Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 41
Filter
Add more filters










Publication year range
1.
Food Sci Biotechnol ; 33(6): 1459-1466, 2024 May.
Article in English | MEDLINE | ID: mdl-38585568

ABSTRACT

In this study, the effect of different moisture levels in extruded plant-based meat on macrophage immunostimulation, and the potential of this meat as a protein source and a solution to environmental and economic challenges associated with conventional meat was investigated. To determine the effects of the extruded plant-based meat, cell viability assay, enzyme-linked immunosorbent assay, flow cytometry, and western blotting were performed. Low-moisture (LMME) and high-moisture meat extracts (HMME) showed higher potential to activate macrophages and regulate cytokine production than raw material extract. Treatment with LMME and HMME resulted in increased expression of CD80, CD86, and MHC class I/II proteins, indicating their potential to activate macrophages. Western blotting suggested that the immune activation observed in a previous study of macrophages was because of the phosphorylation of MAPKs and NF-κB. These findings suggest that extruded plant-based meat can potentially be used as an immunostimulatory food ingredient.

2.
Nutrients ; 16(5)2024 Feb 25.
Article in English | MEDLINE | ID: mdl-38474770

ABSTRACT

Sepsis, a leading cause of death worldwide, is a harmful inflammatory condition that is primarily caused by an endotoxin released by Gram-negative bacteria. Effective targeted therapeutic strategies for sepsis are lacking. In this study, using an in vitro and in vivo mouse model, we demonstrated that CM1, a derivative of the natural polyphenol chrysin, exerts an anti-inflammatory effect by inducing the expression of the ubiquitin-editing protein TNFAIP3 and the NAD-dependent deacetylase sirtuin 1 (SIRT1). Interestingly, CM1 attenuated the Toll-like receptor 4 (TLR4)-induced production of inflammatory cytokines by inhibiting the extracellular-signal-regulated kinase (ERK)/MAPK and nuclear factor kappa B (NF-κB) signalling pathways. In addition, CM1 induced the expression of TNFAIP3 and SIRT1 on TLR4-stimulated primary macrophages; however, the anti-inflammatory effect of CM1 was abolished by the siRNA-mediated silencing of TNFAPI3 or by the genetic or pharmacologic inhibition of SIRT1. Importantly, intravenous administration of CM1 resulted in decreased susceptibility to endotoxin-induced sepsis, thereby attenuating the production of pro-inflammatory cytokines and neutrophil infiltration into the lung compared to control mice. Collectively, these findings demonstrate that CM1 has therapeutic potential for diverse inflammatory diseases, including sepsis.


Subject(s)
Flavonoids , Sepsis , Shock, Septic , Mice , Animals , Sirtuin 1/metabolism , Toll-Like Receptor 4/metabolism , Lipopolysaccharides/pharmacology , NF-kappa B/metabolism , Shock, Septic/drug therapy , Endotoxins , Cytokines/metabolism , Sepsis/drug therapy , Anti-Inflammatory Agents/therapeutic use
3.
J Med Food ; 26(12): 927-938, 2023 Dec.
Article in English | MEDLINE | ID: mdl-38064431

ABSTRACT

Cisplatin, a potent and prominent chemotherapeutic drug, has considerable side effects, including nephrotoxicity, which limits its therapeutic application and efficacy. Therefore, the development of agents that protect normal cells while preserving cisplatin's chemotherapeutic properties is of utmost importance. This study aimed to explore the protective effects of Bombyx batryticatus protein-rich extract (BBPE) against cisplatin-induced nephrotoxicity in a cisplatin-treated mouse model and human embryonic kidney (HEK293) cells. Apoptosis was assessed in HEK293 cells to determine the cytoprotective effects of BBPE and its effects on the generation of cisplatin-induced reactive oxygen species (ROS) and mitochondrial transmembrane potential (MTP) collapse. Although cisplatin induced nephrotoxicity in HEK293 cells, pretreatment with BBPE showed significant protective effects against cisplatin-induced nephrotoxicity by regulating the expression levels of pro- and antiapoptotic proteins. The cytoprotective effects of BBPE were mediated by decreased ROS production and MTP loss in cisplatin-treated HEK293 cells. The in vitro results were confirmed in the cisplatin-treated mouse model. Pretreatment with BBPE protected against cisplatin-induced nephrotoxicity by restoring malondialdehyde, superoxide dismutase, and catalase levels in kidney tissue and blood urea nitrogen and creatinine serum levels. Furthermore, histopathological assessment and terminal dUTP nick end-labeling staining showed that BBPE mitigated cisplatin-induced nephrotoxicity in kidney tissues. Overall, BBPE may act as a potent agent for alleviating cisplatin-induced nephrotoxicity, thereby increasing the safety of cisplatin-based chemotherapy.


Subject(s)
Bombyx , Cisplatin , Mice , Animals , Humans , Cisplatin/adverse effects , HEK293 Cells , Reactive Oxygen Species/metabolism , Bombyx/metabolism , Oxidative Stress , Glutathione/metabolism , Kidney , Apoptosis
4.
Biol Proced Online ; 25(1): 17, 2023 Jun 16.
Article in English | MEDLINE | ID: mdl-37328878

ABSTRACT

BACKGROUND: Deinococcus radiodurans is a robust bacterium that can withstand harsh environments that cause oxidative stress to macromolecules due to its cellular structure and physiological functions. Cells release extracellular vesicles for intercellular communication and the transfer of biological information; their payload reflects the status of the source cells. Yet, the biological role and mechanism of Deinococcus radiodurans-derived extracellular vesicles remain unclear. AIM: This study investigated the protective effects of membrane vesicles derived from D. radiodurans (R1-MVs) against H2O2-induced oxidative stress in HaCaT cells. RESULTS: R1-MVs were identified as 322 nm spherical molecules. Pretreatment with R1-MVs inhibited H2O2-mediated apoptosis in HaCaT cells by suppressing the loss of mitochondrial membrane potential and reactive oxygen species (ROS) production. R1-MVs increased the superoxide dismutase (SOD) and catalase (CAT) activities, restored glutathione (GSH) homeostasis, and reduced malondialdehyde (MDA) production in H2O2-exposed HaCaT cells. Moreover, the protective effect of R1-MVs against H2O2-induced oxidative stress in HaCaT cells was dependent on the downregulation of mitogen-activated protein kinase (MAPK) phosphorylation and the upregulation of the nuclear factor E2-related factor 2 (Nrf2)/antioxidant response element (ARE) pathway. Furthermore, the weaker protective capabilities of R1-MVs derived from ΔDR2577 mutant than that of the wild-type R1-MVs confirmed our inferences and indicated that SlpA protein plays a crucial role in R1-MVs against H2O2-induced oxidative stress. CONCLUSION: Taken together, R1-MVs exert significant protective effects against H2O2-induced oxidative stress in keratinocytes and have the potential to be applied in radiation-induced oxidative stress models.

5.
Front Immunol ; 14: 1182927, 2023.
Article in English | MEDLINE | ID: mdl-37304302

ABSTRACT

Low-dose radiation therapy (LDRT) can suppress intractable inflammation, such as that in rheumatoid arthritis, and is used for treating more than 10,000 rheumatoid arthritis patients annually in Europe. Several recent clinical trials have reported that LDRT can effectively reduce the severity of coronavirus disease (COVID-19) and other cases of viral pneumonia. However, the therapeutic mechanism of LDRT remains unelucidated. Therefore, in the current study, we aimed to investigate the molecular mechanism underlying immunological alterations in influenza pneumonia after LDRT. Mice were irradiated to the whole lung 1 day post-infection. The changes in levels of inflammatory mediators (cytokines and chemokines) and immune cell populations in the bronchoalveolar lavage (BALF), lungs, and serum were examined. LDRT-treated mice displayed markedly increased survival rates and reduced lung edema and airway and vascular inflammation in the lung; however, the viral titers in the lungs were unaffected. Levels of primary inflammatory cytokines were reduced after LDRT, and transforming growth factor-ß (TGF-ß) levels increased significantly on day 1 following LDRT. Levels of chemokines increased from day 3 following LDRT. Additionally, M2 macrophage polarization or recruitment was increased following LDRT. We found that LDRT-induced TGF-ß reduced the levels of cytokines and polarized M2 cells and blocked immune cell infiltration, including neutrophils, in BALF. LDRT-induced early TGF-ß production was shown to be a key regulator involved in broad-spectrum anti-inflammatory activity in virus-infected lungs. Therefore, LDRT or TGF-ß may be an alternative therapy for viral pneumonia.


Subject(s)
Arthritis, Rheumatoid , COVID-19 , Pneumonia, Viral , Animals , Mice , COVID-19/radiotherapy , Inflammation , Cytokines , Dimercaprol , Transforming Growth Factors
6.
Mar Biotechnol (NY) ; 25(2): 314-327, 2023 Apr.
Article in English | MEDLINE | ID: mdl-37002465

ABSTRACT

Marine microbes, particularly Bacteroidetes, are a rich source of enzymes that can degrade diverse marine polysaccharides. Aquimarina sp. ERC-38, which belongs to the Bacteroidetes phylum, was isolated from seawater in South Korea. It showed agar-degrading activity and required an additional carbon source for growth on marine broth 2216. Here, the genome of the strain was sequenced to understand its agar degradation mechanism, and 3615 protein-coding sequences were predicted, which were assigned putative functions according to their annotated functional feature categories. In silico genome analysis revealed that the ERC-38 strain has several carrageenan-degrading enzymes but could not degrade carrageenan because it lacked genes encoding κ-carrageenanase and S1_19A type sulfatase. Moreover, the strain possesses multiple genes predicted to encode enzymes involved in agarose degradation, which are located in a polysaccharide utilization locus. Among the enzymes, Aq1840, which is closest to ZgAgaC within the glycoside hydrolase 16 family, was characterized using a recombinant enzyme expressed in Escherichia coli BL21 (DE3) cells. An enzyme assay revealed that recombinant Aq1840 mainly converts agarose to NA4. Moreover, recombinant Aq1840 could weakly hydrolyze A5 into A3 and NA2. These results showed that Aq1840 is involved in at least the initial agar degradation step prior to the metabolic pathway that uses agarose as a carbon source for growth of the strain. Thus, this enzyme can be applied to development and manufacturing industry for prebiotic and antioxidant food additive. Furthermore, our genome sequence analysis revealed that the strain is a potential resource for research on marine polysaccharide degradation mechanisms and carbon cycling.


Subject(s)
Flavobacteriaceae , Polysaccharides , Sepharose/metabolism , Carrageenan/metabolism , Agar/metabolism , Polysaccharides/metabolism , Flavobacteriaceae/genetics , Glycoside Hydrolases/metabolism
7.
Mol Med Rep ; 27(1)2023 Jan.
Article in English | MEDLINE | ID: mdl-36453199

ABSTRACT

Cisplatin is a prominent chemotherapeutic agent that can induce significant damage to normal cells. Therefore, it is important to develop agents that protect normal cells without influencing the chemotherapeutic effect of cisplatin. The present study was conducted to explore the protective effects of Annona muricata leaf polysaccharides (ALPS) against cisplatin­induced toxicity in macrophages. Apoptosis was assessed in macrophages and lung cancer cells to investigate the cytoprotective effect of ALPS, their effect on the production of cisplatin­induced reactive oxygen species (ROS) and the loss of the mitochondrial transmembrane potential (MTP). Cisplatin, when used alone or in combination with ALPS, showed significant toxicity against A549 and H460 lung cancer cells. However, cisplatin­induced cytotoxicity was suppressed by cotreatment of RAW 264.7 macrophages with ALPS. ALPS significantly inhibited the upregulation of Bax, cytosolic cytochrome c and caspases­3, ­8 and ­9. Moreover, ALPS resulted in the cleavage of PARP and downregulation of Bcl­2 levels in a concentration­dependent manner, which ultimately led to a reduction in the apoptotic and necrotic populations of cisplatin­treated RAW 264.7 macrophages. The suppression of the apoptotic signaling pathways was mediated through the reduction of ROS and MTP loss in cisplatin­treated RAW 264.7 macrophages. In addition, ALPS alleviated cell damage by suppressing the mitochondrial apoptotic pathways in cisplatin­treated bone marrow­derived macrophages. Together, these findings suggested that ALPS may alleviate the toxic side effects of chemotherapeutic agents and act as a potential candidate for use as an effective adjuvant therapy.


Subject(s)
Annona , Lung Neoplasms , Cisplatin/pharmacology , Reactive Oxygen Species , Polysaccharides/pharmacology , Macrophages , Plant Leaves , Mitochondria
8.
J Microbiol Biotechnol ; 32(7): 835-843, 2022 Jul 28.
Article in English | MEDLINE | ID: mdl-35719091

ABSTRACT

Deinococcus radiodurans is an extremophilic bacterium that can thrive in harsh environments. This property can be attributed to its unique metabolites that possess strong antioxidants and other pharmacological properties. To determine the potential of D. radiodurans R1 lysate (DeinoLys) as a pharmacological candidate for inflammatory bowel disease (IBD), we investigated the anti-inflammatory activity of DeinoLys in bone marrow-derived dendritic cells (BMDCs) and a colitis mice model. Lipopolysaccharide (LPS)-stimulated BMDCs treated with DeinoLys exhibited alterations in their phenotypic and functional properties by changing into tolerogenic DCs, including strongly inhibited proinflammatory cytokines (TNF-α and IL-12p70) and surface molecule expression and activated DC-induced T cell proliferation/activation with high IL-10 production. These phenotypic and functional changes in BMDCs induced by DeinoLys in the presence of LPS were abrogated by IL-10 neutralization. Furthermore, oral administration of DeinoLys significantly reduced clinical symptoms against dextran sulfate sodium-induced colitis, including body weight loss, disease activity index, histological severity in colon tissue, and lower myeloperoxidase level in mice. Our results establish DeinoLys as a potential anti-inflammatory candidate for IBD therapy.


Subject(s)
Colitis , Deinococcus , Inflammatory Bowel Diseases , Animals , Anti-Inflammatory Agents/pharmacology , Colitis/chemically induced , Colitis/drug therapy , Colitis/metabolism , Cytokines/metabolism , Deinococcus/metabolism , Dendritic Cells/metabolism , Dextran Sulfate/toxicity , Disease Models, Animal , Interleukin-10/metabolism , Lipopolysaccharides/pharmacology , Mice , Mice, Inbred C57BL
9.
Int J Mol Sci ; 22(19)2021 Sep 30.
Article in English | MEDLINE | ID: mdl-34638974

ABSTRACT

Extracellular vesicles (EVs) have recently been isolated from different plants. Plant-derived EVs have been proposed as potent therapeutics and drug-delivery nanoplatforms for delivering biomolecules, including proteins, RNAs, DNAs, and lipids. Herein, Petasites japonicus-derived EVs (PJ-EVs) were isolated through a series of centrifugation steps and characterized using dynamic light scattering and transmission electron microscopy. Immunomodulatory effects of PJ-EVs were assessed using dendritic cells (DCs). PJ-EVs exhibited a spherical morphology with an average size of 122.6 nm. They induced the maturation of DCs via an increase in the expression of surface molecules (CD80, CD86, MHC-I, and MHC-II), production of Th1-polarizing cytokines (TNF-α and IL-12p70), and antigen-presenting ability; however, they reduced the antigen-uptake ability. Furthermore, maturation of DCs induced by PJ-EVs was dependent on the activation and phosphorylation of MAPK and NF-κB signal pathways. Notably, PJ-EV-treated DCs strongly induced the proliferation and differentiation of naïve T cells toward Th1-type T cells and cytotoxic CD8+ T cells along with robust secretion of IFN-γ and IL-2. In conclusion, our study indicates that PJ-EVs can be potent immunostimulatory candidates with an ability of strongly inducing the maturation of DCs.


Subject(s)
Adjuvants, Immunologic/pharmacology , Cell Differentiation/drug effects , Dendritic Cells/immunology , Extracellular Vesicles/immunology , Petasites/cytology , Plants, Edible/cytology , Animals , CD8-Positive T-Lymphocytes/immunology , Cell Differentiation/immunology , Cell Proliferation/drug effects , Cells, Cultured , Cytokines/metabolism , Dendritic Cells/drug effects , Female , Lymphocyte Activation/drug effects , MAP Kinase Signaling System/drug effects , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mitogen-Activated Protein Kinases/metabolism , NF-kappa B/metabolism , Th1 Cells/immunology
10.
Int J Mol Sci ; 22(14)2021 Jul 13.
Article in English | MEDLINE | ID: mdl-34299130

ABSTRACT

Although cisplatin is one of most effective chemotherapeutic drugs that is widely used to treat various types of cancer, it can cause undesirable damage in immune cells and normal tissue because of its strong cytotoxicity and non-selectivity. This study was conducted to investigate the cytoprotective effects of Cudrania tricuspidata fruit-derived polysaccharides (CTPS) against cisplatin-induced cytotoxicity in macrophages, lung cancer cell lines, and a mouse model, and to explore the possibility of application of CTPS as a supplement for anticancer therapy. Both cisplatin alone and cisplatin with CTPS induced a significant cytotoxicity in A549 and H460 lung cancer cells, whereas cytotoxicity was suppressed by CTPS in cisplatin-treated RAW264.7 cells. CTPS significantly attenuated the apoptotic and necrotic population, as well as cell penetration in cisplatin-treated RAW264.7 cells, which ultimately inhibited the upregulation of Bcl-2-associated X protein (Bax), cytosolic cytochrome c, poly (adenosine diphosphateribose) polymerase (PARP) cleavage, and caspases-3, -8, and -9, and the downregulation of B cell lymphoma-2 (Bcl-2). The CTPS-induced cytoprotective action was mediated with a reduction in reactive oxygen species production and mitochondrial transmembrane potential loss in cisplatin-treated RAW264.7 cells. In agreement with the results obtained above, CTPS induced the attenuation of cell damage in cisplatin-treated bone marrow-derived macrophages (primary cells). In in vivo studies, CTPS significantly inhibited metastatic colonies and bodyweight loss as well as immunotoxicity in splenic T cells compared to the cisplatin-treated group in lung metastasis-induced mice. Furthermore, CTPS decreased the level of CRE and BUN in serum. In summation, these results suggest that CTPS-induced cytoprotective action may play a role in alleviating the side effects induced by chemotherapeutic drugs.


Subject(s)
Cisplatin/toxicity , Fruit/chemistry , Macrophages/drug effects , Melanoma, Experimental/drug therapy , Moraceae/chemistry , Plant Extracts/pharmacology , Polysaccharides/pharmacology , Animals , Antineoplastic Agents/toxicity , Apoptosis , Cell Proliferation , Female , Humans , Lung Neoplasms/chemically induced , Lung Neoplasms/drug therapy , Lung Neoplasms/secondary , Macrophages/pathology , Melanoma, Experimental/chemically induced , Melanoma, Experimental/pathology , Membrane Potential, Mitochondrial , Mice , Mice, Inbred C57BL , Mice, Nude , Protective Agents/pharmacology , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
11.
Molecules ; 26(6)2021 Mar 11.
Article in English | MEDLINE | ID: mdl-33799689

ABSTRACT

Although our previous study revealed that gamma-irradiated chrysin enhanced anti-inflammatory activity compared to intact chrysin, it remains unclear whether the chrysin derivative, CM1, produced by gamma irradiation, negatively regulates toll-like receptor (TLR) signaling. In this study, we investigated the molecular basis for the downregulation of TLR4 signal transduction by CM1 in macrophages. We initially determined the appropriate concentration of CM1 and found no cellular toxicity below 2 µg/mL. Upon stimulation with lipopolysaccharide (LPS), CM1 modulated LPS-stimulated inflammatory action by suppressing the release of proinflammatory mediators (cytokines TNF-α and IL-6) and nitric oxide (NO) and downregulated the mitogen-activated protein kinase (MAPK) and nuclear factor-κB (NF-κB) signaling pathways. Furthermore, CM1 markedly elevated the expression of the TLR negative regulator toll-interacting protein (Tollip) in dose- and time-dependent manners. LPS-induced expression of cell surface molecules (CD80, CD86, and MHC class I/II), proinflammatory cytokines (TNF-α and IL-6), COX-2, and iNOS-mediated NO were inhibited by CM1; these effects were prevented by the knockdown of Tollip expression. Additionally, CM1 did not affect the downregulation of LPS-induced expression of MAPKs and NF-κB signaling in Tollip-downregulated cells. These findings provide insight into effective therapeutic intervention of inflammatory disease by increasing the understanding of the negative regulation of TLR signaling induced by CM1.


Subject(s)
Flavonoids/pharmacology , Intracellular Signaling Peptides and Proteins/metabolism , Macrophages/metabolism , Animals , Anti-Inflammatory Agents/pharmacology , Flavonoids/metabolism , Flavonoids/radiation effects , Inflammation/drug therapy , Interleukin-6 , Lipopolysaccharides/adverse effects , Lipopolysaccharides/pharmacology , Macrophages/drug effects , Mice , Mitogen-Activated Protein Kinases/metabolism , NF-kappa B/metabolism , Nitric Oxide , RAW 264.7 Cells , Signal Transduction/drug effects , Toll-Like Receptor 4/drug effects , Toll-Like Receptor 4/metabolism , Toll-Like Receptors/metabolism , Tumor Necrosis Factor-alpha
12.
Int Immunopharmacol ; 95: 107513, 2021 Jun.
Article in English | MEDLINE | ID: mdl-33756223

ABSTRACT

The objective of the current study was to demonstrate the immunostimulatory effects of a polysaccharide isolated from Chrysanthemum zawadskii Herbich var. latilobum leaves (CP) and evaluate its potential as a vaccine adjuvant. Results showed that CP induced maturation of the dendritic cells (DCs). In addition, CP-treated DCs activated naïve T cells to polarized CD4+ and CD8+ T cells and substantially induced the production of IFN-γ and IL-2 in vitro. Furthermore, CP initiated the maturation of DCs via the activation of MAPK and NF-κB signaling pathways. Interestingly, systemic administration of CP-treated DCs pulsed with ovalbumin (OVA) peptides significantly enhanced the immune response in vivo, which included the generation of antigen (OVA)-specific polyfunctional T cells, increased cytotoxic T lymphocyte activity, induction of Th1-mediated humoral immunity, and suppression of tumor growth. Taken together, our study highlighted the immunoregulatory activity of CP as well as its potential as a candidate vaccine adjuvant.


Subject(s)
Adjuvants, Immunologic/pharmacology , Chrysanthemum , Dendritic Cells/drug effects , Polysaccharides/pharmacology , T-Lymphocytes/drug effects , Animals , Dendritic Cells/immunology , Female , Mice, Inbred BALB C , Mice, Inbred C57BL , NF-kappa B/immunology , Ovalbumin/immunology , Plant Leaves , T-Lymphocytes/immunology , Vaccines
13.
Int Immunopharmacol ; 95: 107523, 2021 Jun.
Article in English | MEDLINE | ID: mdl-33706053

ABSTRACT

Although we previously identified a new hydroxymethoxyl chrysin derivative (HMOC) using ionizing radiation, the anti-inflammatory mechanism of HMOC in dendritic cells remains unclear. In this study, we investigate the effects of HMOC on phenotypic and functional changes in activated bone marrow-derived dendritic cells (BMDCs). In lipopolysaccharide (LPS)-stimulated BMDCs, HMOC treatment inhibited pro-inflammatory cytokines (TNF-α, IL-12p70, and IL-1ß), surface molecules (CD80, CD86, MHC-I, and MHC-II), and antigen-presentation to MHC-I and II without a decrease in IL-10. Furthermore, HMOC increased indoleamine 2,3-dioxygenase-1 (IDO1) activity via activation of JNK and p38 signaling in the presence of LPS. Interestingly, LPS-stimulated DCs treated with HMOC inhibited the proliferation and activation of CD4+ and CD8+ T cells, as well as differentiation of CD4+ T cells into Th1-, Th2- and Th17 cells. In addition, LPS-stimulated DCs treated with HMOC induced an increase in CD4+CD25+Foxp3+ regulatory T cells (Tregs). Collectively, our results suggest that HMOC confers tolerogenic properties in BMDCs, which are responsible for inducing Th cell differentiation to Tregs. Our findings provide a better understanding of the anti-inflammatory mechanism of HMOC in DCs and may contribute to development of a valuable therapeutic candidate for atopic dermatitis.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Dendritic Cells/drug effects , Flavonoids/pharmacology , Animals , Antigen Presentation , Cell Survival/drug effects , Cells, Cultured , Coculture Techniques , Cytokines/immunology , Dendritic Cells/immunology , Female , Lipopolysaccharides , Mice, Inbred C57BL , Ovalbumin/immunology , T-Lymphocytes/drug effects , T-Lymphocytes/immunology
14.
Molecules ; 26(2)2021 Jan 18.
Article in English | MEDLINE | ID: mdl-33477499

ABSTRACT

Bombyx batryticatus, a protein-rich edible insect, is widely used as a traditional medicine in China. Several pharmacological studies have reported the anticancer activity of B. batryticatus extracts; however, the capacity of B. batryticatus extracts as immune potentiators for increasing the efficacy of cancer immunotherapy is still unverified. In the present study, we investigated the immunomodulatory role of B. batryticatus protein-rich extract (BBPE) in bone marrow-derived dendritic cells (BMDCs) and DC vaccine-immunized mice. BBPE-treated BMDCs displayed characteristics of mature immune status, including high expression of surface molecules (CD80, CD86, major histocompatibility complex (MHC)-I, and MHC-II), increased production of proinflammatory cytokines (tumor necrosis factor-α and interleukin-12p70), enhanced antigen-presenting ability, and reduced endocytosis. BBPE-treated BMDCs promoted naive CD4+ and CD8+ T-cell proliferation and activation. Furthermore, BBPE/ovalbumin (OVA)-pulsed DC-immunized mice showed a stronger OVA-specific multifunctional T-cell response in CD4+ and CD8+ T cells and a stronger Th1 antibody response than mice receiving differently treated DCs, which showed the enhanced protective effect against tumor growth in E.G7 tumor-bearing mice. Our data demonstrate that BBPE can be a novel immune potentiator for a DC-based vaccine in anticancer therapy.


Subject(s)
Adjuvants, Immunologic , Antigen Presentation/immunology , Cancer Vaccines/immunology , Dendritic Cells/physiology , Insect Proteins/metabolism , Th1 Cells/immunology , Tissue Extracts/pharmacology , Animals , Bombyx , Cell Proliferation , Dendritic Cells/cytology , Dendritic Cells/drug effects , Dendritic Cells/immunology , Female , Lymphocyte Activation , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL
15.
J Nutr Biochem ; 87: 108524, 2021 01.
Article in English | MEDLINE | ID: mdl-33039583

ABSTRACT

Tolerogenic dendritic cells (tolDCs) can induce the differentiation of immunosuppressive regulatory T cells and are therefore candidates for the prevention or treatment of various inflammatory diseases. Galangin, a major component of propolis and Alpinia officinarum, has well-established anti-inflammatory effects, but its ability to induce a tolerogenic state in DCs has not been demonstrated. In this study, we investigated the effects of galangin on DC differentiation and immune responses. In particular, we compared phenotypic and functional differences between DCs (Gal-DCs) generated by galangin treatment during DC differentiation and bone marrow-derived DCs. Gal-DCs were generated by adding culture medium containing various doses of galangin (1.8-18.5 µM) on 3 and 6 day. Upon lipopolysaccharide (100 ng/mL) stimulation for 24 h, Gal-DCs generated with 7.4 µM galangin treatment showed lower levels of CD86 and lower major histocompatibility complex class II antigen-presentation than those of bone marrow-derived DCs. Furthermore, Gal-DCs showed markedly increased programmed death ligand 1 expression and IL-10 production via the activation of mitogen-activated protein kinases. Interestingly, Gal-DCs co-cultured with allogeneic CD4 T cells exhibited the reduced cell proliferation and differentiation into Th1-, Th2-, and Th17-type cell; instead, Gal-DCs contributed to the induction of CD4+CD25+Foxp3+ Tregs. Taken together, our data suggest that exposure to galangin during DC differentiation confers tolerogenic properties, efficiently inducing Th cell differentiation to immunosuppressive Tregs. These findings provide new insights into the molecular mechanism underlying the anti-inflammatory effects of galangin on DCs.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Cell Differentiation/drug effects , Dendritic Cells/drug effects , Flavonoids/pharmacology , Lipopolysaccharides/immunology , Alpinia/chemistry , Animals , Anti-Inflammatory Agents/chemistry , Cells, Cultured , Dendritic Cells/cytology , Dendritic Cells/immunology , Female , Flavonoids/chemistry , Immune Tolerance/drug effects , Mice, Inbred BALB C , Mice, Inbred C57BL , T-Lymphocytes, Regulatory/cytology , T-Lymphocytes, Regulatory/drug effects , T-Lymphocytes, Regulatory/immunology
16.
Vaccines (Basel) ; 8(4)2020 Nov 19.
Article in English | MEDLINE | ID: mdl-33228229

ABSTRACT

Cancer cells can secrete exosomes under various stressful conditions, whose functions are involved in the delivery of various biologically active materials into host cells and/or modulation of host immune responses. Therefore, an improved understanding of the immunological interventions that stress-induced tumor exosomes have may provide novel therapeutic approaches and more effective vaccine designs. Here, we confirmed the phenotypical and functional alterations of dendritic cells (DCs), which act as a bridge between the innate and adaptive arms of immunity, following non-irradiated (N-exo) and gamma-irradiated melanoma cancer cell-derived exosome (G-exo) stimulation, and evaluated the N-exo- and G-exo-stimulated DCs as therapeutic cancer vaccine candidates. We demonstrated that G-exo-stimulated DCs result in DC maturation by the upregulation of surface molecule expression, pro-inflammatory cytokine release, and antigen-presenting ability, and the downregulation of endocytic capacity. In addition, these cells promoted T cell proliferation and the generation of T helper type 1 (Th1) and interferon (IFN)-γ-producing CD8+ T cells. However, N-exo-stimulated DCs induced semi-mature phenotypes and functions, eventually inhibiting T cell proliferation, decreasing IFN-γ, and increasing IL-10-producing CD4+ T cells. In addition, although N-exo and G-exo stimulations showed similar levels of antigen-specific IFN-γ production, which served as tumor antigen sources in melanoma-specific T cells, G-exo-stimulated DC vaccination conferred a stronger tumor growth inhibition than N-exo-stimulated DC vaccination; further, this was accompanied by a high frequency of tumor-specific, multifunctional effector T cells. These results suggest that gamma irradiation could provide important clues for designing and developing effective exosome vaccines that can induce strong immunogenicity, especially tumor-specific multifunctional T cell responses.

17.
Nutrients ; 12(11)2020 Oct 22.
Article in English | MEDLINE | ID: mdl-33105813

ABSTRACT

Oxya chinensis sinuosa (Ocs) is consumed as representative edible insects in Asia, but its function in various immune systems remains unclear. This study aimed to demonstrate the immunomodulatory effect, particularly on the innate and adaptive immune response, of Ocs protein (Ocs-P) and to investigate its function as a potent anticancer immunostimulant when administered during the progression stage of colon carcinoma in tumor-bearing mice. Our in vitro results demonstrated that Ocs-P treatment induces phenotypic alteration (increased expression of surface molecules and production of Th1-polarizing cytokines and decreased antigen uptake ability) of dendritic cells (DCs) through the activation of MAPK and NF-κB-dependent signaling pathways. Additionally, Ocs-P-stimulated DCs initiated differentiation of naive T cells into IFN-γ-producing Th1-type T cells effectively and activated cytotoxic CD8+ T cell response. In colon carcinoma-bearing mouse models, oral administration of Ocs-P inhibited tumor growth and restored the expression of decreased surface molecules in lineage-CD11c+MHC-II+ splenic DCs. Furthermore, Ocs-P administration enhanced the generation of multifunctional CD4+ and CD8+ T cells expressing Th1-type cytokines (TNF-α, IFN-γ, and IL-2) and the degranulation marker (CD107a). Collectively, these results suggest that Ocs-P demonstrates an immunostimulatory effect and may induce powerful anticancer immunity.


Subject(s)
Colonic Neoplasms/immunology , Dietary Supplements , Edible Insects/chemistry , Grasshoppers/chemistry , Insect Proteins/immunology , Insect Proteins/pharmacology , Adaptive Immunity , Adjuvants, Immunologic , Animals , Colonic Neoplasms/diet therapy , Colonic Neoplasms/pathology , Cytokines/metabolism , Dendritic Cells/immunology , Female , Immunity, Innate , Lymphocyte Activation , MAP Kinase Signaling System , Mice , Mice, Inbred BALB C , NF-kappa B p50 Subunit/metabolism , Signal Transduction , T-Lymphocytes, Cytotoxic/immunology , Th1 Cells/immunology
18.
Nutrients ; 12(6)2020 May 29.
Article in English | MEDLINE | ID: mdl-32486094

ABSTRACT

Dendritic cells (DCs) are powerful antigen-presenting cells that are often used to evaluate adjuvants, particularly for adjuvant selection for various vaccines. Here, polysaccharides (named ALP) isolated from leaves of Annona muricata L., which are used in traditional medicine such as for bacterial infections and inflammatory diseases, were evaluated as an adjuvant candidate that can induce anti-tumor activity. We first confirmed the phenotypic (surface molecules, cytokines, antigen uptake, and antigen-presenting ability) and functional alterations (T cell proliferation/activation) of DCs in vitro. We also confirmed the adjuvant effect by evaluating anti-tumor activity and immunity using an ALP-treated DC-immunized mouse model. ALP functionally induced DC maturation by up-regulating the secretion of Th1-polarizing pro-inflammatory cytokines, the expression of surface molecules, and antigen-presenting ability. ALP triggered DC maturation, which is dependent on the activation of the MAPK and NF-κB signaling pathways. ALP-activated DCs showed an ample capacity to differentiate naive T cells to Th1 and activated CD8+ T cells effectively. The systemic administration of DCs that pulse ALP and ovalbumin peptides strongly increased cytotoxic T lymphocyte (CTL) activity (by 9.5% compared to that in the control vaccine groups), the generation of CD107a-producing multifunctional T cells, and Th1-mediated humoral immunity, and caused a significant reduction (increased protection by 29% over that in control vaccine groups) in tumor growth. ALP, which triggers the Th1 and CTL response, provides a basis for a new adjuvant for various vaccines.


Subject(s)
Adjuvants, Immunologic , Annonaceae/chemistry , Cancer Vaccines , Dendritic Cells/immunology , Polysaccharides , Thymoma/immunology , Thymus Neoplasms/immunology , Animals , Antigen Presentation , Cytokines/metabolism , Female , Mice, Inbred BALB C , Mice, Inbred C57BL , Mitogen-Activated Protein Kinases/metabolism , NF-kappa B/metabolism , Polysaccharides/immunology , Polysaccharides/isolation & purification , Polysaccharides/pharmacology , Signal Transduction/drug effects , Signal Transduction/immunology , T-Lymphocytes/immunology , Thymoma/therapy , Thymus Neoplasms/therapy
19.
Korean J Parasitol ; 58(1): 7-14, 2020 Feb.
Article in English | MEDLINE | ID: mdl-32145722

ABSTRACT

Toxoplasma gondii is an intracellular protozoan parasite that infects approximately one third of the human popu- lation worldwide. Considering the toxicity and side effects of anti-toxoplasma medications, it is important to develop effec- tive drug alternatives with fewer and less severe off-target effects. In this study, we found that 4-hydroxybenzaldehyde (4- HBA) induced autophagy and the expression of NAD-dependent protein deacetylase sirtuin-1 (SIRT1) in primary murine bone marrow-derived macrophages (BMDMs). Interestingly, treatment of BMDMs with 4-HBA significantly reduced the number of macrophages infected with T. gondii and the proliferation of T. gondii in infected cells. This effect was impaired by pretreating the macrophages with 3-methyladenine or wortmannin (selective autophagy inhibitors) or with sirtinol or EX527 (SIRT1 inhibitors). Moreover, we found that pharmacological inhibition of SIRT1 prevented 4-HBA-mediated expres- sion of LC3-phosphatidylethanolamine conjugate (LC3-II) and the colocalization of T. gondii parasitophorous vacuoles with autophagosomes in BMDMs. These data suggest that 4-HBA promotes antiparasitic host responses by activating SIRT1- mediated autophagy, and 4-HBA might be a promising therapeutic alternative for the treatment of toxoplasmosis.


Subject(s)
Autophagy , Benzaldehydes/pharmacology , Macrophages/physiology , Sirtuin 1 , Toxoplasma/growth & development , Animals , Cells, Cultured , Depression, Chemical , Mice, Inbred C57BL
20.
Biosci Biotechnol Biochem ; 84(5): 1001-1012, 2020 May.
Article in English | MEDLINE | ID: mdl-31960754

ABSTRACT

Crude extracts and phytochemical compounds derived from Annona muricata leaves have been demonstrated to exert neuroprotective effects. However, the neuroprotective effects of Annona muricata leaves-derived polysaccharide extracts (ALPs) have not been investigated. ALP treatment was shown to induce concentration-dependent antioxidant activity in HT22 cells, and to increase cell viability in H2O2-treated HT22 cells. These effects were correlated with a decrease in major components of oxidation, including: Ca2+, ROS, and malondialdehyde (MDA). Mediators of the intracellular response to oxidation, including Bax, cytochrome c, and cleaved caspases-3, -8, -9, MAPKs, and NF-κB, were positively influenced by ALP treatment under conditions of H2O2-mediated oxidative stress. In addition, ALP restored the expression of superoxide dismutase (SOD) and associated signaling pathways (PARP, PI3K/AKT and Nrf2-mediated HO-1/NQO-1) following H2O2 treatment. These results provide new pharmacological evidence that ALP facilitates neuroprotection via prevention of neuronal oxidative stress and promotion of cell survival signaling pathways.Abbreviations: ABTS: 2,2'-azino-bis-(3-ethylbenzothiazoline-6-sulfonicacid); AD: Alzheimer's disease; ALP: polysaccharide extracts isolated from Annona muricata leaves; ARE: antioxidant response element; DPPH: 1,1-diphenyl-picrylhydrazyl; DCFH-DA: 2',7'-dichlorofluorescin diacetate; ECL: electrochemiluminescence; ERK: extracellular regulated kinase; FBS: Fetal bovine serum; FITC: fluorescein isothiocyanate; FRAP: ferric reducing antioxidant power; HO-1: Heme oxygenase-1; JNK: c-jun N-terminal kinase; MAPKs: mitogen-activated protein kinases; MDA: malondialdehyde; MMP: mitochondrial membrane potential; MTT: 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazoliumbromide; NQO1: NAD(P)H:quinine oxidoreductase 1, Nrf2: nuclear factor-E2-related factor 2; PD: parkinson's disease; PI3K: phosphatidylinositol-3kinase; PVDF: polyvinylidene difluoride; ROS: reactive oxygen species; SOD: Superoxidedismutase; TPTZ: tripydyltriazine.


Subject(s)
Annona/chemistry , Antioxidants/pharmacology , Hydrogen Peroxide/pharmacology , Neurons/drug effects , Neuroprotective Agents/pharmacology , Plant Extracts/pharmacology , Polysaccharides/pharmacology , Animals , Apoptosis/drug effects , Calcium/metabolism , Cell Cycle/drug effects , Cell Line , Cell Survival/drug effects , Malondialdehyde/analysis , Malondialdehyde/metabolism , Membrane Potential, Mitochondrial/drug effects , Mice , Oxidative Stress/drug effects , Reactive Oxygen Species/analysis , Reactive Oxygen Species/metabolism , Signal Transduction/drug effects , Superoxide Dismutase/analysis , Superoxide Dismutase/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...