Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
1.
Food Res Int ; 163: 112140, 2023 01.
Article in English | MEDLINE | ID: mdl-36596095

ABSTRACT

Systemic lupus erythematosus (SLE) is a multiorgan disorder with a deregulated immune-inflammatory response. Nutritional therapy has been considered a promising approach to SLE management. Oleocanthal (OLE), the main extra virgin olive oil (EVOO)-derived secoiridoid, has shown to regulate the immune-inflammatory response in various disease contexts; however, its possible beneficial effects on SLE remain unclear. This study sought to evaluate the effects of OLE enriched diet on renal damage and aortic endothelial dysfunction in murine pristane-induced SLE, focusing on the action mechanisms and signaling pathways involved. BALB/c mice were injected with pristane and fed with OLE supplemented diet (0.01 % (w/w)) for six months. Levels of cytokines were measured by ELISA in lipopolysaccharide (LPS)-stimulated peritoneal macrophages and splenocytes. Presence of immunoglobulin G (IgG) and IgM immune complexes were examined by immunofluorescence and immunohistochemistry. Thoracic aortas were used to evaluate endothelial dysfunction. Western blotting was employed to detect signaling pathways and oxidative-inflammatory-related mediators. Dietary OLE supplementation reduced Th1/Th17 pro-inflammatory cytokines production and alleviated renal damage by decreasing immunoglobulin complexes deposition, and inflammation-mediating enzymes expression. The mechanisms underlying these protective effects could be related to the regulation of nuclear factor erythroid 2-related factor 2/Haem oxygenase 1 (Nrf-2/HO-1), mitogen-activated protein kinases (MAPKs), signal transducer and transcription activator of transcription (STAT-3), inflammasome and, nuclear factor kappa B (NF-κB) signaling pathways. Also, dietary OLE improved aortic endothelial dysfunction and vascular reactivity, normalizing endothelial nitric oxide synthase (eNOS) uncoupling, and NADPH oxidase-1 (NOX-1) overexpression. This study shows the immunomodulatory effects of OLE in an in vivo model of SLE by improving renal damage and regulating aortic endothelial dysfunction. These preliminary results provide OLE as a new therapeutic strategy in SLE management.


Subject(s)
Lupus Erythematosus, Systemic , Animals , Mice , Diet , Dietary Supplements , Cytokines/metabolism
2.
Nutrients ; 13(2)2021 Jan 22.
Article in English | MEDLINE | ID: mdl-33499113

ABSTRACT

: Oleuropein (OL), an olive tree secoiridoid and its peracetylated derivate (Per-OL) have exhibited several beneficial effects on LPS-stimulated macrophages and murine experimental systemic lupus erythematosus (SLE). This study was designed to evaluate dietary Per-OL in comparison with OL supplementation effects on collagen-induced arthritis (CIA) murine model. Three-weeks-old DBA-1/J male mice were fed from weaning with a standard commercial diet or experimental enriched-diets in 0.05 % (w/w) OL, 0.05% and 0.025% Per-OL. After six weeks of pre-treatment, arthritis was induced by bovine collagen type II by tail base injection (day 0) and on day 21, mice received a booster injection. Mice were sacrificed 42 days after the first immunization. Both Per-OL and OL diets significantly prevented histological damage and arthritic score development, although no statistically significant differences were observed between both compounds. Also, serum collagen oligomeric matrix protein (COMP), metalloprotease (MMP)-3 and pro-inflammatory cytokines levels were ameliorated in paws from secoiridoids fed animals. Mitogen-activated protein kinases (MAPK)s and nuclear transcription factor-kappa-B (NF-κB) activations were drastically down-regulated whereas nuclear factor E2-related factor 2 (Nrf2) and heme-oxygenase-1 (HO-1) protein expressions were up-regulated in those mice fed with OL and Per-OL diets. We conclude that both Per-OL and its parent compound, OL, supplements might provide a basis for developing a new dietary strategy for the prevention of rheumatoid arthritis.


Subject(s)
Arthritis, Experimental/diet therapy , Inflammasomes/drug effects , Iridoid Glucosides/pharmacology , Animals , Arthritis, Experimental/chemically induced , Arthritis, Experimental/metabolism , Dietary Supplements , Disease Models, Animal , Heme Oxygenase-1/metabolism , Inflammasomes/metabolism , MAP Kinase Signaling System/drug effects , Male , Matrix Metalloproteinase 3/metabolism , Membrane Proteins/metabolism , Mice , Mice, Inbred DBA , NF-E2-Related Factor 2/metabolism , NF-kappa B/drug effects , Signal Transduction/drug effects
3.
Biol Blood Marrow Transplant ; 26(4): 615-624, 2020 04.
Article in English | MEDLINE | ID: mdl-31756538

ABSTRACT

Polyphenols are a group of chemical substances found in plants, with immunomodulatory, antiproliferative, and anti-inflammatory properties that might be useful in the prophylaxis and treatment of graft-versus-host disease (GVHD). Polyphenolic extract (PE) obtained from extra virgin olive oil (EVOO) decreased the activation and proliferation of activated T cells. In addition, a decreased production of proinflammatory cytokines was observed upon exposure to PE. Western blot assays showed a marked inhibition of Akt phosphorylation and nuclear translocation of NF-κB in activated T cells. In a murine model of acute GVHD, we observed that mice that received a diet supplemented in PE (600 ppm) presented a higher survival rate and lower risk of developing GVHD when compared with the group that received a control diet. Histopathologic examination showed a significantly lower gut involvement in mice receiving PE, with a decrease in proinflammatory cytokines (IL-2, IL-17, and TNF-α) in serum and the reestablishment of butyrate concentration in the gut. In conclusion, PE obtained from EVOO exerted a potent immunomodulatory effect, reducing the activation and proliferation of activated T cells and the production of proinflammatory cytokines. In a murine model of acute GVHD, a PE-supplemented diet reduced the incidence and severity of the disease and increased survival after transplantation.


Subject(s)
Graft vs Host Disease , Animals , Disease Models, Animal , Graft vs Host Disease/prevention & control , Mice , NF-kappa B , Olive Oil , Plant Extracts
4.
Planta Med ; 85(8): 670-677, 2019 May.
Article in English | MEDLINE | ID: mdl-31018218

ABSTRACT

Quercus ilex L. (Fagaceae) is one of the most commonly used plants in folk medicine in the Mediterranean region to treat gastrointestinal disorders. The aim of the present study was to evaluate the effects of a polyphenol extract from mature leaves of Q. ilex on acute 2,4,6-trinitrobenzene sulfonic acid-induced colitis in rats. A polyphenol extract from mature leaves of Q. ilex (250 and 500 mg/kg/day) was administered by gavage 48, 24, and 1 h prior to the induction of colitis with 2,4,6-trinitrobenzene sulfonic acid as well as 24 h later. The inflammation response was assessed by histology, myeloperoxidase activity, and Th1 proinflammatory cytokine production. The protein expression of cyclooxygenase-2 and inducible nitric oxide synthase, and signaling pathways were studied by Western blotting in the colon tissues. The polyphenol extract from mature leaves of Q. ilex decreased neutrophil infiltration, interleukin-1ß and TNF-α production, and proinflammatory proteins cyclooxygenase-2 and inducible nitric oxide synthase overexpression. Also, the polyphenol extract from mature leaves of Q. ilex was capable of blocking the activation of mitogen-activated protein kinases and nuclear transcription factor-kappa B. Furthermore, the reduction of inflammation by polyphenol extract from mature leaves of Q. ilex treatment was accompanied by a recovery of Nrf2 and heme oxygenase-1 protein expression levels. In conclusion, this study demonstrates that a polyphenol extract from mature leaves of Q. ilex improves 2,4,6-trinitrobenzene sulfonic acid-induced colitis, probably through mitogen-activated protein kinase/nuclear transcription factor-kappa B inhibition and Nrf2/heme oxygenase-1 activation signaling pathways, thus reducing the production of Th1 proinflammatory cytokines and cyclooxygenase-2 and inducible nitric oxide synthase overexpression.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Colitis/drug therapy , Phytotherapy , Plant Extracts/therapeutic use , Quercus/chemistry , Animals , Anti-Inflammatory Agents, Non-Steroidal/isolation & purification , Colitis/chemically induced , Inflammation Mediators/metabolism , MAP Kinase Signaling System/drug effects , Male , NF-E2-Related Factor 2/metabolism , Plant Leaves/chemistry , Protein Serine-Threonine Kinases/antagonists & inhibitors , Rats , Trinitrobenzenesulfonic Acid , NF-kappaB-Inducing Kinase
5.
Br J Nutr ; 121(1): 55-62, 2019 01.
Article in English | MEDLINE | ID: mdl-30360768

ABSTRACT

The polyphenolic extract (PE) from extra virgin olive oil (EVOO) has been shown to possess important anti-inflammatory and joint protective properties in murine models of rheumatoid arthritis (RA). This study was designed to evaluate the effects of PE on IL-1ß-activated human synovial fibroblasts SW982 cell line. PE from EVOO treatment inhibited IL-1ß-induced matrix metalloproteases (P<0·001), TNF-α and IL-6 production (P<0·001). Similarly, IL-1ß-induced cyclo-oxygenase-2 and microsomal PGE synthase-1 up-regulations were down-regulated by PE (P<0·001). Moreover, IL-1ß-induced mitogen-activated protein kinase (MAPK) phosphorylation and NF-κB activation were ameliorated by PE (P<0·001). These results suggest that PE from EVOO reduces the production of proinflammatory mediators in human synovial fibroblasts; particularly, these protective effects could be related to the inhibition of MAPK and NF-κB signalling pathways. Taken together, PE from EVOO probably could provide an attractive complement in management of diseases associated with over-activation of synovial fibroblasts, such as RA.


Subject(s)
Inflammation/drug therapy , Interleukin-1beta/pharmacology , Olive Oil/chemistry , Polyphenols/pharmacology , Synovial Membrane/drug effects , Anti-Inflammatory Agents , Arthritis, Rheumatoid/drug therapy , Cell Line , Cyclooxygenase 2/genetics , Down-Regulation/drug effects , Fibroblasts/drug effects , Humans , Inflammation/prevention & control , Interleukin-6/antagonists & inhibitors , Matrix Metalloproteinase Inhibitors/pharmacology , Matrix Metalloproteinases , Mitogen-Activated Protein Kinases/metabolism , NF-kappa B/metabolism , Plant Extracts/pharmacology , Polyphenols/analysis , Polyphenols/isolation & purification , Prostaglandin-E Synthases/genetics , Signal Transduction/drug effects , Synovial Membrane/cytology , Synovitis/prevention & control , Tumor Necrosis Factor-alpha/antagonists & inhibitors
6.
Br J Nutr ; 120(6): 681-692, 2018 09.
Article in English | MEDLINE | ID: mdl-30060774

ABSTRACT

Monocytes and macrophages are critical effectors and regulators of inflammation and innate immune response, which appear altered in different autoimmune diseases such as systemic lupus erythematosus (SLE). Recent studies suggested that virgin olive oil (VOO) and particularly its phenol compounds might possess preventive effects on different immune-inflammatory diseases, including SLE. Here, we evaluated the effects of VOO (and sunflower oil) on lipopolysaccharide (LPS)-activated peritoneal macrophages from a model of pristane-induced SLE in BALB/c mice, as well as those of the phenol fraction (PF) from VOO on the immune-inflammatory activity and plasticity in monocytes and monocyte-derived macrophages from healthy volunteers. The release of nitrite and inflammatory cytokines was lower in LPS-treated peritoneal macrophages from pristane-SLE mice fed the VOO diet when compared with the sunflower oil diet. PF from VOO similarly decreased the secretion of nitrite and inflammatory cytokines and expression of inducible nitric oxide, PPARγ and Toll-like receptor 4 in LPS-treated human monocytes. PF from VOO also prevented the deregulation of human monocyte subset distribution by LPS and blocked the genetic signature of M1 macrophages while favouring the phenotype of M2 macrophages upon canonical polarisation of naïve human macrophages. For the first time, our study provides several lines of in vivo and in vitro evidence that VOO and PF from VOO target and counteract inflammatory pathways in the monocyte-macrophage lineage of mice with pristane-induced SLE and of healthy subjects, which is a meaningful foundation for further development and application in preclinical and clinical use of PF from VOO in patients with SLE.


Subject(s)
Diet , Inflammation/prevention & control , Macrophages, Peritoneal/drug effects , Macrophages/drug effects , Olive Oil/chemistry , Phenols/pharmacology , Animals , Cytokines/metabolism , Female , Humans , Immunity, Innate/drug effects , Inflammation/chemically induced , Inflammation/metabolism , Lipopolysaccharides , Lupus Erythematosus, Systemic/diet therapy , Lupus Erythematosus, Systemic/immunology , Lupus Erythematosus, Systemic/metabolism , Lupus Erythematosus, Systemic/pathology , Macrophages/immunology , Macrophages/metabolism , Macrophages, Peritoneal/immunology , Macrophages, Peritoneal/metabolism , Mice, Inbred BALB C , Monocytes/drug effects , Monocytes/metabolism , Nitric Oxide/metabolism , Nitrites/metabolism , Olea/chemistry , PPAR gamma/metabolism , Phenol , Plant Extracts/chemistry , Plant Extracts/pharmacology , Plant Extracts/therapeutic use , Terpenes , Toll-Like Receptor 4/metabolism
7.
J Ethnopharmacol ; 209: 210-218, 2017 Sep 14.
Article in English | MEDLINE | ID: mdl-28698002

ABSTRACT

ETHNOPHARMACOLOGICAL RELEVANCE: Cistus albidus L. (Cistaceae) has been traditionally used to treat various inflammatory diseases, but no systematic studies on the anti-inflammatory and anti-nociceptive actions of C. albidus and its putative mechanism have been reported. We aimed to explore the anti-inflammatory and anti-nociceptive effects of this plant and to characterize its polyphenolic composition by liquid chromatography coupled to mass spectrometry (MS). MATERIALS AND METHODS: A chloroform extract derived from C. albidus leaves was obtained by solid-liquid and liquid-liquid extraction. The tail immersion test and acetic-acid-induced writhing test were used to evaluate the anti-nociceptive action, while the experimental λ-carrageenan-induced paw edema model was used to test the anti-inflammatory action. Changes in cyclooxygenase (COX)-2 and inducible nitric oxide synthase (iNOS) expression, as well as the role of mitogen-activated protein kinases (MAPKs) and the nuclear transcription factor kappa B (NF-kB) signaling pathways on lipopolysaccharide (LPS)-stimulated murine peritoneal macrophages were analyzed by western blotting. HPLC with diode array detection coupled to tandem mass spectrometry detection with electrospray ionization (HPLC-DAD-ESI-MS/MS) was performed to determine the phytochemical profile of the extract. RESULTS: Significant anti-nociceptive activity was observed both in the tail immersion (59.63% reduction at 120min) and in the acetic acid (65.94% inhibition) tests at 100mg/kg. The extract (50mg/kg) exhibited a substantial reduction in paw edema (51.6%) and significantly inhibited nitrite generation (72.62%) without affecting cell viability of LPS-stimulated murine peritoneal macrophages. These results were concomitant with a down-regulation of the pro-inflammatory enzymes COX-2 and iNOS in extract-treated macrophages and a decrease in p38 MAPK phosphorylation. HPLC-DAD-ESI-MS/MS analysis revealed that flavonols such as kaempferol and quercetin derivatives were potentially responsible for such effects. CONCLUSION: These results support the widespread use of C. albidus in popular medicine and indicate that this plant has therapeutic potential with analgesic and anti-inflammatory properties based on the presence of flavonol derivatives.


Subject(s)
Analgesics/therapeutic use , Anti-Inflammatory Agents/pharmacology , Chloroform/chemistry , Cistus/chemistry , Flavonols/pharmacology , Plant Extracts/pharmacology , Analgesics/chemistry , Animals , Anti-Inflammatory Agents/chemistry , Cell Survival/drug effects , Cells, Cultured , Chromatography, High Pressure Liquid , Edema/drug therapy , Flavonols/chemistry , Gene Expression Regulation, Enzymologic/drug effects , Lipopolysaccharides , Macrophages, Peritoneal/drug effects , Male , Mice , Mitogen-Activated Protein Kinases/genetics , Mitogen-Activated Protein Kinases/metabolism , Pain/drug therapy , Plant Extracts/chemistry , Tandem Mass Spectrometry
8.
Mol Nutr Food Res ; 59(12): 2537-46, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26382723

ABSTRACT

SCOPE: Hydroxytyrosol acetate (HTy-Ac), an extra virgin olive oil (EVOO) polyphenol, has recently been reported to exhibit antioxidant and anti-inflammatory effects on LPS-stimulated macrophagesand ulcerative colitis. This study was designed to evaluate dietary HTy-Ac supplementation effects on collagen-induced arthritis (CIA) in mice. METHODS AND RESULTS: DBA-1/J mice were fed from weaning with 0.05% HTy-Ac. After 6 weeks, arthritis was induced by type II collagen. Mice were sacrificed 42 days after first immunization. Blood was recollected and paws were histological and biochemically processed. HTy-Ac diet significantly prevent edarthritis development and decreased serum IgG1 and IgG2a, cartilage olimeric matrix protein (COMP) and metalloproteinase-3 (MMP-3) levels, as well as, pro-inflammatory cytokines levels (TNF-α, IFN-γ, IL-1ß, IL-6 and IL-17A). The activation of Janus kinase-signal transducer and activator of transcription (JAK/STAT), mitogen-activated protein kinases (MAPKs) and nuclear transcription factor-kappa B (NF-κB) pathways were drastically ameliorated whereas nuclear factor E2-related factor 2 (Nrf2) and heme oxygenase-1 (HO-1) protein expressions were significantly up-regulated in those mice fed with HTy-Ac. CONCLUSION: HTy-Ac improved the oxidative events and returned pro-inflammatory proteins expression to basal levels probably through JAK/STAT, MAPKs and NF-κB pathways. HTy-Ac supplement might provide a basis for developing a new dietary strategy for the prevention of rheumatoid arthritis.


Subject(s)
Acetates/pharmacology , Arthritis, Experimental/metabolism , Arthritis, Experimental/prevention & control , Catechols/pharmacology , Olive Oil/pharmacology , Animals , Arthritis, Experimental/chemically induced , Autoantibodies/metabolism , Cartilage Oligomeric Matrix Protein/blood , Collagen/toxicity , Cyclooxygenase 2/metabolism , Cytokines/metabolism , Dietary Supplements , Matrix Metalloproteinase 3/blood , Mice, Inbred DBA , Prostaglandin-E Synthases/metabolism , STAT3 Transcription Factor/metabolism
9.
J Nutr Biochem ; 25(12): 1275-81, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25294776

ABSTRACT

The consumption of extra virgin olive oil (EVOO) in Mediterranean countries has shown beneficial effects. A wide range of evidence indicates that phenolic compounds present in EVOO are endowed with anti-inflammatory properties. In this work, we evaluated the effects of EVOO-polyphenol extract (PE) in a model of rheumatoid arthritis, the collagen-induced arthritis model in mice. On day 0, DBA-1/J mice were immunized with bovine type II collagen. On day 21, mice received a booster injection. PE (100 and 200 mg/kg) was orally administered once a day from days 29 to 41 to arthritic mice. We have demonstrated that PE decreases joint edema, cell migration, cartilage degradation and bone erosion. PE significantly reduced the levels of proinflammatory cytokines and prostaglandin E2 in the joint as well as the expression of cyclooxygenase-2 and microsomal prostaglandin E synthase-1. Our data indicate that PE inhibits c-Jun N-terminal kinase, p38 and signal transducer and activator of transcription-3. In addition, PE decreases nuclear factor κB translocation leading to the down-regulation of the arthritic process. These results support the interest of natural diet components in the development of therapeutic products for arthritic conditions.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Arthritis, Experimental/drug therapy , Plant Oils/pharmacology , Polyphenols/pharmacology , Activating Transcription Factor 3/genetics , Activating Transcription Factor 3/metabolism , Administration, Oral , Animals , Cyclooxygenase 2/genetics , Cyclooxygenase 2/metabolism , Cytokines/blood , Dinoprostone/blood , Down-Regulation , Intramolecular Oxidoreductases/genetics , Intramolecular Oxidoreductases/metabolism , JNK Mitogen-Activated Protein Kinases/metabolism , Male , Mice , Mice, Inbred DBA , NF-kappa B/genetics , NF-kappa B/metabolism , Olive Oil , Phosphorylation , Prostaglandin-E Synthases , STAT3 Transcription Factor/genetics , STAT3 Transcription Factor/metabolism , Signal Transduction , p38 Mitogen-Activated Protein Kinases/genetics , p38 Mitogen-Activated Protein Kinases/metabolism
10.
J. physiol. biochem ; 70(1): 163-172, mar. 2014.
Article in English | IBECS | ID: ibc-121615

ABSTRACT

Inflammatory bowel disease (IBD), which includes ulcerative colitis and Crohn’s disease, is a chronic intestinal disorder resultant from a dysfunctional epithelial, innate and adaptive immune response to intestinal microorganisms. Current IBD treatment presents limitations in both efficacy and safety that stimulated for new active drugs. Retama spp. have been traditionally used in the Mediterranean region in treatment of pain and inflammation. In this study, the anti-inflammatory and protective properties of a standardised aqueous extract from Retama monosperma (RmE) was evaluated in vivo, by intra-colonic administration of trinitrobenzene sulfonic acid (TNBS) in rats as a Crohn’s disease model. The qualitative and quantitative analysis of flavonoids from RmE was performed by high-performance liquid chromatography-tandem mass spectrometry. Oral administration of RmE diminished the severity and extension of the intestinal injuries induced by TNBS. In addition, RmE increased mucus production in goblet cells in colon mucosa, decreased neutrophil infiltration and cyclooxygenase-2 (COX-2) and inducible nitric oxide synthase (iNOS) overexpression. Similarly, RmE significantly reduced p38 mitogen-activated protein kinase activation, preventing the inhibitory protein IêB degradation in colonic mucosa. RmE anti-inflammatory effects seem to be related to impairment of neutrophil function and COX-2 and iNOS down-regulation possibly through p38 MAPK and nuclear transcription factor kappa B signalling pathways. These results suggest that RmE might contribute to the development of new pharmaceutical products for inflammatory bowel disease


Subject(s)
Animals , Rats , Anti-Inflammatory Agents/pharmacokinetics , Cytisus , Colitis, Ulcerative/drug therapy , Protective Agents/pharmacokinetics , Disease Models, Animal , Plant Extracts/therapeutic use
11.
Food Chem ; 147: 117-23, 2014 Mar 15.
Article in English | MEDLINE | ID: mdl-24206694

ABSTRACT

Extra virgin olive oil (EVOO) has demonstrated great anti-inflammatory properties. Nowadays, it is clear that its minor components have a key role in these beneficial effects. However, the contribution of the unsaponifiable fraction (UF) to these healthy effects remains unknown. The present study was designed to evaluate UF in LPS stimulated peritoneal macrophages isolated from mice. NO production was analysed by the Griess method and intracellular ROS by fluorescence analysis. In addition, MAPK family activation, IKBα degradation, NFκB-p65, iNOS and COX-2 expression were determined by Western blot. UF exerted anti-inflammatory and anti-oxidant effects inhibiting LPS-induced intracellular ROS and nitrite production. Additionally, UF decreased COX-2 and iNOS protein expression. These effects were related with a down-regulation in NFκB signal signalling pathways and in MAPK phosphorylation. UF of EVOO compounds could play an important role in the anti-inflammatory effect of virgin olive oils and probably provide an attractive complement in management of inflammatory diseases.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Macrophages, Peritoneal/drug effects , Macrophages, Peritoneal/immunology , Plant Oils/pharmacology , Animals , Anti-Inflammatory Agents/chemistry , Cells, Cultured , Cyclooxygenase 2/genetics , Cyclooxygenase 2/immunology , Gene Expression/drug effects , Lipopolysaccharides/adverse effects , Lipopolysaccharides/immunology , Macrophage Activation , Male , Mice , NF-kappa B/genetics , NF-kappa B/immunology , Olive Oil , Plant Oils/chemistry
12.
J Physiol Biochem ; 70(1): 163-72, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24057513

ABSTRACT

Inflammatory bowel disease (IBD), which includes ulcerative colitis and Crohn's disease, is a chronic intestinal disorder resultant from a dysfunctional epithelial, innate and adaptive immune response to intestinal microorganisms. Current IBD treatment presents limitations in both efficacy and safety that stimulated for new active drugs. Retama spp. have been traditionally used in the Mediterranean region in treatment of pain and inflammation. In this study, the anti-inflammatory and protective properties of a standardised aqueous extract from Retama monosperma (RmE) was evaluated in vivo, by intra-colonic administration of trinitrobenzene sulfonic acid (TNBS) in rats as a Crohn's disease model. The qualitative and quantitative analysis of flavonoids from RmE was performed by high-performance liquid chromatography-tandem mass spectrometry. Oral administration of RmE diminished the severity and extension of the intestinal injuries induced by TNBS. In addition, RmE increased mucus production in goblet cells in colon mucosa, decreased neutrophil infiltration and cyclooxygenase-2 (COX-2) and inducible nitric oxide synthase (iNOS) overexpression. Similarly, RmE significantly reduced p38 mitogen-activated protein kinase activation, preventing the inhibitory protein IκB degradation in colonic mucosa. RmE anti-inflammatory effects seem to be related to impairment of neutrophil function and COX-2 and iNOS down-regulation possibly through p38 MAPK and nuclear transcription factor kappa B signalling pathways. These results suggest that RmE might contribute to the development of new pharmaceutical products for inflammatory bowel disease.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Colitis, Ulcerative/drug therapy , Fabaceae/chemistry , Plant Extracts/pharmacology , Acute Disease , Animals , Anti-Inflammatory Agents/chemistry , Anti-Inflammatory Agents/therapeutic use , Colitis, Ulcerative/chemically induced , Colitis, Ulcerative/metabolism , Colon/drug effects , Colon/enzymology , Colon/pathology , Cyclooxygenase 2/metabolism , Drug Evaluation, Preclinical , Ethanol , Gene Expression , I-kappa B Proteins/genetics , I-kappa B Proteins/metabolism , MAP Kinase Signaling System , Male , NF-kappa B/metabolism , Nitric Oxide Synthase Type II/metabolism , Plant Extracts/chemistry , Plant Extracts/therapeutic use , Rats , Rats, Wistar , Trinitrobenzenesulfonic Acid
13.
Nutr Cancer ; 65(6): 908-18, 2013.
Article in English | MEDLINE | ID: mdl-23909736

ABSTRACT

Extra virgin olive oil (EVOO) has demonstrated great oncostatic and antiinflammatory properties. Nowadays, it is clear that unsaponifiable fraction (UF) as well as other minor EVOO components have a key role in these beneficial effects. The present study was designed to evaluate UF effect in HT-29 human colon adenocarcinoma cells. Cell growth and viability assays were determined by sulphorhodamine B test at different time points (24, 48, and 72 h). The proapoptotic effect was evaluated by flow cytometric studies and different protein expression were determined by immunoblotting. UF µg/mL concentrations' range significantly reduced the growth of HT-29 cell line. Moreover UF induced intrinsic apoptotic pathway in HT-29 human colon adenocarcinoma cells through PPARγ and NFκB signaling pathways coming up to COX-2 downregulation and modulating p53 suppressor protein levels. The results suggest that UF of EVOO may exert an important role in the anticancer effect of EVOO and provide a natural resource for the prevention or treatment of human colon cancer.


Subject(s)
Apoptosis/drug effects , Cell Proliferation/drug effects , Plant Oils/pharmacology , Antineoplastic Agents/pharmacology , Cell Survival/drug effects , Cyclooxygenase 2/genetics , Cyclooxygenase 2/metabolism , Down-Regulation , HT29 Cells , Humans , NF-kappa B/genetics , NF-kappa B/metabolism , Olive Oil , PPAR gamma/genetics , PPAR gamma/metabolism , Polyphenols/administration & dosage , Rhodamines/metabolism , Signal Transduction , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism
15.
Mol Nutr Food Res ; 52(9): 1040-61, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18655004

ABSTRACT

Curcumin is a polyphenol derived from Curcuma longa. Over the last few years, a number of studies have provided evidence of its main pharmacological properties including chemosensitizing, radiosensitizing, wound healing activities, antimicrobial, antiviral, antifungical, immunomodulatory, antioxidant and anti-inflammatory. More recent data provide interesting insights into the effect of this compound on cancer chemoprevention and chemotherapy. In fact, preclinical studies have shown its ability to inhibit carcinogenesis in various types of cancer including colorectal cancer (CRC). Curcumin has the capacity of interact with multiple molecular targets affecting the multistep process of carcinogenesis. Also, curcumin is able to arrest the cell cycle, to inhibit the inflammatory response and the oxidative stress and to induce apoptosis in cancer cells. Likewise, it has been shown to possess marked antiangiogenic properties. Furthermore, curcumin potentiates the growth inhibitory effect of cyclo-oxygenase (COX)-2 inhibitors and traditional chemotherapy agents implicating another promising therapy regimen in the future treatment of CRC. However, its clinical advance has been hindered by its short biological half-life and low bioavailability after oral administration. This review is intended to provide the reader an update of the bioavailability and pharmacokinetics of curcumin and describes the recently identified molecular pathways responsible of its anticancer potential in CRC.


Subject(s)
Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/prevention & control , Curcumin/therapeutic use , Animals , Anticarcinogenic Agents/therapeutic use , Antineoplastic Agents/therapeutic use , Cell Cycle/drug effects , Clinical Trials as Topic , Disease Models, Animal , Humans , Mice , Rats , Wound Healing/drug effects
16.
Int Immunopharmacol ; 3(13-14): 1731-41, 2003 Dec.
Article in English | MEDLINE | ID: mdl-14636824

ABSTRACT

In this study, we have evaluated the efficacy of dosmalfate, a new flavonoid derivative compound, for the prevention and treatment of experimental colitis. To induce colitis, BALB/c mice received 5% dextran sulphate sodium (DSS) in their drinking water continuously for 7 days. Colitis was quantified by a clinical damage score, colon length, weight loss, stool consistency and rectal bleeding. Inflammatory response was assessed by neutrophil infiltration, determined by histology and myeloperoxidase (MPO) activity. Interleukin (IL)-1 beta, prostaglandins (PG)E(2) and (PG)D(2) concentrations in colonic tissue, histological and histochemical analysis of the lesions were also measured. Dosmalfate (400-800 mg/kg body weight, p.o.) ameliorated severe colitis reduced the degree of inflammation through reduction of neutrophil infiltration and IL-1 beta levels. (PG)E(2) and (PG)D(2) synthesis were significantly reduced in colitis control group and treatment with dosmalfate abolished the decrease in PG synthesis in colon mucosa. We conclude that dosmalfate is protective in acute DSS-induced colitis. The beneficial effects seem to be related to a decrease of neutrophil infiltration, absence of up-regulation of IL-1 beta and increase of PG production in colon mucosa.


Subject(s)
Colitis, Ulcerative/chemically induced , Dextran Sulfate/adverse effects , Dextran Sulfate/antagonists & inhibitors , Diosmin/analogs & derivatives , Diosmin/therapeutic use , Administration, Oral , Animals , Colitis, Ulcerative/drug therapy , Colon, Descending/drug effects , Colon, Descending/injuries , Colon, Descending/ultrastructure , Colon, Transverse/drug effects , Colon, Transverse/injuries , Colon, Transverse/ultrastructure , Dextran Sulfate/administration & dosage , Dinoprostone/biosynthesis , Diosmin/administration & dosage , Diosmin/pharmacokinetics , Disease Models, Animal , Drinking , Flavonoids/administration & dosage , Flavonoids/chemistry , Flavonoids/pharmacokinetics , Interleukin-1/biosynthesis , Intestinal Mucosa/drug effects , Intestinal Mucosa/enzymology , Intestinal Mucosa/injuries , Male , Mice , Mice, Inbred BALB C , Neutrophil Infiltration/drug effects , Peroxidase/metabolism , Prostaglandin D2/biosynthesis , Time Factors , Water
SELECTION OF CITATIONS
SEARCH DETAIL