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











Publication year range
1.
Lab Anim Res ; 39(1): 5, 2023 Mar 08.
Article in English | MEDLINE | ID: mdl-36890604

ABSTRACT

BACKGROUND: Orthotopic liver transplantation is the only option for patients with end-stage liver disease and hepatocellular carcinoma. Post-transplant immunosuppressive therapy is important to prevent graft failure. We investigated the effectiveness of tacrolimus (FK506) and their mechanisms for liver transplant immune tolerance in an outbred rat LT model. RESULTS: To investigate the therapeutic effect of the FK506 on outbred rat LT model, FK506 and postoperative therapy were administered subcutaneously once or twice daily to transplanted rats. Histopathological and immunohistochemical analyses were conducted for all groups. The regulation of inflammatory cytokine signaling in the spleen was analyzed by flow cytometry. FK506 attenuated allograft rejection and increased survival in rat orthotopic liver transplantation models. The FK506-treated group had reduced serum levels of alanine aminotransferase, aspartate aminotransferase and alkaline phosphatase. Furthermore, FK506 decreased the expression of inflammatory cytokines and the activation of pathogenic Th1 and Th17 cells in the liver. CONCLUSIONS: Taken together, we revealed that FK506 ameliorated strong allograft rejection in outbred liver transplantation model by anti-inflammatory effect and inhibitory peroperty of pathogenic T cells.

2.
Exp Mol Med ; 54(9): 1577-1585, 2022 09.
Article in English | MEDLINE | ID: mdl-36175484

ABSTRACT

Systemic sclerosis (SSc) is a chronic autoimmune disease characterized by inflammation, microangiopathy, and progressive fibrosis in the skin and internal organs. To evaluate the pathophysiologic mechanisms and efficacies of potential therapeutics for SSc, a preclinical model recapitulating the disease phenotypes is needed. Here, we introduce a novel animal model for SSc using immunodeficient mice injected with peripheral blood mononuclear cells (PBMCs) from SSc patients. Human PBMCs acquired from SSc patients and healthy controls were transferred into NOD.Cg-PrkdcscidIl2rgtm1Wjl (NSG) mice with concurrent bleomycin injection. Blood, skin, and lung tissues were acquired and analyzed after PBMC engraftment. In addition, we investigated whether the humanized murine model could be used to assess the efficacy of potential therapeutics for SSc. Human PBMCs from SSc patients and healthy controls were engrafted into the blood, skin, and lung tissues of NSG mice. Histological analysis of affected tissues from mice treated with SSc PBMCs (SSc hu-mice) demonstrated substantial inflammation, fibrosis and vasculopathy with human immune cell infiltration and increased expression of IL-17, TGF-ß, CCL2, CCL3, and CXCL9. The proportions of circulating and tissue-infiltrating T helper 17 (Th17) cells were elevated in SSc hu-mice. These cells showed increased expression of CXCR3 and phosphorylated STAT3. SSc hu-mice treated with rebamipide and other potential Th17-cell-modulating drugs presented significantly reduced tissue fibrosis. Mice injected with patient-derived PBMCs show promise as an animal model of SSc.


Subject(s)
Interleukin-17 , Scleroderma, Systemic , Animals , Bleomycin , Disease Models, Animal , Fibrosis , Humans , Inflammation/metabolism , Leukocytes, Mononuclear/metabolism , Lung/pathology , Mice , Mice, Inbred NOD , Mice, SCID , Scleroderma, Systemic/metabolism , Scleroderma, Systemic/pathology , Skin/metabolism , Th17 Cells , Transforming Growth Factor beta/metabolism
3.
Front Immunol ; 13: 817006, 2022.
Article in English | MEDLINE | ID: mdl-35418987

ABSTRACT

Although rejection or tolerance can occur in liver transplantation (LT) patients, there are no reliable non-invasive methods for predicting immune homeostasis. In this study, we developed a humanized mouse model to predict liver immune homeostasis in patients who underwent LT. The patient-derived avatar model was developed by injecting peripheral blood mononuclear cells from healthy controls (HCs) or LT patients with stable, rejection, or tolerance into NOD.Cg-PrkdcscidIL2rgtm1Wjl/SzJ (NSG) mice, followed by injection of human hepatic stellate cells and Carbone tetrachloride (CCl4). After 7 weeks, the patient's T-cell engraftment and liver inflammation in the avatar model were evaluated and compared with the liver histology of LT patients. Changes in liver inflammation following treatment with tacrolimus and/or biguanide derivatives were also examined. The C-X-C Motif Chemokine Receptor 3 (CXCR3)-dependently engrafted patient T cells led to differences in liver inflammation in our model according to the status of LT patients. The livers of avatar models from rejection patients had severe inflammation with more T helper 17 cells and fewer regulatory T cells compared to those of models from tolerance and HCs showing only mild inflammation. Moreover, our model classified stable post-LT patients into severe and mild inflammation groups, which correlated well with liver immunity in these patients. Our models revealed alleviation of inflammation after combination treatment with tacrolimus and biguanide derivatives or monotherapy. Consequently, using our new patient-derived avatar model, we predicted liver immune homeostasis in patients with stable LT without biopsy. Moreover, our avatar model may be useful for preclinical analysis to evaluate treatment responses while reducing risks to patients.


Subject(s)
Liver Transplantation , Animals , Biguanides , Disease Models, Animal , Homeostasis , Humans , Inflammation , Leukocytes, Mononuclear , Liver , Liver Transplantation/adverse effects , Mice , Mice, Inbred NOD , Tacrolimus
4.
Front Immunol ; 12: 606024, 2021.
Article in English | MEDLINE | ID: mdl-33986739

ABSTRACT

Multiple studies have explored the potential role of programmed death-ligand 1 (PD-L1) as a mediator of Myeloid-derived suppressor cells (MDSCs) effects in various cancers. However, the role PD-L1 expression in MDSCs on autoimmune disease is still largely unknown.This study was undertaken to whether MDSC expressing PD-L1 have more potent immunoregulatory activity and control autoimmunity more effectively in two murine models of lupus (MRL/lpr mice and Roquinsan/san mice). The populations of MDSC were increased in peripheral blood of lupus patients. The mRNA levels of immunosuppressive molecules were profoundly decreased in MDSCs from lupus patients and mice. Co-culture with splenocytes showed that PD-L1 expressing MDSCs from control mice expand both Treg cells and regulatory B cells more potently. Infusion of PD-L1 expressing MDSCs reduced autoantibody levels and degree of proteinuria and improved renal pathology of two animal models of lupus. Moreover, PD-L1 expressing MDSCs therapy can suppress double negative (CD4-CD8-CD3+) T cells, the major pathogenic immune cells and follicular helper T cells in MRL/lpr mice, and podocyte damage. Our results indicate PD-L1 expressing MDSCs have more potent immunoregualtory activity and ameliorate autoimmunity more profoundly. These findings suggest PD-L1 expressing MDSCs be a promising therapeutic strategy targeting systemic autoimmune diseases.


Subject(s)
B7-H1 Antigen/genetics , Gene Expression Regulation , Immunomodulation/genetics , Lupus Erythematosus, Systemic/etiology , Lupus Erythematosus, Systemic/metabolism , Myeloid-Derived Suppressor Cells/immunology , Myeloid-Derived Suppressor Cells/metabolism , Animals , Autoimmunity/genetics , B-Lymphocyte Subsets/immunology , B-Lymphocyte Subsets/metabolism , B7-H1 Antigen/metabolism , Biomarkers , Cell- and Tissue-Based Therapy , Disease Models, Animal , Disease Susceptibility , Female , Humans , Immunophenotyping , Lupus Erythematosus, Systemic/pathology , Lupus Erythematosus, Systemic/therapy , Mice , Mice, Inbred MRL lpr , Mice, Transgenic , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism
5.
J Transl Med ; 18(1): 483, 2020 12 14.
Article in English | MEDLINE | ID: mdl-33317573

ABSTRACT

BACKGROUND: Myeloid-derived suppressor cells (MDSCs) play a critical role in modulating the immune response and promoting immune tolerance in models of autoimmunity and transplantation. Regulatory T cells (Tregs) exert therapeutic potential due to their immunomodulatory properties, which have been demonstrated both in vitro and in clinical trials. Cell-based therapy for acute graft-versus-host disease (aGVHD) may enable induction of donor-specific tolerance in the preclinical setting. METHODS: We investigated whether the immunoregulatory activity of the combination of MDSCs and Tregs on T cell and B cell subset and alloreactive T cell response. We evaluated the therapeutic effects of combined cell therapy for a murine aGVHD model following MHC-mismatched bone marrow transplantation. We compared histologic analysis from the target tissues of each groups were and immune cell population by flow cytometric analysis. RESULTS: We report a novel approach to inducing immune tolerance using a combination of donor-derived MDSCs and Tregs. The combined cell-therapy modulated in vitro the proliferation of alloreactive T cells and the Treg/Th17 balance in mice and human system. Systemic infusion of MDSCs and Tregs ameliorated serverity and inflammation of aGVHD mouse model by reducing the populations of proinflammatory Th1/Th17 cells and the expression of proinflammatory cytokines in target tissue. The combined therapy promoted the differentiation of allogeneic T cells toward Foxp3 + Tregs and IL-10-producing regulatory B cells. The combination treatment control also activated human T and B cell subset. CONCLUSIONS: Therefore, the combination of MDSCs and Tregs has immunomodulatory activity and induces immune tolerance to prevent of aGVHD severity. This could lead to the development of new clinical approaches to the prevent aGVHD.


Subject(s)
Graft vs Host Disease , Myeloid-Derived Suppressor Cells , Acute Disease , Animals , Graft vs Host Disease/therapy , Immunity , Mice , Mice, Inbred C57BL , T-Lymphocytes, Regulatory , Th17 Cells
6.
J Transl Med ; 18(1): 225, 2020 06 06.
Article in English | MEDLINE | ID: mdl-32505218

ABSTRACT

BACKGROUND: Fibrosis is the formation of excess connective tissue in an organ or tissue during a reparative or reactive process. Graft-versus-host disease (GvHD) is a medical complication of allogeneic tissue transplantation with transplanted donor T cell-mediated inflammatory response; it is characterized by a severe immune response with fibrosis in the final stage of the inflammatory process. T helper 17 cells play a critical role in the pathogenesis of GvHD. Fingolimod (FTY720), an analogue of sphingosine-1-phosphate (S1P), is an effective immunosuppressive agent in experimental transplantation models. METHODS: In this study, we evaluated the effects of FTY720 as a treatment for an animal GvHD model with inflammation and fibrosis. The splenocytes, lymph nodes, blood, tissues from Syngeneic mice and GvHD-induced mice treated vehicle or FTY720 were compared using flow cytometry, hematological analyses, histologic analyses. RESULTS: FTY720 reduced clinical scores based on the following five clinical parameters: weight loss, posture, activity, fur texture, and skin integrity. FACS data showed that T lymphocyte numbers increased in mesenteric lymph nodes and decreased in splenocytes of FTY720-treated mice. Tissue analysis showed that FTY720 reduced skin, intestinal inflammation, and fibrotic markers. FTY720 dramatically decreased α-smooth muscle actin, connective tissue growth factor, and fibronectin protein levels in keloid skin fibroblasts. CONCLUSIONS: Thus, FTY720 suppressed migration of pathogenic T cells to target organs, reducing inflammation. FTY720 also inhibited fibrogenesis marker expression in vitro and in vivo. Together, these results suggest that FTY720 prevents GvHD progression via immunosuppression of TH17 and simultaneously acts an anti-fibrotic agent.


Subject(s)
Fingolimod Hydrochloride , Graft vs Host Disease , Animals , Fibrosis , Fingolimod Hydrochloride/pharmacology , Fingolimod Hydrochloride/therapeutic use , Graft vs Host Disease/drug therapy , Immunosuppressive Agents/pharmacology , Mice , Mice, Inbred C57BL , Propylene Glycols/pharmacology
7.
Front Immunol ; 10: 1526, 2019.
Article in English | MEDLINE | ID: mdl-31379809

ABSTRACT

Optimizing Treg function and improving Treg stability are attractive treatment strategies for treating autoimmune rheumatoid arthritis (RA). However, the limited number of circulating Tregs and questions about the functional stability of in vitro-expanded Tregs are potential limitations of Treg-based cell therapy. The aim of this study was to analyze the regulatory effect of daurinol, a catalytic inhibitor of topoisomerase IIα, on Th cell differentiation and to evaluate their therapeutic potential in a preclinical experimental model of RA. We investigated the effect of daurinol on T cell differentiation by flow cytometry. Foxp3 stability and methylation were analyzed by suppression assays and bisulfite pyrosequencing. Daurinol was treated in the collagen-induced arthritis (CIA) model, and the effects in vivo were determined. We found that daurinol can promote Treg differentiation and reciprocally inhibit Th17 differentiation. This Treg-inducing property of daurinol was associated with decreased activity of Akt-mTOR and reciprocally increased activity of neuropilin-1 (Nrp1)-PTEN. Daurinol treatment inhibited aerobic glycolysis in Th17 conditions, indicating the metabolic changes by daurinol. We found that the daurinol increase the Treg stability was achieved by Foxp3 hypomethylation. In vivo daurinol treatment in CIA mice reduced the clinical arthritis severity and histological inflammation. The Treg population frequency increased and the Th17 cells decreased in the spleens of arthritis mice treated with daurinol. These results showed the anti-arthritic and immunoregulating properties of daurinol is achieved by increased differentiation and stabilization of Tregs. Our study provides first evidence for daurinol as a treatment for RA.


Subject(s)
Arthritis, Rheumatoid/immunology , Autoimmune Diseases/immunology , Forkhead Transcription Factors/immunology , Neuropilin-1/immunology , PTEN Phosphohydrolase/immunology , Signal Transduction/immunology , T-Lymphocytes, Regulatory/immunology , Animals , Arthritis, Experimental/immunology , Cell Differentiation/immunology , Cytokines/immunology , Humans , Inflammation/immunology , Lymphocyte Activation/immunology , Mice , Mice, Inbred C57BL , Mice, Inbred DBA , Th17 Cells/immunology
8.
J Immunol ; 203(1): 127-136, 2019 07 01.
Article in English | MEDLINE | ID: mdl-31142603

ABSTRACT

Mesenchymal stem cells (MSCs) can protect against cartilage breakdown in osteoarthritis (OA) via their immunomodulatory capacities. However, the optimization strategy for using MSCs remains challenging. This study's objective was to identify the in vivo effects of metformin-stimulated adipose tissue-derived human MSCs (Ad-hMSCs) in OA. An animal model of OA was established by intra-articular injection of monosodium iodoacetate into rats. OA rats were divided into a control group and two therapy groups (treated with Ad-hMSCs or metformin-stimulated Ad-hMSCs). Limb nociception was assessed by measuring the paw withdrawal latency and threshold. Our data show that metformin increased IL-10 and IDO expression in Ad-hMSCs and decreased high-mobility group box 1 protein, IL-1ß, and IL-6 expression. Metformin increased the migration capacity of Ad-hMSCs with upregulation of chemokine expression. In cocultures, metformin-stimulated Ad-hMSCs inhibited the mRNA expression of RUNX2, COL X, VEGF, MMP1, MMP3, and MMP13 in IL-1ß-stimulated OA chondrocytes and increased the expression of TIMP1 and TIMP3. The antinociceptive activity and chondroprotective effects were greater in OA rats treated with metformin-stimulated Ad-hMSCs than in those treated with unstimulated Ad-hMSCs. TGF-ß expression in subchondral bone of OA joints was attenuated more in OA rats treated with metformin-stimulated Ad-hMSCs. Our findings suggest that metformin offers a promising option for the clinical application of Ad-hMSCs as a cell therapy for OA.


Subject(s)
Adipose Tissue/cytology , Anti-Inflammatory Agents/metabolism , Chondrocytes/physiology , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells/physiology , Metformin/metabolism , Osteoarthritis/therapy , Animals , Cell Movement , Cells, Cultured , Cytoprotection , Diphosphates , Disease Models, Animal , Humans , Imidazoles , Indoleamine-Pyrrole 2,3,-Dioxygenase/metabolism , Interleukin-10/metabolism , Male , Nociception , Rats , Rats, Wistar
10.
Immunol Lett ; 201: 45-51, 2018 09.
Article in English | MEDLINE | ID: mdl-30395870

ABSTRACT

Inflammatory bowel disease (IBD) is caused by chronic inflammation of the gastrointestinal tract. The pathogenesis of IBD remains unclear. The inflammation is associated with activation of T helper (Th) lymphocytes and chronic production of inflammatory cytokines. Ro60 suppresses the expression of tumor necrosis factor α, interleukin (IL)-6, and interferon α by inhibiting Alu transcription; control of Ro60 mRNA expression may thus be therapeutically useful. However, few studies have evaluated the anti-inflammatory activity of Ro60. The Ro60 level is decreased in IBD patients; we thus hypothesized that Ro60 was involved in the development of this autoimmune disease. We subjected mice with dextran sodium sulfate (DSS)-induced colitis to gene therapy using a vector that overexpressed Ro60 threefold. We scored IBD progression by repeatedly weighing the mice. Ro60 ameliorated colitis severity and reduced the levels of tumor necrosis factor α, IL-6, IL-17, IL-8, and vascular endothelial growth factor. Ro60 overexpression decreased the levels of α-smooth muscle actin (a marker of activated myofibroblasts) and type I collagen. The anti-inflammatory and anti-fibrotic activities of Ro60 ameliorated the severity of DSS-induced colitis in mice by repressing inflammation, fibrosis, angiogenesis, and the production of reactive oxygen species.


Subject(s)
Autoantigens/metabolism , Colitis/immunology , Colon/pathology , Inflammation/immunology , Inflammatory Bowel Diseases/immunology , RNA, Small Cytoplasmic/metabolism , Ribonucleoproteins/metabolism , Actins/metabolism , Animals , Autoantigens/genetics , Colitis/therapy , Collagen Type I/metabolism , Colon/physiology , Cytokines/metabolism , Dextran Sulfate , Disease Models, Animal , Fibrosis , Genetic Therapy , Genetic Vectors , Humans , Inflammation/therapy , Inflammation Mediators/metabolism , Inflammatory Bowel Diseases/therapy , Mice , Mice, Inbred C57BL , RNA, Small Cytoplasmic/genetics , Ribonucleoproteins/genetics
11.
Sci Rep ; 8(1): 3753, 2018 02 28.
Article in English | MEDLINE | ID: mdl-29491381

ABSTRACT

Myeloid-derived suppressor cells (MDSCs) are heterogenous populations of immature myeloid progenitor cells with immunoregulatory function. MDSCs play critical roles in controlling the processes of autoimmunity but their roles in rheumatoid arthritis (RA) are controversial. The present study was undertaken to investigate whether MDSCs have therapeutic impact in mice with collagen-induced arthritis (CIA), an animal model of RA. We also examined the mechanisms underlying the anti-arthritic effect of MDSCs. In vitro treatment with MDSCs repressed IL-17 but increased FOXP3 in CD4+ T cells in mice. In vivo infusion of MDSCs markedly ameliorated inflammatory arthritis. Th17 cells and Th1 cells were decreased while Tregs were increased in the spleens of MDSCs-treated mice. MDSCs profoundly inhibited T cell proliferation. Addition of anti-IL-10 almost completely blocked the anti-proliferative effects of MDSCs on T cells. Anti-IL-10 blocked the expansion of Tregs by MDSCs. However, infusion of MDSCs from IL-10 KO mice failed to suppress inflammatory arthritis. MDSCs could reciprocally regulate Th17/Treg cells and suppress CIA via IL-10, suggesting that MDSCs might be a promising therapeutic strategy for T cell mediated autoimmune diseases including RA.


Subject(s)
Arthritis, Rheumatoid/immunology , Arthritis, Rheumatoid/metabolism , Interleukin-10/biosynthesis , Myeloid-Derived Suppressor Cells/metabolism , T-Lymphocytes, Regulatory/cytology , Animals , Arginase/metabolism , Cell Proliferation , Forkhead Transcription Factors/metabolism , Interleukin-10/metabolism , Male , Mice , Myeloid-Derived Suppressor Cells/cytology , Th1 Cells/cytology , Th17 Cells/cytology
12.
Front Immunol ; 9: 2881, 2018.
Article in English | MEDLINE | ID: mdl-30619261

ABSTRACT

Osteoarthritis (OA) is a degenerative disease that induces pain, cartilage deformation, and joint inflammation. Mesenchymal stem cells (MSCs) are potential therapeutic agents for treatment of OA. However, MSC therapy can cause excessive inflammation. Signal transducer and activator of transcription 3 (STAT3) modulates secretion of many proinflammatory cytokines. Experimental OA was induced by intra-articular (IA) injection of monosodium iodoacetate (MIA) to the right knee of rats. MSCs from OA patients (OA-MSCs) were treated with STA21, a small molecule that blocks STAT3 signaling, by IA or intravenous (IV) injection after MIA injection. Pain severity was quantified by assessment of secondary tactile allodynia using the von Frey assessment test. Cartilage degradation was measured by microcomputed tomography image analysis, histological analysis, and the Mankin score. Protein and gene expression was evaluated by enzyme-linked immunosorbent assay, immunohistochemistry, and real-time polymerase chain reaction. MSCs increased production of proinflammatory cytokines under inflammatory conditions. STA21 significantly decreased expression of these proinflammatory molecules via inhibition of STAT3 activity but increased gene expression of molecules related to migration potential and immunomodulation in OA-MSCs. STAT3-inhibited OA-MSCs administrated by IV or IA injection decreased pain severity and cartilage damage in rats with MIA-induced OA rats by decreasing proinflammatory cytokines in the joints. Combined IA and IV-injected STAT3-inhibited OA-MSCs had an additive effect of pain relief in MIA-induced OA rats. STAT3 inhibition may optimize the therapeutic activities of MSCs for treating OA by attenuating pain and progression of MIA by inhibiting inflammation and cartilage damage.


Subject(s)
Arthritis, Experimental/metabolism , Cartilage, Articular/metabolism , Mesenchymal Stem Cells/metabolism , Osteoarthritis/metabolism , Pain/metabolism , STAT3 Transcription Factor/metabolism , Administration, Intravenous , Animals , Arthritis, Experimental/chemically induced , Arthritis, Experimental/therapy , Cartilage, Articular/pathology , Cells, Cultured , Cytokines/metabolism , Disease Models, Animal , Heterografts , Humans , Injections, Intra-Articular , Iodoacetic Acid , Male , Mesenchymal Stem Cell Transplantation/methods , Mesenchymal Stem Cells/drug effects , Osteoarthritis/chemically induced , Osteoarthritis/therapy , Pain/physiopathology , Pain/prevention & control , Rats, Wistar , STAT3 Transcription Factor/antagonists & inhibitors , STAT3 Transcription Factor/genetics , Small Molecule Libraries/administration & dosage , Small Molecule Libraries/chemistry , Small Molecule Libraries/pharmacology
13.
Tissue Eng Regen Med ; 14(4): 453-464, 2017 Aug.
Article in English | MEDLINE | ID: mdl-30603501

ABSTRACT

Human embryonic stem cell (hESC) culture system has been changing culture conditions from conventional to xeno-free for therapeutic cell applications, and N-glycolylneuraminic acid (Neu5Gc) could be a useful indicator of xenogeneic contaminations in hESCs because human cells can no longer produce it genetically. We set up the humanized culture condition using commercially available humanized materials and two different adaptation methods: sequential or direct. SNUhES4 and H1 hESC lines, previously established in conventional culture conditions, were maintained using the humanized culture condition and were examined for the presence of Neu5Gc. The hESCs showed the same morphology and character as those of the conventional culture condition. Moreover, they were negative for Neu5Gc within two passages without loss of pluripotency. This study suggested that this method can effectively cleanse previously established hESC lines, bringing them one step closer to being clinical-grade hESCs.

14.
Dev Reprod ; 20(1): 63-71, 2016 Mar.
Article in English | MEDLINE | ID: mdl-27294211

ABSTRACT

Human embryonic stem cells (hESCs) have been routinely cultured on mouse embryonic fibroblast feederlayers with a medium containing animal materials. For clinical application of hESCs, animal-derived products from the animal feeder cells, animal substrates such as gelatin or Matrigel and animal serum are strictly to be eliminated in the culture system. In this study, we performed that SNUhES32 and H1 were cultured on human amniotic fluid cells (hAFCs) with KOSR XenoFree and a humanized substrate. All of hESCs were relatively well propagated on hAFCs feeders with xeno-free conditions and they expressed pluripotent stem cell markers, alkaline phosphatase, SSEA-4, TRA1-60, TRA1-81, Oct-4, and Nanog like hESCs cultured on STO or human foreskin fibroblast feeders. In addition, we observed the expression of nonhuman N-glycolylneuraminic acid (Neu5GC) molecules by flow cytometry, which was xenotransplantation components of contamination in hESCs cultured on animal feeder conditions, was not detected in this xeno-free condition. In conclusion, SNUhES32 and H1 could be maintained on hAFCs for humanized culture conditions, therefore, we suggested that new xenofree conditions for clinical grade hESCs culture will be useful data in future clinical studies.

15.
Nitric Oxide ; 20(2): 122-8, 2009 Mar.
Article in English | MEDLINE | ID: mdl-18976718

ABSTRACT

Nitric oxide (NO) is a marker of pulmonary inflammation. In asthma, the levels of exhaled NO are elevated and the source of this increased NO is inducible nitric oxide synthase (iNOS) within airway epithelial cells. Epimagnolin and fargesin are compounds isolated from the ethanol extract of Magnoliae flos, the seed of the Magnolia plant and are used to treat nasal congestion, headache and sinusitis in Asian countries. This study investigated whether epimagnolin and fargesin inhibit extracellular signal-regulated kinase (ERK) activation and decrease iNOS expression and NO production in stimulated human respiratory epithelial cells. An immortal Type II alveolar cell line of human origin (A549) was stimulated by cytomix (CM), composed of IL-1beta, TNF-alpha and IFN-gamma, with or without concurrent exposure to M. flos extract (epimagnolin or fargesin). CM-induced levels of NO production, iNOS expression and ERK activation were evaluated. A549 cells stimulated with CM showed increases in iNOS mRNA and protein expression, and NO synthesis. However, treatment with epimagnolin or fargesin decreased levels of iNOS mRNA and protein expression, and NO synthesis. CM stimulated a rapid increase in the activity of ERK, whereas epimagnolin and fargesin inhibited ERK phosphorylation. Epimagnolin and fargesin inhibit iNOS expression and decrease production of NO via ERK pathway in cytokine-stimulated human respiratory epithelial cells.


Subject(s)
Benzodioxoles/pharmacology , Extracellular Signal-Regulated MAP Kinases/metabolism , Lignans/pharmacology , Magnolia/chemistry , Nitric Oxide Synthase Type II/antagonists & inhibitors , Plant Extracts/pharmacology , Respiratory Mucosa/metabolism , Analysis of Variance , Animals , Benzodioxoles/isolation & purification , Cell Line , Cell Line, Tumor , Cell Survival , Flavonoids/pharmacology , Gene Expression/drug effects , Gene Expression/physiology , Humans , Interferon-gamma/metabolism , Interleukin-1beta/metabolism , Lignans/isolation & purification , Mice , Nitric Oxide/metabolism , Nitric Oxide Synthase Type II/genetics , Nitric Oxide Synthase Type II/metabolism , Plant Extracts/chemistry , RNA, Messenger/metabolism , Seeds/chemistry , Statistics, Nonparametric , Tumor Necrosis Factor-alpha/metabolism
16.
Chromosome Res ; 16(8): 1075-84, 2008.
Article in English | MEDLINE | ID: mdl-18937039

ABSTRACT

Human embryonic stem cells (hESCs) are pluripotent and hold great promise as useful tools in basic scientific research and in the field of regenerative medicine. However, several studies have recently reported chromosomal abnormalities such as gains of chromosomes 12, 17 and X in hESCs. This genetic instability presents an obstacle in the application of hESCs as sources of cell therapies. We found that trisomy 12 was correlated with changes in hESC colony morphology during hESC maintenance. In this study, we investigated whether normal and trisomy 12 cells could be separated in hESC cultures displaying trisomy 12 mosaicism with two types of colony morphology using a mechanical transfer technique. Eight sublines were cultured from eight hESC colonies displaying normal or abnormal morphology. Four sublines with normal morphology had normal chromosome 12 numbers, whereas the four sublines with abnormal morphology displayed trisomy 12. These results indicate that a hESC colony with a minor degree of chromosomal mosaicism and normal morphology could proceed to a colony with normal chromosomes after prolonged cultures with mechanical transfer. Therefore, analysis of cultures for chromosomal abnormalities when changes in colony morphology are observed during culture is essential for maintaining normal hESC lines.


Subject(s)
Cell Separation/methods , Cell- and Tissue-Based Therapy/methods , Chromosomes, Human, Pair 12/genetics , Embryonic Stem Cells/cytology , Mosaicism , Trisomy , Alkaline Phosphatase/metabolism , Cell Line , Humans , In Situ Hybridization, Fluorescence , Karyotyping
17.
J Mol Cell Cardiol ; 39(5): 823-32, 2005 Nov.
Article in English | MEDLINE | ID: mdl-16198370

ABSTRACT

Human vascular smooth muscle cells (HVSMCs) are resistant to Fas-mediated death under normal physiological conditions. However, HVSMC death by activation of the receptor pathway was reported in the atherosclerotic lesions. In this study, we investigated whether 7-ketocholesterol, one of the major cholesterol oxides in the lesions, altered resistance of HVSMC to Fas-mediated death pathway. Cross-linking of Fas receptor with agonistic anti-Fas antibody (CH11) in the presence of 7-ketocholesterol induced death in human aorta smooth muscle cells (HAoSMC) as detected by morphology, viability, and DNA fragmentation. The agonistic anti-Fas antibody, however, did not induce death in the presence of 7alpha-hydroxycholesterol or cholesterol. The HAoSMC death was significantly inhibited by an antagonistic Fas receptor (FasR) antibody and by expression of dominant negative Fas-associated death domain containing protein (DN-FADD) using adenoviruses. Activation of caspase-3 was observed in HAoSMC destined to death. HAoSMC death was significantly inhibited by pharmacological caspase inhibitor, z-VAD and z-DEVD, and baculovirus caspase inhibitor p35. 7-Ketocholesterol impaired mitochondrial transmembrane potential and ATP production. Overexpression of bcl-xL also significantly inhibited HAoSMC death. In dying HAoSMC, bax was translocated from the cytosol to mitochondria and cytochrome c was released from mitochondria into the cytosol. This is the first report demonstrating implication of the oxysterol in Fas-mediated death pathway. The present study proposes that 7-ketocholesterol would contribute to loss of HVSMC in the atherosclerotic lesions by altering resistance to receptor-mediated death pathway.


Subject(s)
Aorta/cytology , Ketocholesterols/pharmacology , Myocytes, Smooth Muscle/cytology , Myocytes, Smooth Muscle/drug effects , fas Receptor/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Caspases/metabolism , Cell Death/drug effects , Cell Shape , Cytochromes c/metabolism , Cytosol/metabolism , Fas-Associated Death Domain Protein , Humans , Membrane Potentials , Mitochondria/drug effects , Mitochondria/metabolism , Myocytes, Smooth Muscle/metabolism , Protein Transport , Time Factors , bcl-2-Associated X Protein/metabolism
18.
Biochem Biophys Res Commun ; 333(4): 1093-9, 2005 Aug 12.
Article in English | MEDLINE | ID: mdl-15993097

ABSTRACT

This study was undertaken to investigate whether a physiologically compatible concentration of 7-ketocholesterol had any effect on human vascular smooth muscle cells (HVSMCs). We found that 7-ketocholesterol changed the viability of human aorta smooth muscle cells (HAoSMC) not by cytotoxicity but by activation of tumor necrosis factor-alpha receptor (TNFR)-mediated death. Whereas TNF-alpha did not affect the viability in the presence of 7alpha-hydroxycholesterol or cholesterol, the cytokine induced HAoSMC death in the presence of 7-ketocholesterol as detected by morphology, viability, and fragmentation of chromosomal DNA. The HAoSMC death was inhibited by a neutralizing anti-TNF receptor 1 (TNFR1) antibody and by the caspase inhibitors of z-VAD and z-DEVD. Activations of caspase-8 and -3 were detected from dying HAoSMCs. 7-Ketocholesterol inhibited translocation of the nuclear factor kappaB (NF-kappaB) subunits of p65 and p50 from the cytosol into the nucleus, increase of NF-kappaB activity, and expression of caspase-8 homolog Fas ligand interleukin-1-converting enzyme inhibitory protein by TNF-alpha. We also found that X-chromosome-linked inhibitor of apoptosis protein was degraded in dying HAoSMC. The present study proposes that 7-ketocholesterol would contribute to the disappearance of HVSMC in the atherosclerotic lesions by enhancing receptor-mediated death. This is the first report demonstrating induction of TNF-alpha-mediated death by oxysterol in cells.


Subject(s)
Aorta/metabolism , Apoptosis/physiology , Ketocholesterols/pharmacology , Muscle, Smooth, Vascular/metabolism , Myocytes, Smooth Muscle/metabolism , Tumor Necrosis Factor-alpha/pharmacology , Aorta/cytology , Aorta/drug effects , Apoptosis/drug effects , Cells, Cultured , Dose-Response Relationship, Drug , Humans , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/drug effects , Myocytes, Smooth Muscle/cytology , Myocytes, Smooth Muscle/drug effects
19.
J Ethnopharmacol ; 97(3): 515-9, 2005 Mar 21.
Article in English | MEDLINE | ID: mdl-15740889

ABSTRACT

The accumulation of circulating monocytes in the arterial wall is an early in atherosclerotic plaque formation. Monocyte chemoattractant protein-1 (MCP-1) promotes the migration of monocytes and would play a role in the development of atherosclerotic lesions. Searching for inhibitors of MCP-1-induced cell migration from natural sources, we isolated one active compound through active-guided fractionations from the MeOH extracts of Sophora flavescens Ait (Leguminosae). On the basis of spectral evidence, the structure of active compound was identified as kurarinone. It inhibited the migration of THP-1 cells induced by MCP-1 with IC50 value of 19.2 microg/mL. In addition, it inhibited the binding of MCP-1 to THP-1 cells and phosphorylation of p42/44 MARK.


Subject(s)
Cell Migration Inhibition , Chemokine CCL2/antagonists & inhibitors , Chemokine CCL2/physiology , Chemotaxis/drug effects , Flavonoids/pharmacology , Sophora , Cell Line , Chemotaxis/physiology , Dose-Response Relationship, Drug , Flavonoids/chemistry , Flavonoids/isolation & purification , Humans , Plant Extracts/chemistry , Plant Extracts/isolation & purification , Plant Extracts/pharmacology , Plant Roots
20.
Arch Pharm Res ; 26(9): 716-8, 2003 Sep.
Article in English | MEDLINE | ID: mdl-14560918

ABSTRACT

In the course of our search for Acyl-CoA: cholesterol acyltransferase (ACAT) inhibitors from natural sources, a new type of ACAT inhibitor was isolated from a methanol extract of Diospyros kaki. On the basis of spectral and structural evidence, the compound was identified as pheophorbide A-methyl ester. Pheophorbide A-methyl ester inhibited ACAT activity in a dose dependent manner with an IC50 value of 1.85 microg/mL.


Subject(s)
Acyl Coenzyme A/chemistry , Chlorophyll/analogs & derivatives , Chlorophyll/chemistry , Diospyros/chemistry , Plant Extracts/pharmacology , Plant Extracts/pharmacokinetics , Plant Leaves/chemistry , Sterol O-Acyltransferase/antagonists & inhibitors , Sterol O-Acyltransferase/isolation & purification , Acyl Coenzyme A/isolation & purification , Acyl Coenzyme A/pharmacokinetics , Animals , Chlorophyll/isolation & purification , Chlorophyll/pharmacokinetics , Chromatography, High Pressure Liquid/methods , Dose-Response Relationship, Drug , Drugs, Chinese Herbal , Gas Chromatography-Mass Spectrometry , Korea , Methanol , Methyl Ethers , Microsomes, Liver/enzymology , Molecular Structure , Rats , Sterol O-Acyltransferase/pharmacokinetics
SELECTION OF CITATIONS
SEARCH DETAIL