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










Publication year range
1.
Eur Respir J ; 61(4)2023 04.
Article in English | MEDLINE | ID: mdl-36549711

ABSTRACT

BACKGROUND: Receptor-interacting protein kinase 1 (RIPK1) is a key mediator of regulated cell death (including apoptosis and necroptosis) and inflammation, both drivers of COPD pathogenesis. We aimed to define the contribution of RIPK1 kinase-dependent cell death and inflammation in the pathogenesis of COPD. METHODS: We assessed RIPK1 expression in single-cell RNA sequencing (RNA-seq) data from human and mouse lungs, and validated RIPK1 levels in lung tissue of COPD patients via immunohistochemistry. Next, we assessed the consequences of genetic and pharmacological inhibition of RIPK1 kinase activity in experimental COPD, using Ripk1 S25D/S25D kinase-deficient mice and the RIPK1 kinase inhibitor GSK'547. RESULTS: RIPK1 expression increased in alveolar type 1 (AT1), AT2, ciliated and neuroendocrine cells in human COPD. RIPK1 protein levels were significantly increased in airway epithelium of COPD patients compared with never-smokers and smokers without airflow limitation. In mice, exposure to cigarette smoke (CS) increased Ripk1 expression similarly in AT2 cells, and further in alveolar macrophages and T-cells. Genetic and/or pharmacological inhibition of RIPK1 kinase activity significantly attenuated airway inflammation upon acute and subacute CS exposure, as well as airway remodelling, emphysema, and apoptotic and necroptotic cell death upon chronic CS exposure. Similarly, pharmacological RIPK1 kinase inhibition significantly attenuated elastase-induced emphysema and lung function decline. Finally, RNA-seq on lung tissue of CS-exposed mice revealed downregulation of cell death and inflammatory pathways upon pharmacological RIPK1 kinase inhibition. CONCLUSIONS: RIPK1 kinase inhibition is protective in experimental models of COPD and may represent a novel promising therapeutic approach.


Subject(s)
Emphysema , Pulmonary Disease, Chronic Obstructive , Pulmonary Emphysema , Humans , Mice , Animals , Lung , Cell Death , Inflammation/metabolism , Mice, Inbred C57BL , Receptor-Interacting Protein Serine-Threonine Kinases/genetics , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism
2.
Front Cardiovasc Med ; 8: 715337, 2021.
Article in English | MEDLINE | ID: mdl-34760938

ABSTRACT

Objective: Atherosclerosis is an arterial occlusive disease with hypercholesterolemia and hypertension as common risk factors. Advanced-stage stenotic plaque, which features inflammation and necrotic core formation, is the major reason for clinical intervention. Receptor interacting serine/threonine-protein kinase 1 (RIPK1) mediates inflammation and cell death and is expressed in atherosclerotic lesions. The role of RIPK1 in advanced-stage atherosclerosis is unknown. Approach and Results: To investigate the effect of RIPK1 inhibition in advanced atherosclerotic plaque formation, we used ApoE SA/SA mice, which exhibit hypercholesterolemia, and develop angiotensin-II mediated hypertension upon administration of doxycycline in drinking water. These mice readily develop severe atherosclerosis, including that in coronary arteries. Eight-week-old ApoE SA/SA mice were randomized to orally receive a highly selective RIPK1 inhibitor (RIPK1i, GSK547) mixed with a western diet, or control diet. RIPK1i administration reduced atherosclerotic plaque lesion area at 2 weeks of treatment, consistent with suppressed inflammation (MCP-1, IL-1ß, TNF-α) and reduced monocyte infiltration. However, administration of RIPK1i unexpectedly exacerbated atherosclerosis at 4 weeks of treatment, concomitant with increased macrophages and lipid deposition in the plaques. Incubation of isolated macrophages with oxidized LDL resulted in foam cell formation in vitro. RIPK1i treatment promoted such foam cell formation while suppressing the death of these cells. Accordingly, RIPK1i upregulated the expression of lipid metabolism-related genes (Cd36, Ppara, Lxrα, Lxrb, Srebp1c) in macrophage foam cells with ABCA1/ABCG1 unaltered. Furthermore, RIPK1i treatment inhibited ApoA1 synthesis in the liver and reduced plasma HDL levels. Conclusion: RIPK1 modulates the development of atherosclerosis in a stage-dependent manner, implicating both pro-atherosclerotic (monocyte infiltration and inflammation) and anti-atherosclerotic effects (suppressing foam cell accumulation and promoting ApoA1 synthesis). It is critical to identify an optimal therapeutic duration for potential clinical use of RIPK1 inhibitor in atherosclerosis or other related disease indications.

3.
Nat Commun ; 12(1): 3364, 2021 06 07.
Article in English | MEDLINE | ID: mdl-34099649

ABSTRACT

Necroptosis is a lytic, inflammatory form of cell death that not only contributes to pathogen clearance but can also lead to disease pathogenesis. Necroptosis is triggered by RIPK3-mediated phosphorylation of MLKL, which is thought to initiate MLKL oligomerisation, membrane translocation and membrane rupture, although the precise mechanism is incompletely understood. Here, we show that K63-linked ubiquitin chains are attached to MLKL during necroptosis and that ubiquitylation of MLKL at K219 significantly contributes to the cytotoxic potential of phosphorylated MLKL. The K219R MLKL mutation protects animals from necroptosis-induced skin damage and renders cells resistant to pathogen-induced necroptosis. Mechanistically, we show that ubiquitylation of MLKL at K219 is required for higher-order assembly of MLKL at membranes, facilitating its rupture and necroptosis. We demonstrate that K219 ubiquitylation licenses MLKL activity to induce lytic cell death, suggesting that necroptotic clearance of pathogens as well as MLKL-dependent pathologies are influenced by the ubiquitin-signalling system.


Subject(s)
Herpesviridae Infections/metabolism , Lysine/metabolism , Protein Kinases/metabolism , Skin/metabolism , Animals , Cell Line , Cells, Cultured , HEK293 Cells , HT29 Cells , Herpesviridae Infections/genetics , Herpesviridae Infections/virology , Humans , Lysine/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Muromegalovirus/physiology , NIH 3T3 Cells , Necroptosis/genetics , Necrosis , Protein Kinases/genetics , Skin/pathology , Ubiquitination
4.
Acta Neuropathol ; 140(5): 737-764, 2020 11.
Article in English | MEDLINE | ID: mdl-32642868

ABSTRACT

Impaired neuronal proteostasis is a salient feature of many neurodegenerative diseases, highlighting alterations in the function of the endoplasmic reticulum (ER). We previously reported that targeting the transcription factor XBP1, a key mediator of the ER stress response, delays disease progression and reduces protein aggregation in various models of neurodegeneration. To identify disease modifier genes that may explain the neuroprotective effects of XBP1 deficiency, we performed gene expression profiling of brain cortex and striatum of these animals and uncovered insulin-like growth factor 2 (Igf2) as the major upregulated gene. Here, we studied the impact of IGF2 signaling on protein aggregation in models of Huntington's disease (HD) as proof of concept. Cell culture studies revealed that IGF2 treatment decreases the load of intracellular aggregates of mutant huntingtin and a polyglutamine peptide. These results were validated using induced pluripotent stem cells (iPSC)-derived medium spiny neurons from HD patients and spinocerebellar ataxia cases. The reduction in the levels of mutant huntingtin was associated with a decrease in the half-life of the intracellular protein. The decrease in the levels of abnormal protein aggregation triggered by IGF2 was independent of the activity of autophagy and the proteasome pathways, the two main routes for mutant huntingtin clearance. Conversely, IGF2 signaling enhanced the secretion of soluble mutant huntingtin species through exosomes and microvesicles involving changes in actin dynamics. Administration of IGF2 into the brain of HD mice using gene therapy led to a significant decrease in the levels of mutant huntingtin in three different animal models. Moreover, analysis of human postmortem brain tissue and blood samples from HD patients showed a reduction in IGF2 level. This study identifies IGF2 as a relevant factor deregulated in HD, operating as a disease modifier that buffers the accumulation of abnormal protein species.


Subject(s)
Huntington Disease/metabolism , Huntington Disease/pathology , Insulin-Like Growth Factor II/metabolism , Protein Aggregation, Pathological/metabolism , Animals , Humans , Insulin-Like Growth Factor II/pharmacology , Mice , Mice, Transgenic , Protein Aggregates/drug effects
5.
Blood ; 135(26): 2388-2401, 2020 06 25.
Article in English | MEDLINE | ID: mdl-32232483

ABSTRACT

A goal in precision medicine is to use patient-derived material to predict disease course and intervention outcomes. Here, we use mechanistic observations in a preclinical animal model to design an ex vivo platform that recreates genetic susceptibility to T-cell-mediated damage. Intestinal graft-versus-host disease (GVHD) is a life-threatening complication of allogeneic hematopoietic cell transplantation. We found that intestinal GVHD in mice deficient in Atg16L1, an autophagy gene that is polymorphic in humans, is reversed by inhibiting necroptosis. We further show that cocultured allogeneic T cells kill Atg16L1-mutant intestinal organoids from mice, which was associated with an aberrant epithelial interferon signature. Using this information, we demonstrate that pharmacologically inhibiting necroptosis or interferon signaling protects human organoids derived from individuals harboring a common ATG16L1 variant from allogeneic T-cell attack. Our study provides a roadmap for applying findings in animal models to individualized therapy that targets affected tissues.


Subject(s)
Graft vs Host Disease/prevention & control , Intestinal Diseases/prevention & control , Organoids , T-Lymphocytes/immunology , Acrylamides/pharmacology , Animals , Autophagy , Autophagy-Related Proteins/deficiency , Autophagy-Related Proteins/genetics , Bone Marrow Transplantation/adverse effects , Coculture Techniques , Colon/abnormalities , Female , Genetic Predisposition to Disease , Graft vs Host Disease/immunology , Graft vs Host Disease/pathology , Humans , Imidazoles/pharmacology , Indoles/pharmacology , Inflammatory Bowel Diseases/pathology , Intestinal Diseases/immunology , Intestinal Diseases/pathology , Intestinal Mucosa/immunology , Intestinal Mucosa/pathology , Male , Mice , Mice, Inbred C57BL , Necroptosis/drug effects , Nitriles , Paneth Cells/pathology , Precision Medicine , Pyrazoles/pharmacology , Pyrimidines , Radiation Chimera , Receptor-Interacting Protein Serine-Threonine Kinases/deficiency , Sulfonamides/pharmacology , T-Lymphocytes/transplantation
6.
J Med Chem ; 62(10): 5096-5110, 2019 05 23.
Article in English | MEDLINE | ID: mdl-31013427

ABSTRACT

RIP1 kinase regulates necroptosis and inflammation and may play an important role in contributing to a variety of human pathologies, including inflammatory and neurological diseases. Currently, RIP1 kinase inhibitors have advanced into early clinical trials for evaluation in inflammatory diseases such as psoriasis, rheumatoid arthritis, and ulcerative colitis and neurological diseases such as amyotrophic lateral sclerosis and Alzheimer's disease. In this paper, we report on the design of potent and highly selective dihydropyrazole (DHP) RIP1 kinase inhibitors starting from a high-throughput screen and the lead-optimization of this series from a lead with minimal rat oral exposure to the identification of dihydropyrazole 77 with good pharmacokinetic profiles in multiple species. Additionally, we identified a potent murine RIP1 kinase inhibitor 76 as a valuable in vivo tool molecule suitable for evaluating the role of RIP1 kinase in chronic models of disease. DHP 76 showed efficacy in mouse models of both multiple sclerosis and human retinitis pigmentosa.


Subject(s)
Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/pharmacology , Nuclear Pore Complex Proteins/antagonists & inhibitors , Pyrazoles/chemical synthesis , Pyrazoles/pharmacology , RNA-Binding Proteins/antagonists & inhibitors , Animals , Biological Availability , Cell Line , Chronic Disease , Drug Design , Encephalomyelitis, Autoimmune, Experimental/drug therapy , Enzyme Inhibitors/pharmacokinetics , Haplorhini , High-Throughput Screening Assays , Humans , Mice , Mice, Inbred C57BL , Models, Molecular , Multiple Sclerosis/drug therapy , Pyrazoles/pharmacokinetics , Rats , Retinitis Pigmentosa/drug therapy , Structure-Activity Relationship
7.
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
8.
BMC Physiol ; 11: 1, 2011 Jan 06.
Article in English | MEDLINE | ID: mdl-21211044

ABSTRACT

BACKGROUND: We and others have demonstrated previously that ghrelin receptor (GhrR) knock out (KO) mice fed a high fat diet (HFD) have increased insulin sensitivity and metabolic flexibility relative to WT littermates. A striking feature of the HFD-fed GhrR KO mouse is the dramatic decrease in hepatic steatosis. To characterize further the underlying mechanisms of glucose homeostasis in GhrR KO mice, we conducted both hyperglycemic (HG) and hyperinsulinemic-euglycemic (HI-E) clamps. Additionally, we investigated tissue glucose uptake and specifically examined liver insulin sensitivity. RESULTS: Consistent with glucose tolerance-test data, in HG clamp experiments, GhrR KO mice showed a reduction in glucose-stimulated insulin release relative to WT littermates. Nevertheless, a robust 1st phase insulin secretion was still achieved, indicating that a healthy ß-cell response is maintained. Additionally, GhrR KO mice demonstrated both a significantly increased glucose infusion rate and significantly reduced insulin requirement for maintenance of the HG clamp, consistent with their relative insulin sensitivity. In HI-E clamps, both LFD-fed and HFD-fed GhrR KO mice showed higher peripheral insulin sensitivity relative to WT littermates as indicated by a significant increase in insulin-stimulated glucose disposal (Rd), and decreased hepatic glucose production (HGP). HFD-fed GhrR KO mice showed a marked increase in peripheral tissue glucose uptake in a variety of tissues, including skeletal muscle, brown adipose tissue and white adipose tissue. GhrR KO mice fed a HFD also showed a modest, but significant decrease in conversion of pyruvate to glucose, as would be anticipated if these mice displayed increased liver insulin sensitivity. Additionally, the levels of UCP2 and UCP1 were reduced in the liver and BAT, respectively, in GhrR KO mice relative to WT mice. CONCLUSIONS: These results indicate that improved glucose homeostasis of GhrR KO mice is characterized by robust improvements of glucose disposal in both normal and metabolically challenged states, relative to WT controls. GhrR KO mice have an intact 1st phase insulin response but require significantly less insulin for glucose disposal. Our experiments reveal that the insulin sensitivity of GhrR KO mice is due to both BW independent and dependent factors. We also provide several lines of evidence that a key feature of the GhrR KO mouse is maintenance of hepatic insulin sensitivity during metabolic challenge.


Subject(s)
Glucose Clamp Technique/methods , Insulin Resistance/genetics , Insulin/blood , Receptors, Ghrelin/deficiency , Animals , Dietary Fats/administration & dosage , Glucose Tolerance Test/methods , Glycemic Index/genetics , Liver/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout
9.
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
10.
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
11.
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
12.
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
13.
J Med Chem ; 48(19): 5888-91, 2005 Sep 22.
Article in English | MEDLINE | ID: mdl-16161992

ABSTRACT

4-(1-Benzo[1,3]dioxol-5-ylmethylpiperidine-4-ylmethyl)-6-chlorochromen-2-one (7) is a potent, orally bioavailable melanin concentrating hormone receptor 1 (MCHr1) antagonist that causes dose-dependent weight loss in diet-induced obese mice. Further evaluation of 7 in an anesthetized dog model of cardiovascular safety revealed adverse hemodynamic effects at a plasma concentration comparable to the minimally effective therapeutic concentration. These results highlight the need for scrutiny of the cardiovascular safety profile of MCHr1 antagonists.


Subject(s)
Coumarins/chemical synthesis , Piperidines/chemical synthesis , Receptors, Pituitary Hormone/antagonists & inhibitors , Receptors, Somatostatin/antagonists & inhibitors , Administration, Oral , Animals , Anti-Obesity Agents/adverse effects , Anti-Obesity Agents/chemical synthesis , Anti-Obesity Agents/pharmacology , Biological Availability , Blood Pressure/drug effects , Brain/metabolism , Cell Line, Tumor , Coumarins/adverse effects , Coumarins/pharmacology , Dogs , Eating/drug effects , Energy Metabolism , Humans , Male , Mice , Mice, Obese , Myocardial Contraction/drug effects , Piperidines/adverse effects , Piperidines/pharmacology , Radioligand Assay , Structure-Activity Relationship
14.
Ann N Y Acad Sci ; 1041: 216-22, 2005 May.
Article in English | MEDLINE | ID: mdl-15956711

ABSTRACT

A recombinant adenovirus containing the human H2 preprorelaxin (hH2) cDNA and a reporter gene was coinjected with a transactivator virus (Ad-tTA) into the lateral cerebral ventricles of female rats. Cardiovascular effects were measured over a 21-day period. Circulating vasopressin in the periphery was significantly greater (P < .0001) in the relaxin-treated group throughout the experimental period, compared with controls. There was a significant decrease in plasma osmolality (P < .05) by approximately 10 mmol/L in the treated group by day 14. Immunofluorescence for hH2 present in cryosections showed rAd transduction and hH2 expression from ependymal cells of the ventricular system. Adenovirus-mediated delivery of hH2 to the brain is capable of producing bioactive relaxin that affects cardiovascular parameters.


Subject(s)
Gene Expression , Relaxin/genetics , Relaxin/metabolism , Adenoviridae/genetics , Animals , Humans , Osmolar Concentration , Protein Precursors/genetics , Rats , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Time Factors , Vasopressins/blood
16.
J Mol Neurosci ; 22(1-2): 93-103, 2004.
Article in English | MEDLINE | ID: mdl-14742914

ABSTRACT

To identify new drug targets for the treatment of obesity, we employed a degenerate reverse transcriptasepolymerase chain reaction technique to isolate novel members of the G-protein coupled receptor superfamily from mouse hypothalamus. One of our clones was found to encode a protein with 90% amino acid identity to human GPR10, which was previously identified as the receptor for prolactin-releasing peptide (PrRP) and has been implicated in lactation, the regulation of food intake and other physiological functions. To investigate the role of GPR10 in food intake and body weight homeostasis, we generated mice carrying a targeted deletion of the GPR10 gene. First, using these knockout animals, we confirmed that GPR10 is the principle receptor for PrRP in the mouse hypothalamus because deletion of GPR10 completely abolished PrRP binding to isolated hypothalamic cell membranes. Second, we investigated the effect of normal and high-fat diets on energy intake, body weight, and glucose homeostasis in wild-type and GPR10 knockout mice. After fasting and refeeding, food intake in knockout animals was unchanged relative to control littermates. However, beginning at 16 wk of age on a normal diet, knockout mice became hyperphagic, obese, and showed significant increases in body fat and the levels of leptin and insulin, as well as decreased glucose tolerance. This metabolic profile was similar to the effect of a high-fat diet on wild-type animals. Our findings provide direct evidence that GPR10 is the receptor for PrRP and that it is involved in the regulation of energy balance in mice. GPR10 knockout mice will also prove useful for investigating other proposed activities for PrRP.


Subject(s)
Appetite Regulation/genetics , Homeostasis/genetics , Hypothalamic Hormones/metabolism , Hypothalamus/metabolism , Neuropeptides/metabolism , Receptors, G-Protein-Coupled/genetics , Animals , Body Weight/genetics , Caloric Restriction , Cell Membrane/genetics , Cell Membrane/metabolism , DNA, Complementary/genetics , Food, Formulated , Glucose/metabolism , Humans , Hyperphagia/genetics , Hyperphagia/metabolism , Mice , Mice, Knockout , Molecular Sequence Data , Obesity/genetics , Obesity/metabolism , Prolactin-Releasing Hormone , Protein Binding/genetics , Receptors, G-Protein-Coupled/isolation & purification , Receptors, G-Protein-Coupled/metabolism , Sequence Homology, Amino Acid , Sequence Homology, Nucleic Acid
17.
Endocrinology ; 144(8): 3683-91, 2003 Aug.
Article in English | MEDLINE | ID: mdl-12865351

ABSTRACT

This study reports the characterization of a recombinant adenoviral vector containing a tetracycline-regulatable promoter, driving the bicistronic expression of the human H2 preprorelaxin (hH2) cDNA and enhanced green fluorescent protein, via an internal ribosomal entry site. An hH2 ELISA was used to measure the secreted levels of recombinant hH2 in transfected canine (CF33.Mt) and human (MDA-MB-435) mammary cancer cell lines over a 6-d period; secreted peptide peaked on d 2 and 4 for the canine and human cell types, respectively. An unprocessed hH2 immunoreactive form of approximately 18 kDa was identified by Western blotting analysis and confirmed by mass spectrometry, suggesting that prorelaxin remains unprocessed in these cell types. The biological activity of the adenovirally expressed human prorelaxin was measured in the established human monocytic cell line THP-1 cAMP ELISA and in an in vitro Transwell cell migration system. Exogenous recombinant hH2 and adenovirally-mediated delivery of prorelaxin to CF33.Mt cells conferred a significant migratory action in the cells, compared with controls. Cell proliferation assays were performed to discount the possibility that the effect of relaxin was mitogenic. Thus, we have demonstrated that prorelaxin has the ability to facilitate cell migration processes exclusive of its ability to stimulate cell proliferation. In validating this adenovirus-based system, we have created a potential tool for further exploration of the physiology of relaxin in mammalian systems.


Subject(s)
Adenoviridae/genetics , Mammary Neoplasms, Animal/pathology , Neoplasm Invasiveness , Protein Precursors/genetics , Relaxin/genetics , Animals , Blotting, Western , Cell Division , Cell Line , Cell Movement , Culture Media, Conditioned/chemistry , Dogs , Gene Expression , Genetic Vectors , Green Fluorescent Proteins , Humans , Luminescent Proteins/genetics , Mass Spectrometry , Monocytes/metabolism , Protein Precursors/analysis , Protein Precursors/physiology , Recombinant Proteins/analysis , Relaxin/analysis , Relaxin/physiology , Transfection
18.
J Biol Chem ; 277(33): 29874-80, 2002 Aug 16.
Article in English | MEDLINE | ID: mdl-12019269

ABSTRACT

PYRIN-containing Apaf1-like proteins (PYPAFs) are members of the nucleotide-binding site/leucine-rich repeat (NBS/LRR) family of signal transduction proteins. We report here that PYPAF7 is a novel PYPAF protein that activates inflammatory signaling pathways. The expression of PYPAF7 is highly restricted to immune cells, and its gene maps to chromosome 19q13.4, a locus that contains a cluster of genes encoding numerous PYPAF family members. Co-expression of PYPAF7 with ASC results in the recruitment of PYPAF7 to distinct cytoplasmic loci and a potent synergistic activation of NF-kappa B. To identify other proteins involved in PYPAF7 and ASC signaling pathways, we performed a mammalian two-hybrid screen and identified pro-caspase-1 as a binding partner of ASC. Co-expression of PYPAF7 and ASC results in the synergistic activation of caspase-1 and a corresponding increase in secretion of interleukin-1 beta. In addition, PYPAF1 induces caspase-1-dependent cytokine processing when co-expressed with ASC. These findings indicate that PYPAF family members participate in inflammatory signaling by regulating the activation of NF-kappa B and cytokine processing.


Subject(s)
Carrier Proteins/physiology , Caspase 1/metabolism , Cytokines/metabolism , Intracellular Signaling Peptides and Proteins , NF-kappa B/metabolism , Amino Acid Sequence , Animals , COS Cells , Carrier Proteins/chemistry , Carrier Proteins/genetics , Carrier Proteins/metabolism , Cell Line , Chromosome Mapping , Chromosomes, Human, Pair 19 , Humans , Molecular Sequence Data , Sequence Homology, Amino Acid
19.
J Biol Chem ; 277(16): 13952-8, 2002 Apr 19.
Article in English | MEDLINE | ID: mdl-11821383

ABSTRACT

Caspase-associated recruitment domains (CARD) are protein-protein interaction modules found extensively in proteins that play important roles in apoptosis, NFkappaB activation, and cytokine regulation. In this study we identified a novel human protein, CARD-8, which contains a C-terminal CARD domain with high similarity to the CARD domain of caspase-1/ICE. We demonstrate that CARD-8 interacts physically with caspase-1 and negatively regulates caspase-1-dependent IL-1beta generation in the THP-1 monocytic cell line. CARD-8 binds also to ICEBERG and pseudo-ICE, two other recently identified proteins, which bind to the CARD domain of caspase-1 and negatively regulate its activity. Reverse transcriptase-PCR analysis revealed that CARD-8 is expressed mainly in monocytes, placenta, lymph nodes, and spleen. This pattern of expression is consistent with caspase-1 expression in the same cells and tissues. CARD-8 was also found to negatively regulate NF-kappaB activation by TNF-alpha stimulation and by ectopically expressed RICK, suggesting that this protein may control cell survival. Consistent with these results, stable expression of CARD-8 in U937 or THP-1 cells sensitizes the cells to differentiation-induced apoptosis. Overexpression of CARD-8 can also induce apoptosis in transfected cells. The results suggest that CARD-8 represents a new signaling molecule involved in the regulation of caspase-1 and NF-kappaB activation.


Subject(s)
Adaptor Proteins, Signal Transducing , Apoptosis , Carrier Proteins/chemistry , Carrier Proteins/metabolism , Caspase 1/metabolism , Neoplasm Proteins , Nucleoside-Phosphate Kinase/metabolism , Amino Acid Sequence , CARD Signaling Adaptor Proteins , Cell Line , DNA, Complementary/metabolism , Enzyme Activation , Humans , Interleukin-1/metabolism , Luciferases/metabolism , Molecular Sequence Data , NF-kappa B/metabolism , Precipitin Tests , Protein Structure, Tertiary , RNA, Messenger/metabolism , Sequence Homology, Amino Acid , Time Factors , Tissue Distribution , Transfection , Tumor Cells, Cultured , Tumor Necrosis Factor-alpha/metabolism
20.
J Biol Chem ; 277(13): 11570-5, 2002 Mar 29.
Article in English | MEDLINE | ID: mdl-11786556

ABSTRACT

The PYRIN domain is a recently identified protein-protein interaction domain that is found at the N terminus of several proteins thought to function in apoptotic and inflammatory signaling pathways. We report here that PYPAF1 (PYRIN-containing Apaf1-like protein 1) is a novel PYRIN-containing signaling protein that belongs to the nucleotide-binding site/leucine-rich repeat (NBS/LRR) family of signaling proteins. The expression of PYPAF1 is highly restricted to immune cells, and its gene maps to chromosome 1q44, a locus that is associated with the rare inflammatory diseases Muckle-Wells syndrome and familial cold urticaria. To identify downstream signaling partners of PYPAF1, we performed a mammalian two-hybrid screen and identified ASC as a PYRIN-containing protein that interacts selectively with the PYRIN domain of PYPAF1. When expressed in cells, ASC recruits PYPAF1 to distinct cytoplasmic loci and induces the activation of NF-kappaB. Furthermore, coexpression of PYPAF1 with ASC results in a potent synergistic activation of NF-kappaB. These findings suggest that PYPAF1 and ASC function as upstream activators of NF-kappaB signaling.


Subject(s)
Carrier Proteins/metabolism , Cytoskeletal Proteins/metabolism , NF-kappa B/metabolism , Amino Acid Sequence , CARD Signaling Adaptor Proteins , Carrier Proteins/chemistry , Carrier Proteins/physiology , Molecular Sequence Data , NLR Family, Pyrin Domain-Containing 3 Protein , Sequence Homology, Amino Acid , Signal Transduction
SELECTION OF CITATIONS
SEARCH DETAIL