Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Language
Publication year range
1.
Am J Physiol Gastrointest Liver Physiol ; 308(7): G591-604, 2015 Apr 01.
Article in English | MEDLINE | ID: mdl-25591865

ABSTRACT

In inflammatory bowel disease (IBD), obesity is associated with worsening of the course of disease. Here, we examined the role of obesity in the development of colitis and studied mesenteric fat-epithelial cell interactions in patients with IBD. We combined the diet-induce obesity with the trinitrobenzene sulfonic acid (TNBS) colitis mouse model to create groups with obesity, colitis, and their combination. Changes in the mesenteric fat and intestine were assessed by histology, myeloperoxidase assay, and cytokine mRNA expression by real-time PCR. Medium from human mesenteric fat and cultured preadipocytes was obtained from obese patients and those with IBD. Histological analysis showed inflammatory cell infiltrate and increased histological damage in the intestine and mesenteric fat of obese mice with colitis compared with all other groups. Obesity also increased the expression of proinflammatory cytokines including IL-1ß, TNF-α, monocyte chemoattractant protein 1, and keratinocyte-derived chemokine, while it decreased the TNBS-induced increases in IL-2 and IFN-γ in mesenteric adipose and intestinal tissues. Human mesenteric fat isolated from obese patients and those with and IBD demonstrated differential release of adipokines and growth factors compared with controls. Fat-conditioned media reduced adiponectin receptor 1 (AdipoR1) expression in human NCM460 colonic epithelial cells. AdipoR1 intracolonic silencing in mice exacerbated TNBS-induced colitis. In conclusion, obesity worsens the outcome of experimental colitis, and obesity- and IBD-associated changes in adipose tissue promote differential mediator release in mesenteric fat that modulates colonocyte responses and may affect the course of colitis. Our results also suggest an important role for AdipoR1 for the fat-intestinal axis in the regulation of inflammation during colitis.


Subject(s)
Abdominal Fat/metabolism , Adipocytes, White/metabolism , Colitis/etiology , Cytokines/metabolism , Inflammation Mediators/metabolism , Obesity/complications , Receptors, Adiponectin/metabolism , Abdominal Fat/immunology , Adipocytes, White/immunology , Adipokines/genetics , Adipokines/metabolism , Animals , Case-Control Studies , Cells, Cultured , Colitis/chemically induced , Colitis/genetics , Colitis/immunology , Colitis/metabolism , Colitis/pathology , Colon/immunology , Colon/metabolism , Colon/pathology , Culture Media, Conditioned/metabolism , Cytokines/genetics , Cytokines/immunology , Disease Models, Animal , Female , Gene Expression Regulation , Gene Knockdown Techniques , Humans , Inflammation Mediators/immunology , Male , Mice, Inbred C57BL , Obesity/genetics , Obesity/immunology , Obesity/metabolism , Obesity/pathology , RNA Interference , RNA, Messenger/metabolism , Severity of Illness Index , Signal Transduction , Time Factors , Trinitrobenzenesulfonic Acid
2.
Am J Gastroenterol ; 107(2): 262-72, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22158028

ABSTRACT

OBJECTIVES: Low-grade colonic mucosal inflammation has been postulated to have an important role in the pathophysiology of irritable bowel syndrome (IBS). The objectives of this study were (i) to identify serum and tissue-based immunological and neuroendocrine markers associated with mucosal inflammation in male (M) and female (F) patients with non-post-infectious IBS (non-PI-IBS) compared with healthy controls and (ii) to assess possible correlations of such markers with IBS symptoms. METHODS: Sigmoid mucosal biopsies were obtained from 45 Rome II positive IBS patients without a history of PI-IBS (26 F, 35.5% IBS-C, 33.3% IBS-D, 31.1% IBS-A/M) and 41 healthy controls (22 F) in order to measure immunological markers (serum cytokine levels, colonic mucosal mRNA levels of cytokines, mucosal immune cell counts) and neuroendocrine markers associated with mucosal inflammation (corticotropin releasing factor- and neurokinin (NK)-related ligands and receptors, enterochromaffin cells). Symptoms were measured using validated questionnaires. RESULTS: Of all the serum and mucosal cytokines measured, only interleukin-10 (IL-10) mRNA expression showed a group difference, with female, but not male, patients showing lower levels compared with female controls (18.0±2.9 vs. 29.5±4.0, P=0.006). Mucosal mRNA expression of NK-1 receptor was significantly lower (1.15±0.19 vs. 2.66±0.56, P=0.008) in female, but not male, patients compared with healthy controls. No other significant differences were observed. CONCLUSIONS: Immune cell counts and levels of cytokines and neuropeptides that are associated with inflammation were not significantly elevated in the colonic mucosa of non-PI-IBS patients, and did not correlate with symptoms. Thus, these findings do not support that colonic mucosal inflammation consistently has a primary role in these patients. However, the finding of decreased IL-10 mRNA expression may be a possible biomarker of IBS and warrants further investigation.


Subject(s)
Biomarkers/metabolism , Colon/metabolism , Intestinal Mucosa/metabolism , Irritable Bowel Syndrome/metabolism , Adolescent , Adult , Colon/immunology , Colon/pathology , Cytokines/metabolism , Female , Humans , Interleukin-10/metabolism , Intestinal Mucosa/immunology , Intestinal Mucosa/pathology , Irritable Bowel Syndrome/immunology , Irritable Bowel Syndrome/pathology , Male , Middle Aged , Receptors, Neurokinin-1/metabolism , Sex Factors , Surveys and Questionnaires
3.
Endocrinology ; 152(6): 2197-205, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21467195

ABSTRACT

Peripheral administration of a specific neurokinin-1 receptor (NK-1R) antagonist to mice leads to reduced weight gain and circulating levels of insulin and leptin after high-fat diet (HFD). Here, we assessed the contribution of substance P (SP) and NK-1R in diet-induced obesity using NK-1R deficient [knockout (KO)] mice and extended our previous findings to show the effects of SP-NK-1R interactions on adipose tissue-associated insulin signaling and glucose metabolic responses. NK-1R KO and wild-type (WT) littermates were fed a HFD for 3 wk, and obesity-associated responses were determined. Compared with WT, NK-1 KO mice show reduced weight gain and circulating levels of leptin and insulin in response to HFD. Adiponectin receptor mRNA levels are higher in mesenteric fat and liver in NK-1 KO animals compared with WT, after HFD. Mesenteric fat from NK-1R KO mice fed with HFD has reduced stress-activated protein kinase/c-Jun N-terminal kinase and protein kinase C activation compared with WT mice. After glucose challenge, NK-1R KO mice remove glucose from the circulation more efficiently than WT and pair-fed controls, suggesting an additional peripheral effect of NK-1R-mediated signaling on glucose metabolism. Glucose uptake experiments in isolated rat adipocytes showed that SP directly inhibits insulin-mediated glucose uptake. Our results further establish a role for SP-NK-1R interactions in adipose tissue responses, specifically as they relate to obesity-associated pathologies such as glucose intolerance and insulin resistance. Our results highlight this pathway as an important therapeutic approach for type 2 diabetes.


Subject(s)
Adipose Tissue/metabolism , Dietary Fats/metabolism , Insulin/metabolism , Obesity/metabolism , Receptors, Neurokinin-1/metabolism , Substance P/metabolism , Animals , Female , Glucose/metabolism , Humans , Leptin/blood , Leptin/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Obesity/genetics , Obesity/physiopathology , Rats , Rats, Inbred F344 , Receptors, Neurokinin-1/genetics , Signal Transduction , Weight Gain
4.
Am J Physiol Gastrointest Liver Physiol ; 296(5): G1012-9, 2009 May.
Article in English | MEDLINE | ID: mdl-19282377

ABSTRACT

White adipose tissue is intimately involved in the regulation of immunity and inflammation. We reported that human mesenteric preadipocytes express the substance P (SP)-mediated neurokinin-1 receptor (NK-1R), which signals proinflammatory responses. Here we tested the hypothesis that SP promotes proliferation and survival of human mesenteric preadipocytes and investigated responsible mechanism(s). Preadipocytes were isolated from mesenteric fat biopsies during gastric bypass surgery. Proliferative and antiapoptotic responses were delineated in 3-(4,5-dimethylthiazol-2-yl)-5-(3-carboxymethoxyphenyl)-2-(4-sulfophenyl)-2H-tetrazolium (MTS), bromodeoxyuridine (BrdU), caspase-3, and TUNEL assays, as well as Western immunoanalysis. SP (10(-7) M) increased MTS and proliferation (BrdU) and time dependently (15-30 min) induced Akt, EGF receptor, IGF receptor, integrin alphaVbeta3, phosphatidylinositol 3-kinase, and PKC-theta phosphorylation. Furthermore, pharmacological antagonism of Akt and PKC-theta activation significantly attenuated SP-induced preadipocyte proliferation. Exposure of preadipocytes to the proapoptotic Fas ligand (FasL, 100 microM) resulted in nuclear DNA fragmentation (TUNEL assay), as well as increased cleaved poly (ADP-ribose) polymerase, cleaved caspase-7, and caspase-3 expression. Cotreatment with SP almost completely abolished these responses in a NK-1R-dependent fashion. SP (10(-7) M) also time dependently stimulated expression 4E binding protein 1 and phosphorylation of p70 S6 kinase, which increased protein translation efficiency. SP increases preadipocyte viability, reduces apoptosis, and stimulates proliferation, possibly via cell cycle upregulation and increased protein translation efficiency. SP-induced proliferative and antiapoptotic pathways in fat depots may contribute to development of the creeping fat and inflammation characteristic of Crohn's disease.


Subject(s)
Adipocytes/metabolism , Apoptosis , Cell Proliferation , Intra-Abdominal Fat/metabolism , Signal Transduction , Substance P/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Adipocytes/drug effects , Adipocytes/enzymology , Adipocytes/pathology , Apoptosis/drug effects , Caspase 3/metabolism , Caspase 7/metabolism , Cell Cycle , Cell Cycle Proteins , Cell Proliferation/drug effects , Cell Survival , Cells, Cultured , ErbB Receptors/metabolism , Fas Ligand Protein/metabolism , Humans , Integrin alphaVbeta3/metabolism , Intra-Abdominal Fat/drug effects , Intra-Abdominal Fat/enzymology , Intra-Abdominal Fat/pathology , Isoenzymes/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Phosphoinositide-3 Kinase Inhibitors , Phosphoproteins/metabolism , Phosphorylation , Poly(ADP-ribose) Polymerases/metabolism , Protein Kinase C/metabolism , Protein Kinase C-theta , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Proto-Oncogene Proteins c-akt/metabolism , Receptor, IGF Type 1/metabolism , Receptors, Neurokinin-1/metabolism , Ribosomal Protein S6 Kinases, 70-kDa/metabolism , Signal Transduction/drug effects , Time Factors , Tumor Necrosis Factor-alpha/metabolism
5.
Am J Pathol ; 173(2): 400-10, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18599605

ABSTRACT

Substance P (SP) regulates important intestinal functions, such as mucosal permeability, motility, chloride secretion, and inflammation via the neurokinin-1 receptor (NK-1R). Previous reports showed that vascularization and expression of angiogenic factors are evident in the colonic mucosa of rats with colitis and patients with inflammatory bowel disease. Here we determined whether SP is associated with angiogenesis. Human NCM460 colonocytes stably transfected with the human NK-1R (NCM460-NK-1R cells) and mice with dextran sodium sulfate-induced colitis were used. We found that expression of the angiogenic factor CCN1 was increased in the colons of patients with Crohn's disease and ulcerative colitis. Mucosal extracts from inflammatory bowel disease patients induced human intestinal microvascular endothelial cell migration that was inhibited by blockade of CCN1 and its receptor integrin alphavbeta3. Both the degree of angiogenesis and CCN1 expression were elevated in the colons of mice with dextran sodium sulfate-induced colitis, which was reduced by treatment with the NK-1R antagonist CJ-12255. SP also increased CCN1 expression in NCM460-NK-1R colonocytes. SP exposure to human intestinal microvascular endothelial cells co-cultured with NCM460-NK-1R cells induced angiogenic activity that was inhibited by CCN1 silencing. In addition, intracolonic overexpression of CCN1 induced angiogenesis in mouse colon. Thus, SP mediates angiogenesis via CCN1 during colitis.


Subject(s)
Colitis, Ulcerative/metabolism , Immediate-Early Proteins/biosynthesis , Intercellular Signaling Peptides and Proteins/biosynthesis , Receptors, Neurokinin-1/metabolism , Substance P/physiology , Animals , Bridged Bicyclo Compounds, Heterocyclic/pharmacology , Cell Movement , Colitis, Ulcerative/chemically induced , Colitis, Ulcerative/pathology , Colon/blood supply , Colon/pathology , Crohn Disease/metabolism , Crohn Disease/pathology , Cysteine-Rich Protein 61 , Dextran Sulfate , Endothelial Cells/physiology , Endothelium, Vascular/pathology , Endothelium, Vascular/physiopathology , Humans , Immediate-Early Proteins/antagonists & inhibitors , Integrin alphaVbeta3/metabolism , Male , Mice , Mice, Inbred C57BL , Microcirculation/pathology , Microcirculation/physiopathology , Neovascularization, Pathologic/metabolism , Substance P/pharmacology
6.
Gastroenterology ; 134(3): 747-55, 2008 Mar.
Article in English | MEDLINE | ID: mdl-18325388

ABSTRACT

BACKGROUND & AIMS: Substance P (SP) is an 11-amino acid peptide that belongs to the tachykinin family of peptides. SP acts in the brain and in the periphery as a neuropeptide, neurotransmitter, and hormone affecting diverse physiologic pathways, mainly via its high-affinity neurokinin-1 receptor (NK-1R). Its presence in the hypothalamus and other areas of the brain that regulate feeding as well as in the stomach and small intestine prompted us to investigate its role on appetite control and energy balance. METHODS: CJ 012,255 (CJ), a SP antagonist that binds to NK-1R, was injected into lean, diet-induced obese (DIO), and genetically obese (ob/ob) mice, and its effects on body weight, adiposity, and insulin sensitivity were investigated. RESULTS: CJ administration prevented weight gain and accumulation of fat after 2 weeks of high-fat feeding, whereas similar CJ treatment in obese mice (following 3 months of high-fat diet) resulted in weight loss, reduction in adiposity, and improvement of insulin sensitivity, in part because of inhibition of food intake. The effects of SP in the control of energy balance are, at least in part, leptin independent because CJ treatment was also effective in leptin-deficient mice. Peripheral SP administration resulted in a mild, dose-dependent increase in food intake, evident 3 hours post-SP injection. CONCLUSIONS: CJ reduces appetite and promotes weight loss in mice. We speculate that NK-1R antagonists, already tested in clinical trials for various diseases, may represent a potential target against obesity.


Subject(s)
Anti-Obesity Agents/pharmacology , Appetite Regulation/drug effects , Bridged Bicyclo Compounds, Heterocyclic/pharmacology , Energy Metabolism/drug effects , Feeding Behavior/drug effects , Neurokinin-1 Receptor Antagonists , Obesity/drug therapy , Substance P/metabolism , Adiposity/drug effects , Animals , Anti-Obesity Agents/administration & dosage , Blood Glucose/drug effects , Body Weight/drug effects , Bridged Bicyclo Compounds, Heterocyclic/administration & dosage , Dietary Fats/administration & dosage , Disease Models, Animal , Dose-Response Relationship, Drug , Injections, Intraperitoneal , Insulin/blood , Leptin/deficiency , Leptin/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Obese , Obesity/metabolism , Obesity/physiopathology , Receptors, Neurokinin-1/metabolism , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...