Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 32
Filter
Add more filters










Publication year range
1.
Diabetes ; 58(9): 2032-8, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19502416

ABSTRACT

OBJECTIVE: An increased expression of RELM-beta (resistin-like molecule-beta), a gut-derived hormone, is observed in animal models of insulin resistance/obesity and intestinal inflammation. Intestinal sugar absorption is modulated by dietary environment and hormones/cytokines. The aim of this study was to investigate the effect of RELM-beta on intestinal glucose absorption. RESEARCH DESIGN AND METHODS: Oral glucose tolerance test was performed in mice and rats in the presence and the absence of RELM-beta. The RELM-beta action on glucose transport in rat jejunal sacs, everted rings, and mucosal strips was explored as well as downstream kinases modulating SGLT-1 and GLUT2 glucose transporters. RESULTS: Oral glucose tolerance test carried out in rodents showed that oral administration of RELM-beta increased glycemia. Studies in rat jejunal tissue indicated that mucosal RELM-beta promoted absorption of glucose from the gut lumen. RELM-beta had no effect on paracellular mannitol transport, suggesting a transporter-mediated transcellular mechanism. In studies with jejunal mucosa mounted in Ussing chamber, luminal RELM-beta inhibited SGLT-1 activity in line with a diminished SGLT-1 abundance in brush border membranes (BBMs). Further, the potentiating effect of RELM-beta on jejunal glucose uptake was associated with an increased abundance of GLUT2 at BBMs. The effects of RELM-beta were associated with an increased amount of protein kinase C betaII in BBMs and an increased phosphorylation of AMP-activated protein kinase (AMPK). CONCLUSIONS: The regulation of SGLT-1 and GLUT2 by RELM-beta expands the role of gut hormones in short-term AMPK/protein kinase C mediated control of energy balance.


Subject(s)
Glucose Transporter Type 2/metabolism , Glucose/pharmacokinetics , Hormones, Ectopic/metabolism , Jejunum/metabolism , Sodium-Glucose Transporter 1/metabolism , AMP-Activated Protein Kinases/metabolism , Animals , Biological Transport, Active/drug effects , Biological Transport, Active/physiology , Glucose Tolerance Test , Hormones, Ectopic/pharmacology , Intercellular Signaling Peptides and Proteins , Intestinal Absorption/drug effects , Intestinal Absorption/physiology , Intestinal Mucosa/drug effects , Intestinal Mucosa/metabolism , Jejunum/drug effects , Male , Mice , Mice, Inbred C57BL , Phosphorylation/drug effects , Phosphorylation/physiology , Protein Kinase C/metabolism , Protein Kinase C beta , Rats , Rats, Wistar , Recombinant Proteins/pharmacology , Sodium/metabolism
2.
Inflamm Bowel Dis ; 14(7): 931-41, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18300276

ABSTRACT

BACKGROUND: Resistin and resistin-like molecule (RELM)beta comprise a novel class of cysteine-rich proteins secreted into the circulation implicated in hepatic insulin resistance and inflammation. RELMbeta is specifically produced by intestinal goblet cells but regulation of its expression and much of its local function are not elucidated. RELMbeta has been suggested to regulate colonic inflammation susceptibility, which is dependent on the mucosal barrier integrity. METHODS: In this work we explored the physiopathological role of RELMbeta in the colon. Among agents tested, carbachol and gastrin were strong inhibitors of RELMbeta mRNA accumulation. We examined the effect of recombinant RELMbeta on mucin secretion by human mucus-secreting HT29-Cl.16E cells in culture and by mouse colonic goblet cells in vivo. RESULTS: RELMbeta upregulated MUC2 and M1/MUC5AC gene expression in HT29-Cl.16E cells. RELMbeta enhanced M1/MUC5AC secretion by human colonic HT29-Cl.16E cells and MUC2 secretion by murine intestinal goblet cells. RELMbeta exerted its action exclusively on the apical side of HT29-Cl.16E cells, in agreement with its luminal mucosecretagogue effect in mice. Its action required calcium, protein kinase C, tyrosine kinases, and extracellular-regulated protein kinase activities and was synergized by carbachol. An intracolonic RELMbeta challenge was performed in the trinitrobenzene sulfonic acid (TNBS)-murine model of colitis and macroscopic and histological scores were monitored. The macroscopic and histopathological severity of TNBS-induced colitis was significantly attenuated by RELMbeta pretreatment. CONCLUSIONS: A direct participation in maintaining the mucosal defense barrier can be ascribed to RELMbeta in line with a regulatory role in intestinal inflammation.


Subject(s)
Colitis/physiopathology , Hormones, Ectopic/physiology , Intestinal Mucosa/metabolism , Mucus/metabolism , Trinitrobenzenesulfonic Acid , Animals , Blotting, Western , Calcium/physiology , Carbachol/pharmacology , Cells, Cultured , Colitis/chemically induced , Colitis/pathology , Gastrins/pharmacology , Gene Expression , Goblet Cells/metabolism , Hormones, Ectopic/genetics , Hormones, Ectopic/pharmacology , Humans , Intercellular Signaling Peptides and Proteins , Intestines , Mice , Mucin 5AC , Mucin-2 , Mucins/genetics , Protein Kinase C/physiology , Protein Kinases/physiology , Protein-Tyrosine Kinases/physiology , RNA, Messenger/drug effects , Recombinant Proteins/pharmacology , Reverse Transcriptase Polymerase Chain Reaction
3.
J Immunol ; 179(10): 7012-20, 2007 Nov 15.
Article in English | MEDLINE | ID: mdl-17982092

ABSTRACT

SAMP1/Fc mice develop spontaneous ileitis that shares many features with human Crohn's disease. One of the earliest features of ileitis in SAMP1/Fc mice is an increase in the number of ileal goblet and intermediate cells. Resistin-like molecule beta (RELMbeta) is a goblet cell-specific, cysteine-rich peptide previously shown to function as part of the innate immune response. In this study, we examined the role of expression of RELMbeta in the initiation of ileal inflammation in SAMP1/Fc mice. RELMbeta was highly induced in the ilea of SAMP1/Fc mice beginning at age 5 wk, coincident with the histological appearance of inflammation. RELMbeta was found in ileal goblet cells and some intermediate and Paneth cells. Surprisingly, RELMbeta mRNA levels were significantly increased in the ilea of 80% of germ-free SAMP1/Fc mice examined compared with specific pathogen-free AKR control mice of similar age. Ileitis was observed in germfree SAMP1/Fc mice, although it was attenuated relative to specific pathogen-free SAMP1/Fc mice. These data suggest that neither the early induction of RELMbeta expression nor ileal inflammation requires the presence of viable intestinal flora. Neither was the induction of RELMbeta dependent on the major Th1 or Th2 cytokines. However, RELMbeta stimulated naive bone marrow-derived macrophages to secrete significant amounts of TNF-alpha, IL-6, and RANTES. Our data suggest that RELMbeta is involved in the initiation of ileitis in SAMP1/Fc mice and may act through the induction of proinflammatory cytokines from resident immune cells within the mucosa.


Subject(s)
Goblet Cells/immunology , Hormones, Ectopic/immunology , Ileitis/immunology , Paneth Cells/immunology , Animals , Crohn Disease/genetics , Crohn Disease/immunology , Crohn Disease/pathology , Cytokines/genetics , Cytokines/immunology , Disease Models, Animal , Gene Expression Regulation/immunology , Goblet Cells/pathology , Hormones, Ectopic/genetics , Hormones, Ectopic/pharmacology , Ileitis/genetics , Ileitis/pathology , Inflammation/genetics , Inflammation/immunology , Inflammation/pathology , Inflammation Mediators/immunology , Intercellular Signaling Peptides and Proteins , Macrophages/immunology , Macrophages/pathology , Mice , Mice, Transgenic , Paneth Cells/pathology , Th1 Cells/immunology , Th1 Cells/pathology , Th2 Cells/immunology , Th2 Cells/pathology
4.
Am J Physiol Lung Cell Mol Physiol ; 293(2): L305-13, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17545488

ABSTRACT

Resistin-like molecule (RELM)-beta is a cysteine-rich cytokine implicated in insulin resistance and asthmatic responses, but its function remains an enigma. We now report that RELM-beta has a role in promoting airway inflammation and lung remodeling in the mouse lung. RELM-beta is strongly induced by diverse allergens and T helper type 2 (Th2) cytokines by an IL-13- and STAT6-dependent mechanism. To understand the in vivo role of RELM-beta, we delivered recombinant murine RELM-beta intratracheally to naïve mice. RELM-beta induced dose-dependent leukocyte accumulation (most prominently involving macrophages) and goblet cell hyperplasia. The most prominent effect induced by RELM-beta was increased perivascular and peribronchial collagen deposition. Mice genetically deficient in RELM-beta had reduced accumulation of collagen and goblet cell hyperplasia in an experimental model of allergic airway inflammation. In vitro experiments demonstrated that RELM-beta had fibroblast motogenic activity. These results identify RELM-beta as a Th2-associated cytokine with potent inflammatory and remodeling activity.


Subject(s)
Allergens/immunology , Hormones, Ectopic/immunology , Lung/immunology , Pneumonia/immunology , Allergens/metabolism , Animals , Asthma/immunology , Asthma/metabolism , Bronchoalveolar Lavage Fluid/cytology , Bronchoalveolar Lavage Fluid/immunology , Cell Movement , Collagen/metabolism , Female , Fibroblasts/immunology , Fibroblasts/metabolism , Fibroblasts/pathology , Goblet Cells/immunology , Hormones, Ectopic/genetics , Hormones, Ectopic/pharmacology , Intercellular Signaling Peptides and Proteins , Interleukin-13/metabolism , Interleukin-4/metabolism , Lung/metabolism , Lung/pathology , Male , Mice , Mice, Inbred BALB C , Mice, Mutant Strains , NIH 3T3 Cells , Pneumonia/metabolism , Pneumonia/pathology , Respiratory Mucosa/immunology , Respiratory Mucosa/metabolism , Respiratory Mucosa/pathology , STAT6 Transcription Factor/metabolism , Th2 Cells/drug effects , Th2 Cells/immunology , Th2 Cells/metabolism
5.
J Clin Invest ; 116(11): 2914-23, 2006 Nov.
Article in English | MEDLINE | ID: mdl-17024245

ABSTRACT

Although inflammatory bowel disease (IBD) is the result of a dysregulated immune response to commensal gut bacteria in genetically predisposed individuals, the mechanism(s) by which bacteria lead to the development of IBD are unknown. Interestingly, deletion of intestinal goblet cells protects against intestinal injury, suggesting that this epithelial cell lineage may produce molecules that exacerbate IBD. We previously reported that resistin-like molecule beta (RELMbeta; also known as FIZZ2) is an intestinal goblet cell-specific protein that is induced upon bacterial colonization whereupon it is expressed in the ileum and colon, regions of the gut most often involved in IBD. Herein, we show that disruption of this gene reduces the severity of colitis in the dextran sodium sulfate (DSS) model of murine colonic injury. Although RELMbeta does not alter colonic epithelial proliferation or barrier function, we show that recombinant protein activates macrophages to produce TNF-alpha both in vitro and in vivo. RELMbeta expression is also strongly induced in the terminal ileum of the SAMP1/Fc model of IBD. These results suggest a model whereby the loss of epithelial barrier function by DSS results in the activation of the innate mucosal response by RELMbeta located in the lumen, supporting the hypothesis that this protein is a link among goblet cells, commensal bacteria, and the pathogenesis of IBD.


Subject(s)
Colitis/metabolism , Colitis/pathology , Dextran Sulfate/pharmacology , Hormones, Ectopic/metabolism , Animals , Cell Movement/drug effects , Cell Proliferation , Colitis/chemically induced , Colitis/microbiology , Disease Models, Animal , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Epithelial Cells/pathology , Gene Expression Regulation , Hormones, Ectopic/genetics , Hormones, Ectopic/pharmacology , Inflammation/chemically induced , Inflammation/pathology , Intercellular Signaling Peptides and Proteins , Leukocyte Count , Lymphocytes/cytology , Lymphocytes/drug effects , Lymphocytes/metabolism , Macrophages/drug effects , Mice , Mice, Knockout
6.
Eur J Endocrinol ; 153(3): R1-5, 2005 Sep.
Article in English | MEDLINE | ID: mdl-16131594

ABSTRACT

OBJECTIVE: Several hormones expressed in white adipose tissue influence food intake at the central level. We sought to determine whether resistin, a circulating adipose-derived hormone in rodents, has actions on the hypothalamus by determining the effects of central resistin injection on food intake and on hypothalamic Fos protein expression. DESIGN: As resistin expression in adipose tissue is influenced by altered nutritional status, we studied the effect of central resistin in both fed and pre-fasted rats. RESULTS: In fasted rats, central injection of resistin decreased food intake acutely and increased the number of cells that express Fos protein in the arcuate nucleus but not in any other hypothalamic structure. The effect on food intake was dose-dependent and did not result in the formation of a conditioned taste aversion. CONCLUSIONS: Taken together, these results provide the first evidence documenting a central action of resistin, which could be involved in a feedback loop targeting the hypothalamus. On the other hand, since we observed resistin mRNA in the arcuate and ventromedial nuclei of the hypothalamus, it is also possible that brain-derived resistin serves as a neuropeptide involved in the regulation of energy homeostasis. However, since resistin-induced satiety was modest and transient, as central administration for several days did not affect body weight, the physiological relevance and therapeutic potential of the observed principal phenomenon may be limited.


Subject(s)
Eating/drug effects , Hormones, Ectopic/pharmacology , Hypothalamus/drug effects , Satiety Response/drug effects , Adiponectin , Animals , Body Weight/drug effects , Eating/physiology , Hypothalamus/metabolism , Immunohistochemistry , In Situ Hybridization , Injections, Intraventricular , Insulin/blood , Intercellular Signaling Peptides and Proteins/blood , Leptin/blood , Male , Proto-Oncogene Proteins c-fos/metabolism , RNA, Messenger/genetics , Rats , Resistin , Satiety Response/physiology , Statistics, Nonparametric , Taste/physiology
7.
J Clin Endocrinol Metab ; 90(8): 4852-7, 2005 Aug.
Article in English | MEDLINE | ID: mdl-15886251

ABSTRACT

CONTEXT: A newly discovered hormone resistin has been shown to be increased in women with polycystic ovary syndrome (PCOS). OBJECTIVE: The purpose of this study was to confirm increased resistin concentrations in women with PCOS and to test the direct effect of resistin on human theca cell androgen production. DESIGN: Resistin was measured in fasting serum samples by RIA. To test the direct effects of resistin on ovarian androgen biosynthesis, human theca cells were cultured with resistin for 3 d in the presence and absence of forskolin and insulin. PATIENTS: Fasting serum samples were obtained from 45 women with PCOS and 74 regularly cycling premenopausal control women in the follicular phase of their menstrual cycles, and ovarian theca cell cultures were established from two control women. RESULTS: The mean serum resistin concentration was increased (40%) in women with PCOS. Serum resistin concentrations correlated positively with body mass index and testosterone in PCOS women but not in controls. There were no significant correlations between resistin and fasting insulin or indicators of insulin resistance when corrected for body mass index. In cultured human theca cells, basal 17alpha-hydroxylase activity was unchanged by resistin alone, but resistin enhanced 17alpha-hydroxylase activity in the presence of forskolin or a combination of forskolin plus insulin. Resistin (> or =1 ng/ml) augmented forskolin and forskolin plus insulin stimulation of CYP17 mRNA expression in a concentration-dependent manner. CONCLUSION: These data indicate that abnormal resistin secretion in PCOS may play a role in causing ovarian hyperandrogenism.


Subject(s)
Hormones, Ectopic/blood , Hyperandrogenism/metabolism , Polycystic Ovary Syndrome/metabolism , Steroid 17-alpha-Hydroxylase/metabolism , Theca Cells/enzymology , Adult , Cells, Cultured , Colforsin/pharmacology , Female , Hormones, Ectopic/pharmacology , Humans , Hypoglycemic Agents/pharmacology , Insulin/pharmacology , Insulin Resistance , Resistin , Theca Cells/drug effects
8.
J Vasc Surg ; 41(4): 691-8, 2005 Apr.
Article in English | MEDLINE | ID: mdl-15874935

ABSTRACT

OBJECTIVE: Resistin, a novel adipocyte-derived cytokine, is involved in the development of insulin resistance and diabetes mellitus. In this study, we determined whether resistin could affect vasomotor function, oxidative stress, and endothelial nitric oxide synthase (eNOS) expression in porcine coronary arteries. METHODS: Porcine coronary arteries were treated with resistin or antioxidant seleno-L-methionine (SeMet). Vasomotor function was studied by using a myograph system. Levels of superoxide anion (O 2 - ) were detected by the lucigenin-enhanced chemiluminescence method. The eNOS mRNA and protein levels were determined by real-time polymerase chain reaction and immunohistochemistry, respectively. Culture of isolated porcine coronary artery endothelial cells (PCAECs) was also included. RESULTS: Endothelium-dependent relaxation in response to bradykinin was reduced by 15% and 30% for the rings treated with 10 and 40 ng/mL of resistin, respectively, as compared with controls ( P < .05). Endothelium-independent relaxation in response to sodium nitroprusside (SNP) was also reduced by 11% after treatment with 40 ng/mL of resistin ( P < .05). The O 2 - level was increased in the 40 ng/mL resistin-treated vessels by 88% as compared with controls ( P < .05). SeMet reversed these effects. The eNOS mRNA levels in PCAEC cultures treated with resistin (10 and 40 ng/mL) were decreased by 27% and 55%, respectively ( P < .05) and by 39% in the endothelial cells purified from porcine coronary artery rings after treatment with 40 ng/mL of resistin ( P < .05). Immunoreactivity of eNOS in the resistin-treated vessel rings was also substantially reduced. CONCLUSIONS: Resistin reduces the endothelium-dependent and endothelium-independent vasorelaxation. This effect is associated with increased superoxide radical production, decreased eNOS expression, and is effectively reversed by the antioxidant SeMet. CLINICAL RELEVANCE: Obesity has been considered to be an independent risk factor for coronary artery disease and other vascular lesions. Resistin is a newly discovered adipocyte-derived cytokine, and its plasma levels are increased in obese individuals. However, it is not clear whether resistin could directly contribute to vascular disease formation. This study showed that resistin can cause endothelial dysfunction in porcine coronary arteries through oxidative stress and down-regulation of eNOS. Thus, this study may suggest a new mechanism of obesity-associated vascular disease and that antioxidants may effectively prevent vascular disease in obese individuals.


Subject(s)
Coronary Vessels/drug effects , Endothelial Cells/drug effects , Endothelial Cells/physiology , Hormones, Ectopic/pharmacology , Vasodilation/drug effects , Vasodilation/physiology , Animals , Cell Culture Techniques , Coronary Vessels/cytology , Models, Animal , Nitric Oxide/metabolism , Nitric Oxide Synthase/metabolism , Recombinant Proteins/pharmacology , Resistin , Swine
9.
Endocrinology ; 146(8): 3334-42, 2005 Aug.
Article in English | MEDLINE | ID: mdl-15905315

ABSTRACT

Beyond their effects on central metabolic functions, leptin, resistin, and adiponectin have profound effects on a number of other physiologic processes, including immune function and inflammation. Although leptin, resistin, and adiponectin are produced in human placenta and adipose tissue, their immunoregulatory actions in these tissues are not known. Therefore, the aim of this study was to determine the effect of leptin, resistin, and adiponectin on the release of proinflammatory mediators in human placenta and sc adipose tissue. Samples were obtained from normal pregnancies at the time of cesarean section. Tissue explants (n = 5) were incubated in the absence (basal control) or presence of a leptin (1, 10, and 100 ng/ml), resistin (1, 10, and 100 ng/ml), and adiponectin (0.1 and 0.5 microg/ml). After 6 h incubation, the medium was collected, and the release of IL-1beta, IL-6, TNFalpha, prostaglandin (PG)F2alpha and PGE2 was quantified by ELISA. There was no effect of resistin on proinflammatory cytokine or prostaglandin release; however, leptin at 100 ng/ml and adiponectin at 0.1 and/or 0.5 microg/ml significantly increased the release of IL-1beta, IL-6, TNFalpha, and PGE2 from human placenta and adipose tissue. Although both leptin and adiponectin significantly increased PGF2alpha release from human placenta, there was no effect of these hormones on PGF2alpha release from adipose tissue. Furthermore, this leptin- and adiponectin-induced proinflammatory response could be abrogated by treatment with the antiinflammatory ERK1/2 MAPK inhibitor U0126, the peroxisomal proliferator-activated receptor-gamma ligand troglitazone, and the nuclear factor-kappaB inhibitor BAY 11-7082. Collectively, these data indicate that leptin and adiponectin activate proinflammatory cytokine release and phospholipid metabolism in human placenta and adipose tissue, and antiinflammatory agents can abrogate leptin- and adiponectin-induced inflammation.


Subject(s)
Adipose Tissue/physiology , Cytokines/metabolism , Inflammation/physiopathology , Intercellular Signaling Peptides and Proteins/pharmacology , Leptin/pharmacology , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , NF-kappa B/metabolism , PPAR gamma/physiology , Placenta/physiology , Prostaglandins/metabolism , Adiponectin , Adipose Tissue/cytology , Adipose Tissue/drug effects , Cell Survival , Female , Hormones, Ectopic/pharmacology , Humans , Organ Culture Techniques , Placenta/cytology , Placenta/drug effects , Pregnancy , Resistin
10.
Peptides ; 26(5): 845-51, 2005 May.
Article in English | MEDLINE | ID: mdl-15808915

ABSTRACT

Obesity is a major risk factor for the development of hypertension. Adipokines may cause hypertension by acting both centrally and directly on the vascular vessels. We wished to clarify whether three adipokines, leptin, resistin and tumor necrosis factor-alpha, affect expression of adrenomedullin and endothelin-1 in vascular endothelial cells. Human umbilical vein endothelial cells were cultured for 24 h with leptin (1-10 nmol/l), resistin (1-10 nmol/l) or tumor necrosis factor-alpha (1-10 ng/ml). Expression of adrenomedullin and endothelin-1 was examined by radioimmunoassay and northern blot analysis. Immunoreactive-adrenomedullin in the medium and adrenomedullin mRNA expression levels were decreased by treatment of tumor necrosis factor-alpha time- and dose-dependently, whereas endothelin-1 secretion was not significantly changed by it. Leptin or resistin had no significant effects on expression of adrenomedullin or endothelin-1 in human umbilical vein endothelial cells. Under hypoxic conditions (1% O2), expression of both adrenomedullin and endothelin-1 was induced in these cells. Immunoreactive-adrenomedullin levels in the medium were decreased by treatment of tumor necrosis factor-alpha under hypoxia. Leptin or resistin had no significant effects on adrenomedullin or endothelin-1 expression also in hypoxia. These findings have raised the possibility that decreased expression of adrenomedullin by tumor necrosis factor-alpha may be related to the increased risk of hypertension and other cardiovascular diseases in obese subjects.


Subject(s)
Endothelin-1/metabolism , Endothelium, Vascular/metabolism , Hormones, Ectopic/pharmacology , Leptin/pharmacology , Peptides/metabolism , Tumor Necrosis Factor-alpha/pharmacology , Adrenomedullin , Cells, Cultured , Endothelin-1/genetics , Endothelium, Vascular/drug effects , Humans , Obesity/etiology , Obesity/metabolism , Peptides/genetics , RNA, Messenger/analysis , RNA, Messenger/metabolism , Resistin
11.
Endocrinology ; 146(5): 2200-9, 2005 May.
Article in English | MEDLINE | ID: mdl-15705777

ABSTRACT

Human FIZZ3 (hFIZZ3) was identified as an ortholog of mouse resistin (mResistin), an adipocyte-specific secreted factor linked to insulin resistance in rodents. Unlike mResistin, hFIZZ3 is expressed in macrophages and monocytes, but is undetectable in adipose tissue. The profound macrophage infiltration of adipose that occurs during obesity suggests that hFIZZ3 may play an important role in adipocyte biology. Using a recombinant protein produced in Escherichia coli, we report here that chronic treatment of cultured human adipocytes with hFIZZ3 results in hypotropic cells with smaller lipid droplets. Recombinant hFIZZ3 facilitates preadipocyte proliferation and stimulates adipocyte triglyceride lipolysis, whereas recombinant mResistin inhibits adipocyte differentiation, with no detectable effect on proliferation or lipolysis. In addition, insulin-stimulated glucose uptake and Akt phosphorylation are not altered in hFIZZ3-treated adipocytes, indicating an intact insulin response. In mouse adipose explants, hFIZZ3 accelerates simultaneously triglyceride lipolysis and fatty acid reesterification, as assessed by measurement of glycerol and fatty acid release. Consistent with the in vitro findings, acute administration of recombinant hFIZZ3 into normal mice caused a significant increase in serum glycerol concentration with no elevation in free fatty acid at 45 min post injection. Taken together, the data suggest that recombinant hFIZZ3 can influence adipose metabolism by regulating preadipocyte cell number, adipocyte lipid content, and energy expenditure via accelerating the fatty acid/triglyceride futile cycle.


Subject(s)
Adipocytes/metabolism , Hormones, Ectopic/pharmacology , Lipolysis/drug effects , Recombinant Proteins/pharmacology , Adipocytes/cytology , Adipocytes/drug effects , Animals , Cell Differentiation , Cell Division/drug effects , Cells, Cultured , Esterification , Fatty Acids/metabolism , Glucose/metabolism , Glycerol/metabolism , Humans , In Vitro Techniques , Insulin/pharmacology , Mice , Phosphorylation , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-akt , Resistin , Signal Transduction/drug effects , Triglycerides/metabolism
12.
Mol Cell Biol ; 25(4): 1569-75, 2005 Feb.
Article in English | MEDLINE | ID: mdl-15684405

ABSTRACT

Resistin is an adipocyte hormone that modulates glucose homeostasis. Here we show that in 3T3-L1 adipocytes, resistin attenuates multiple effects of insulin, including insulin receptor (IR) phosphorylation, IR substrate 1 (IRS-1) phosphorylation, phosphatidylinositol-3-kinase (PI3K) activation, phosphatidylinositol triphosphate production, and activation of protein kinase B/Akt. Remarkably, resistin treatment markedly induces the gene expression of suppressor of cytokine signaling 3 (SOCS-3), a known inhibitor of insulin signaling. The 50% effective dose for resistin induction of SOCS-3 is approximately 20 ng/ml, close to levels of resistin in serum. Association of SOCS-3 protein with the IR is also increased by resistin. Inhibition of SOCS function prevented resistin from antagonizing insulin action in adipocytes. SOCS-3 induction is the first cellular effect of resistin that is independent of insulin and is a likely mediator of resistin's inhibitory effect on insulin signaling in adipocytes.


Subject(s)
Adipocytes/metabolism , Enzyme Activation/physiology , Hormones, Ectopic/pharmacology , Insulin Resistance/physiology , Repressor Proteins/metabolism , Transcription Factors/metabolism , 3T3-L1 Cells , Adipocytes/cytology , Adipocytes/drug effects , Animals , Cell Differentiation/physiology , Cells, Cultured , Enzyme Activation/drug effects , Hormones, Ectopic/administration & dosage , Insulin/metabolism , Insulin Receptor Substrate Proteins , Mice , Phosphatidylinositol 3-Kinases/metabolism , Phosphoproteins/metabolism , Phosphorylation/drug effects , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-akt , Receptor, Insulin/metabolism , Resistin , Signal Transduction/drug effects , Signal Transduction/physiology , Suppressor of Cytokine Signaling 3 Protein , Suppressor of Cytokine Signaling Proteins
13.
Endocrinology ; 146(1): 273-9, 2005 Jan.
Article in English | MEDLINE | ID: mdl-15471967

ABSTRACT

The adipocyte-derived hormone resistin has been proposed as a possible link between obesity and insulin resistance in murine models. Many recent studies have reported physiological roles for resistin in glucose homeostasis, one of which is enhancement of glucose production from the liver by up-regulating gluconeogenic enzymes such as glucose-6-phosphatase and phosphoenolpyruvate carboxykinase. However, its in vivo roles in lipid metabolism still remain to be clarified. In this study, we investigated the effects of resistin overexpression on insulin action and lipid metabolism in C57BL/6 mice using an adenoviral gene transfer technique. Elevated plasma resistin levels in mice treated with the resistin adenovirus (AdmRes) were confirmed by Western blotting analysis and RIAs. Fasting plasma glucose levels did not differ between AdmRes-treated mice and controls, but the basal insulin concentration was significantly elevated in AdmRes-treated mice. In AdmRes-treated mice, the glucose-lowering effect of insulin was impaired, as evaluated by insulin tolerance tests. Furthermore, total cholesterol and triglyceride concentrations were significantly higher, whereas the high-density lipoprotein cholesterol level was significantly lower. Lipoprotein analysis revealed that low-density lipoprotein was markedly increased in AdmRes-treated mice, compared with controls. In addition, in vivo Triton WR-1339 studies showed evidence of enhanced very low-density lipoprotein production in AdmRes-treated mice. The expressions of genes involved in lipoprotein metabolism, such as low-density lipoprotein receptor and apolipoprotein AI in the liver, were decreased. These results suggest that resistin overexpression induces dyslipidemia in mice, which is commonly seen in the insulin-resistant state, partially through enhanced secretion of lipoproteins.


Subject(s)
Hormones, Ectopic , Hyperlipidemias/chemically induced , Adenoviridae/genetics , Animals , Cholesterol/blood , Gene Expression , Gene Transfer Techniques , Genetic Vectors , Hormones, Ectopic/blood , Hormones, Ectopic/genetics , Hormones, Ectopic/pharmacology , Hyperlipidemias/blood , Hyperlipidemias/physiopathology , Insulin Resistance , Lipoproteins/metabolism , Lipoproteins, LDL/blood , Lipoproteins, VLDL/biosynthesis , Liver/metabolism , Male , Mice , Mice, Inbred BALB C , Muscle, Skeletal/metabolism , Resistin , Time Factors
14.
Acta Pharmacol Sin ; 25(12): 1705-11, 2004 Dec.
Article in English | MEDLINE | ID: mdl-15569419

ABSTRACT

AIM: To detect the resistin expression of white adipose tissue in diet-induced obese (DIO) versus diet-resistant (DR) rats, and to investigate the relationship of mutated resistin and 3T3-L1 preadipocytes differentiation. METHODS: RT-PCR and Western Blot were used to detect gene /protein expression. 3T3-L1 cells were cultured, transfected, and induced to differentiation using 0.5 mmol/L 3-isobutyl-1-methylxanthine (MIX), 1 mg/L insulin, and 1 micromol/L dexamethasone. Oil red O staining was applied to detect the degree of preadipocytes differentiation. RESULTS: Expression of resistin mRNA was upregulated in DIO rats and downregulated in DR rats. However, the expression levels varied greatly within the groups. Sequencing of the resistin genes from DIO and DR rats revealed a Leu9Val (C25G) missense mutation within the signal peptide in one DR rat. The mutant resistin inhibited preadipocyte differentiation. Local experiments and Western blotting with tagged resistin fusion proteins identified both mutant and wild type proteins in the cytoplasm and secreted into the culture medium. Computer predictions using the Proscan and Subloc programs revealed four putative phosphorylation sites and a possible leucine zipper motif within the rat resistin protein. CONCLUSION: Resistin-increased differentiation may be inhibited by the mutation-containing precursor protein, or by the mutant non-secretory resistin isoform.


Subject(s)
Adipocytes/cytology , Hormones, Ectopic/genetics , Mutation, Missense , Protein Sorting Signals/genetics , 3T3-L1 Cells , Adipocytes/drug effects , Animals , Cell Differentiation/drug effects , Dietary Fats , Gene Expression/drug effects , Genetic Vectors , Hormones, Ectopic/pharmacology , Male , Mice , Obesity/genetics , Obesity/metabolism , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Rats , Rats, Sprague-Dawley , Resistin , Transfection
15.
Circulation ; 110(21): 3335-40, 2004 Nov 23.
Article in English | MEDLINE | ID: mdl-15545519

ABSTRACT

BACKGROUND: Resistin, a novel adipokine, is elevated in patients with type 2 diabetes and may play a role in the vascular complications of this disorder. One recent study has shown that resistin has a proinflammatory effect on endothelial cells. However, there is no information on whether resistin could also affect vascular smooth muscle cells (SMCs). Thus, the purpose of this study was to assess whether resistin could induce SMC proliferation and to study the mechanisms whereby resistin signals in SMCs. METHODS AND RESULTS: Human aortic smooth muscle cells (HASMCs) were stimulated with increasing concentrations of resistin for 48 hours. Cell proliferation was induced by resistin in a dose-dependent manner as assessed by direct cell counting. To gain more insights into the mechanism of action of resistin, we investigated the extracellular signal-regulated kinase (ERK) and/or phosphatidylinositol 3-kinase (PI3K) signaling pathways. Transient phosphorylation of the p42/44 mitogen-activated protein kinase (ERK 1/2) occurred after addition of resistin to HASMCs. U0126, a specific inhibitor of ERK phosphorylation, significantly inhibited ERK 1/2 phosphorylation and reduced resistin-simulated proliferation of HASMCs. LY294002, a specific PI3K inhibitor, also significantly inhibited HASMC proliferation after resistin stimulation. CONCLUSIONS: Our results demonstrate that resistin induces HASMC proliferation through both ERK 1/2 and Akt signaling pathways. The proliferative action exerted by resistin on HASMCs may account in part for the increased incidence of restenosis in diabetes patients.


Subject(s)
Hormones, Ectopic/pharmacology , Mitogen-Activated Protein Kinase 1/physiology , Mitogen-Activated Protein Kinase 3/physiology , Muscle, Smooth, Vascular/drug effects , Myocytes, Smooth Muscle/drug effects , Protein Serine-Threonine Kinases/physiology , Proto-Oncogene Proteins/physiology , Signal Transduction/drug effects , Aorta/cytology , Arteriosclerosis/metabolism , Butadienes/pharmacology , Cell Division/drug effects , Cells, Cultured/cytology , Cells, Cultured/drug effects , Cells, Cultured/enzymology , Chromones/pharmacology , Diabetes Complications/metabolism , Hormones, Ectopic/antagonists & inhibitors , Imidazoles/pharmacology , Mitogen-Activated Protein Kinase 1/antagonists & inhibitors , Mitogen-Activated Protein Kinase 3/antagonists & inhibitors , Morpholines/pharmacology , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/enzymology , Myocytes, Smooth Muscle/cytology , Myocytes, Smooth Muscle/enzymology , Nitriles/pharmacology , Phosphatidylinositol 3-Kinases/physiology , Phosphoinositide-3 Kinase Inhibitors , Protein Serine-Threonine Kinases/antagonists & inhibitors , Proto-Oncogene Proteins/antagonists & inhibitors , Proto-Oncogene Proteins c-akt , Pyridines/pharmacology , Recombinant Proteins/pharmacology , Resistin , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors , p38 Mitogen-Activated Protein Kinases/physiology
16.
Eur J Endocrinol ; 150(6): 885-92, 2004 Jun.
Article in English | MEDLINE | ID: mdl-15191360

ABSTRACT

OBJECTIVE: To investigate the relationship between resistin (a potential link between obesity and type 2 diabetes) and preadipocyte differentiation. DESIGN: A rat resistin expression vector was transfected into 3T3-L1 preadipocytes and differentiation was compared between normal 3T3-L1 cells, rat resistin-transfected cells and non-transfected cells grown in conditioned medium taken from resistin-expressing cultures. METHODS: The rat resistin gene was inserted into the pDual GC and pEFGP-N2 expression vectors for examination of the effects of resistin overexpression in 3T3-L1 cells before and after differentiation was stimulated with 3-isobutyl-1-methyxanthine (MIX), insulin and dexamethasone (DEX). Smaller conserved fragments were inserted into short interference RNA (siRNA) expression vectors, for examination of the effect of targeted resistin inhibition on differentiation of resistin-overexpressing 3T3-L1 cells. RESULTS: Prior to stimulation, the resistin-transfected 3T3-L1 cells contained many more small lipid droplets than did non-transfected 3T3-L1 cells. Following stimulation, differentiation in the resistin-transfected 3T3-L1 cells was dramatically promoted, especially in the early stages. Stimulation of differentiation was also observed in non-transfected 3T3-L1 cells grown in resistin protein-containing conditioned medium. The expression of adipocyte differentiation-associated markers such as CCAAT enhancer binding protein (C/EBPalpha), retinoid X receptor (RXRalpha) and lipoprotein lipase (LPL) was upregulated in resistin-overexpressing cells, whereas expression of preadipocyte factor-1 (Pref-1), an inhibitor of preadipocyte differentiation, was downregulated. In addition, expression of two of the three tested siRNAs inhibited the adipoconversion process, providing further evidence that resistin promotes the differentiation of preadipocytes to adipocytes. CONCLUSION: Resistin can promote preadipocyte differentiation. Based on this, we propose that resistin may be an important candidate mediator of obesity-induced insulin resistance.


Subject(s)
Adipocytes/cytology , Cell Differentiation/drug effects , Hormones, Ectopic/pharmacology , Stem Cells/cytology , 1-Methyl-3-isobutylxanthine/pharmacology , 3T3-L1 Cells , Animals , Cytoplasm/chemistry , Dexamethasone/pharmacology , Gene Expression , Hormones, Ectopic/analysis , Hormones, Ectopic/genetics , Insulin/pharmacology , Mice , RNA, Small Interfering/genetics , RNA, Small Interfering/physiology , Rats , Resistin , Transfection
17.
Eur J Pharmacol ; 493(1-3): 41-4, 2004 Jun 16.
Article in English | MEDLINE | ID: mdl-15189762

ABSTRACT

Adiponectin (Adipocyte Complement-Related Protein of 30 kDa, ACRP30) and resistin are adipocyte-derived polypeptide hormones playing a role in metabolic homeostasis. Their plasma levels are inversely (adiponectin) or directly (resistin) correlated to obesity and they have opposite effects on insulin sensitivity. Adipose tissue hormones such as leptin have been shown to modulate neurotransmitters which control feeding in the hypothalamus. We have studied the effects of adiponectin and resistin on dopamine, norepinephrine and serotonin release from hypothalamic neuronal endings (synaptosomes) in vitro. We have found that adiponectin does not modify either basal or depolarization-induced amine release, while resistin inhibits the stimulated release of dopamine and norepinephrine, leaving unaffected serotonin release. We can conclude that, similarly to leptin, but differently from adiponectin, the adipose tissue hormone resistin could affect the central mechanisms of feeding by inhibiting catecholamine release in the hypothalamus.


Subject(s)
Dopamine/metabolism , Hormones, Ectopic/pharmacology , Hypothalamus/metabolism , Intercellular Signaling Peptides and Proteins/physiology , Norepinephrine/antagonists & inhibitors , Adiponectin , Animals , Area Under Curve , Dopamine Antagonists/pharmacology , Drug Evaluation, Preclinical/methods , Hormones, Ectopic/chemistry , Hypothalamus/cytology , Hypothalamus/drug effects , Intercellular Signaling Peptides and Proteins/chemistry , Italy , Male , Norepinephrine/metabolism , Perfusion , Rats , Rats, Wistar , Resistin , Serotonin/metabolism , Serotonin/pharmacology , Synaptosomes/drug effects , Synaptosomes/metabolism , Tritium
18.
Science ; 304(5674): 1154-8, 2004 May 21.
Article in English | MEDLINE | ID: mdl-15155948

ABSTRACT

Resistin, founding member of the resistin-like molecule (RELM) hormone family, is secreted selectively from adipocytes and induces liver-specific antagonism of insulin action, thus providing a potential molecular link between obesity and diabetes. Crystal structures of resistin and RELMbeta reveal an unusual multimeric structure. Each protomer comprises a carboxy-terminal disulfide-rich beta-sandwich "head" domain and an amino-terminal alpha-helical "tail" segment. The alpha-helical segments associate to form three-stranded coiled coils, and surface-exposed interchain disulfide linkages mediate the formation of tail-to-tail hexamers. Analysis of serum samples shows that resistin circulates in two distinct assembly states, likely corresponding to hexamers and trimers. Infusion of a resistin mutant, lacking the intertrimer disulfide bonds, in pancreatic-insulin clamp studies reveals substantially more potent effects on hepatic insulin sensitivity than those observed with wild-type resistin. This result suggests that processing of the intertrimer disulfide bonds may reflect an obligatory step toward activation.


Subject(s)
Disulfides/chemistry , Hormones, Ectopic/chemistry , Hormones, Ectopic/metabolism , Intercellular Signaling Peptides and Proteins , Adipocytes/metabolism , Adiponectin , Amino Acid Sequence , Animals , Cell Line , Crystallization , Crystallography, X-Ray , Culture Media, Conditioned , Glucose/metabolism , Hormones, Ectopic/genetics , Hormones, Ectopic/pharmacology , Humans , Insulin/administration & dosage , Insulin/blood , Insulin Resistance , Liver/metabolism , Mice , Molecular Sequence Data , Molecular Weight , Mutation , Protein Folding , Protein Structure, Quaternary , Protein Structure, Secondary , Protein Structure, Tertiary , Proteins/chemistry , Proteins/metabolism , Resistin
19.
J Clin Endocrinol Metab ; 88(12): 6098-106, 2003 Dec.
Article in English | MEDLINE | ID: mdl-14671216

ABSTRACT

Resistin, an adipocyte secreted factor, has been suggested to link obesity with type 2 diabetes in rodent models, but its relevance to human diabetes remains uncertain. Although previous studies have suggested a role for this adipocytokine as a pathogenic factor, its functional effects, regulation by insulin, and alteration of serum resistin concentration by diabetes status remain to be elucidated. Therefore, the aims of this study were to analyze serum resistin concentrations in type 2 diabetic subjects; to determine the in vitro effects of insulin and rosiglitazone (RSG) on the regulation of resistin, and to examine the functional effects of recombinant human resistin on glucose and lipid metabolism in vitro. Serum concentrations of resistin were analyzed in 45 type 2 diabetic subjects and 34 nondiabetic subjects. Subcutaneous human adipocytes were incubated in vitro with insulin, RSG, and insulin in combination with RSG to examine effects on resistin secretion. Serum resistin was increased by approximately 20% in type 2 diabetic subjects compared with nondiabetic subjects (P = 0.004) correlating with C-reactive protein. No other parameters, including adiposity and fasting insulin levels, correlated with serum resistin in this cohort. However, in vitro, insulin stimulated resistin protein secretion in a concentration-dependent manner in adipocytes [control, 1215 +/- 87 pg/ml (mean +/- SEM); 1 nM insulin, 1414.0 +/- 89 pg/ml; 1 microM insulin, 1797 +/- 107 pg/ml (P < 0.001)]. RSG (10 nM) reduced the insulin-mediated rise in resistin protein secretion (1 nM insulin plus RSG, 971 +/- 35 pg/ml; insulin, 1 microM insulin plus RSG, 1019 +/- 28 pg/ml; P < 0.01 vs. insulin alone). Glucose uptake was reduced after treatment with 10 ng/ml recombinant resistin and higher concentrations (P < 0.05). Our in vitro studies demonstrated a small, but significant, reduction in glucose uptake with human recombinant resistin in differentiated preadipocytes. In human abdominal sc adipocytes, RSG blocks the insulin-mediated release of resistin secretion in vitro. In conclusion, elevated serum resistin in human diabetes reflects the subclinical inflammation prevalent in type 2 diabetes. Our in vitro studies suggest a modest effect of resistin in reducing glucose uptake, and suppression of resistin expression may contribute to the insulin-sensitizing and glucose-lowering actions of the thiazolidinediones.


Subject(s)
Adipocytes/cytology , Adipocytes/metabolism , Diabetes Mellitus, Type 2/metabolism , Glucose/metabolism , Hormones, Ectopic/metabolism , Intercellular Signaling Peptides and Proteins , Lipid Metabolism , Adipocytes/drug effects , Adult , Blotting, Western , Case-Control Studies , Cell Differentiation , Cross Reactions , Diabetes Mellitus, Type 2/pathology , Female , Gene Expression , Glucose/pharmacokinetics , Hormones, Ectopic/blood , Hormones, Ectopic/genetics , Hormones, Ectopic/pharmacology , Humans , Hypoglycemic Agents/pharmacology , Insulin/pharmacology , Leptin/metabolism , Lipolysis/drug effects , Male , Middle Aged , Recombinant Proteins/pharmacology , Resistin , Reverse Transcriptase Polymerase Chain Reaction , Rosiglitazone , Stem Cells/metabolism , Subcutaneous Tissue/drug effects , Subcutaneous Tissue/metabolism , Thiazolidinediones/pharmacology
20.
Eur J Endocrinol ; 149(3): 231-8, 2003 Sep.
Article in English | MEDLINE | ID: mdl-12943526

ABSTRACT

DESIGN: It has recently been shown that deficiency of adrenomedullin (AM), a potent vasodilator peptide, leads to insulin resistance. We studied expression of AM in NIH 3T3-L1 adipocytes and compared it with expression of resistin, an adipocyte-derived peptide hormone that is proposed to cause insulin resistance. Moreover, we studied the effects of tumor necrosis factor-alpha (TNF-alpha), a known mediator of insulin resistance, on the expression of AM and resistin in 3T3-L1 adipocytes. METHODS: 3T3-L1 cells were induced to differentiate to adipocytes by insulin, dexamethasone and 3-isobutyl-1-methylxanthine. Expression of AM mRNA and resistin mRNA was examined by Northern blot analysis. Immunoreactive AM in the medium was measured by RIA. RESULTS: AM mRNA was expressed in preadipocytes, but barely detectable in adipocytes. Immunoreactive AM was detected in the medium of both preadipocytes and adipocytes, with about 2.5 times higher levels found in preadipocytes. In contrast, resistin mRNA was expressed in adipocytes, whereas it was not detected in preadipocytes. Treatment with TNF-alpha increased AM expression in both adipocytes and preadipocytes, whereas it decreased resistin mRNA levels in adipocytes. CONCLUSIONS: The present study has shown that AM expression was down-regulated and resistin expression was up-regulated during adipocyte differentiation of 3T3-L1 cells. TNF-alpha acted as a potent negative regulator of resistin expression and a potent positive regulator of AM expression in adipocytes, raising the possibility that in addition to its known actions in causing insulin resistance, TNF-alpha may also have actions against insulin resistance through AM and resistin.


Subject(s)
Adipocytes/drug effects , Adipocytes/metabolism , Hormones, Ectopic/biosynthesis , Intercellular Signaling Peptides and Proteins , Peptides/metabolism , Proteins , Tumor Necrosis Factor-alpha/pharmacology , 1-Methyl-3-isobutylxanthine/pharmacology , 3T3 Cells , Adrenomedullin , Animals , Blotting, Northern , Cell Differentiation/drug effects , Cell Differentiation/physiology , Dexamethasone/pharmacology , Dose-Response Relationship, Drug , Gene Expression Regulation/drug effects , Glucocorticoids/pharmacology , Hormones, Ectopic/genetics , Hormones, Ectopic/pharmacology , Humans , Insulin/pharmacology , Insulin Resistance/physiology , Interferon-alpha/pharmacology , Interleukin-1/pharmacology , Mice , Nerve Growth Factor , Peptides/genetics , Peptides/pharmacology , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Recombinant Proteins/pharmacology , Resistin
SELECTION OF CITATIONS
SEARCH DETAIL
...