Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
Am J Physiol Renal Physiol ; 316(6): F1211-F1217, 2019 06 01.
Article in English | MEDLINE | ID: mdl-30864840

ABSTRACT

The gut microbiome is composed of a diverse population of bacteria that have beneficial and adverse effects on human health. The microbiome has recently gained attention and is increasingly noted to play a significant role in health and a number of disease states. Increasing urea concentration during chronic kidney disease (CKD) leads to alterations in the intestinal flora that can increase production of gut-derived toxins and alter the intestinal epithelial barrier. These changes can lead to an acceleration of the process of kidney injury. A number of strategies have been proposed to interrupt this pathway of injury in CKD. The purpose of this review is to summarize the role of the gut microbiome in CKD, tools used to study this microbial population, and attempts to alter its composition for therapeutic purposes.


Subject(s)
Bacteria/metabolism , Gastrointestinal Microbiome , Intestines/microbiology , Kidney/metabolism , Renal Insufficiency, Chronic/microbiology , Urea/metabolism , Uremia/microbiology , Animals , Dietary Supplements , Host-Pathogen Interactions , Humans , Intestinal Mucosa/metabolism , Intestinal Mucosa/microbiology , Intestines/physiopathology , Kidney/physiopathology , Permeability , Renal Insufficiency, Chronic/metabolism , Renal Insufficiency, Chronic/physiopathology , Renal Insufficiency, Chronic/therapy , Uremia/metabolism , Uremia/physiopathology , Uremia/therapy
2.
Am J Cardiol ; 120(9): 1501-1507, 2017 Nov 01.
Article in English | MEDLINE | ID: mdl-28847594

ABSTRACT

Cardiac myosin binding protein-C (cMyBP-C) is a heart muscle-specific thick filament protein. Elevated level of serum cMyBP-C is an indicator of early myocardial infarction (MI), but its value as a predictor of future cardiovascular disease is unknown. Based on the presence of significant amount of cMyBP-C in the serum of previous study subjects independent of MI, we hypothesized that circulating cMyBP-C is a sensitive indicator of ongoing cardiovascular stress and disease. To test this hypothesis, 75 men and 83 women of similar ages were recruited for a prospective study. They underwent exercise stress echocardiography to provide pre- and poststress blood samples for subsequent determination of serum cMyBP-C levels. The subjects were followed for 1 to 1.5 years. Exercise stress increased serum cMyBP-C in all subjects. Twenty-seven primary events (such as death, MI, revascularization, invasive cardiovascular procedure, or cardiovascular-related hospitalization) and 7 critical events (CE; such as death, MI, stroke, or pulmonary embolism) occurred. After adjusting for sex and cardiovascular risk factors with multivariate Cox regression, a 96% sensitive prestress cMyBP-C threshold carried a hazard ratio of 8.1 with p = 0.041 for primary events. Most subjects (6 of 7) who had CE showed normal ejection fraction on echocardiography. Prestress cMyBP-C demonstrated area under receiver operating curve of 0.91 and multivariate Cox regression hazard ratio of 13.8 (p = 0.000472) for CE. Thus, basal cMyBP-C levels reflected susceptibility for a variety of cardiovascular diseases. Together with its high sensitivity, cMyBP-C holds potential as a screening biomarker for the existence of severe cardiovascular diseases.


Subject(s)
Cardiovascular Diseases/blood , Cardiovascular Diseases/etiology , Carrier Proteins/blood , Aged , Cardiovascular Diseases/diagnosis , Exercise Test , Female , Humans , Male , Middle Aged , Predictive Value of Tests , Prospective Studies , ROC Curve , Stroke Volume
3.
Aging (Albany NY) ; 8(11): 3065-3090, 2016 11 21.
Article in English | MEDLINE | ID: mdl-27875806

ABSTRACT

This study aimed to establish mechanistic links between the aging-associated changes in the functional status of mast cells and the altered responses of mesenteric tissue and mesenteric lymphatic vessels (MLVs) to acute inflammation. We used an in vivo model of acute peritoneal inflammation induced by lipopolysaccharide treatment of adult (9-month) and aged (24-month) F-344 rats. We analyzed contractility of isolated MLVs, mast cell activation, activation of nuclear factor-κB (NF-κB) without and with stabilization of mast cells by cromolyn or blockade of all types of histamine receptors and production of 27 major pro-inflammatory cytokines in adult and aged perilymphatic mesenteric tissues and blood. We found that the reactivity of aged contracting lymphatic vessels to LPS-induced acute inflammation was abolished and that activated mast cells trigger NF-κB signaling in the mesentery through release of histamine. The aging-associated basal activation of mesenteric mast cells limits acute inflammatory NF-κB activation in aged mesentery. We conclude that proper functioning of the mast cell/histamine/NF-κB axis is necessary for reactions of the lymphatic vessels to acute inflammatory stimuli as well as for interaction and trafficking of immune cells near and within the collecting lymphatics.


Subject(s)
Cytokines/metabolism , Histamine/metabolism , Inflammation/metabolism , Mast Cells/metabolism , NF-kappa B/metabolism , Peritoneal Diseases/metabolism , Animals , Cromolyn Sodium/pharmacology , Inflammation/chemically induced , Inflammation Mediators/metabolism , Lipopolysaccharides , Lymphatic Vessels/metabolism , Male , Mesentery/metabolism , Peritoneal Diseases/chemically induced , Rats , Rats, Inbred F344
4.
J Neurochem ; 135(3): 565-76, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26179031

ABSTRACT

Hepatic encephalopathy (HE) is a serious neurological complication of acute and chronic liver failure. Expression of the neurosteroid/bile acid receptor Takeda G protein-coupled receptor 5 (TGR5) has been demonstrated in the brain and is thought to be neuroprotective. However, it is unknown how TGR5 signaling can influence the progression and associated neuroinflammation of HE. HE was induced in C57Bl/6 mice via intraperitoneal injection of azoxymethane (AOM) and tissue was collected throughout disease progression. TGR5 expression was elevated in the frontal cortex following AOM injection in mice. The cellular localization of TGR5 was found in both neurons and microglia in the cortex of C57Bl/6 mice. Central infusion of the TGR5 agonist, betulinic acid, prior to AOM injection delayed neurological decline, increased cortical cyclic adenosine monophosphate concentrations, reduced microglia activation and proliferation, and reduced proinflammatory cytokine production. Betulinic acid treatment in vitro reduced the neuronal expression of chemokine ligand 2, a chemokine previously demonstrated to contribute to HE pathogenesis. Lastly, treatment of the microglia cell line EOC-20 with conditioned media from betulinic acid-treated primary neurons decreased phagocytic activity and cytokine production. Together, these data identify that activation of TGR5, which is up-regulated during HE, alleviates neuroinflammation and improves outcomes of AOM-treated mice through neuron and microglia paracrine signaling.


Subject(s)
Hepatic Encephalopathy/metabolism , Hepatic Encephalopathy/prevention & control , Receptors, G-Protein-Coupled/metabolism , Signal Transduction/physiology , Animals , Cells, Cultured , Inflammation/metabolism , Inflammation/prevention & control , Male , Mice , Mice, Inbred C57BL , Pentacyclic Triterpenes , Receptors, G-Protein-Coupled/agonists , Signal Transduction/drug effects , Triterpenes/administration & dosage , Betulinic Acid
5.
Circ Heart Fail ; 8(3): 582-94, 2015 May.
Article in English | MEDLINE | ID: mdl-25740839

ABSTRACT

BACKGROUND: Heart failure (HF) with preserved ejection fraction (HFpEF) accounts for ≈50% of all cases of HF and currently has no effective treatment. Diastolic dysfunction underlies HFpEF; therefore, elucidation of the mechanisms that mediate relaxation can provide new potential targets for treatment. Cardiac myosin-binding protein-C (cMyBP-C) is a thick filament protein that modulates cross-bridge cycling rates via alterations in its phosphorylation status. Thus, we hypothesize that phosphorylated cMyBP-C accelerates the rate of cross-bridge detachment, thereby enhancing relaxation to mediate diastolic function. METHODS AND RESULTS: We compared mouse models expressing phosphorylation-deficient cMyBP-C(S273A/S282A/S302A)-cMyBP-C(t3SA), phosphomimetic cMyBP-C(S273D/S282D/S302D)-cMyBP-C(t3SD), and wild-type-control cMyBP-C(tWT) to elucidate the functional effects of cMyBP-C phosphorylation. Decreased voluntary running distances, increased lung/body weight ratios, and increased brain natriuretic peptide levels in cMyBP-C(t3SA) mice demonstrate that phosphorylation deficiency is associated with signs of HF. Echocardiography (ejection fraction and myocardial relaxation velocity) and pressure/volume measurements (-dP/dtmin, pressure decay time constant τ-Glantz, and passive filling stiffness) show that cMyBP-C phosphorylation enhances myocardial relaxation in cMyBP-C(t3SD) mice, whereas deficient cMyBP-C phosphorylation causes diastolic dysfunction with HFpEF in cMyBP-C(t3SA) mice. Simultaneous force and [Ca(2+)]i measurements on intact papillary muscles show that enhancement of relaxation in cMyBP-C(t3SD) mice and impairment of relaxation in cMyBP-C(t3SA) mice are not because of altered [Ca(2+)]i handling, implicating that altered cross-bridge detachment rates mediate these changes in relaxation rates. CONCLUSIONS: cMyBP-C phosphorylation enhances relaxation, whereas deficient phosphorylation causes diastolic dysfunction and phenotypes resembling HFpEF. Thus, cMyBP-C is a potential target for treatment of HFpEF.


Subject(s)
Carrier Proteins/metabolism , Heart Failure/metabolism , Ventricular Dysfunction, Left/metabolism , Ventricular Function, Left , Animals , Blood Pressure , Carrier Proteins/genetics , Cyclic AMP-Dependent Protein Kinases/metabolism , Diastole , Genotype , Heart Failure/genetics , Heart Failure/physiopathology , Kinetics , Mice, Transgenic , Mutation , Phenotype , Phosphorylation , Protein Processing, Post-Translational , Ventricular Dysfunction, Left/genetics , Ventricular Dysfunction, Left/physiopathology
6.
Transl Gastrointest Cancer ; 2(1)2013 Jan 01.
Article in English | MEDLINE | ID: mdl-24312749

ABSTRACT

Melatonin exerts a multitude of physiological functions including the regulation of the sleep cycle and circadian rhythm. Although the synthesis of melatonin in the pineal gland is regulated by changes in the light/dark cycle, the release of melatonin in the gastrointestinal tract is related to food consumption. Melatonin regulates antioxidative processes and it improves T-helper cell response by stimulating the production of specific cytokines. Melatonin is directly involved in preventing tumor initiation, promotion, and progression in a variety of cancers of the gastrointestinal tract including colorectal cancer, cholangiocarcinoma, hepatocarcinoma, and pancreatic carcinoma. This paper is a review of the literature regarding melatonin in the gastrointestinal tract and as a potential therapy for gastrointestinal cancers.

7.
Gastroenterology Res ; 6(4): 124-133, 2013 Aug.
Article in English | MEDLINE | ID: mdl-27785242

ABSTRACT

BACKGROUND: To elucidate the effects of a solution containing interleukin-10 and anti-IL-1 antibody in modulating experimental intestinal inflammation. METHODS: Colitis was induced in BALB/c mice by oral administration of dextran sodium sulphate; mice were then treated with interleukin-10 plus anti-IL-1 antibody at low dosage. Transepithelial electrical resistance of isolated mouse colon and colon lengths were evaluated. Cytokines concentrations in organocultures supernatants and plasma samples were evaluated by Enzyme-Linked Immuno Sorbent Assay. Tight junction proteins were evaluated by immunofluorescence, respectively. RESULTS: Oral administration of tested products restores intestinal barrier function during experimental intestinal inflammation in association with reduced levels of proinflammatory cytokines, increased interleukin-10 plasma concentrations and a tight junction architecture restoration. CONCLUSION: Obtained results may contribute to modelling an interesting strategy for the treatment of patients with inflammatory bowel diseases.

8.
Hepatology ; 57(3): 1130-41, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23080076

ABSTRACT

UNLABELLED: Secretin stimulates ductal secretion by interacting with secretin receptor (SR) activating cyclic adenosine 3',5'-monophosphate/cystic fibrosis transmembrane conductance regulator/chloride bicarbonate anion exchanger 2 (cAMP⇒CFTR⇒Cl(-) /HCO 3- AE2) signaling that is elevated by biliary hyperplasia. Cholangiocytes secrete several neuroendocrine factors regulating biliary functions by autocrine mechanisms. Melatonin inhibits biliary growth and secretin-stimulated choleresis in cholestatic bile-duct-ligated (BDL) rats by interaction with melatonin type 1 (MT1) receptor through down-regulation of cAMP-dependent signaling. No data exist regarding the role of melatonin synthesized locally by cholangiocytes in the autocrine regulation of biliary growth and function. In this study, we evaluated the (1) expression of arylalkylamine N-acetyltransferase (AANAT; the rate-limiting enzyme for melatonin synthesis from serotonin) in cholangiocytes and (2) effect of local modulation of biliary AANAT expression on the autocrine proliferative/secretory responses of cholangiocytes. In the liver, cholangiocytes (and, to a lesser extent, BDL hepatocytes) expressed AANAT. AANAT expression and melatonin secretion (1) increased in BDL, compared to normal rats and BDL rats treated with melatonin, and (2) decreased in normal and BDL rats treated with AANAT Vivo-Morpholino, compared to controls. The decrease in AANAT expression, and subsequent lower melatonin secretion by cholangiocytes, was associated with increased biliary proliferation and increased SR, CFTR, and Cl(-) /HCO 3- AE2 expression. Overexpression of AANAT in cholangiocyte cell lines decreased the basal proliferative rate and expression of SR, CFTR, and Cl(-) /HCO 3- AE2 and ablated secretin-stimulated biliary secretion in these cells. CONCLUSION: Local modulation of melatonin synthesis may be important for management of the balance between biliary proliferation/damage that is typical of cholangiopathies. (HEPATOLOGY 2013).


Subject(s)
Arylalkylamine N-Acetyltransferase/metabolism , Autocrine Communication/physiology , Bile Ducts, Intrahepatic/cytology , Bile Ducts, Intrahepatic/enzymology , Cholestasis/metabolism , Cholestasis/pathology , Animals , Anion Transport Proteins/genetics , Anion Transport Proteins/metabolism , Antioxidants/metabolism , Antioxidants/pharmacology , Antiporters/genetics , Antiporters/metabolism , Apoptosis/drug effects , Apoptosis/physiology , Arylalkylamine N-Acetyltransferase/genetics , Autocrine Communication/drug effects , Bile Ducts, Intrahepatic/drug effects , Cell Line, Transformed , Cell Proliferation , Gene Knockdown Techniques , Male , Melatonin/blood , Melatonin/pharmacology , Mice , Proliferating Cell Nuclear Antigen/genetics , Proliferating Cell Nuclear Antigen/metabolism , Rats , Rats, Inbred F344 , Receptors, Serotonin/genetics , Receptors, Serotonin/metabolism , SLC4A Proteins
9.
Dig Dis Sci ; 57(5): 1227-37, 2012 May.
Article in English | MEDLINE | ID: mdl-22278339

ABSTRACT

BACKGROUND: Vasostatin-1 (VS-1), the N-terminal fragment of chromogranin A (CgA), decreases the permeability of endothelial cells in vitro and in vivo. AIMS: Here, we investigated whether a similar effect could be observed also on intestinal epithelial cells (IECs) in vitro and whether VS-1 could have favorable effects on animal models of acute or chronic colitis, which are characterized by increased permeability of the intestinal epithelium. METHODS: In vitro, VS-1 was tested on IEC monolayers showing increased permeability, on mechanically injured IEC monolayers, and on the production of the chemokine IL-8/KC by lipopolysaccharide (LPS)-stimulated IECs. In vivo, VS-1 was tested in animal models of dextran sodium salt (DSS)-induced acute or chronic colitis. RESULTS: In vitro, VS-1 inhibited increased permeability of IECs induced by interferon-γ and tumor necrosis factor-α. Moreover, VS-1 promoted healing of mechanically injured IEC monolayers, most likely through stimulation of cell migration, rather than cell proliferation. Eventually, VS-1 inhibited LPS-induced production of IL-8. In vivo, VS-1 exerted protective effects in animal models of acute or chronic colitis upon oral, but not systemic administration. CONCLUSIONS: VS-1 is therapeutically active in animal models of acute or chronic, DSS-induced colitis. The mechanisms underlying this effect are likely to be multiple, and may include inhibition of enhanced intestinal permeability, repair of injured intestinal mucosae, and inhibition of the production of IL-8/KC and possibly other inflammatory cytokines.


Subject(s)
Cell Membrane Permeability/drug effects , Chromogranin A , Colitis , Colon/metabolism , Epithelial Cells/metabolism , Peptide Fragments , Administration, Oral , Animals , Cell Line, Tumor , Cell Movement/drug effects , Chromogranin A/administration & dosage , Chromogranin A/pharmacokinetics , Chronic Disease , Colitis/metabolism , Colitis/pathology , Colon/drug effects , Colon/pathology , Disease Models, Animal , Epithelial Cells/drug effects , Epithelial Cells/pathology , Female , Humans , Interferon-gamma/metabolism , Interleukin-8/antagonists & inhibitors , Interleukin-8/metabolism , Lipopolysaccharides/metabolism , Mice , Mice, Inbred C57BL , Peptide Fragments/administration & dosage , Peptide Fragments/pharmacokinetics , Protective Agents/administration & dosage , Protective Agents/pharmacokinetics , Treatment Outcome , Tumor Necrosis Factor-alpha/metabolism
10.
Genes Chromosomes Cancer ; 51(2): 111-26, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22045652

ABSTRACT

To highlight possible similarities and differences in receptor tyrosine kinase (RTK) and downstream signalling activation profiles between clear-cell sarcomas (CCS) and metastatic melanomas (MM), frozen, and paired-matched fixed samples of six CCS with EWSR1 rearrangement (EWSR1+), five CCS without EWSR1 rearrangement (EWSR1-), and seven MM were investigated by means of biochemical, immunohistochemical, FISH, molecular analyses, and immunofluorescence confocal microscopy. Fixed samples of a further 10 CCS and 14 MM were investigated by means of sequencing for BRAF, NRAS, and KRAS mutations and FISH analyses for the gain of chromosomes 22 and 8. RTK analysis of all CCS/MM samples showed activation of short-form (sf) recepteur d'origine nantais (RON) RTK and of PDGFRB, MET, and HER3. Analysis of downstream signaling revealed consistent phosphorylation patterns of PI3K/AKT, RSK, and the mTOR targets S6 and 4EBP1. Analysis of frozen and fixed material from 21 CCS and 21 MM showed the presence of the V600E BRAF mutation in 2/12 EWSR1+ and 3/9 EWSR1- CCS and 9/21 MM and demonstrated a significant (P < 0.001) correlation between the gain of chromosomes 22 and 8 and EWSR1- CCS. Our results show that BRAF mutation can also be present in CCS and support the proposed aberration of chromosomes 22 and 8 as a possibly useful nonrandom hallmark of EWSR1- CCS. Besides, they broaden the spectrum of the similarities of RTK pathway activation between CCS and MM, thus suggesting that new drugs found to be active in melanoma and RON inhibitors could have a role in CCS treatment. © 2011 Wiley Periodicals, Inc.


Subject(s)
Biomarkers, Tumor/metabolism , Melanoma/secondary , Receptor Protein-Tyrosine Kinases/metabolism , Sarcoma, Clear Cell/metabolism , Adult , Aged , Aged, 80 and over , Base Sequence , Biomarkers, Tumor/genetics , Calmodulin-Binding Proteins/genetics , Calmodulin-Binding Proteins/metabolism , Chromosome Duplication , Chromosomes, Human, Pair 22 , Chromosomes, Human, Pair 8 , Female , Gene Expression , Humans , Lymphatic Metastasis , Male , Melanoma/genetics , Melanoma/metabolism , Middle Aged , Phosphorylation , RNA-Binding Protein EWS , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , Receptor Protein-Tyrosine Kinases/genetics , Sarcoma, Clear Cell/genetics , Sarcoma, Clear Cell/pathology , Sequence Analysis, DNA , Trisomy , Young Adult
11.
Immunol Cell Biol ; 89(5): 630-9, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21102537

ABSTRACT

The innate immune system is present throughout the female reproductive tract and functions in synchrony with the adaptive immune system to provide protection in a way that enhances the chances for fetal survival, while protecting against potential pathogens. Recent data show that activation of Toll-like receptor (TLR)2 and 4 by low-molecular weight hyaluronic acid (LMW-HA) in the epidermis induces secretion of the antimicrobial peptide ß-defensin 2. In the present work, we show that LMW-HA induces vaginal epithelial cells to release different antimicrobial peptides, via activation of TLR2 and TLR4. Further, we found that LMW-HA favors repair of vaginal epithelial injury, involving TLR2 and TLR4, and independently from its classical receptor CD44. This wound-healing activity of LMW-HA is dependent from an Akt/phosphatidylinositol 3 kinase pathway. Therefore, these findings suggest that the vaginal epithelium is more than a simple physical barrier to protect against invading pathogens: on the contrary, this surface acts as efficient player of innate host defense, which may modulate its antimicrobial properties and injury restitution activity, following LMW-HA stimulation; this activity may furnish an additional protective activity to this body compartment, highly and constantly exposed to microbiota, ameliorating the self-defense of the vaginal epithelium in both basal and pathological conditions.


Subject(s)
Adjuvants, Immunologic/pharmacology , Epithelium/drug effects , Hyaluronic Acid/pharmacology , Toll-Like Receptor 2/immunology , Toll-Like Receptor 4/immunology , Vagina/drug effects , Vagina/immunology , Cell Line, Transformed , Epithelial Cells/drug effects , Epithelial Cells/immunology , Epithelial Cells/metabolism , Epithelium/immunology , Epithelium/metabolism , Female , Gene Expression Regulation, Archaeal/drug effects , Humans , Hyaluronic Acid/metabolism , Immunity, Innate , Immunologic Factors/immunology , Immunologic Factors/metabolism , Inflammation Mediators/metabolism , Ligands , Phosphatidylinositol 3-Kinase/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction/drug effects , Wound Healing/drug effects , Wound Healing/genetics , beta-Defensins/genetics , beta-Defensins/metabolism
12.
Am J Pathol ; 175(3): 1066-76, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19700751

ABSTRACT

We have recently observed that oral administration of D-glucose saves animals from lipopolysaccharide (LPS)-induced death. This effect is the likely consequence of glucose-induced activation of the sodium-dependent glucose transporter-1. In this study, we investigated possible hepatoprotective effects of glucose-induced, sodium-dependent, glucose transporter-1 activation. We show that oral administration of D-glucose, but not of either D-fructose or sucrose, prevents LPS-induced liver injury, as well as liver injury and death induced by an overdose of acetaminophen. In both of these models, physiological liver morphology is maintained and organ protection is confirmed by unchanged levels of the circulating markers of hepatotoxicity, such as alanine transaminase or lactate dehydrogenase. In addition, D-glucose was found to protect the liver from alpha-amanitin-induced liver injury. In this case, in contrast to the previously described models, a second signal had to be present in addition to glucose to achieve protective efficacy. Toll-like receptor 4 stimulation that was induced by low doses of LPS was identified as such a second signal. Eventually, the protective effect of orally administered glucose on liver injury induced by LPS, overdose of acetaminophen, or alpha-amanitin was shown to be mediated by the anti-inflammatory cytokine interleukin-10. These findings, showing glucose-induced protective effects in several animal models of liver injury, might be relevant in view of possible therapeutic interventions against different forms of acute hepatic injury.


Subject(s)
Chemical and Drug Induced Liver Injury , Glucose/administration & dosage , Liver Diseases/prevention & control , Acetaminophen/adverse effects , Administration, Oral , Alpha-Amanitin/toxicity , Animals , Fructose/administration & dosage , Galactosamine/adverse effects , Interleukin-10 , Intestinal Mucosa/metabolism , Lipopolysaccharides/toxicity , Liver Diseases/metabolism , Mice , Mice, Inbred C57BL , Signal Transduction , Sodium-Glucose Transporter 1/metabolism , Sucrose/administration & dosage , Toll-Like Receptor 4/metabolism
13.
Pulm Pharmacol Ther ; 22(6): 497-510, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19464382

ABSTRACT

Many inflammatory diseases are characterized by an imbalance among lymphocyte populations, in particular Th1, Th2 and the recently described Th17 cells. The Th1/Th2 imbalance is linked to many factors, but certainly the role of cytokines is essential. In Th2 diseases IL-4 expression is predominant, while Th1 pathologies are characterized by high expression of IFN-gamma and IL-12. Though today the therapeutical proposal for many inflammatory diseases aims to re-establish normal levels of Th1/Th2 cytokines, the pharmacological use of cytokines, which are very active molecules, is limited by the possible collateral effects. Therefore, our study aims to determine, in a murine model of allergic asthma, the possible therapeutic activity of low dose cytokines solutions, mechanically activated. We found that oral administration of low doses IL-12 plus IFN-gamma is able to solve the bronchial hyperresponsiveness condition of mice, establishing normal cytokine levels. The anti-asthma activity was confirmed by histological analysis of lungs and broncho-alveolar lavage fluid cell count. Serum ovalbumin-specific IgE was also significantly inhibited by treatment with low dose activated cytokines solution. These findings may suggest a novel approach to diseases which involve a Th1/Th2 imbalance.


Subject(s)
Asthma/drug therapy , Cytokines/therapeutic use , Interferon-gamma/therapeutic use , Interleukin-12/therapeutic use , Respiratory Hypersensitivity/drug therapy , Animals , Asthma/pathology , Bronchial Hyperreactivity/drug therapy , Bronchial Hyperreactivity/physiopathology , Bronchoalveolar Lavage Fluid/cytology , CD11c Antigen/immunology , Cytokines/administration & dosage , Dendritic Cells/drug effects , Dose-Response Relationship, Drug , Enzyme-Linked Immunosorbent Assay , Interferon-gamma/administration & dosage , Interleukin-12/administration & dosage , Lung/immunology , Lung/pathology , Male , Mice , Recombinant Proteins/therapeutic use , Respiratory Hypersensitivity/pathology , Solutions , Spleen/cytology , Spleen/drug effects , Th1 Cells/drug effects , Th1 Cells/immunology , Th2 Cells/drug effects , Th2 Cells/immunology
14.
J Immunol ; 181(5): 3126-36, 2008 Sep 01.
Article in English | MEDLINE | ID: mdl-18713983

ABSTRACT

In this study, we demonstrate the protective effect of the activation of sodium-dependent glucose transporter-1 (SGLT-1) on damages induced by TLR ligands, in intestinal epithelial cells and in a murine model of septic shock. In intestinal epithelial cell lines, glucose inhibited the IL-8/keratinocyte-derived chemokine production and the activation of the TLR-related transcription factor NF-kappaB stimulated by LPS or CpG-oligodeoxynucleotide. Oral ingestion of glucose was found to protect 100% of mice from lethal endotoxic shock induced by i.p. LPS administration; protection was only observed when glucose was administered orally, not by i.p. route, suggesting the important role of intestinal epithelial cells in this protection. In addition, we observed that the in vivo protection depends on an increase of anti-inflammatory cytokine IL-10. The cornerstone of the observed immunomodulatory and life-saving effects resides in activation of SGLT-1; in fact, the glucose analog 3-O-methyl-d-gluco-pyranose, which induces the transporter activity, but is not metabolized, exerted the same inhibitory effects as glucose both in vitro and in vivo. Thus, we propose that activated SGLT-1, apart from its classical metabolic function, may be a promising target for inhibition of bacteria-induced inflammatory processes and life-saving treatments, assuming a novel role as an immunological player.


Subject(s)
Intestinal Mucosa/immunology , Sodium-Glucose Transporter 1/immunology , Animals , Cell Line , Chemokines/antagonists & inhibitors , Glucose/administration & dosage , Glucose/pharmacology , Humans , Inflammation , Interleukin-10/immunology , Lipopolysaccharides/pharmacology , Mice , NF-kappa B/antagonists & inhibitors , Oligodeoxyribonucleotides/pharmacology , Shock, Septic/chemically induced , Shock, Septic/drug therapy , Shock, Septic/pathology
15.
Int Immunol ; 20(5): 709-18, 2008 May.
Article in English | MEDLINE | ID: mdl-18397908

ABSTRACT

Toll-like receptors (TLRs) 4, 5, 7 and 9 belong to a family of proteins that recognize mainly conserved microbial motifs. Though each TLR has a highly specific ability to recognize a particular microbial pattern, recent papers suggest that some ligands are able to affect the expression of different TLRs. In this paper, we have investigated TLR4, 5, 7 and 9 expression, both at mRNA and protein level, following treatment of different intestinal epithelial cell lines with LPS, flagellin, loxiribine, CpG-oligodeoxynucleotide and peptidoglycan, to assess if the different TLR ligands may modulate the expression of the respective TLR and of the unrelated ones. Our results show that a cross-talk exists between TLRs and various ligands, indicating a cross-regulation among these pattern recognition receptors. In particular, TLR4 was generally down-regulated by treatment with ligands other than LPS, while flagellin and unrelated microbial-associated molecular patterns exerted a general stimulatory activity as regards TLR5 expression. Concerning TLR7 and 9, we have observed a more variable behaviour of the various cell lines with the different ligands. Together, our results demonstrate that the expression of TLRs in intestinal cells is highly dynamic and tightly regulated in response to encountered microbial stimuli.


Subject(s)
Antigens, Bacterial/immunology , Interleukin-8/immunology , Receptor Cross-Talk , Toll-Like Receptors/metabolism , Animals , Antigens, Bacterial/metabolism , Cell Line, Tumor , Epithelial Cells/immunology , Epithelial Cells/metabolism , HT29 Cells , Humans , Interleukin-8/biosynthesis , Ligands , Macrophages/immunology , Macrophages/metabolism , Mice , Oligodeoxyribonucleotides/immunology , Oligodeoxyribonucleotides/metabolism , Peptidoglycan/immunology , Peptidoglycan/metabolism , Toll-Like Receptors/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...