Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 26
1.
Clin Rheumatol ; 41(1): 255-264, 2022 Jan.
Article En | MEDLINE | ID: mdl-34387761

OBJECTIVES: Adiponectin (AD) highly expressed in synovial tissue correlates closely with progressive bone erosion in rheumatoid arthritis (RA). However, it remains unknown whether AD receptor mediates the pathogenesis of RA. This study aimed to investigate the effects of adiponectin receptor 1 (AdipoR1) signaling on synovial inflammation and joint damage in collagen-induced arthritis. METHODS: AD and AdipoR1 expression in synovial tissue of RA and osteoarthritis (OA) patients were tested by PCR and western blotting. The frequency of AdipoR1 on RA synovial fibroblasts (RASFs) was examined by flow cytometry after stimulation with AD, IL-6, or TNF-α. AdipoR1 was knocked down in human RASF cell line (MH7A) and CIA mice joints using lentiviral particles carrying the AdipoR1 short hairpin RNA (shAdipoR1). Both the proliferation and apoptosis of MH7A and the secretion of inflammatory factors from MH7A were examined in vitro. The therapeutic effect of local AdipoR1 inhibition on CIA mice was assessed in vivo. RESULTS: Both the expression of AD and AdipoR1 were significantly higher in RA synovial tissue. AdipoR1 on RASFs was upregulated by AD. Silencing AdipoR1 remarkably reduced lipopolysaccharides-induced proliferation and promoted the apoptosis of MH7A. Moreover, AdipoR1 knockdown inhibited the release of inflammatory factors in vitro. In CIA mice, local AdipoR1 inhibition effectively decreased joint inflammation and alleviated bone destruction via suppressing RANKL and RANKL/OPG ratio in vivo. CONCLUSIONS: AdipoR1 signaling participates in the process of synovial inflammation and joint damage in CIA. Local blockade of AdipoR1 might be a new target for the clinical treatment of RA. Key points • Local AdipoR1 inhibition decreased joint inflammation and alleviated bone destruction in CIA.


Arthritis, Experimental , Arthritis, Rheumatoid , Receptors, Adiponectin/antagonists & inhibitors , Animals , Arthritis, Experimental/drug therapy , Arthritis, Rheumatoid/drug therapy , Fibroblasts , Humans , Inflammation , Mice , Receptors, Adiponectin/genetics , Signal Transduction , Synovial Membrane
2.
Mol Neurodegener ; 16(1): 23, 2021 04 13.
Article En | MEDLINE | ID: mdl-33849621

BACKGROUND: Recently, we and other researchers reported that brain metabolic disorders are implicated in Alzheimer's disease (AD), a progressive, devastating and incurable neurodegenerative disease. Hence, novel therapeutic approaches are urgently needed to explore potential and novel therapeutic targets/agents for the treatment of AD. The neuronal adiponectin receptor 1 (AdipoR1) is an emerging potential target for intervention in metabolic-associated AD. We aimed to validate this hypothesis and explore in-depth the therapeutic effects of an osmotin-derived adiponectin-mimetic novel nonapeptide (Os-pep) on metabolic-associated AD. METHODS: We used an Os-pep dosage regimen (5 µg/g, i.p., on alternating days for 45 days) for APP/PS1 in amyloid ß oligomer-injected, transgenic adiponectin knockout (Adipo-/-) and AdipoR1 knockdown mice. After behavioral studies, brain tissues were subjected to biochemical and immunohistochemical analyses. In separate cohorts of mice, electrophysiolocal and Golgi staining experiments were performed. To validate the in vivo studies, we used human APP Swedish (swe)/Indiana (ind)-overexpressing neuroblastoma SH-SY5Y cells, which were subjected to knockdown of AdipoR1 and APMK with siRNAs, treated with Os-pep and other conditions as per the mechanistic approach, and we proceeded to perform further biochemical analyses. RESULTS: Our in vitro and in vivo results show that Os-pep has good safety and neuroprotection profiles and crosses the blood-brain barrier. We found reduced levels of neuronal AdipoR1 in human AD brain tissue. Os-pep stimulates AdipoR1 and its downstream target, AMP-activated protein kinase (AMPK) signaling, in AD and Adipo-/- mice. Mechanistically, in all of the in vivo and in vitro studies, Os-pep rescued aberrant neuronal metabolism by reducing neuronal insulin resistance and activated downstream insulin signaling through regulation of AdipoR1/AMPK signaling to consequently improve the memory functions of the AD and Adipo-/- mice, which was associated with improved synaptic function and long-term potentiation via an AdipoR1-dependent mechanism. CONCLUSION: Our findings show that Os-pep activates AdipoR1/AMPK signaling and regulates neuronal insulin resistance and insulin signaling, which subsequently rescues memory deficits in AD and adiponectin-deficient models. Taken together, the results indicate that Os-pep, as an adiponectin-mimetic novel nonapeptide, is a valuable and promising potential therapeutic candidate to treat aberrant brain metabolism associated with AD and other neurodegenerative diseases.


Alzheimer Disease/drug therapy , Memory Disorders/prevention & control , Neuroprotective Agents/pharmacology , Receptors, Adiponectin/antagonists & inhibitors , AMP-Activated Protein Kinases/metabolism , Adiponectin/deficiency , Alzheimer Disease/metabolism , Alzheimer Disease/psychology , Amyloid beta-Peptides/genetics , Animals , Cell Line, Tumor , Drug Evaluation, Preclinical , Humans , Insulin Resistance , Male , Maze Learning , Memory Disorders/drug therapy , Memory Disorders/etiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Neurons/drug effects , Neuroprotective Agents/therapeutic use , Presenilin-1/genetics , RNA Interference , RNA, Small Interfering/genetics , Receptors, Adiponectin/genetics , Signal Transduction
3.
Sci Rep ; 11(1): 2923, 2021 02 03.
Article En | MEDLINE | ID: mdl-33536560

The antidiabetic adiponectin receptor agonist AdipoRon has been shown to suppress the tumour growth of human pancreatic cancer cells. Because obesity and diabetes affect pancreatic cancer progression and chemoresistance, we investigated the effect of AdipoRon on orthotopic tumour growth of Panc02 pancreatic cancer cells in DIO (diet-induced obese) prediabetic mice. Administration of AdipoRon into DIO mice fed high-fat diets, in which prediabetic conditions were alleviated to some extent, did not reduce either body weight or tumour growth. However, when the DIO mice were fed low-fat diets, body weight and the blood leptin level gradually decreased, and importantly, AdipoRon became effective in suppressing tumour growth, which was accompanied by increases in necrotic areas and decreases in Ki67-positive cells and tumour microvessels. AdipoRon inhibited cell growth and induced necrotic cell death of Panc02 cells and suppressed angiogenesis of endothelial MSS31 cells. Insulin and IGF-1 only slightly reversed the AdipoRon-induced suppression of Panc02 cell survival but had no effect on the AdipoRon-induced suppression of MSS31 cell angiogenesis. Leptin significantly ameliorated AdipoRon-induced suppression of angiogenesis through inhibition of ERK1/2 activation. These results suggest that obesity-associated factors weaken the anticancer effect of AdipoRon, which indicates the importance of weight loss in combating pancreatic cancer.


Obesity/complications , Pancreatic Neoplasms/drug therapy , Piperidines/pharmacology , Receptors, Adiponectin/antagonists & inhibitors , Animals , Cell Line, Tumor/transplantation , Diet, High-Fat/adverse effects , Disease Models, Animal , Drug Resistance, Neoplasm , Humans , Male , Mice , Obesity/metabolism , Pancreatic Neoplasms/complications , Pancreatic Neoplasms/pathology , Piperidines/therapeutic use , Receptors, Adiponectin/metabolism
4.
Int J Mol Sci ; 22(2)2021 Jan 17.
Article En | MEDLINE | ID: mdl-33477324

Adiponectin plays multiple critical roles in modulating various physiological processes by binding to its receptors. The functions of PEG-BHD1028, a potent novel peptide agonist to AdipoRs, was evaluated using in vitro and in vivo models based on the reported action spectrum of adiponectin. To confirm the design concept of PEG-BHD1028, the binding sites and their affinities were analyzed using the SPR (Surface Plasmon Resonance) assay. The results revealed that PEG-BHD1028 was bound to two heterogeneous binding sites of AdipoR1 and AdipoR2 with a relatively high affinity. In C2C12 cells, PEG-BHD1028 significantly activated AMPK and subsequent pathways and enhanced fatty acid ß-oxidation and mitochondrial biogenesis. Furthermore, it also facilitated glucose uptake by lowering insulin resistance in insulin-resistant C2C12 cells. PEG-BHD1028 significantly reduced the fasting plasma glucose level in db/db mice following a single s.c. injection of 50, 100, and 200 µg/Kg and glucose tolerance at a dose of 50 µg/Kg with significantly decreased insulin production. The animals received 5, 25, and 50 µg/Kg of PEG-BHD1028 for 21 days significantly lost their weight after 18 days in a range of 5-7%. These results imply the development of PEG-BHD1028 as a potential adiponectin replacement therapeutic agent.


Peptides/pharmacology , Receptors, Adiponectin/metabolism , Adiponectin/metabolism , Animals , Binding Sites , Cell Culture Techniques , Fatty Acids/metabolism , Insulin/metabolism , Insulin Resistance , Male , Mice , Mice, Inbred C57BL , Mitochondria/metabolism , Myoblasts/metabolism , Organelle Biogenesis , Oxidation-Reduction , Peptides/chemistry , Polyethylene Glycols/pharmacology , Receptors, Adiponectin/antagonists & inhibitors , Signal Transduction
5.
J Neuroinflammation ; 16(1): 110, 2019 May 25.
Article En | MEDLINE | ID: mdl-31128596

BACKGROUND: Microglia-mediated neuroinflammation is important in Alzheimer's disease (AD) pathogenesis. Extracellular deposition of ß-amyloid (Aß), a major pathological hallmark of AD, can induce microglia activation. Adiponectin (APN), an adipocyte-derived adipokine, exerts anti-inflammatory effects in the periphery and brain. Chronic APN deficiency leads to cognitive impairment and AD-like pathologies in aged mice. Here, we aim to study the role of APN in regulating microglia-mediated neuroinflammation in AD. METHODS: Inflammatory response of cultured microglia (BV2 cells) to AßO and effects of APN were studied by measuring levels of proinflammatory cytokines (tumor necrosis factor α [TNFα] and interleukin-1ß [IL-1ß]) in cultured medium before and after exposure to AßO, with and without APN pretreatment. Adiponectin receptor 1 (AdipoR1) and receptor 2 (AdipoR2) were targeted by small interference RNA. To study the neuroprotective effect of APN, cultured HT-22 hippocampal cells were treated with conditioned medium of AßO-exposed BV2 cells or were co-cultured with BV2 cells in transwells. The cytotoxicity of HT-22 hippocampal cells was assessed by MTT reduction. We generated APN-deficient AD mice (APN-/-5xFAD) by crossing APN-knockout mice with 5xFAD mice to determine the effects of APN deficiency on microglia-mediated neuroinflammation in AD. RESULTS: AdipoR1 and AdipoR2 were expressed in BV2 cells and microglia of mice. Pretreatment with APN for 2 h suppressed TNFα and IL-1ß release induced by AßO in BV2 cells. Additionally, APN rescued the decrease of AMPK phosphorylation and suppressed nuclear translocation of nuclear factor kappa B (NF-κB) induced by AßO. Compound C, an inhibitor of AMPK, abolished these effects of APN. Knockdown of AdipoR1, but not AdipoR2 in BV2 cells, inhibited the ability of APN to suppress proinflammatory cytokine release induced by AßO. Moreover, pretreatment with APN inhibited the cytotoxicity of HT-22 cells co-cultured with AßO-exposed BV2 cells. Lastly, APN deficiency exacerbated microglia activation in 9-month-old APN-/-5xFAD mice associated with upregulation of TNFα and IL-1ß in the cortex and hippocampus. CONCLUSIONS: Our findings demonstrate that APN inhibits inflammatory response of microglia to AßO via AdipoR1-AMPK-NF-κB signaling, and APN deficiency aggravates microglia activation and neuroinflammation in AD mice. APN may be a novel therapeutic agent for inhibiting neuroinflammation in AD.


AMP-Activated Protein Kinases/biosynthesis , Adiponectin/pharmacology , Amyloid beta-Peptides/toxicity , Microglia/metabolism , NF-kappa B/biosynthesis , Peptide Fragments/toxicity , Receptors, Adiponectin/biosynthesis , AMP-Activated Protein Kinases/antagonists & inhibitors , Amyloid beta-Peptides/antagonists & inhibitors , Animals , Dose-Response Relationship, Drug , Inflammation/chemically induced , Inflammation/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Microglia/drug effects , NF-kappa B/antagonists & inhibitors , Peptide Fragments/antagonists & inhibitors , Receptors, Adiponectin/antagonists & inhibitors , Signal Transduction/drug effects , Signal Transduction/physiology
6.
Int J Mol Sci ; 20(7)2019 Apr 10.
Article En | MEDLINE | ID: mdl-30974901

Adiponectin, an adipokine secreted by adipocytes, exerts favorable effects in the milieu of diabetes and metabolic syndrome through its anti-inflammatory, antifibrotic, and antioxidant effects. It mediates fatty acid metabolism by inducing AMP-activated protein kinase (AMPK) phosphorylation and increasing peroxisome proliferative-activated receptor (PPAR)-α expression through adiponectin receptor (AdipoR)1 and AdipoR2, respectively, which in turn activate PPAR gamma coactivator 1 alpha (PGC-1α), increase the phosphorylation of acyl CoA oxidase, and upregulate the uncoupling proteins involved in energy consumption. Moreover, adiponectin potently stimulates ceramidase activity associated with its two receptors and enhances ceramide catabolism and the formation of its anti-apoptotic metabolite, sphingosine 1 phosphate (S1P), independently of AMPK. Low circulating adiponectin levels in obese patients with a risk of insulin resistance, type 2 diabetes, and cardiovascular diseases, and increased adiponectin expression in the state of albuminuria suggest a protective and compensatory role for adiponectin in mitigating further renal injury during the development of overt diabetic kidney disease (DKD). We propose AdipoRon, an orally active synthetic adiponectin receptor agonist as a promising drug for restoration of DKD without inducing systemic adverse effects. Its renoprotective role against lipotoxicity and oxidative stress by enhancing the AMPK/PPARα pathway and ceramidase activity through AdipoRs is revealed here.


Adiponectin/metabolism , Diabetes Mellitus, Type 2 , Obesity , Oxidative Stress/drug effects , Piperidines/therapeutic use , Receptors, Adiponectin/antagonists & inhibitors , Animals , Diabetes Mellitus, Type 2/drug therapy , Diabetes Mellitus, Type 2/metabolism , Diabetes Mellitus, Type 2/pathology , Humans , Kidney/metabolism , Kidney/pathology , Lipid Metabolism/drug effects , Obesity/drug therapy , Obesity/metabolism , Obesity/pathology , Receptors, Adiponectin/metabolism
7.
Biomed Pharmacother ; 109: 2293-2304, 2019 Jan.
Article En | MEDLINE | ID: mdl-30551487

EndMT plays an important role in the relationship between endothelial dysfunction and atherosclerosis. This work will elucidate the biofunction induced by miR-449a and lipid rafts in EndMT and development of atherosclerosis. The differential miRNA expression between atherosclerotic plaques and normal arteries were analyzed. The luciferase activities of AdipoR2 3' UTR treated with miR-449a were determined. ECs were dealt with miR-449a mimics or inhibitors, then cell proliferation and migration were assessed. Moreover, the expression of AdipoR2 and mesenchymal cell markers were analyzed. The influences of lipid rafts related to reciprocity between E-cadherin and AdipoR2 on TNF-α-induced damage in ECs were investigated. ApoE KO diabetic mice were used to explore the potential roles of miR-449a on atherosclerosis. Our results indicated that compared with normal arteries, 17 miRNAs were upregulated and 3 miRNAs were down-regulated in atherosclerotic plaques. The relative expression of miR-449a in plaques was significantly higher than that in normal arteries. MiR-449a suppressed AdipoR2 expression, additionally its interaction protein E-cadherin in ECs. MiR-449a enhanced expression of mesenchymal cell markers, induced cell proliferation and migration of ECs, regulated the interaction between E-cadherin and AdipoR2 interceded by lipid rafts. The miR-449a antagomir could protect against the development process of atherosclerosis in ApoE KO diabetic mice. In conclusion, miR-449a targeted to AdipoR2, and was a crucial mediator of EndMT and atherosclerosis in ECs through regulating E-cadherin bindability with AdipoR2 in lipid rafts. These results suggested that aim to lipid rafts and miR-449a in chronic EC inflammation response, was a feasible therapy strategy for atherosclerosis.


Atherosclerosis/metabolism , Cadherins/metabolism , Epithelial-Mesenchymal Transition/physiology , Membrane Microdomains/metabolism , MicroRNAs/biosynthesis , Receptors, Adiponectin/biosynthesis , Animals , Atherosclerosis/pathology , Cadherins/antagonists & inhibitors , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Experimental/pathology , Humans , Male , Membrane Microdomains/pathology , Mice , Mice, Knockout , Receptors, Adiponectin/antagonists & inhibitors
8.
Drug Discov Today ; 23(6): 1305-1311, 2018 06.
Article En | MEDLINE | ID: mdl-29747002

Given the paradigm of anti-insulin resistance in therapies for metabolic syndrome, there has been considerable interest in adiponectin (APN), an adipocyte-derived sensitizer of insulin receptor signaling. In contrast to hypoadiponectinemia in metabolic syndrome, evidence suggests that Alzheimer's disease (AD) and other diseases, including chronic heart failure (CHF) and chronic kidney disease (CKD), are characterized by hyperadiponectinemia as well as the APN/obesity paradoxes, indicating that a decrease in APN might also be beneficial for these diseases. Thus, distinct from metabolic syndrome, it is anticipated that APN receptor antagonists rather than agonists might be effective in therapy for some chronic diseases.


Aging/metabolism , Receptors, Adiponectin/metabolism , Adiponectin/deficiency , Adiponectin/metabolism , Animals , Chronic Disease , Humans , Metabolic Syndrome/drug therapy , Metabolic Syndrome/metabolism , Metabolism, Inborn Errors/drug therapy , Metabolism, Inborn Errors/metabolism , Obesity/metabolism , Receptors, Adiponectin/agonists , Receptors, Adiponectin/antagonists & inhibitors , Signal Transduction
9.
Int J Biochem Cell Biol ; 94: 133-145, 2018 01.
Article En | MEDLINE | ID: mdl-29197627

Resistin is an adipocyte-derived cytokine and was named for its role in the development of insulin resistance. Increased serum resistin levels are also associated with steatohepatitis and non-alcoholic fatty liver disease. In a previous study, resistin was observed to reduce mitochondrial content and upregulate miR-34a significantly in the liver. In this study, male C57BL/6 mice were injected with agomir-34a or control agomir, and HepG2 cells were transfected with miR-34a mimics or inhibitors to assess their role in resistin-induced fat deposition. The overexpression of miR-34a increased liver and HepG2 cell TAG content, decreased mitochondrial content, changed mitochondrial morphology and impaired mitochondrial function. In contrast, a miR-34a inhibitor significantly restored the TAG content and mitochondrial transmembrane potential. A study of transcriptional regulation revealed that C/EBPß is essential for upregulating miR-34a by resistin. Furthermore, miR-34a inhibited the PPARα signaling pathway by binding to sites in the 3'UTR of AdipoR2 genes and the AMPK pathway. Consequently, this increased the fat content and decreased the mitochondrial content in HepG2 cells. This paper reveals a novel mechanism for mitochondrial regulation, which suggests that normal mitochondrial content and function is crucial for lipid metabolism in the liver.


AMP-Activated Protein Kinases/metabolism , Hepatocytes/metabolism , Lipid Metabolism , MicroRNAs/metabolism , Mitochondrial Dynamics , PPAR alpha/agonists , Resistin/metabolism , 3' Untranslated Regions/drug effects , AMP-Activated Protein Kinases/chemistry , Animals , Antagomirs/administration & dosage , Antagomirs/pharmacology , Gene Expression Regulation/drug effects , Hep G2 Cells , Hepatocytes/drug effects , Hepatocytes/ultrastructure , Humans , Injections, Intravenous , Lipid Metabolism/drug effects , Male , Mice, Inbred C57BL , MicroRNAs/agonists , MicroRNAs/antagonists & inhibitors , MicroRNAs/chemistry , Microscopy, Electron, Transmission , Mitochondrial Dynamics/drug effects , Non-alcoholic Fatty Liver Disease/chemically induced , Non-alcoholic Fatty Liver Disease/etiology , Non-alcoholic Fatty Liver Disease/metabolism , Non-alcoholic Fatty Liver Disease/pathology , PPAR alpha/metabolism , RNA/metabolism , RNA Interference , Receptors, Adiponectin/antagonists & inhibitors , Receptors, Adiponectin/genetics , Receptors, Adiponectin/metabolism , Recombinant Proteins/administration & dosage , Recombinant Proteins/metabolism , Recombinant Proteins/pharmacology , Resistin/administration & dosage , Resistin/genetics , Resistin/pharmacology , Signal Transduction/drug effects
10.
Sci Rep ; 7(1): 12435, 2017 09 29.
Article En | MEDLINE | ID: mdl-28963462

Recent studies on neurodegeneration have focused on dysfunction of CNS energy metabolism as well as proteinopathies. Adiponectin (ADPN), an adipocyte-derived hormone, plays a major role in the regulation of insulin sensitivity and glucose homeostasis in peripheral organs via adiponectin receptors. In spite of accumulating evidence that adiponectin has neuroprotective properties, the underlying role of adiponectin receptors has not been illuminated. Here, using gene therapy-mediated suppression with shRNA, we found that adiponectin receptor 1 (AdipoR1) suppression induces neurodegeneration as well as metabolic dysfunction. AdipoR1 knockdown mice exhibited increased body weight and abnormal plasma chemistry and also showed spatial learning and memory impairment in behavioural studies. Moreover, AdipoR1 suppression resulted in neurodegenerative phenotypes, diminished expression of the neuronal marker NeuN, and increased expression and activity of caspase 3. Furthermore, AD-like pathologies including insulin signalling dysfunction, abnormal protein aggregation and neuroinflammatory responses were highly exhibited in AdipoR1 knockdown groups, consistent with brain pathologies in ADPN knockout mice. Together, these results suggest that ADPN-AdipoR1 signalling has the potential to alleviate neurodegenerative diseases such as Alzheimer's diseases.


Adiponectin/genetics , Alzheimer Disease/genetics , Caspase 3/genetics , Memory Disorders/genetics , Receptors, Adiponectin/genetics , Adiponectin/metabolism , Alzheimer Disease/metabolism , Alzheimer Disease/physiopathology , Animals , Body Weight , Caspase 3/metabolism , DNA-Binding Proteins , Disease Models, Animal , Gene Expression Regulation , Insulin/metabolism , Male , Memory Disorders/metabolism , Memory Disorders/physiopathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Receptors, Adiponectin/antagonists & inhibitors , Receptors, Adiponectin/deficiency , Signal Transduction , Spatial Learning/physiology
11.
Mol Metab ; 6(3): 267-275, 2017 03.
Article En | MEDLINE | ID: mdl-28271033

OBJECTIVE: Adiponectin and the signaling induced by its cognate receptors, AdipoR1 and AdipoR2, have garnered attention for their ability to promote insulin sensitivity and oppose steatosis. Activation of these receptors promotes the deacylation of ceramide, a lipid metabolite that appears to play a causal role in impairing insulin signaling. METHODS: Here, we have developed transgenic mice that overexpress AdipoR1 or AdipoR2 under the inducible control of a tetracycline response element. These represent the first inducible genetic models that acutely manipulate adiponectin receptor signaling in adult mouse tissues, which allows us to directly assess AdipoR signaling on glucose and lipid metabolism. RESULTS: Overexpression of either adiponectin receptor isoform in the adipocyte or hepatocyte is sufficient to enhance ceramidase activity, whole body glucose metabolism, and hepatic insulin sensitivity, while opposing hepatic steatosis. Importantly, metabolic improvements fail to occur in an adiponectin knockout background. When challenged with a leptin-deficient genetic model of type 2 diabetes, AdipoR2 expression in adipose or liver is sufficient to reverse hyperglycemia and glucose intolerance. CONCLUSION: These observations reveal that adiponectin is critical for AdipoR-induced ceramidase activation which enhances hepatic glucose and lipid metabolism via rapidly acting "cross-talk" between liver and adipose tissue sphingolipids.


Ceramidases/metabolism , Receptors, Adiponectin/physiology , Adipocytes/metabolism , Adiponectin/genetics , Adipose Tissue/metabolism , Animals , Ceramidases/genetics , Ceramidases/physiology , Fatty Liver/metabolism , Glucose/metabolism , Glucose/physiology , Hepatocytes/metabolism , Homeostasis/genetics , Insulin/metabolism , Insulin Resistance/genetics , Leptin/metabolism , Lipid Metabolism/genetics , Lipid Metabolism/physiology , Lipids/physiology , Liver/metabolism , Mice , Mice, Transgenic , Piperidines/metabolism , Receptors, Adiponectin/antagonists & inhibitors , Receptors, Adiponectin/genetics
12.
J Mol Endocrinol ; 58(4): 155-165, 2017 05.
Article En | MEDLINE | ID: mdl-28219936

It has been reported that ischemic preconditioning (IPC) and adiponectin (APN) are cardioprotective in many cardiovascular disorders. However, whether APN mediates the effect of IPC on myocardial injury has not been elucidated. This study was conducted to investigate whether IPC affects myocardial ischemic injury by increasing APN expression. Male adult rats with cardiac knockdowns of APN and its receptors via intramyocardial small-interfering RNA injection were subjected to IPC and then myocardial infarction (MI) at 24 h after IPC. Globular APN (gAd) was injected at 10 min before MI. APN mRNA and protein levels in myocardium as well as the plasma APN concentration were markedly high at 6 and 12 h after IPC. IPC ameliorated myocardial injury as evidenced by improved cardiac functions and a reduced infarct size. Compared with the control MI group, rats in the IPC + MI group had elevated levels of left ventricular ejection fraction and fractional shortening and a smaller MI size (P < 0.05). However, the aforementioned protective effects were ameliorated in the absence of APN and APN receptors, followed by the inhibition of AMP-activated protein kinase (AMPK) phosphorylation, but reversed by gAd treatment in wild-type rats, and AMPK phosphorylation increased (P < 0.05). Overall, our results suggest that the cardioprotective effects of IPC are partially due to upregulation of APN and provide a further insight into IPC-mediated signaling effects.


Adiponectin/genetics , Ischemic Preconditioning, Myocardial , Myocardial Infarction/prevention & control , Myocardial Reperfusion Injury/prevention & control , Myocardium/metabolism , Receptors, Adiponectin/genetics , AMP-Activated Protein Kinases/genetics , AMP-Activated Protein Kinases/metabolism , Adiponectin/antagonists & inhibitors , Adiponectin/metabolism , Animals , Gene Expression Regulation , Heart Function Tests , Male , Myocardial Infarction/genetics , Myocardial Infarction/metabolism , Myocardial Infarction/pathology , Myocardial Reperfusion Injury/genetics , Myocardial Reperfusion Injury/metabolism , Myocardial Reperfusion Injury/pathology , Myocardium/pathology , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Rats , Rats, Sprague-Dawley , Receptors, Adiponectin/antagonists & inhibitors , Receptors, Adiponectin/metabolism , Signal Transduction , Stroke Volume/physiology
13.
J Cachexia Sarcopenia Muscle ; 8(3): 370-385, 2017 Jun.
Article En | MEDLINE | ID: mdl-27897419

BACKGROUND: Exercise train (ET) stimulates muscle response in pathological conditions, including aging. The molecular mechanisms by which exercise improves impaired adiponectin/adiponectin receptor 1 (AdipoR1)-related muscle actions associated with aging are poorly understood. Here we observed that in a senescence-accelerated mouse prone 10 (SAMP10) model, long-term ET modulated muscle-regenerative actions. METHODS: 25-week-old male SAMP10 mice were randomly assigned to the control and the ET (45 min/time, 3/week) groups for 4 months. Mice that were maintained in a sedentary condition served controls. RESULTS: ET ameliorated aging-related muscle changes in microstructure, mitochondria, and performance. The amounts of proteins or mRNAs for p-AMPKα, p-Akt, p-ERK1/2, p-mTOR, Bcl-XL, p-FoxO3, peroxisome proliferators-activated receptor-γ coactivator, adiponectin receptor1 (adpoR1), and cytochrome c oxidase-IV, and the numbers of CD34+ /integrin-α7+ muscle stem cells (MuSCs) and proliferating cells in the muscles and bone-marrow were enhanced by ET, whereas the levels of p-GSK-3α and gp91phox proteins and apoptotic cells were reduced by ET. The ET also resulted in increased levels of plasma adiponectin and the numbers of bone-marrow (BM)-derived circulating CD34+ /integrin-α7+ MuSCs and their functions. Integrin-α7+ MuSCs of exercised mice had improved changes of those beneficial molecules. These ET-mediated aged muscle benefits were diminished by adiponectin and AdipoR1 blocking as well as AMPK inhibition. Finally, recombinant mouse adiponectin enhanced AMPK and mTOR phosphorylations in BM-derived integrin-α7+ cells. CONCLUSIONS: These findings suggest that ET can improve aging-related impairments of BM-derived MuSC regenerative capacity and muscle metabolic alterations via an AMPK-dependent mechanism that is mediated by an adiponectin/AdipoR1 axis in SAMP10 mice.


Adiponectin/metabolism , Muscles/physiology , Myoblasts/metabolism , Physical Conditioning, Animal , Receptors, Adiponectin/metabolism , Regeneration , Animals , Biomarkers , Body Weight , Cell Movement , Cytokines/blood , Lipids/blood , MAP Kinase Signaling System/drug effects , Male , Mice , Mitochondria, Muscle/drug effects , Mitochondria, Muscle/metabolism , Muscle Strength , Muscle, Skeletal/physiology , Receptors, Adiponectin/antagonists & inhibitors , Signal Transduction/drug effects
14.
Acta Biochim Biophys Sin (Shanghai) ; 48(6): 509-19, 2016 Jun.
Article En | MEDLINE | ID: mdl-27125977

The induced lipogenesis and its regulation in C2C12 myoblasts remain largely unclear. Here, we found that the cocktail method could significantly induce lipogenesis through regulating lipid metabolic genes and Erk1/2 phosphorylation in myoblasts. Meanwhile, the expression and secretion of CTRP6 were increased during ectopic lipogenesis. Moreover, CTRP6 knockdown down-regulated the levels of lipogenic genes and phosphorylated Erk1/2 (p-Erk1/2) in the early lipogenic stage, whereas up-regulated p-Erk1/2 in the terminal differentiation. Interestingly, the effect of CTRP6 siRNA was attenuated by U0126 (a special p-Erk1/2 inhibitor) in myoblasts. Furthermore, AdipoR1, not AdipoR2, was first identified as a receptor of CTRP6 during the process of mitotic clonal expansion. Collectively, we suggest that CTRP6 mediates the ectopic lipogenesis through AdipoR1/Erk/PPARγ signaling pathway in myoblasts. Our findings will shed light on the novel biological function of CTRP6 during myoblast lipogenesis and provide a hopeful direction of improving meat quality of domestic animal by lipogenic regulation in skeletal muscle myoblasts.


Adipokines/metabolism , Lipogenesis/physiology , Myoblasts, Skeletal/metabolism , Adipokines/antagonists & inhibitors , Adipokines/genetics , Animals , Cell Differentiation , Cell Line , Gene Knockdown Techniques , Lipogenesis/genetics , MAP Kinase Signaling System , Mice , Models, Biological , Myoblasts, Skeletal/cytology , PPAR gamma/metabolism , RNA, Small Interfering/genetics , Receptors, Adiponectin/antagonists & inhibitors , Receptors, Adiponectin/genetics , Receptors, Adiponectin/metabolism , Signal Transduction
15.
Cell Physiol Biochem ; 38(3): 909-25, 2016.
Article En | MEDLINE | ID: mdl-26919192

BACKGROUND/AIMS: Poor viability of transplanted mesenchymal stem cells (MSCs) within the ischemic heart limits their therapeutic potential for cardiac repair. Globular adiponectin (gAPN) exerts anti-apoptotic effects on several types of stem cells. Herein, we investigated the effect of gAPN on the MSCs against apoptosis induced by hypoxia and serum deprivation (H/SD). METHODS: MSCs exposed to H/SD conditions were treated with different concentrations of gAPN. To identify the main type of receptor, MSCs were transfected with siRNA targeting adiponectin receptor 1 or 2 (AdipoR1 or AdipoR2). To elucidate the downstream pathway, MSCs were pre-incubated with AMPK inhibitor Compound C. Apoptosis, caspase-3 activity and mitochondrial membrane potential were evaluated. RESULTS: H/SD-induced MSCs apoptosis and caspase-3 activation were attenuated by gAPN in a concentration-dependent manner. gAPN increased Bcl-2 and decreased Bax expressions. The loss of mitochondrial membrane potential induced by H/SD was also abolished by gAPN. The protective effect of gAPN was significantly attenuated after the knockdown of AdipoR1 rather than AdipoR2. Moreover, Compound C partly suppressed the anti-apoptotic effect of gAPN. CONCLUSIONS: gAPN inhibits H/SD-induced apoptosis in MSCs via AdipoR1-mediated pathway, possibly linked to the activation of AMPK. gAPN may be a novel survival factor for MSCs in the ischemic engraftment environment.


Adiponectin/pharmacology , Culture Media, Serum-Free/pharmacology , Mesenchymal Stem Cells/cytology , Receptors, Adiponectin/metabolism , Animals , Apoptosis/drug effects , Caspase 3/metabolism , Cell Hypoxia/drug effects , Cell Survival/drug effects , Cells, Cultured , Gene Expression Regulation/drug effects , Male , Mesenchymal Stem Cells/metabolism , Pyrazoles/pharmacology , Pyrimidines/pharmacology , RNA, Small Interfering/pharmacology , Rats , Receptors, Adiponectin/antagonists & inhibitors
16.
Curr Mol Med ; 15(10): 905-31, 2015.
Article En | MEDLINE | ID: mdl-26592248

Atherosclerosis and its complications represent the leading death cause worldwide, despite many therapeutic developments. Atherosclerosis is a complex, multistage disease whereby perturbed lipid metabolism leads to cholesterol accumulation into the vascular walls and plaque formation. Generation of apoE-/- and LDLR-/- atherosclerosis mouse models opened the avenue for investigating the mechanisms of action for specific molecules. We focus herein on the involvement of non-lipoprotein receptors in atherogenesis, as revealed by their total or site-specific ablation in the aforementioned murine models. The receptors reviewed span a broad range, from molecules related to lipid metabolism (adiponectin receptors) to molecules whose connection with atherogenesis is less obvious (cannabinoid receptors). We also outline cross-transplantation studies which allowed uncoupling the lipid modulating effects from the inflammatory ones. For certain receptors, since knockouts were unavailable, pharmacological data are presented instead. We emphasize the contribution of the receptors to the pathology, based on functional criteria, such as oxidative stress, immune response, inflammation, angiogenesis. Controversial aspects regarding the pro- or anti- atherogenic activity of some receptors are highlighted. We assume these discrepancies are due to the experimental setup, animal models used, tissue-specific action, various isoforms analyzed, divergent signaling or cross-talk between metabolic and immune pathways. Understanding the influences of cellular receptors in the progression of atherosclerosis allows their modulation towards an antiatherogenic phenotype. The experimental studies in animal models were in some cases successfully extrapolated to humans leading to atheroma reduction, and we expect this to occur even to a greater extent, based on the newest achievements.


Anticholesteremic Agents/therapeutic use , Atherosclerosis/drug therapy , Molecular Targeted Therapy/methods , Plaque, Atherosclerotic/drug therapy , Animals , Apolipoproteins E/deficiency , Apolipoproteins E/genetics , Atherosclerosis/genetics , Atherosclerosis/metabolism , Atherosclerosis/pathology , Disease Models, Animal , Gene Expression Regulation , Humans , Lipid Metabolism/drug effects , Mice , Mice, Knockout , Oxidative Stress/drug effects , Plaque, Atherosclerotic/genetics , Plaque, Atherosclerotic/metabolism , Plaque, Atherosclerotic/pathology , Receptors, Adiponectin/antagonists & inhibitors , Receptors, Adiponectin/genetics , Receptors, Adiponectin/metabolism , Receptors, Cannabinoid/genetics , Receptors, Cannabinoid/metabolism , Receptors, LDL/deficiency , Receptors, LDL/genetics , Receptors, Leptin/agonists , Receptors, Leptin/genetics , Receptors, Leptin/metabolism , Receptors, Nicotinic/genetics , Receptors, Nicotinic/metabolism , Signal Transduction
17.
Sci Rep ; 5: 17310, 2015 Nov 27.
Article En | MEDLINE | ID: mdl-26611106

Recent studies have demonstrated that adiponectin (APN) attenuates cerebral ischemic/reperfusion via globular adiponectin (gAD). However, the therapeutic role of gAD in cerebral ischemic injury in type 1 diabetes mellitus (T1DM) remains unclear. Our results showed that gAD improved neurological scores and reduced the infarct volumes in the 8-week T1DM (T1DM-8W) mice, but not in the 2-week T1DM (T1DM-2W) mice. Moreover, the ischemic penumbra APN levels increased and peaked in T1DM-2W mice, and reduced to normal in T1DM-8W mice, while the APN receptor 1 (AdipoR1) expression change was the opposite. Administration of rosiglitazone in T1DM-2W mice up-regulated the expression of AdipoR1 and restored the neuroprotection of gAD, while intracerebroventricular injection of AdipoR1 small interfering RNA (siRNA) in T1DM-8W mice reversed it. Furthermore, the expression of p-PERK, p-IRE1 and GRP78 were increased whereas the expressions of CHOP and cleaved caspase-12 as well as the number of apoptotic neurons were decreased after gAD treatment in T1DM-8W mice. These beneficial effects of gAD were reversed by pretreatment with AdipoR1 siRNA. These results demonstrated a dynamic dysfunction of APN/AdipoR1 accompanying T1DM progression. Interventions bolstering AdipoR1 expression during early stages and gAD supplementation during advanced stages may potentially reduce the cerebral ischemic injury in diabetic patients.


Adiponectin/pharmacology , Brain Ischemia/drug therapy , Diabetes Mellitus, Experimental/drug therapy , Receptors, Adiponectin/genetics , Reperfusion Injury/drug therapy , Adiponectin/genetics , Adiponectin/metabolism , Animals , Apoptosis/drug effects , Brain Ischemia/complications , Brain Ischemia/genetics , Brain Ischemia/metabolism , Caspase 12/genetics , Caspase 12/metabolism , Diabetes Mellitus, Experimental/complications , Diabetes Mellitus, Experimental/genetics , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Type 1/drug therapy , Diabetes Mellitus, Type 1/genetics , Diabetes Mellitus, Type 1/metabolism , Diabetes Mellitus, Type 1/pathology , Endoplasmic Reticulum Chaperone BiP , Gene Expression Regulation , Heat-Shock Proteins/genetics , Heat-Shock Proteins/metabolism , Humans , Hypoglycemic Agents/pharmacology , Injections, Intraventricular , Male , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Mice, Inbred C57BL , Neurons/drug effects , Neurons/metabolism , Neurons/pathology , Neuroprotective Agents/pharmacology , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , RNA, Small Interfering/administration & dosage , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Receptors, Adiponectin/agonists , Receptors, Adiponectin/antagonists & inhibitors , Receptors, Adiponectin/metabolism , Reperfusion Injury/complications , Reperfusion Injury/genetics , Reperfusion Injury/metabolism , Rosiglitazone , Signal Transduction , Thiazolidinediones/pharmacology , Transcription Factor CHOP/genetics , Transcription Factor CHOP/metabolism , eIF-2 Kinase/genetics , eIF-2 Kinase/metabolism
18.
Methods Mol Biol ; 1081: 195-209, 2013.
Article En | MEDLINE | ID: mdl-24014441

Receptor-ligand interactions represent one of the most basic processes in biological systems. Receptor activation and deactivation induce or prevent a series of downstream signaling events that ultimately result in normal or abnormal cellular functions. Contemporary biology is in continuous search for the identification of novel receptors and their ligands. The adipose tissue participates in the regulation of energy homeostasis as an important endocrine organ that secretes a number of biologically active adipokines, including leptin and adiponectin. A recent discovery and design process for leptin and adiponectin receptor response modifier peptides can be generalized to a series of transmembrane receptor ligands. A family of 11-13 amino acid residue-long leptin receptor (ObR) agonists has been identified by analyzing the effect of peptides corresponding to the three presumed active sites of leptin on the growth of leptin-responsive cancer cells. In the case of adiponectin, overlapping peptides were walked across the entire globular domain of the protein to identify the active site and derive adiponectin receptor (AdipoR) agonist peptides. In both sets, native residues were replaced by nonnatural analogs to improve the pharmacological properties including stability, efficacy and targeting. Later the ObR analogs were converted into true ObR antagonists that show antagonist-agonist selectivity of 1,000 in cellular assays. The design process of ObR antagonists included shortening of the peptide length and incorporating additional nonnatural residues. Here I take a look into this receptor agonist and antagonist discovery process from a practical point of view.


Receptors, Adiponectin/agonists , Receptors, Adiponectin/antagonists & inhibitors , Adiponectin/chemistry , Adiponectin/metabolism , Adiponectin/pharmacology , Amino Acid Sequence , Animals , Binding Sites , Cell Line , Humans , Inhibitory Concentration 50 , Leptin/chemistry , Leptin/metabolism , Leptin/pharmacology , Ligands , Peptide Fragments/chemistry , Peptide Fragments/metabolism , Peptide Fragments/pharmacology , Receptors, Adiponectin/chemistry , Receptors, Adiponectin/metabolism
19.
PLoS One ; 8(7): e68641, 2013.
Article En | MEDLINE | ID: mdl-23874700

Adipokines secreted from adipose tissue are key regulators of metabolism in animals. Adiponectin, one of the adipokines, modulates pancreatic beta cell function to maintain energy homeostasis. Recently, significant conservation between Drosophila melanogaster and mammalian metabolism has been discovered. Drosophila insulin like peptides (Dilps) regulate energy metabolism similarly to mammalian insulin. However, in Drosophila, the regulatory mechanism of insulin producing cells (IPCs) by adipokine signaling is largely unknown. Here, we describe the discovery of the Drosophila adiponectin receptor and its function in IPCs. Drosophila adiponectin receptor (dAdipoR) has high homology with the human adiponectin receptor 1. The dAdipoR antibody staining revealed that dAdipoR was expressed in IPCs of larval and adult brains. IPC- specific dAdipoR inhibition (Dilp2>dAdipoR-Ri) showed the increased sugar level in the hemolymph and the elevated triglyceride level in whole body. Dilps mRNA levels in the Dilp2>dAdipoR-Ri flies were similar with those of controls. However, in the Dilp2>dAdipoR-Ri flies, Dilp2 protein was accumulated in IPCs, the level of circulating Dilp2 was decreased, and insulin signaling was reduced in the fat body. In ex vivo fly brain culture with the human adiponectin, Dilp2 was secreted from IPCs. These results indicate that adiponectin receptor in insulin producing cells regulates insulin secretion and controls glucose and lipid metabolism in Drosophila melanogaster. This study demonstrates a new adipokine signaling in Drosophila and provides insights for the mammalian adiponectin receptor function in pancreatic beta cells, which could be useful for therapeutic application.


Brain/cytology , Drosophila Proteins/metabolism , Drosophila melanogaster/cytology , Drosophila melanogaster/metabolism , Glucose/metabolism , Insulin/metabolism , Lipid Metabolism , Receptors, Adiponectin/metabolism , Adiponectin/pharmacology , Amino Acid Sequence , Animals , Drosophila Proteins/antagonists & inhibitors , Drosophila Proteins/chemistry , Drosophila melanogaster/drug effects , Female , Gene Knockdown Techniques , Humans , Insulin Secretion , Larva/cytology , Larva/drug effects , Larva/metabolism , Lipid Metabolism/drug effects , Molecular Sequence Data , Phenotype , Receptors, Adiponectin/antagonists & inhibitors , Receptors, Adiponectin/chemistry , Sequence Homology, Amino Acid , Signal Transduction/drug effects
20.
FEBS J ; 280(16): 3920-7, 2013 Aug.
Article En | MEDLINE | ID: mdl-23773620

Adiponectin is an adipocyte hormone that is predominantly secreted by adipocytes, and has important roles in glucose and lipid homeostasis. Recent studies have shown that adiponectin is also involved in the regulation of many endocrine organs, such as the ovary, adrenal gland, and pituitary. However, its biological role in male testes is largely unexplored. The present findings demonstrate the presence of adeponectin receptors (adiponectin receptor 1 and adiponectin receptor 2) in TM3 cells derived from mouse Leydig cells. Proinflammatory cytokine treatment significantly downregulated mRNA and protein levels of adiponectin receptor 1 and adiponectin receptor 2. However, adiponectin pretreatment successfully inhibited the signaling pathway mediated by proinflammatory cytokines. At the molecular level, we provide compelling evidence that adeponectin achieves this by suppressing nuclear factor-κB activation through promotion of AMP-activated protein kinase phosphorylation. Thus, our data clearly indicate that adiponectin plays a protective role in Leydig cells through its anti-inflammatory actions.


Adiponectin/metabolism , Cytokines/antagonists & inhibitors , Down-Regulation , Leydig Cells/metabolism , NF-kappa B/antagonists & inhibitors , Receptors, Adiponectin/metabolism , Signal Transduction , 3-Hydroxysteroid Dehydrogenases/antagonists & inhibitors , 3-Hydroxysteroid Dehydrogenases/chemistry , 3-Hydroxysteroid Dehydrogenases/genetics , 3-Hydroxysteroid Dehydrogenases/metabolism , AMP-Activated Protein Kinases/metabolism , Animals , Cell Line , Cyclooxygenase 2/chemistry , Cyclooxygenase 2/genetics , Cyclooxygenase 2/metabolism , Cytokines/metabolism , Gene Silencing , Leydig Cells/immunology , Male , Mice , Mice, Inbred C57BL , NF-kappa B/metabolism , Nitric Oxide Synthase Type II/antagonists & inhibitors , Nitric Oxide Synthase Type II/chemistry , Nitric Oxide Synthase Type II/genetics , Nitric Oxide Synthase Type II/metabolism , Phosphoproteins/agonists , Phosphoproteins/antagonists & inhibitors , Phosphoproteins/genetics , Phosphoproteins/metabolism , Phosphorylation , Protein Processing, Post-Translational , Receptors, Adiponectin/antagonists & inhibitors , Receptors, Adiponectin/genetics , Up-Regulation
...