Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 30
Filter
1.
Pharm Biol ; 61(1): 541-555, 2023 Dec.
Article in English | MEDLINE | ID: mdl-36994890

ABSTRACT

CONTEXT: Gouty arthritis (GA) is a characteristically inflammatory disease often associated with lipid metabolism disorder. Huangqin Qingrechubi capsule (HQC) has been used for the treatment of GA. OBJECTIVE: To explore the mechanism of HQC in the treatment of GA. MATERIALS AND METHODS: A total of 30 GA patients (GA group) and 30 healthy subjects [normal control (NC) group] were recruited. The GA group was treated with HQC (3.6 g/d) for 10 days. Lipid metabolism and inflammation indexes were detected. Five herbal names of HQC, or 'gouty arthritis', 'hyperlipidemia' and 'inflammation' were used as key words to search related databases for network pharmacological analysis. Subsequently, GA-fibroblast-like synoviocytes (FLSs) were stimulated with GA-peripheral blood mononuclear cells (PBMCs) (3:1) and treated with HQC drug-containing serum (20%). RT-qPCR, Western blot, and ELISA were conducted to further explore the mechanism of HQC in improving GA. RESULTS: In clinical observation, HQC decreased the expression of lncRNA H19 and IL-1ß, and increased the expression of adiponectin (APN) and IL-4 in the GA group (about half). Through network pharmacology, the PI3K/AKT signaling pathway was identified. Cell experiments showed that HQC treatment reduced the viability of GA-FLSs (49.61%), up-regulated the expression of IL-4 (155.18%), IL-10 (165.13%), and APN (31.24%), and down-regulated the expression of lncRNA H19 (33.70%), IL-1ß (64.70%), TNF-α (78.32%), p-PI3K (48.80%), and p-AKT (53.48%). DISCUSSION AND CONCLUSIONS: HQC improved lipid metabolism disorder and inflammatory response of GA by regulating the lncRNA H19/APN/PI3K/AKT. Maintaining the stability of lipid metabolism may be an effective way to alleviate GA.


Subject(s)
Arthritis, Gouty , Lipid Metabolism Disorders , RNA, Long Noncoding , Humans , Proto-Oncogene Proteins c-akt/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Scutellaria baicalensis , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , Adiponectin/metabolism , Adiponectin/pharmacology , Lipid Metabolism , Leukocytes, Mononuclear/metabolism , Interleukin-4/pharmacology , Signal Transduction , Inflammation/drug therapy , Arthritis, Gouty/drug therapy
2.
Biomed Res Int ; 2022: 2369650, 2022.
Article in English | MEDLINE | ID: mdl-36193302

ABSTRACT

Conventional breeding of wild (Cucumis melo var. makuwa Makino (CM)) and cultivated (Cucumis melo var. reticulatus (CR)) melons is aimed at improving their biological traits. Here, we prepared a nontoxic, bioactive extract of vitalmelon (F1 hybrid) and evaluated its antiadipogenic and antiobesity effects in fully differentiated 3T3-L1 adipocytes and high-fat diet- (HFD-) induced obese C57BL/6 mice. In fully differentiated 3T3-L1 adipocytes, the vitalmelon extract reduced the DMI- (dexamethasone, 3-isobutyl-1-methylxanthine, and insulin-) induced increases in lipid droplet number and intracellular glucose and triglyceride levels. In addition, the extract inhibited 3T3-L1 preadipocyte differentiation by downregulating PPAR-γ and target genes LPL, CD36, HMGCR, and L-FABP. To investigate the inhibitory effects of the vitalmelon extract on lipid metabolism, we measured serum lipid, hormone, and cytokine concentrations; lipolytic activity; lipid accumulation; and adipogenesis in HFD-fed mice treated with the extract. The HFD+vitalmelon-fed mice showed lower blood cholesterol, free fatty acid, sugar, leptin, and insulin concentrations but higher blood adiponectin concentrations than the HFD-fed mice. Moreover, the HFD+vitalmelon-fed mice showed lower abdominal fat levels, smaller fat cells, lower weight, and fewer lipid droplets in the liver tissue than the HFD-fed mice. Therefore, in HFD-fed mice, vitalmelon regulated lipid metabolism through PPAR-γ, highlighting its potential as a promising antiobesity functional food.


Subject(s)
Adipogenesis , Anti-Obesity Agents , 1-Methyl-3-isobutylxanthine/pharmacology , 3T3-L1 Cells , Adiponectin/pharmacology , Animals , Anti-Obesity Agents/pharmacology , Cholesterol , Cytokines/pharmacology , Dexamethasone/pharmacology , Diet, High-Fat/adverse effects , Fatty Acids, Nonesterified , Fruit/metabolism , Glucose/pharmacology , Insulin , Leptin/pharmacology , Mice , Mice, Inbred C57BL , Obesity/metabolism , PPAR gamma/metabolism , Plant Extracts/pharmacology , Sugars , Triglycerides
3.
Sci Rep ; 12(1): 444, 2022 01 10.
Article in English | MEDLINE | ID: mdl-35013505

ABSTRACT

Although islet transplantation (ITx) is a promising therapy for severe diabetes mellitus, further advancements are necessary. Adiponectin, an adipokine that regulates lipid and glucose metabolism, exerts favorable effects on islets, such as reinforcement of the insulin-releasing function. This study evaluated the possibility of adiponectin use to improve ITx outcomes. We treated mouse islets with 10 µg/mL recombinant mouse adiponectin by overnight culture and then assessed the insulin-releasing, angiogenic, and adhesion functions of the islets. Furthermore, 80 syngeneic islet equivalents with or without adiponectin treatment were transplanted into the renal subcapsular space of diabetic mice. In in vitro assessment, released insulin at high glucose stimulation, insulin content, and expressions of vascular endothelial growth factor and integrin ß1 were improved in adiponectin-treated islets. Furthermore, adiponectin treatment improved the therapeutic effect of ITx on blood glucose levels and promoted angiogenesis of the transplanted islets. However, the therapeutic effect was not pronounced in glucose tolerance test results. In conclusion, adiponectin treatment had preferable effects in the insulin-releasing, angiogenic, and adhesion functions of islets and contributed to the improvement of ITx. The future use of adiponectin treatment in clinical settings to improve ITx outcomes should be investigated.


Subject(s)
Adiponectin/therapeutic use , Islets of Langerhans Transplantation , Islets of Langerhans/drug effects , Adiponectin/pharmacology , Animals , Cell Adhesion/drug effects , Drug Evaluation, Preclinical , Insulin Secretion/drug effects , Male , Mice, Inbred C57BL , Neovascularization, Physiologic/drug effects
4.
Dig Dis Sci ; 67(6): 2232-2243, 2022 06.
Article in English | MEDLINE | ID: mdl-34009553

ABSTRACT

BACKGROUND: Intestinal fibrosis is a common complication of Crohn's disease (CD). Adiponectin reportedly exerts anti-inflammatory effects in various disease models, including colitis models. AIMS: In this study, we aimed to determine the effects of adiponectin on intestinal fibrosis and the underlying mechanisms. METHODS: A murine model of intestinal fibrosis was established by administering increasing doses of 2,4,6-trinitrobenzene sulfonic acid to Balb/c mice via enema for 7 weeks. Primary human fibroblasts were isolated from the colon tissues of patients with CD. The fibroblasts were incubated with transforming growth factor (TGF)-ß1 to establish a fibrosis model in vitro. Pathway inhibitors were used to verify the potential signaling pathways involved in the anti-fibrogenic effect of adiponectin. RESULTS: Compared with the normal mesentery, adiponectin expression was significantly increased in the hypertrophic mesentery of patients with CD. Intraperitoneal injection of adiponectin significantly decreased the activity of myeloperoxidase and the expression of pro-inflammatory cytokines (tumor necrosis factor α and interleukin 6) in the colon of fibrosis model mice, whereas the expression of the anti-inflammatory cytokine interleukin 10 was substantially increased. Moreover, adiponectin treatment inhibited colon shortening, decreased colon weight, and reduced fibrotic protein deposition in the model mice. Adiponectin reduced the phosphorylation of Smad2 and collagen deposition induced by TGF-ß1 in primary human intestinal fibroblasts, with an increase in AMP-activated protein kinase (AMPK) phosphorylation. Furthermore, this phenomenon was reversed by the AMPK inhibitor. CONCLUSIONS: Adiponectin can protect against intestinal fibrosis by enhancing the phosphorylation of AMPK and inhibiting the activity of the TGF-ß1/Smad signaling pathway.


Subject(s)
AMP-Activated Protein Kinases , Adiponectin , Crohn Disease , AMP-Activated Protein Kinases/metabolism , Adiponectin/metabolism , Adiponectin/pharmacology , Animals , Crohn Disease/pathology , Cytokines/metabolism , Fibroblasts/metabolism , Fibrosis , Humans , Mice , Phosphorylation , Transforming Growth Factor beta1/metabolism
5.
J Nutr Sci Vitaminol (Tokyo) ; 68(6): 533-539, 2022.
Article in English | MEDLINE | ID: mdl-36596552

ABSTRACT

Oral ingestion of collagen hydrolysate has various benecial effects. We developed a novel fermented collagen peptide (FCP), different from the conventional collagen peptides, by fermenting gelatin with Aspergillus sojae. This study aimed to investigate the effect of FCP in inhibiting fat accumulation under high-fat loading. Male C57BL/6J mice were fed a low- or high-fat diet, or a high-fat diet including 5% FCP for 28 d. Body weight, visceral fat weight, adiponectin levels, leptin concentration, fatty acid synthase (FAS) activity, and carnitine palmitoyltransferase 1A (CPT) activity were determined. FCP supplementation was found to significantly decrease the body weight, visceral fat weight, leptin concentration, and FAS activity, and increase adiponectin levels and CPT activity compared to that in the high-fat diet-fed group. In conclusion, FCP intake reduced visceral fat weight and body weight in high-fat diet-fed mice.


Subject(s)
Diet, High-Fat , Leptin , Mice , Male , Animals , Diet, High-Fat/adverse effects , Mice, Obese , Adiponectin/pharmacology , Intra-Abdominal Fat , Mice, Inbred C57BL , Body Weight , Collagen/pharmacology , Eating
6.
Int J Mol Sci ; 20(22)2019 Nov 15.
Article in English | MEDLINE | ID: mdl-31731705

ABSTRACT

Adiponectin, an adipokine derived from the adipose tissue, manifests anti-inflammatory effects in the metabolically active organs and is, therefore, beneficial in various metabolic diseases associated with inflammation. However, the role of adiponectin in alleviating the hypothalamic inflammation connected to the pathogenesis of obesity has not yet been clearly interrogated. Here, we identified that the systemic administration of adiponectin suppresses the activation of microglia and thereby reverses the hypothalamic inflammation during short-term exposure to a high-fat diet. Additionally, we show that adiponectin induces anti-inflammatory effects in the microglial cell line subjected to an exogenous treatment with a saturated free fatty acid. In conclusion, the current study suggests that adiponectin suppresses the saturated free fatty acid-triggered the hypothalamic inflammation by modulating the microglial activation and thus maintains energy homeostasis.


Subject(s)
Adiponectin/therapeutic use , Diet, High-Fat/adverse effects , Hypothalamus/metabolism , Inflammation/drug therapy , Microglia/metabolism , Adiponectin/pharmacology , Animals , Cell Line , Cells, Cultured , Hypothalamus/drug effects , Hypothalamus/immunology , Immunoblotting , Immunohistochemistry , Inflammation/etiology , Inflammation/immunology , Mice , Mice, Inbred C57BL , Microglia/drug effects , Microglia/immunology , Real-Time Polymerase Chain Reaction
7.
Cells ; 8(6)2019 05 29.
Article in English | MEDLINE | ID: mdl-31146419

ABSTRACT

Adiponectin and leptin play roles in the hunger response, and they can induce the inflammatory process as the initial mechanism of the innate immune response. It is possible for alterations in the levels of these adipokines to compromise the functional activity of human colostrum phagocytes. Therefore, the objective of this study is to analyze the effects of adiponectin and leptin on colostrum mononuclear (MN) cells. Colostrum was collected from 80 healthy donors, who were divided into two groups: the control group and the high body mass index (BMI) group. MN cells were used to analyze phagocytosis by flow cytometry, and reactive oxygen species (ROS), intracellular calcium, and apoptosis were assessed by fluorimetry using a microplate reader. Adipokines restored the levels of phagocytosis to the high BMI group (p < 0.05), with a mechanism that is action-dependent on the release of ROS and intracellular calcium. However, adiponectin and leptin simultaneously contributed to better microbicidal activity, thus reflecting an increase in the apoptosis level (p < 0.05) in the high BMI group. Probably, the maintenance of the balance between adiponectin and leptin levels enhances the protection and decreases the indices of neonatal infection in the breastfeeding infants of women with high BMI values. Therefore, policies that support pre-gestational weight control should be encouraged.


Subject(s)
Adiponectin/pharmacology , Inflammation/pathology , Leptin/pharmacology , Milk, Human/metabolism , Obesity/pathology , Adult , Apoptosis/drug effects , Body Mass Index , Calcium/metabolism , Colostrum/drug effects , Female , Humans , Phagocytosis/drug effects , Pregnancy , Respiratory Burst/drug effects
8.
Br J Nutr ; 119(5): 486-495, 2018 03.
Article in English | MEDLINE | ID: mdl-29508690

ABSTRACT

At birth, when immune responses are insufficient, there begins the development of the defence capability against pathogens. Leptin and adiponectin, adipokines that are present in breast milk, have been shown to play a role in the regulation of immune responses. We report here, for the first time, the influence of in vivo adipokine supplementation on the intestinal immune system in early life. Suckling Wistar rats were daily supplemented with leptin (0·7 µg/kg per d, n 36) or adiponectin (35 µg/kg per d, n 36) during the suckling period. The lymphocyte composition, proliferation and cytokine secretion from mesenteric lymph node lymphocytes (on days 14 and 21), as well as intestinal IgA and IgM concentration (day 21), were evaluated. At day 14, leptin supplementation significantly increased the TCRαß + cell proportion in mesenteric lymph nodes, in particular owing to an increase in the TCRαß + CD8+ cell population. Moreover, the leptin or adiponectin supplementation promoted the early development CD8+ cells, with adiponectin being the only adipokine capable of enhancing the lymphoproliferative ability at the end of the suckling period. Although leptin decreased intestinal IgA concentration, it had a trophic effect on the intestine in early life. Supplementation of both adipokines modulated the cytokine profile during (day 14) and at the end (day 21) of the suckling period. These results suggest that leptin and adiponectin during suckling play a role in the development of mucosal immunity in early life.


Subject(s)
Adiponectin/pharmacology , Animals, Suckling , Dietary Supplements , Intestines/drug effects , Leptin/pharmacology , Lymph Nodes/drug effects , Lymphocytes/metabolism , Animals , Animals, Newborn/growth & development , Animals, Newborn/immunology , Animals, Suckling/growth & development , Animals, Suckling/immunology , CD8 Antigens/metabolism , Immunity, Mucosal/drug effects , Immunoglobulin A/metabolism , Immunoglobulin M/metabolism , Intestinal Mucosa/drug effects , Intestines/immunology , Mesentery/immunology , Rats, Wistar , Receptors, Antigen, T-Cell, alpha-beta/metabolism
9.
Reprod Fertil Dev ; 30(4): 571-584, 2018 Mar.
Article in English | MEDLINE | ID: mdl-28911368

ABSTRACT

The present study examined the efficacy of adiponectin for regulating the reproductive, metabolic and fertility status of mice with polycystic ovary syndrome (PCOS). PCOS was induced in prepubertal (21- to 22-day-old) mice using dehydroepiandrosterone (6mg 100g-1day-1 for 25days), after which mice were administered either a low or high dose of adiponectin (5 or 15µgmL-1, s.c., respectively). PCOS mice exhibited typical features, including the presence of numerous cystic follicles, increased circulating androgens, increased body mass, altered steroidogenesis, decreased insulin receptor expression and increased serum triglycerides, serum glucose, Toll-like receptor (TLR)-4 (a marker of inflammation) and vascular endothelial growth factor (VEGF; a marker of angiogenesis). These parameters were significantly correlated with a reduction in adiponectin in PCOS mice compared with vehicle-treated control mice. Exogenous adiponectin treatment of PCOS mice restored body mass and circulating androgen, triglyceride and glucose levels. Adiponectin also restored ovarian expression of steroidogenic markers (LH receptors, steroidogenic acute regulatory protein and 3ß-hydroxysteroid dehydrogenase), insulin receptor, TLR-4 and VEGF levels in control mice. Adiponectin restored ovulation in PCOS mice, as indicated by the presence of a corpus luteum and attainment of pregnancy. These findings suggest that adiponectin effectively facilitates fertility in anovulatory PCOS. We hypothesise that systemic adiponectin treatment may be a promising therapeutic strategy for the management of PCOS.


Subject(s)
Adiponectin/therapeutic use , Body Mass Index , Ovary/drug effects , Polycystic Ovary Syndrome/drug therapy , Adiponectin/pharmacology , Androgens/blood , Animals , Dehydroepiandrosterone , Disease Models, Animal , Female , Mice , Ovary/metabolism , Phosphoproteins/metabolism , Polycystic Ovary Syndrome/chemically induced , Polycystic Ovary Syndrome/metabolism , Receptor, Insulin/metabolism , Receptors, LH/metabolism , Triglycerides/blood , Vascular Endothelial Growth Factor A/metabolism
10.
J Mol Endocrinol ; 58(4): 179-191, 2017 05.
Article in English | MEDLINE | ID: mdl-28283578

ABSTRACT

CTRP9 is a member of the C1q/TNF-related protein (CTRP) superfamily and has been studied in mammals, whereas the comparative studies of CTRP9 in non-mammalian species are still absent. In this study, ctrp9 was isolated and characterized from the orange-spotted grouper (Epinephelus coioides). The full-length cDNA of ctrp9 was 1378 bp in size with an ORF (open reading frame) of 1020 bp that encodes a 339 amino acid pre-pro hormone. The mRNA expression of ctrp9 showed a rather high level in the kidney and brain, but a low level in other tissues. Furthermore, the mRNA expression of ctrp9 decreased significantly in the liver after fasting for 7 days and restored to the normal levels after refeeding. In contrast, the ctrp9 mRNA level increased in the hypothalamus after fasting. The recombinant gCtrp9 (globular Ctrp9) was prepared using the Pichia pastoris expression system and was verified by Western blot as well as mass spectrometry assays. In the primary hepatocytes culture, the recombinant gCtrp9 could inhibit the glucose production after 12-h treatment. After i.p. (intraperitoneal) injection with recombinant gCtrp9, in hypothalamus, mRNA expression levels of npy and orexin (orexigenic factors) decreased, whereas the expression levels of crh and pomc (anorexigenic factors) increased. Moreover, i.p. injection with the recombinant gCtrp9 could reduce the serum concentrations of glucose, TG and low-density lipoprotein cholesterol but increase the content of high-density lipoprotein cholesterol. Our studies for the first time unveil the structure of Ctrp9 and its potential role as a regulatory factor of metabolism and food intake in teleost.


Subject(s)
Adiponectin/genetics , Eating/genetics , Fish Proteins/genetics , Hepatocytes/metabolism , Hypothalamus/metabolism , Perciformes/genetics , Adiponectin/metabolism , Adiponectin/pharmacology , Amino Acid Sequence , Animals , Corticotropin-Releasing Hormone/genetics , Corticotropin-Releasing Hormone/metabolism , Fish Proteins/metabolism , Fish Proteins/pharmacology , Gene Expression Regulation , Glucose/metabolism , Hepatocytes/cytology , Hepatocytes/drug effects , Kidney/metabolism , Liver/metabolism , Neuropeptide Y/genetics , Neuropeptide Y/metabolism , Open Reading Frames , Orexins/genetics , Orexins/metabolism , Perciformes/classification , Perciformes/metabolism , Phylogeny , Pichia/genetics , Pichia/metabolism , Primary Cell Culture , Pro-Opiomelanocortin/genetics , Pro-Opiomelanocortin/metabolism , Protein Precursors/genetics , Protein Precursors/metabolism , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Recombinant Proteins/pharmacology , Sequence Alignment , Sequence Homology, Amino Acid
11.
J Med Invest ; 62(3-4): 199-203, 2015.
Article in English | MEDLINE | ID: mdl-26399348

ABSTRACT

Enhancing exercise motivation is the best way to prevent obesity and diabetes. In this study, we examined whether adiponectin affects locomotion activity in Wister and Spontaneously-Running Tokushima-Shikoku (SPORTS) rats using two types of behavioral assays: home cage and wheel running activity. SPORTS rats were established from an original line from Wister strain that had shown high level of wheel running activity in our laboratory. Injection of adiponectin into the lateral ventricle of Wister rats and SPORTS rats decreased home cage activity, but no change was observed in the food intake and oxygen consumption. This result indicates the possibility that adiponectin can reduce non-exercise activity thermogenesis (NEAT) and physical activity via the central nervous system. In contrast, injection of adiponectin did not change wheel running activity in SPORTS rats. We produced hypothalamus-destructed model rat using monosodium glutamate (MSG) to elucidate the regulation site of adiponectin. Injection of adiponectin into MSG-treated SPORTS rats did not change amount of home cage activity and food intake, suggesting that adiponectin action on home cage activity was in the hypothalamic area. These results suggest that adiponectin regulates locomotion activity through mediobasal hypothalamus.


Subject(s)
Adiponectin/pharmacology , Hypothalamus/drug effects , Motor Activity/drug effects , Adiponectin/administration & dosage , Animals , Hypothalamus/physiology , Injections, Intraventricular , Male , Rats , Rats, Wistar , Sodium Glutamate/pharmacology
12.
Mol Med Rep ; 11(4): 2695-702, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25483913

ABSTRACT

This study was performed to evaluate whether endocan expression, which is known to be involved in tumor angiogenesis, was increased in rheumatoid arthritic tissues. In addition, the involvement of adiponectin in the regulation of endocan expression in arthritic joints was examined. Arthritic synovial tissues from patients with rheumatoid arthritis (RA) or osteoarthritis (OA) were immunostained with antibodies to endocan and vascular endothelial growth factor (VEGF). Subsequently, synovial cells and human umbilical vein endothelial cells were cultured and stimulated with interleukin-1 ß (IL-1ß) or adiponectin. The mRNA and protein levels of endocan were evaluated by polymerase chain reaction and ELISA, respectively. Endocan expression was markedly increased in the inflammatory sites of RA synovial tissues. In OA tissues, endocan expression was higher in tissues displaying moderate and severe inflammation than in those with mild inflammation. In vitro expression levels of endocan and VEGF in endothelial and synovial cells were differentially increased in response to IL-1ß stimulation. Adiponectin was a more potent stimulant of endocan than IL-1ß at their respective physiological concentrations in synovial cells. Endocan silencing by small interfering RNA transfection of synovial cells decreased in vitro cell migration and invasion. In conclusion, adiponectin is an important factor in the stimulation of endocan expression in synovial cells. Adiponectin-induced endocan expression in synovial cells may stimulate cell migration and invasion as well as angiogenesis in the pannus of arthritic joints.


Subject(s)
Arthritis/genetics , Gene Expression , Neoplasm Proteins/genetics , Proteoglycans/genetics , Synovial Membrane/metabolism , Adiponectin/metabolism , Adiponectin/pharmacology , Arthritis/metabolism , Arthritis/pathology , Arthritis, Rheumatoid/genetics , Arthritis, Rheumatoid/metabolism , Arthritis, Rheumatoid/pathology , Cell Movement/drug effects , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Fibroblasts/drug effects , Fibroblasts/metabolism , Gene Knockdown Techniques , Humans , Immunohistochemistry , Neoplasm Proteins/metabolism , Osteoarthritis/genetics , Osteoarthritis/metabolism , Osteoarthritis/pathology , Proteoglycans/metabolism , RNA Interference , RNA, Small Interfering/genetics , Synovial Membrane/cytology , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor A/metabolism
13.
Am J Physiol Endocrinol Metab ; 306(12): E1418-30, 2014 Jun 15.
Article in English | MEDLINE | ID: mdl-24780611

ABSTRACT

Adiponectin (APN), the most abundant adipocyte-secreted adipokine, regulates energy homeostasis and exerts well-characterized insulin-sensitizing properties. The peripheral or central effects of APN regulating bone metabolism are beginning to be explored but are still not clearly understood. In the present study, we found that APN-knockout (APN-KO) mice fed a normal diet exhibited decreased trabecular structure and mineralization and increased bone marrow adiposity compared with wild-type (WT) mice. APN intracerebroventricular infusions decreased uncoupling protein 1 (UCP1) expression in brown adipose tissue, epinephrine and norepinephrine serum levels, and osteoclast numbers, whereas osteoblast osteogenic marker expression and trabecular bone mass increased in APN-KO and WT mice. In addition, centrally administered APN increased hypothalamic tryptophan hydroxylase 2 (TPH2), cocaine- and amphetamine-regulated transcript (CART), and 5-hydroxytryptamine (serotonin) receptor 2C (Htr2C) expressions but decreased hypothalamic cannabinoid receptor-1 expression. Treatment of immortalized mouse neurons with APN demonstrated that APN-mediated effects on TPH2, CART, and Htr2C expression levels were abolished by downregulating adaptor protein containing pleckstrin homology domain, phosphotyrosine domain, and leucine zipper motif (APPL)-1 expression. Pharmacological increase in sympathetic activity stimulated adipogenic differentiation of bone marrow stromal cells (BMSC) and reversed APN-induced expression of the lysine-specific demethylases involved in regulating their commitment to the osteoblastic lineage. In conclusion, we found that APN regulates bone metabolism via central and peripheral mechanisms to decrease sympathetic tone, inhibit osteoclastic differentiation, and promote osteoblastic commitment of BMSC.


Subject(s)
Adiponectin/pharmacology , Bone Density Conservation Agents/pharmacology , Bone Marrow/drug effects , Bone and Bones/drug effects , Osteogenesis/drug effects , Peptide Fragments/pharmacology , Recombinant Proteins/pharmacology , Adiponectin/antagonists & inhibitors , Adiponectin/chemistry , Adiponectin/genetics , Adiposity/drug effects , Animals , Bone Density Conservation Agents/administration & dosage , Bone Density Conservation Agents/antagonists & inhibitors , Bone Density Conservation Agents/chemistry , Bone Marrow/metabolism , Bone and Bones/diagnostic imaging , Bone and Bones/metabolism , Cell Line , Gene Expression Regulation/drug effects , Gene Silencing , Humans , Hypothalamus/cytology , Hypothalamus/drug effects , Hypothalamus/metabolism , Infusions, Intraventricular , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Neurons/cytology , Neurons/drug effects , Neurons/metabolism , Peptide Fragments/antagonists & inhibitors , Peptide Fragments/chemistry , Peptide Fragments/genetics , Protein Conformation , Radiography , Random Allocation , Recombinant Proteins/administration & dosage , Recombinant Proteins/chemistry
14.
PLoS One ; 9(4): e94545, 2014.
Article in English | MEDLINE | ID: mdl-24718591

ABSTRACT

Obstructive sleep apnea syndrome (OSAS) is associated with many cardiovascular disorders such as heart failure, hypertension, atherosclerosis, and arrhythmia and so on. Of the many associated factors, chronic intermittent hypoxia (CIH) in particular is the primary player in OSAS. To assess the effects of CIH on cardiac function secondary to OSAS, we established a model to study the effects of CIH on Wistar rats. Specifically, we examined the possible underlying cellular mechanisms of hypoxic tissue damage and the possible protective role of adiponectin against hypoxic insults. In the first treatment group, rats were exposed to CIH conditions (nadir O2, 5-6%) for 8 hours/day, for 5 weeks. Subsequent CIH-induced cardiac dysfunction was measured by echocardiograph. Compared with the normal control (NC) group, rats in the CIH-exposed group experienced elevated levels of left ventricular end-systolic dimension and left ventricular end-systolic volume and depressed levels of left ventricular ejection fraction and left ventricular fractional shortening (p<0.05). However, when adiponectin (Ad) was added in CIH + Ad group, we saw a rescue in the elevations of the aforementioned left ventricular function (p<0.05). To assess critical cardiac injury, we detected myocardial apoptosis by Terminal deoxynucleotidyl transfer-mediated dUTP nick end-labeling (TUNEL) analysis. It was showed that the apoptosis percentage in CIH group (2.948%) was significantly higher than that in NC group (0.4167%) and CIH + Ad group (1.219%) (p<0.05). Protein expressions of cleaved caspase-3, cleaved caspase-9, and cleaved-caspase-12 validated our TUNEL results (p<0.05). Mechanistically, our results demonstrated that the proteins expressed with endoplasmic reticulum stress and the expression of reactive oxygen species (ROS) were significantly elevated under CIH conditions, whereas Ad supplementation partially decreased them. Overall, our results suggested that Ad augmentation could improve CIH-induced left ventricular dysfunction and associated myocardial apoptosis by inhibition of ROS-dependent ER stress.


Subject(s)
Adiponectin/pharmacology , Cardiotonic Agents/pharmacology , Endoplasmic Reticulum Stress/drug effects , Hypoxia/drug therapy , Myocardium/pathology , Adiponectin/blood , Adiponectin/therapeutic use , Animals , Apoptosis/drug effects , Cardiotonic Agents/therapeutic use , Caspases/metabolism , Electrocardiography , Hypoxia/blood , In Situ Nick-End Labeling , Male , Membrane Proteins/metabolism , Mitogen-Activated Protein Kinases/metabolism , Protein Serine-Threonine Kinases/metabolism , Rats, Wistar , Reactive Oxygen Species/metabolism , Signal Transduction/drug effects , Unfolded Protein Response/drug effects
15.
Leukemia ; 28(10): 2080-9, 2014 Oct.
Article in English | MEDLINE | ID: mdl-24646889

ABSTRACT

Obesity increases the risk of developing multiple myeloma (MM). Adiponectin is a cytokine produced by adipocytes, but paradoxically decreased in obesity, that has been implicated in MM progression. Herein, we evaluated how prolonged exposure to adiponectin affected the survival of MM cells as well as putative signaling mechanisms. Adiponectin activates protein kinase A (PKA), which leads to decreased AKT activity and increased AMP-activated protein kinase (AMPK) activation. AMPK, in turn, induces cell cycle arrest and apoptosis. Adiponectin-induced apoptosis may be mediated, at least in part, by the PKA/AMPK-dependent decline in the expression of the enzyme acetyl-CoA-carboxylase (ACC), which is essential to lipogenesis. Supplementation with palmitic acid, the preliminary end product of fatty acid synthesis, rescues MM cells from adiponectin-induced apoptosis. Furthermore, 5-(tetradecyloxy)-2-furancarboxylic acid (TOFA), an ACC inhibitor, exhibited potent antiproliferative effects on MM cells that could also be inhibited by fatty acid supplementation. Thus, adiponectin's ability to reduce survival of MM cells appears to be mediated through its ability to suppress lipogenesis. Our findings suggest that PKA/AMPK pathway activators, or inhibitors of ACC, may be useful adjuvants to treat MM. Moreover, the antimyeloma effect of adiponectin supports the concept that hypoadiponectinemia, as occurs in obesity, promotes MM tumor progression.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Adiponectin/pharmacology , Cyclic AMP-Dependent Protein Kinases/metabolism , Multiple Myeloma/metabolism , Acetyl-CoA Carboxylase/metabolism , Adipokines/metabolism , Adiponectin/deficiency , Adiponectin/metabolism , Animals , Apoptosis , Cell Cycle , Cell Line, Tumor , Cell Proliferation , Disease Progression , Fatty Acids/chemistry , Furans/chemistry , Humans , Lipogenesis , Metabolism, Inborn Errors/metabolism , Mice , Multiple Myeloma/drug therapy , Obesity/metabolism , Signal Transduction
16.
Nature ; 503(7477): 493-9, 2013 Nov 28.
Article in English | MEDLINE | ID: mdl-24172895

ABSTRACT

Adiponectin secreted from adipocytes binds to adiponectin receptors AdipoR1 and AdipoR2, and exerts antidiabetic effects via activation of AMPK and PPAR-α pathways, respectively. Levels of adiponectin in plasma are reduced in obesity, which causes insulin resistance and type 2 diabetes. Thus, orally active small molecules that bind to and activate AdipoR1 and AdipoR2 could ameliorate obesity-related diseases such as type 2 diabetes. Here we report the identification of orally active synthetic small-molecule AdipoR agonists. One of these compounds, AdipoR agonist (AdipoRon), bound to both AdipoR1 and AdipoR2 in vitro. AdipoRon showed very similar effects to adiponectin in muscle and liver, such as activation of AMPK and PPAR-α pathways, and ameliorated insulin resistance and glucose intolerance in mice fed a high-fat diet, which was completely obliterated in AdipoR1 and AdipoR2 double-knockout mice. Moreover, AdipoRon ameliorated diabetes of genetically obese rodent model db/db mice, and prolonged the shortened lifespan of db/db mice on a high-fat diet. Thus, orally active AdipoR agonists such as AdipoRon are a promising therapeutic approach for the treatment of obesity-related diseases such as type 2 diabetes.


Subject(s)
Diabetes Mellitus, Type 2/drug therapy , Longevity/drug effects , Obesity/physiopathology , Piperidines/pharmacology , Receptors, Adiponectin/agonists , Adenylate Kinase/metabolism , Adiponectin/metabolism , Adiponectin/pharmacology , Adipose Tissue, White/drug effects , Adipose Tissue, White/metabolism , Adipose Tissue, White/pathology , Administration, Oral , Animals , Diabetes Mellitus, Type 2/complications , Diabetes Mellitus, Type 2/metabolism , Diabetes Mellitus, Type 2/prevention & control , Diet, High-Fat , Drug Evaluation, Preclinical , Dyslipidemias/drug therapy , Enzyme Activation/drug effects , Glucose Intolerance/drug therapy , Inflammation/drug therapy , Insulin Resistance , Liver/drug effects , Liver/metabolism , Liver/pathology , Mice , Mitochondria/drug effects , Mitochondria/metabolism , Muscle Fibers, Skeletal/cytology , Muscle Fibers, Skeletal/drug effects , Muscles/cytology , Obesity/complications , Obesity/drug therapy , Obesity/genetics , Oxidative Stress/drug effects , PPAR alpha/metabolism , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha , Piperidines/administration & dosage , Piperidines/metabolism , Piperidines/therapeutic use , Receptors, Adiponectin/deficiency , Receptors, Adiponectin/genetics , Receptors, Adiponectin/metabolism , Signal Transduction/drug effects , Small Molecule Libraries/chemistry , Transcription Factors/biosynthesis , Triglycerides/metabolism
17.
PLoS One ; 8(4): e61343, 2013.
Article in English | MEDLINE | ID: mdl-23630583

ABSTRACT

The aims of the present study were to elucidate a possible mechanism of kidney crystal formation by using a metabolic syndrome (MetS) mouse model and to assess the effectiveness of adiponectin treatment for the prevention of kidney crystals. Further, we performed genome-wide expression analyses for investigating novel genetic environmental changes. Wild-type (+/+) mice showed no kidney crystal formation, whereas ob/ob mice showed crystal depositions in their renal tubules. However, this deposition was remarkably reduced by adiponectin. Expression analysis of genes associated with MetS-related kidney crystal formation identified 259 genes that were >2.0-fold up-regulated and 243 genes that were <0.5-fold down-regulated. Gene Ontology (GO) analyses revealed that the up-regulated genes belonged to the categories of immunoreaction, inflammation, and adhesion molecules and that the down-regulated genes belonged to the categories of oxidative stress and lipid metabolism. Expression analysis of adiponectin-induced genes related to crystal prevention revealed that the numbers of up- and down-regulated genes were 154 and 190, respectively. GO analyses indicated that the up-regulated genes belonged to the categories of cellular and mitochondrial repair, whereas the down-regulated genes belonged to the categories of immune and inflammatory reactions and apoptosis. The results of this study provide compelling evidence that the mechanism of kidney crystal formation in the MetS environment involves the progression of an inflammation and immunoresponse, including oxidative stress and adhesion reactions in renal tissues. This is the first report to prove the preventive effect of adiponectin treatment for kidney crystal formation by renoprotective activities and inhibition of inflammation and apoptosis.


Subject(s)
Adiponectin/pharmacology , Anti-Inflammatory Agents/pharmacology , Apoptosis/drug effects , Kidney Calculi/prevention & control , Metabolic Syndrome/complications , Adiponectin/genetics , Adiponectin/metabolism , Adiponectin/therapeutic use , Animals , Anti-Inflammatory Agents/therapeutic use , Calcium Oxalate/metabolism , Chemokine CCL2/genetics , Chemokine CCL2/metabolism , Cholesterol/metabolism , Disease Models, Animal , Drug Evaluation, Preclinical , Kidney/drug effects , Kidney Calculi/etiology , Kidney Calculi/metabolism , Male , Metabolic Syndrome/metabolism , Mice , Mice, Inbred C57BL , Mice, Obese , Oligonucleotide Array Sequence Analysis , Osteopontin/genetics , Osteopontin/metabolism , Superoxide Dismutase/genetics , Superoxide Dismutase/metabolism , Transcriptome
18.
J Endocrinol ; 215(1): 107-17, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22859860

ABSTRACT

Accelerated atherosclerosis is the primary cardiovascular manifestation of diabetes and correlates inversely with levels of circulating adiponectin, an anti-atherosclerotic adipokine that declines in diabetes. We therefore initiated a study to examine the mechanisms by which adiponectin, a hormone released from adipose tissue, influences the proliferation of vascular smooth muscle cells (SMCs). Addition of adiponectin to quiescent porcine coronary artery SMCs increased both protein and DNA synthesis and concurrently activated ERK1/2 and Akt. By contrast, globular adiponectin, a truncated form of this protein, exhibited anti-mitogenic properties as indicated by the inhibition of protein and DNA synthesis in SMCs stimulated with platelet-derived growth factor (PDGF). Whereas globular adiponectin did not stimulate growth-related signal transduction pathways, it was able to block the PDGF-dependent phosphorylation of eukaryotic elongation factor 2 kinase, a regulator of protein synthesis. Proteolysis of adiponectin with trypsin, which produces globular adiponectin, reversed the growth-stimulating actions of the undigested protein. As the existence of globular adiponectin remains controversial, western blotting was used to establish its presence in rat serum. We found that globular adiponectin was detectable in rat serum, but this result was not obtained with all antibodies. The contrasting properties of adiponectin and its globular form with respect to SMC proliferation suggest that protection against atherosclerosis may therefore be mediated, in part, by the level of globular adiponectin.


Subject(s)
Adiponectin/chemistry , Adiponectin/metabolism , Adiponectin/pharmacology , Cell Proliferation/drug effects , Myocytes, Smooth Muscle/drug effects , Protein Folding , Adenylate Kinase/metabolism , Adiponectin/blood , Animals , Cells, Cultured , Down-Regulation/drug effects , Drug Evaluation, Preclinical , Myocytes, Smooth Muscle/metabolism , Myocytes, Smooth Muscle/physiology , Protein Isoforms/chemistry , Protein Isoforms/metabolism , Protein Isoforms/pharmacology , Proteolysis/drug effects , Rats , Rats, Sprague-Dawley , Signal Transduction/drug effects , Signal Transduction/physiology , Swine
19.
J Endocrinol ; 214(2): 177-89, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22582096

ABSTRACT

Adiponectin secreted from adipose tissues plays a role in the regulation of energy homeostasis, food intake, and reproduction in the hypothalamus. We have previously demonstrated that adiponectin significantly inhibited GNRH secretion from GT1-7 hypothalamic GNRH neuron cells. In this study, we further investigated the effect of adiponectin on hypothalamic KISS1 gene transcription, which is the upstream signal of GNRH. We found that globular adiponectin (gAd) or AICAR, an artificial AMPK activator, decreased KISS1 mRNA transcription and promoter activity. Conversely, inhibition of AMPK by Compound C or AMPKα1-SiRNA augmented KISS1 mRNA transcription and promoter activity. Additionally, gAd and AICAR decreased the translocation of specificity protein-1 (SP1) from cytoplasm to nucleus; however, Compound C and AMPKα1-siRNA played an inverse role. Our experiments in vivo demonstrated that the expression of Kiss1 mRNA was stimulated twofold in the Compound C-treated rats and decreased about 60-70% in gAd- or AICAR-treated rats compared with control group. The numbers of kisspeptin immunopositive neurons in the arcuate nucleus region of Sprague Dawley rats mimicked the same trend seen in Kiss1 mRNA levels in animal groups with different treatments. In conclusion, our results provide the first evidence that adiponectin reduces Kiss1 gene transcription in GT1-7 cells through activation of AMPK and subsequently decreased translocation of SP1.


Subject(s)
Adenylate Kinase/physiology , Adiponectin/pharmacology , Hypothalamus/drug effects , Kisspeptins/genetics , Neurons/drug effects , Sp1 Transcription Factor/physiology , Adenylate Kinase/metabolism , Adiponectin/physiology , Aminoimidazole Carboxamide/analogs & derivatives , Aminoimidazole Carboxamide/pharmacology , Animals , Cells, Cultured , Down-Regulation/drug effects , Down-Regulation/genetics , Female , Hypothalamus/cytology , Hypothalamus/metabolism , Kisspeptins/metabolism , Neurons/metabolism , Phosphorylation/drug effects , Protein Transport/drug effects , Protein Transport/genetics , Protein Transport/physiology , Rats , Rats, Sprague-Dawley , Ribonucleotides/pharmacology , Sp1 Transcription Factor/metabolism , Transcription, Genetic/drug effects
20.
PLoS One ; 7(12): e52193, 2012.
Article in English | MEDLINE | ID: mdl-23284930

ABSTRACT

A high fat (HF) diet rapidly impairs the ability of adiponectin (Ad) to stimulate fatty acid (FA) oxidation in oxidative soleus muscle, but the underlying mechanism remains elusive. Mere days of HF feeding also increase the muscle's production and accumulation of reactive oxygen species (ROS) and shift cellular redox to a more oxidized state. It seems plausible that this shift towards a more oxidized state might act as negative feedback to suppress the ability of Ad to stimulate FA oxidation and generate more ROS. Therefore, we sought to determine whether i) a shift towards a more oxidized redox state (reduction in GSH/2GSSG) coincided with impaired Ad-stimulated palmitate oxidation in oxidative and glycolytic rodent muscle after 5 days of HF feeding (60% kCal), and ii) if supplementation with the antioxidant, N-acetylcysteine (NAC) could prevent the HF-diet induced impairment in Ad-response. Globular Ad (gAd) increased palmitate oxidation in isolated soleus and EDL muscles by 42% and 34%, respectively (p<0.05) but this was attenuated with HF feeding in both muscles. HF feeding decreased total GSH (-26%, p<0.05) and GSH/2GSSG (-49%, p<0.05) in soleus, but not EDL. Supplementation with NAC prevented the HF diet-induced reductions in GSH and GSH/2GSSG in soleus, but did not prevent the loss of Ad response in either muscle. Furthermore, direct incubations with H(2)O(2) did not impair Ad-stimulated FA oxidation in either muscle. In conclusion, our data indicates that skeletal muscle Ad resistance is rapidly induced in both oxidative and glycolytic muscle, independently of altered cellular redox state.


Subject(s)
Adiponectin/pharmacology , Fatty Acids/metabolism , Muscle, Skeletal/drug effects , Muscle, Skeletal/metabolism , Animals , Blood Glucose/drug effects , Body Weight/drug effects , Diet, High-Fat/adverse effects , Female , Glutathione/metabolism , Lipid Metabolism/drug effects , Oxidation-Reduction/drug effects , Protein Carbonylation/drug effects , Rats , Rats, Sprague-Dawley
SELECTION OF CITATIONS
SEARCH DETAIL