Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
J Immunol Methods ; 529: 113682, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38705372

ABSTRACT

BACKGROUND: The measurement of antigen-specific serum IgE is common in clinical assessments of type I allergies. However, the interaction between antigens and IgE won't invariably trigger mast cell activation. We previously developed the IgE crosslinking-induced luciferase expression (EXiLE) method using the RS-ATL8 mast cell line; however, the method may not be sensitive enough in some cases. METHODS: In this study, we introduced an NF-AT-regulated luciferase reporter gene into the RBL-2H3 rat mast cell line and expressed a chimeric high-affinity IgE receptor (FcεRI) α chain gene, comprising an extracellular domain from humans and transmembrane/intracellular domains from rats. RESULTS: We generated multiple clones expressing the chimeric receptor. Based on their responsiveness and proliferation, we selected the HuRa-40 clone. This cell line exhibited significantly elevated human α chain expression compared to RS-ATL8 cells, demonstrating a 10-fold enhancement of antigen-specific reactivity. Reproducibility across different batches and operators was excellent. Moreover, we observed a detectable response inhibition by an anti-allergy drugs (omalizumab and cyclosporin A). CONCLUSIONS: HuRa-40 cells-which carry the human-rat chimeric IgE receptor-comprise a valuable reporter cell line for the EXiLE method. Their versatility extends to various applications and facilitates high-throughput screening of anti-allergy drugs.


Subject(s)
Immunoglobulin E , Luciferases , Mast Cells , Receptors, IgE , Receptors, IgE/metabolism , Receptors, IgE/genetics , Receptors, IgE/immunology , Animals , Humans , Mast Cells/immunology , Mast Cells/metabolism , Rats , Immunoglobulin E/immunology , Luciferases/genetics , Luciferases/metabolism , Cell Line , Genes, Reporter , Reproducibility of Results , Receptors, Chimeric Antigen/genetics , Receptors, Chimeric Antigen/immunology , Receptors, Chimeric Antigen/metabolism
2.
Shokuhin Eiseigaku Zasshi ; 62(6): 193-202, 2021.
Article in Japanese | MEDLINE | ID: mdl-34955470

ABSTRACT

We examined the effects of cooking and processing on the quantitation of soy protein in various soy-based foods. For the phosphate-buffered saline (PBS) extraction, the total protein content was measured by bicinchoninic acid assay, and the buffer extraction containing sodium dodecyl sulfate (SDS) and 2-mercaptoethanol (ME) was measured by the 2-D Quant Kit, and SDS-polyacrylamide gel electrophoresis analysis (SDS-PAGE) of each extraction was performed. Furthermore, measurements were performed by various ELISAs. During the tofu cooking process, the protein concentrations of soaked soybeans and Seigo (soybean homogenized with water) fluctuated- the change in protein solubility due to the amount of water during sample homogenization was considered to be a factor. It was thought that the decrease in protein concentration due to heating of Seigo during soymilk production was considered to indicate thermal denaturation of the protein, and that SDS and ME extraction in tofu may affect the measurement system. In cooking excluding roasted beans, proteins with a mass of 50 kDa or above and around 20 kDa were denatured, and in twice-fried tofu, proteins around 40 kDa were denatured, but the protein concentration excluding boiled soybeans did not decreased. Furthermore, the protein concentration from roasted beans, yuba, roasted okara and fried tofu increased with the cooking time, suggesting that the denaturation temperature of the protein shifted to a high temperature as the water content decreased. Both of the two types of ELISAs that comply with the official labeling system of foods containing allergenic substances were useful for detecting soybean protein by detecting proteins and peptides in processed soybean products, fermented foods excluding natto, and health foods.


Subject(s)
Soy Foods , Soybean Proteins , Allergens/analysis , Cooking , Soy Foods/analysis , Soybean Proteins/analysis , Glycine max
4.
Allergol Int ; 68(3): 342-351, 2019 Jul.
Article in English | MEDLINE | ID: mdl-30803853

ABSTRACT

BACKGROUND: The reported prevalences of IgG autoantibodies (AAbs) to FcεRIα and IgE in sera from patients with chronic spontaneous urticaria (CSU) have varied, and these AAbs are also often observed in healthy control subjects. Regarding the histamine release activity of purified IgG from patients with CSU, the number of examined patients has been small. Thus, we sought to determine the prevalence and FcεRI crosslinking ability of these AAbs in a large number of patients with CSU and non-atopic control (NC) subjects. METHODS: We compared the concentrations of anti-IgE and anti-FcεRIα AAbs and the abilities of these AAbs to cause FcεRI aggregation in patients with CSU (n = 134) and NC subjects (n = 55) using ELISA and an in vitro elicitation test, respectively. RESULTS: The concentration of anti-IgE AAbs was significantly different between the NC subjects and the CSU patients (P < 0.0001, cutoff value: 0.558 µg/mL), whereas the concentration of anti-FcεRIα AAbs was not. A significant difference in the duration of illness was noted between patients with lower and those with higher concentrations of anti-IgE AAbs relative to the cutoff value. The abilities of anti-IgE AAbs, but not anti-FcεRIα AAbs, to induce FcεRI crosslinking were significantly higher in CSU patients than in NC subjects (P = 0.0106). CONCLUSIONS: In the Japanese population of CSU patients studied, the ability of the anti-IgE AAbs to induce FcεRI crosslinking differed significantly between NC subjects and CSU patients, suggesting the involvement of anti-IgE AAbs in the pathogenesis of CSU in the Japanese population.


Subject(s)
Autoantibodies/immunology , Immunoglobulin E/immunology , Receptors, IgE/immunology , Urticaria/immunology , Adolescent , Adult , Aged , Aged, 80 and over , Animals , Autoantibodies/blood , Basophils/immunology , Cells, Cultured , Chronic Disease , Female , Histamine Release , Humans , Immunoglobulin G/blood , Immunologic Capping , Male , Mast Cells/immunology , Middle Aged , Young Adult
5.
Exp Hematol ; 40(2): 143-54, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22056679

ABSTRACT

Novel roles for the serine protease plasmin have been implicated recently in physiological and pathological processes. However, whether plasmin is involved in erythropoiesis is not known. In the present study, we studied the consequences of plasminogen deficiency on erythropoiesis in plasminogen-deficient (Plg knockout [KO]) mice. Erythroid differentiation was attenuated in male Plg KO mice and resulted in erythroblastic accumulation within the spleen and bone marrow, with increased apoptosis in the former, erythrocytosis, and splenomegaly, whereas similar erythropoietic defect was less prominent in female Plg KO mice. In addition, erythrocyte lifespan was shorter in both male and female Plg KO mice. Erythropoietin levels were compensatory increased in both male and female Plg KO mice, and resulted in a higher frequency of burst-forming units-erythroid within the spleen and bone marrow. Surprisingly, we found that male Plg KO mice, but not their female counterparts, exhibited normochromic normocytic anemia. The observed sex-linked erythropoietic defect was attributed to decreased serum testosterone levels in Plg KO mice as a consequence of impaired secretion of the pituitary luteinizing hormone (LH) under steady-state condition. Surgical castration causing testosterone deficiency and stimulating LH release attenuated erythroid differentiation and induced anemia in wild-type animals, but did not further decrease the hematocrit levels in Plg KO mice. In addition, complementation of LH using human choriogonadotropin, which increases testosterone production, improved the erythropoietic defect and anemia in Plg KO mice. The present results identify a novel role for plasmin in the hormonal regulation of postnatal erythropoiesis by the LH-testosterone axis.


Subject(s)
Erythropoiesis , Luteinizing Hormone/physiology , Plasminogen/physiology , Testosterone/physiology , Animals , Erythropoietin/blood , Female , Fibrinolysin/physiology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Plasminogen/deficiency
6.
Int Arch Allergy Immunol ; 156(2): 148-58, 2011.
Article in English | MEDLINE | ID: mdl-21576985

ABSTRACT

BACKGROUND: CD4(+)CD25(+) regulatory T (T(reg)) cells can control the allergic response to allergen, airway eosinophilia and airway hypersensitivity. We speculated that chronic inflammation persisting in asthma airways is dependent on abnormalities of these T(reg) cells. There are differences in the pathology of asthma in adults and children, and the airways of pediatric asthma are considered to be more naive than those of adults. Therefore, we analyzed the functionality of T(reg) cells in pediatric asthma and the relationship between T(reg) function and asthma symptoms. METHODS: The anergic state, which is one of the defining properties of T(reg), was analyzed by measuring intracellular Ca(2+) influx following T cell receptor (TCR) stimulation. FOXP3-positive cells and FOXP3 mRNA expression were measured by flow analysis and real-time PCR with the SYBR method, respectively. RESULTS: CD45RO(+) cells make up approximately 99% of CD4(+)CD25(high) T cells and 89% of CD4(+)CD25(low) T cells in human adult blood. The proportion of CD45RO(+) cells in CD4(+)CD25(+) (high + low) T cells from pediatric asthma was much smaller (about 56%). Interestingly, our data indicated that CD45RO(+) T(reg) cells from pediatric asthma aberrantly increased intracellular Ca(2+) concentrations following TCR activation compared with pediatric nonasthma controls. CONCLUSION: These impaired CD45RO(+) T(reg) cell functions were correlated with asthma symptoms. The correlation was observed in the group with a highly expressed atopic phenotype and longer duration of asthma. We suggest that chronic inflammation in pediatric asthma airways may be the result of impaired regulatory functions of CD45RO(+) T(reg) cells.


Subject(s)
Asthma/immunology , Calcium/immunology , Forkhead Transcription Factors/immunology , Leukocyte Common Antigens/immunology , T-Lymphocytes, Regulatory/immunology , Adolescent , Child , Female , Flow Cytometry , Forkhead Transcription Factors/genetics , Humans , Infant , Male , Microscopy, Fluorescence , RNA, Messenger/chemistry , RNA, Messenger/genetics , Reverse Transcriptase Polymerase Chain Reaction
7.
Blood ; 115(21): 4302-12, 2010 May 27.
Article in English | MEDLINE | ID: mdl-20110420

ABSTRACT

Ischemia of the heart, brain, and limbs is a leading cause of morbidity and mortality worldwide. Treatment with tissue type plasminogen activator (tPA) can dissolve blood clots and can ameliorate the clinical outcome in ischemic diseases. But the underlying mechanism by which tPA improves ischemic tissue regeneration is not well understood. Bone marrow (BM)-derived myeloid cells facilitate angiogenesis during tissue regeneration. Here, we report that a serpin-resistant form of tPA by activating the extracellular proteases matrix metalloproteinase-9 and plasmin expands the myeloid cell pool and mobilizes CD45(+)CD11b(+) proangiogenic, myeloid cells, a process dependent on vascular endothelial growth factor-A (VEGF-A) and Kit ligand signaling. tPA improves the incorporation of CD11b(+) cells into ischemic tissues and increases expression of neoangiogenesis-related genes, including VEGF-A. Remarkably, transplantation of BM-derived tPA-mobilized CD11b(+) cells and VEGFR-1(+) cells, but not carrier-mobilized cells or CD11b(-) cells, accelerates neovascularization and ischemic tissue regeneration. Inhibition of VEGF signaling suppresses tPA-induced neovascularization in a model of hind limb ischemia. Thus, tPA mobilizes CD11b(+) cells from the BM and increases systemic and local (cellular) VEGF-A, which can locally promote angiogenesis during ischemic recovery. tPA might be useful to induce therapeutic revascularization in the growing field of regenerative medicine.


Subject(s)
Myeloid Cells/drug effects , Myeloid Cells/physiology , Neovascularization, Physiologic/drug effects , Regeneration/drug effects , Tissue Plasminogen Activator/pharmacology , Animals , Base Sequence , Bone Marrow Transplantation , CD11b Antigen/metabolism , DNA Primers/genetics , Female , Gene Expression/drug effects , Ischemia/drug therapy , Ischemia/pathology , Ischemia/physiopathology , Male , Matrix Metalloproteinase 9/deficiency , Matrix Metalloproteinase 9/genetics , Matrix Metalloproteinase 9/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Mutant Proteins/pharmacology , Neovascularization, Physiologic/genetics , Plasminogen/deficiency , Plasminogen/genetics , Plasminogen/metabolism , Recombinant Proteins/pharmacology , Regeneration/physiology , Signal Transduction , Stem Cell Factor/metabolism , Tissue Plasminogen Activator/deficiency , Tissue Plasminogen Activator/genetics , Tissue Plasminogen Activator/physiology , Transplantation Chimera , Vascular Endothelial Growth Factor A/biosynthesis , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor Receptor-1/metabolism
8.
Cell Stem Cell ; 1(6): 658-70, 2007 Dec 13.
Article in English | MEDLINE | ID: mdl-18371407

ABSTRACT

Hematopoietic stem cells within the bone marrow exist in a quiescent state. They can differentiate and proliferate in response to hematopoietic stress (e.g., myelosuppression), thereby ensuring a well-regulated supply of mature and immature hematopoietic cells within the circulation. However, little is known about how this stress response is coordinated. Here, we show that plasminogen (Plg), a classical fibrinolytic factor, is a key player in controlling this stress response. Deletion of Plg in mice prevented hematopoietic stem cells from entering the cell cycle and undergoing multilineage differentiation after myelosuppression, leading to the death of the mice. Activation of Plg by administration of tissue-type plasminogen activator promoted matrix metalloproteinase-mediated release of Kit ligand from stromal cells, thereby promoting hematopoietic progenitor cell proliferation and differentiation. Thus, activation of the fibrinolytic cascade is a critical step in regulating the hematopoietic stress response.


Subject(s)
Fibrinolysis , Hematopoiesis , Hematopoietic Stem Cells/cytology , Plasminogen/genetics , Animals , Bone Marrow Transplantation , Cell Differentiation , Cell Proliferation , Cells, Cultured , Gene Deletion , Hematopoiesis/genetics , Hematopoiesis/physiology , Matrix Metalloproteinases/drug effects , Mice , Mice, Knockout , Plasminogen/pharmacology , Signal Transduction , Stem Cell Factor/drug effects , Stem Cell Factor/metabolism
9.
J Lipid Res ; 47(7): 1572-82, 2006 Jul.
Article in English | MEDLINE | ID: mdl-16603722

ABSTRACT

Human plasma contains at least three forms of adiponectin: a trimer, a hexamer, and a high-molecular-weight (HMW) multimer. We purified HMW adiponectin from human plasma using its affinity to gelatin and obtained monoclonal antibodies against it. On Western blot analysis, the reactivity of these monoclonal antibodies was shown to be restricted to a non-heat-denatured form of adiponectin molecules. On heating, the collagen-like domain of adiponectin molecules became denatured, and thus the trimer form could not be maintained. From these, monoclonal antibodies against HMW adiponectin were suggested to react with the intact trimer of adiponectin. With these monoclonal antibodies, we developed a sandwich ELISA system for quantifying adiponectin in human serum. Its specificity was verified by analysis of serum fractions separated by gel-filtration chromatography, and our ELISA system was found to be HMW adiponectin-specific. With this novel ELISA, the HMW adiponectin concentrations were 8.4 +/- 5.5 microg/ml (mean +/- SD) in healthy women and 6.2 +/- 3.6 microg/ml in healthy men. Also, serum with a lower HMW adiponectin concentration was shown to have a lower HMW ratio (i.e., HMW adiponectin/total adiponectin).


Subject(s)
Enzyme-Linked Immunosorbent Assay/methods , Adiponectin/analysis , Adiponectin/blood , Adiponectin/chemistry , Adiponectin/immunology , Adolescent , Aged , Aged, 80 and over , Animals , Antibodies, Monoclonal , CHO Cells , Cricetinae , Female , Humans , Male , Mice , Middle Aged , Molecular Structure , Molecular Weight , Multiprotein Complexes , Protein Structure, Quaternary , Recombinant Proteins/analysis , Recombinant Proteins/chemistry
10.
FASEB J ; 19(14): 2005-7, 2005 Dec.
Article in English | MEDLINE | ID: mdl-16223785

ABSTRACT

The granulocyte colony-stimulating factor (G-CSF) promotes angiogenesis. However, the exact mechanism is not known. We demonstrate that vascular endothelial growth factor (VEGF) was released by Gr-1+CD11b- neutrophils but not Gr-1-CD11b+ monocytes prestimulated with G-CSF in vitro and in vivo. Similarly, in vivo, concomitant with an increase in neutrophil numbers in circulation, G-CSF augmented plasma VEGF level in vivo. Local G-CSF administration into ischemic tissue increased capillary density and provided a functional vasculature and contributed to neovascularization of ischemic tissue. Blockade of the VEGF pathway abrogated G-CSF-induced angiogenesis. On the other hand, as we had shown previously, VEGF can induce endothelial progenitor cell (EPC) mobilization. Here, we show that G-CSF also augmented the number of circulating VEGF receptor-2 (VEGFR2) EPCs as compared with untreated controls. Blocking the VEGF/VEGFR1, but to a much lesser extent, the VEGF/VEGFR2 pathway in G-CSF-treated animals delayed tissue revascularization in a hindlimb model. These data clearly show that G-CSF modulates angiogenesis by increasing myelomonocytic cells (VEGFR1+ neutrophils) and their release of VEGF. Our results indicated that administration of G-CSF into ischemic tissue provides a novel and safe therapeutic strategy to improve neovascularization.


Subject(s)
Granulocyte Colony-Stimulating Factor/physiology , Analysis of Variance , Animals , CD11b Antigen/biosynthesis , Capillaries/pathology , Cells, Cultured , Collagen/chemistry , Drug Combinations , Endothelium, Vascular/cytology , Endothelium, Vascular/metabolism , Granulocyte Colony-Stimulating Factor/metabolism , Hematopoietic Stem Cells/cytology , Humans , Immunohistochemistry , In Situ Hybridization, Fluorescence , Ischemia/pathology , Laminin/chemistry , Leukocytes, Mononuclear/cytology , Male , Mice , Mice, Inbred BALB C , Models, Statistical , Monocytes/cytology , Neovascularization, Pathologic , Neutrophils/metabolism , Proteoglycans/chemistry , Recombinant Proteins/chemistry , Stem Cells/cytology , Time Factors , Vascular Endothelial Growth Factor A/blood , Vascular Endothelial Growth Factor A/metabolism , Vascular Endothelial Growth Factor Receptor-1/metabolism , Vascular Endothelial Growth Factor Receptor-2/metabolism
11.
J Exp Med ; 202(6): 739-50, 2005 Sep 19.
Article in English | MEDLINE | ID: mdl-16157686

ABSTRACT

Mast cells accumulate in tissues undergoing angiogenesis during tumor growth, wound healing, and tissue repair. Mast cells can secrete angiogenic factors such as vascular endothelial growth factor (VEGF). Ionizing irradiation has also been shown to have angiogenic potential in malignant and nonmalignant diseases. We observed that low-dose irradiation fosters mast cell-dependent vascular regeneration in a limb ischemia model. Irradiation promoted VEGF production by mast cells in a matrix metalloproteinase-9 (MMP-9)-dependent manner. Irradiation, through MMP-9 up-regulated by VEGF in stromal and endothelial cells, induced the release of Kit-ligand (KitL). Irradiation-induced VEGF promoted migration of mast cells from the bone marrow to the ischemic site. Irradiation-mediated release of KitL and VEGF was impaired in MMP-9-deficient mice, resulting in a reduced number of tissue mast cells and delayed vessel formation in the ischemic limb. Irradiation-induced vasculogenesis was abrogated in mice deficient in mast cells (steel mutant, Sl/Sl(d) mice) and in mice in which the VEGF pathway was blocked. Irradiation did not induce progenitor mobilization in Sl/Sl(d) mice. We conclude that increased recruitment and activation of mast cells following irradiation alters the ischemic microenvironment and promotes vascular regeneration in an ischemia model. These data show a novel mechanism of neovascularization and suggest that low-dose irradiation may be used for therapeutic angiogenesis to augment vasculogenesis in ischemic tissues.


Subject(s)
Gamma Rays , Hematopoietic Stem Cells/radiation effects , Mast Cells/metabolism , Mast Cells/radiation effects , Matrix Metalloproteinase 9/physiology , Neovascularization, Physiologic/radiation effects , Vascular Endothelial Growth Factor A/metabolism , Vascular Endothelial Growth Factor A/radiation effects , Animals , Bone Marrow/radiation effects , Cell Proliferation/radiation effects , Extremities/blood supply , Hematopoietic Stem Cells/enzymology , Ischemia/radiotherapy , Mast Cells/enzymology , Matrix Metalloproteinase 9/deficiency , Matrix Metalloproteinase 9/genetics , Matrix Metalloproteinase 9/radiation effects , Mice
SELECTION OF CITATIONS
SEARCH DETAIL
...