Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 79
Filter
1.
Proc Natl Acad Sci U S A ; 114(42): 11027-11033, 2017 10 17.
Article in English | MEDLINE | ID: mdl-28973871

ABSTRACT

The mammalian intestine is colonized by trillions of bacteria that perform essential metabolic functions for their hosts. The mutualistic nature of this relationship depends on maintaining spatial segregation between these bacteria and the intestinal epithelial surface. This segregation is achieved in part by the presence of a dense mucus layer at the epithelial surface and by the production of antimicrobial proteins that are secreted by epithelial cells into the mucus layer. Here, we show that resistin-like molecule ß (RELMß) is a bactericidal protein that limits contact between Gram-negative bacteria and the colonic epithelial surface. Mouse and human RELMß selectively killed Gram-negative bacteria by forming size-selective pores that permeabilized bacterial membranes. In mice lacking RELMß, Proteobacteria were present in the inner mucus layer and invaded mucosal tissues. Another RELM family member, human resistin, was also bactericidal, suggesting that bactericidal activity is a conserved function of the RELM family. Our findings thus identify the RELM family as a unique family of bactericidal proteins and show that RELMß promotes host-bacterial mutualism by regulating the spatial segregation between the microbiota and the intestinal epithelium.


Subject(s)
Gastrointestinal Microbiome , Gram-Negative Bacteria , Hormones, Ectopic/physiology , Intestinal Mucosa/microbiology , Animals , Humans , Immunity, Innate , Intercellular Signaling Peptides and Proteins , Intestinal Mucosa/immunology , Lipid Metabolism , Mice , Resistin/physiology , Symbiosis
2.
J Immunol ; 187(1): 450-61, 2011 Jul 01.
Article in English | MEDLINE | ID: mdl-21602491

ABSTRACT

Found in inflammatory zone (FIZZ) 2, also known as resistin-like molecule (RELM)-ß, belongs to a novel cysteine-rich secreted protein family named FIZZ/RELM. Its function is unclear, but a closely related family member, FIZZ1, has profibrotic activities. The human ortholog of rodent FIZZ1 has not been identified, but human FIZZ2 has significant sequence homology to both rodent FIZZ2 (59%) and FIZZ1 (50%). Given the greater homology to rodent FIZZ2, analyzing the role of FIZZ2 in a rodent model of bleomycin-induced pulmonary fibrosis would be of greater potential relevance to human fibrotic lung disease. The results showed that FIZZ2 was highly induced in lungs of rodents with bleomycin-induced pulmonary fibrosis and of human patients with idiopathic pulmonary fibrosis. FIZZ2 expression was induced in rodent and human lung epithelial cells by Th2 cytokines, which was mediated via STAT6 signaling. The FIZZ2 induction in murine lungs was found to be essential for pulmonary fibrosis, as FIZZ2 deficiency significantly suppressed pulmonary fibrosis and associated enhanced extracellular matrix and cytokine gene expression. In vitro analysis indicated that FIZZ2 could stimulate type I collagen and α-smooth muscle actin expression in lung fibroblasts. Furthermore, FIZZ2 was shown to have chemoattractant activity for bone marrow (BM) cells, especially BM-derived CD11c(+) dendritic cells. Notably, lung recruitment of BM-derived cells was impaired in FIZZ2 knockout mice. These findings suggest that FIZZ2 is a Th2-associated multifunctional mediator with potentially important roles in the pathogenesis of fibrotic lung diseases.


Subject(s)
Hormones, Ectopic/biosynthesis , Intercellular Signaling Peptides and Proteins/biosynthesis , Pulmonary Fibrosis/immunology , Pulmonary Fibrosis/metabolism , Animals , Bone Marrow Cells/pathology , Cell Movement/immunology , Cell Proliferation , Cells, Cultured , Disease Models, Animal , Female , Fibroblasts/pathology , Hormones, Ectopic/genetics , Hormones, Ectopic/physiology , Humans , Intercellular Signaling Peptides and Proteins/genetics , Intercellular Signaling Peptides and Proteins/physiology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Inbred CBA , Mice, Knockout , Mice, Transgenic , Myofibroblasts/pathology , Pulmonary Fibrosis/pathology , Rats , Rats, Inbred F344 , Th2 Cells/immunology , Th2 Cells/metabolism , Th2 Cells/pathology
3.
J Immunol ; 186(12): 7205-14, 2011 Jun 15.
Article in English | MEDLINE | ID: mdl-21555532

ABSTRACT

Guanylate cyclase C (GUCY2C or GC-C) and its ligands, guanylin (GUCA2A or Gn) and uroguanylin (GUCA2B or Ugn), are expressed in intestinal epithelial cells and regulate ion secretion, intestinal barrier function, and epithelial monolayer homeostasis via cGMP-dependent signaling pathways. The aim of this study was to determine whether GC-C and its ligands direct the course of intestinal inflammation. In this article, we show that dextran sodium sulfate (DSS)-induced clinical disease and histological damage to the colonic mucosa were significantly less severe in GC-C(-/-) mice and moderately reduced in Gn(-/-) animals. Relative to wild-type controls, GC-C(-/-) and Gn(-/-) mice had reduced apoptosis and increased proliferation of intestinal epithelial cells during DSS colitis. Basal and DSS-induced production of resistin-like molecule ß (RELMß) was substantially diminished in GC-C(-/-) mice. RELMß is thought to stimulate cytokine production in macrophages in this disease model and, consistent with this, TNF-α and IFN-γ production was minimal in GC-C(-/-) animals. RELMß and cytokine levels were similar to wild-type in Gn(-/-) mice, however. Colonic instillation of recombinant RELMß by enema into GC-C(-/-) mice restores sensitivity to DSS-mediated mucosal injury. These findings demonstrate a novel role for GC-C signaling in facilitating mucosal wounding and inflammation, and further suggest that this may be mediated, in part, through control of RELMß production.


Subject(s)
Guanylate Cyclase/physiology , Animals , Colonic Diseases/etiology , Colonic Diseases/pathology , Gastrointestinal Hormones/physiology , Hormones, Ectopic/biosynthesis , Hormones, Ectopic/physiology , Inflammation/etiology , Intercellular Signaling Peptides and Proteins , Interferon-gamma/biosynthesis , Intestinal Mucosa/pathology , Mice , Mice, Knockout , Natriuretic Peptides/physiology , Tumor Necrosis Factor-alpha/biosynthesis
4.
J Exp Med ; 206(13): 2947-57, 2009 Dec 21.
Article in English | MEDLINE | ID: mdl-19995957

ABSTRACT

Th2 cells drive protective immunity against most parasitic helminths, but few mechanisms have been demonstrated that facilitate pathogen clearance. We show that IL-4 and IL-13 protect against intestinal lumen-dwelling worms primarily by inducing intestinal epithelial cells (IECs) to differentiate into goblet cells that secrete resistin-like molecule (RELM) beta. RELM-beta is essential for normal spontaneous expulsion and IL-4-induced expulsion of Nippostrongylus brasiliensis and Heligmosomoides polygyrus, which both live in the intestinal lumen, but it does not contribute to immunity against Trichinella spiralis, which lives within IEC. RELM-beta is nontoxic for H. polygyrus in vitro but directly inhibits the ability of worms to feed on host tissues during infection. This decreases H. polygyrus adenosine triphosphate content and fecundity. Importantly, RELM-beta-driven immunity does not require T or B cells, alternative macrophage activation, or increased gut permeability. Thus, we demonstrate a novel mechanism for host protection at the mucosal interface that explains how stimulation of epithelial cells by IL-4 and IL-13 contributes to protection against parasitic helminthes that dwell in the intestinal lumen.


Subject(s)
Gastrointestinal Tract/parasitology , Hormones, Ectopic/physiology , Intestinal Mucosa/immunology , Nematospiroides dubius/immunology , Nippostrongylus/immunology , Animals , Intercellular Signaling Peptides and Proteins , Interleukin-13/physiology , Interleukin-4/physiology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL
5.
Gastroenterology ; 137(5): 1716-24.e1-2, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19706296

ABSTRACT

BACKGROUND & AIMS: The composition of the gut microbiome is affected by host phenotype, genotype, immune function, and diet. Here, we used the phenotype of RELMbeta knockout (KO) mice to assess the influence of these factors. METHODS: Both wild-type and RELMbeta KO mice were lean on a standard chow diet, but, upon switching to a high-fat diet, wild-type mice became obese, whereas RELMbeta KO mice remained comparatively lean. To investigate the influence of diet, genotype, and obesity on microbiome composition, we used deep sequencing to characterize 25,790 16S rDNA sequences from uncultured bacterial communities from both genotypes on both diets. RESULTS: We found large alterations associated with switching to the high-fat diet, including a decrease in Bacteroidetes and an increase in both Firmicutes and Proteobacteria. This was seen for both genotypes (ie, in the presence and absence of obesity), indicating that the high-fat diet itself, and not the obese state, mainly accounted for the observed changes in the gut microbiota. The RELMbeta genotype also modestly influenced microbiome composition independently of diet. Metagenomic analysis of 537,604 sequence reads documented extensive changes in gene content because of a high-fat diet, including an increase in transporters and 2-component sensor responders as well as a general decrease in metabolic genes. Unexpectedly, we found a substantial amount of murine DNA in our samples that increased in proportion on a high-fat diet. CONCLUSIONS: These results demonstrate the importance of diet as a determinant of gut microbiome composition and suggest the need to control for dietary variation when evaluating the composition of the human gut microbiome.


Subject(s)
Diet , Dietary Fats/administration & dosage , Intestines/microbiology , Obesity/metabolism , Obesity/microbiology , RNA, Ribosomal, 16S/genetics , Animals , Female , Hormones, Ectopic/physiology , Intercellular Signaling Peptides and Proteins , Metagenome , Mice , Mice, Inbred C57BL , Mice, Knockout , Obesity/genetics
6.
J Immunol ; 181(7): 4709-15, 2008 Oct 01.
Article in English | MEDLINE | ID: mdl-18802073

ABSTRACT

The secreted goblet cell-derived protein resistin-like molecule beta (RELMbeta) has been implicated in divergent functions, including a direct effector function against parasitic helminths and a pathogenic function in promoting inflammation in models of colitis and ileitis. However, whether RELMbeta influences CD4(+) T cell responses in the intestine is unknown. Using a natural model of intestinal inflammation induced by chronic infection with gastrointestinal helminth Trichuris muris, we identify dual functions for RELMbeta in augmenting CD4(+) Th1 cell responses and promoting infection-induced intestinal inflammation. Following exposure to low-dose Trichuris, wild-type C57BL/6 mice exhibit persistent infection associated with robust IFN-gamma production and intestinal inflammation. In contrast, infected RELMbeta(-/-) mice exhibited a significantly reduced expression of parasite-specific CD4(+) T cell-derived IFN-gamma and TNF-alpha and failed to develop Trichuris-induced intestinal inflammation. In in vitro T cell differentiation assays, recombinant RELMbeta activated macrophages to express MHC class II and secrete IL-12/23p40 and enhanced their ability to mediate Ag-specific IFN-gamma expression in CD4(+) T cells. Taken together, these data suggest that goblet cell-macrophage cross-talk, mediated in part by RELMbeta, can promote adaptive CD4(+) T cell responses and chronic inflammation following intestinal helminth infection.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Goblet Cells/immunology , Hormones, Ectopic/physiology , Inflammation Mediators/physiology , Interferon-gamma/biosynthesis , Intestinal Diseases, Parasitic/immunology , Intestinal Diseases, Parasitic/pathology , Trichuriasis/immunology , Acute Disease , Adjuvants, Immunologic/physiology , Animals , CD4-Positive T-Lymphocytes/cytology , CD4-Positive T-Lymphocytes/metabolism , Cell Communication/immunology , Chronic Disease , Goblet Cells/metabolism , Hormones, Ectopic/genetics , Intercellular Signaling Peptides and Proteins , Intestinal Diseases, Parasitic/metabolism , Macrophages/immunology , Macrophages/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Resistin/genetics , Resistin/physiology , Trichuriasis/metabolism , Trichuriasis/pathology , Trichuris/immunology
8.
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
9.
J Allergy Clin Immunol ; 118(1): 257-68, 2006 Jul.
Article in English | MEDLINE | ID: mdl-16815164

ABSTRACT

BACKGROUND: Resistin-like molecule (RELM) beta is a cysteine-rich cytokine expressed in the gastrointestinal tract and implicated in insulin resistance and gastrointestinal nematode immunity; however, its function primarily remains an enigma. OBJECTIVE: We sought to elucidate the function of RELM-beta in the gastrointestinal tract. METHODS: We generated RELM-beta gene-targeted mice and examined colonic epithelial barrier function, gene expression profiles, and susceptibility to acute colonic inflammation. RESULTS: We show that RELM-beta is constitutively expressed in the colon by goblet cells and enterocytes and has a role in homeostasis, as assessed by alterations in colon mRNA transcripts and epithelial barrier function in the absence of RELM-beta. Using acute colonic inflammatory models, we demonstrate that RELM-beta has a central role in the regulation of susceptibility to colonic inflammation. Mechanistic studies identify that RELM-beta regulates expression of type III regenerating gene (REG) (REG3beta and gamma), molecules known to influence nuclear factor kappaB signaling. CONCLUSIONS: These data define a critical role for RELM-beta in the maintenance of colonic barrier function and gastrointestinal innate immunity. CLINICAL IMPLICATIONS: These findings identify RELM-beta as an important molecule in homeostatic gastrointestinal function and colonic inflammation, and as such, these results have implications for a variety of human inflammatory gastrointestinal conditions, including allergic gastroenteropathies.


Subject(s)
Colitis/etiology , Colon/physiology , Hormones, Ectopic/physiology , Animals , Intercellular Signaling Peptides and Proteins , Interleukin-13/physiology , Mice , Mice, Inbred C57BL , Pancreatitis-Associated Proteins , Permeability , Proteins/genetics
10.
J Endocrinol ; 186(3): 457-65, 2005 Sep.
Article in English | MEDLINE | ID: mdl-16135665

ABSTRACT

The aim of this study was to determine the release and regulation of leptin, resistin and adiponectin from human placenta and fetal membranes, and maternal subcutaneous adipose tissue and skeletal muscle obtained from normal and gestational diabetes mellitus (GDM)-complicated pregnancies at the time of Cesarean section. Tissue explants were incubated in the absence (basal control) or presence of 10 mug/ml lipopolysaccharide (LPS), 10, 20 or 40 ng/ml tumor necrosis factor-alpha (TNF-alpha), interleukin (IL)-6 and IL-8, 1 microM phorbol myristate acetate, 10, 20 and 40 mM glucose, 0.1, 1 and 10 microM insulin and 0.1 1 and 10 microM dexamethasone, progesterone and estrogen. After an 18-h incubation, the medium was collected and the release of leptin, resistin and adiponectin was quantified by ELISA. Human gestational tissues and maternal tissues released immunoreactive leptin, resistin and adiponectin; however, there was no difference in the release of either resistin or adiponectin between normal pregnant women and women with gestational diabetes. The release of leptin was significantly higher in placenta, amnion and choriodecidua obtained from normal pregnant women compared with women with GDM. However, in maternal tissues, the situation was reversed, with adipose tissue and skeletal muscle obtained from women with GDM releasing significantly greater amounts of leptin. In adipose tissue and skeletal muscle the release of leptin was significantly greater in insulin-controlled GDM compared with diet-controlled GDM, and leptin release from adipose tissue was significantly correlated with maternal body mass index. In all tissues tested, there was no effect of incubation with LPS, IL-6, IL-8 or TNF-alpha on leptin, resistin or adiponectin release. PMA significantly increased the release of resistin from placenta and adipose tissue. Insulin increased placental resistin release, whereas the hormones dexamethasone, progesterone and estrogen significantly decreased placental resistin release. The data presented in this study demonstrate that dysregulation of leptin metabolism and/or function in the placenta may be implicated in the pathogenesis of GDM. Furthermore, resistin release is greatly affected by a variety of inflammatory mediators and hormones.


Subject(s)
Diabetes, Gestational/physiopathology , Extraembryonic Membranes/metabolism , Hormones/physiology , Placenta/metabolism , Adiponectin , Adipose Tissue/metabolism , Adult , Anti-Inflammatory Agents/pharmacology , Case-Control Studies , Cytokines/pharmacology , Dexamethasone/pharmacology , Diabetes, Gestational/metabolism , Estrogens/pharmacology , Female , Glucose/pharmacology , Hormones, Ectopic/physiology , Humans , Insulin/pharmacology , Intercellular Signaling Peptides and Proteins/physiology , Leptin/physiology , Lipopolysaccharides/pharmacology , Muscle, Skeletal/metabolism , Organ Culture Techniques , Pregnancy , Progesterone/pharmacology , Resistin , Tetradecanoylphorbol Acetate/pharmacology
11.
J Biol Chem ; 280(36): 31679-85, 2005 Sep 09.
Article in English | MEDLINE | ID: mdl-15983036

ABSTRACT

The adipocytokine resistin impairs glucose tolerance and insulin sensitivity in rodents. Here, we examined the effect of resistin on glucose uptake in isolated adult mouse cardiomyocytes. Murine resistin reduced insulin-stimulated glucose uptake, establishing the heart as a resistin target tissue. Notably, human resistin also impaired insulin action in mouse cardiomyocytes, providing the first evidence that human and mouse resistin homologs have similar functions. Resistin is a cysteine-rich molecule that circulates as a multimer of a dimeric form dependent upon a single intermolecular disulfide bond, which, in the mouse, involves Cys26; mutation of this residue to alanine (C26A) produces a monomeric molecule that appears to be bioactive in the liver. Remarkably, unlike native resistin, monomeric C26A resistin had no effect on basal or insulin-stimulated glucose uptake in mouse cardiomyocytes. Resistin impairs glucose uptake in cardiomyocytes by mechanisms that involve altered vesicle trafficking. Thus, in cardiomyocytes, both mouse and human resistins directly impair glucose transport; and in contrast to effects on the liver, these actions of resistin require oligomerization.


Subject(s)
Glucose/metabolism , Hormones, Ectopic/physiology , Myocytes, Cardiac/metabolism , Animals , Biological Transport, Active/physiology , Dimerization , Exocytosis/physiology , Glucose/antagonists & inhibitors , Glucose Transporter Type 4 , Hormones, Ectopic/genetics , Hormones, Ectopic/metabolism , Humans , Insulin/physiology , Male , Mice , Mice, Inbred C57BL , Monosaccharide Transport Proteins/metabolism , Muscle Proteins/metabolism , Mutation , Phosphorylation , Protein Serine-Threonine Kinases/metabolism , Protein Transport/physiology , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-akt , Resistin , Transferrin/metabolism
12.
Circ Res ; 96(10): 1042-52, 2005 May 27.
Article in English | MEDLINE | ID: mdl-15920027

ABSTRACT

For many years adipose tissue was viewed as the site where excess energy was stored, in the form of triglycerides (TGs), and where that energy, when needed elsewhere in the body, was released in the form of fatty acids (FAs). Recently, it has become clear that when the regulation of the storage and release of energy by adipose tissue is impaired, plasma FA levels become elevated and excessive metabolism of FA, including storage of TGs, occurs in nonadipose tissues. Most recently, work by several laboratories has made it clear that in addition to FA, adipose tissue communicates with the rest of the body by synthesizing and releasing a host of secreted molecules, collectively designated as adipokines. Several recent reviews have described how these molecules, along with FA, significantly effect total body glucose metabolism and insulin sensitivity. Relatively little attention has been paid to the effects of adipokines on lipid metabolism. In this review, we will describe, in detail, the effects of molecules secreted by adipose tissue, including FA, leptin, adiponectin, resistin, TNF-alpha, IL-6, and apolipoproteins, on lipid homeostasis in several nonadipose tissues, including liver, skeletal muscle, and pancreatic beta cells.


Subject(s)
Adipocytes/metabolism , Adipose Tissue/metabolism , Insulin Resistance , Lipid Metabolism , Adiponectin , Animals , Apolipoproteins/metabolism , Fatty Acids/metabolism , Homeostasis , Hormones, Ectopic/physiology , Humans , Insulin/metabolism , Insulin Secretion , Intercellular Signaling Peptides and Proteins/physiology , Interleukin-6/pharmacology , Islets of Langerhans/metabolism , Leptin/physiology , Lipoproteins, VLDL/metabolism , Liver/metabolism , Resistin , Tumor Necrosis Factor-alpha/pharmacology
13.
J Surg Res ; 126(1): 121-9, 2005 Jun 01.
Article in English | MEDLINE | ID: mdl-15916985

ABSTRACT

Adipose tissue has recently emerged as an active endocrine organ that secretes a variety of metabolically important substances, collectively called adipocytokines or adipokines. In this review we summarize the effects of the adipokines leptin, adiponectin, and resistin on the vasculature and their potential role for pathogenesis of vascular disease. Leptin is associated with arterial wall thickness, decreased vessel distensibility, and elevated C reactive protein (CRP) levels. Leptin possesses procoagulant and antifibrinolytic properties, and it promotes thrombus and atheroma formation, probably through the leptin receptors by promoting vascular inflammation, proliferation, and calcification, and by increasing oxidative stress. Research for development of pharmacologic antagonism for the leptin receptor is currently under way. Adiponectin inhibits the expression of the adhesion molecules ICAM-1, VCAM-1, and P selectin. Therefore, it interferes with monocyte adherence to endothelial cells and their subsequent migration to the subendothelial space, one of the initial events in the development of atherosclerosis. Adiponectin also inhibits the transformation of macrophages to foam cells in vitro and decreases their phagocytic activity. Resistin, discovered in 2001, represents the newest of the adipokines and was named for its ability to promote insulin resistance. Resistin increases the expression of the adhesion molecules VCAM-1 and ICAM-1, up-regulates the monocyte chemoattractant chemokine-1, and promotes endothelial cell activation via ET-1 release. Although many aspects of its function need further clarification, it appears that resistin will add significantly to our knowledge of the pathophysiology of vascular disease and the metabolic syndrome.


Subject(s)
Endothelium, Vascular/physiology , Hormones, Ectopic/physiology , Intercellular Signaling Peptides and Proteins/physiology , Leptin/physiology , Vascular Diseases/etiology , Adiponectin , Animals , Arteriosclerosis/etiology , Humans , Resistin , Thrombosis/etiology
14.
J Immunol ; 174(9): 5789-95, 2005 May 01.
Article in English | MEDLINE | ID: mdl-15843582

ABSTRACT

The adipokine resistin is suggested to be an important link between obesity and insulin resistance. In the present study, we assessed the impact of resistin as inflammatogenic cytokine in the setting of arthritis. In vitro experiments on human PBMC were performed to assess cytokine response and transcription pathways of resistin-induced inflammation. Proinflammatory properties of resistin were evaluated in animal model by intra-articular injection of resistin followed by histological evaluation of the joint. Levels of resistin were assessed by ELISA in 74 paired blood and synovial fluid samples of patients with rheumatoid arthritis. Results were compared with the control group comprised blood samples from 34 healthy individuals and 21 synovial fluids from patients with noninflammatory joint diseases. We now show that resistin displays potent proinflammatory properties by 1) strongly up-regulating IL-6 and TNF-alpha, 2) responding to TNF-alpha challenge, 3) enhancing its own activity by a positive feedback, and finally 4) inducing arthritis when injected into healthy mouse joints. Proinflammatory properties of resistin were abrogated by NF-kappaB inhibitor indicating the importance of NF-kappaB signaling pathway for resistin-induced inflammation. Resistin is also shown to specifically accumulate in the inflamed joints of patients with rheumatoid arthritis and its levels correlate with other markers of inflammation. Our results indicate that resistin is a new and important member of the cytokine family with potent regulatory functions. Importantly, the identified properties of resistin make it a novel and interesting therapeutic target in chronic inflammatory diseases such as rheumatoid arthritis.


Subject(s)
Adipocytes/immunology , Adipocytes/metabolism , Hormones, Ectopic/physiology , Inflammation Mediators/physiology , Adolescent , Adult , Aged , Aged, 80 and over , Animals , Arthritis, Experimental/immunology , Arthritis, Experimental/metabolism , Arthritis, Experimental/pathology , Arthritis, Rheumatoid/immunology , Arthritis, Rheumatoid/metabolism , Arthritis, Rheumatoid/pathology , Cells, Cultured , Female , Hormones, Ectopic/administration & dosage , Humans , Inflammation Mediators/administration & dosage , Injections, Intra-Articular , Interleukin-1/biosynthesis , Interleukin-1/genetics , Interleukin-6/biosynthesis , Interleukin-6/genetics , Intracellular Fluid/immunology , Intracellular Fluid/metabolism , Male , Mice , Middle Aged , NF-kappa B/metabolism , NF-kappa B/physiology , Recombinant Proteins/administration & dosage , Recombinant Proteins/pharmacology , Resistin , Signal Transduction/immunology , Synovial Membrane/immunology , Synovial Membrane/metabolism , Synovial Membrane/pathology , Tumor Necrosis Factor-alpha/biosynthesis , Tumor Necrosis Factor-alpha/genetics
15.
Am J Physiol Endocrinol Metab ; 288(3): E625-32, 2005 Mar.
Article in English | MEDLINE | ID: mdl-15522996

ABSTRACT

Resistin is an adipocyte-secreted hormone proposed to link obesity with insulin resistance and diabetes, but no previous study has performed a joint quantitative evaluation of white adipose tissue (WAT) resistin mRNA expression and serum levels in relation to insulinemia and glycemia in mice. We have thus comparatively assessed WAT resistin mRNA expression and serum resistin levels in lean C57BL/6J mice and various mouse models of obesity, including diet-induced obese (DIO) C57BL/6J mice, high fat-fed TNF-alpha-/- mice, and brown adipose tissue (BAT)-deficient uncoupling protein-diphtheria toxin A chain (UCP1-DTA) mice. We also studied whether treatment with the weight-reducing and insulin-sensitizing compounds, MTII, an alpha-melanocyte-stimulating hormone analog, or CNTF(Ax15), a ciliary neurotrophic factor analog, alters resistin mRNA expression and/or circulating levels in lean and DIO C57BL/6J mice. We find that resistin mRNA expression is similar in DIO and lean C57BL/6J mice, as well as in TNF-alpha-/- and wild-type (WT) mice. Circulating resistin levels, however, are higher in DIO C57BL/6J, high fat-fed TNF-alpha-/-, and UCP1-DTA mice compared with lean controls. Moreover, although resistin mRNA expression is upregulated by MTII treatment for 24 h and downregulated by CNTF(Ax15) treatment for 3 or 7 days, circulating resistin levels are not altered by MTII or CNTF(Ax15) treatment. In addition, serum resistin levels, but not resistin mRNA expression levels, are correlated with body weight, and neither resistin mRNA expression nor serum resistin levels are correlated with serum insulin or glucose levels. We conclude that transcriptional regulation of resistin in WAT does not correlate with circulating resistin levels and that circulating resistin is unlikely to play a major endocrine role in insulin resistance or glycemia in mice.


Subject(s)
Blood Glucose/physiology , Hormones, Ectopic/physiology , Insulin Resistance/physiology , alpha-MSH/analogs & derivatives , Adipose Tissue/chemistry , Adipose Tissue/drug effects , Adipose Tissue/metabolism , Animals , Blood Glucose/drug effects , Body Weight/drug effects , Carrier Proteins/genetics , Ciliary Neurotrophic Factor/analogs & derivatives , Ciliary Neurotrophic Factor/pharmacology , Diet , Diphtheria Toxin/genetics , Disease Models, Animal , Eating/drug effects , Energy Intake/drug effects , Gene Expression/genetics , Hormones, Ectopic/blood , Hormones, Ectopic/genetics , Insulin/blood , Ion Channels , Male , Membrane Proteins/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Obese , Mice, Transgenic , Mitochondrial Proteins , Multivariate Analysis , Obesity/chemically induced , Obesity/metabolism , Peptide Fragments/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Resistin , Reverse Transcriptase Polymerase Chain Reaction , Tumor Necrosis Factor-alpha/genetics , Uncoupling Protein 1 , alpha-MSH/agonists , alpha-MSH/pharmacology
16.
Cell Mol Life Sci ; 61(19-20): 2485-96, 2004 Oct.
Article in English | MEDLINE | ID: mdl-15526156

ABSTRACT

Resistin is a newly discovered adipocyte hormone. It is related to resistin-like molecules alpha, beta and gamma in structure and function. Resistin is produced by white and brown adipose tissues but has also has been identified in several other tissues, including the hypothalamus, pituitary and adrenal glands, pancreas, gastrointestinal tract, myocytes, spleen, white blood cells and plasma. The tissue level of resistin is decreased by insulin, cytokines such as tumour necrosis factor alpha, endothelin-1 and increased by growth and gonadal hormones, hyperglycaemia, male gender and some proinflammatory cytokines, such as interleukin-6 and lipopolysaccharide. Resistin antagonizes insulin action, and it is downregulated by rosiglitazone and peroxisome proliferator-activated receptor gamma agonists. Since evidence of a direct link between resistin genotype and human diabetes is still weak, more molecular, physiological and clinical studies are needed to determine the role of resistin in the aetiology of type 2 diabetes.


Subject(s)
Hormones, Ectopic/chemistry , Hormones, Ectopic/physiology , Adipose Tissue/metabolism , Amino Acid Sequence , Animals , Cytokines/metabolism , Diabetes Mellitus, Type 2/metabolism , Female , Glucose/metabolism , Humans , Insulin/metabolism , Insulin Resistance , Interleukin-6/metabolism , Lipopolysaccharides/metabolism , Male , Models, Biological , Molecular Sequence Data , Rats , Recombinant Proteins/chemistry , Resistin , Sequence Homology, Amino Acid , Signal Transduction , Tissue Distribution
17.
Trends Endocrinol Metab ; 15(6): 247-9, 2004 Aug.
Article in English | MEDLINE | ID: mdl-15358276

ABSTRACT

The role of adipocyte-secreted resistin/adipocyte-specific secretory factor (ADSF)/FIZZ3 in obesity and diabetes has been controversial at best. Recently generated resn knockout mice showed normal glucose and insulin sensitivity with lower fasting glucose levels. Upon feeding with a high-fat diet, the knockout mice exhibited increased glucose tolerance with decreased hepatic glucose output, possibly due to phosphorylation and activation of AMP-activated protein kinase and suppression of gluconeogenic genes. In comparison, transgenic mice overexpressing a dominant negative form of resistin/ADSF/FIZZ3 showed increased adiposity with elevated leptin and adiponectin levels, accompanying enhanced glucose tolerance and insulin sensitivity both on chow and high-fat diets. Although its underlying mechanisms need further elucidation, the in vivo studies demonstrate a role of resistin/ADSF/FIZZ3 in obesity and insulin resistance.


Subject(s)
Aldosterone/metabolism , Diabetes Mellitus/metabolism , Energy Metabolism/physiology , Hormones, Ectopic/physiology , Insulin Resistance/physiology , Obesity/metabolism , Proteins/metabolism , Animals , Diabetes Mellitus/genetics , Hormones, Ectopic/genetics , Insulin Resistance/genetics , Mice , Mice, Knockout , Mice, Transgenic , Obesity/genetics , Resistin
18.
Clin Chem ; 50(9): 1511-25, 2004 Sep.
Article in English | MEDLINE | ID: mdl-15265818

ABSTRACT

BACKGROUND: Recent studies point to the adipose tissue as a highly active endocrine organ secreting a range of hormones. Leptin, ghrelin, adiponectin, and resistin are considered to take part in the regulation of energy metabolism. APPROACH: This review summarizes recent knowledge on leptin and its receptor and on ghrelin, adiponectin, and resistin, and emphasizes their roles in pathobiochemistry and clinical chemistry. CONTENT: Leptin, adiponectin, and resistin are produced by the adipose tissue. The protein leptin, a satiety hormone, regulates appetite and energy balance of the body. Adiponectin could suppress the development of atherosclerosis and liver fibrosis and might play a role as an antiinflammatory hormone. Increased resistin concentrations might cause insulin resistance and thus could link obesity with type II diabetes. Ghrelin is produced in the stomach. In addition to its role in long-term regulation of energy metabolism, it is involved in the short-term regulation of feeding. These hormones have important roles in energy homeostasis, glucose and lipid metabolism, reproduction, cardiovascular function, and immunity. They directly influence other organ systems, including the brain, liver, and skeletal muscle, and are significantly regulated by nutritional status. This newly discovered secretory function has extended the biological relevance of adipose tissue, which is no longer considered as only an energy storage site. SUMMARY: The functional roles, structures, synthesis, analytical aspects, and clinical significance of leptin, ghrelin, adiponectin, and resistin are summarized.


Subject(s)
Energy Metabolism/physiology , Hormones, Ectopic/physiology , Intercellular Signaling Peptides and Proteins , Leptin/physiology , Peptide Hormones/physiology , Proteins/physiology , Adiponectin , Adipose Tissue/metabolism , Animals , Gastric Mucosa/metabolism , Ghrelin , Hormones, Ectopic/blood , Hormones, Ectopic/chemistry , Hormones, Ectopic/metabolism , Humans , Leptin/blood , Leptin/chemistry , Leptin/metabolism , Liver/metabolism , Muscle, Skeletal/metabolism , Obesity/metabolism , Peptide Hormones/blood , Peptide Hormones/chemistry , Peptide Hormones/metabolism , Proteins/chemistry , Proteins/metabolism , Receptors, Cell Surface/blood , Receptors, Cell Surface/chemistry , Receptors, Cell Surface/metabolism , Receptors, Leptin , Resistin
19.
Proc Natl Acad Sci U S A ; 101(17): 6780-5, 2004 Apr 27.
Article in English | MEDLINE | ID: mdl-15090646

ABSTRACT

Adipocyte-specific secretory factor (ADSF)/resistin is a small cysteine-rich protein secreted from adipose tissue that belongs to a gene family found in inflammatory zone (FIZZ) or found in resistin-like molecule (RELM). ADSF has been implicated in modulating adipogenesis and insulin resistance. To examine the long-term function of ADSF in adipogenesis and glucose homeostasis, we constructed an expression vector for a dominant inhibitory form of ADSF by fusing it to the human IgGgamma constant region (hFc). ADSF-hFc not only homodimerizes but heterooligomerizes with ADSF/resistin and prevents ADSF/resistin inhibition of adipocyte differentiation of 3T3-L1 cells in a dominant negative manner. Transgenic mice overexpressing ADSF-hFc in adipose tissue show increased adiposity with elevated expression of adipocyte markers as well as enlarged adipocyte size. This finding clearly demonstrates in vivo the inhibitory role of ADSF in adipogenesis. ADSF-hFc transgenic mice with impaired ADSF function exhibit improved glucose tolerance and insulin sensitivity either on chow or high-fat diets. Because of the enhanced adipocyte differentiation, the ADSF-hFc transgenic mice show increased expression of leptin and adiponectin in adipose tissue. The elevated circulating levels for these adipocyte-derived hormones with decreased plasma triglyceride and free fatty acid levels may account for the improved glucose and insulin tolerance in these transgenic mice.


Subject(s)
Adipocytes/cytology , Cell Division/physiology , Hormones, Ectopic/physiology , 3T3-L1 Cells , Animals , Blotting, Northern , Blotting, Western , Cell Differentiation/physiology , Glucose Tolerance Test , Insulin Resistance , Mice , Mice, Transgenic , Resistin , Reverse Transcriptase Polymerase Chain Reaction
SELECTION OF CITATIONS
SEARCH DETAIL
...