Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
Add more filters










Database
Language
Publication year range
1.
Int Immunopharmacol ; 95: 107476, 2021 Jun.
Article in English | MEDLINE | ID: mdl-33676147

ABSTRACT

BACKGROUND: Vitamin C (ascorbic acid, AscH2) has been shown to enhance immunity. Here, we studied its immunomodulatory effect on human endothelial cells (ECs) during S. aureus infection. MATERIALS AND METHODS: The ex vivo effects of AscH2 were performed on primary human umbilical vein endothelial cells (HUVECs) infected or not with S. aureus. RESULTS: AscH2 treatment induced a marked downregulation of nitric oxide (NO) production and a moderate upregulation of arginase activity in S. aureus-infected HUVECs (respectively, p < 0.05 and p > 0.05). Although the upregulated release levels of soluble intercellular adhesion molecular 1 (sICAM-1/sCD54) and sE-selectin (sCD62E) molecules were not significantly different between treated and untreated S. aureus-infected HUVECs, AscH2 treatment induced reversing effect on sICAM-1 release when comparing to uninfected control HUVECs. Moreover, AscH2 treatment appears to have a significant effect on preventing HUVEC necrosis induced by S. aureus infection (p < 0.05). Furthermore, AscH2 treatment induced a significant upregulation of cell protective redox biomarker in S. aureus-infected, as shown by superoxide dismutase (SOD) activity (p < 0.05), but not by catalase activity (p > 0.05). Additionally, S. aureus infection markedly downregulated total bound calcium ions (bCa2+) levels as compared to control HUVECs, whereas, AscH2 treatment induced a slight upregulation of bCa2+ levels in infected HUVECs as compared to infected and untreated HUVECs (p > 0.05). On the other hand, AscH2 treatment downregulated increased total cellular cholesterol content (tccCHOL) levels in HUVECs induced by S. aureus infection (p < 0.05). In addition, AscH2 treatment markedly reversed S. aureus effect on upregulation of intracellular glucose (iGLU) levels within infected HUVECs (p < 0.05). Moreover, AscH2 treatment significantly downregulated S. aureus growth (p < 0.05), and significantly upregulated bacterial internalization and intracellular killing by HUVECs (p < 0.05), as well as their cell cycle activation (p < 0.01). Finally, AscH2 treatment has a slight effect on the production of interleukin 6 (IL-6), but induced a marked downregulation of that of IL-1ß in S. aureus-infected HUVECs (respectively, p > 0.05, and p < 0.05). CONCLUSIONS: Our outcomes demonstrated that, during S. aureus infection, AscH2 treatment promotes human ECs survival and function, as well as prevents inflammatory response exacerbation, while inducing bactericidal activity.


Subject(s)
Anti-Bacterial Agents/pharmacology , Ascorbic Acid/analogs & derivatives , Ascorbic Acid/pharmacology , Human Umbilical Vein Endothelial Cells/drug effects , Immunologic Factors/pharmacology , Staphylococcus aureus , Cell Survival/drug effects , Cells, Cultured , Human Umbilical Vein Endothelial Cells/immunology , Human Umbilical Vein Endothelial Cells/microbiology , Humans , Intercellular Adhesion Molecule-1/immunology , Interleukin-1beta/immunology , Interleukin-6/immunology , Nitric Oxide/immunology , Staphylococcal Infections/drug therapy , Staphylococcal Infections/immunology
2.
PLoS One ; 15(10): e0240982, 2020.
Article in English | MEDLINE | ID: mdl-33108409

ABSTRACT

BACKGROUND: Immune activities of monocytes (MOs) can be altered within the microenvironment of solid malignancies, including breast cancer. Metformin (1,1-dimethylbiguanide hydrochloride, MET), has been shown to decrease tumor cell proliferation, but its effects have yet to be explored with respect to MOs (monocytes) activity during their crosstalk with breast cancer cells. Here, we investigated the effects of MET on overall phenotypic functional activities, including cellular immunometabolism and protective redox signaling based-biomarkers, intracellular free calcium ions (ifCa2+), phagocytosis and co-operative cytokines (IFN-γ and IL-10) of autologous MOs before and during their interplay with primary ER-/PR-/HER2+ breast cancer cells. METHODS: Human primary breast cancer cells were either cultured alone or co-cultured with autologous MOs before treatment with MET. RESULTS: MET downregulated breast cancer cell proliferation and phagocytosis, while having no significant effect on the ratio of phosphorylated Akt (p-Akt) to total Akt. Additionally, we observed that, in the absence of MET treatment, the levels of lactate dehydrogenase (LDH)-based cytotoxicity, catalase, ifCa2+, IL-10 and arginase activity were significantly reduced in co-cultures compared to levels in MOs cultured alone whereas levels of inducible nitric oxide synthase (iNOS) activity were significantly increased. In contrast, MET treatment reduced the effects measured in co-culture on the levels of LDH-based cytotoxicity, arginase activity, catalase, ifCa2+, and IFN-γ. MET also induced upregulation of both iNOS and arginase in MO cells, although the increase did not reach significant difference for iNOS activity. Moreover, MET induced a robust increase of superoxide dismutase (SOD) activity in MOs, but not in MOs co-cultured with breast cancer cells. Furthermore, MET markedly upregulated the levels of IFN-γ production and downregulated those of IL-10 in isolated MOs, while inducing a slight opposing up-regulation of IL-10 production in co-cultures. CONCLUSIONS: Our results show that the biomarkers of phenotypic functional activities of MOs are modified after co-culturing with primary human breast cancer cells. Treatment of co-cultures with MET resulted in increased release of antitumor cytokine IFN-γ and ifCa2+, and increased cell necrosis during breast cancer cells-MOs crosstalk.


Subject(s)
Biomarkers/metabolism , Breast Neoplasms/metabolism , Metformin/pharmacology , Monocytes/cytology , Breast Neoplasms/drug therapy , Cell Proliferation/drug effects , Cells, Cultured , Coculture Techniques , Female , Gene Expression Regulation, Neoplastic/drug effects , Humans , L-Lactate Dehydrogenase/metabolism , Receptor, ErbB-2/metabolism , Receptors, Estrogen/metabolism , Receptors, Progesterone/metabolism
3.
Int Immunopharmacol ; 83: 106349, 2020 Jun.
Article in English | MEDLINE | ID: mdl-32172203

ABSTRACT

BACKGROUND: Exacerbation of CD16 as molecule marker of both intermediate and non-classical monocytes (MOs) has been shown to be involved in the pathogenesis of myocardial infarction (MI). In this study, we have tried to evaluate the aspirin (acetylsalicylic acid, ASA) treatment effect on the CD16-expressed MOs and activation-associated CD40 in MI. METHODS: MOs were isolated from the whole blood of healthy controls and patients with MI. The cells were stimulated and treated with different doses of ASA. RESULTS: ASA significantly decreased nitric oxide (NO) production and inducible NO synthase (iNOS) activity, but significantly increased arginase activity. Levels of interleukin (IL)-1ß, IL-6 and interferon-γ (IFN-γ) were downregulated, whereas those of IL-10 were upregulated. Additionally, ASA induced a markedly increase in both phagocytosis and intracellular pathogen killing activities. Moreover, ASA treatment induced significantly upregulation of intracellular levels of glucose (iGlu), and free calcium ions (ifCa2+), and, covertly, significantly downregulation of total cellular cholesterol content (tccCHOL). Furthermore, the expression levels of CD16 and CD40 were significantly downregulated in ASA-treated MOs. CONCLUSIONS: We show for the first time that ASA immunomodulates the functional activities of MOs during MI and promotes their switching toward a classical phenotype, exhibiting low CD16 expression levels and thereby anti-inflammatory properties.


Subject(s)
Anti-Inflammatory Agents/therapeutic use , Aspirin/therapeutic use , Autoimmune Diseases/drug therapy , Inflammation/drug therapy , Monocytes/immunology , Myocardial Infarction/drug therapy , CD40 Antigens/metabolism , Calcium/metabolism , Cells, Cultured , Cytokines/metabolism , Down-Regulation , Humans , Immunomodulation , Nitric Oxide Synthase Type II/metabolism , Receptors, IgG/metabolism
4.
Immunol Lett ; 179: 122-130, 2016 11.
Article in English | MEDLINE | ID: mdl-27717877

ABSTRACT

BACKGROUND: Type 1 diabetes (T1D) is associated with an imbalance between inflammation and repair. Recently, the biologically active form of vitamin D3, i.e. 1,25(OH)2D3, has been reported to have potent immunomodulatory effects on both innate and adaptive immune cells, as well as on the production of their specific cytokines. METHODS: We examined the effect of 1,25(OH)2D3 on the production of proinflammatory Th1/Th17 and anti-inflammatory Th2/Treg related cytokines, as well as on the phosphorylation of monocyte-expressed STAT4 and STAT6 at the recent-onset human T1D. RESULTS: The levels of IFN-γ, IL-17 and nitric oxide (NO) production were significantly increased in peripheral blood mononuclear cells (PBMCs) from patients with T1D compared to controls. Similarly, STAT4 tyrosine phosphorylation (p-STAT4, Tyr693) levels were significantly increased in monocytes from patients when compared to controls. Conversely, the levels of IL-4, IL-10 and p-STAT6 (Tyr641) were significantly decreased in type 1 diabetic patients than in controls. Treatment with 1,25(OH)2D3 resulted in significant up-regulation of IL-4, IL-10, arginase activity, and p-STAT6 and, conversely, down-regulation of IFN-γ, IL-17 and NO production levels, as well as p-STAT4. Additionally, 1,25(OH)2D3 significantly enhanced Treg-to-Th17 ratio, and induced a significant decrease in Th1-to-Th2, NO production-to-arginase activity and p-STAT4-to-p-STAT6 ratios. CONCLUSIONS: Our study suggests that the biologically active form of vitamin D can reverse the activation of inflammatory pathways at the onset of T1D. Additionally, its immunomodulation properties may vary depending on the overall patterns of cytokines. From a therapeutic point of view, vitamin D may potentially be suggested as an immunological adjuvant and a potential anti-inflammatory agent in individuals at risk of T1D.


Subject(s)
Calcitriol/metabolism , Cytokines/metabolism , Diabetes Mellitus, Type 1/metabolism , Inflammation Mediators/metabolism , Monocytes/metabolism , Nitric Oxide/metabolism , STAT6 Transcription Factor/metabolism , Arginase/metabolism , Calcitriol/pharmacology , Diabetes Mellitus, Type 1/immunology , Humans , Leukocytes, Mononuclear/drug effects , Leukocytes, Mononuclear/immunology , Leukocytes, Mononuclear/metabolism , Monocytes/drug effects , Monocytes/immunology , Phosphorylation/drug effects , STAT4 Transcription Factor/metabolism , T-Lymphocyte Subsets/drug effects , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism
5.
Int Immunopharmacol ; 30: 94-101, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26655879

ABSTRACT

BACKGROUND: The bioactive form of vitamin D3, i.e.1,25-dihydroxyvitamin D3 (1,25(OH)2D3) vitamin D has been shown to modulate monocytes/macrophages physiology and its response against bacterial infections. Pseudomonas aeruginosa (P. aeruginosa) is an opportunistic bacterial pathogen that can most frequently be fatal in immunocompromised infected people. METHODS: We investigated the ex vivo effect of 1,25(OH)2D3 on monocyte-derived macrophages function against P. aeruginosa infection. RESULTS: Relative vitamin D receptor (VDR) mRNA expression was significantly increased in infected and 1,25(OH)2D3-treated macrophages compared to controls (p<0.01). Treatment with 1,25(OH)2D3 markedly resulted in up-regulation of nitric oxide (NO) and IL-1ß production and down-regulation of IL-10 levels (respectively, p=0.029, p=0.048 and p=0.008). Additionally, 1,25(OH)2D3 significantly increased M1/M2 macrophage ratio (p<0.05) and slightly reduced intracellular bacterial development. Furthermore, the arginase activity, P. aeruginosa phagocytosis and killing were significantly increased in cells that were both infected and 1,25(OH)2D3-treated compared to the infected, but not 1,25(OH)2D3-treated macrophages (respectively, p<0.001, p<0.01 and p<0.001). CONCLUSIONS: We show in this study that bioactive from of vitamin D [1,25-dihydroxyvitamin D3 (1,25D3)] can enhance M1 macrophage polarization and their bactericidal protective activity against P. aeruginosa. Future works would involve improving the treatment response through dose-dependent effect studies, both in ex vivo and in vivo models.


Subject(s)
Calcitriol/pharmacology , Macrophages/drug effects , Pseudomonas Infections/immunology , Pseudomonas aeruginosa/physiology , Receptors, Calcitriol/metabolism , Cell Differentiation/drug effects , Cells, Cultured , Cytotoxicity, Immunologic/drug effects , Gene Expression Regulation/drug effects , Humans , Interleukin-10/genetics , Interleukin-10/metabolism , Interleukin-1beta/genetics , Interleukin-1beta/metabolism , Macrophages/immunology , Phagocytosis/drug effects , Pseudomonas Infections/therapy , Pseudomonas aeruginosa/drug effects , Receptors, Calcitriol/genetics , Th1 Cells/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...