Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
Cell Death Dis ; 12(5): 458, 2021 05 07.
Article in English | MEDLINE | ID: mdl-33963174

ABSTRACT

Lipid metabolism-related gene mutations can cause retinitis pigmentosa, a currently untreatable blinding disease resulting from progressive neurodegeneration of the retina. Here, we demonstrated the influence of adiponectin receptor 1 (ADIPOR1) deficiency in retinal neurodegeneration using Adipor1 knockout (KO) mice. Adipor1 mRNA was observed to be expressed in photoreceptors, predominately within the photoreceptor inner segment (PIS), and increased after birth during the development of the photoreceptor outer segments (POSs) where photons are received by the visual pigment, rhodopsin. At 3 weeks of age, visual function impairment, specifically photoreceptor dysfunction, as recorded by electroretinography (ERG), was evident in homozygous, but not heterozygous, Adipor1 KO mice. However, although photoreceptor loss was evident at 3 weeks of age and progressed until 10 weeks, the level of visual dysfunction was already substantial by 3 weeks, after which it was retained until 10 weeks of age. The rhodopsin mRNA levels had already decreased at 3 weeks, suggesting that reduced rhodopsin may have contributed to early visual loss. Moreover, inflammation and oxidative stress were induced in homozygous KO retinas. Prior to observation of photoreceptor loss via optical microscopy, electron microscopy revealed that POSs were present; however, they were misaligned and their lipid composition, including docosahexaenoic acid (DHA), which is critical in forming POSs, was impaired in the retina. Importantly, the expression of Elovl2, an elongase of very long chain fatty acids expressed in the PIS, was significantly reduced, and lipogenic genes, which are induced under conditions of reduced endogenous DHA synthesis, were increased in homozygous KO mice. The causal relationship between ADIPOR1 deficiency and Elovl2 repression, together with upregulation of lipogenic genes, was confirmed in vitro. Therefore, ADIPOR1 in the retina appears to be indispensable for ELOVL2 induction, which is likely required to supply sufficient DHA for appropriate photoreceptor function and survival.


Subject(s)
Fatty Acid Elongases/metabolism , Lipid Metabolism/genetics , Photoreceptor Cells, Vertebrate/metabolism , Receptors, Adiponectin/deficiency , Vision Disorders/metabolism , Animals , Docosahexaenoic Acids/metabolism , Mice , Mice, Knockout , Transfection
2.
Sci Rep ; 8(1): 14339, 2018 09 25.
Article in English | MEDLINE | ID: mdl-30254279

ABSTRACT

The knockout (KO) of the adiponectin receptor 1 (AdipoR1) gene causes retinal degeneration. Here we report that ADIPOR1 protein is primarily found in the eye and brain with little expression in other tissues. Further analysis of AdipoR1 KO mice revealed that these animals exhibit early visual system abnormalities and are depleted of RHODOPSIN prior to pronounced photoreceptor death. A KO of AdipoR1 post-development either in photoreceptors or the retinal pigment epithelium (RPE) resulted in decreased expression of retinal proteins, establishing a role for ADIPOR1 in supporting vision in adulthood. Subsequent analysis of the Mfrprd6 mouse retina demonstrated that these mice are lacking ADIPOR1 in their RPE layer alone, suggesting that loss of ADIPOR1 drives retinal degeneration in this model. Moreover, we found elevated levels of IRBP in both the AdipoR1 KO and the Mfrprd6 models. The spatial distribution of IRBP was also abnormal. This dysregulation of IRBP hypothesizes a role for ADIPOR1 in retinoid metabolism.


Subject(s)
Gene Expression Regulation , Gene Knockout Techniques , Receptors, Adiponectin/deficiency , Receptors, Adiponectin/metabolism , Retinal Pigment Epithelium/metabolism , Vision, Ocular , Animals , Eye Proteins/metabolism , Humans , Mice , Receptors, Adiponectin/genetics , Retinoids/metabolism , Retinol-Binding Proteins/metabolism
3.
Sci Rep ; 7(1): 12435, 2017 09 29.
Article in English | MEDLINE | ID: mdl-28963462

ABSTRACT

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.


Subject(s)
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
4.
Sci Rep ; 7: 43771, 2017 03 03.
Article in English | MEDLINE | ID: mdl-28256562

ABSTRACT

The incidence of benign prostatic hyperplasia (BPH) is increasing among obese individuals, but few studies have fully explained the underlying mechanisms. We aimed to elucidate the relationship between obesity and BPH. Herein, we show that in prostatic epithelial and stromal cells, adiponectin exerts multifunctional effects including anti-proliferation, blocking of G1/S-phase progression and the promotion of apoptosis via inhibiting the MEK-ERK-p90RSK axis. Furthermore, we found that a high-fat diet (HFD) led to adiponectin deficiency and microscopic BPH in a mouse model of obesity. And an adiponectin supplement protected the obese mice from microscopic BPH. The present study provides evidence that adiponectin is a protective regulator in the development and progression of BPH and that adiponectin deficiency causally links BPH with obesity.


Subject(s)
Adiponectin/pharmacology , Obesity/prevention & control , Prostate/drug effects , Prostatic Hyperplasia/prevention & control , Animals , Cell Cycle/drug effects , Cell Line , Cell Proliferation/drug effects , Diet, High-Fat/adverse effects , Disease Progression , Humans , Male , Mice, Inbred C57BL , Obesity/etiology , Obesity/metabolism , Prostate/metabolism , Prostate/pathology , Prostatic Hyperplasia/etiology , Prostatic Hyperplasia/metabolism , RNA Interference , Receptors, Adiponectin/deficiency , Receptors, Adiponectin/genetics , Receptors, Adiponectin/metabolism
5.
Zhonghua Nan Ke Xue ; 23(11): 975-981, 2017 Nov.
Article in Chinese | MEDLINE | ID: mdl-29738161

ABSTRACT

OBJECTIVE: To investigate the relationship of the serum adiponectin level with the development of prostate cancer (PCa) and its influence on the biological characteristics of PCa cells. METHODS: This retrospective case-control study included 45 cases of PCa and 50 non-PCa controls. We analyzed the relationship of the serum adiponectin level with the development of PCa. Using the lentiviral vector, we constructed a stably transfected cell line with adiponectin receptor deficiency, treated the PCa cells with adiponectin, and determined the influence of the adiponectin level on the biological behavior of the PC-3 cells by CCK-8 assay, flow cytometry, tube formation assay and scratch wound healing assay. RESULTS: The level of serum adiponectin was decreased in the PCa patients as compared with the controls and it was negatively correlated with the Gleason scores. Adiponectin suppressed the proliferation, migration and tube formation of the PC-3 cells and inhibited them from transforming into S-stage cells. In the condition of adiponectin receptor deficiency, the PC-3 cells exhibited even greater abilities of proliferation, migration and pro-angiogenesis. CONCLUSIONS: The decreased level of the serum adiponectin or deficiency of adiponectin receptor may contribute to the development of prostate cancer.


Subject(s)
Adiponectin/blood , Prostatic Neoplasms/blood , Prostatic Neoplasms/etiology , Case-Control Studies , Cell Line, Tumor , Cell Proliferation , Humans , Male , Neoplasm Grading , Receptors, Adiponectin/deficiency , Retrospective Studies , Transfection
6.
Am J Physiol Heart Circ Physiol ; 310(9): H1164-75, 2016 05 01.
Article in English | MEDLINE | ID: mdl-26921438

ABSTRACT

Skeletal muscle metabolic changes are common in patients with chronic heart failure (HF). Previously, we demonstrated a functional skeletal muscle adiponectin resistance in HF patients with reduced left ventricular ejection fraction (HFrEF). We aimed to examine the impact of adiponectin receptor 1 (AdipoR1) deficiency and TNF-α treatment on adiponectin signaling, proliferative capacity, myogenic differentiation, and mitochondrial biogenesis in primary human skeletal muscle cells. Primary cultures of myoblasts and myotubes were initiated from the musculus vastus lateralis of 10 HFrEF patients (left ventricular ejection fraction; 31.30 ± 2.89%) and 10 age- and gender-matched healthy controls. Healthy control cultures were transfected with siAdipoR1 and/or exposed to TNF-α (10 ng/ml; 72 h). Primary cultures from HFrEF patients preserved the features of adiponectin resistance in vivo. AdipoR1 mRNA was negatively correlated with time to reach maximal cell index (r = -0.7319, P = 0.003). SiRNA-mediated AdipoR1 silencing reduced pAMPK (P < 0.01), AMPK activation (P = 0.046), and myoblast proliferation rate (xCELLigence Real-Time Cellular Analysis; P < 0.0001). Moreover, TNF-α decreased the mRNA expression of genes involved in glucose (APPL1, P = 0.0002; AMPK, P = 0.021), lipid (PPARα, P = 0.025; ACADM, P = 0.003), and mitochondrial (FOXO3, P = 0.018) metabolism, impaired myogenesis (MyoD1, P = 0.053; myogenin, P = 0.048) and polarized cytokine secretion toward a growth-promoting phenotype (IL-10, IL-1ß, IFN-γ, P < 0.05 for all; Meso Scale Discovery Technology). Major features of adiponectin resistance are retained in primary cultures from the skeletal muscle of HFrEF patients. In addition, our results suggest that an increased inflammatory constitution contributes to adiponectin resistance and confers alterations in skeletal muscle differentiation, growth, and function.


Subject(s)
Adiponectin/metabolism , Heart Failure/metabolism , Mitochondria, Muscle/drug effects , Muscle Development/drug effects , Muscle Fibers, Skeletal/drug effects , Organelle Biogenesis , Quadriceps Muscle/drug effects , Receptors, Adiponectin/metabolism , Signal Transduction/drug effects , Tumor Necrosis Factor-alpha/pharmacology , Case-Control Studies , Cell Proliferation/drug effects , Cells, Cultured , Female , Gene Expression Regulation , Heart Failure/genetics , Heart Failure/pathology , Humans , Inflammation/genetics , Inflammation/metabolism , Inflammation/pathology , Male , Middle Aged , Mitochondria, Muscle/metabolism , Mitochondria, Muscle/pathology , Muscle Fibers, Skeletal/metabolism , Muscle Fibers, Skeletal/pathology , Phenotype , Primary Cell Culture , Quadriceps Muscle/metabolism , Quadriceps Muscle/pathology , RNA Interference , Receptors, Adiponectin/deficiency , Receptors, Adiponectin/genetics , Time Factors , Transfection
7.
PLoS One ; 8(11): e80330, 2013.
Article in English | MEDLINE | ID: mdl-24324556

ABSTRACT

Adiponectin has been shown to have beneficial cardiovascular effects and to signal through the adiponectin receptors, AdipoR1 and AdipoR2. The original aim of this study was to investigate the effect of combined AdipoR1 and AdipoR2 deficiency (AdipoR1(-/-)AdipoR2(-/-)) on atherosclerosis. However, we made the interesting observation that AdipoR1(-/-) AdipoR2(-/-) leads to embryonic lethality demonstrating the critical importance of the adiponectin signalling system during development. We then investigated the effect of AdipoR2-ablation on the progression of atherosclerosis in apolipoprotein E deficient (ApoE(-/-)) mice. AdipoR2(-/-)ApoE(-/-) mice fed an atherogenic diet had decreased plaque area in the brachiocephalic artery compared with AdipoR2(+/+) ApoE(-/-) littermate controls as visualized in vivo using an ultrasound biomicroscope and confirmed by histological analyses. The decreased plaque area in the brachiocephalic artery could not be explained by plasma cholesterol levels or inflammatory status. However, accumulation of neutral lipids was decreased in peritoneal macrophages from AdipoR2(-/-)ApoE(-/-) mice after incubation with oxidized LDL. This effect was associated with lower CD36 and higher ABCA1 mRNA levels in peritoneal macrophages from AdipoR2(-/-)ApoE(-/-) mice compared with AdipoR2(+/+)ApoE(-/-) controls after incubation with oxidized LDL. In summary, we show that adiponectin receptors are crucial during embryonic development and that AdipoR2-deficiency slows down the progression of atherosclerosis in the brachiocephalic artery of ApoE-deficient mice.


Subject(s)
Apolipoproteins E/deficiency , Apolipoproteins E/metabolism , Atherosclerosis/metabolism , Receptors, Adiponectin/metabolism , Animals , Apolipoproteins E/genetics , Atherosclerosis/genetics , Blotting, Western , Body Weight/genetics , Body Weight/physiology , Female , Genotype , Heterozygote , Macrophages/metabolism , Male , Mice , Mice, Knockout , Receptors, Adiponectin/deficiency , Receptors, Adiponectin/genetics
8.
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
9.
J Invest Dermatol ; 133(3): 812-821, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23096717

ABSTRACT

Skin wounds comprise a serious medical issue for which few pharmacological interventions are available. Moreover, the inflammatory, angiogenic, and proliferative facets of a typical response to a wound each have broader relevance in other pathological conditions. Here we describe a genomics-driven approach to identify secreted proteins that modulate wound healing in a mouse ear punch model. We show that adiponectin, when injected into the wound edge, accelerates wound healing. Notably, adiponectin injection causes upregulation of keratin gene transcripts within hours of treatment, and subsequently promotes collagen organization, formation of pilosebaceous units, and proliferation of cells in the basal epithelial cell layer and pilosebaceous units of healing tissue. The globular domain of adiponectin is sufficient to mediate accelerated dorsal skin wound closure, and the effects are lost in mice that are homozygous null for the adiponectin receptor 1 gene. These findings extend recent observations of a protective role of adiponectin in other tissue injury settings, suggest modulation of AdipoR1 for the clinical management of wounds, and demonstrate a new approach to the identification of regulators of a wound healing response.


Subject(s)
Adiponectin/physiology , Skin/injuries , Skin/physiopathology , Wound Healing/physiology , Adiponectin/pharmacology , Animals , Collagen/metabolism , Keratins/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Models, Animal , Receptors, Adiponectin/deficiency , Receptors, Adiponectin/genetics , Receptors, Adiponectin/physiology , Skin/metabolism , Up-Regulation/drug effects , Wound Healing/drug effects
10.
Brain Res ; 1423: 1-9, 2011 Nov 14.
Article in English | MEDLINE | ID: mdl-22000082

ABSTRACT

Adiponectin can act in the brain to increase energy expenditure and reduce body weight by mechanisms not entirely understood. We found that adiponectin type 1 and type 2 receptors (AdipoR1 and AdipoR2) are expressed in warm sensitive neurons of the hypothalamic preoptic area (POA) which play a critical role in the regulation of core body temperature (CBT) and energy balance. Thus, we tested the ability of adiponectin to influence CBT in wild-type mice and in mice deficient for AdipoR1 or AdipoR2. Local injection of adiponectin into the POA induced prolonged elevation of core body temperature and decreased respiratory exchange ratio (RER) indicating that increased energy expenditure is associated with increased oxidation of fat over carbohydrates. In AdipoR1 deficient mice, the ability of adiponectin to raise CBT was significantly blunted and its ability to decrease RER was completely lost. In AdipoR2 deficient mice, adiponectin had only diminished hyperthermic effects but reduced RER similarly to wild type mice. These results indicate that adiponectin can contribute to energy homeostasis by regulating CBT by direct actions on AdipoR1 and R2 in the POA.


Subject(s)
Adiponectin/pharmacology , Body Temperature/drug effects , Preoptic Area/cytology , Receptors, Adiponectin/metabolism , Sensory Receptor Cells/physiology , Analysis of Variance , Animals , Calorimetry, Indirect , Energy Metabolism/drug effects , Gene Expression Regulation/drug effects , Gene Expression Regulation/genetics , In Vitro Techniques , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptors, Adiponectin/deficiency , Sensory Receptor Cells/drug effects , Telemetry , Thermosensing/drug effects , Thermosensing/physiology
11.
PLoS One ; 6(4): e19143, 2011 Apr 28.
Article in English | MEDLINE | ID: mdl-21552570

ABSTRACT

Cardiomyocyte apoptosis is an important remodeling event contributing to heart failure and adiponectin may mediate cardioprotective effects at least in part via attenuating apoptosis. Here we used hypoxia-reoxygenation (H/R) induced apoptosis in H9c2 cells to examine the effect of adiponectin and cellular mechanisms of action. We first used TUNEL labeling in combination with laser scanning cytometry to demonstrate that adiponectin prevented H/R-induced DNA fragmentation. The anti-apoptotic effect of adiponectin was also verified via attenuation of H/R-induced phosphatidylserine exposure using annexin V binding. H/R-induced apoptosis via the mitochondrial-mediated intrinsic pathway of apoptosis as assessed by cytochrome c release into cytosol and caspase-3 activation, both of which were attenuated by adiponectin. Mechanistically, we demonstrated that adiponectin enhanced anti-oxidative potential in these cells which led to attenuation of the increase in intracellular reactive oxygen species (ROS) caused by H/R. To further address the mechanism of adiponctins anti-apoptotic effects we used siRNA to efficiently knockdown adiponectin receptor (AdipoR1) expression and found that this attenuated the protective effects of adiponectin on ROS production and caspase 3 activity. Knockdown of APPL1, an important intracellular binding partner for AdipoR, also significantly reduced the ability of adiponectin to prevent H/R-induced ROS generation and caspase 3 activity. In summary, H/R-induced ROS generation and activation of the intrinsic apoptotic pathway was prevented by adiponectin via AdipoR1/APPL1 signaling and increased anti-oxidant potential.


Subject(s)
Adiponectin/pharmacology , Apoptosis/drug effects , Carrier Proteins/metabolism , Nerve Tissue Proteins/metabolism , Oxygen/metabolism , Receptors, Adiponectin/metabolism , Adaptor Proteins, Signal Transducing , Animals , Annexin A5/metabolism , Antioxidants/metabolism , Base Sequence , Carrier Proteins/genetics , Caspase 3/metabolism , Cell Hypoxia/drug effects , Cell Line , Cytochromes c/metabolism , DNA Fragmentation/drug effects , Gene Knockdown Techniques , Mitochondria/drug effects , Mitochondria/metabolism , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Nerve Tissue Proteins/deficiency , Nerve Tissue Proteins/genetics , RNA, Small Interfering/genetics , Rats , Reactive Oxygen Species/metabolism , Receptors, Adiponectin/deficiency , Receptors, Adiponectin/genetics , Reperfusion Injury/pathology
12.
Nature ; 464(7293): 1313-9, 2010 Apr 29.
Article in English | MEDLINE | ID: mdl-20357764

ABSTRACT

Adiponectin is an anti-diabetic adipokine. Its receptors possess a seven-transmembrane topology with the amino terminus located intracellularly, which is the opposite of G-protein-coupled receptors. Here we provide evidence that adiponectin induces extracellular Ca(2+) influx by adiponectin receptor 1 (AdipoR1), which was necessary for subsequent activation of Ca(2+)/calmodulin-dependent protein kinase kinase beta (CaMKKbeta), AMPK and SIRT1, increased expression and decreased acetylation of peroxisome proliferator-activated receptor gamma coactivator-1alpha (PGC-1alpha), and increased mitochondria in myocytes. Moreover, muscle-specific disruption of AdipoR1 suppressed the adiponectin-mediated increase in intracellular Ca(2+) concentration, and decreased the activation of CaMKK, AMPK and SIRT1 by adiponectin. Suppression of AdipoR1 also resulted in decreased PGC-1alpha expression and deacetylation, decreased mitochondrial content and enzymes, decreased oxidative type I myofibres, and decreased oxidative stress-detoxifying enzymes in skeletal muscle, which were associated with insulin resistance and decreased exercise endurance. Decreased levels of adiponectin and AdipoR1 in obesity may have causal roles in mitochondrial dysfunction and insulin resistance seen in diabetes.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Adiponectin/metabolism , Calcium/metabolism , Mitochondria/metabolism , Receptors, Adiponectin/metabolism , Sirtuin 1/metabolism , Trans-Activators/metabolism , Animals , Calcium Signaling , Calcium-Calmodulin-Dependent Protein Kinase Kinase/metabolism , Cell Line , Glucose/metabolism , Homeostasis , Insulin/metabolism , Insulin Resistance , Mice , Muscle Cells/cytology , Muscle Cells/metabolism , Muscle, Skeletal/cytology , Muscle, Skeletal/metabolism , Oocytes/metabolism , Oxidative Stress , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha , Physical Conditioning, Animal , Receptors, Adiponectin/deficiency , Transcription Factors , Xenopus laevis
13.
Gut ; 57(11): 1531-8, 2008 Nov.
Article in English | MEDLINE | ID: mdl-18676419

ABSTRACT

BACKGROUND AND AIMS: The effect of adiponectin on colorectal carcinogenesis has been proposed but not fully investigated. We investigated the effect of adiponectin deficiency on the development of colorectal cancer. METHODS: We generated three types of gene-deficient mice (adiponectin-deficient, adiponectin receptor 1-deficient, and adiponectin receptor 2-deficient) and investigated chemical-induced colon polyp formation and cell proliferation in colon epithelium. Western blot analysis was performed to elucidate the mechanism which affected colorectal carcinogenesis by adiponectin deficiency. RESULTS: The numbers of colon polyps were significantly increased in adiponectin-deficient mice compared with wild-type mice fed a high-fat diet. However, no difference was observed between wild-type and adiponectin-deficient mice fed a basal diet. A significant increase in cell proliferative activity was also observed in the colonic epithelium of the adiponectin-deficient mice when compared with wild-type mice fed a high-fat diet; however, no difference was observed between wild-type and adiponectin-deficient mice fed a basal diet. Similarly, an increase in epithelial cell proliferation was observed in adiponectin receptor 1-deficient mice, but not in adiponectin receptor 2-deficient mice. Western blot analysis revealed activation of mammalian target of rapamycin, p70 S6 kinase, S6 protein and inactivation of AMP-activated protein kinase in the colon epithelium of adiponectin-deficient mice fed with high-fat diet. CONCLUSIONS: Adiponectin suppresses colonic epithelial proliferation via inhibition of the mammalian target of the rapamycin pathway under a high-fat diet, but not under a basal diet. These studies indicate a novel mechanism of suppression of colorectal carcinogenesis induced by a Western-style high-fat diet.


Subject(s)
Adiponectin/deficiency , Colorectal Neoplasms/etiology , Dietary Fats/administration & dosage , Obesity/metabolism , Precancerous Conditions/etiology , Receptors, Adiponectin/deficiency , AMP-Activated Protein Kinase Kinases , Animals , Apoptosis/physiology , Azoxymethane , Blotting, Western , Cell Proliferation , Cell Transformation, Neoplastic/chemically induced , Cell Transformation, Neoplastic/metabolism , Colonic Polyps/etiology , Colonic Polyps/pathology , Colorectal Neoplasms/pathology , Colorectal Neoplasms/physiopathology , Dietary Fats/adverse effects , Intestinal Mucosa/cytology , Intestinal Mucosa/metabolism , Mice , Mice, Knockout , Obesity/physiopathology , Precancerous Conditions/chemically induced , Precancerous Conditions/pathology , Protein Biosynthesis/physiology , Protein Kinases/metabolism , TOR Serine-Threonine Kinases
SELECTION OF CITATIONS
SEARCH DETAIL
...