vivo. Treatment with P. gingivalis LPS induced the expression of ATG5, Beclin1 and microtubule-associated protein 1 light chain 3 (LC3) and elevated the expression of pro-inflammatory cytokine IL-1ß and Dec1 in human PDLFs. Knockdown of Dec1 partly reversed the detrimental influences of LPS on these autophagy markers in human PDLFs. The inhibition of autophagy with Dec1 siRNA suppressed the inflammatory effect of AKT/mTOR signaling pathways following treatment with P. gingivalis LPS. P. gingivalis-treated Dec1KO mice partly reduced autophagy expression. These findings suggest that a Dec1 deficiency can modulate the interaction between autophagy and inflammation in PDLFs.


Subject(s)
Autophagy/genetics , Basic Helix-Loop-Helix Transcription Factors/genetics , Homeodomain Proteins/genetics , Inflammation/genetics , Periodontal Ligament/metabolism , Tumor Suppressor Proteins/genetics , Animals , Autophagy-Related Protein 5/genetics , Basic Helix-Loop-Helix Transcription Factors/antagonists & inhibitors , Beclin-1/genetics , Fibroblasts/metabolism , Fibroblasts/pathology , Gene Expression Regulation/genetics , Homeodomain Proteins/antagonists & inhibitors , Humans , Inflammation/chemically induced , Inflammation/pathology , Lipopolysaccharides/toxicity , Mice , Mice, Knockout , Microtubule-Associated Proteins/genetics , Periodontal Ligament/microbiology , Periodontal Ligament/pathology , Porphyromonas gingivalis/pathogenicity , Proto-Oncogene Proteins c-akt/genetics , RNA, Small Interfering/genetics , RNA, Small Interfering/pharmacology , Signal Transduction/genetics , TOR Serine-Threonine Kinases/genetics
4.
Anal Chem ; 92(13): 8715-8721, 2020 07 07.
Article in English | MEDLINE | ID: mdl-32449357

ABSTRACT

Mass spectrometry imaging (MSI) based on matrix-assisted laser desorption/ionization (MALDI) provides information on the identification and spatial distribution of biomolecules. Quantitative analysis, however, has been challenging largely due to heterogeneity in both the size of the matrix crystals and the extraction area. In this work, we present a compartmentalized elastomeric stamp for quantitative MALDI-MSI of adsorbed peptides. Filling the compartments with matrix solution and stamping onto a planar substrate extract and concentrate analytes adsorbed in each compartment into a single analyte-matrix cocrystal over the entire stamped area. Walls between compartments help preserve spatial information on the adsorbates. The mass intensity of the cocrystals directly correlates with the surface coverage of analytes, which enables not only quantitative analysis but estimation of an equilibrium constant for the adsorption. We demonstrate via MALDI-MSI relative quantitation of peptides adsorbed along a microchannel with varying surface coverages.


Subject(s)
Peptides/chemistry , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization/methods , Adsorption , Fluorescein-5-isothiocyanate/chemistry , Lab-On-A-Chip Devices , Microscopy, Fluorescence
5.
Adv Biol Regul ; 75: 100654, 2020 01.
Article in English | MEDLINE | ID: mdl-31558431

ABSTRACT

During neural development, growing neuronal cells consistently sense and communicate with their surroundings through the use of signaling molecules. In this process, spatiotemporally well-coordinated intracellular signaling is a prerequisite for proper neuronal network formation. Thus, intense interest has focused on investigating the signaling mechanisms in neuronal structure formation that link the activation of receptors to the control of cell shape and motility. Recent studies suggest that Phospholipase C gamma1 (PLCγ1), a signal transducer, plays key roles in nervous system development by mediating specific ligand-receptor systems. In this overview of the most recent advances in the field, we discuss the mechanisms by which extracellular stimuli trigger PLCγ1 signaling and, the role PLCγ1 in nervous system development.


Subject(s)
Nerve Net/enzymology , Phospholipase C gamma/metabolism , Signal Transduction/physiology , Animals , Mice
6.
J Nanosci Nanotechnol ; 16(2): 1396-9, 2016 Feb.
Article in English | MEDLINE | ID: mdl-27433593

ABSTRACT

This study examined the effect of the immobilization of the Gly-Arg-Gly-Asp-Ser (GRGDS) peptide on titanium dioxide (TiO2) nanotube via chemical grafting on osteoblast-like cell (MG-63) viability and differentiation. The specimens were divided into two groups; TiO2 nanotubes and GRGDS-immobilized TiO2 nanotubes. The surface characteristics of GRGDS-immobilized TiO2 nanotubes were observed by using X-ray photoelectron spectroscopy (XPS) and a field emission scanning electron microscope (FE-SEM). The morphology of cells on specimens was observed by FE-SEM after 2 hr and 24 hr. The level of cell viability was investigated via a tetrazolium (XTT) assay after 2 and 4 days. Alkaline phosphatase (ALP) activity was evaluated to measure the cell differentiation after 4 and 7 days. The presence of nitrogen up-regulation or C==O carbons con- firmed that TiO2 nanotubes were immobilized with GRGDS peptides. Cell adhesion was enhanced on the GRGDS-immobilized TiO2 nanotubes compared to TiO2 nanotubes. Furthermore, significantly increased cell spreading and proliferation were observed with the cells grown on GRGDS-immobilized TiO2 nanotubes (P < .05). However, there was no significant difference in ALP activity between GRGDS-immobilized TiO2 nanotubes and TiO2 nanotubes. These results suggest that the GRGDS-immobilized TiO2 nanotubes might be effective in improving the osseointegration of dental implants.


Subject(s)
Cell Differentiation/drug effects , Nanotubes/chemistry , Oligopeptides , Osteoblasts/metabolism , Titanium , Cell Line, Tumor , Cell Survival/drug effects , Humans , Oligopeptides/chemistry , Oligopeptides/pharmacology , Osteoblasts/cytology , Titanium/chemistry , Titanium/pharmacology
7.
Diabetologia ; 58(12): 2867-76, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26342595

ABSTRACT

AIMS/HYPOTHESIS: O-GlcNAcylation plays a role as a metabolic sensor regulating cellular signalling, transcription and metabolism. Transcription factors and signalling pathways related to metabolism are modulated by N-acetyl-glucosamine (O-GlcNAc) modification. Aberrant regulation of O-GlcNAcylation is closely linked to insulin resistance, type 2 diabetes and obesity. Current evidence shows that increased O-GlcNAcylation negatively regulates insulin signalling, which is associated with insulin resistance and type 2 diabetes. Here, we aimed to evaluate the effects of Oga (also known as Mgea5) haploinsufficiency, which causes hyper-O-GlcNAcylation, on metabolism. METHODS: We examined whether Oga(+/-) mice developed insulin resistance. Metabolic variables were determined including body weight, glucose and insulin tolerance, metabolic rate and thermogenesis. RESULTS: Oga deficiency does not affect insulin signalling even at hyper-O-GlcNAc levels. Oga(+/-) mice are lean with reduced fat mass and improved glucose tolerance. Furthermore, Oga(+/-) mice resist high-fat diet-induced obesity with ameliorated hepatic steatosis and improved glucose metabolism. Oga haploinsufficiency potentiates energy expenditure through the enhancement of brown adipocyte differentiation from the stromal vascular fraction of subcutaneous white adipose tissue (WAT). CONCLUSIONS/INTERPRETATION: Our observations suggest that O-GlcNAcase (OGA) is essential for energy metabolism via regulation of the thermogenic WAT program.


Subject(s)
Adipose Tissue, Brown/metabolism , Adipose Tissue, White/metabolism , Energy Metabolism/genetics , Obesity/genetics , beta-N-Acetylhexosaminidases/genetics , Acetylglucosamine/metabolism , Adipocytes, Brown/metabolism , Adipocytes, Brown/pathology , Animals , Blood Glucose/metabolism , Body Weight/genetics , Cell Differentiation , Diabetes Mellitus, Type 2/genetics , Glucose Intolerance/genetics , Insulin Resistance/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Signal Transduction , Thermogenesis/genetics
8.
Cell Signal ; 27(12): 2488-95, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26385316

ABSTRACT

Chronic inflammation in adipose tissue is highly associated with insulin resistance. Herein, we demonstrate that a novel modification of PPARγ is strongly associated with inflammatory responses in adipose tissue. c-Src kinase directly phosphorylated PPARγ at Tyr78, and this process was reversed by protein tyrosine phosphatase-1B (PTP-1B). In adipocytes, phosphorylation of PPARγ suppressed the expression of pro-inflammatory genes as well as the secretion of chemokines and cytokines, thus reducing macrophage migration. Importantly, pharmacological inhibition of c-Src kinase aggravated insulin resistance in obese mice with a concomitant increase in the expression of pro-inflammatory genes in adipose tissue. These data strongly suggest that PPARγ phosphorylation is the key regulatory mechanism of the inflammatory response in adipose tissue, which is highly associated with glucose tolerance and insulin sensitivity. Furthermore, these data increase our understanding of the mechanical aspects of developing novel anti-diabetic drugs targeting PPARγ phosphorylation.


Subject(s)
Insulin Resistance , Obesity/metabolism , PPAR gamma/metabolism , Protein Processing, Post-Translational , Adipose Tissue/immunology , Adipose Tissue/metabolism , Amino Acid Sequence , Animals , CSK Tyrosine-Protein Kinase , Chemokines/genetics , Chemokines/metabolism , Gene Expression Regulation , HEK293 Cells , Humans , Male , Mice , Mice, Inbred C57BL , Obesity/immunology , Phosphorylation , Protein Tyrosine Phosphatase, Non-Receptor Type 1/metabolism , RAW 264.7 Cells , src-Family Kinases/physiology
9.
Placenta ; 36(10): 1063-8, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26286378

ABSTRACT

INTRODUCTION: Placental vasculogenesis is essential for fetal growth and development, and is affected profoundly by oxygen tension (hypoxia). Hypoxia-inducible factor-1α (HIF-1α), which is stabilized at the protein level in response to hypoxia, is essential for vascular morphogenesis in the placenta. Many studies suggested that responses to hypoxia is influenced by O-GlcNAcylation. O-GlcNAcylation is regulated by O-GlcNAc transferase (OGT) and O-GlcNAcase (OGA) that catalyze the addition and removal of O-GlcNAc respectively. METHODS: We generated OGA deficient mice and evaluated OGA(-/-) placentas. The analysis of OGA(-/-) placentas was focused on morphological change and placental vasculogenesis. HIF-1α protein stability or transcriptional activity under dysregulation of O-GlcNAcylation were evaluated by Western blot, RT-qPCR and luciferase reporter gene assays in MEFs or MS1 cell line. RESULTS: Deletion of OGA results in defective placental vasculogenesis. OGA(-/-) placentas showed an abnormal placental shape and reduced vasculature in the labyrinth, which caused a developmental delay in the embryos. OGA deletion, which elevates O-GlcNAcylation and downregulates O-GlcNAc transferase (OGT), suppressed HIF-1α stabilization and the transcription of its target genes. In contrast, the overexpression of O-GlcNAc cycling enzymes enhanced the expression and transcriptional activity of HIF-1α. DISCUSSION: These results suggest that OGA plays a critical role in placental vasculogenesis by modulating HIF-1α stabilization. Control of O-GlcNAcylation is essential for placental development.


Subject(s)
Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , N-Acetylglucosaminyltransferases/metabolism , Neovascularization, Physiologic , Placenta/blood supply , beta-N-Acetylhexosaminidases/metabolism , Animals , Cell Line , Female , Hypoxia/enzymology , Male , Mice, Inbred C57BL , Placenta/embryology , Placenta/enzymology , Placental Circulation , Pregnancy
10.
Oncotarget ; 6(14): 12529-42, 2015 May 20.
Article in English | MEDLINE | ID: mdl-25915426

ABSTRACT

O-GlcNAcylation is a reversible post-translational modification. O-GlcNAc addition and removal is catalyzed by O-GlcNAc transferase (OGT) and O-GlcNAcase (OGA), respectively. More recent evidence indicates that regulation of O-GlcNAcylation is important for inflammatory diseases and tumorigenesis. In this study, we revealed that O-GlcNAcylation was increased in the colonic tissues of dextran sodium sulfate (DSS)-induced colitis and azoxymethane (AOM)/DSS-induced colitis-associated cancer (CAC) animal models. Moreover, the O-GlcNAcylation level was elevated in human CAC tissues compared with matched normal counterparts. To investigate the functional role of O-GlcNAcylation in colitis, we used OGA heterozygote mice, which have an increased level of O-GlcNAcylation. OGA(+/-) mice have higher susceptibility to DSS-induced colitis than OGA(+/+) mice. OGA(+/-) mice exhibited a higher incidence of colon tumors than OGA(+/+) mice. In molecular studies, elevated O-GlcNAc levels were shown to enhance the activation of NF-κB signaling through increasing the binding of RelA/p65 to its target promoters. We also found that Thr-322 and Thr352 in the p65-O-GlcNAcylation sites are critical for p65 promoter binding. These results suggest that the elevated O-GlcNAcylation level in colonic tissues contributes to the development of colitis and CAC by disrupting regulation of NF-κB-dependent transcriptional activity.


Subject(s)
Colitis/genetics , Colonic Neoplasms/genetics , Gene Expression Regulation, Neoplastic/genetics , NF-kappa B/metabolism , beta-N-Acetylhexosaminidases/metabolism , Animals , Blotting, Western , Cell Transformation, Neoplastic/genetics , Colitis/metabolism , Colitis/pathology , Colonic Neoplasms/metabolism , Colonic Neoplasms/pathology , Disease Models, Animal , Humans , Mice , Mice, Inbred C57BL , Mice, Transgenic , Protein Processing, Post-Translational
11.
Sci Rep ; 4: 4805, 2014 Jun 13.
Article in English | MEDLINE | ID: mdl-24925581

ABSTRACT

Adipose tissue functions as an endocrine organ, and the development of systemic inflammation in adipose tissue is closely associated with metabolic diseases, such as obesity and insulin resistance. Accordingly, the fine regulation of the inflammatory response caused by obesity has therapeutic potential for the treatment of metabolic syndrome. In this study, we analyzed the role of DJ-1 (PARK7) in adipogenesis and inflammation related to obesity in vitro and in vivo. Many intracellular functions of DJ-1, including oxidative stress regulation, are known. However, the possibility of DJ-1 involvement in metabolic disease is largely unknown. Our results suggest that DJ-1 deficiency results in reduced adipogenesis and the down-regulation of pro-inflammatory cytokines in vitro. Furthermore, DJ-1-deficient mice show a low-level inflammatory response in the high-fat diet-induced obesity model. These results indicate previously unknown functions of DJ-1 in metabolism and therefore suggest that precise regulation of DJ-1 in adipose tissue might have a therapeutic advantage for metabolic disease treatment.


Subject(s)
Adipogenesis/physiology , Adipose Tissue/pathology , Disease Models, Animal , Inflammation/etiology , Obesity/complications , Oncogene Proteins/physiology , Peroxiredoxins/physiology , 3T3-L1 Cells , Adipose Tissue/metabolism , Animals , Apoptosis , Cell Proliferation , Cells, Cultured , Cytokines/genetics , Cytokines/metabolism , Diet, High-Fat/adverse effects , Immunoenzyme Techniques , Inflammation/metabolism , Inflammation/pathology , Insulin Resistance , Mice , Mice, Knockout , Obesity/metabolism , Obesity/pathology , Protein Deglycase DJ-1 , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction
12.
J Clin Periodontol ; 41(3): 262-8, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24256094

ABSTRACT

AIM: We assessed the association of periodontal disease and number of missing teeth with subclinical atherosclerosis in an adult Korean population. MATERIALS AND METHODS: Cross-sectional data from 5404 individuals aged ≥50 years were obtained from the 2008-2010 Dong-gu study. Periodontal examinations were conducted to determine the number of missing teeth, pocket depth (PD), clinical attachment loss (CAL), and bleeding on probing (BOP). The percentages of sites with PD ≥ 4 mm (PD 4%), CAL ≥ 4 mm (CAL 4%), and BOP (BOP%) were recorded for each participant. B-mode ultrasound was performed to determine common carotid artery intima-media thickness (CCA IMT) and the presence of carotid plaques. Multivariate linear regression models were used to assess the associations between periodontal parameters and CCA IMT and carotid plaque. RESULTS: Number of missing teeth was associated with increased CCA IMT, and BOP% was associated with increased CCA IMT in females only. This association was robust in never smokers. CONCLUSIONS: The number of missing teeth was associated with CCA IMT, and BOP% was associated with CCA IMT in females only. These associations were robust in never smokers. Our results suggest that tooth loss due to oral disease may play a role in subclinical carotid atherosclerosis.


Subject(s)
Periodontal Diseases/epidemiology , Plaque, Atherosclerotic/epidemiology , Tooth Loss/epidemiology , Aged , Aged, 80 and over , Carotid Artery Diseases/diagnostic imaging , Carotid Artery Diseases/epidemiology , Carotid Artery, Common/diagnostic imaging , Cross-Sectional Studies , Female , Humans , Male , Middle Aged , Periodontal Attachment Loss/epidemiology , Periodontal Index , Periodontal Pocket/epidemiology , Periodontitis/epidemiology , Plaque, Atherosclerotic/diagnostic imaging , Republic of Korea/epidemiology , Risk Factors , Sex Factors , Smoking/epidemiology , Tunica Intima/diagnostic imaging , Tunica Media/diagnostic imaging , Ultrasonography
13.
Mol Ther ; 21(5): 1004-13, 2013 May.
Article in English | MEDLINE | ID: mdl-23511245

ABSTRACT

Periostin is an extracellular matrix (ECM) protein that is overexpressed in a variety of human cancers, and its functions appear to be linked to tumor growth, metastasis, and angiogenesis. Recent clinical evidence suggests that aberrant periostin expression is correlated with poor outcome in patients with breast cancer. To identify novel tools to regulate the functional role of periostin, we generated benzyl-d(U)TP-modified DNA aptamers that were directed against human periostin (PNDAs) and characterized their functional roles in breast cancer progression. PNDA-3 selectively bound to the FAS-1 domain of periostin with nanomolar affinity and disrupted the interaction between periostin and its cell surface receptors, αvß3 and αvß5 integrins. PNDA-3 markedly antagonized the periostin-induced adhesion, migration, and invasion of breast cancer cells and blocked the activation of various components of the αvß3 and αvß5 integrin signal transduction pathways. In a 4T1 orthotopic mouse model, PNDA-3 administration significantly reduced primary tumor growth and distant metastasis. Thus, our results demonstrated that periostin-integrin signaling regulates breast cancer progression at multiple levels in tumor cells and the tumor microenvironment. DNA aptamers targeting periostin may potentially be used to inhibit breast cancer progression.


Subject(s)
Aptamers, Nucleotide/genetics , Aptamers, Nucleotide/metabolism , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Cell Adhesion Molecules/metabolism , Animals , Aptamers, Nucleotide/chemistry , Base Sequence , Breast Neoplasms/pathology , Cell Adhesion/genetics , Cell Line, Tumor , Cell Movement/genetics , Cell Proliferation , Disease Models, Animal , Female , Humans , Integrins/metabolism , Mice , Molecular Sequence Data , Neoplasm Metastasis , Protein Binding , Signal Transduction , Tumor Burden/drug effects , Xenograft Model Antitumor Assays
14.
J Biol Chem ; 287(49): 41268-76, 2012 Nov 30.
Article in English | MEDLINE | ID: mdl-23043110

ABSTRACT

Protrudin is a FYVE (Fab 1, YOTB, Vac 1, and EEA1) domain-containing protein involved in transport of neuronal cargoes and implicated in the onset of hereditary spastic paraplegia. Our image-based screening of the lipid binding domain library revealed novel plasma membrane localization of the FYVE domain of protrudin unlike canonical FYVE domains that are localized to early endosomes. The membrane binding study by surface plasmon resonance analysis showed that this FYVE domain preferentially binds phosphatidylinositol 4,5-bisphosphate (PtdIns(4,5)P(2)), phosphatidylinositol 3,4-bisphosphate (PtdIns(3,4)P(2)), and phosphatidylinositol 3,4,5-trisphosphate (PtdIns(3,4,5)P(3)) unlike canonical FYVE domains that specifically bind phosphatidylinositol 3-phosphate (PtdIns(3)P). Furthermore, we found that these phosphoinositides (PtdInsP) differentially regulate shuttling of protrudin between endosomes and plasma membrane via its FYVE domain. Protrudin mutants with reduced PtdInsP-binding affinity failed to promote neurite outgrowth in primary cultured hippocampal neurons. These results suggest that novel PtdInsP selectivity of the protrudin-FYVE domain is critical for its cellular localization and its role in neurite outgrowth.


Subject(s)
Carrier Proteins/biosynthesis , Phosphatidylinositol Phosphates/chemistry , Phosphatidylinositols/metabolism , Amino Acid Sequence , Animals , Carrier Proteins/chemistry , Carrier Proteins/genetics , Carrier Proteins/metabolism , Cell Membrane/metabolism , Endosomes/metabolism , Kinetics , Lipids/chemistry , Mice , Models, Biological , Models, Molecular , Molecular Sequence Data , Mutation , NIH 3T3 Cells , Neurites/metabolism , Protein Conformation , Protein Structure, Tertiary , Sequence Homology, Amino Acid , Surface Plasmon Resonance/methods , Vesicular Transport Proteins
15.
PLoS One ; 4(9): e7090, 2009 Sep 18.
Article in English | MEDLINE | ID: mdl-19763255

ABSTRACT

BACKGROUND: Upon ligand binding, cell surface signaling receptors are internalized through a process tightly regulated by endocytic proteins and adaptor protein 2 (AP2) to orchestrate them. Although the molecular identities and roles of endocytic proteins are becoming clearer, it is still unclear what determines the receptor endocytosis kinetics which is mainly regulated by the accumulation of endocytic apparatus to the activated receptors. METHODOLOGY/PRINCIPAL FINDINGS: Here we employed the kinetic analysis of endocytosis and adaptor recruitment to show that mu2, a subunit of AP2 interacts directly with phospholipase D (PLD)1, a receptor-associated signaling protein and this facilitates the membrane recruitment of AP2 and the endocytosis of epidermal growth factor receptor (EGFR). We also demonstrate that the PLD1-mu2 interaction requires the binding of PLD1 with phosphatidic acid, its own product. CONCLUSIONS/SIGNIFICANCE: These results suggest that the temporal regulation of EGFR endocytosis is achieved by auto-regulatory PLD1 which senses the receptor activation and triggers the translocation of AP2 near to the activated receptor.


Subject(s)
Adaptor Protein Complex 2/metabolism , Adaptor Protein Complex mu Subunits/metabolism , ErbB Receptors/metabolism , Phospholipase D/metabolism , Endocytosis , Glutathione Transferase/metabolism , HeLa Cells , Humans , Kinetics , Ligands , Models, Biological , Phosphatidic Acids/metabolism , Protein Binding , Protein Transport , RNA Interference , Signal Transduction
16.
Cell Signal ; 19(8): 1784-96, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17531443

ABSTRACT

Phospholipase C-gamma1 (PLC-gamma1), which generates two second messengers, namely, inositol-1, 4, 5-trisphosphate and diacylglycerol, is implicated in growth factor-mediated chemotaxis. However, the exact role of PLC-gamma1 in integrin-mediated cell adhesion and migration remains poorly understood. In this study, we demonstrate that PLC-gamma1 is required for actin cytoskeletal organization and cell motility through the regulation of Pyk2 and paxillin activation. After fibronectin stimulation, PLC-gamma1 directly interacted with the cytoplasmic tail of integrin beta1. In PLC-gamma1-silenced cells, integrin-induced Pyk2 and paxillin phosphorylation were significantly reduced and PLC-gamma1 potentiated the integrin-induced Pyk2/paxillin activation in its enzymatic activity-dependent manner. In addition, specific knock-down of PLC-gamma1 resulted in a failure to form focal adhesions dependent on fibronectin stimulation, which appeared to be caused by the suppression of Pyk2 and paxillin phosphorylation. Interestingly, PLC-gamma1 potentiated the activations of Rac, thus integrin-induced lamellipodia formation was up-regulated. Consequently, the strength of cell-substratum interaction and cell motility were profoundly up-regulated by PLC-gamma1. Taken together, these results suggest that PLC-gamma1 is a key player in integrin-mediated cell spreading and motility achieved by the activation of Pyk2/paxillin/Rac signaling.


Subject(s)
Cell Movement/physiology , Focal Adhesion Kinase 2/metabolism , Integrins/metabolism , Paxillin/metabolism , Phospholipase C gamma/metabolism , Animals , Cell Adhesion/physiology , Enzyme Activation , Fibroblasts/metabolism , Mice , Models, Biological , NIH 3T3 Cells , Phospholipase C gamma/isolation & purification , RNA, Small Interfering/metabolism
17.
Cell Signal ; 18(12): 2283-91, 2006 Dec.
Article in English | MEDLINE | ID: mdl-16837165

ABSTRACT

Mammalian target-of-rapamycin (mTOR), which is a master controller of cell growth, senses a mitogenic signal in part through the lipid second messenger phosphatidic acid (PA), generated by phospholipase D (PLD). To understand further which isozymes of PLD are involved in this process, we compared the effect of PLD isozymes on mTOR activation. We found that PLD2 has an essential role in mitogen-induced mTOR activation as the siRNA-mediated knockdown of PLD2, not of PLD1, profoundly reduced the phosphorylations of S6K1 and 4EBP1, well-known mTOR effectors. Furthermore, exogenous PA-induced mTOR activation was abrogated by PLD2 knockdown, but not by PLD1 knockdown. This abrogation was found to be the result of complex formation between PLD2 and mTOR/raptor. PLD2 possesses a TOS-like motif (Phe-Glu-Val-Gln-Val, a.a. 265-269), through which it interacts with raptor independently of the other TOS motif-containing proteins, S6K1 and 4EBP1. PLD2-dependent mTOR activation appears to require PLD2 binding to mTOR/raptor with lipase activity, since lipase-inactive PLD2 cannot trigger mTOR activation despite its ability to interact with mTOR/raptor. Abrogation of mitogen-dependent mTOR activation by PLD2 knockdown was rescued only by wild type PLD2, but not by raptor binding-deficient and lipase-inactive PLD2. Our results demonstrate the importance of localized PA generation for the mitogen-induced activation of mTOR, which is achieved by a specific interaction between PLD2 and mTOR/raptor.


Subject(s)
Mitogens/metabolism , Phospholipase D/metabolism , Protein Kinases/metabolism , Signal Transduction , Amino Acid Sequence , Animals , Binding Sites/genetics , Blotting, Western , COS Cells , Cell Line , Chlorocebus aethiops , Genetic Vectors/genetics , Humans , Immunoprecipitation , Mutation/genetics , Phospholipase D/genetics , Protein Binding , Protein Kinases/genetics , TOR Serine-Threonine Kinases , Transfection
19.
J Cell Sci ; 117(Pt 17): 3785-95, 2004 Aug 01.
Article in English | MEDLINE | ID: mdl-15252117

ABSTRACT

Phospholipase C-gamma1 (PLC-gamma1), which interacts with a variety of signaling molecules through its two Src homology (SH) 2 domains and a single SH3 domain has been implicated in the regulation of many cellular functions. We demonstrate that PLC-gamma1 acts as a guanine nucleotide exchange factor (GEF) of dynamin-1, a 100 kDa GTPase protein, which is involved in clathrin-mediated endocytosis of epidermal growth factor (EGF) receptor. Overexpression of PLC-gamma1 increases endocytosis of the EGF receptor by increasing guanine nucleotide exchange activity of dynamin-1. The GEF activity of PLC-gamma1 is mediated by the direct interaction of its SH3 domain with dynamin-1. EGF-dependent activation of ERK and serum response element (SRE) are both up-regulated in PC12 cells stably overexpressing PLC-gamma1, but knockdown of PLC-gamma1 by siRNA significantly reduces ERK activation. These results establish a new role for PLC-gamma1 in the regulation of endocytosis and suggest that endocytosis of activated EGF receptors may mediate PLC-gamma1-dependent proliferation.


Subject(s)
Dynamin I/metabolism , ErbB Receptors/metabolism , Guanine Nucleotide Exchange Factors/metabolism , Type C Phospholipases/physiology , Animals , Brain/metabolism , Cell Proliferation , Clathrin/metabolism , Dose-Response Relationship, Drug , Endocytosis , Genes, Reporter , Guanosine Triphosphate/metabolism , Immunoprecipitation , PC12 Cells , Phospholipase C gamma , Protein Structure, Tertiary , RNA, Small Interfering/metabolism , Rats , Signal Transduction , Time Factors , Transcription, Genetic , Up-Regulation , src Homology Domains
20.
J Biol Chem ; 279(16): 16339-48, 2004 Apr 16.
Article in English | MEDLINE | ID: mdl-14744865

ABSTRACT

Mammalian phospholipase D (PLD) has been reported to be a key enzyme for epidermal growth factor (EGF)-induced cellular signaling, however, the regulatory mechanism of PLD is still unclear. In this report, we found that Munc-18-1 is a potent negative regulator of PLD in the basal state and that its inhibition is abolished by EGF stimulation. We investigated PLD-binding proteins obtained from rat brain extract, and identified a 67-kDa protein as Munc-18-1 by peptide-mass finger-printing. The direct association between PLD and Munc-18-1 was confirmed by in vitro binding analysis using the purified proteins, and their binding sites were identified as the phox homology domain of PLD and multiple sites of Munc-18-1. PLD activity was potently inhibited by Munc-18-1 in vitro (IC50 = 2-5 nm), and the cotransfection of COS-7 cells with Munc-18-1 and PLD inhibited basal PLD activity in vivo. In the basal state, Munc-18-1 coprecipitated with PLD and colocalized with PLD2 at the plasma membrane of COS-7 cells. EGF treatment triggered the dissociation of Munc-18-1 from PLD when PLD was activated by EGF. The dissociation of the endogenous interaction between Munc-18-1 and PLD, and the activation of PLD by EGF were also observed in primary cultured chromaffin cells. These results suggest that Munc-18-1 is a potent negative regulator of basal PLD activity and that EGF stimulation abolishes this interaction.


Subject(s)
Epidermal Growth Factor/metabolism , Nerve Tissue Proteins/metabolism , Phospholipase D/metabolism , Vesicular Transport Proteins/metabolism , Amino Acid Sequence , Animals , Binding Sites/genetics , Down-Regulation , Enzyme Activation , Humans , Molecular Sequence Data , Munc18 Proteins , Nerve Tissue Proteins/genetics , Phospholipase D/antagonists & inhibitors , Phospholipase D/genetics , Protein Binding , Protein Structure, Tertiary , Rats , Sequence Deletion , Vesicular Transport Proteins/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...