Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters










Publication year range
1.
Cell Rep Methods ; 3(1): 100370, 2023 01 23.
Article in English | MEDLINE | ID: mdl-36814843

ABSTRACT

Islet transplantation is an effective treatment for type 1 diabetes (T1D). However, a shortage of donors and the need for immunosuppressants are major issues. The ideal solution is to develop a source of insulin-secreting cells and an immunoprotective method. No bioartificial pancreas (BAP) devices currently meet all of the functions of long-term glycemic control, islet survival, immunoprotection, discordant xenotransplantation feasibility, and biocompatibility. We developed a device in which porcine islets were encapsulated in a highly stable and permeable hydrogel and a biocompatible immunoisolation membrane. Discordant xenotransplantation of the device into diabetic mice improved glycemic control for more than 200 days. Glycemic control was also improved in new diabetic mice "relay-transplanted" with the device after its retrieval. The easily retrieved devices exhibited almost no adhesion or fibrosis and showed sustained insulin secretion even after the two xenotransplantations. This device has the potential to be a useful BAP for T1D.


Subject(s)
Diabetes Mellitus, Experimental , Diabetes Mellitus, Type 1 , Islets of Langerhans Transplantation , Animals , Mice , Swine , Diabetes Mellitus, Type 1/surgery , Transplantation, Heterologous , Diabetes Mellitus, Experimental/surgery , Pancreas
2.
Eur J Pharmacol ; 811: 110-116, 2017 Sep 15.
Article in English | MEDLINE | ID: mdl-28587776

ABSTRACT

Recently, we identified a novel phosphodiesterase 2A (PDE2A) inhibitor, PDM-631 ((S)-3-cyclopropyl-6-methyl-1-(1-(4-(trifluoromethoxy)phenyl)propan-2-yl)-1,5-dihydro-4H-pyrazolo[3,4-d]pyrimidin-4-one). PDM-631 showed potent inhibitory activities for human and rat PDE2A with IC50 values of 1.5 and 4.2nM, respectively and more than 2000-fold selectivity against other phosphodiesterases. In rat studies, PDM-631 showed oral bioavailability and good brain penetration, and increased the cGMP levels in the cortex. These data indicate that PDM-631 is a potent, selective, orally active, and brain-penetrable PDE2A inhibitor. In behavioral studies using rat models, PDM-631 (3-30mg/kg) resulted in better discrimination between a novel object and a familiar one 48h after the acquisition phase in the novel object recognition test, thus indicating that PDM-631 increased object recognition memory. In contrast, PDM-631 did not attenuate the conditioned avoidance response at the same dose range (3-30mg/kg) in rats, indicating that PDM-631 did not show an antipsychotic-like effect. In test for extrapyramidal side effect, PDM-631 had no effect on catalepsy at the effective doses (10 and 30mg/kg) in the novel object recognition test, while haloperidol caused catalepsy at a dose of 3mg/kg. Our results suggest that PDM-631 is a good pharmacological tool that can be used to investigate the role of PDE2A and may have therapeutic potential for the treatment of cognitive impairments associated with schizophrenia and neurodegenerative disorders, without any extrapyramidal side effects.


Subject(s)
Cyclic Nucleotide Phosphodiesterases, Type 2/antagonists & inhibitors , Pyrazoles/administration & dosage , Pyrazoles/pharmacology , Administration, Oral , Animals , Avoidance Learning/drug effects , Conditioning, Psychological/drug effects , Cyclic GMP/metabolism , Cyclic Nucleotide Phosphodiesterases, Type 2/metabolism , Dose-Response Relationship, Drug , Drug Design , Male , Pyrazoles/pharmacokinetics , Pyrimidines , Rats , Rats, Sprague-Dawley , Recognition, Psychology/drug effects
4.
Eur J Pharmacol ; 802: 60-68, 2017 May 05.
Article in English | MEDLINE | ID: mdl-28249709

ABSTRACT

Severe sepsis is a complex, multifactorial, and rapidly progressing disease characterized by excessive inflammation and coagulation following bacterial infection. To simultaneously suppress pro-inflammatory and pro-coagulant responses, we genetically engineered a novel fusion protein (MR1007) consisting of an anti-CD14 antibody and the modified second domain of bikunin, and evaluated the potential of MR1007 as an anti-sepsis agent. Suppressive effects of MR1007 on lipopolysaccharide (LPS)-induced inflammatory responses were assessed using peripheral blood mononuclear cells or endothelial cells. Its inhibitory activity against the coagulation factor XIa was assessed using a purified enzyme and a chromogenic substrate. Anticoagulant activity was assessed using human or rabbit plasma. Anti-inflammatory and anti-coagulant effects and/or survival benefits were evaluated in an endotoxemia model and a cecal ligation and puncture model. MR1007 inhibited LPS-induced cytokine production in peripheral blood mononuclear cells and endothelial cells, inhibited factor XIa, and exhibited anticoagulant activity. In an endotoxemia model, 0.3-3mg/kg MR1007 suppressed pro-inflammatory and pro-coagulant responses in a dose-dependent manner; at a dose of 3mg/kg, the protein improved survival even when administered 8h after the LPS injection. In addition, 10mg/kg MR1007 administered 2h post cecal ligation and puncture improved survival. However, MR1007 administered at doses up to 30mg/kg did not increase ear bleeding time or bacterial counts in the cecal ligation and puncture model. Thus, simultaneous targeting of CD14 and factor XIa improves survival in the rabbit endotoxemia and sepsis models and represents a promising approach for the treatment of severe sepsis.


Subject(s)
Alpha-Globulins/chemistry , Antibodies, Monoclonal/pharmacology , Endotoxemia/drug therapy , Factor XIa/antagonists & inhibitors , Lipopolysaccharide Receptors/immunology , Molecular Targeted Therapy , Recombinant Fusion Proteins/pharmacology , Animals , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/therapeutic use , Anticoagulants/immunology , Anticoagulants/pharmacology , Anticoagulants/therapeutic use , Bleeding Time , Disease Models, Animal , E-Selectin/metabolism , Endotoxemia/metabolism , Gene Expression Regulation/drug effects , Humans , Interleukin-6/biosynthesis , Protein Domains , Rabbits , Recombinant Fusion Proteins/immunology , Recombinant Fusion Proteins/therapeutic use , Survival Analysis
5.
Eur J Pharmacol ; 802: 76-84, 2017 May 05.
Article in English | MEDLINE | ID: mdl-28246026

ABSTRACT

The transcription factor NF-E2-related factor 2 (Nrf2) is a key regulator of cellular defense mechanisms against oxidative stress. Multiple sclerosis (MS), a chronic inflammatory disease of the central nervous system, is characterized by progressive demyelination and neurodegeneration induced by inflammation and oxidative stress. The induction of Nrf2 signaling has been shown to inhibit disease development and progression in the experimental autoimmune encephalomyelitis (EAE) model of MS in mice. In the present study, we performed a high-throughput screening assay using a chimeric construct of the N-terminal portion of Nrf2 fused to LacZ. Using this approach, we identified the novel Nrf2 inducer TFM-735. Using human primary cell profiling systems, we found that TFM-735 inhibited T cell proliferation and exerted immuno-modulatory effects by inhibiting the production of IL-6 and IL-17. TFM-735 also inhibited IL-17 secretion from human peripheral blood mononuclear cells stimulated with anti-CD3 and anti-CD28. In EAE mice treated with TFM-735, the expression of the Nrf2 target gene Nqo1 increased in the brain and spleen, disease severity was ameliorated, and plasma IL-17 levels decreased. Furthermore, TFM-735 inhibited luciferase activity in Wim-6 transgenic EAE mice expressing the human interleukin 6-luciferase (hIL6-BAC-Luc) reporter. Therefore, these findings indicate that TFM-735 is a potent Nrf2 inducer that inhibits inflammatory cytokine production and disease progression in mice with EAE and that TFM-735 is a promising therapeutic agent for MS.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental/drug therapy , Encephalomyelitis, Autoimmune, Experimental/metabolism , NF-E2-Related Factor 2/metabolism , Pyrazoles/pharmacology , Thiazoles/pharmacology , Animals , HEK293 Cells , Humans , Interleukin-17/metabolism , Interleukin-6/genetics , Mice , Mice, Inbred C57BL , Pyrazoles/therapeutic use , Thiazoles/therapeutic use
6.
Eur J Pharmacol ; 806: 18-24, 2017 Jul 05.
Article in English | MEDLINE | ID: mdl-28322834

ABSTRACT

Cluster of differentiation 14 (CD14), a pattern recognition receptor expressed on myeloid cells and a critical component of the innate immune system, mediates local and systemic host responses to gram-negative bacterial products, including lipopolysaccharide (LPS). Therefore, CD14 is an attractive target for development of sepsis therapies, and several monoclonal anti-CD14 antibodies have been reported. In this study, we prepared an anti-human CD14 monoclonal antibody, F1024-1-3, which suppressed LPS-induced upregulation of pro-inflammatory cytokines and an adhesion molecule in human peripheral mononuclear cells and human vascular endothelial cells. Half-maximal inhibitory concentrations in these assays ranged from 0.1 to 1µg/ml. In rabbits, intravenous administration (3mg/kg) as well as in vitro exposure of F1024-1-3 suppressed LPS-induced cytokine production in whole blood. In endotoxemia models generated by three sequential injections of LPS, intravenous administration of F1024-1-3 at 0.3-3mg/kg sharply reduced pro-inflammatory responses in a dose-dependent manner and moderately attenuated pro-coagulant responses; at 1mg/kg, the protein protected rabbits from lethality even when administered 2h after the initial LPS injection. However, F1024-1-3 (10mg/kg) given 2h post-surgery did not prevent death of rabbits in a cecal ligation and puncture model. Thus, suppression of CD14-mediated activation of leukocytes and endothelial cells alone may not be clinically efficacious for the treatment of severe sepsis and septic shock.


Subject(s)
Antibodies, Monoclonal/immunology , Endotoxins/toxicity , Lipopolysaccharide Receptors/immunology , Sepsis/immunology , Animals , Blood Coagulation/drug effects , Coinfection/drug therapy , Coinfection/microbiology , Epitopes/immunology , Humans , Interleukin-6/biosynthesis , Leukocytes/drug effects , Leukocytes/immunology , Leukocytes/metabolism , Male , Rabbits , Rats , Sepsis/chemically induced , Sepsis/metabolism , Sepsis/physiopathology , Survival Analysis
7.
J Immunol Methods ; 418: 52-60, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25655782

ABSTRACT

Soluble Glycoprotein VI (GPVI) is an attractive marker for disorders marked by platelet activation, such as thrombotic microangiopathy, myocardial infarction, and stroke. Several groups have already developed an immunoassay for soluble GPVI; however, there are several discrepancies between the groups' assays. In this study, we prepared the two types of recombinant soluble GPVI, the monomeric form GPVI (GPVI-His) and the dimeric form of GPVI (GPVI-Fc), moreover, we generated four anti-GPVI antibodies, F1232-7-1 (7S1), F1232-10-2 (10S2), F1232-19-1 (19D1), and F1232-21-1 (21D1). The former 2 antibodies (7S1 and 10S2) had a high affinity for both GPVI-His and GPVI-Fc, while the latter 2 antibodies (19D1 and 21D1) showed a high affinity for GPVI-Fc but low affinity for GPVI-His. All of the antibodies comparably recognized surface GPVI on resting platelets. Furthermore, we established two immunoassays for soluble GPVI, 7S1/10S2-HRP and 19D1/21D1-HRP (capture antibody/detection antibody). 7S1/10S2-HRP showed equivalent reactivity with GPVI-His and GPVI-Fc, whereas 19D1/21D1-HRP had high affinity for GPVI-Fc but low reactivity with GPVI-His. In terms of reactivity with platelet-derived soluble GPVI, 7S1/10S2-HRP demonstrated sensitive detection whereas 19D1/21D1-HRP was nonreactive. Taken together, 7S1/10S2-HRP is a suitable candidate for a reliable soluble GPVI immunoassay as it has a high affinity for monomeric GPVI.


Subject(s)
Blood Platelets/immunology , Immunoassay , Platelet Membrane Glycoproteins/analysis , Platelet Membrane Glycoproteins/immunology , Antibodies/immunology , Biomarkers/analysis , Enzyme-Linked Immunosorbent Assay , Humans , Recombinant Proteins/immunology
8.
Platelets ; 26(8): 745-50, 2015.
Article in English | MEDLINE | ID: mdl-25549161

ABSTRACT

Glycoprotein VI (GPVI) plays a critical role in the platelet response to collagen. Clinical studies suggest that the plasma level of soluble GPVI (sGPVI) is a highly specific and useful platelet activation marker. However, many properties of sGPVI have not been fully characterized, such as its sensitivity in detecting platelet activation and its elimination rate from the blood. In this study we established a sandwich enzyme-linked immunosorbent assay for human sGPVI, which cross-reacts to cynomolgus monkey sGPVI, and evaluated the time course of sGPVI production in a cynomolgus monkey model of lipopolysaccharide (LPS)-induced thrombocytopenia. The sGPVI levels in this model were dramatically elevated and returned to baseline by 24 hours after LPS injection, the change was more pronounced than the existing platelet activation biomarker, soluble P-selectin (sP-selectin) levels. The elimination half-life of recombinant human sGPVI was about 2.5 hours following intravenous administration to monkeys. These results suggest that plasma sGPVI closely reflects platelet activation in the bloodstream and has a short half-life. sGPVI would be a useful biomarker for disorders marked by platelet activation and for monitoring anti-platelet therapy.


Subject(s)
Blood Platelets/metabolism , Platelet Activation , Platelet Membrane Glycoproteins/metabolism , Amino Acid Sequence , Animals , Biomarkers , Disease Models, Animal , Enzyme-Linked Immunosorbent Assay , Humans , Macaca fascicularis , Molecular Sequence Data , P-Selectin/blood , Platelet Membrane Glycoproteins/chemistry , Platelet Membrane Glycoproteins/pharmacokinetics , Recombinant Proteins/administration & dosage , Recombinant Proteins/pharmacokinetics , Reproducibility of Results , Sequence Alignment , Thrombocytopenia/blood , Thrombocytopenia/metabolism , Time Factors
9.
PLoS One ; 9(11): e112027, 2014.
Article in English | MEDLINE | ID: mdl-25405858

ABSTRACT

OBJECTIVE: Diacylglycerol O-acyltransferase 1 (DGAT1) catalyzes the final committed step in triglyceride biosynthesis. DGAT1 null mice are known to be resistant to diet-induced obesity, and more insulin sensitive relative to the wild-type; however, the mice exhibit abnormalities in the skin. This work determined whether the intestine-targeted DGAT1 inhibitor could improve obesity and insulin resistance without skin aberrations in mice. DESIGN AND METHODS: We synthesized 2 DGAT1 inhibitors: Compound A, described in the patent application from the Japan Tobacco, and Compound B (A-922500), reported by Abbott Laboratories. Both compounds were evaluated for inhibitory activities against DGAT1 enzymes and effects on the skin in mice in vivo. Compound B was further investigated for effects on obesity and insulin resistance in diet-induced-obese (DIO) mice. RESULTS: The 2 compounds comparably inhibited the DGAT1 enzyme activity and the cellular triglyceride synthesis in vitro, while they showed different distribution patterns in mice in vivo. Compound A, which distributed systemically, caused skin aberrations, while Compound B, which preferentially distributed to the intestine, improved obesity and insulin resistance without skin aberrations in DIO mice. CONCLUSIONS: Our results suggest that the intestine is the key tissue in which DGAT1 plays a role in promoting obesity and insulin resistance.


Subject(s)
Biphenyl Compounds/therapeutic use , Diacylglycerol O-Acyltransferase/antagonists & inhibitors , Enzyme Inhibitors/therapeutic use , Insulin Resistance , Intestines/drug effects , Obesity/drug therapy , Phenylurea Compounds/therapeutic use , Animals , Biphenyl Compounds/adverse effects , Biphenyl Compounds/chemical synthesis , Biphenyl Compounds/pharmacokinetics , Diacylglycerol O-Acyltransferase/metabolism , Enzyme Inhibitors/adverse effects , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/pharmacokinetics , HT29 Cells , Hep G2 Cells , Humans , Intestinal Mucosa/metabolism , Male , Mice , Mice, Inbred C57BL , Phenylurea Compounds/adverse effects , Phenylurea Compounds/chemical synthesis , Phenylurea Compounds/pharmacokinetics , Skin/drug effects , Tissue Distribution
10.
Genes Cells ; 18(10): 899-908, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23890231

ABSTRACT

Nrf2 is a transcription factor that regulates the antioxidant and detoxification enzyme genes and provides defense against oxidative and electrophilic stresses in various tissues. In brain, while neuroprotective functions of Nrf2 have been well documented, Nrf2 contribution to the brain function remains to be elucidated. To address this issue, we investigated whether Nrf2 deficiency affects psychological behaviors, neurotransmitter systems and gene expressions in mice. We conducted four behavioral tests, social interaction, open-field, rotarod and forced swimming tests and found that Nrf2 knockout mice exhibited reduced immobility in the forced swimming test. Neurochemical analyses revealed that the dopamine and serotonin metabolites increased in the brains of Nrf2 knockout mice. We also present a catalog of genes whose expression is Nrf2-dependent in brain under unstressed conditions, which includes a number of xenobiotic-metabolizing enzyme genes. These results thus support our contention that Nrf2 regulates its target genes in brain under unstressed conditions and loss of Nrf2 affects various brain functions.


Subject(s)
Behavior, Animal , Brain/metabolism , NF-E2-Related Factor 2/genetics , NF-E2-Related Factor 2/metabolism , Amino Acids/metabolism , Animals , Anxiety , Dopamine Agents/metabolism , Locomotion , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Motor Activity , Oxidative Stress , Phenotype , Rotarod Performance Test , Serine/metabolism , Serotonin Agents/metabolism , Social Behavior
12.
J Clin Invest ; 118(5): 1785-95, 2008 May.
Article in English | MEDLINE | ID: mdl-18382762

ABSTRACT

Platelet adhesion to vascular subendothelium, mediated in part by interactions between collagen and glycoprotein VI (GPVI) complexed with Fc receptor gamma-chain, is crucial for thrombus formation. Antiplatelet therapy benefits patients with various thrombotic and ischemic diseases, but the safety and efficacy of existing treatments are limited. Recent data suggest GPVI as a promising target for a novel antiplatelet therapy, for example, GPVI-specific Abs that deplete GPVI from the surface of platelets. Here, we characterized GPVI-specific auto-Abs (YA-Abs) from the first reported patient with ongoing platelet GPVI deficiency caused by the YA-Abs. To obtain experimentally useful human GPVI-specific mAbs with characteristics similar to YA-Abs, we generated human GPVI-specific mouse mAbs and selected 2 representative mAbs, mF1201 and mF1232, whose binding to GPVI was inhibited by YA-Abs. In vitro, mF1201, but not mF1232, induced human platelet activation and GPVI shedding, and mF1232 inhibited collagen-induced human platelet aggregation. Administration of mF1201 and mF1232 to monkeys caused GPVI immunodepletion with and without both significant thrombocytopenia and GPVI shedding, respectively. When a human/mouse chimeric form of mF1232 (cF1232) was labeled with a fluorescent endocytosis probe and administered to monkeys, fluorescence increased in circulating platelets and surface GPVI was lost. Loss of platelet surface GPVI mediated by cF1232 was successfully reproduced in vitro in the presence of a cAMP-elevating agent. Thus, we have characterized cAMP-dependent endocytosis of GPVI mediated by a human GPVI-specific mAb as what we believe to be a novel antiplatelet therapy.


Subject(s)
Antibodies, Monoclonal , Blood Platelets/immunology , Cyclic AMP/metabolism , Endocytosis/physiology , Platelet Aggregation Inhibitors , Platelet Membrane Glycoproteins/metabolism , Receptors, IgG/metabolism , Amino Acid Sequence , Animals , Antibodies, Monoclonal/genetics , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/therapeutic use , Autoantibodies/genetics , Autoantibodies/immunology , Female , Humans , Macaca fascicularis , Male , Mice , Molecular Sequence Data , Platelet Adhesiveness/physiology , Platelet Aggregation Inhibitors/immunology , Platelet Aggregation Inhibitors/therapeutic use , Platelet Membrane Glycoproteins/genetics , Receptors, IgG/genetics , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism
13.
Eur J Pharmacol ; 569(3): 237-43, 2007 Aug 27.
Article in English | MEDLINE | ID: mdl-17588563

ABSTRACT

Sepsis occurs when microbes activate toll-like receptors (TLRs) stimulating widespread inflammation and activating coagulation cascades. TLR4 signal transduction has been recognized as a key pathway for lipopolysaccharide (LPS)-induced activation of various cells and an attractive target for treatment of sepsis. We found a new benzisothiazole derivative, M62812 that inhibits TLR4 signal transduction. This compound suppressed LPS-induced upregulation of inflammatory cytokines, adhesion molecules and procoagulant activity in human vascular endothelial cells and peripheral mononuclear cells. The half maximal inhibitory concentrations in these assays ranged from 1 to 3 microg/ml. Single intravenous administration of M62812 (10-20 mg/kg) protected mice from lethality and reduced inflammatory and coagulatory parameters in a murine d-galactosamine-sensitized endotoxin shock model. M62812 (20 mg/kg) also prevented mice from lethality in a murine cecal ligation and puncture model. These results suggest that inhibition of TLR4 signal transduction can suppress coagulation as well as inflammation during sepsis and may be clinically beneficial in sepsis treatment.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Shock, Septic/prevention & control , Signal Transduction/drug effects , Thiazoles/pharmacology , Toll-Like Receptor 4/metabolism , Animals , Anti-Inflammatory Agents/administration & dosage , Anticoagulants/administration & dosage , Anticoagulants/pharmacology , Blood Coagulation/drug effects , Disease Models, Animal , Dose-Response Relationship, Drug , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Endotoxins , Humans , Inflammation Mediators/metabolism , Injections, Intravenous , Leukocytes, Mononuclear/drug effects , Leukocytes, Mononuclear/metabolism , Lipopolysaccharides , Male , Mice , Mice, Inbred BALB C , Thiazoles/administration & dosage , Toll-Like Receptor 4/antagonists & inhibitors , Toll-Like Receptor 4/drug effects , Umbilical Veins , Up-Regulation
14.
J Immunol ; 175(11): 7348-56, 2005 Dec 01.
Article in English | MEDLINE | ID: mdl-16301641

ABSTRACT

An orally active antiallergic agent, M50367, skews the Th1/Th2 balance toward Th1 dominance by suppressing naive Th cell differentiation into Th2 cells in vitro. Administration results in the suppression of IgE synthesis and peritoneal eosinophilia in vivo. In this report, we determined that M50354 (an active metabolite of M50367) was a ligand for the aryl hydrocarbon receptor (AhR); the immunological effects of this compound on in vitro Th1/Th2 differentiation from naive Th cells and Th1/Th2 balance in vivo were manifested through binding to AhR. These effects were completely abolished in AhR-deficient mice. AhR expression in the naive Th cell was significantly up-regulated by costimulation of TCR and CD28. Suppression of naive Th cell differentiation into Th2 cells via binding of M50354 to AhR was associated with inhibition of GATA-3 expression in Th cells. In addition, forced expression of a constitutively active form of AhR or activation of AhR by the addition of representative ligands suppressed naive Th cell differentiation into Th2 cells. Based on these results, we conclude that AhR functions as a modulator of the in vivo Th1/Th2 balance through activation in naive Th cells.


Subject(s)
Receptors, Aryl Hydrocarbon/metabolism , Signal Transduction/immunology , Th1 Cells/immunology , Th2 Cells/immunology , Animals , Anti-Allergic Agents/pharmacology , Benzimidazoles/metabolism , Benzimidazoles/pharmacology , Blotting, Western , Cell Differentiation/immunology , Cytokines/drug effects , Cytokines/immunology , Cytokines/metabolism , Electrophoretic Mobility Shift Assay , Mice , Mice, Mutant Strains , Rats , Receptors, Aryl Hydrocarbon/immunology , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/drug effects , Th1 Cells/cytology , Th1 Cells/drug effects , Th2 Cells/cytology , Th2 Cells/drug effects , Th2 Cells/metabolism , Transfection
15.
J Infect Chemother ; 11(5): 234-8, 2005 Oct.
Article in English | MEDLINE | ID: mdl-16258819

ABSTRACT

CD14, a high-affinity receptor for lipopolysaccharide (LPS), is a glycoprotein expressed on the surface membranes of monocytes/macrophages. We have identified a previously unknown form of soluble CD14, named soluble CD14 subtype (sCD14-ST), that is increased in patients with sepsis. To measure sCD14-ST concentrations in plasma, we prepared anti-sCD14-ST antibodies and developed an enzyme immunoassay (EIA) for this soluble form of CD14. With this assay, quantitative measurements are available within 4 h, and we compared the levels of sCD14-ST in plasma from normal subjects (healthy controls), patients with systemic inflammatory response syndrome (SIRS), and sepsis patients. The level of sCD14-ST in subjects with sepsis was much higher than the levels in subjects with SIRS and the healthy controls. Additionally, when a subject's sCD14-ST level was used as a diagnostic marker for sepsis, the area under the receiver operating characteristic (ROC) curve was 0.817, thereby demonstrating that elevated sCD14-ST levels were a better marker for sepsis than the other molecular markers we tested. sCD14-ST levels also correlated with procalcitonin (PCT) levels and with sequential organ failure assessment (SOFA) scores. Finally, changes in sCD14-ST concentration correlated with the severity of sepsis. Taken together, these results indicate that sCD14-ST is a useful marker for the rapid diagnosis of sepsis and for monitoring the severity of the disease.


Subject(s)
Biomarkers/blood , Lipopolysaccharide Receptors/blood , Sepsis/diagnosis , Aged , Humans , Immunoenzyme Techniques/methods , Male , ROC Curve , Severity of Illness Index , Systemic Inflammatory Response Syndrome/diagnosis
16.
Cell Immunol ; 224(1): 29-37, 2003 Jul.
Article in English | MEDLINE | ID: mdl-14572798

ABSTRACT

We previously reported an orally active anti-allergic agent, M50367, modulated Th1/Th2 balance to down-regulate Th2 response in a murine model of atopic asthma. In this study, we examined the effect of M50354, the active metabolite of M50367, on the differentiation of naïve Th cells into Th1/Th2 cells. M50354 at 3 microM decreased the generation of Th2 cells by 0.2-fold and increased that of Th1 cells by 1.6-fold from naïve Th cells primed with antigenic peptide and antigen-presenting cells. Its effect was also seen when naïve Th cells were primed with anti-T cell receptor and anti-CD28 agonistic antibodies instead of antigen and antigen-presenting cells. M50354 decreased early endogenous IL-4 production in the nai;ve Th cell priming culture without affecting interferon-gamma production and proliferation. In contrast, M50354 had no effect on interferon-gamma and IL-4 production from mature Th1 and Th2 cells. These results suggest that M50354 directly acts on naïve Th cells to suppress their differentiation into Th2 cells.


Subject(s)
Anti-Allergic Agents/pharmacology , Benzimidazoles/pharmacology , Cell Differentiation/drug effects , Th1 Cells/drug effects , Th2 Cells/drug effects , Administration, Oral , Animals , Antibodies/immunology , Antibodies/pharmacology , Antigen-Presenting Cells/immunology , Antigens/immunology , Antigens/pharmacology , Asthma/drug therapy , Asthma/immunology , Benzimidazoles/administration & dosage , Cell Differentiation/immunology , Cell Division/drug effects , Cell Division/immunology , Dose-Response Relationship, Drug , Interferon-gamma/biosynthesis , Interferon-gamma/immunology , Interleukin-4/biosynthesis , Interleukin-4/immunology , Mice , Mice, Inbred BALB C , Mice, Transgenic , Reaction Time/drug effects , Reaction Time/immunology , Th1 Cells/immunology , Th2 Cells/immunology
SELECTION OF CITATIONS
SEARCH DETAIL