Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 33
1.
Am J Physiol Renal Physiol ; 326(1): F30-F38, 2024 01 01.
Article En | MEDLINE | ID: mdl-37916286

Plasma nucleosides-pseudouridine (PU) and N2N2-dimethyl guanosine (DMG) predict the progression of type 2 diabetic kidney disease (DKD) to end-stage renal disease, but the mechanisms underlying this relationship are not well understood. We used a well-characterized model of type 2 diabetes (db/db mice) and control nondiabetic mice (db/m mice) to characterize the production and excretion of PU and DMG levels using liquid chromatography-mass spectrometry. The fractional excretion of PU and DMG was decreased in db/db mice compared with control mice at 24 wk before any changes to renal function. We then examined the dynamic changes in nucleoside metabolism using in vivo metabolic flux analysis with the injection of labeled nucleoside precursors. Metabolic flux analysis revealed significant decreases in the ratio of urine-to-plasma labeling of PU and DMG in db/db mice compared with db/m mice, indicating significant tubular dysfunction in diabetic kidney disease. We observed that the gene and protein expression of the renal tubular transporters involved with nucleoside transport in diabetic kidneys in mice and humans was reduced. In conclusion, this study strongly suggests that tubular handling of nucleosides is altered in early DKD, in part explaining the association of PU and DMG with human DKD progression observed in previous studies.NEW & NOTEWORTHY Tubular dysfunction explains the association between the nucleosides pseudouridine and N2N2-dimethyl guanosine and diabetic kidney disease.


Diabetes Mellitus, Type 2 , Diabetic Nephropathies , Humans , Mice , Animals , Diabetic Nephropathies/metabolism , Pseudouridine/metabolism , Diabetes Mellitus, Type 2/complications , Diabetes Mellitus, Type 2/metabolism , Nucleosides/metabolism , Renal Elimination , Kidney/metabolism , Guanosine/metabolism
2.
J Biol Chem ; 299(4): 103057, 2023 04.
Article En | MEDLINE | ID: mdl-36822331

CLEC16A is an E3 ubiquitin ligase that regulates mitochondrial quality control through mitophagy and is associated with over 20 human diseases. CLEC16A forms a complex with another E3 ligase, RNF41, and a ubiquitin-specific peptidase, USP8; however, regions that regulate CLEC16A activity or the assembly of the tripartite mitophagy regulatory complex are unknown. Here, we report that CLEC16A contains an internal intrinsically disordered protein region (IDPR) that is crucial for CLEC16A function and turnover. IDPRs lack a fixed secondary structure and possess emerging yet still equivocal roles in protein stability, interactions, and enzymatic activity. We find that the internal IDPR of CLEC16A is crucial for its degradation. CLEC16A turnover was promoted by RNF41, which binds and acts upon the internal IDPR to destabilize CLEC16A. Loss of this internal IDPR also destabilized the ubiquitin-dependent tripartite CLEC16A-RNF41-USP8 complex. Finally, the presence of an internal IDPR within CLEC16A was confirmed using NMR and CD spectroscopy. Together, our studies reveal that an IDPR is essential to control the reciprocal regulatory balance between CLEC16A and RNF41, which could be targeted to improve mitochondrial health in disease.


Intrinsically Disordered Proteins , Mitophagy , Humans , Intrinsically Disordered Proteins/genetics , Intrinsically Disordered Proteins/metabolism , Ubiquitin-Protein Ligases/metabolism , Ubiquitin/metabolism , Monosaccharide Transport Proteins/metabolism , Lectins, C-Type/metabolism
3.
Autophagy ; 19(2): 525-543, 2023 02.
Article En | MEDLINE | ID: mdl-35604110

CLEC16A regulates mitochondrial health through mitophagy and is associated with over 20 human diseases. However, the key structural and functional regions of CLEC16A, and their relevance for human disease, remain unknown. Here, we report that a disease-associated CLEC16A variant lacks a C-terminal intrinsically disordered protein region (IDPR) that is critical for mitochondrial quality control. IDPRs comprise nearly half of the human proteome, yet their mechanistic roles in human disease are poorly understood. Using carbon detect NMR, we find that the CLEC16A C terminus lacks secondary structure, validating the presence of an IDPR. Loss of the CLEC16A C-terminal IDPR in vivo impairs mitophagy, mitochondrial function, and glucose-stimulated insulin secretion, ultimately causing glucose intolerance. Deletion of the CLEC16A C-terminal IDPR increases CLEC16A ubiquitination and degradation, thus impairing assembly of the mitophagy regulatory machinery. Importantly, CLEC16A stability is dependent on proline bias within the C-terminal IDPR, but not amino acid sequence order or charge. Together, we elucidate how an IDPR in CLEC16A regulates mitophagy and implicate pathogenic human gene variants that disrupt IDPRs as novel contributors to diabetes and other CLEC16A-associated diseases.Abbreviations : CAS: carbon-detect amino-acid specific; IDPR: intrinsically disordered protein region; MEFs: mouse embryonic fibroblasts; NMR: nuclear magnetic resonance.


Intrinsically Disordered Proteins , Mitophagy , Humans , Animals , Mice , Mitophagy/genetics , Intrinsically Disordered Proteins/genetics , Intrinsically Disordered Proteins/chemistry , Intrinsically Disordered Proteins/metabolism , Autophagy , Fibroblasts/metabolism , Ubiquitination , Monosaccharide Transport Proteins/metabolism , Lectins, C-Type/genetics , Lectins, C-Type/metabolism
4.
JCI Insight ; 5(24)2020 12 17.
Article En | MEDLINE | ID: mdl-33232298

Inflammatory damage contributes to ß cell failure in type 1 and 2 diabetes (T1D and T2D, respectively). Mitochondria are damaged by inflammatory signaling in ß cells, resulting in impaired bioenergetics and initiation of proapoptotic machinery. Hence, the identification of protective responses to inflammation could lead to new therapeutic targets. Here, we report that mitophagy serves as a protective response to inflammatory stress in both human and rodent ß cells. Utilizing in vivo mitophagy reporters, we observed that diabetogenic proinflammatory cytokines induced mitophagy in response to nitrosative/oxidative mitochondrial damage. Mitophagy-deficient ß cells were sensitized to inflammatory stress, leading to the accumulation of fragmented dysfunctional mitochondria, increased ß cell death, and hyperglycemia. Overexpression of CLEC16A, a T1D gene and mitophagy regulator whose expression in islets is protective against T1D, ameliorated cytokine-induced human ß cell apoptosis. Thus, mitophagy promotes ß cell survival and prevents diabetes by countering inflammatory injury. Targeting this pathway has the potential to prevent ß cell failure in diabetes and may be beneficial in other inflammatory conditions.


Insulin-Secreting Cells/metabolism , Lectins, C-Type/metabolism , Mitophagy/physiology , Monosaccharide Transport Proteins/metabolism , Animals , Apoptosis , Cell Survival , Diabetes Complications , Diabetes Mellitus/metabolism , Diabetes Mellitus, Type 1/metabolism , Diabetes Mellitus, Type 2/metabolism , Disease Models, Animal , Female , Humans , Inflammation/metabolism , Insulin-Secreting Cells/physiology , Male , Mice , Mice, Inbred C57BL , Mitochondria/metabolism , Oxidative Stress , Primary Cell Culture , Protective Agents/metabolism , Signal Transduction
5.
Diabetes ; 67(2): 265-277, 2018 02.
Article En | MEDLINE | ID: mdl-29180353

Mitophagy is a cellular quality-control pathway, which is essential for elimination of unhealthy mitochondria. While mitophagy is critical to pancreatic ß-cell function, the posttranslational signals governing ß-cell mitochondrial turnover are unknown. Here, we report that ubiquitination is essential for the assembly of a mitophagy regulatory complex, comprised of the E3 ligase Nrdp1, the deubiquitinase enzyme USP8, and Clec16a, a mediator of ß-cell mitophagy with unclear function. We discover that the diabetes gene Clec16a encodes an E3 ligase, which promotes nondegradative ubiquitin conjugates to direct its mitophagy effectors and stabilize the Clec16a-Nrdp1-USP8 complex. Inhibition of the Clec16a pathway by the chemotherapeutic lenalidomide, a selective ubiquitin ligase inhibitor associated with new-onset diabetes, impairs ß-cell mitophagy, oxygen consumption, and insulin secretion. Indeed, patients treated with lenalidomide develop compromised ß-cell function. Moreover, the ß-cell Clec16a-Nrdp1-USP8 mitophagy complex is destabilized and dysfunctional after lenalidomide treatment as well as after glucolipotoxic stress. Thus, the Clec16a-Nrdp1-USP8 complex relies on ubiquitin signals to promote mitophagy and maintain mitochondrial quality control necessary for optimal ß-cell function.


Endopeptidases/metabolism , Endosomal Sorting Complexes Required for Transport/metabolism , Insulin-Secreting Cells/metabolism , Lectins, C-Type/metabolism , Mitophagy , Monosaccharide Transport Proteins/metabolism , Ubiquitin Thiolesterase/metabolism , Ubiquitin-Protein Ligases/metabolism , Angiogenesis Inhibitors/pharmacology , Animals , Cell Line , Cells, Cultured , Crosses, Genetic , Endopeptidases/chemistry , Endopeptidases/genetics , Endosomal Sorting Complexes Required for Transport/antagonists & inhibitors , Endosomal Sorting Complexes Required for Transport/chemistry , Endosomal Sorting Complexes Required for Transport/genetics , Enzyme Inhibitors/pharmacology , Glucose/metabolism , Humans , Insulin/metabolism , Insulin Secretion , Insulin-Secreting Cells/cytology , Insulin-Secreting Cells/drug effects , Lectins, C-Type/antagonists & inhibitors , Lectins, C-Type/chemistry , Lectins, C-Type/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Mitophagy/drug effects , Monosaccharide Transport Proteins/antagonists & inhibitors , Monosaccharide Transport Proteins/chemistry , Monosaccharide Transport Proteins/genetics , Protein Multimerization/drug effects , Protein Stability/drug effects , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/metabolism , Tissue Banks , Tissue Culture Techniques , Ubiquitin Thiolesterase/antagonists & inhibitors , Ubiquitin Thiolesterase/chemistry , Ubiquitin Thiolesterase/genetics , Ubiquitin-Protein Ligases/antagonists & inhibitors , Ubiquitin-Protein Ligases/chemistry , Ubiquitin-Protein Ligases/genetics , Ubiquitination/drug effects
6.
Neurosci Lett ; 595: 7-11, 2015 May 19.
Article En | MEDLINE | ID: mdl-25841790

The mechanisms underlying alterations in brain functions in response to physical exercise are not fully understood. The present study examined the central effect of irisin, a 112 amino acid polypeptide hormone secreted from the skeletal muscle after exercise, on the locomotion in rats. Central administration of irisin significantly increased the locomotion. Relative to control animals treated with IgG Fc peptide, rats receiving irisin demonstrated a marked increase in total travel distance, ambulatory counts and time, and vertical counts and time. These changes were associated with a significant decrease in resting time. Central treatment of irisin also induced significant increases in oxygen consumption, carbon dioxide production and heat production, indicating an increase in metabolic activity. Our study suggests that physical activity may signal to the central nervous system to coordinate locomotion with metabolic activity via irisin.


Fibronectins/pharmacology , Motor Activity/drug effects , Animals , Carbon Dioxide/metabolism , Fibronectins/metabolism , Injections, Intraventricular , Locomotion/drug effects , Male , Oxygen Consumption/drug effects , Rats, Sprague-Dawley
7.
Cardiovasc Drugs Ther ; 29(2): 121-7, 2015 Apr.
Article En | MEDLINE | ID: mdl-25820670

INTRODUCTION: Irisin is a newly identified 112 amino acid hormone, derived as a product of fibronectin type III domain containing 5 (FNDC5), which is highly related to metabolic activity in skeletal muscle and brown fat. The effects of irisin on cardiovascular functions are unknown. PURPOSE: To explore the effects of central and peripheral irisin on cardiovascular functions. METHODS: Irisin was either administrated into 3rd ventricle of rats or intravenously, and its effects on blood pressure and cardiac contractibility measured. RESULTS: Administration of recombinant human irisin into the 3rd brain ventricle of rats activated neurons in the paraventricular nuclei of the hypothalamus. Central administration of irisin increased blood pressure and cardiac contractibility. Exogenous irisin reversed atenolol-induced inhibition of cardiac contractibility. In contrast, peripheral administration of irisin reduced blood pressure in both control and spontaneously hypertensive rats. Irisin dilated mesenteric artery rings through ATP-sensitive potassium channels. CONCLUSION: Our studies indicate that central and peripheral irisin may differentially regulate cardiovascular activities.


Blood Pressure/physiology , Fibronectins/physiology , Paraventricular Hypothalamic Nucleus/drug effects , Administration, Intravenous , Animals , Blood Pressure/drug effects , Fibronectins/administration & dosage , Fibronectins/pharmacology , Heart/drug effects , Heart/physiology , Humans , Infusions, Intraventricular , KATP Channels/drug effects , KATP Channels/physiology , Male , Mesenteric Arteries/drug effects , Mesenteric Arteries/physiology , Neurons/drug effects , Neurons/physiology , Paraventricular Hypothalamic Nucleus/physiology , Rats , Rats, Inbred SHR , Recombinant Proteins/administration & dosage , Recombinant Proteins/pharmacology , Vasodilation/drug effects , Vasodilation/physiology
8.
J Gastrointest Surg ; 18(8): 1495-506, 2014 Aug.
Article En | MEDLINE | ID: mdl-24916589

BACKGROUND: In inflammatory bowel disease, autonomic dysfunction contributes to symptoms, morbidity, and health care resource utilization. Efferent vagal neurons, which provide the primary parasympathetic input to the gastrointestinal tract, are housed in the dorsal motor nucleus of the vagus (DMV) in the brainstem. This study seeks to characterize the effects of IBD on DMV neuronal survival and function. METHODS: TNBS (picrylsulfonic acid) was administered by enema to induce colitis in rats. Brain sections through the DMV were examined for neuronal apoptosis using TUNEL labeling, and for glial cell activation by immunofluorescence. Prothrombin production was evaluated via quantitative RT-PCR from DMV tissue, as well as by double immunofluorescence in DMV sections. To investigate the effects of thrombin in the DMV, thrombin or thrombin and an antagonist to its receptor were administered into the fourth ventricle via a stereotactically placed cannula. DMV sections were then examined for apoptosis by TUNEL assay. To evaluate the effect of thrombin on DMV neuronal function, we examined calcium signaling in primary DMV neuron cultures following exposure to thrombin and other neurotransmitters. RESULTS: TNBS colitis is associated with significantly increased rates of DMV neuronal apoptosis, affecting 12.7 % of DMV neurons in animals with colitis, compared to 3.4 % in controls. There was a corresponding increase in DMV neuron activated caspase-3 immunoreactivity (14.8 vs. 2.6 % of DMV neurons). TNBS-treated animals also demonstrated significantly increased DMV astrocyte and microglial immunoreactivity, indicating glial cell activation. DMV prothrombin production was significantly increased in TNBS colitis, with a close anatomic relationship between prothrombin and microglia. Direct DMV exposure to thrombin replicated the apoptosis and activation of caspase-3 seen in TNBS colitis; these effects were prevented by coadministration of the PAR-1 inhibitor FR171113. Cultured DMV neurons exhibited impaired calcium signaling in response to neurotransmitters following exposure to thrombin. Glutamate-induced calcium transients decreased by 59 %, and those triggered by GABA were reduced by 61 %. PAR-1 antagonism prevented these thrombin-induced changes in calcium signaling. CONCLUSIONS: IBD is associated with DMV microglial activation and production of prothrombin. Thrombin in the DMV causes vagal neuron apoptosis and decreased sensitivity to neurotransmitters.


Apoptosis , Brain Stem/physiopathology , Colitis, Ulcerative/physiopathology , Neurons/physiology , Prothrombin/metabolism , Thrombin/metabolism , Vagus Nerve/physiopathology , Animals , Biomarkers/metabolism , Brain Stem/metabolism , Colitis, Ulcerative/chemically induced , Colitis, Ulcerative/metabolism , Fluorescent Antibody Technique , In Situ Nick-End Labeling , Inflammatory Bowel Diseases/chemically induced , Inflammatory Bowel Diseases/metabolism , Inflammatory Bowel Diseases/physiopathology , Male , Rats , Rats, Sprague-Dawley , Reverse Transcriptase Polymerase Chain Reaction , Trinitrobenzenesulfonic Acid
9.
J Gastrointest Surg ; 18(9): 1632-41, 2014 Sep.
Article En | MEDLINE | ID: mdl-24961441

BACKGROUND: The role of peripheral tumor necrosis factor alpha (TNFα) in inflammatory bowel disease (IBD) is well established, but its central nervous system (CNS) effects are not understood. Thrombin, another mediator of inflammation in IBD, has been implicated in CNS vagal neuron apoptosis in the dorsal motor nucleus of the vagus (DMV). This study evaluates DMV TNFα exposure, characterizes effects of TNFα on DMV neurons, and identifies a relationship between DMV TNFα and thrombin in IBD. METHODS: 2,4,6-Trinitrobenzene sulfonic acid was administered via enema to induce colonic inflammation in rats. TNFα in serum, cerebrospinal fluid (CSF), and DMV tissues were determined by ELISA and DMV TNFα expression by quantitative reverse transcription PCR (RT-PCR). TNFα was administered into the fourth intracerebral ventricle (4 V) adjacent to the DMV, with and without blockade of TNF receptor 1 (TNFR1) and the thrombin receptor proteinase-activated receptor 1 (PAR1). Immunofluorescence was used to evaluate microglial activation (Cd11b) and prothrombin presence in DMV sections. Apoptosis was examined using terminal deoxynucleotidyl transferase deoxyuridine triphosphate nick end labeling (TUNEL) and activated caspase-3 immunofluorescence. RESULTS: IBD is associated with increased TNFα protein in serum, CSF, and DMV tissue; DMV TNFα transcription is also increased. TNFα (4 V) caused a 54 % increase in microglial activation, a 27 % increase in DMV prothrombin protein, and a 31 % increase in vagal neuron apoptosis by TUNEL. There was a 52 % increase in activated caspase-3 immunofluorescence in TNFα-treated animals (p < 0.05). All effects of 4 V TNFα were prevented by TNFR1 blockade. TNFα-induced apoptosis was prevented by PAR1 blockade. CONCLUSIONS: IBD is associated with DMV exposure to TNFα, causing excess DMV prothrombin and vagal apoptosis.


Apoptosis/drug effects , Inflammatory Bowel Diseases/metabolism , Neurons, Efferent/drug effects , Neurons, Efferent/metabolism , Thrombin/metabolism , Tumor Necrosis Factor-alpha/metabolism , Tumor Necrosis Factor-alpha/pharmacology , Animals , CD11b Antigen/metabolism , Caspase 3/metabolism , Inflammatory Bowel Diseases/chemically induced , Male , Microglia/drug effects , Microglia/metabolism , Prothrombin/metabolism , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Receptor, PAR-1/antagonists & inhibitors , Receptors, Tumor Necrosis Factor, Type I/antagonists & inhibitors , Trinitrobenzenesulfonic Acid , Tumor Necrosis Factor-alpha/genetics , Vagus Nerve
10.
Endocrinology ; 155(2): 429-40, 2014 Feb.
Article En | MEDLINE | ID: mdl-24280058

The hypothalamus plays a key role in the regulation of feeding behavior. Several hypothalamic nuclei, including the arcuate nucleus (ARC), paraventricular nucleus, and ventromedial nucleus of the hypothalamus (VMH), are involved in energy homeostasis. Analysis of microarray data derived from ARC revealed that leucine-rich repeat-containing G protein-coupled receptor 4 (LGR4) is highly expressed. LGR4, LGR5, and LGR6 form a subfamily of closely related receptors. Recently, R-spondin (Rspo) family proteins were identified as ligands of the LGR4 subfamily. In the present study, we investigated the distribution and function of LGR4-LGR6 and Rspos (1-4) in the brain of male rat. In situ hybridization showed that LGR4 is expressed in the ARC, VMH, and median eminence of the hypothalamus. LGR4 colocalizes with neuropeptide Y, proopiomelanocortin, and brain-derived neurotrophic factor neurons. LGR5 is not detectable with in situ hybridization; LGR6 is only expressed in the epithelial lining of the lower portion of the third ventricle and median eminence. Rspo1 is expressed in the VMH and down-regulated with fasting. Rspo3 is expressed in the paraventricular nucleus and also down-regulated with fasting. Rspos 1 and 3 colocalize with the neuronal marker HuD, indicating that they are expressed by neurons. Injection of Rspo1 or Rspo3 into the third brain ventricle inhibited food intake. Rspo1 decreased neuropeptide Y and increased proopiomelanocortin expression in the ARC. Rspo1 and Rspo3 mRNA is up-regulated by insulin. These data indicate that Rspo1 and Rspo3 and their receptor LGR4 form novel circuits in the brain to regulate energy homeostasis.


Eating/physiology , Hypothalamus/metabolism , Neurons/metabolism , Receptors, G-Protein-Coupled/metabolism , Thrombospondins/metabolism , Animals , Brain-Derived Neurotrophic Factor/metabolism , Down-Regulation/drug effects , Down-Regulation/physiology , Eating/drug effects , Fasting , Hypothalamus/drug effects , Insulin/pharmacology , Male , Neurons/drug effects , Neuropeptide Y/metabolism , Pro-Opiomelanocortin/metabolism , Rats , Thrombospondins/pharmacology , Up-Regulation/drug effects , Up-Regulation/physiology
11.
Exp Physiol ; 98(12): 1696-704, 2013 Dec.
Article En | MEDLINE | ID: mdl-23955309

Previous studies have demonstrated that mammalian target of rapamycin (mTOR) signalling in the hypothalamus is involved in the control of energy homeostasis. The aim of this study was to characterize the effect of mTOR signalling in the dorsal motor nucleus of the vagus (DMNV) on energy intake. Phospho-mTOR was detected in the DMNV neurons, and its levels were increased by energy deprivation. Rapamycin significantly inhibited mTOR activity and reduced food intake when administrated into the fourth ventricle. Exposure of DMNV neurons to ghrelin increased the phosphorylation of mTOR. Injection of ghrelin into the fourth ventricle significantly increased food intake relative to the control vehicle. Pretreatment with rapamycin for 15 min attenuated the orexigenic effect of ghrelin. A reduction in the phosphorylation of mTOR was observed following injection of nesfatin-1 into the fourth ventricle. When administrated by injection into the fourth ventricle, nesfatin-1 suppressed food intake in comparison with the control vehicle. The anorexigenic effect of nesfatin-1 was significantly attenuated by pretreatment with leucine for 15 min. All these findings suggest that mTOR signalling in the DMNV neurons regulates both the nutrient and the hormonal signals for the modulation of food intake.


Calcium-Binding Proteins/metabolism , DNA-Binding Proteins/metabolism , Eating/physiology , Ghrelin/metabolism , Motor Neurons/metabolism , Nerve Tissue Proteins/metabolism , TOR Serine-Threonine Kinases/metabolism , Vagus Nerve/metabolism , Animals , Animals, Newborn , Cells, Cultured , Energy Intake , Fasting/physiology , Male , Nucleobindins , Rats , Rats, Sprague-Dawley
12.
Am J Physiol Gastrointest Liver Physiol ; 303(5): G570-7, 2012 Sep 01.
Article En | MEDLINE | ID: mdl-22723266

Nesfatin-1, a novel hypothalamic peptide, inhibits nocturnal feeding behavior and gastrointestinal motility in rodents. The effects of nesfatin-1 on gastrointestinal secretory function, including gastric acid production, have not been evaluated. Nesfatin-1 was injected into the fourth intracerebral ventricle (4V) of chronically cannulated rats to identify a nesfatin dose sufficient to inhibit food intake. Nesfatin-1 (2 µg) inhibited dark-phase food intake, in a dose-dependent fashion, for >3 h. Gastric acid production was evaluated in urethane-anesthetized rats. Nesfatin-1 (2 µg) was introduced via the 4V following endocrine stimulation of gastric acid secretion by pentagastrin (2 µg·kg(-1)·h(-1) iv), vagal stimulation with 2-deoxy-D-glucose (200 mg/kg sc), or no stimulus. Gastric secretions were collected via gastric cannula and neutralized by titration to determine acid content. Nesfatin-1 did not affect basal and pentagastrin-stimulated gastric acid secretion, whereas 2-deoxy-D-glucose-stimulated gastric acid production was inhibited by nesfatin-1 in a dose-dependent manner. c-Fos immunofluorescence in brain sections was used to evaluate in vivo neuronal activation by nesfatin-1 administered via the 4V. Nesfatin-1 caused activation of efferent vagal neurons, as evidenced by a 16-fold increase in the mean number of c-Fos-positive neurons in the dorsal motor nucleus of the vagus (DMNV) in nesfatin-1-treated animals vs. controls (P < 0.01). Finally, nesfatin-induced Ca(2+) signaling was evaluated in primary cultured DMNV neurons from neonatal rats. Nesfatin-1 caused dose-dependent Ca(2+) increments in 95% of cultured DMNV neurons. These studies demonstrate that central administration of nesfatin-1, at doses sufficient to inhibit food intake, results in inhibition of vagally stimulated secretion of gastric acid. Nesfatin-1 activates DMNV efferent vagal neurons in vivo and triggers Ca(2+) signaling in cultured DMNV neurons.


Calcium-Binding Proteins/pharmacology , DNA-Binding Proteins/pharmacology , Eating/drug effects , Gastric Acid/metabolism , Gastric Mucosa/metabolism , Nerve Tissue Proteins/pharmacology , Vagus Nerve/drug effects , Animals , Calcium/metabolism , Male , Nucleobindins , Proto-Oncogene Proteins c-fos/metabolism , Rats , Rats, Sprague-Dawley , Vagus Nerve/physiology
13.
BMC Neurosci ; 12: 95, 2011 Sep 28.
Article En | MEDLINE | ID: mdl-21955513

BACKGROUND: The arcuate nucleus of the hypothalamus regulates food intake. Ankyrin repeat and SOCS box containing protein 4 (Asb-4) is expressed in neuropeptide Y and proopiomelanocortin (POMC) neurons in the arcuate nucleus, target neurons in the regulation of food intake and metabolism by insulin and leptin. However, the target protein(s) of Asb-4 in these neurons remains unknown. Insulin receptor substrate 4 (IRS4) is an adaptor molecule involved in the signal transduction by both insulin and leptin. In the present study we examined the colocalization and interaction of Asb-4 with IRS4 and the involvement of Asb-4 in insulin signaling. RESULTS: In situ hybridization showed that the expression pattern of Asb-4 was consistent with that of IRS4 in the rat brain. Double in situ hybridization showed that IRS4 colocalized with Asb-4, and both Asb-4 and IRS4 mRNA were expressed in proopiomelanocortin (POMC) and neuropeptide Y (NPY) neurons within the arcuate nucleus of the hypothalamus. In HEK293 cells co-transfected with Myc-tagged Asb-4 and Flag-tagged IRS4, Asb-4 co-immunoprecipitated with IRS4; In these cells endogenous IRS4 also co-immunoprecipitated with transfected Myc-Asb-4; Furthermore, Asb-4 co-immunoprecipitated with IRS4 in rat hypothalamic extracts. In HEK293 cells over expression of Asb-4 decreased IRS4 protein levels and deletion of the SOCS box abolished this effect. Asb-4 increased the ubiquitination of IRS4; Deletion of SOCS box abolished this effect. Expression of Asb-4 decreased both basal and insulin-stimulated phosphorylation of AKT at Thr308. CONCLUSIONS: These data demonstrated that Asb-4 co-localizes and interacts with IRS4 in hypothalamic neurons. The interaction of Asb-4 with IRS4 in cell lines mediates the degradation of IRS4 and decreases insulin signaling.


Hypothalamus/cytology , Hypothalamus/metabolism , Insulin Receptor Substrate Proteins/antagonists & inhibitors , Insulin Receptor Substrate Proteins/metabolism , Neurons/metabolism , Suppressor of Cytokine Signaling Proteins/metabolism , Animals , CHO Cells , Cell Line , Cricetinae , HEK293 Cells , Humans , Insulin/metabolism , Insulin/physiology , Insulin Receptor Substrate Proteins/genetics , Male , Mice , Neurons/cytology , Rats , Rats, Sprague-Dawley , Signal Transduction/physiology , Suppressor of Cytokine Signaling Proteins/physiology
14.
Peptides ; 31(10): 1888-93, 2010 Oct.
Article En | MEDLINE | ID: mdl-20603172

The melanocortin signaling system is integral in regulating energy homeostasis. The melanocortin-4 receptor (MC4R) activates several signaling pathways in performance of this function. The effect of MC4R on insulin-stimulated mammalian target of rapamycin (mTOR), a cellular energy sensor, signaling was investigated. The GT1-1 cell line which expresses MC4R expression was utilized. mTOR signaling was measured by Western blotting analysis using Phospho-mTOR (Ser2448) antibody. NDP-MSH dose-dependently enhanced insulin-stimulated mTOR phosphorylation. The MC4R antagonist SHU9119 blocked this effect, demonstrating specificity. The protein kinase A - cyclic AMP pathway and the MAP kinase pathway were not involved in NDP-MSH actions on insulin-stimulated mTOR phosphorylation. In contrast, the AMP-activated protein kinase agonist, AICAR, attenuated this effect. MC4R activation potentiates insulin-stimulated mTOR signaling via the AMPK pathway.


Insulin/metabolism , Receptor, Melanocortin, Type 4/metabolism , Signal Transduction/physiology , TOR Serine-Threonine Kinases/metabolism , Affinity Labels/metabolism , Affinity Labels/pharmacology , Aminoimidazole Carboxamide/analogs & derivatives , Aminoimidazole Carboxamide/metabolism , Animals , Cell Line , Cyclic AMP/metabolism , Cyclic AMP-Dependent Protein Kinases/metabolism , Enzyme Activation , Hypoglycemic Agents/metabolism , Mitogen-Activated Protein Kinases/metabolism , Ribonucleotides/metabolism , Signal Transduction/drug effects , alpha-MSH/analogs & derivatives , alpha-MSH/metabolism , alpha-MSH/pharmacology
15.
Endocrinology ; 151(1): 134-42, 2010 Jan.
Article En | MEDLINE | ID: mdl-19934378

Ankyrin repeat and suppressor of cytokine signaling box-containing protein 4 (Asb-4) is specifically expressed in the energy homeostasis-related brain areas and colocalizes with proopiomelanocortin (POMC) neurons of the arcuate nucleus (ARC). Injection of insulin into the third ventricle of the rat brain increased Asb-4 mRNA expression in the paraventricular nucleus but not in the ARC of the hypothalamus, whereas injection of leptin (ip) increased Asb-4 expression in both mouse paraventricular nucleus and ARC. A transgenic mouse in which Myc-tagged Asb-4 is specifically expressed in POMC neurons of the ARC was made and used to study the effects of Asb-4 on ingestive behavior and metabolic rate. Animals with overexpression of Asb-4 in POMC neurons demonstrated an increase in food intake. However, POMC-Asb-4 transgenic animals gained significantly less weight from 6-30 wk of age. The POMC-Asb-4 mice had reduced fat mass and increased lean mass and lower levels of blood leptin. The transgenic animals were resistant to high-fat diet-induced obesity. Transgenic mice had significantly higher rates of oxygen consumption and carbon dioxide production than wild-type mice during both light and dark periods. The locomotive activity of transgenic mice was increased. The overexpression of Asb-4 in POMC neurons increased POMC mRNA expression in the ARC. The transgenic animals had no observed effect on peripheral glucose metabolism and the activity of the autonomic nervous system. These results indicate that Asb-4 is a key regulatory protein in the central nervous system, involved in the control of feeding behavior and metabolic rate.


Arcuate Nucleus of Hypothalamus/metabolism , Hyperphagia/genetics , Neurons/metabolism , Pro-Opiomelanocortin/metabolism , Suppressor of Cytokine Signaling Proteins/genetics , Thinness/genetics , Animals , Arcuate Nucleus of Hypothalamus/drug effects , Female , Gene Expression/drug effects , Gene Expression/physiology , Hyperphagia/metabolism , Insulin/pharmacology , Leptin/pharmacology , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neurons/drug effects , Phenotype , Pro-Opiomelanocortin/genetics , Rats , Rats, Sprague-Dawley , Suppressor of Cytokine Signaling Proteins/metabolism , Thinness/metabolism
16.
Peptides ; 30(6): 1098-104, 2009 Jun.
Article En | MEDLINE | ID: mdl-19463742

The melanocortin system is crucial to regulation of energy homeostasis. The melanocortin receptor type 4 (MC4R) modulates insulin signaling via effects on c-Jun N-terminal kinase (JNK). The melanocortin agonist NDP-MSH dose-dependently inhibited JNK activity in HEK293 cells stably expressing the human MC4R; effects were reversed by melanocortin receptor antagonist. NDP-MSH time- and dose-dependently inhibited IRS-1(ser307) phosphorylation, effects also reversed by a specific melanocortin receptor antagonist. NDP-MSH augmented insulin-stimulated AKT phosphorylation in vitro. The melanocortin agonist melanotan II increased insulin-stimulated AKT phosphorylation in the rat hypothalamus in vivo. NDP-MSH increased insulin-stimulated glucose uptake in hypothalamic GT1-1 cells. The current study shows that the melanocortinergic system interacts with insulin signaling via novel effects on JNK activity.


Insulin/metabolism , JNK Mitogen-Activated Protein Kinases/antagonists & inhibitors , Receptor, Melanocortin, Type 4/agonists , Receptor, Melanocortin, Type 4/metabolism , Signal Transduction , alpha-MSH/analogs & derivatives , Animals , Cell Line , Dose-Response Relationship, Drug , Humans , Hypothalamus/metabolism , Insulin/pharmacology , Insulin Receptor Substrate Proteins/antagonists & inhibitors , Insulin Receptor Substrate Proteins/metabolism , JNK Mitogen-Activated Protein Kinases/metabolism , Peptides, Cyclic/pharmacology , Phosphorylation , Proto-Oncogene Proteins c-akt/drug effects , Proto-Oncogene Proteins c-akt/metabolism , Rats , Rats, Sprague-Dawley , Receptor, Melanocortin, Type 4/biosynthesis , Time Factors , alpha-MSH/pharmacology
17.
Surgery ; 144(2): 149-58, 2008 Aug.
Article En | MEDLINE | ID: mdl-18656620

BACKGROUND: The effects of intestinal inflammation on the central nervous system are unknown. The dorsal motor nucleus of the vagus (DMNV) integrates peripheral and central signals and sends efferent signals to the gastrointestinal system. The purpose of this study was to determine the effects of intestinal inflammation on the DMNV in an animal model and in vitro. METHODS: Carbocyanine dye (DiI) was injected into the stomach wall of rats to label retrogradely the neurons of the DMNV. Colitis was induced with trinitrobenzene sulfonic acid (TNBS). Tissue was examined under fluorescent microscopy. In vitro studies were performed using primary culture of DMNV neurons. Cell proliferation was measured by BrdU incorporation. Apoptosis was measured by an enzyme sandwich-linked immunosorbent assay. Single-cell cytoplasmic calcium transients were determined using the fluorescence dye fura-2-AM. Reverse transcriptase-polymerase chain reaction of glutamate receptor was performed. RESULTS: Animals treated with TNBS ate less and lost weight compared with controls. Microscopic analysis demonstrated a 77% decrease in DiI labeling in the DMNV of TNBS animals compared with controls. Cell proliferation in DMNV neurons after 24-hour exposure to the cytokines interleukin- (IL)-1 beta, IL-6, or tumor necrosis factor- (TNF)-alpha was significantly decreased. Similarly, apoptosis of DMNV neurons after 24 hours of incubation with IL-1 beta or TNF-alpha was significantly increased, but no changes resulted with IL-6. Exposure to each cytokine resulted in decreased glutamate-induced intracellular calcium transients. Transcription of glutamate receptor was decreased after 24-hour exposure to TNF-alpha. CONCLUSIONS: DMNV neurons projecting to the stomach are reduced in number after induction of colitis in rats. In vitro, proinflammatory cytokines diminish DMNV cellular proliferation, increase apoptosis, and alter calcium responses to glutamate. These results indicate that intestinal inflammation affects adversely neuronal survival and function in the DMNV.


Colitis/pathology , Medulla Oblongata/pathology , Neurons/pathology , Vagus Nerve/pathology , Animals , Apoptosis/drug effects , Calcium/analysis , Cell Proliferation/drug effects , Cells, Cultured , Colitis/chemically induced , Cytokines/pharmacology , Immunohistochemistry , Male , Rats , Rats, Sprague-Dawley , Stomach/innervation , Trinitrobenzenesulfonic Acid
18.
Surgery ; 144(2): 159-67, 2008 Aug.
Article En | MEDLINE | ID: mdl-18656621

BACKGROUND: The effects of intestinal inflammation on the central neurons projecting to the enteric nervous system are unknown. The dorsal motor nucleus of the vagus signals to the gastrointestinal system. Ghrelin is elevated in patients with inflammatory bowel disease and has been implicated as an inflammatory mediator. The purpose of this study was to investigate the effects of gastrointestinal inflammation on the dorsal motor nucleus of the vagus in rats, as well as the effects of proinflammatory cytokines and ghrelin on neurons from the dorsal motor nucleus of the vagus in vitro. METHODS: DiI was injected into the stomach wall of rats to retrogradely label neurons of the dorsal motor nucleus of the vagus. Intestinal inflammation was induced with indomethacin injection. Serial serum ghrelin measurements were performed. Tissue was examined under fluorescent microscopy. In vitro studies using primary culture of neurons from the dorsal motor nucleus of the vagus were performed. Reverse transcriptase-polymerase chain reaction for cytokine transcripts and immunohistochemistry for cytokine receptors were performed. Cell proliferation and apoptosis were measured by enzyme-linked immunosorbent assay. RESULTS: A significant decrease of DiI labeling was demonstrated in the dorsal motor nucleus of the vagus of animals injected with indomethacin. Serum levels of ghrelin were significantly elevated 2 days after induction of inflammation. In vitro, apoptosis and cell proliferation were measured after 24-hour exposure to experimental conditions. Ghrelin alone had no effect on apoptosis. Exposure to interleukin (IL)-1 beta or tumor necrosis factor (TNF)-alpha increased apoptosis. The addition of ghrelin to cytokine resulted in significant decreases in apoptosis compared to cytokine alone. Ghrelin significantly increased neuronal proliferation. Exposure to IL-1 beta, IL-6, or TNF-alpha significantly decreased proliferation. The addition of ghrelin to TNF-alpha or IL-6 significantly increased cellular proliferation compared to cytokine alone. CONCLUSIONS: Neurons from the dorsal motor nucleus of the vagus that project to the stomach are reduced in number after induction of colitis in rats. In vitro, proinflammatory cytokines increase apoptosis and decrease cell proliferation of neurons from the dorsal motor nucleus of the vagus. These effects are attenuated by ghrelin.


Apoptosis/drug effects , Cell Proliferation/drug effects , Colitis/pathology , Ghrelin/pharmacology , Neurons, Efferent/drug effects , Vagus Nerve/drug effects , Animals , Cells, Cultured , Colitis/chemically induced , Gastrointestinal Tract/innervation , Ghrelin/blood , Indomethacin , Male , Medulla Oblongata/drug effects , Medulla Oblongata/pathology , Neural Pathways , Neurons, Efferent/chemistry , Neurons, Efferent/pathology , Rats , Rats, Sprague-Dawley , Receptors, Interleukin-1/analysis , Receptors, Interleukin-6/analysis , Trinitrobenzenesulfonic Acid , Tumor Necrosis Factor-alpha/analysis , Vagus Nerve/pathology
19.
Endocrinology ; 149(9): 4710-6, 2008 Sep.
Article En | MEDLINE | ID: mdl-18535105

Ghrelin, a gastric peptide hormone, has been reported to regulate GH secretion and energy homeostasis. Here, we examined the effect of des-acyl ghrelin driven from the fatty acid-binding protein-4 (FABP4) promoter on adiposity and glucose metabolism. A high level of expression of des-acyl ghrelin (692 +/- 293 fmol/g fat) in adipose tissue was detected in FABP4-ghrelin transgenic mice, but not in wild-type littermates. Circulating des-acyl ghrelin was significantly higher in FABP4-ghrelin transgenic mice (8409 +/- 3390 pm) compared with wild-type mice (513 +/- 58 pm). No significant change was observed for plasma acylated ghrelin and obestatin. Epididymal and perirenal fat masses decreased 35 +/- 9 and 52 +/- 9%, respectively, in FABP4-ghrelin transgenic mice. FABP4-ghrelin transgenic mice are resistant to obesity induced by high-fat diet. Brown fat mass was not affected by overexpression of ghrelin in adipose tissue. Glucose tolerance tests showed glucose levels to be significantly lower in FABP4-ghrelin transgenic mice than in controls after glucose administration. Insulin sensitivity testing showed that FABP4-ghrelin transgenic mice had a 28 +/- 5% greater hypoglycemic response to insulin. Our study demonstrates that overexpression of ghrelin from the FABP4 promoter impairs the development of white adipose tissues, and alters glucose tolerance and insulin sensitivity in mice.


Adiposity/ethnology , Energy Metabolism/genetics , Ghrelin/genetics , Glucose/metabolism , Adipose Tissue/anatomy & histology , Adipose Tissue/metabolism , Animals , Body Weight/genetics , Eating/genetics , Fatty Acid-Binding Proteins/genetics , Ghrelin/metabolism , Glucose Intolerance/genetics , Insulin/blood , Insulin Resistance/genetics , Leptin/blood , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Promoter Regions, Genetic
20.
Cell Signal ; 19(6): 1185-92, 2007 Jun.
Article En | MEDLINE | ID: mdl-17276034

Asb-4 is a gene that is specifically expressed in the hypothalamic energy homeostasis-associated areas and is down-regulated in the arcuate nucleus of fasted Sprague Dawley and obese Zucker rats. It has two functional domains, the ankyrin repeat and the SOCS box. The function of Asb-4 is unclear. We used yeast two hybridization to search for protein(s) that interact with Asb-4. With Asb-4 minus its SOCS box (Asb-4/Deltasb) as a bait, we screened mouse testis and arcuate nucleus cDNA libraries and identified G-protein pathway suppressor 1 (GPS1, also known as CSN1) as an Asb-4 interacting protein. GPS1 co-immunoprecipitated with Asb-4 both in vitro and in human HEK293 cells. When Asb-4 and GPS1 were co-transfected into HEK293 cells, expression of Asb-4 reduced the protein level of GPS1. Deletion of the SOCS box (Asb4/Deltasb) did not abolish the inhibitory effect of Asb-4 on GPS1, indicating that the SOCS box was not needed for its inhibitory effect. In NIH 3T3 L1 cells, expression of GPS1 enhanced c-Jun NH2-terminal kinase (JNK) activity. Co-expression of Asb-4 with GPS1 inhibited JNK activity. Treatment of the cells with insulin (20 nM) stimulated JNK activity. Expression of GPS1 potentiated the stimulatory effect of insulin, whereas co-expression of Asb-4 along with GPS1 inhibited JNK activity. In HEK293 cells expression of GPS1 elevated phosphorylation of insulin receptor substrate 1 (IRS-1) at serine307, co-expression of Asb-4 with GPS1 reduced the IRS-1ser307 phosphorylation. The present study demonstrates that Asb-4 interacts with GPS1 and inhibits JNK activity.


Ankyrin Repeat , Intracellular Signaling Peptides and Proteins/metabolism , JNK Mitogen-Activated Protein Kinases/antagonists & inhibitors , Suppressor of Cytokine Signaling Proteins/metabolism , Animals , Cell Line , Humans , Immunoprecipitation , Insulin Receptor Substrate Proteins , JNK Mitogen-Activated Protein Kinases/metabolism , Mice , Neurons/cytology , Neurons/metabolism , Phosphoproteins/metabolism , Phosphorylation , Phosphoserine/metabolism , Protein Binding , Protein Transport , Rats , Recombinant Fusion Proteins/metabolism , Sequence Deletion , Two-Hybrid System Techniques , Ubiquitin/metabolism
...