Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 103
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Nat Immunol ; 14(6): 554-63, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23624557

ABSTRACT

Microenvironment-based alterations in phenotypes of mast cells influence the susceptibility to anaphylaxis, yet the mechanisms underlying proper maturation of mast cells toward an anaphylaxis-sensitive phenotype are incompletely understood. Here we report that PLA2G3, a mammalian homolog of anaphylactic bee venom phospholipase A2, regulates this process. PLA2G3 secreted from mast cells is coupled with fibroblastic lipocalin-type PGD2 synthase (L-PGDS) to provide PGD2, which facilitates mast-cell maturation via PGD2 receptor DP1. Mice lacking PLA2G3, L-PGDS or DP1, mast cell-deficient mice reconstituted with PLA2G3-null or DP1-null mast cells, or mast cells cultured with L-PGDS-ablated fibroblasts exhibited impaired maturation and anaphylaxis of mast cells. Thus, we describe a lipid-driven PLA2G3-L-PGDS-DP1 loop that drives mast cell maturation.


Subject(s)
Group III Phospholipases A2/immunology , Mast Cells/immunology , Paracrine Communication/immunology , Prostaglandin D2/immunology , Receptors, Prostaglandin/immunology , Animals , Blotting, Western , Bone Marrow Cells/immunology , Bone Marrow Cells/metabolism , Cell Differentiation/genetics , Cell Differentiation/immunology , Cells, Cultured , Fibroblasts/cytology , Fibroblasts/immunology , Fibroblasts/metabolism , Gene Expression Profiling , Group III Phospholipases A2/genetics , Group III Phospholipases A2/metabolism , Humans , Intramolecular Oxidoreductases/genetics , Intramolecular Oxidoreductases/immunology , Intramolecular Oxidoreductases/metabolism , Lipocalins/genetics , Lipocalins/immunology , Lipocalins/metabolism , Mast Cells/metabolism , Mast Cells/ultrastructure , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , Microscopy, Electron, Transmission , Oligonucleotide Array Sequence Analysis , Paracrine Communication/genetics , Prostaglandin D2/metabolism , Receptors, Prostaglandin/genetics , Receptors, Prostaglandin/metabolism , Reverse Transcriptase Polymerase Chain Reaction
2.
Mol Biol Rep ; 49(7): 5875-5882, 2022 Jul.
Article in English | MEDLINE | ID: mdl-35347543

ABSTRACT

AIMS: Although skin manifestations are common in diabetic patients, its characteristics are poorly identified. This study explored the differentiation process of keratinocytes in type 2 diabetes mellitus (T2DM) in vivo. METHODS: Back skin of T2DM model KKAy/TaJcl mice (KKAy) and C57BL/6JJcl mice (control) aged 8 and 12 weeks was used. The mRNA expression of differentiation markers of keratinocytes was measured by quantitative real-time polymerase chain reaction (qRT-PCR). The expression of each marker in situ was examined immunohistochemically. RESULTS: KKAy mice showed hyperglycemia versus control mice. The histological findings showed increased thickness and structural impairment of epidermal tissue in KKAy mice. The qRT-PCR revealed that the expression of integrin beta 1 and keratin 14 in KKAy and control mice was identical. However, the expression of involucrin at 8 weeks, keratin 10 at 12 weeks, and filaggrin and loricrin at 8 and 12 weeks was decreased in KKAy mice. Immunohistochemical findings showed that filaggrin was markedly decreased in KKAy mice, though Ki-67 remained unchanged. CONCLUSION: The terminal differentiation process was impaired in the diabetic skin, while keratinocyte proliferation was preserved. Damaged terminal differentiation of keratinocytes may contribute to impairment of the skin barrier function in diabetic dermatoses.


Subject(s)
Diabetes Mellitus, Experimental , Diabetes Mellitus, Type 2 , Animals , Cell Differentiation , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Type 2/metabolism , Epidermis/metabolism , Keratinocytes/metabolism , Mice , Mice, Inbred C57BL
3.
Biosci Biotechnol Biochem ; 86(4): 490-501, 2022 Mar 21.
Article in English | MEDLINE | ID: mdl-35040954

ABSTRACT

The first layer of active plant immunity relies upon the recognition of pathogen-associated molecular patterns (PAMPs), and the induction of PTI. Flagellin is the major protein component of the bacterial flagellum. Flagellin-derived peptide fragments such as CD2-1, flg22, and flgII-28 function as PAMPs in most higher plants. To determine the distribution of CD2-1, flg22, and flgII-28 recognition systems within plant species, the inducibility of PTI by CD2-1, flg22, and flgII-28 in 8 plant species, including monocotyledonous and dicotyledonous plants, was investigated. CD2-1 caused PTI responses in Oryza sativa, Brachypodium distachyon, and Asparagus persicus; flg22 caused PTI responses in Phyllostachys nigra, A. persicus, Arabidopsis thaliana, Nicotiana tabacum, Solanum lycopersicum, and Lotus japonicus; and flgII-28 caused PTI responses only in S. lycopersicum. Furthermore, quantitative analysis of FLS2 receptor revealed that the responsiveness of flg22 in plants was dependent on the expression level of the receptor.


Subject(s)
Flagellin , Plant Immunity , Plants/immunology , Flagellin/genetics , Flagellin/metabolism , Gene Expression Regulation, Plant , Plant Diseases/microbiology
4.
Mol Plant Microbe Interact ; 34(2): 186-197, 2021 Feb.
Article in English | MEDLINE | ID: mdl-33135963

ABSTRACT

Microbial pathogens deliver effectors into plant cells to suppress plant immune responses and modulate host metabolism in order to support infection processes. We sought to determine if the Acidovorax avenae rice-virulent K1 strain can suppress pathogen-associated molecular pattern-triggered immunity (PTI) induced by flagellin isolated from the rice-avirulent N1141 strain. The flagellin-triggered PTI, including H2O2 generation, callose deposition, and expression of several immune-related genes were strongly suppressed in K1 preinoculated cultured rice cells in a type III secretion system (T3SS)-dependent manner. By screening 4,562 transposon-tagged mutants based on their suppression ability, we found that 156 transposon-tagged K1 mutants lost the ability to suppress PTI induction. Mutant sequence analysis, comprehensive expression analysis using RNA sequencing, and the prediction of secretion through T3SS showed that a protein named A. avenae K1 suppression factor 1 (AKSF1) suppresses flagellin-triggered PTI in rice. Translocation of AKSF1 protein into rice cells is dependent on the T3SS during infection, an AKSF1-disruption mutant lost the ability to suppress PTI responses, and expression of AKSF1 in the AKSF1-disruption mutant complemented the suppression activity. When AKSF1-disruption mutants were inoculated into the host rice plant, reduction of the disease symptoms and suppression of bacterial growth were observed. Taken together, our results demonstrate that AKSF1 is a novel effector that can suppress the PTI in a host rice plant.[Formula: see text] The author(s) have dedicated the work to the public domain under the Creative Commons CC0 "No Rights Reserved" license by waiving all of his or her rights to the work worldwide under copyright law, including all related and neighboring rights, to the extent allowed by law. 2021.


Subject(s)
Comamonadaceae , Oryza , Pathogen-Associated Molecular Pattern Molecules , Plant Immunity , Comamonadaceae/genetics , Comamonadaceae/pathogenicity , Oryza/immunology , Oryza/microbiology , Pathogen-Associated Molecular Pattern Molecules/immunology , Plant Diseases/microbiology
5.
Int J Mol Sci ; 22(13)2021 Jun 28.
Article in English | MEDLINE | ID: mdl-34203170

ABSTRACT

Acidovorax avenae is a flagellated, pathogenic bacterium to various plant crops that has also been found in human patients with haematological malignancy, fever, and sepsis; however, the exact mechanism for infection in humans is not known. We hypothesized that the human innate immune system could be responsive to the purified flagellin isolated from A. avenae, named FLA-AA. We observed the secretion of inflammatory cytokines such as tumor necrosis factor-alpha (TNF-α), interleukin (IL)-6, and IL-8 by treating FLA-AA to human dermal fibroblasts, as well as macrophages. This response was exclusively through TLR5, which was confirmed by using TLR5-overexpression cell line, 293/hTLR5, as well as TLR5-specific inhibitor, TH1020. We also observed the secretion of inflammatory cytokine, IL-1ß, by the activation of NLRC4 with FLA-AA. Overall, our results provide a molecular basis for the inflammatory response caused by FLA-AA in cell-based assays.


Subject(s)
Comamonadaceae/chemistry , Flagellin/pharmacology , Immunity, Innate/physiology , CARD Signaling Adaptor Proteins/metabolism , Calcium-Binding Proteins/metabolism , Fibroblasts/drug effects , Fibroblasts/metabolism , Humans , Immunity, Innate/genetics , Interleukin-18/metabolism , Interleukin-6/metabolism , Macrophages/drug effects , Macrophages/metabolism , Signal Transduction/drug effects , Tumor Necrosis Factor-alpha/metabolism
6.
Cardiovasc Diabetol ; 19(1): 85, 2020 06 13.
Article in English | MEDLINE | ID: mdl-32534578

ABSTRACT

BACKGROUND: Type 2 diabetes (T2D) is associated with renal impairment and vascular endothelial dysfunction. Therefore, this pathological connection is an important therapeutic target. Recent cardiovascular and renal outcome trials demonstrated that sodium glucose cotransporter 2 inhibitors (SGLT2is) consistently reduced the risks of cardiovascular and renal events and mortality in patients with T2D and various other background risks including chronic kidney disease (CKD). However, the precise mechanisms by which SGLT2is accords these therapeutic benefits remain uncertain. It is also unknown whether these SGLT2is-associated benefits are associated with the amelioration of endothelial dysfunction in patients with T2D and CKD. METHODS: The PROCEED trial is an investigator-initiated, prospective, multicenter, open-label, randomized-controlled trial. The target sample size is 110 subjects. After they furnish informed consent and their endothelial dysfunction is confirmed from their decreased reactive hyperemia indices (RHI), eligible participants with T2D (HbA1c, 6.0-9.0%) and established CKD (30 mL/min/1.73 m2 ≤ estimated glomerular filtration ratio [eGFR] < 60 and/or ≥ urine albumin-to-creatinine ratio 30 mg/g Cr) will be randomized (1:1) to receive either 50 mg ipragliflozin daily or continuation of background treatment (non-SGLT2i). The primary endpoint is the change in RHI from baseline after 24 weeks. To compare the treatment effects between groups, the baseline-adjusted means and their 95% confidence intervals will be estimated by analysis of covariance adjusted for HbA1c (< 7.0% or ≥ 7.0%), age (< 70 y or ≥ 70 y), RHI (< 1.67 or ≥ 1.67), eGFR (< 45 mL/min/1.73 m2 or ≥ 45 mL/min/1.73 m2), and smoking status. Prespecified responder analyses will be also conducted to determine the proportions of patients with clinically meaningful changes in RHI at 24 weeks. DISCUSSION: PROCEED is the first trial to examine the effects of ipragliflozin on endothelial dysfunction in patients with T2D and CKD. This ongoing trial will establish whether endothelial dysfunction is a therapeutic target of SGLT2is in this population. It will also provide deep insights into the potential mechanisms by which SGLT2is reduced the risks of cardiovascular and renal events in recent outcome trials. Trial registration Unique Trial Number, jRCTs071190054 (https://jrct.niph.go.jp/en-latest-detail/jRCTs071190054).


Subject(s)
Diabetes Mellitus, Type 2/drug therapy , Diabetic Angiopathies/drug therapy , Diabetic Nephropathies/drug therapy , Endothelium, Vascular/drug effects , Glucosides/therapeutic use , Renal Insufficiency, Chronic/drug therapy , Sodium-Glucose Transporter 2 Inhibitors/therapeutic use , Thiophenes/therapeutic use , Diabetes Mellitus, Type 2/diagnosis , Diabetes Mellitus, Type 2/epidemiology , Diabetes Mellitus, Type 2/physiopathology , Diabetic Angiopathies/diagnosis , Diabetic Angiopathies/epidemiology , Diabetic Angiopathies/physiopathology , Diabetic Nephropathies/diagnosis , Diabetic Nephropathies/epidemiology , Diabetic Nephropathies/physiopathology , Endothelium, Vascular/physiopathology , Glucosides/adverse effects , Humans , Japan/epidemiology , Multicenter Studies as Topic , Prospective Studies , Randomized Controlled Trials as Topic , Renal Insufficiency, Chronic/diagnosis , Renal Insufficiency, Chronic/epidemiology , Renal Insufficiency, Chronic/physiopathology , Sodium-Glucose Transporter 2 Inhibitors/adverse effects , Thiophenes/adverse effects , Time Factors , Treatment Outcome
7.
Am J Respir Cell Mol Biol ; 60(3): 289-298, 2019 03.
Article in English | MEDLINE | ID: mdl-30326727

ABSTRACT

Chemoattractant receptor homologous with T-helper cell type 2 cells (CRTH2), a receptor for prostaglandin D2, is preferentially expressed on T-helper cell type 2 lymphocytes, group 2 innate lymphoid cells, eosinophils, and basophils, and elicits the production of type 2 cytokines, including profibrotic IL-13. We hypothesized that lack of CRTH2 might protect against fibrotic lung disease, and we tested this hypothesis using a bleomycin-induced lung inflammation and fibrosis model in CRTH2-deficient (CRTH2-/-) or wild-type BALB/c mice. Compared with wild-type mice, CRTH2-/- mice treated with bleomycin exhibited significantly higher mortality, enhanced accumulation of inflammatory cells 14-21 days after bleomycin injection, reduced pulmonary compliance, and increased levels of collagen and total protein in the lungs. These phenotypes were associated with decreased levels of IFN-γ, IL-6, IL-10, and IL-17A in BAL fluid. Adoptive transfer of splenocytes from wild-type, but not CRTH2-/-, mice 2 days before injection of bleomycin resolved the sustained inflammation as well as the increased collagen and protein accumulation in the lungs of CRTH2-/- mice. We consider that the disease model is driven by γδT cells that express CRTH2; thus, the adoptive transfer of γδT cells could ameliorate bleomycin-induced alveolar inflammation and fibrosis.


Subject(s)
Bleomycin/pharmacology , Pneumonia/chemically induced , Pneumonia/metabolism , Pulmonary Fibrosis/chemically induced , Pulmonary Fibrosis/metabolism , Receptors, Immunologic/deficiency , Receptors, Prostaglandin/deficiency , Animals , Basophils/immunology , Basophils/metabolism , Cytokines/immunology , Cytokines/metabolism , Eosinophils/immunology , Eosinophils/metabolism , Immunity, Innate/immunology , Intraepithelial Lymphocytes/immunology , Intraepithelial Lymphocytes/metabolism , Lymphocytes/immunology , Lymphocytes/metabolism , Male , Mice , Mice, Inbred BALB C , Pneumonia/immunology , Pulmonary Fibrosis/immunology , Receptors, Immunologic/immunology , Receptors, Prostaglandin/immunology
8.
Arterioscler Thromb Vasc Biol ; 38(9): 2191-2197, 2018 09.
Article in English | MEDLINE | ID: mdl-30026272

ABSTRACT

Objective- To identify the transcription factors that could contribute to direct reprogramming of fibroblasts toward smooth muscle cell fate. Approach and Results- We screened various combinations of transcription factors, including Myocd (myocardin), Mef2C (myocyte enhancer factor 2C), Mef2B (myocyte enhancer factor 2B), Mkl1 (MKL [megakaryoblastic leukemia]/Myocd-like 1), Gata4 (GATA-binding protein 4), Gata5 (GATA-binding protein 5), Gata6 (GATA-binding protein 6), Ets1 (E26 avian leukemia oncogene 1, 5' domain), and their corresponding carboxyterminal fusions to the transactivation domain of MyoD (myogenic differentiation 1)-indicated by *-for their effects on reprogramming mouse embryonic fibroblasts and human adult dermal fibroblasts to the smooth muscle cell fate as determined by the expression of specific markers. The combination of 3 transcription factors, Myocd (or Myocd*) with Mef2C (or Mef2C*) and Gata6, was the most efficient in enhancing the expression of smooth muscle marker genes and decreasing fibroblast gene expression. Additionally, the derived induced smooth muscle-like cells showed a contractile phenotype in response to carbachol. Conclusions- Combination of Myocd and Gata6 with Mef2C* (MG2*) could sufficiently and efficiently direct differentiation of mouse embryonic and human dermal fibroblasts into induced smooth muscle-like cells, thus opening new opportunities for disease modeling, tissue engineering, and personalized medicine.


Subject(s)
Cellular Reprogramming Techniques/methods , Fibroblasts/cytology , Muscle, Smooth, Vascular/cytology , Myocytes, Smooth Muscle/cytology , Transcription Factors , Animals , Carbachol/pharmacology , Cell Differentiation , Cells, Cultured , Cellular Reprogramming , Embryo, Mammalian , Gene Expression , Humans , Mice, Inbred C57BL , Myocytes, Smooth Muscle/drug effects , Phenotype , Skin/cytology
9.
Stem Cells ; 35(2): 351-361, 2017 02.
Article in English | MEDLINE | ID: mdl-27571517

ABSTRACT

Vascular smooth muscle cells (VSMCs) derived from cardiovascular progenitor cell (CVPC) lineage populate the tunica media of the aortic root. Understanding differentiation of VSMCs from CVPC will further our understanding of the molecular mechanisms contributing to aortic root aneurysms, and thus, facilitate the development of novel therapeutic agents to prevent this devastating complication. It is established that the yes-associated protein (YAP) and Hippo pathway is important for VSMC proliferation and phenotype switch. To determine the role of YAP in differentiation of VSMCs from CVPCs, we utilized the in vitro monolayer lineage specific differentiation method by differentiating human embryonic stem cells into CVPCs, and then, into VSMCs. We found that expression of YAP decreased during differentiation of VSMC from CVPCs. Overexpression of YAP attenuated expression of VSMC contractile markers and impaired VSMC function. Knockdown of YAP increased expression of contractile proteins during CVPC-VSMCs differentiation. Importantly, expression of YAP decreased transcription of myocardin during this process. Overexpression of YAP in PAC1 SMC cell line inhibited luciferase activity of myocardin proximal promoter in a dose dependent and NKX2.5 dependent manners. YAP protein interacted with NKX2.5 protein and inhibited binding of NKX2.5 to the 5'-proximal promoter region of myocardin in CVPC-derived VSMCs. In conclusion, YAP negatively regulates differentiation of VSMCs from CVPCs by decreasing transcription of myocardin in a NKX2.5-dependent manner. Stem Cells 2017;35:351-361.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Cell Differentiation/genetics , Cell Lineage , Myocytes, Cardiac/cytology , Myocytes, Smooth Muscle/cytology , Nuclear Proteins/genetics , Phosphoproteins/metabolism , Stem Cells/cytology , Trans-Activators/genetics , Transcription, Genetic , Biomarkers/metabolism , Cell Line , Gene Knockdown Techniques , Homeobox Protein Nkx-2.5/metabolism , Humans , Models, Biological , Muscle, Smooth, Vascular/cytology , Nuclear Proteins/metabolism , Promoter Regions, Genetic , Protein Binding , Trans-Activators/metabolism , Transcription Factors , YAP-Signaling Proteins
10.
Allergol Int ; 67(2): 234-242, 2018 Apr.
Article in English | MEDLINE | ID: mdl-28919488

ABSTRACT

BACKGROUND: Several methods have been developed to detect allergen-specific IgE in sera. The passive IgE sensitization assay using human IgE receptor-expressing rat cell line RBL-2H3 is a powerful tool to detect biologically active allergen-specific IgE in serum samples. However, one disadvantage is that RBL-2H3 cells are vulnerable to high concentrations of human sera. Only a few human cultured cell lines are easily applicable to the passive IgE sensitization assay. However, the use of human induced pluripotent stem cells (iPSCs) to generate human mast cells (MCs) has not yet been reported. METHODS: The nuclear factor-kappa B (NF-κB)-responsive luciferase reporter gene was stably introduced into a human iPSC line 201B7, and the transfectants were induced to differentiate into MCs (iPSC-MCs). The iPSC-MCs were sensitized overnight with sera from subjects who were allergic to cedar pollen, ragweed pollen, mites, or house dust, and then stimulated with an extract of corresponding allergens. Activation of iPSC-MCs was evaluated by ß-hexosaminidase release, histamine release, or luciferase intensity. RESULTS: iPSCs-MCs stably expressed high-affinity IgE receptor and functionally responded to various allergens when sensitized with human sera from relevant allergic subjects. This passive IgE sensitization system, which we termed the induced mast cell activation test (iMAT), worked well even with undiluted human sera. CONCLUSIONS: iMAT may serve as a novel determining system for IgE/allergens in the clinical and research settings.


Subject(s)
Basophil Degranulation Test/methods , Hypersensitivity/diagnosis , Induced Pluripotent Stem Cells/cytology , Mast Cells/cytology , Mast Cells/immunology , Adult , Allergens , Cell Culture Techniques , Cell Differentiation , Cells, Cultured , Female , Humans , Male , Middle Aged
11.
Development ; 141(18): 3561-71, 2014 Sep.
Article in English | MEDLINE | ID: mdl-25142465

ABSTRACT

Through intercellular signalling, the somatic compartment of the foetal testis is able to program primordial germ cells to undergo spermatogenesis. Fibroblast growth factor 9 and several members of the transforming growth factor ß superfamily are involved in this process in the foetal testis, counteracting the induction of meiosis by retinoic acid and activating germinal mitotic arrest. Here, using in vitro and in vivo approaches, we show that prostaglandin D2 (PGD2), which is produced through both L-Pgds and H-Pgds enzymatic activities in the somatic and germ cell compartments of the foetal testis, plays a role in mitotic arrest in male germ cells by activating the expression and nuclear localization of the CDK inhibitor p21(Cip1) and by repressing pluripotency markers. We show that PGD2 acts through its Dp2 receptor, at least in part through direct effects in germ cells, and contributes to the proper differentiation of male germ cells through the upregulation of the master gene Nanos2. Our data identify PGD2 signalling as an early pathway that acts in both paracrine and autocrine manners, and contributes to the differentiation of germ cells in the foetal testis.


Subject(s)
Cell Differentiation/physiology , DNA-Binding Proteins/metabolism , Fetus/physiology , Gene Expression Regulation, Developmental/physiology , Germ Cells/physiology , Prostaglandin D2/metabolism , Testis/metabolism , Transcription Factors/metabolism , Analysis of Variance , Animals , Blotting, Western , Cell Nucleus/metabolism , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Fluorescent Antibody Technique , Germ Cells/metabolism , Male , Mice , Mice, Knockout , RNA-Binding Proteins/metabolism , Real-Time Polymerase Chain Reaction , Testis/growth & development
12.
J Immunol ; 193(12): 5835-40, 2014 Dec 15.
Article in English | MEDLINE | ID: mdl-25362177

ABSTRACT

Although the cyclooxygenase metabolites PGs are known to be involved in the progression of arthritis, the role of PGD2 remains unclear. In this study, we evaluated the contribution of signaling mediated through a PGD2 receptor, chemoattractant receptor-homologous molecule expressed on Th2 cells (CRTH2), in the progression of adjuvant-induced joint inflammation. Injection of CFA into the ankle joint stimulated PGD2 production and induced paw swelling in both CRTH2-naive (WT) and CRTH2(-/-) mice. CRTH2(-/-) mice presented more severe arthritic manifestations than did WT mice. Through bone marrow transplantation experiments between WT and CRTH2(-/-) mice, we showed that CRTH2 deficiency in bone marrow-derived immune cells is involved in disease progression. Morphological studies showed that CRTH2 deficiency accelerated the infiltration of macrophages into the inflamed paw. Consistent with this finding, we observed that treatment with the macrophage inactivator GdCl3 or the macrophage-depleting agent liposomal clodronate improved arthritis symptoms in CRTH2(-/-) mice. Adoptive transfer of CRTH2(-/-) macrophages exacerbated joint inflammation in WT mice. In addition, CRTH2 deficiency accelerated, whereas CRTH2 agonism inhibited, the expression of a macrophage-activating cytokine (GM-CSF) and a chemokine receptor (CXCR2) in CFA-treated peritoneal macrophages. Together, these observations demonstrate that PGD2-CRTH2 signaling plays a protective role in joint inflammation by attenuating the infiltration of macrophages.


Subject(s)
Arthritis, Experimental/genetics , Arthritis, Experimental/immunology , Receptors, Immunologic/deficiency , Receptors, Prostaglandin/deficiency , Adjuvants, Immunologic/adverse effects , Animals , Arthritis, Experimental/metabolism , Arthritis, Experimental/pathology , Dinoprostone/metabolism , Disease Progression , Macrophage Activation/genetics , Macrophage Activation/immunology , Macrophages/immunology , Macrophages/metabolism , Macrophages/pathology , Male , Mice , Mice, Knockout , Prostaglandin D2/metabolism , Receptors, Immunologic/genetics , Receptors, Immunologic/metabolism , Receptors, Prostaglandin/genetics , Receptors, Prostaglandin/metabolism , Signal Transduction
13.
J Immunol ; 192(1): 459-65, 2014 Jan 01.
Article in English | MEDLINE | ID: mdl-24298012

ABSTRACT

The effects of PGD2 are extremely context dependent. It can have pro- or anti-inflammatory effects in clinically important pathological conditions. A greater mechanistic insight into the determinants of PGD2 activity during inflammation is thus required. In this study, we investigated the role of PGD2 in croton oil-induced dermatitis using transgenic (TG) mice overexpressing hematopoietic PGD synthase. Administration of croton oil caused tissue swelling and vascular leakage in the mouse ear. Compared with wild-type animals, TG mice produced more PGD2 and showed decreased inflammation in the early phase, but more severe manifestations during the late phase. Data obtained from bone marrow transplantation between wild-type and TG mice indicated that PGD2 produced by tissue resident cells in the TG mice attenuated early-phase inflammation, whereas PGD2 produced from hematopoietic lineage cells exacerbated late-phase inflammation. There are two distinct PGD2 receptors: D-prostanoid receptor (DP) and chemoattractant receptor-homologous molecule expressed on Th2 cells (CRTH2). In TG mice, treatment with a DP antagonist exacerbated inflammation in the early phase, whereas treatment with a CRTH2 antagonist attenuated inflammation during the late phase. In vitro experiments showed that DP agonism enhanced vascular endothelial barrier formation, whereas CRTH2 agonism stimulated neutrophil migration. Collectively, these results show that when hematopoietic PGD synthase is overexpressed, tissue resident cell-derived PGD2 suppresses skin inflammation via DP in the early phase, but hematopoietic lineage cell-derived PGD2 stimulates CRTH2 and promotes inflammation during the late phase. DP-mediated vascular barrier enhancement or CRTH2-mediated neutrophil activation may be responsible for these effects. Thus, PGD2 represents opposite roles in inflammation, depending on the disease phase in vivo.


Subject(s)
Dermatitis/immunology , Dermatitis/metabolism , Immunologic Factors/metabolism , Prostaglandin D2/metabolism , Animals , Capillary Permeability/drug effects , Chemotaxis, Leukocyte/drug effects , Chemotaxis, Leukocyte/immunology , Dermatitis/genetics , Disease Models, Animal , Disease Progression , Gene Expression , Immunologic Factors/pharmacology , Intramolecular Oxidoreductases/genetics , Intramolecular Oxidoreductases/metabolism , Leukocytes/immunology , Leukocytes/metabolism , Lipocalins/genetics , Lipocalins/metabolism , Mice , Neutrophils/drug effects , Neutrophils/immunology , Prostaglandin D2/pharmacology , Receptors, Immunologic/metabolism , Receptors, Prostaglandin/metabolism , Signal Transduction
14.
Proc Natl Acad Sci U S A ; 110(13): 5205-10, 2013 Mar 26.
Article in English | MEDLINE | ID: mdl-23479612

ABSTRACT

We investigated the role of prostaglandin D2 (PGD2) signaling in acute lung injury (ALI), focusing on its producer-effector interaction in vivo. Administration of endotoxin increased edema and neutrophil infiltration in the WT mouse lung. Gene disruption of hematopoietic PGD synthase (H-PGDS) aggravated all of the symptoms. Experiments involving bone marrow transplantation between WT and H-PGDS-deficient mice showed that PGD2 derived from alveolar nonhematopoietic lineage cells (i.e., endothelial cells and epithelial cells) promotes vascular barrier function during the early phase (day 1), whereas neutrophil-derived PGD2 attenuates its own infiltration and cytokine expression during the later phase (day 3) of ALI. Treatment with either an agonist to the PGD2 receptor, DP, or a degradation product of PGD2, 15-deoxy-Δ(12,14)-PGJ2, exerted a therapeutic action against ALI. Data obtained from bone marrow transplantation between WT and DP-deficient mice suggest that the DP signal in alveolar endothelial cells is crucial for the anti-inflammatory reactions of PGD2. In vitro, DP agonism directly enhanced endothelial barrier formation, and 15-deoxy-Δ(12,14)-PGJ2 attenuated both neutrophil migration and cytokine expression. These observations indicate that the PGD2 signaling between alveolar endothelial/epithelial cells and infiltrating neutrophils provides anti-inflammatory effects in ALI, and suggest the therapeutic potential of these signaling enhancements.


Subject(s)
Acute Lung Injury/metabolism , Endothelial Cells/metabolism , Prostaglandin D2/metabolism , Pulmonary Alveoli/metabolism , Receptors, Immunologic/isolation & purification , Receptors, Prostaglandin/isolation & purification , Acute Disease , Acute Lung Injury/drug therapy , Acute Lung Injury/genetics , Acute Lung Injury/pathology , Animals , Bone Marrow Transplantation , Endothelial Cells/pathology , Epithelial Cells/metabolism , Epithelial Cells/pathology , Female , Intramolecular Oxidoreductases/genetics , Intramolecular Oxidoreductases/metabolism , Lipocalins/genetics , Lipocalins/metabolism , Mice , Mice, Knockout , Neutrophil Infiltration/drug effects , Neutrophils/metabolism , Neutrophils/pathology , Pneumonia/drug therapy , Pneumonia/genetics , Pneumonia/metabolism , Pneumonia/pathology , Prostaglandin D2/genetics , Pulmonary Alveoli/pathology , Receptors, Immunologic/antagonists & inhibitors , Receptors, Immunologic/genetics , Receptors, Prostaglandin/antagonists & inhibitors , Receptors, Prostaglandin/genetics , Signal Transduction/drug effects , Signal Transduction/genetics , Time Factors , Transplantation, Homologous
15.
J Neurosci ; 34(7): 2514-23, 2014 Feb 12.
Article in English | MEDLINE | ID: mdl-24523542

ABSTRACT

Chemoattractant receptor-homologous molecule expressed on T helper type 2 cells (CRTH2) is a second prostaglandin D2 receptor involved in mediating the allergic response; however, its central function is not yet known. Here, we demonstrate that central CRTH2 mediates emotional impairment. Lipopolysaccharide (LPS)-induced decreases in social interaction and novel exploratory behavior were observed in wild-type (CRTH2(+/+)) mice but not CRTH2-deficient (CRTH2(-/-)) mice, but both genotypes showed hypolocomotion and anorexia following LPS injection. Tumor (colon 26) inoculation, a more pathologically relevant model, induced decreases in social interaction and novel exploratory behavior in CRTH2(+/+), but not CRTH2(-/-) mice. In addition, the CRTH2 antagonists including clinically available ramatroban reversed impaired social interaction and novel exploratory behavior after either LPS or tumor inoculation in CRTH2(+/+) mice. Finally, LPS-induced c-Fos expression in the hypothalamic paraventricular nucleus (PVN) and central amygdala (CeA) was selectively abolished in CRTH2(-/-) mice. These results show that CRTH2 participates in LPS-induced emotional changes and activation in the PVN and CeA. Our study provides the first evidence that central CRTH2 regulates specific emotional behaviors, and that CRTH2 antagonism has potential as a therapeutic target for behavioral symptoms associated with tumors and infectious diseases.


Subject(s)
Brain/metabolism , Illness Behavior/physiology , Receptors, Immunologic/metabolism , Receptors, Prostaglandin/metabolism , Stress, Psychological/metabolism , Animals , Disease Models, Animal , Immunohistochemistry , Lipopolysaccharides/toxicity , Male , Mice , Mice, Inbred BALB C , Mice, Knockout , Neoplasms, Experimental/psychology , Reverse Transcriptase Polymerase Chain Reaction
16.
Mol Plant Microbe Interact ; 28(6): 648-58, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25625819

ABSTRACT

Flagellin from the rice avirulent N1141 strain of Acidovorax avenae functions as a pathogen-associated molecular pattern (PAMP) and induces PAMP-triggered immunity (PTI) in rice. To study the recognition mechanism of flagellin in rice, we attempted to define one or more regions of the flagellin protein required to activate the PTI response. Based on domain classification, we produced four fragments of N1141 flagellin: N-terminal D0, D1. and D2 domains (ND0-2), N-terminal D2, D3, and C-terminal D2 domains (ND2-CD2), C-terminal D2, D1, and D0 domains (CD2-0), and C-terminal D2 and D1 domains (CD2-1). The C-terminal CD2-1 and CD2-0 fragments induced PTI responses in cultured rice cells. Synthetic flg22, which is sufficient to produce the flagellin response in Arabidopsis, and the N-terminal flagellin fragments containing flg22 region elicited very weak immune responses in rice. OsFLS2, the rice ortholog of AtFLS2, which mediates flg22 recognition, was not involved in CD2-0 or CD2-1 recognition in rice. In addition, CD2-0 triggered resistance to coinfection with pathogenic bacteria. Taken together, these data suggest that rice mainly recognizes flagellin CD2-1 by a receptor distinct from OsFLS2 and that this epitope recognition leads to PTI responses.


Subject(s)
Arabidopsis/immunology , Comamonadaceae/physiology , Flagellin/immunology , Host-Pathogen Interactions , Oryza/immunology , Plant Diseases/immunology , Amino Acid Sequence , Arabidopsis/genetics , Arabidopsis/microbiology , Arabidopsis Proteins/genetics , Arabidopsis Proteins/metabolism , Bacterial Proteins/genetics , Bacterial Proteins/immunology , Bacterial Proteins/metabolism , Cells, Cultured , Comamonadaceae/genetics , Epitopes , Flagellin/genetics , Flagellin/metabolism , Gene Expression Profiling , Gene Expression Regulation, Plant , Glucans/metabolism , Hydrogen Peroxide/metabolism , Oligonucleotide Array Sequence Analysis , Oryza/genetics , Oryza/microbiology , Plant Diseases/microbiology , Plant Immunity , Plant Proteins/genetics , Plant Proteins/metabolism , Plants, Genetically Modified , Protein Kinases/genetics , Protein Kinases/metabolism , Recombinant Proteins
17.
Mol Plant Microbe Interact ; 27(2): 113-24, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24200076

ABSTRACT

Plants sense potential pathogens by recognizing conserved pathogen-associated molecular patterns (PAMPs) that cause PAMP-triggered immunity (PTI). We previously reported that rice recognizes flagellin from the rice-incompatible N1141 strain of Acidovorax avenae and subsequently induces immune responses. Cell extracts isolated from flagellin-deficient N1141 (Δfla1141) still induced PTI responses, suggesting that Δfla1141 possesses an additional PAMP distinct from flagellin. Here, we show that elongation factor Tu (EF-Tu), one of the most abundant bacterial proteins, acts as a PAMP in rice and causes several PTI responses. In Brassicaceae species, EF-Tu and an N-acetylated peptide comprising the first 18 amino acids of the N-terminus, termed elf18, are fully active as inducers of PTI responses. By contrast, elf18 did not cause any immune responses in rice, whereas an EF-Tu middle region comprising Lys176 to Gly225, termed EFa50, is fully active as a PAMP in rice. In the leaves of rice plants, EF-Tu induced H2O2 generation and callose deposition, and also triggered resistance to coinfection with pathogenic bacteria. Taken together, these data demonstrate that rice recognizes EFa50, which is distinct from elf18, and that this epitope induces PTI responses.


Subject(s)
Arabidopsis/immunology , Cell Extracts/pharmacology , Comamonadaceae/metabolism , Oryza/immunology , Peptide Elongation Factor Tu/metabolism , Plant Diseases/immunology , Amino Acid Sequence , Arabidopsis/genetics , Arabidopsis/microbiology , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Bacterial Proteins/pharmacology , Cell Extracts/isolation & purification , Epitopes/immunology , Flagellin/genetics , Flagellin/metabolism , Gene Expression Profiling , Gene Expression Regulation, Plant , Glucans/metabolism , Hydrogen Peroxide/metabolism , Molecular Sequence Data , Oligonucleotide Array Sequence Analysis , Oryza/genetics , Oryza/microbiology , Peptide Elongation Factor Tu/genetics , Peptide Elongation Factor Tu/pharmacology , Plant Diseases/microbiology , Plant Immunity , Plant Leaves/genetics , Plant Leaves/immunology , Plant Leaves/microbiology , Plant Proteins/genetics , Recombinant Proteins , Sequence Deletion
18.
Cancer Sci ; 105(11): 1472-9, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25230301

ABSTRACT

Basophil activation was observed in patients with a history of carboplatin-induced severe hypersensitivity reaction (HR). However, the precise mechanism by which carboplatin induces basophil activation and the associated surrogate markers remains to be elucidated. To investigate whether IgE-dependent mechanisms, including the overexpression of FcεRI, participate in carboplatin-induced basophil activation, 13 ovarian cancer patients were enrolled: 5 with a history of carboplatin-induced severe hypersensitivity reaction within the past 2 years, and 8 with no such history. The expression levels of FcεRI, IgE, and CD203c on basophils were measured using a flow cytometer. Immunoglobulin E-dependent basophil activation was evaluated by testing for IgE passive sensitization using lactic acid, and by testing for phosphatidylinositol 3-kinase inhibition, using wortmannin. In three patients positive for carboplatin hypersensitivity, pretreatment with wortmannin almost completely inhibited carboplatin-induced basophil activation (P < 0.05). In a healthy control subject, whose own IgE showed no response to carboplatin, acquired reactivity to carboplatin when exposed to plasma from patients positive for carboplatin hypersensitivity. This did not occur when the same experiment was carried out using plasma from the patients negative for carboplatin hypersensitivity. Moreover, pretreatment with omalizumab, a monoclonal anti-IgE antibody, almost completely blocked carboplatin-induced basophil activation in the plasma of patients positive for carboplatin hypersensitivity. On further investigation, the HR-positive group had significantly higher levels of FcεRI compared with the negative group (P < 0.05). In conclusion, an IgE-dependent mechanism incorporating FcεRI overexpression participates in carboplatin-induced severe HR. These results establish the relevance of monitoring the pharmacodynamic changes of basophils to prevent carboplatin-induced severe HR.


Subject(s)
Antineoplastic Agents/adverse effects , Basophils/immunology , Carboplatin/adverse effects , Drug Hypersensitivity/immunology , Drug Hypersensitivity/metabolism , Immunoglobulin E/immunology , Receptors, IgE/metabolism , Aged , Androstadienes/pharmacology , Antineoplastic Agents/therapeutic use , Basophils/drug effects , Basophils/metabolism , Carboplatin/therapeutic use , Drug Hypersensitivity/genetics , Female , Gene Expression , Humans , Immunization , Immunoglobulin E/metabolism , Immunophenotyping , Middle Aged , Ovarian Neoplasms/complications , Ovarian Neoplasms/drug therapy , Phosphoinositide-3 Kinase Inhibitors , Phosphoric Diester Hydrolases/metabolism , Pyrophosphatases/metabolism , Receptors, IgE/genetics , Wortmannin
19.
J Immunol ; 188(11): 5655-64, 2012 Jun 01.
Article in English | MEDLINE | ID: mdl-22544936

ABSTRACT

Although arachidonic acid cascade has been shown to be involved in sepsis, little is known about the role of PGD(2) and its newly found receptor, chemoattractant receptor-homologous molecule expressed on Th2 cells (CRTH2), on the septic response. Severe sepsis is associated with the failure of neutrophil migration. To investigate whether CRTH2 influences neutrophil recruitment and the lethality during sepsis, sepsis was induced by cecal ligation and puncture (CLP) surgery in mice. CRTH2 knockout (CRTH2(-/-)) mice were highly resistant to CLP-induced sepsis, which was associated with lower bacterial load and lower production of TNF-α, IL-6, and CCL3. IL-10, an anti-inflammatory cytokine, was higher in CRTH2(-/-) mice, blunting CLP-induced lethality in CRTH2(-/-) mice. Neutrophil accumulation in the peritoneum was more pronounced after CLP in CRTH2(-/-) mice, which was associated with higher CXCR2 levels in circulating neutrophils. Furthermore, sepsis caused a decrease in the level of acetylation of histone H3, an activation mark, at the CXCR2 promoter in wild-type neutrophils, suggesting that CXCR2 expression levels are epigenetically regulated. Finally, both pharmacological depletion of neutrophils and inhibition of CXCR2 abrogated the survival benefit in CRTH2(-/-) mice. These results demonstrate that genetic ablation of CRTH2 improved impaired neutrophil migration and survival during severe sepsis, which was mechanistically associated with epigenetic-mediated CXCR2 expression. Thus, CRTH2 is a potential therapeutic target for polymicrobial sepsis.


Subject(s)
Cell Movement/immunology , Neutrophils/immunology , Receptors, Immunologic/physiology , Receptors, Prostaglandin/physiology , Sepsis/immunology , Animals , Bacterial Load/immunology , Cecum/surgery , Cell Movement/genetics , Cell Survival/genetics , Cell Survival/immunology , Cytokines/physiology , Disease Models, Animal , Disease Resistance/genetics , Disease Resistance/immunology , Female , Inflammation Mediators/physiology , Ligation , Mice , Mice, Inbred BALB C , Mice, Knockout , Punctures , Receptors, Immunologic/deficiency , Receptors, Prostaglandin/deficiency , Sepsis/microbiology , Sepsis/prevention & control
20.
Clin Case Rep ; 12(3): e8652, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38464587

ABSTRACT

An elderly patient with upper back pain and hypertension was diagnosed as having spontaneous spinal epidural hematoma (SSEH) after excluding artery dissection. The initial symptoms of SSEH mimic those of artery dissection, and the symptoms of spinal damage frequently appear later. Physicians should, therefore, be mindful of SSEH.

SELECTION OF CITATIONS
SEARCH DETAIL