Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
J Biol Chem ; 300(1): 105566, 2024 Jan.
Article in English | MEDLINE | ID: mdl-38103643

ABSTRACT

Macrophages play critical roles in inflammation and tissue homeostasis, and their functions are regulated by various autocrine, paracrine, and endocrine factors. We have previously shown that CTRP6, a secreted protein of the C1q family, targets both adipocytes and macrophages to promote obesity-linked inflammation. However, the gene programs and signaling pathways directly regulated by CTRP6 in macrophages remain unknown. Here, we combine transcriptomic and phosphoproteomic analyses to show that CTRP6 activates inflammatory gene programs and signaling pathways in mouse bone marrow-derived macrophages (BMDMs). Treatment of BMDMs with CTRP6 upregulated proinflammatory, and suppressed the antiinflammatory, gene expression. We also showed that CTRP6 activates p44/42-MAPK, p38-MAPK, and NF-κB signaling pathways to promote inflammatory cytokine secretion from BMDMs, and that pharmacologic inhibition of these signaling pathways markedly attenuated the effects of CTRP6. Pretreatment of BMDMs with CTRP6 also sensitized and potentiated the BMDMs response to lipopolysaccharide (LPS)-induced inflammatory signaling and cytokine secretion. Consistent with the metabolic phenotype of proinflammatory macrophages, CTRP6 treatment induced a shift toward aerobic glycolysis and lactate production, reduced oxidative metabolism, and elevated mitochondrial reactive oxygen species production in BMDMs. Importantly, in accordance with our in vitro findings, BMDMs from CTRP6-deficient mice were less inflammatory at baseline and showed a marked suppression of LPS-induced inflammatory gene expression and cytokine secretion. Finally, loss of CTRP6 in mice also dampened LPS-induced inflammation and hypothermia. Collectively, our findings suggest that CTRP6 regulates and primes the macrophage response to inflammatory stimuli and thus may have a role in modulating tissue inflammatory tone in different physiological and disease contexts.


Subject(s)
Adipokines , Gene Expression Profiling , Inflammation , Lipopolysaccharides , Macrophages , Phosphoproteins , Proteomics , Animals , Mice , Adipokines/deficiency , Adipokines/genetics , Adipokines/metabolism , Bone Marrow Cells/cytology , Cytokines/metabolism , Glycolysis , Hypothermia/complications , Inflammation/complications , Inflammation/genetics , Inflammation/immunology , Inflammation/metabolism , Lactic Acid/biosynthesis , Lipopolysaccharides/immunology , Macrophages/cytology , Macrophages/immunology , Macrophages/metabolism , Mice, Inbred C57BL , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , NF-kappa B/metabolism , Phosphoproteins/analysis , Phosphoproteins/metabolism , Signal Transduction , Reactive Oxygen Species/metabolism
2.
Mol Biol Rep ; 49(2): 1617-1622, 2022 Feb.
Article in English | MEDLINE | ID: mdl-34811637

ABSTRACT

BACKGROUND: Adipose tissue secretes various adipocytokines that play important roles in lipid and glucose metabolism. C1q and tumor necrosis factor-related protein 3 (CTRP3) is a paralog of adiponectin, which has been extensively studied. Previously, we showed that epididymal white adipose tissue size is decreased in high fat diet-fed Ctrp3 knockout (KO) mice. Here, I examined metabolic roles of CTRP3 in non-obese mice under starvation conditions. METHODS AND RESULTS: Serum alanine aminotransferase (ALT) and aspartate aminotransferase (AST) levels were increased in 20-h-fasted standard chow-fed Ctrp3 KO mice compared with wild-type (WT) controls. RT-qPCR analysis revealed that ALT1, AST2, and glucose-6-phosphatase mRNA expressions were increased in the liver of Ctrp3 KO mice after a 20-h fast. Upon intraperitoneal alanine administration, Ctrp3 KO mice showed a modest but significant increase in the conversion of alanine to glucose. To characterize hepatic metabolism in fasted Ctrp3 KO mice, I further analyzed metabolomic profiles in the liver. Unexpectedly, metabolome analysis of the liver of 20-h-fasted Ctrp3 KO mice revealed that the relative concentrations of 10 of the 20 amino acids were lower than in WT controls. The relative concentrations of ornithine and argininosuccinate, which are urea cycle intermediates, were also decreased in the Ctrp3 KO liver. CONCLUSIONS: Taken together, my results indicate that CTRP3 has novel roles in regulating both gluconeogenesis and amino acid metabolism in the liver during starvation.


Subject(s)
Adipokines/genetics , Adipokines/metabolism , Gluconeogenesis/genetics , Adipokines/deficiency , Adiponectin/metabolism , Adipose Tissue/metabolism , Adipose Tissue, White/metabolism , Alanine Transaminase/metabolism , Amino Acids/metabolism , Animals , Diet, High-Fat , Gluconeogenesis/physiology , Glucose/metabolism , Lipid Metabolism/genetics , Liver/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout
3.
Cytokine ; 148: 155663, 2021 12.
Article in English | MEDLINE | ID: mdl-34388476

ABSTRACT

BACKGROUND AND AIM: CAMP (Cathelicidin antimicrobial peptide) expression in adipocytes is regulated by Toll-like receptor (TLR) agonists. Secreted adipokines such as CTRP-3 have been suggested to participate in innate immune signaling in adipose tissue (AT). This study investigates whether TLR-induced CAMP expression in adipocytes is antagonized by CTRP-3. METHODS: 3T3-L1 adipocytes were co-stimulated with TLR agonists (LPS, MALP-2, Pam3CSK4, pI:C) and recombinant CTRP-3. In a SIRS model, C57BL/6 wild-type mice were intraperitoneally (ip) injected with recombinant CTRP-3 prior to LPS. CAMP expression was analyzed by real-time PCR in AT of wild-type mice and in AT and primary adipocytes from transgenic mice lacking adipocyte CTRP-3 expression. Comparative transcriptome analysis by RNA seq. was applied in CTRP-3 KO adipocytes. RESULTS: In vitro, CTRP-3 antagonized TLR4- and TLR1/2-induced CAMP expression in adipocytes whereas TLR3- and TLR2/6-mediated induction of CAMP was not affected. in vivo, application of exogenous CTRP-3 dose-dependently antagonized LPS-induced CAMP expression in intra-abdominal AT. CAMP expression in total AT and in primary adipocytes of subcutaneous and intra-abdominal AT did not differ between wild-type mice and transgenic mice lacking adipocyte CTRP-3 expression. CONCLUSIONS: The study suggests a hypothetical role of CAMP in host defense not only against Gram-positive bacteria sensed by TLR1/2 and TLR2/6 but also against Gram-negative bacteria sensed by TLR4 and potentially against viruses sensed by TLR3. The machinery of TLR-mediated pro-inflammatory activation of the CAMP gene in adipocytes seems to be partly modulated by secreted adipokines belonging to the growing family of C1q/TNF-related proteins such as CTRP-3.


Subject(s)
Adipocytes/metabolism , Adipokines/metabolism , Antimicrobial Peptides/genetics , Gene Expression Regulation , Toll-Like Receptors/metabolism , 3T3-L1 Cells , Adipokines/deficiency , Adipose Tissue/metabolism , Animals , Antimicrobial Peptides/metabolism , Disease Models, Animal , Lipopolysaccharides , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Models, Biological , Systemic Inflammatory Response Syndrome/genetics , Transcriptome/genetics
4.
Am J Physiol Regul Integr Comp Physiol ; 320(1): R19-R35, 2021 01 01.
Article in English | MEDLINE | ID: mdl-33085906

ABSTRACT

C1q/TNF-related protein 1 (CTRP1) is an endocrine factor with metabolic, cardiovascular, and renal functions. We previously showed that aged Ctrp1-knockout (KO) mice fed a control low-fat diet develop renal hypertrophy and dysfunction. Since aging and obesity adversely affect various organ systems, we hypothesized that aging, in combination with obesity induced by chronic high-fat feeding, would further exacerbate renal dysfunction in CTRP1-deficient animals. To test this, we fed wild-type and Ctrp1-KO mice a high-fat diet for 8 mo or longer. Contrary to our expectation, no differences were observed in blood pressure, heart function, or vascular stiffness between genotypes. Loss of CTRP1, however, resulted in an approximately twofold renal enlargement (relative to body weight), ∼60% increase in urinary total protein content, and elevated pH, and changes in renal gene expression affecting metabolism, signaling, transcription, cell adhesion, solute and metabolite transport, and inflammation. Assessment of glomerular integrity, the extent of podocyte foot process effacement, as well as renal response to water restriction and salt loading did not reveal significant differences between genotypes. Interestingly, blood platelet, white blood cell, neutrophil, lymphocyte, and eosinophil counts were significantly elevated, whereas mean corpuscular volume and hemoglobin were reduced in Ctrp1-KO mice. Cytokine profiling revealed increased circulating levels of CCL17 and TIMP-1 in KO mice. Compared with our previous study, current data suggest that chronic high-fat feeding affects renal phenotypes differently than similarly aged mice fed a control low-fat diet, highlighting a diet-dependent contribution of CTRP1 deficiency to age-related changes in renal structure and function.


Subject(s)
Adipokines/deficiency , Aging/metabolism , Diet, High-Fat/adverse effects , Kidney Diseases/etiology , Kidney/metabolism , Obesity/etiology , Adipokines/genetics , Age Factors , Aging/genetics , Aging/pathology , Animals , Chemokine CCL17/blood , Female , Gene Expression Regulation , Genotype , Hypertrophy , Kidney/ultrastructure , Kidney Diseases/genetics , Kidney Diseases/metabolism , Kidney Diseases/pathology , Male , Mice, Inbred C57BL , Mice, Knockout , Phenotype , Tissue Inhibitor of Metalloproteinase-1/blood
5.
PLoS One ; 15(12): e0243483, 2020.
Article in English | MEDLINE | ID: mdl-33275602

ABSTRACT

BACKGROUND: Myocardial infarction (MI) is a leading cause of death worldwide. We previously identified adipolin, also known as C1q/Tnf-related protein 12, as an anti-inflammatory adipokine with protective features against metabolic and vascular disorders. Here, we investigated the effect of adipolin on myocardial remodeling in a mouse model of MI. METHODS: Male adipolin-knockout (APL-KO) and wild-type (WT) mice were subjected to the permanent ligation of the left anterior descending coronary artery to create MI. RESULTS: APL-KO mice exhibited increased ratios of heart weight/body weight and lung weight/body weight after MI compared with WT mice. APL-KO mice showed increased left ventricular diastolic diameter and decreased fractional shortening after MI compared with WT mice. APL-KO mice exhibited increased expression of pro-inflammatory mediators and enhanced cardiomyocyte apoptosis in the post-MI hearts compared with WT mice. Systemic administration of adenoviral vectors expressing adipolin to WT mice after MI surgery improved left ventricular contractile dysfunction and reduced cardiac expression of pro-inflammatory genes. Treatment of cultured cardiomyocytes with adipolin protein reduced lipopolysaccharide-induced expression of pro-inflammatory mediators and hypoxia-induced apoptosis. Treatment with adipolin protein increased Akt phosphorylation in cardiomyocytes. Inhibition of PI3 kinase/Akt signaling reversed the anti-inflammatory and anti-apoptotic effects of adipolin in cardiomyocytes. CONCLUSION: Our data indicate that adipolin ameliorates pathological remodeling of myocardium after MI, at least in part, by its ability to reduce myocardial inflammatory response and apoptosis.


Subject(s)
Adipokines/metabolism , Myocardial Infarction/pathology , Ventricular Remodeling , Adipokines/deficiency , Adipokines/genetics , Animals , Apoptosis/drug effects , Disease Models, Animal , Inflammation Mediators/metabolism , Lipopolysaccharides/pharmacology , Mice , Mice, Inbred C57BL , Mice, Knockout , Myocardial Infarction/metabolism , Myocardium/metabolism , Myocardium/pathology , Myocytes, Cardiac/cytology , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Phosphorylation/drug effects , Proto-Oncogene Proteins c-akt/metabolism , Rats , Recombinant Proteins/biosynthesis , Recombinant Proteins/pharmacology , Signal Transduction/drug effects , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism
6.
Mol Biol Rep ; 47(11): 9219-9224, 2020 Nov.
Article in English | MEDLINE | ID: mdl-33057992

ABSTRACT

Adipokines are important regulators of lipid and glucose metabolism. A family of adiponectin paralogs is known as C1q and tumor necrosis factor (TNF)-related proteins (CTRPs). One line of Ctrp3-deficient mice shows reduced liver size in response to obesity. We generated and characterized another line of Ctrp3 knockout (KO) mice to reveal novel physiological functions of CTRP3. Interestingly, high fat diet (HFD)-fed Ctrp3 KO mice displayed a decrease in the epididymal white adipose tissue (WAT) weight to total body weight ratio. Histologically, adipocyte size was significantly smaller in the epididymal WAT of HFD-fed Ctrp3 KO mice than wild-type (WT) controls. The expression of several genes involved in lipogenesis, lipolysis and adipogenesis in the epididymal WAT of Ctrp3 KO mice fed a HFD was decreased. The present findings provide new insight into the role of CTRP3 as adipokine in the regulation of adipose tissue in obesity.


Subject(s)
Adipokines/genetics , Adipose Tissue, White/metabolism , Gene Expression , Lipogenesis/genetics , Obesity/genetics , Adipogenesis/genetics , Adipokines/deficiency , Adipose Tissue, White/growth & development , Age Factors , Animals , Diet, High-Fat/adverse effects , Lipolysis/genetics , Male , Mice, Inbred C57BL , Mice, Knockout , Obesity/etiology , Obesity/metabolism , Organ Size/genetics , Weight Gain/genetics
7.
Article in English | MEDLINE | ID: mdl-32360289

ABSTRACT

The regulation of porcine subcutaneous (SC) and intramuscular (IM) fat deposition significantly affects pork quality and the lean meat percentage of the carcass, respectively. The adipokine C1q/tumor necrosis factor-related protein 6 (CTRP6), plays a significant role in regulating animal fat deposition. The purpose of this study was to understand the effects of CTRP6 gene knockdown in IM and SC adipocytes by RNA-seq analysis. A total of 1830 and 2936 differentially expressed genes (DEGs) were identified in SC and IM adipocytes, respectively. 844 were down- and 2092 were upregulated in SC adipocytes, while 648 were down- and 1182 were upregulated in IM adipocytes. Furthermore, 1778 DEGs were detected only in SC adipocytes, 672 DEGs only in IM adipocytes, and 1158 DEGs in both types of adipocytes. GO analysis indicated that DEGs involved in adipocyte differentiation were significantly enriched in both SC and IM adipocytes following treatment with CTRP6-siRNA. Moreover, KEGG pathway enrichment analysis revealed differences of metabolic regulation between IM and SC adipocytes. With CTRP6-silencing, the signaling pathways related to Ras and arachidonic acid metabolism were significantly enriched in IM adipocytes, while four other signaling pathways, encompassing the TNF, MAPK, p53 and adipokine pathway were specifically enriched in SC adipocytes. Interestingly, the effect of CTRP6-siRNA treatment was attenuated by the specific Ras activator ML-097 in IM adipocytes, while the specific p53 activator SJ-172550 had the corresponding effect in SC adipocytes. Altogether, we suggest that CTRP6 may be a differential regulator of the development and metabolism of IM and SC adipose tissues.


Subject(s)
Adipokines/metabolism , Subcutaneous Fat/metabolism , Adipocytes/metabolism , Adipokines/deficiency , Adipokines/genetics , Animals , Male , Signal Transduction , Swine
8.
FASEB J ; 34(2): 2657-2676, 2020 02.
Article in English | MEDLINE | ID: mdl-31908037

ABSTRACT

Local and systemic factors that influence renal structure and function in aging are not well understood. The secretory protein C1q/TNF-related protein 1 (CTRP1) regulates systemic metabolism and cardiovascular function. We provide evidence here that CTRP1 also modulates renal physiology in an age- and sex-dependent manner. In mice lacking CTRP1, we observed significantly increased kidney weight and glomerular hypertrophy in aged male but not female or young mice. Although glomerular filtration rate, plasma renin and aldosterone levels, and renal response to water restriction did not differ between genotypes, CTRP1-deficient male mice had elevated blood pressure. Echocardiogram and pulse wave velocity measurements indicated normal heart function and vascular stiffness in CTRP1-deficient animals, and increased blood pressure was not due to greater salt retention. Paradoxically, CTRP1-deficient mice had elevated urinary sodium and potassium excretion, partially resulting from reduced expression of genes involved in renal sodium and potassium reabsorption. Despite renal hypertrophy, markers of inflammation, fibrosis, and oxidative stress were reduced in CTRP1-deficient mice. RNA sequencing revealed alterations and enrichments of genes in metabolic processes in CTRP1-deficient animals. These results highlight novel contributions of CTRP1 to aging-associated changes in renal physiology.


Subject(s)
Adipokines/deficiency , Hypertension/metabolism , Hypertrophy/metabolism , Kidney/metabolism , Adipokines/metabolism , Animals , Blood Pressure/physiology , Hypertension/physiopathology , Hypertrophy/physiopathology , Inflammation/metabolism , Inflammation/physiopathology , Mice, Knockout , Signal Transduction/physiology
9.
Cardiovasc Res ; 116(1): 237-249, 2020 01 01.
Article in English | MEDLINE | ID: mdl-30874788

ABSTRACT

AIMS: Secreted factors produced by adipose tissue are involved in the pathogenesis of cardiovascular disease. We previously identified adipolin, also known as C1q/TNF-related protein 12, as an insulin-sensitizing adipokine. However, the role of adipolin in vascular disease remains unknown. Here, we investigated whether adipolin modulates pathological vascular remodelling. METHODS AND RESULTS: Adipolin-knockout (APL-KO) and wild-type (WT) mice were subjected to wire-induced injury of the femoral artery. APL-KO mice showed increased neointimal thickening after vascular injury compared with WT mice, which was accompanied by an enhanced inflammatory response and vascular cell proliferation in injured arteries. Adipolin deficiency also led to a reduction in transforming growth factor-ß (TGF-ß) 1 protein levels in injured arteries. Treatment of cultured macrophages with adipolin protein led to a reduction in lipopolysaccharide-stimulated expression of inflammatory mediators, including tumour necrosis factor (TNF)-α, interleukin (IL) 6, and monocyte chemotactic protein (MCP)-1. These effects were reversed by inhibition of TGF-ß receptor II (TGF-ßRII)/Smad2 signalling. Adipolin also reduced platelet-derived growth factor (PDGF)-BB-stimulated proliferation of vascular smooth muscle cells (VSMCs) through a TGF-ßRII/Smad2-dependent pathway. Furthermore, adipolin treatment significantly increased TGF-ß1 concentration in media from cultured VSMCs and macrophages. CONCLUSION: These data indicate that adipolin protects against the development of pathological vascular remodelling by attenuating macrophage inflammatory responses and VSMC proliferation.


Subject(s)
Adipokines/metabolism , Cell Proliferation , Inflammation Mediators/metabolism , Macrophages, Peritoneal/metabolism , Muscle, Smooth, Vascular/metabolism , Myocytes, Smooth Muscle/metabolism , Vascular Remodeling , Vascular System Injuries/metabolism , Adipokines/deficiency , Adipokines/genetics , Animals , Disease Models, Animal , Femoral Artery/injuries , Femoral Artery/metabolism , Femoral Artery/pathology , Femoral Artery/physiopathology , HEK293 Cells , Human Umbilical Vein Endothelial Cells/metabolism , Human Umbilical Vein Endothelial Cells/pathology , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , Muscle, Smooth, Vascular/injuries , Muscle, Smooth, Vascular/pathology , Muscle, Smooth, Vascular/physiopathology , Myocytes, Smooth Muscle/pathology , Neointima , Phosphorylation , RAW 264.7 Cells , Receptor, Transforming Growth Factor-beta Type II/genetics , Receptor, Transforming Growth Factor-beta Type II/metabolism , Signal Transduction , Smad2 Protein/genetics , Smad2 Protein/metabolism , Vascular System Injuries/genetics , Vascular System Injuries/pathology , Vascular System Injuries/physiopathology
10.
Skinmed ; 17(4): 248-253, 2019.
Article in English | MEDLINE | ID: mdl-31627786

ABSTRACT

Zinc-α2-glycoprotein (ZAG), a recently identified adipokine, is a multidisciplinary protein, which is secreted in various body fluids. The ZAG plays roles in lipolysis, regulation of metabolism, cell proliferation and differentiation, regulation of melanin synthesis, cell adhesion, and immunoregulation. The aim of this study is to estimate serum and tissue levels of ZAG in patients with vitiligo. The study included 30 vitiligo patients and 30 healthy controls. Lesional skin biopsy was performed, and blood sample was retrieved to determine the level of ZAG in blood using ELISA kit. In this study, the mean level of ZAG was found to be significantly lower in the vitiligo patients' tissue in comparison with the healthy control subjects' tissue ( p=0.001); the level of ZAG was also lower in vitiligo patients' serum in comparison with the healthy control subjects' serum ( p=0.001). A highly significant correlation was observed between the duration of the disease and the level of ZAG in the tissue of patients (r =0.9; p=0.001). Also a highly significant positive correlation was observed between the age of patients and the level of ZAG in the tissue (r =0.5; p=0.006). Diminishing of ZAG in serum and tissue of vitiligo patients is another important player sharing in the complex pathogenesis of vitiligo.


Subject(s)
Adipokines/deficiency , Seminal Plasma Proteins/analysis , Seminal Plasma Proteins/physiology , Vitiligo/etiology , Vitiligo/metabolism , Adolescent , Adult , Child , Female , Humans , Male , Middle Aged , Seminal Plasma Proteins/blood , Skin/chemistry , Vitiligo/blood , Young Adult , Zn-Alpha-2-Glycoprotein
11.
J Biol Chem ; 292(36): 14836-14850, 2017 09 08.
Article in English | MEDLINE | ID: mdl-28726640

ABSTRACT

Obesity is associated with chronic low-grade inflammation, and metabolic regulators linking obesity to inflammation have therefore received much attention. Secreted C1q/TNF-related proteins (CTRPs) are one such group of regulators that regulate glucose and fat metabolism in peripheral tissues and modulate inflammation in adipose tissue. We have previously shown that expression of CTRP6 is up-regulated in leptin-deficient mice and, conversely, down-regulated by the anti-diabetic drug rosiglitazone. Here, we provide evidence for a novel role of CTRP6 in modulating both inflammation and insulin sensitivity. We found that in obese and diabetic humans and mouse models, CTRP6 expression was markedly up-regulated in adipose tissue and that stromal vascular cells, such as macrophages, are a major CTRP6 source. Overexpressing mouse or human CTRP6 impaired glucose disposal in peripheral tissues in response to glucose and insulin challenge in wild-type mice. Conversely, Ctrp6 gene deletion improved insulin action and increased metabolic rate and energy expenditure in diet-induced obese mice. Mechanistically, CTRP6 regulates local inflammation and glucose metabolism by targeting macrophages and adipocytes, respectively. In cultured macrophages, recombinant CTRP6 dose-dependently up-regulated the expression and production of TNF-α. Conversely, CTRP6 deficiency reduced circulating inflammatory cytokines and pro-inflammatory macrophages in adipose tissue. CTRP6-overexpressing mice or CTRP6-treated adipocytes had reduced insulin-stimulated Akt phosphorylation and glucose uptake. In contrast, loss of CTRP6 enhanced insulin-stimulated Akt activation in adipose tissue. Together, these results establish CTRP6 as a novel metabolic/immune regulator linking obesity to adipose tissue inflammation and insulin resistance.


Subject(s)
Adipokines/metabolism , Adipose Tissue/metabolism , Collagen/metabolism , Inflammation/metabolism , Insulin Resistance , Obesity/metabolism , 3T3-L1 Cells , Adipokines/deficiency , Animals , Cells, Cultured , Disease Models, Animal , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Obese , RAW 264.7 Cells
12.
Atherosclerosis ; 257: 138-145, 2017 02.
Article in English | MEDLINE | ID: mdl-28131048

ABSTRACT

BACKGROUND AND AIMS: Obesity contributes to the progression of vascular disorders. C1q/TNF-related protein (CTRP) 1 is a circulating adipokine, which is upregulated in obese complications including coronary artery disease. Here, we investigated the role of CTRP1 in regulation of vascular remodeling after mechanical injury and evaluated its potential mechanism. METHODS: Mice were subjected to wire-induced injury of left femoral arteries. An adenoviral vector encoding CTRP1 (Ad-CTRP1) or ß-galactosidase as a control was injected into the jugular vein of mice 3 days prior to surgery. RESULTS: Systemic administration of Ad-CTRP1 to wild-type mice led to reduction of the neointimal thickening after wire-induced arterial injury and the number of bromodeoxyuridine-positive cells in injured vessels as compared with treatment with control vectors. Treatment of vascular smooth muscle cells (VSMCs) with CTRP1 protein attenuated proliferative activity and ERK phosphorylation in response to PDGF-BB. CTRP1 treatment increased cyclic AMP (cAMP) levels in VSMCs, and inhibition of adenylyl cyclase reversed the inhibitory effect of CTRP1 on VSMC growth and ERK phosphorylation. Antagonization of sphingosine-1-phosphaterote (S1P) receptor 2 blocked the effects of CTRP1 on cAMP production and VSMC growth. Furthermore, CTRP1-knockout mice had enhanced neointimal thickening following injury and increased numbers of proliferating cells in neointima compared to control WT mice. CONCLUSIONS: These findings indicate that CTRP1 functions to prevent the development of pathological vascular remodeling by reducing VSMC growth through the cAMP-dependent pathway.


Subject(s)
Adipokines/metabolism , Cell Proliferation , Muscle, Smooth, Vascular/metabolism , Myocytes, Smooth Muscle/metabolism , Neointima , Vascular System Injuries/metabolism , Adipokines/deficiency , Adipokines/genetics , Animals , Cells, Cultured , Cyclic AMP/metabolism , Disease Models, Animal , Extracellular Signal-Regulated MAP Kinases/metabolism , Femoral Artery/injuries , Femoral Artery/metabolism , Femoral Artery/pathology , Genetic Predisposition to Disease , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Male , Mice, Inbred C57BL , Mice, Knockout , Muscle, Smooth, Vascular/injuries , Muscle, Smooth, Vascular/pathology , Myocytes, Smooth Muscle/pathology , Phenotype , Phosphorylation , Proteins/metabolism , Receptors, Lysosphingolipid/metabolism , Signal Transduction , Vascular System Injuries/genetics , Vascular System Injuries/pathology
13.
Am J Physiol Endocrinol Metab ; 311(4): E678-E697, 2016 10 01.
Article in English | MEDLINE | ID: mdl-27555298

ABSTRACT

C1q/TNF-related protein 1 (CTRP1) is a conserved plasma protein of the C1q family with notable metabolic and cardiovascular functions. We have previously shown that CTRP1 infusion lowers blood glucose and that transgenic mice with elevated circulating CTRP1 are protected from diet-induced obesity and insulin resistance. Here, we used a genetic loss-of-function mouse model to address the requirement of CTRP1 for metabolic homeostasis. Despite similar body weight, food intake, and energy expenditure, Ctrp1 knockout (KO) mice fed a low-fat diet developed insulin resistance and hepatic steatosis. Impaired glucose metabolism in Ctrp1 KO mice was associated with increased hepatic gluconeogenic gene expression and decreased skeletal muscle glucose transporter glucose transporter 4 levels and AMP-activated protein kinase activation. Loss of CTRP1 enhanced the clearance of orally administered lipids but did not affect intestinal lipid absorption, hepatic VLDL-triglyceride export, or lipoprotein lipase activity. In contrast to triglycerides, hepatic cholesterol levels were reduced in Ctrp1 KO mice, paralleling the reduced expression of cholesterol synthesis genes. Contrary to expectations, when challenged with a high-fat diet to induce obesity, Ctrp1 KO mice had increased physical activity and reduced body weight, adiposity, and expression of lipid synthesis and fibrotic genes in adipose tissue; these phenotypes were linked to elevated FGF-21 levels. Due in part to increased hepatic AMP-activated protein kinase activation and reduced expression of lipid synthesis genes, Ctrp1 KO mice fed a high-fat diet also had reduced liver and serum triglyceride and cholesterol levels. Taken together, these results provide genetic evidence to establish the significance of CTRP1 to systemic energy metabolism in different metabolic and dietary contexts.


Subject(s)
Adipokines/deficiency , Adipokines/genetics , Glucose/metabolism , Homeostasis , Lipid Metabolism/genetics , AMP-Activated Protein Kinases/metabolism , Animals , Blood Glucose/metabolism , Body Weight/genetics , Cholesterol/blood , Diet, High-Fat , Eating , Energy Metabolism/genetics , Gluconeogenesis/genetics , Glucose Transporter Type 4/metabolism , Homeostasis/genetics , Liver/metabolism , Mice, Inbred C57BL , Mice, Knockout , Obesity/genetics , Triglycerides/blood
14.
J Pharm Pharmacol ; 61(11): 1493-8, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19903374

ABSTRACT

OBJECTIVES: It has been recently reported that blockade of type 1 cannabinoid (CB1) receptors by specific antagonists or genetic manipulation alleviates dyslipidaemia, hyperglycaemia and insulin resistance in animal models of obesity and type 2 diabetes. However, the precise role of adipokines in the insulin-sensitising effects of the CB1 antagonist rimonabant is not clear. METHODS: ob/ob mice were treated with different doses of rimonabant and then subjected to an oral glucose tolerance test. The expression of different adipokines in white adipose tissue was analysed by quantitative real-time PCR. KEY FINDINGS: Rimonabant (30 mg/kg) significantly inhibited body weight and fat pad weight gain (P < 0.05) and improved glucose tolerance. Gene expression analysis indicated that tumour necrosis factor-alpha, visfatin and retinol binding protein-4 were downregulated in the adipose tissue of ob/ob mice treated with rimonabant compared with controls, whereas adiponectin was significantly upregulated. CONCLUSIONS: Rimonabant-mediated alteration of adipokines in white adipose tissues may play a role in improving insulin sensitivity in obese animals.


Subject(s)
Adipokines/physiology , Adipose Tissue, White/drug effects , Anti-Obesity Agents/therapeutic use , Glucose Intolerance/drug therapy , Insulin Resistance , Obesity/drug therapy , Piperidines/therapeutic use , Pyrazoles/therapeutic use , Adipokines/blood , Adipokines/deficiency , Adiponectin/blood , Adipose Tissue/drug effects , Adipose Tissue, White/metabolism , Animals , Anti-Obesity Agents/pharmacology , Blood Glucose/metabolism , Body Weight/drug effects , Down-Regulation , Female , Gene Expression , Glucose Intolerance/metabolism , Glucose Tolerance Test , Insulin/blood , Leptin/deficiency , Mice , Mice, Knockout , Models, Animal , Nicotinamide Phosphoribosyltransferase/genetics , Nicotinamide Phosphoribosyltransferase/metabolism , Organ Size/drug effects , Piperidines/pharmacology , Pyrazoles/pharmacology , RNA, Messenger/metabolism , Receptor, Cannabinoid, CB1/antagonists & inhibitors , Retinol-Binding Proteins, Plasma/genetics , Retinol-Binding Proteins, Plasma/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Rimonabant , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...