Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 48
Filter
1.
Cells ; 11(6)2022 03 09.
Article in English | MEDLINE | ID: mdl-35326386

ABSTRACT

At inflammatory loci, pro-inflammatory activation of macrophages produces large amounts of reactive oxygen species (ROS) that induce DNA breaks and apoptosis. Given that M-CSF and GM-CSF induce two different pathways in macrophages, one for proliferation and the other for survival, in this study we wanted to determine if these growth factors are able to protect against the DNA damage produced during macrophage activation. In macrophages treated with DNA-damaging agents we found that GM-CSF protects better against DNA damage than M-CSF. Treatment with GM-CSF resulted in faster recovery of DNA damage than treatment with M-CSF. The number of apoptotic cells induced after DNA damage was higher in the presence of M-CSF. Protection against DNA damage by GM-CSF is not related to its higher capacity to induce proliferation. GM-CSF induces differentiation markers such as CD11c and MHCII, as well as the pro-survival Bcl-2A1 protein, which make macrophages more resistant to DNA damage.


Subject(s)
Granulocyte-Macrophage Colony-Stimulating Factor , Macrophage Colony-Stimulating Factor , Cell Differentiation , DNA Damage , Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Granulocyte-Macrophage Colony-Stimulating Factor/pharmacology , Macrophage Colony-Stimulating Factor/metabolism , Macrophage Colony-Stimulating Factor/pharmacology , Macrophages/metabolism
2.
EMBO Mol Med ; 10(7)2018 07.
Article in English | MEDLINE | ID: mdl-29907597

ABSTRACT

The protein kinase p38α plays a key role in cell homeostasis, and p38α signaling in intestinal epithelial cells protects against colitis-induced tumorigenesis. However, little is known on the contribution of p38α signaling in intestinal stromal cells. Here, we show that myeloid cell-specific downregulation of p38α protects mice against inflammation-associated colon tumorigenesis. The reduced tumorigenesis correlates with impaired detection in the colon of crucial chemokines for immune cell recruitment. We identify insulin-like growth factor-1 (IGF-1) as a novel mediator of the p38α pathway in macrophages. Moreover, using genetic and pharmacological approaches, we confirm the implication of IGF-1 produced by myeloid cells in colon inflammation and tumorigenesis. We also show a correlation between IGF-1 pathway activation and the infiltration of myeloid cells with active p38α in colon samples from patients with ulcerative colitis or colon cancer. Altogether, our results uncover an important role for myeloid IGF-1 downstream of p38α in colitis-associated tumorigenesis and suggest the interest in evaluating IGF-1 therapies for inflammation-associated intestinal diseases, taking into consideration IGF-1 signaling and immune cell infiltration in patient biopsies.


Subject(s)
Carcinogenesis/metabolism , Colitis/complications , Colonic Neoplasms/etiology , Insulin-Like Growth Factor I/metabolism , Intestinal Mucosa/metabolism , Mitogen-Activated Protein Kinase 14/metabolism , Myeloid Cells/metabolism , Animals , Carcinogenesis/immunology , Chemokines/metabolism , Colitis/immunology , Colitis/metabolism , Colonic Neoplasms/immunology , Colonic Neoplasms/metabolism , Down-Regulation , Female , Humans , Intestines , Male , Mice , Mice, Inbred C57BL , Signal Transduction
3.
Front Mol Neurosci ; 10: 14, 2017.
Article in English | MEDLINE | ID: mdl-28174520

ABSTRACT

Medical marijuana is increasingly prescribed as an analgesic for a growing number of indications, amongst which terminal cancer and multiple sclerosis. However, the mechanistic aspects and properties of cannabis remain remarkably poorly characterized. In this study we aimed to investigate the immune-cell modulatory properties of medical cannabis. Healthy volunteers were asked to ingest medical cannabis, and kinome profiling was used to generate comprehensive descriptions of the cannabis challenge on inflammatory signal transduction in the peripheral blood of these volunteers. Results were related to both short term and long term effects in patients experimentally treated with a medical marijuana preparation for suffering from abdominal pain as a result of chronic pancreatitis or other causes. The results reveal an immunosuppressive effect of cannabinoid preparations via deactivation of signaling through the pro-inflammatory p38 MAP kinase and mTOR pathways and a concomitant deactivation of the pro-mitogenic ERK pathway. However, long term cannabis exposure in two patients resulted in reversal of this effect. While these data provide a powerful mechanistic rationale for the clinical use of medical marijuana in inflammatory and oncological disease, caution may be advised with sustained use of such preparations.

4.
Methods Mol Biol ; 1417: 169-83, 2016.
Article in English | MEDLINE | ID: mdl-27221489

ABSTRACT

Intracellular signaling and cellular activation have been demonstrated to reside on multi-protein complexes rather than in isolated proteins. Consequently, techniques to resolve these complexes have gained much attention over the last few years. Förster Resonance Energy Transfer (FRET) coupled with Fluorescence Lifetime Imaging Microscopy (FLIM) is a powerful tool to discriminate direct interactions between two proteins within a multi-protein complex. Here, we present the use of FRET-FLIM as an experimental tool for the interpretation of the inflammasome composition. We also introduce some considerations required for the correct use of this technique and the control experiments that should be implemented.


Subject(s)
Fluorescence Resonance Energy Transfer/methods , Inflammasomes/metabolism , NLR Proteins/chemistry , Inflammasomes/chemistry , Microscopy, Fluorescence/methods , NLR Proteins/metabolism , Protein Interaction Mapping , Protein Multimerization
5.
Food Funct ; 6(4): 1176-84, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25700085

ABSTRACT

Probiotics have been used as alternative therapies in intestinal inflammatory disorders. Many studies have shown that different bacterial probiotic strains possess immuno-modulatory and anti-inflammatory properties. However, there is an increasing interest in the use of non-viable bacteria to reduce the risk of microbial translocation and infection. The aim of this study was to evaluate whether the viability of L. fermentum CECT5716 is essential to exert its intestinal anti-inflammatory effect. We compared the preventative effects of viable and non-viable probiotic in the TNBS model of rat colitis. In vitro studies were also performed in Caco-2 and RAW 264.7 cells to evaluate the probiotic effects on IL-8, IL-1ß and nitrite production, and p44/42 and p38 MAP kinase protein expressions. In vitro results revealed a decrease in the stimulated production of pro-inflammatory mediators regardless of the viability of the probiotic. Likewise, both forms of the probiotic administered to colitic rats produced a significant reduction of IL-1ß and TNF-α levels and colonic iNOS expression. In conclusion, both live and dead L. fermentum CECT5716 have been demonstrated to attenuate the inflammatory process and diminish the production of some of the inflammatory mediators. In fact, the viability of this probiotic did not affect its immuno-modulatory and anti-inflammatory properties.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Limosilactobacillus fermentum , Microbial Viability , Probiotics , Animals , Caco-2 Cells , Colitis/microbiology , Colitis/therapy , Female , Gastrointestinal Microbiome , Humans , Immunomodulation , Interleukin-1beta/antagonists & inhibitors , Interleukin-1beta/metabolism , Interleukin-8/antagonists & inhibitors , Interleukin-8/metabolism , Intestinal Mucosa/metabolism , Intestines/microbiology , Mice , Mitogen-Activated Protein Kinase 1/antagonists & inhibitors , Mitogen-Activated Protein Kinase 1/genetics , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/antagonists & inhibitors , Mitogen-Activated Protein Kinase 3/genetics , Mitogen-Activated Protein Kinase 3/metabolism , Nitric Oxide/antagonists & inhibitors , Nitric Oxide/metabolism , Nitric Oxide Synthase Type II/genetics , Nitric Oxide Synthase Type II/metabolism , RAW 264.7 Cells , Rats , Rats, Wistar , Trinitrobenzenesulfonic Acid/adverse effects , Tumor Necrosis Factor-alpha/metabolism , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors , p38 Mitogen-Activated Protein Kinases/genetics , p38 Mitogen-Activated Protein Kinases/metabolism
6.
Int Arch Allergy Immunol ; 162(3): 214-24, 2013.
Article in English | MEDLINE | ID: mdl-24021980

ABSTRACT

BACKGROUND: Previous studies have indicated that colitis increases intestinal permeability to food antigens. This condition also generates an immunoreactive milieu in the gut, which may exacerbate or counteract allergy reactions. This, along with the fact that both colitis and allergy are being codiagnosed more frequently, means the scientific interest on the immune relation between these pathologies is increasing. We evaluated the immune response to an internalized food antigen that was initiated during a concomitant active intestinal inflammatory response. METHODS: An ovalbumin (OVA)-induced immune response was analyzed in healthy mice and in mice suffering from colitis induced by the administration of dinitrofluorobenzene/dinitrosulfonic acid (DNFB/DNS) at the moment of OVA challenge. The OVA-induced clinical score and allergy response both in plasma and in splenocyte cultures from these animals were compared. RESULTS: Although no differences were observed in the allergy clinical score, the concomitant active colitis led to an increase in the immune response to OVA antigen, as shown by increased spleen size and OVA-induced splenocyte proliferation, exacerbated expression of total and OVA-specific IgG1 levels, increased colonic IL-4 expression and OVA-induced IL-4 and IL-5 cytokine expression in spleen cells. CONCLUSIONS: Our results indicate that animals with active colitis undergo an exacerbated immune response to an internalized antigen. This finding could be relevant for the allergy management of patients presenting simultaneously with chronic colitis.


Subject(s)
Antigens/immunology , Colitis/immunology , Ovalbumin/immunology , Animals , Colitis/chemically induced , Cytokines/biosynthesis , Disease Models, Animal , Female , Hypersensitivity/immunology , Immunoglobulin E/blood , Immunoglobulin E/immunology , Immunoglobulin G/blood , Immunoglobulin G/immunology , Lymphocyte Activation , Lymphocytes/immunology , Mice , Spleen/immunology
7.
Eur J Immunol ; 42(8): 1938-48, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22865045

ABSTRACT

Macrophages play a central role in the immune response. These cells either proliferate in response to, for example, growth factors or become activated in response to, for example, LPS and develop functional activities. Experiments carried out in mice showed that macrophage proliferation requires a short period of ERK phosphorylation, while an extended period is required for macrophage activation. The length of phosphorylation is controlled by the MAPK phosphatase-1 (MKP-1), a nuclear-localized dual-specificity phosphatase that dephosphorylates the MAPKs ERK, p38, and c-Jun NH(2) -terminal kinase (JNK). MKP-1 is induced in macrophages by growth factors, as well as by activators such as LPS, but with different kinetics; to achieve the different functional outcomes (proliferation versus activation), the inhibition of MKP-1 by cytokines such as IFN-γ blocks macrophage proliferation and induces activation. The data presented in this review show that this phosphatase is the switch between macrophage proliferation and activation.


Subject(s)
Dual Specificity Phosphatase 1/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , Macrophage Activation , Macrophages/immunology , Macrophages/metabolism , Animals , Cell Proliferation , Dual Specificity Phosphatase 1/antagonists & inhibitors , Humans , Interferon-gamma , MAP Kinase Signaling System , Mice , Phosphorylation
8.
Eur J Immunol ; 42(11): 3028-37, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22865229

ABSTRACT

The amount of arginine available at inflammatory loci is a limiting factor for the growth of several cells of the immune system. IL-4-induced activation of macrophages produced arginase-1, which converts arginine into ornithine, a precursor of polyamines and proline. Trichostatin A (TSA), a pan-inhibitor of histone deacetylases (HDACs), inhibited IL-4-induced arginase-1 expression. TSA showed promoter-specific effects on the IL-4-responsive genes. While TSA inhibited the expression of arginase-1, fizz1, and mrc1, other genes, such as ym,1 mgl1, and mgl2, were not affected. The inhibition of arginase-1 occurred at the transcriptional level with the inhibition of polymerase II binding to the promoter. IL-4 induced STAT6 phosphorylation and binding to DNA. These activities were not affected by TSA treatment. However, TSA inhibited C/EBPß DNA binding. This inhibitor induced acetylation on lysine residues 215-216, which are critical for DNA binding. Finally, using macrophages from STAT6 KO mice we showed that STAT6 is required for the DNA binding of C/EBPß. These results demonstrate that the acetylation/deacetylation balance strongly influences the expression of arginase-1, a gene of alternative activation of macrophages. These findings also provide a molecular mechanism to explain the control of gene expression through deacetylase activity.


Subject(s)
Arginase/biosynthesis , CCAAT-Enhancer-Binding Protein-beta/metabolism , Histone Deacetylase Inhibitors/pharmacology , Interleukin-4/pharmacology , Macrophages/immunology , Acetylation , Animals , Arginase/genetics , Arginase/immunology , CCAAT-Enhancer-Binding Protein-beta/immunology , Hydroxamic Acids/pharmacology , Macrophage Activation/drug effects , Macrophage Activation/immunology , Macrophages/drug effects , Macrophages/enzymology , Male , Mice , Mice, Inbred BALB C , Mice, Knockout , Phosphorylation , RNA/chemistry , RNA/genetics , Real-Time Polymerase Chain Reaction , STAT6 Transcription Factor/immunology , Statistics, Nonparametric
9.
J Immunol Methods ; 381(1-2): 41-9, 2012 Jul 31.
Article in English | MEDLINE | ID: mdl-22542400

ABSTRACT

Cow's milk protein allergy (CMPA) is one of the most prevalent human food-borne allergies, particularly in children. Experimental animal models have become critical tools with which to perform research on new therapeutic approaches and on the molecular mechanisms involved. However, oral food allergen sensitization in mice requires several weeks and is usually associated with unspecific immune responses. To overcome these inconveniences, we have developed a new food allergy model that takes only two weeks while retaining the main characters of allergic response to food antigens. The new model is characterized by oral sensitization of weaned Balb/c mice with 5 doses of purified cow's milk protein (CMP) plus cholera toxin (CT) for only two weeks and posterior challenge with an intraperitoneal administration of the allergen at the end of the sensitization period. In parallel, we studied a conventional protocol that lasts for seven weeks, and also the non-specific effects exerted by CT in both protocols. The shorter protocol achieves a similar clinical score as the original food allergy model without macroscopically affecting gut morphology or physiology. Moreover, the shorter protocol caused an increased IL-4 production and a more selective antigen-specific IgG1 response. Finally, the extended CT administration during the sensitization period of the conventional protocol is responsible for the exacerbated immune response observed in that model. Therefore, the new model presented here allows a reduction not only in experimental time but also in the number of animals required per experiment while maintaining the features of conventional allergy models. We propose that the new protocol reported will contribute to advancing allergy research.


Subject(s)
Cholera Toxin/immunology , Disease Models, Animal , Food Hypersensitivity/immunology , Milk Proteins/immunology , Administration, Oral , Animals , Cattle , Cholera Toxin/administration & dosage , Diarrhea/etiology , Diarrhea/immunology , Enzyme-Linked Immunosorbent Assay , Female , Food Hypersensitivity/complications , Food Hypersensitivity/diagnosis , Histamine/blood , Histamine/immunology , Humans , Immunoglobulin G/blood , Immunoglobulin G/immunology , Injections, Intraperitoneal , Interleukin-4/blood , Interleukin-4/immunology , Mice , Mice, Inbred BALB C , Milk Hypersensitivity/complications , Milk Hypersensitivity/diagnosis , Milk Hypersensitivity/immunology , Milk Proteins/administration & dosage , Sensitivity and Specificity , Time Factors
10.
Methods Mol Biol ; 844: 223-35, 2012.
Article in English | MEDLINE | ID: mdl-22262446

ABSTRACT

In order to perform their functions, macrophages must be activated either by Th1-type cytokines, such as interferon-gamma which is called classical activation or M1, or by Th2-type cytokines, such as IL-4, IL-10, IL-13, etc. referred as alternative activation or M2. In all of these conditions, macrophages require the uptake of exogenous arginine to meet their metabolic demands. Depending on the intracellular availability of this amino acid, the activities of these cells are differentially modulated. In this regard, macrophage activation requires this amino acid for the synthesis of proteins, production of nitric oxide via classical activation, and production of polyamines and proline through alternative activation. Therefore, the study of the arginine transport for amino acid system transporters may be a key regulatory step for physiological responses in macrophages. In this chapter, we present simple and direct methods to determine the mRNA expression and activity of arginine transporters. Moreover, we describe a direct method to measure the arginine catabolism using thin-layer chromatography.


Subject(s)
Arginine/metabolism , Macrophage Activation/immunology , Macrophages/immunology , Amino Acid Transport System y+/genetics , Amino Acid Transport System y+/metabolism , Animals , Cells, Cultured , Macrophage Activation/genetics , Macrophages/metabolism , Mice , RNA, Messenger/metabolism
11.
Eur J Nutr ; 51(3): 365-74, 2012 Apr.
Article in English | MEDLINE | ID: mdl-21688122

ABSTRACT

PURPOSE: The aim of this study was to better characterise the biological effects of Lactobacillus salivarius ssp. salivarius CECT5713, a probiotic with immunomodulatory properties. METHODS: Live or dead probiotic was assayed in the TNBS model of rat colitis to determine whether viability was a requisite to exert the beneficial effects. In vitro studies were also performed in Caco-2 cells to evaluate its effects on epithelial cell recovery and IL-8 production. Finally, the probiotic was assayed in the LPS model of septic shock in mice to establish its effects when there is an altered systemic immune response. RESULTS: The viability of the probiotic was required for its anti-inflammatory activity. The probiotic inhibited IL-8 production in stimulated Caco-2 cells and facilitated the recovery of damaged intestinal epithelium. In LPS-treated mice, the probiotic inhibited the production of TNFα in plasma and lungs and increased the hepatic glutathione content. These effects were associated with an improvement in the altered production of the T-cell cytokines in splenocytes, by reducing IL-2 and IL-5 and by increasing IL-10. Finally, it reduced the increased plasma IgG production in LPS-treated mice. CONCLUSION: The anti-inflammatory effects of viable L. salivarius ssp. salivarius CECT5713 are not restricted to the gastrointestinal tract.


Subject(s)
Colitis/therapy , Immunologic Factors/administration & dosage , Intestine, Large/microbiology , Lactobacillus/metabolism , Probiotics/administration & dosage , Animals , Anti-Inflammatory Agents/administration & dosage , Caco-2 Cells , Female , Glutathione/analysis , Humans , Immunoglobulin G/metabolism , Interleukin-10/metabolism , Interleukin-5/metabolism , Interleukin-8/metabolism , Intestinal Mucosa/metabolism , Lactobacillus/growth & development , Lipopolysaccharides/metabolism , Liver/metabolism , Male , Mice , Mice, Inbred BALB C , Rats , Rats, Wistar , Shock, Septic/pathology , Shock, Septic/prevention & control , Tumor Necrosis Factor-alpha/antagonists & inhibitors , Tumor Necrosis Factor-alpha/blood
12.
Biochem Pharmacol ; 82(12): 1891-900, 2011 Dec 15.
Article in English | MEDLINE | ID: mdl-21930116

ABSTRACT

Antibiotics have been empirically used for human inflammatory bowel disease, being limited to short periods. Probiotics are able to attenuate intestinal inflammation due to its immunomodulatory properties, being considered as safe when chronically administered. The aim was to test the association of minocycline, a tetracycline with immunomodulatory properties, and the probiotic Escherichia coli Nissle 1917 (EcN) in a mouse model of reactivated colitis. For this purpose, female C57BL/6J mice were assigned to different groups: non-colitic and dextran sodium sulfate (DSS)-control groups (without treatment), minocycline (50 mg/kg/day; p.o.), EcN (5×10(8) CFU/day; p.o.), and minocycline plus EcN treated groups. Colitis was induced by adding DSS in the drinking water (3%) for 5 days; 2 weeks later, colitis was reactivated by subsequent exposure to DSS. The inflammatory status was evaluated daily by a disease activity index (DAI); colonic damage was assessed histologically and biochemically by evaluating mRNA relative expression of different mediators by qPCR. Finally, a microbiological analysis of the colonic contents was performed. Minocycline and EcN exerted intestinal anti-inflammatory effect and attenuated the reactivation of the colitis, as shown by the reduced DAI values, being these effects greater when combining both treatments. This was evidenced histologically and biochemically, by reduced expression of TNFα, IL-1ß, IL-2, MIP-2, MCP-1, ICAM-1, iNOS and MMP-9, together with increased MUC-3 and ZO-1 expression. Finally, the altered microbiota composition of colitic mice was partially restored after the different treatments. In conclusion, EcN supplementation to minocycline treatment improves the recovery of the intestinal damage and prevents the reactivation of experimental colitis.


Subject(s)
Colitis/chemically induced , Dextran Sulfate/toxicity , Escherichia coli/classification , Escherichia coli/physiology , Minocycline/pharmacology , Probiotics/pharmacology , Animals , Anti-Bacterial Agents/therapeutic use , Colitis/drug therapy , Colon/drug effects , Colon/pathology , Cytokines/genetics , Cytokines/metabolism , Disease Models, Animal , Drug Therapy, Combination , Female , Gene Expression Regulation/drug effects , Inflammation/drug therapy , Matrix Metalloproteinase 9/genetics , Matrix Metalloproteinase 9/metabolism , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Mice, Inbred C57BL , Mucin-3/genetics , Mucin-3/metabolism , Nitric Oxide Synthase Type II/genetics , Nitric Oxide Synthase Type II/metabolism , Phosphoproteins/genetics , Phosphoproteins/metabolism , Probiotics/classification , Zonula Occludens-1 Protein
13.
Inflamm Bowel Dis ; 17(10): 2087-101, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21910170

ABSTRACT

BACKGROUND: The dinitrofluorobenzene/dinitrosulfonic acid (DNFB/DNS) model was originally described as an experimental model of intestinal inflammation resembling human ulcerative colitis (UC). Due to the absence of acceptable UC experimental models for pharmacological preclinical assays, here we examine the immune response induced in this model. METHODS: Balb/c mice were sensitized by skin application of DNFB on day 1, followed by an intrarectal challenge with DNS on day 5. We further expanded this model by administering a second DNS challenge on day 15. The features of colonic inflammation and immune response were evaluated. RESULTS: The changes observed in colonic tissue corresponded, in comparison to the trinitrobenzene sulfonic acid (TNBS) colitis model, to a mild mucosal effect in the colon, which spontaneously resolved in less than 5 days. Furthermore, the second hapten challenge did not exacerbate the inflammatory response. In contrast to other studies, we did not observe any clear involvement of tumor necrosis factor alpha (TNF-α) or other Th1 cytokines during the initial inflammatory response; however, we found that a more Th2-humoral response appeared to mediate the first contact with the hapten. An increased humoral response was detected during the second challenge, although an increased Th1/Th17-cytokine expression profile was also simultaneously observed. CONCLUSIONS: On the basis of these results, although the DNFB/DNS model can display some features found in human UC, it should be considered as a model for the study of the intestinal hypersensitivity seen, for example, during food allergy or irritable bowel syndrome but not intestinal inflammation per se.


Subject(s)
Benzenesulfonates/toxicity , Colitis/chemically induced , Dinitrofluorobenzene/toxicity , Disease Models, Animal , Haptens/toxicity , Inflammation/chemically induced , Intestinal Mucosa/drug effects , Animals , Colitis/immunology , Colitis/pathology , Cytokines/genetics , Cytokines/metabolism , Drug Hypersensitivity , Humans , Immunoenzyme Techniques , Inflammation/immunology , Inflammation/pathology , Intestinal Mucosa/immunology , Intestinal Mucosa/pathology , Lymphocytes/immunology , Lymphocytes/metabolism , Lymphocytes/pathology , Male , Mast Cells/immunology , Mast Cells/metabolism , Mast Cells/pathology , Mice , Mice, Inbred BALB C
14.
Pharmacol Res ; 63(4): 308-19, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21193045

ABSTRACT

Some antibiotics, including minocycline, have recently been reported to display immunomodulatory properties in addition to their antimicrobial activity. The use of a compound with both immunomodulatory and antibacterial properties could be very interesting in the treatment of inflammatory bowel disease (IBD), so the aim of our study was to evaluate the anti-inflammatory effect of minocycline in several experimental models of IBD. Firstly, the immunomodulatory activity of the antibiotic was tested in vitro using Caco-2 intestinal epithelial cells and RAW 264.7 macrophages; minocycline was able to inhibit IL-8 and nitrite production, respectively. In vivo studies were performed in trinitrobenzenesulfonic acid (TNBS)-induced rat colitis and dextran sodium sulfate (DSS)-induced mouse colitis. The results revealed that minocycline exerted an intestinal anti-inflammatory effect when administered as a curative treatment in the TNBS model, modulating both immune and microbiological parameters, being confirmed in the DSS model; whereas none of the other antibiotics tested (tetracycline and metronidazole) showed anti-inflammatory effect. However, minocycline administration before the colitis induction was not able to prevent the development of the intestinal inflammation, thus showing that only its antimicrobial activity is not enough for the anti-inflammatory effect. In conclusion, minocycline displays an anti-inflammatory effect on different models of rodent colitis which could be attributed to the association of its antibacterial and immunomodulatory properties.


Subject(s)
Anti-Inflammatory Agents/therapeutic use , Colitis/drug therapy , Immunologic Factors/therapeutic use , Minocycline/therapeutic use , Animals , Anti-Inflammatory Agents/pharmacology , Caco-2 Cells , Cell Line , Colitis/chemically induced , Colitis/pathology , Dextran Sulfate , Female , Humans , Immunologic Factors/pharmacology , Inflammatory Bowel Diseases/drug therapy , Macrophages/drug effects , Mice , Mice, Inbred C57BL , Minocycline/pharmacology , Rats , Rats, Wistar , Trinitrobenzenesulfonic Acid
16.
Adv Immunol ; 108: 1-20, 2010.
Article in English | MEDLINE | ID: mdl-21056727

ABSTRACT

Macrophages play key roles in inflammation. During the onset of the inflammatory process, these phagocytic cells become activated and have destructive effects. Macrophage activation, which involves the induction of more than 400 genes, results in an increased capacity to eliminate bacteria and to regulate many other cells through the release of cytokines and chemokines. However, excessive activation has damaging effects, such as septic shock, which can lead to multiple organ dysfunction syndrome and death. In other situations, persistence of proinflammatory activity results in the development of chronic inflammation, such as rheumatoid arthritis, psoriasis, and inflammatory bowel disease. To prevent undesirable effects, several mechanisms have evolved to control the excess of activation, thereby leading to macrophage deactivation and the resolution of inflammation. In this review, we discuss several mechanisms that mediate macrophage deactivation.


Subject(s)
Inflammation/physiopathology , Macrophages/cytology , Macrophages/immunology , Animals , Humans , Inflammation Mediators/immunology , Macrophage Activation
17.
Clin Nutr ; 29(6): 832-9, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20605664

ABSTRACT

Different types of dietary fiber can be distinguished considering their rate of fermentability, thus determining the location of the large intestine where they exert their beneficial effect. Their combination could be interesting to obtain health-promoting effects throughout the entire colon. The aim of the present study was to evaluate the synergistic effect of two dietary fibers with different fermentation patterns, fructooligosaccharides (FOS) (Beneo(®)-95) and resistant starch (Fibersol(®)-2), after their administration to healthy rats or in trinitrobenzenesulphonic acid-(TNBS) colitic rats, with an altered colonic immune response. In healthy rats, the administration of the combination of FOS and resistant starch induced changes in the intestinal microbiota, by increasing lactobacilli and bifidobacteria in caecum and colonic contents. Furthermore, its administration up-regulated the expression of the trefoil factor-3 and MUC-2 in comparison with untreated rats, thus improving the intestinal barrier function. The beneficial effects observed with this combination were confirmed in the TBNS model of rat colitis, since it was able to exert intestinal anti-inflammatory effect, associated with an increase of protective bacteria and up-regulation of epithelial defense mechanisms. In conclusion, the combination of two different dietary fibers may result in a synergistic prebiotic effect, and may confer greater health benefits to the host.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Colitis/microbiology , Oligosaccharides/metabolism , Prebiotics/microbiology , Starch/metabolism , Animals , Colitis/metabolism , Colon/metabolism , Colon/microbiology , Dietary Fiber/pharmacology , Disease Models, Animal , Female , Fermentation , Lactobacillus/isolation & purification , Rats , Rats, Wistar , Up-Regulation
18.
Immunobiology ; 215(11): 863-73, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20149475

ABSTRACT

Survival and proliferation signals are two processes closely interrelated and finely controlled in most cell types, whose deregulation may lead to carcinogenesis. In the last decade, different studies have suggested that both cellular functions are also intimately associated with other cellular activities such as differentiation and cellular activation, especially in immune cells. The aim of this study was to evaluate the effects of the short-chain fatty acid (SCFA) butyrate on the proliferation and activation state of different cell types involved in inflammatory bowel disease. We focused on intestinal epithelial cells, macrophages and T-lymphocytes, using both primary non-transformed cultures and established cell lines. The results showed that low concentrations of butyrate inhibited the proliferation of all the immune cell types tested in this work, whereas it only induced apoptosis in activated T-lymphocytes, non-differentiated epithelial cells and macrophage cell lines, but not in differentiated epithelial cells or primary macrophages. Butyrate apoptosis induction was mediated by caspase-3/7 activation. This SCFA was only able to modify cell activation, measured as expression of inflammatory cytokines, in those cell types in which apoptosis was induced. In conclusion, our results suggest a cell type-specificity of the immune-modulatory effects of butyrate based on the proliferation/activation characteristic physiology of these processes in different cells types.


Subject(s)
Apoptosis/physiology , Butyrates/pharmacology , Cell Proliferation/drug effects , Immunologic Factors/pharmacology , Inflammatory Bowel Diseases/immunology , Lymphocyte Activation/drug effects , Animals , Caspase 3/metabolism , Caspase 7/metabolism , Cells, Cultured , Dose-Response Relationship, Immunologic , Epithelial Cells/immunology , Epithelial Cells/pathology , HT29 Cells , Humans , Inflammatory Bowel Diseases/pathology , Macrophages/immunology , Male , Mice , Organ Specificity , T-Lymphocytes/immunology
19.
J Pharmacol Sci ; 109(2): 315-8, 2009 Feb.
Article in English | MEDLINE | ID: mdl-19234369

ABSTRACT

The aim of the present study was to evaluate the inmunomodulatory effects of UR-1505, a new salicylate derivative, on the T helper (Th)2/humoral response produced during dextran sodium sulfate (DSS)-induced rat colitis. In the in vitro studies, UR-1505 (300 microM) inhibited both the production of interleukin (IL)-10 and IL-5 in concanavalin A (Con A)-activated splenocytes and the production of immunoglobulin (Ig) G and IgA by B-lymphocytes. However, in contrast to the in vitro results, the administration of UR-1505 (10 and 30 mg/kg per day) to rats with established DSS-colitis enhanced both IL-10 and IgA production, whereas it inhibited IgG production, thus ameliorating the intestinal inflammation.


Subject(s)
Anti-Inflammatory Agents/therapeutic use , Colitis/drug therapy , Salicylates/therapeutic use , Th2 Cells/immunology , Animals , Colitis/chemically induced , Colitis/immunology , Colitis, Ulcerative/drug therapy , Colitis, Ulcerative/immunology , Colon/metabolism , Cytokines/antagonists & inhibitors , Cytokines/metabolism , Dextran Sulfate , Disease Models, Animal , Immunoglobulin A/metabolism , Immunoglobulin G/metabolism , Rats
20.
Br J Nutr ; 101(1): 51-8, 2009 Jan.
Article in English | MEDLINE | ID: mdl-18445307

ABSTRACT

The preventative effects of the probiotic Lactobacillus fermentum CECT5716 were evaluated in the lipopolysaccharide (LPS) model of septic shock in mice. The probiotic was administered suspended in drinking water at the final concentration of 108 colony-forming units/ml for 2 weeks before the induction of an endotoxic shock by an intraperitoneal injection of LPS (400 microg/200 microl per mouse). Blood and different organs were collected after 24 h to evaluate the severity of the endotoxic shock and the preventative effects of the probiotic. L. fermentum reduced TNF-alpha levels in blood, which promotes the major alterations observed during septic shock, as well as the infiltration of activated neutrophils into the lungs. Furthermore, free radical overproduction and oxidative stress were associated with a significant decrease in hepatic glutathione levels in septic mice, and with an excessive NO production attributed to the induction of the inducible isoform of NO synthase (iNOS). In fact, hepatic glutathione levels were significantly increased in the group of mice receiving the probiotic, and the increased iNOS expression both in the colon and lungs was down-regulated in those mice treated with L. fermentum. Finally, pre-treatment with L. fermentum may also exert its protective action modulating the expression of different cytokines in splenocyte-derived T cells such us IL-2, IL-5, IL-6 or IL-10. In conclusion, pre-treatment with L. fermentum may exert its protective action against LPS-induced organ damage in mice by a combination of several actions including its antioxidant properties and by reduction of the synthesis of the pro-inflammatory TNF-alpha and IL-6.


Subject(s)
Limosilactobacillus fermentum , Probiotics/therapeutic use , Shock, Septic/prevention & control , Animals , Cells, Cultured , Disease Models, Animal , Lipopolysaccharides , Lung/metabolism , Male , Mice , Mice, Inbred BALB C , Neutrophil Activation , Nitric Oxide Synthase Type II/metabolism , Spleen/metabolism , Tumor Necrosis Factor-alpha/biosynthesis , Tumor Necrosis Factor-alpha/blood
SELECTION OF CITATIONS
SEARCH DETAIL
...