Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 34
1.
Sci Rep ; 13(1): 13074, 2023 08 11.
Article En | MEDLINE | ID: mdl-37567910

Nephritis is common in systemic lupus erythematosus patients and is associated with hyper-activation of immune and renal cells. Although mesenchymal stem cells (MSCs) ameliorate nephritis by inhibiting T and B cells, whether MSCs directly affect renal cells is unclear. To address this issue, we examined the direct effect of MSCs on renal cells with a focus on chemokines. We found that expression of CCL2, CCL3, CCL4, CCL5, CCL8, CCL19, and CXCL10 increased 1.6-5.6-fold in the kidney of lupus-prone MRL.Faslpr mice with advancing age from 9 to 16 weeks. Although MSCs inhibited the increase in the expression of most chemokines by 52-95%, they further increased CCL8 expression by 290%. Using renal cells, we next investigated how MSCs enhanced CCL8 expression. CCL8 was expressed by podocytes, but not by tubular cells. MSCs enhanced CCL8 expression by podocytes in a contact-dependent manner, which was proved by transwell assay and blocking with anti-VCAM-1 antibody. Finally, we showed that CCL8 itself activated MSCs to produce more immunosuppressive factors (IL-10, IDO, TGF-ß1, and iNOS) and to inhibit more strongly IFN-γ production by T cells. Taken together, our data demonstrate that MSCs activate podocytes to produce CCL8 in a contact-dependent manner and conversely, podocyte-derived CCL8 might potentiate immunosuppressive activity of MSCs in a paracrine fashion. Our study documents a previously unrecognized therapeutic mechanism of MSCs in nephritis.


Lupus Erythematosus, Systemic , Lupus Nephritis , Mesenchymal Stem Cells , Podocytes , Animals , Mice , Chemokines/metabolism , Mice, Inbred MRL lpr , Podocytes/metabolism
2.
Arch Pharm Res ; 45(1): 1-10, 2022 Jan.
Article En | MEDLINE | ID: mdl-34905179

The susceptibility of cancer cells to natural killer (NK) cell-mediated cytotoxicity depends on the balance of activating and inhibitory ligands expressed on their surface. Although many types of cancer cells are killed by NK cells, non-small-cell lung cancer (NSCLC) cells are relatively resistant to NK cell-mediated cytotoxicity. In this study, we showed that several NSCLC cell lines have differential sensitivity to NK cell-mediated cytotoxicity: NCI-H522 cells were highly sensitive, but A549, NCI-H23, NCI-H1915, and NCI-H1299 were resistant. Among activating ligands such as CD48, HLA-A/B/G, ICAM-1, MICA/B, and ULBPs, only CD48 rendered NCI-H522 cells susceptible to NK cell-mediated cytotoxicity, which was proved by using CD48 siRNA and neutralizing antibody. CD48-positive NCI-H522 cells established a more stable contact with NK cells than did CD48-negative A549 and CD48 siRNA cell-transfected NCI-H522 cells. Taken together, these data demonstrate that CD48-positive NSCLC cells might be susceptible to NK cell-mediated cytotoxicity, which provide information on how to stratify NSCLC patients potentially responsive to NK-cell therapy.


CD48 Antigen/metabolism , Carcinoma, Non-Small-Cell Lung/immunology , Killer Cells, Natural/physiology , Lung Neoplasms/immunology , Blotting, Western , CD48 Antigen/immunology , Carcinoma, Non-Small-Cell Lung/metabolism , Cell Line, Tumor , Flow Cytometry , Humans , Killer Cells, Natural/immunology , Lung Neoplasms/metabolism , Polymerase Chain Reaction
3.
Theranostics ; 10(22): 10186-10199, 2020.
Article En | MEDLINE | ID: mdl-32929342

Rationale: Systemic lupus erythematosus (SLE) is a multi-organ autoimmune disease characterized by autoantibody production by hyper-activated B cells. Although mesenchymal stem cells (MSCs) ameliorate lupus symptoms by inhibiting T cells, whether they inhibit B cells has been controversial. Here we address this issue and reveal how to prime MSCs to inhibit B cells and improve the efficacy of MSCs in SLE. Methods: We examined the effect of MSCs on purified B cells in vitro and the therapeutic efficacy of MSCs in lupus-prone MRL.Faslpr mice. We screened chemicals for their ability to activate MSCs to inhibit B cells. Results: Mouse bone marrow-derived MSCs inhibited mouse B cells in a CXCL12-dependent manner, whereas human bone marrow-derived MSCs (hMSCs) did not inhibit human B (hB) cells. We used a chemical approach to overcome this hurdle and found that phorbol myristate acetate (PMA), phorbol 12,13-dibutyrate, and ingenol-3-angelate rendered hMSCs capable of inhibiting IgM production by hB cells. As to the mechanism, PMA-primed hMSCs attracted hB cells in a CXCL10-dependent manner and induced hB cell apoptosis in a PD-L1-dependent manner. Finally, we showed that PMA-primed hMSCs were better than naïve hMSCs at ameliorating SLE progression in MRL.Faslpr mice. Conclusion: Taken together, our data demonstrate that phorbol esters might be good tool compounds to activate MSCs to inhibit B cells and suggest that our chemical approach might allow for improvements in the therapeutic efficacy of hMSCs in SLE.


B-Lymphocytes/drug effects , Lupus Erythematosus, Systemic/drug therapy , Mesenchymal Stem Cells/drug effects , Phorbol Esters/pharmacology , Animals , Apoptosis/drug effects , Cells, Cultured , Female , Humans , Male , Mesenchymal Stem Cell Transplantation , Mice , Mice, Inbred C3H , T-Lymphocytes/drug effects
4.
Stem Cells Int ; 2020: 5617192, 2020.
Article En | MEDLINE | ID: mdl-32215018

Systemic lupus erythematosus (SLE) is an autoimmune disease, which is characterized by hyperactivation of T and B cells. Human mesenchymal stem cells (hMSCs) ameliorate the progression of SLE in preclinical studies using lupus-prone MRL.Fas lpr mice. However, whether hMSCs inhibit the functions of xenogeneic mouse T and B cells is not clear. To address this issue, we examined the in vitro effects of hMSCs on T and B cells isolated from MRL.Fas lpr mice. Naïve hMSCs inhibited the functions of T cells but not B cells. hMSCs preconditioned with IFN-γ (i) inhibited the proliferation of and IgM production by B cells, (ii) attracted B cells for cell-cell interactions in a CXCL10-dependent manner, and (iii) inhibited B cells by producing indoleamine 2,3-dioxygenase. In summary, our data demonstrate that hMSCs exert therapeutic activity in mice in three steps: first, naïve hMSCs inhibit the functions of T cells, hMSCs are then activated by IFN-γ, and finally, they inhibit B cells.

5.
Immune Netw ; 19(5): e36, 2019 Oct.
Article En | MEDLINE | ID: mdl-31720047

Mesenchymal stem cells (MSCs) ameliorate the renal injury in Adriamycin (ADR)-induced nephropathy, but the mechanisms underlying their efficacy remain incompletely understood. In this study, we demonstrated that MSCs increased the survival, recovered body weight loss, and decreased proteinuria and serum creatinine levels in ADR-treated mice. MSCs also prevented podocyte damage and renal fibrosis by decreasing the expression of fibronectin, collagen 1α1, and α-smooth muscle actin. From a mechanistic perspective, MSCs inhibited renal inflammation by lowering the expression of CCL4, CCL7, CCL19, IFN-α/ß, TGF-ß, TNF-α, and chitinase 3-like 1. In summary, our data demonstrate that MSCs improve renal functions by inhibiting renal inflammation in ADR-induced nephropathy.

6.
Int Immunopharmacol ; 67: 366-371, 2019 Feb.
Article En | MEDLINE | ID: mdl-30583235

Natural killer (NK) cell killing of melanoma cells involves perforin-mediated delivery of granzymes from NK cells to cancer cells; however, how melanoma cells die remains poorly characterized. Here, we examined the dying process of melanoma cells by using time-lapse imaging. Upon contact with NK cells, B16-F10 cells rounded and most of them showed membrane rupture (98 min); however, B16 parent cells showed writhing and delayed membrane rupture (235 min). This morphological difference depended on the expression levels of myosin regulatory light chain 9 (MYL9) but not activating ligands (CD112, CD155, Rae-1, and MULT-1), SPI, FasL, or PD-L1. Taken together, our data show that melanoma cells show two distinct types of morphological changes upon contact with NK cells and suggest that a strategy to decrease MYL9 expression by melanoma cells may improve the efficacy of NK cell-based immunotherapy.


Killer Cells, Natural/physiology , Melanoma, Experimental , Animals , Cell Line, Tumor , Gene Expression Regulation, Neoplastic , Humans , Mice , Mice, Inbred C57BL , Myosin Light Chains/genetics , Myosin Light Chains/metabolism
7.
Int Immunopharmacol ; 63: 66-73, 2018 Oct.
Article En | MEDLINE | ID: mdl-30075430

Natural killer (NK) cells eliminate cancer cells in a contact-dependent manner. However, how NK cells find cancer cells remain unclear. Here, using time-lapse imaging, we investigated how individual NK cells migrate toward cancer cells. Although naïve B16F10 cancer cells produce low levels of chemokines, IFN-γ-treated B16F10 cells secreted high levels of CXCL10, low levels of CCL5, but did not secrete CCL2, CCL7, or CXCL12. Wild-type NK cells migrated well toward cancer cells and killed them, whereas NK cells deficient in CXCR3 did not. CXCR3-deficient NK cells also showed slower migration speed than did wild-type NK cells. Taken together, our data show that NK cells find cancer cells, at least in part, by sensing CXCL10 produced by cancer cells and suggest that a strategy to increase CXCL10 secretion by cancer cells may improve the efficacy of NK cell-based immunotherapy.


Chemokine CXCL10/immunology , Chemotaxis, Leukocyte/immunology , Killer Cells, Natural/immunology , Melanoma, Experimental/immunology , Receptors, CXCR3/immunology , Animals , Cell Line, Tumor , Chemokine CXCL10/genetics , Mice, Inbred C57BL , Mice, Knockout , Receptors, CXCR3/genetics
8.
Stem Cells Int ; 2018: 4273107, 2018.
Article En | MEDLINE | ID: mdl-30057623

The combination of immunosuppressants and mesenchymal stem cells (MSCs) is a promising therapeutic strategy for systemic lupus erythematosus, since this approach reduces doses of immunosuppressants while maintaining the same therapeutic outcome. However, it is unavoidable for MSCs to be exposed to immunosuppressants. Here, we examined the combination effect of prednisone (PD) or mycophenolate mofetil (MMF) and MSCs. We showed that PD or MMF in combination with MSCs showed better therapeutic effect than single therapy in lupus-prone MRL.Faslpr mice, as assessed by using the following readouts: prolongation of survival, decrease in anti-dsDNA and total IgG levels in serum, decrease in cytokine gene expression in spleen cells, and decrease in inflammatory cell infiltration into the kidney. In vitro, immunosuppressants and MSCs inhibited T cell proliferation in a synergistic manner. However, immunosuppressants did not affect MSC viability and functions such as TGF-ß1 and PGE2 production, migration, and immunosuppressive capacity. In summary, our study demonstrates that a combination of immunosuppressants and MSCs is a good strategy to reduce the side effects of PD and MMF without the loss of therapeutic outcome.

9.
Oncoimmunology ; 6(8): e1338994, 2017.
Article En | MEDLINE | ID: mdl-28920003

CD226 is an activating receptor expressed on natural killer (NK) cells, CD8+ T cells, and other immune cells. Upon binding to its ligands expressed on target cells, CD226 activates intracellular signaling that triggers cytokine production and degranulation in NK cells. However, the role of CD226 in contact dynamics between NK and cancer cells has remained unclear. Our time-lapse images showed that individual wild-type CD226+ NK cells contacted B16F10 melanoma cells for 23.7 min, but Cd226-/- NK cells only for 12.8 min, although both NK cell subsets showed equal contact frequency over 4 h. On the surface of B16F10 cells, CD226+ cells stayed at the same site with oscillating movement (named stable contact), while Cd226-/- NK cells moved around at a velocity of 4 µm/min (named unstable contact). Consequently, Cd226-/- NK cells did not kill B16F10 cells in vitro and did not inhibit their metastasis into the lung in vivo. Taken together, our data demonstrate that CD226 enables prolonged stable interaction between NK and cancer cells, which is needed for efficient killing of cancer cells.

10.
Sci Rep ; 7: 41258, 2017 01 24.
Article En | MEDLINE | ID: mdl-28117437

Systemic lupus erythematosus (SLE) is a multi-organ autoimmune disease characterized by autoantibody production. Mesenchymal stem cells (MSCs) ameliorate SLE symptoms by targeting T cells, whereas the mechanisms of their efficacy remain incompletely understood. In this study, we show that transfer of human MSCs increased MRL.Faslpr mouse survival, decreased T cell infiltration in the kidneys, and reduced T cell cytokine expression. In vitro, allogeneic mouse MSCs inhibited MRL.Faslpr T cell proliferation and cytokine production. Time-lapse imaging revealed that MSCs recruited MRL.Faslpr T cells establishing long-lasting cellular contacts by enhancing T cell VCAM-1 expression in a CCL2-dependent manner. In contrast, CCL2 deficient MSCs did not induce T cell migration and VCAM-1 expression, resulting in insufficient cell-cell contact. Consequently, CCL2 deficient MSCs did not inhibit IFN-γ production by T cells and upon transfer no longer prolonged survival of MRL.Faslpr mice. Taken together, our imaging study demonstrates that CCL2 enables the prolonged MSC-T cell interactions needed for sufficient suppression of autoreactive T cells and helps to understand how MSCs ameliorate symptoms in lupus-prone MRL.Faslpr mice.


Cell Communication , Chemokine CCL2/deficiency , Lupus Erythematosus, Systemic/immunology , Lupus Erythematosus, Systemic/pathology , Mesenchymal Stem Cells/metabolism , T-Lymphocytes/metabolism , Animals , Cell Movement , Humans , Mice, Inbred C57BL , Mice, Inbred MRL lpr , Solubility , Vascular Cell Adhesion Molecule-1/metabolism
11.
Cancer Immunol Immunother ; 66(2): 193-202, 2017 Feb.
Article En | MEDLINE | ID: mdl-27888293

Cytotoxicity of cytokine-induced killer (CIK) cells depends mainly on their encounters with target cells, but how many CIK cells are required to kill an individual cancer cell is unknown. Here we used time-lapse imaging to quantify the critical effector cell number required to kill an individual target cell. CIK cells killed MHC-I-negative and MHC-I-positive cancer cells, but natural killer (NK) cells destroyed MHC-I-negative cells only. The average threshold number of CIK cells required to kill an individual cancer cell was 6.7 for MHC-I-negative cells and 6.9 for MHC-I-positive cells. That of NK cells was 2.4 for MHC-I-negative cells. Likely due to the higher threshold numbers, killing by CIK cells was delayed in comparison with NK cells: 40% of MHC-negative target cells were killed after 5 h when co-cultured with CIK cells and after 2 h with NK cells. Our data have implications for the rational design of CIK cell-based immunotherapy of cancer patients.


Cytokine-Induced Killer Cells/immunology , Immunotherapy, Adoptive/methods , Melanoma, Experimental/immunology , Melanoma, Experimental/therapy , Animals , Cytokine-Induced Killer Cells/pathology , Disease Models, Animal , Female , Humans , Melanoma, Experimental/pathology , Mice , Mice, Inbred C57BL
12.
Int Immunopharmacol ; 39: 71-78, 2016 Oct.
Article En | MEDLINE | ID: mdl-27449326

Curdlan, a ß-1,3-glucan isolated from Alcaligenes faecalis, is an agonist of dectin-1 in various immune cells, including dendritic cells (DCs). However, whether curdlan also activates DCs through other receptors remains unknown. In this study, we found that curdlan activates DCs through dectin-1 and toll-like receptor 4 (TLR4). Curdlan increased the expression levels of surface molecules (CD40, CD80, CD86, and MHC-I/II), the production of cytokines (IL-12, IL-1ß, TNF-α, and IFN-ß), migration toward MIP-3ß, and allogeneic T cell stimulation activity of DCs. Curdlan increased the phosphorylation of Syk, Raf-1, Akt, MAPKs, IKK, and NF-κB p65 in DCs. However, curdlan only slightly activated DCs transfected with small interfering RNAs against dectin-1 or TLR4 and C3H/HeJ DCs, which have non-functional TLR4, in comparison with control DCs. Curdlan increased antitumor activity of DCs in a syngeneic tumor model. In summary, our data show that curdlan activates DCs through dectin-1 and TLR4 signaling and the combination of curdlan and DCs efficiently inhibit tumor growth in mice.


Alcaligenes faecalis/immunology , Antineoplastic Agents/therapeutic use , Dendritic Cells/drug effects , Lectins, C-Type/metabolism , Skin Neoplasms/drug therapy , T-Lymphocytes/immunology , Toll-Like Receptor 4/metabolism , beta-Glucans/therapeutic use , Animals , Cell Differentiation/drug effects , Cell Differentiation/genetics , Cell Movement/drug effects , Cytokines/metabolism , Dendritic Cells/physiology , Inflammation Mediators/metabolism , Lectins, C-Type/genetics , Lymphocyte Activation , Melanoma, Experimental , Mice , Mice, Inbred C3H , Mice, Inbred C57BL , Mice, Knockout , RNA, Small Interfering/genetics , Toll-Like Receptor 4/genetics
13.
Immune Netw ; 16(2): 99-108, 2016 Apr.
Article En | MEDLINE | ID: mdl-27162526

Colorectal cancer is the third leading cancer worldwide. Although incidence and mortality of colorectal cancer are gradually decreasing in the US, patients with metastatic colorectal cancer have poor prognosis with an estimated 5-year survival rate of less than 10%. Over the past decade, advances in combination chemotherapy regimens for colorectal cancer have led to significant improvement in progression-free and overall survival. However, patients with metastatic disease gain little clinical benefit from conventional therapy, which is associated with grade 3~4 toxicity with negative effects on quality of life. In previous clinical studies, cell-based immunotherapy using dendritic cell vaccines and sentinel lymph node T cell therapy showed promising therapeutic results for metastatic colorectal cancer. In our preclinical and previous clinical studies, cytokine-induced killer (CIK) cells treatment for colorectal cancer showed favorable responses without toxicities. Here, we review current treatment options for colorectal cancer and summarize available clinical studies utilizing cell-based immunotherapy. Based on these studies, we recommend the use CIK cell therapy as a promising therapeutic strategy for patients with metastatic colorectal cancer.

14.
Cancer Lett ; 378(2): 142-9, 2016 08 10.
Article En | MEDLINE | ID: mdl-27216980

The antitumor activity of cytokine-induced killer (CIK) cells can be increased by co-culturing them with tumor lysate-pulsed dendritic cells (tDCs); this phenomenon has been studied mainly at the population level. Using time-lapse imaging, we examined how CIK cells gather information from tDCs at the single-cell level. tDCs highly expressed CCL5, which bound CCR5 expressed on CIK cells. tDCs strongly induced migration of Ccr5(+/+) CIK cells, but not that of Ccr5(-/-) CIK cells or Ccr5(+/+) CIK cells treated with the CCR5 antagonist Maraviroc. Individual tDCs contacted Ccr5(+/+) CIK cells more frequently and lengthily than with Ccr5(-/-) CIK cells. Consequently, tDCs increased the antitumor activity of Ccr5(+/+) CIK cells in vitro and in vivo, but did not increase that of Ccr5(-/-) CIK cells. Taken together, our data provide insight into the mechanism of CIK cell activation by tDCs at the single-cell level.


Cell Communication , Cytokine-Induced Killer Cells/metabolism , Cytotoxicity, Immunologic , Dendritic Cells/metabolism , Melanoma, Experimental/metabolism , Receptors, CCR5/metabolism , Signal Transduction , Animals , Cells, Cultured , Chemokine CCL5/immunology , Chemokine CCL5/metabolism , Chemotaxis, Leukocyte , Coculture Techniques , Cytokine-Induced Killer Cells/immunology , Cytokine-Induced Killer Cells/transplantation , Dendritic Cells/immunology , Dendritic Cells/transplantation , Genotype , Melanoma, Experimental/immunology , Melanoma, Experimental/pathology , Mice, Inbred C57BL , Mice, Knockout , Phenotype , RNA Interference , Receptors, CCR5/deficiency , Receptors, CCR5/genetics , Time Factors , Time-Lapse Imaging , Transfection , Tumor Burden
15.
Immune Netw ; 15(2): 58-65, 2015 Apr.
Article En | MEDLINE | ID: mdl-25922594

Melanoma is the most aggressive skin cancer and its incidence is gradually increasing worldwide. Patients with metastatic melanoma have a very poor prognosis (estimated 5-year survival rate of <16%). In the last few years, several drugs have been approved for malignant melanoma, such as tyrosine kinase inhibitors and immune checkpoint blockades. Although new therapeutic agents have improved progression-free and overall survival, their use is limited by drug resistance and drug-related toxicity. At the same time, adoptive cell therapy of metastatic melanoma with tumor-infiltrating lymphocytes has shown promising results in preclinical and clinical studies. In this review, we summarize the currently available drugs for treatment of malignant melanoma. In addition, we suggest cytokine-induced killer (CIK) cells as another candidate approach for adoptive cell therapy of melanoma. Our preclinical study and several previous studies have shown that CIK cells have potent anti-tumor activity against melanomas in vitro and in an in vivo human tumor xenograft model without any toxicity.

16.
J Ethnopharmacol ; 166: 92-101, 2015 May 26.
Article En | MEDLINE | ID: mdl-25792017

ETHNOPHARMACOLOGICAL RELEVANCE: Saururus chinensis is a medicinal plant used to treat jaundice, pneumonia, edema, fever, and several inflammatory diseases. Saucerneol D (SD), a lignan constituent of this plant, has antioxidant, anti-asthmatic, and anti-inflammatory activities. SD has been previously reported to inhibit the pro-inflammatory responses of RAW264.7 cells and primary mast cells. In this study, we investigated the effect of SD on the functions of dendritic cells (DCs). MATERIALS AND METHODS: SD was isolated from methanol extract of the roots of S. chinensis. Bone marrow-derived DCs were used as target cells. The effects of SD on the following DC functions were examined: surface molecule expression, cytokine expression, migration, allogenic T cell activation, heme oxygenase-1 expression, and Toll-like receptor 4 signaling. RESULTS: In lipopolysaccharide (LPS)-treated DCs, SD inhibited the expression of cell surface molecules (MHC I/II, CD40, CD80, and CD86), the production of inflammatory mediators (nitric oxide, IL-12, IL-1ß, and TNF-α), and allogenic T cell activation capacity. SD also inhibited DC migration toward MIP-3ß by down-regulating CCR7 expression. SD attenuated LPS-induced activation of NF-κB and MAPK signaling in DCs, but did not directly inhibit kinase activities of IRAK1, IRAK4, TAK1, or IKKß in enzymatic assays. SD did not inhibit LPS binding to myeloid differentiation protein-2, co-receptor of TLR4. SD increased the production of reactive oxygen species, Nrf-2, and heme oxygenase (HO)-1, which degrades the heme to immunosuppressive carbon monoxide and biliverdin, which may underlie the anti-inflammatory effects in SD-treated DCs. CONCLUSIONS: Taken together, these data suggest that SD suppresses LPS-induced activation of DCs through the induction of HO-1, but not by directly affecting Toll-like receptor 4 signaling.


Dendritic Cells/drug effects , Heme Oxygenase-1/metabolism , Lignans/pharmacology , Signal Transduction/drug effects , Toll-Like Receptor 4/antagonists & inhibitors , Animals , Anti-Inflammatory Agents/pharmacology , Cell Movement/drug effects , Cytokines/metabolism , Dendritic Cells/metabolism , Female , Lymphocyte Activation/drug effects , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Plant Roots/chemistry , Saururaceae/chemistry , T-Lymphocytes/drug effects , T-Lymphocytes/metabolism
17.
Arch Pharm Res ; 38(5): 813-25, 2015.
Article En | MEDLINE | ID: mdl-25074039

4-O-methylhonokiol (MH) is known to inhibit inflammation by partially understood mechanisms. Here, the anti-inflammatory mechanisms of MH were examined using enzymatic, cellular, and animal assays. In enzymatic assays, MH inhibited COX-2 activity with an IC50 of 0.062 µM, and also COX-1 with an IC50 of 2.4 µM. In cellular assays, MH was immunotoxic above 10 µM. At non-toxic concentrations (below 3 µM), MH strongly inhibited COX-2-mediated prostaglandin production with an IC50 of 0.1 µM, whereas did not or slightly affect other functions of B cells, T cells, dendritic cells, and macrophages. In an animal model, MH inhibited the increase in footpad thickness and popliteal lymph node weight in zymosan-injected mice. When analyzed the draining pLNs of zymosan-injected mice on day 5, MH inhibited the overall inflammatory responses. However, MH inhibited cyclooxygenase (COX)-2-mediated prostaglandin production without affecting tumor necrosis factor-α production in inflamed tissues within 6 h after zymosan injection. In summary, our data suggest that COX-2 may be a direct anti-inflammatory target of MH in vitro and in vivo.


Anti-Inflammatory Agents, Non-Steroidal/metabolism , Biphenyl Compounds/metabolism , Cyclooxygenase 2/metabolism , Disease Models, Animal , Drug Delivery Systems/methods , Lignans/metabolism , Zymosan/toxicity , Animals , Anti-Inflammatory Agents, Non-Steroidal/administration & dosage , Biphenyl Compounds/administration & dosage , Dose-Response Relationship, Drug , Female , Inflammation/chemically induced , Inflammation/drug therapy , Inflammation/enzymology , Lignans/administration & dosage , Macrophages/drug effects , Macrophages/metabolism , Mice , Mice, Inbred C57BL
18.
Int Immunopharmacol ; 22(2): 400-8, 2014 Oct.
Article En | MEDLINE | ID: mdl-25091622

Sesquiterpenoid tussilagone (TUS) has a variety of pharmacological activities, such as anti-oxidant, anti-cancer, and anti-inflammatory activities. In this study, we investigated the effects of TUS on dendritic cell (DC) functions and the underlying mechanisms. TUS inhibited lipopolysaccharide (LPS)-induced activation of DCs, as shown by decrease in surface molecule expression, cytokine production, cell migration, and allo-T cell activation. In addition, TUS inhibited LPS-induced activation of NF-κB, MAPKs, and IRF-3 signalings in DCs, although it did not directly affect kinase activities of IRAK1/4, TAK1, and IKK, which suggests that TUS might indirectly inhibit TLR signaling in DCs. As a critical mechanism, we showed that TUS activated heme oxygenase-1 (HO-1), which degrades heme to immunosuppressive products, such as carbon monoxide and bilirubin. HO-1 inhibitor reversed the inhibitory activity of TUS in DCs. In conclusion, this study suggests that TUS inhibits DC function through the induction of HO-1.


Dendritic Cells/drug effects , Heme Oxygenase-1/biosynthesis , Membrane Proteins/biosynthesis , Sesquiterpenes/pharmacology , Animals , Cytokines/genetics , Cytokines/metabolism , Dendritic Cells/immunology , Dendritic Cells/metabolism , Female , Lymphocyte Culture Test, Mixed , Mice, Inbred BALB C , Mice, Inbred C57BL , Nitric Oxide/metabolism , Protein Kinases/metabolism , Reactive Oxygen Species/metabolism , Spleen/cytology , T-Lymphocytes/drug effects , T-Lymphocytes/immunology , Toll-Like Receptor 4/antagonists & inhibitors
19.
Food Chem Toxicol ; 72: 212-20, 2014 Oct.
Article En | MEDLINE | ID: mdl-25019244

Dendritic cell (DC) maturation is critical for initiation of the adoptive immune response. DC maturation is often attenuated in several pathological conditions including cancer. In this study, we report the effect of Platycodon grandiflorum polysaccharide (PG) on DC maturation. PG induced phenotypic maturation of DCs, as proved by the increase in the expression of CD40, CD80, CD86, and major histocompatibility complex (MHC)-I/II on the cell surface. PG also induced functional maturation of DCs, as proved by elevated production of interleukin (IL)-12, tumor necrosis factor-α, IL-1ß, IL-6, IL-10, and interferon-ß, and by enhanced allogeneic T cell stimulation ability of PG-treated DCs. PG efficiently induced maturation of DCs from C3H/HeN mice, which have normal Toll-like receptor-4 (TLR4), but not that of DCs from C3H/HeJ mice, which have mutated TLR4, suggesting that TLR4 might be one of the PG receptors in DCs. In line with TLR4 activation, PG increased the phosphorylation of ERK, p38, and JNK, and the nuclear translocation of p-c-Jun, p-CREB, and c-Fos. PG also activated NF-κB signaling, as evidenced by degradation of IκBα/ß and nuclear translocation of p65 and p50. In summary, our data suggest that PG induces DC maturation by activating MAPK and NF-κB signaling downstream of TLR4.


Cell Differentiation/drug effects , Dendritic Cells/drug effects , Platycodon/chemistry , Polysaccharides/pharmacology , Toll-Like Receptor 4/metabolism , Animals , Cell Line , Cell Proliferation/drug effects , Dendritic Cells/metabolism , Endocytosis/drug effects , Female , Interferon-beta/metabolism , Interleukin-10/metabolism , Interleukin-12/metabolism , Interleukin-1beta/metabolism , Interleukin-6/metabolism , Mice , Mice, Inbred BALB C , Mice, Inbred C3H , Mice, Inbred C57BL , NF-kappa B/metabolism , Nitrites/metabolism , Phosphorylation/drug effects , Signal Transduction , T-Lymphocytes/drug effects , T-Lymphocytes/metabolism , Toll-Like Receptor 4/genetics , Tumor Necrosis Factor-alpha/metabolism
20.
Immune Netw ; 14(2): 81-8, 2014 Apr.
Article En | MEDLINE | ID: mdl-24851097

Mesenchymal stem cells (MSCs) are present in diverse tissues and organs, including bone marrow, umbilical cord, adipose tissue, and placenta. MSCs can expand easily in vitro and have regenerative stem cell properties and potent immunoregulatory activity. They inhibit the functions of dendritic cells, B cells, and T cells, but enhance those of regulatory T cells by producing immunoregulatory molecules such as transforming growth factor-ß, hepatic growth factors, prostaglandin E2, interleukin-10, indolamine 2,3-dioxygenase, nitric oxide, heme oxygenase-1, and human leukocyte antigen-G. These properties make MSCs promising therapeutic candidates for the treatment of autoimmune diseases. Here, we review the preclinical studies of MSCs in animal models for systemic lupus erythematosus, rheumatoid arthritis, Crohn's disease, and experimental autoimmune encephalomyelitis, and summarize the underlying immunoregulatory mechanisms.

...