Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
PLoS One ; 8(1): e53717, 2013.
Article in English | MEDLINE | ID: mdl-23308279

ABSTRACT

A key feature of the innate antiviral immune response is a rapid nonspecific response to virus infection largely mediated by the induction and extracellular secretion of type I interferons (IFNs) that restrict virus replication. Cytoplasmic sensors such as RIG-I recognize viral RNA and trigger antiviral signaling pathways that upregulate IFN transcription. However, it remains largely unknown how antiviral signaling is negatively regulated to maintain homeostasis after the elimination of virus. In this report, we have identified the RING domain-containing protein RING finger 11 (RNF11) as a novel negative regulator of innate antiviral signaling. Overexpression of RNF11 downregulated IFN-ß expression and enhanced viral replication whereas siRNA-mediated knockdown of RNF11 suppressed viral replication. RNF11 interacted with the noncanonical IKK kinases TBK1/IKKi and attenuated their Lys63-linked polyubiquitination by blocking interactions with the E3 ligase TRAF3. The inhibitory function of RNF11 was dependent on the ubiquitin-binding adaptor molecule TAX1BP1 which was required for RNF11 to target TBK1/IKKi. Collectively, these results indicate that RNF11 functions together with TAX1BP1 to restrict antiviral signaling and IFN-ß production.


Subject(s)
Carrier Proteins/genetics , Fibroblasts/immunology , Gene Expression Regulation , Immunity, Innate , Intracellular Signaling Peptides and Proteins/genetics , Neoplasm Proteins/genetics , Signal Transduction , Animals , Carrier Proteins/antagonists & inhibitors , Carrier Proteins/immunology , Cell Line , DNA-Binding Proteins , Fibroblasts/virology , HEK293 Cells , Humans , Interferon-beta/biosynthesis , Interferon-beta/immunology , Intracellular Signaling Peptides and Proteins/immunology , Mice , Neoplasm Proteins/immunology , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/immunology , RNA, Small Interfering/genetics , TNF Receptor-Associated Factor 3/genetics , TNF Receptor-Associated Factor 3/immunology , Ubiquitination , Vesiculovirus/immunology , Virus Replication
3.
J Pharmacol Exp Ther ; 339(1): 115-24, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21775475

ABSTRACT

Ghrelin influences a variety of metabolic functions through a direct action at its receptor, the GhrR (GhrR-1a). Ghrelin knockout (KO) and GhrR KO mice are resistant to the negative effects of high-fat diet (HFD) feeding. We have generated several classes of small-molecule GhrR antagonists and evaluated whether pharmacologic blockade of ghrelin signaling can recapitulate the phenotype of ghrelin/GhrR KO mice. Antagonist treatment blocked ghrelin-induced and spontaneous food intake; however, the effects on spontaneous feeding were absent in GhrR KO mice, suggesting target-specific effects of the antagonists. Oral administration of antagonists to HFD-fed mice improved insulin sensitivity in both glucose tolerance and glycemic clamp tests. The insulin sensitivity observed was characterized by improved glucose disposal with dramatically decreased insulin secretion. It is noteworthy that these results mimic those obtained in similar tests of HFD-fed GhrR KO mice. HFD-fed mice treated for 56 days with antagonist experienced a transient decrease in food intake but a sustained body weight decrease resulting from decreased white adipose, but not lean tissue. They also had improved glucose disposal and a striking reduction in the amount of insulin needed to achieve this. These mice had reduced hepatic steatosis, improved liver function, and no evidence of systemic toxicity relative to controls. Furthermore, GhrR KO mice placed on low- or high-fat diets had lifespans similar to the wild type, emphasizing the long-term safety of ghrelin receptor blockade. We have therefore demonstrated that chronic pharmacologic blockade of the GhrR is an effective and safe strategy for treating metabolic syndrome.


Subject(s)
Insulin Resistance/physiology , Insulin/metabolism , Receptors, Ghrelin/antagonists & inhibitors , Signal Transduction/drug effects , Animals , Anti-Obesity Agents/pharmacology , Blood Glucose/metabolism , Body Weight/drug effects , CHO Cells , Cricetinae , Cricetulus , Dietary Fats/pharmacology , Eating/drug effects , Ghrelin/antagonists & inhibitors , Ghrelin/pharmacology , Glucose Clamp Technique , Immunohistochemistry , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Obesity/drug therapy , Receptors, Ghrelin/physiology , Stress, Physiological/physiology
4.
J Virol ; 85(14): 6955-62, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21593151

ABSTRACT

Human T cell leukemia virus type 1 (HTLV-1) inhibits host antiviral signaling pathways although the underlying mechanisms are unclear. Here we found that the HTLV-1 Tax oncoprotein induced the expression of SOCS1, an inhibitor of interferon signaling. Tax required NF-κB, but not CREB, to induce the expression of SOCS1 in T cells. Furthermore, Tax interacted with SOCS1 in both transfected cells and in HTLV-1-transformed cell lines. Although SOCS1 is normally a short-lived protein, in the presence of Tax, the stability of SOCS1 was greatly increased. Accordingly, Tax enhanced the replication of a heterologous virus, vesicular stomatitis virus (VSV), in a SOCS1-dependent manner. Surprisingly, Tax required SOCS1 to inhibit RIG-I-dependent antiviral signaling, but not the interferon-induced JAK/STAT pathway. Inhibition of SOCS1 by RNA-mediated interference in the HTLV-1-transformed cell line MT-2 resulted in increased IFN-ß expression accompanied by reduced HTLV-1 replication and p19(Gag) levels. Taken together, our results reveal that Tax inhibits antiviral signaling, in part, by hijacking an interferon regulatory protein.


Subject(s)
Gene Products, tax/physiology , Human T-lymphotropic virus 1/physiology , Immunity, Innate/physiology , NF-kappa B/metabolism , Signal Transduction/physiology , Suppressor of Cytokine Signaling Proteins/biosynthesis , Base Sequence , Cell Line , DNA Primers , Enzyme-Linked Immunosorbent Assay , Humans , Reverse Transcriptase Polymerase Chain Reaction , Suppressor of Cytokine Signaling 1 Protein , Two-Hybrid System Techniques , Virus Replication
5.
Growth Horm IGF Res ; 20(1): 73-9, 2010 Feb.
Article in English | MEDLINE | ID: mdl-19747867

ABSTRACT

The goal of this study was to examine factors that contribute to energy balance in female GHR -/- mice. We measured energy intake, energy expenditure (EE), fuel utilization, body mass (M(b)) changes and physical activity in 17month-old female GHR -/- mice and their age-matched wild type littermates. The GHR -/- mice were smaller, consumed more food per unit M(b), had greater EE per unit M(b) and had an increase in 24-h EE/M(b) that was similar to the increase in their surface-area-to-volume ratio. Locomotor activity (LMA) was reduced in the GHR -/- mice, but the energetic cost associated with their LMA was greater than in wild type controls. Furthermore, M(b) and LMA were independent explanatory covariates of most of the variance in EE, and when adjusted for M(b) and LMA, the GHR -/- mice had higher EE during both the light and dark phases of the daily cycle. Respiratory quotient was lower in GHR -/- mice during the light phase, which indicated a greater utilization of lipid relative to carbohydrate in these mice. Additionally, GHR -/- mice had higher ratios of caloric intake to EE at several intervals during the dark phase, and this effect was greater and more sustained in the final 3h of the dark phase. Therefore, we conclude that GHR -/- mice are able to overcome the substantial energetic challenges of dwarfism through several mechanisms that promote stable M(b). Relative to wild type mice, the GHR -/- mice consumed more calories per unit M(b), which offset the disproportionate increase in their daily energy expenditure. While GHR -/- mice oxidized a greater proportion of lipid during the light phase in order to meet their energy requirements, they achieved greater energy efficiency and storage during the dark phase through a combination of higher energy consumption and lower LMA.


Subject(s)
Energy Metabolism/physiology , Receptors, Somatotropin/metabolism , Activity Cycles/physiology , Animals , Dwarfism/metabolism , Energy Intake/physiology , Female , Lipid Metabolism/physiology , Mice , Mice, Mutant Strains , Motor Activity/physiology , Receptors, Somatotropin/genetics , Receptors, Somatotropin/physiology
6.
Am J Physiol Regul Integr Comp Physiol ; 298(3): R747-54, 2010 Mar.
Article in English | MEDLINE | ID: mdl-20018821

ABSTRACT

To define the relationship between the respiratory quotient (RQ) and energy intake (EI) and to determine the impact of spontaneous locomotor activity (LMA) in the development of diet-induced obesity (DIO), we fed C57BL/6 mice a high-fat diet (HFD) for either 4 days or 17 wk and analyzed them using indirect calorimetry. Importantly, changes in body mass during calorimetry (DeltaM(b)) significantly covaried with RQ and EI; adjusting the data for DeltaM(b) permitted an analysis of the energy-balanced state. The 24-h RQ strongly predicted 24-h EI, and the slope of this relationship was diet dependent (HFD or chow) but independent of the HFD feeding period. Early-stage DIO was characterized by dark-period hyperphagia and fat storage, offset by greater light-period lipid oxidation; later stage DIO mice had a milder hyperphagia and lower substrate flexibility. Consequently, whereas 24-h RQ equaled the food quotient of the HFD in both early- and late-stage DIO, the range of RQ values was negatively correlated with, and mostly explained by, 24-h EI only in late-stage DIO. Lean and early-stage DIO mice had similar LMA values that were reduced in late-stage DIO. However, LMA significantly explained variance in total energy expenditure (EE) in only early-stage DIO mice. This indicated that the link between LMA and EE was a transient adaptive response to early DIO, whereas the later loss of LMA did not explain body weight gain in C57BL/6 DIO mice.


Subject(s)
Body Weight/physiology , Energy Metabolism/physiology , Hyperphagia/metabolism , Obesity/metabolism , Oxygen Consumption/physiology , Animals , Calorimetry, Indirect , Dietary Fats/pharmacology , Mice , Mice, Inbred C57BL , Motor Activity/physiology , Predictive Value of Tests
7.
J Pharmacol Exp Ther ; 329(3): 1178-86, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19252061

ABSTRACT

The orexigenic peptide ghrelin has been shown to have prokinetic activity in the gastrointestinal (GI) system of several species, including humans. In this series of experiments, we have evaluated the prokinetic activity of novel, small-molecule ghrelin receptor (GhrR) agonists after parenteral and peroral dosing in mice and rats. Gastric emptying, small intestinal transport, and fecal output were determined after intraperitoneal and intracerebroventricular dosing of GhrR agonists, using ghrelin as a positive control. These same parameters were evaluated after oral gavage dosing of the synthetic agonists. Regardless of dose route, GhrR agonist treatment increased gastric emptying, small intestinal transit, and fecal output. However, fecal output was only increased by GhrR agonist treatment if mice were able to feed during the stimulatory period. Thus, GhrR agonists can stimulate upper GI motility, and the orexigenic action of the compounds can indirectly contribute to prokinetic activity along the entire GI tract. The orexigenic and prokinetic effects of either ghrelin or small-molecule GhrR agonists were selective for the GhrR because they were absent when evaluated in GhrR knockout mice. We next evaluated the efficacy of the synthetic GhrR agonists dosed in a model of opiate-induced bowel dysfunction induced by a single injection of morphine. Oral dosing of a GhrR agonist normalized GI motility in opiate-induced dysmotility. These data demonstrate the potential utility of GhrR agonists for treating gastrointestinal hypomotility disorders.


Subject(s)
Gastrointestinal Motility/drug effects , Ghrelin/administration & dosage , Ghrelin/pharmacology , Peptide Hormones/administration & dosage , Peptide Hormones/pharmacology , Receptors, Ghrelin/agonists , Administration, Oral , Animals , Body Weight/drug effects , Bowen's Disease/chemically induced , Bowen's Disease/drug therapy , Bowen's Disease/physiopathology , Central Nervous System/drug effects , Defecation/drug effects , Eating/drug effects , Gastric Emptying/drug effects , Gastrointestinal Transit/drug effects , Intestine, Small/drug effects , Intestine, Small/physiology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Morphine/pharmacology , Peptide Hormones/blood , Rats , Rats, Sprague-Dawley , Receptors, Ghrelin/genetics , Receptors, Ghrelin/metabolism
8.
Mol Cancer ; 7: 39, 2008 May 19.
Article in English | MEDLINE | ID: mdl-18489761

ABSTRACT

BACKGROUND: We have used a mouse model based on overexpression of c-Myc in B cells genetically engineered to be self-reactive to test the hypothesis that farnesyl transferase inhibitors (FTIs) can effectively treat mature B cell lymphomas. FTIs are undergoing clinical trials to treat both lymphoid and non-lymphoid malignancies and we wished to obtain evidence to support the inclusion of B cell lymphomas in future trials. RESULTS: We report that two FTIs, L-744,832 and SCH66336, blocked the growth of mature B cell lymphoma cells in vitro and in vivo. The FTI treatment affected the proliferation and survival of the transformed B cells to a greater extent than naïve B cells stimulated with antigen. In syngeneic mice transplanted with the transgenic lymphoma cells, L-744,832 treatment prevented the growth of the tumor cells and the morbidity associated with the resulting lymphoma progression. Tumors that arose from transplantation of the lymphoma cells regressed with as little as three days of treatment with L-744,832 or SCH66336. Treatment of these established lymphomas with L-744,832 for seven days led to long-term remission of the disease in approximately 25% of animals. CONCLUSION: FTI treatment can block the proliferation and survival of self-reactive transformed B cells that overexpress Myc. In mice transplanted with mature B cell lymphomas, we found that FTI treatment led to regression of disease. FTIs warrant further consideration as therapeutic agents for mature B cell lymphomas and other lymphoid tumors.


Subject(s)
Enzyme Inhibitors/pharmacology , Farnesyltranstransferase/antagonists & inhibitors , Lymphoma, B-Cell/drug therapy , Animals , Cell Line, Tumor , Cell Proliferation/drug effects , Enzyme Inhibitors/therapeutic use , Farnesyltranstransferase/metabolism , Flow Cytometry , Lymphocytes/drug effects , Lymphocytes/metabolism , Lymphocytes/pathology , Lymphoma, B-Cell/genetics , Lymphoma, B-Cell/pathology , Methionine/analogs & derivatives , Methionine/pharmacology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Piperidines/pharmacology , Pyridines/pharmacology , Remission Induction
9.
Regul Pept ; 150(1-3): 55-61, 2008 Oct 09.
Article in English | MEDLINE | ID: mdl-18453014

ABSTRACT

Stimulation of the ghrelin receptor (GhrR) by ghrelin results in a variety of metabolic changes including increased food intake, fat storage and insulin resistance. Loss of ghrelin signaling is protective against diet-induced obesity, suggesting that ghrelin plays a significant homeostatic role in conditions of metabolic stress. We examined glycemic control in GhrR -/- mice fed a high-fat diet, and used indirect calorimetry to assess fuel substrate usage and energy expenditure. GhrR -/- mice fed a high-fat diet had several measures of greater insulin sensitivity, including: lower fasted blood glucose and plasma insulin, lower %Hb(A1c), lower insulin levels during glucose tolerance tests, and improved performance in hyperinsulinemic-euglycemic and hyperglycemic clamp studies. GhrR -/- mice fed a high-fat diet did not develop hepatic steatosis and had lower total cholesterol, relative to controls. Furthermore, GhrR -/- mice demonstrated a lower intestinal triglyceride secretion rate of dietary lipid. GhrR -/- mice have higher respiratory quotients (RQ), indicating a preference for carbohydrate as fuel. The range of RQ values was wider in GhrR -/- mice, indicating greater metabolic flexibility and insulin sensitivity in these animals. We therefore propose that loss of ghrelin signaling promotes insulin sensitivity and metabolic flexibility, and protects against several fatty diet-induced features of metabolic syndrome due to convergent changes in the intake, absorption and utilization of energy.


Subject(s)
Dietary Fats/metabolism , Energy Metabolism/physiology , Insulin Resistance/physiology , Receptors, Ghrelin/genetics , Animals , Blood Glucose/analysis , Calorimetry, Indirect/methods , Cholesterol/metabolism , Dietary Fats/administration & dosage , Dietary Fats/pharmacology , Fasting , Glucose Tolerance Test , Glycated Hemoglobin/analysis , Glycated Hemoglobin/metabolism , Insulin/blood , Mice , Mice, Knockout , Triglycerides/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...