Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Sci Rep ; 13(1): 13074, 2023 08 11.
Artigo em Inglês | MEDLINE | ID: mdl-37567910

RESUMO

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.


Assuntos
Lúpus Eritematoso Sistêmico , Nefrite Lúpica , Células-Tronco Mesenquimais , Podócitos , Animais , Camundongos , Quimiocinas/metabolismo , Camundongos Endogâmicos MRL lpr , Podócitos/metabolismo
2.
Arch Pharm Res ; 45(1): 1-10, 2022 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-34905179

RESUMO

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.


Assuntos
Antígeno CD48/metabolismo , Carcinoma Pulmonar de Células não Pequenas/imunologia , Células Matadoras Naturais/fisiologia , Neoplasias Pulmonares/imunologia , Western Blotting , Antígeno CD48/imunologia , Carcinoma Pulmonar de Células não Pequenas/metabolismo , Linhagem Celular Tumoral , Citometria de Fluxo , Humanos , Células Matadoras Naturais/imunologia , Neoplasias Pulmonares/metabolismo , Reação em Cadeia da Polimerase
3.
Theranostics ; 10(22): 10186-10199, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32929342

RESUMO

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.


Assuntos
Linfócitos B/efeitos dos fármacos , Lúpus Eritematoso Sistêmico/tratamento farmacológico , Células-Tronco Mesenquimais/efeitos dos fármacos , Ésteres de Forbol/farmacologia , Animais , Apoptose/efeitos dos fármacos , Células Cultivadas , Feminino , Humanos , Masculino , Transplante de Células-Tronco Mesenquimais , Camundongos , Camundongos Endogâmicos C3H , Linfócitos T/efeitos dos fármacos
4.
Int Immunopharmacol ; 67: 366-371, 2019 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-30583235

RESUMO

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.


Assuntos
Células Matadoras Naturais/fisiologia , Melanoma Experimental , Animais , Linhagem Celular Tumoral , Regulação Neoplásica da Expressão Gênica , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Cadeias Leves de Miosina/genética , Cadeias Leves de Miosina/metabolismo
5.
Int Immunopharmacol ; 63: 66-73, 2018 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-30075430

RESUMO

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.


Assuntos
Quimiocina CXCL10/imunologia , Quimiotaxia de Leucócito/imunologia , Células Matadoras Naturais/imunologia , Melanoma Experimental/imunologia , Receptores CXCR3/imunologia , Animais , Linhagem Celular Tumoral , Quimiocina CXCL10/genética , Camundongos Endogâmicos C57BL , Camundongos Knockout , Receptores CXCR3/genética
6.
Oncoimmunology ; 6(8): e1338994, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28920003

RESUMO

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.

7.
Sci Rep ; 7: 41258, 2017 01 24.
Artigo em Inglês | MEDLINE | ID: mdl-28117437

RESUMO

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.


Assuntos
Comunicação Celular , Quimiocina CCL2/deficiência , Lúpus Eritematoso Sistêmico/imunologia , Lúpus Eritematoso Sistêmico/patologia , Células-Tronco Mesenquimais/metabolismo , Linfócitos T/metabolismo , Animais , Movimento Celular , Humanos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos MRL lpr , Solubilidade , Molécula 1 de Adesão de Célula Vascular/metabolismo
8.
Cancer Immunol Immunother ; 66(2): 193-202, 2017 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-27888293

RESUMO

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.


Assuntos
Células Matadoras Induzidas por Citocinas/imunologia , Imunoterapia Adotiva/métodos , Melanoma Experimental/imunologia , Melanoma Experimental/terapia , Animais , Células Matadoras Induzidas por Citocinas/patologia , Modelos Animais de Doenças , Feminino , Humanos , Melanoma Experimental/patologia , Camundongos , Camundongos Endogâmicos C57BL
9.
Int Immunopharmacol ; 39: 71-78, 2016 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-27449326

RESUMO

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.


Assuntos
Alcaligenes faecalis/imunologia , Antineoplásicos/uso terapêutico , Células Dendríticas/efeitos dos fármacos , Lectinas Tipo C/metabolismo , Neoplasias Cutâneas/tratamento farmacológico , Linfócitos T/imunologia , Receptor 4 Toll-Like/metabolismo , beta-Glucanas/uso terapêutico , Animais , Diferenciação Celular/efeitos dos fármacos , Diferenciação Celular/genética , Movimento Celular/efeitos dos fármacos , Citocinas/metabolismo , Células Dendríticas/fisiologia , Mediadores da Inflamação/metabolismo , Lectinas Tipo C/genética , Ativação Linfocitária , Melanoma Experimental , Camundongos , Camundongos Endogâmicos C3H , Camundongos Endogâmicos C57BL , Camundongos Knockout , RNA Interferente Pequeno/genética , Receptor 4 Toll-Like/genética
10.
Immune Netw ; 16(2): 99-108, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-27162526

RESUMO

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.

11.
Cancer Lett ; 378(2): 142-9, 2016 08 10.
Artigo em Inglês | MEDLINE | ID: mdl-27216980

RESUMO

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.


Assuntos
Comunicação Celular , Células Matadoras Induzidas por Citocinas/metabolismo , Citotoxicidade Imunológica , Células Dendríticas/metabolismo , Melanoma Experimental/metabolismo , Receptores CCR5/metabolismo , Transdução de Sinais , Animais , Células Cultivadas , Quimiocina CCL5/imunologia , Quimiocina CCL5/metabolismo , Quimiotaxia de Leucócito , Técnicas de Cocultura , Células Matadoras Induzidas por Citocinas/imunologia , Células Matadoras Induzidas por Citocinas/transplante , Células Dendríticas/imunologia , Células Dendríticas/transplante , Genótipo , Melanoma Experimental/imunologia , Melanoma Experimental/patologia , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fenótipo , Interferência de RNA , Receptores CCR5/deficiência , Receptores CCR5/genética , Fatores de Tempo , Imagem com Lapso de Tempo , Transfecção , Carga Tumoral
12.
Immune Netw ; 15(2): 58-65, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25922594

RESUMO

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.

13.
Arch Pharm Res ; 38(5): 813-25, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25074039

RESUMO

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.


Assuntos
Anti-Inflamatórios não Esteroides/metabolismo , Compostos de Bifenilo/metabolismo , Ciclo-Oxigenase 2/metabolismo , Modelos Animais de Doenças , Sistemas de Liberação de Medicamentos/métodos , Lignanas/metabolismo , Zimosan/toxicidade , Animais , Anti-Inflamatórios não Esteroides/administração & dosagem , Compostos de Bifenilo/administração & dosagem , Relação Dose-Resposta a Droga , Feminino , Inflamação/induzido quimicamente , Inflamação/tratamento farmacológico , Inflamação/enzimologia , Lignanas/administração & dosagem , Macrófagos/efeitos dos fármacos , Macrófagos/metabolismo , Camundongos , Camundongos Endogâmicos C57BL
14.
Int Immunopharmacol ; 22(2): 400-8, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-25091622

RESUMO

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.


Assuntos
Células Dendríticas/efeitos dos fármacos , Heme Oxigenase-1/biossíntese , Proteínas de Membrana/biossíntese , Sesquiterpenos/farmacologia , Animais , Citocinas/genética , Citocinas/metabolismo , Células Dendríticas/imunologia , Células Dendríticas/metabolismo , Feminino , Teste de Cultura Mista de Linfócitos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Óxido Nítrico/metabolismo , Proteínas Quinases/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Baço/citologia , Linfócitos T/efeitos dos fármacos , Linfócitos T/imunologia , Receptor 4 Toll-Like/antagonistas & inibidores
15.
Food Chem Toxicol ; 72: 212-20, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-25019244

RESUMO

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.


Assuntos
Diferenciação Celular/efeitos dos fármacos , Células Dendríticas/efeitos dos fármacos , Platycodon/química , Polissacarídeos/farmacologia , Receptor 4 Toll-Like/metabolismo , Animais , Linhagem Celular , Proliferação de Células/efeitos dos fármacos , Células Dendríticas/metabolismo , Endocitose/efeitos dos fármacos , Feminino , Interferon beta/metabolismo , Interleucina-10/metabolismo , Interleucina-12/metabolismo , Interleucina-1beta/metabolismo , Interleucina-6/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C3H , Camundongos Endogâmicos C57BL , NF-kappa B/metabolismo , Nitritos/metabolismo , Fosforilação/efeitos dos fármacos , Transdução de Sinais , Linfócitos T/efeitos dos fármacos , Linfócitos T/metabolismo , Receptor 4 Toll-Like/genética , Fator de Necrose Tumoral alfa/metabolismo
16.
Food Chem Toxicol ; 59: 26-33, 2013 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-23727177

RESUMO

Bisabolangelone (BISA), isolated from the roots of Angelica koreana, has many pharmacological activities, such as anti-tumor, anti-microbial, antioxidant, and anti-inflammatory activities. In this study, we investigated the anti-inflammatory mechanisms of BISA in dendritic cells (DCs), which play an essential role in innate and adaptive immune responses. BISA attenuated the production of pro-inflammatory cytokines including interleukin (IL)-12, IL-1ß, and tumor necrosis factor-alpha (TNF-α), migration to macrophage inflammatory protein-3 beta, and allo-T cell activating ability of DCs. In addition, BISA affected endocytosis of DCs. Molecular studies showed that BISA suppressed MAPK phosphorylation and nuclear translocation of NF-κB p50/p65. Taken together, our data suggest that BISA inhibited DC functions by blocking MAPK and NF-κB signaling.


Assuntos
Anti-Inflamatórios não Esteroides/farmacologia , Células Dendríticas/efeitos dos fármacos , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , NF-kappa B/antagonistas & inibidores , Inibidores de Proteínas Quinases/farmacologia , Sesquiterpenos/farmacologia , Angelica/química , Animais , Anti-Inflamatórios não Esteroides/química , Anti-Inflamatórios não Esteroides/isolamento & purificação , Células Cultivadas , Citocinas/metabolismo , Citocinese/efeitos dos fármacos , Células Dendríticas/citologia , Células Dendríticas/imunologia , Células Dendríticas/metabolismo , Endocitose/efeitos dos fármacos , Etnofarmacologia , Feminino , Ativação Linfocitária/efeitos dos fármacos , Medicina Tradicional Coreana , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , NF-kappa B/metabolismo , Raízes de Plantas/química , Inibidores de Proteínas Quinases/química , Inibidores de Proteínas Quinases/isolamento & purificação , República da Coreia , Sesquiterpenos/química , Sesquiterpenos/isolamento & purificação , Linfócitos T/citologia , Linfócitos T/efeitos dos fármacos , Linfócitos T/imunologia
17.
Int Immunopharmacol ; 15(3): 467-73, 2013 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-23415872

RESUMO

Sophoricoside (SOPH) is an isoflavone isolated from Sophora japonica (Leguminosae). In this study, the inhibitory effect of SOPH on contact dermatitis was investigated. At dosages of 3 and 10 mg/kg, SOPH ameliorated 2,4-dinitrochlorobenzene-induced acute and chronic contact dermatitis by 50-70%. As cellular targets, SOPH mainly affected the functions of B cells rather than T cells, macrophages and dendritic cells. As signaling targets, SOPH inhibited the phosphorylation and degradation of IκBα/ß and the nuclear translocation of NF-κB p65 in B cells, but not in dendritic cells and macrophages. SOPH did not affect the phosphorylation of ERK, p38, and JNK MAPKs, in B cells, dendritic cells, and macrophages. Taken together, these results suggest that SOPH ameliorates contact dermatitis by inhibiting mainly NF-κB signaling in B cells.


Assuntos
Linfócitos B/efeitos dos fármacos , Benzopiranos/administração & dosagem , Núcleo Celular/metabolismo , Dermatite de Contato/tratamento farmacológico , NF-kappa B/metabolismo , Sophora/química , Transporte Ativo do Núcleo Celular/efeitos dos fármacos , Animais , Linfócitos B/imunologia , Benzopiranos/química , Benzopiranos/isolamento & purificação , Células Cultivadas , Células Dendríticas/efeitos dos fármacos , Células Dendríticas/imunologia , Dermatite de Contato/imunologia , Dinitroclorobenzeno/imunologia , Macrófagos/efeitos dos fármacos , Macrófagos/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Transdução de Sinais/efeitos dos fármacos
18.
Int Immunopharmacol ; 15(1): 84-8, 2013 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-23159337

RESUMO

Effusanin C, a constituent of Isodon japonicus, has been used in oriental countries as a traditional folk medicine to treat inflammatory diseases, but its mechanism of action remains unknown. Here, we investigate the inhibitory activity of effusanin C in inflammatory monocytes. Effusanin C markedly inhibited the production of inflammatory mediators including nitric oxide, IL-1ß, and TNF-α in macrophages and dendritic cells. Furthermore, molecular studies showed that effusanin C inhibited phosphorylation of p38, JNK, and ERK, degradation of IκBß, and nuclear translocation of NF-κB p50/p65 in these cells. Taken together, these data show that effusanin C inhibits inflammatory responses by blocking NF-κB and MAPK signalings in monocytes.


Assuntos
Anti-Inflamatórios/farmacologia , Diterpenos/farmacologia , Proteínas Quinases Ativadas por Mitógeno/antagonistas & inibidores , NF-kappa B/metabolismo , Inibidores de Proteínas Quinases/farmacologia , Animais , Linhagem Celular , Células Dendríticas/efeitos dos fármacos , Células Dendríticas/metabolismo , Feminino , Interleucina-1beta/genética , Isodon , Macrófagos/efeitos dos fármacos , Macrófagos/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Monócitos/efeitos dos fármacos , Monócitos/metabolismo , NF-kappa B/antagonistas & inibidores , Óxido Nítrico/metabolismo , Óxido Nítrico Sintase Tipo II/genética , Componentes Aéreos da Planta , Fator de Necrose Tumoral alfa/genética
19.
Int Immunopharmacol ; 15(1): 138-43, 2013 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-23159603

RESUMO

Kamebakaurin (KA) has anti-cancer and anti-inflammatory activities through direct inhibition of DNA-binding activity of nuclear factor-kappa B (NF-κB) p50. We suggest here another molecular target of KA by the use of lipopolysaccharide-treated dendritic cells. In cell- and enzyme-based assays, KA directly inhibited autophosphorylation and kinase activity of TAK1, followed by the inhibition of TAK1-downstream signaling cascades, such as IKK phosphorylation-IκBα degradation-nuclear translocation of NF-κB, phosphorylation of MEK3/6-p38 mitogen activated protein kinase (MAPK), and MKK4/7-c-Jun N-terminal kinase MAPK. These results demonstrated that TAK1 might be the direct molecular target of KA.


Assuntos
Anti-Inflamatórios/farmacologia , Células Dendríticas/efeitos dos fármacos , Diterpenos/farmacologia , MAP Quinase Quinase Quinases/metabolismo , Animais , Citocinas/metabolismo , Células Dendríticas/metabolismo , Feminino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Proteínas Quinases Ativadas por Mitógeno/antagonistas & inibidores , NF-kappa B/antagonistas & inibidores , Óxido Nítrico/metabolismo , Proteínas Recombinantes de Fusão/metabolismo
20.
Food Chem Toxicol ; 51: 411-8, 2013 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-23108216

RESUMO

Maturation of dendritic cells (DCs) is usually attenuated in the tumor microenvironment, which is an important immunological problem in DC-based immunotherapy of cancer. In this study, we report the effect of a Mori fructus polysaccharide (MFP) on DC maturation. MFP was treated to DCs generated from mouse BM cells. MFP induced phenotypic maturation of DCs, as proven by the increased expression of CD40, CD80/86, and MHC-I/II molecules. MFP induced functional maturation of DCs, in that MFP increased the expression of IL-12, IL-1ß, TNF-α, and IFN-ß, decreased antigen capture capacity, and enhanced allogenic T cell stimulation. MFP efficiently induced maturation of DCs from C3H/HeN mice having normal toll-like receptor4 (TLR4), but not DCs from C3H/HeJ mice having mutated TLR4, suggesting that TLR4 might be one of the membrane receptors of MFP. As a mechanism of action, MFP increased phosphorylation of mitogen-activated protein kinase (MAPKs), and nuclear translocation of NF-κB p65 subunit, which were important signal molecules downstream from TLR4. These data suggest that MFP induces DC maturation through TLR4 and MFP can be used as an adjuvant in DC-based cancer immunotherapy.


Assuntos
Células Dendríticas/efeitos dos fármacos , Células Dendríticas/fisiologia , Morus/química , Polissacarídeos/farmacologia , Adjuvantes Imunológicos/farmacologia , Animais , Antígeno B7-1/metabolismo , Antígenos CD40/metabolismo , Proliferação de Células/efeitos dos fármacos , Citocinas/metabolismo , Feminino , Ativação Linfocitária/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C3H , Camundongos Endogâmicos C57BL , Fosforilação/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos , Receptor 4 Toll-Like/genética , Receptor 4 Toll-Like/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA