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.
Cell Mol Gastroenterol Hepatol ; 12(4): 1297-1310, 2021.
Article in English | MEDLINE | ID: mdl-34118488

ABSTRACT

BACKGROUND & AIMS: Patients with simple steatosis (SS) and nonalcoholic steatohepatitis can develop progressive liver fibrosis, which is associated with liver-related mortality. The mechanisms contributing to liver fibrosis development in SS, however, are poorly understood. SS is characterized by hepatocellular free fatty acid (FFA) accumulation without lobular inflammation seen in nonalcoholic steatohepatitis. Because the Hippo signaling transcriptional coactivator YAP1 (YAP) has previously been linked with nonalcoholic fatty liver disease (NAFLD)-related fibrosis, we sought to explore how hepatocyte FFAs activate a YAP-mediated profibrogenic program. METHODS: We analyzed RNA sequencing data from a GEO DataSet (accession: GSE162694) consisting of 143 patients with NAFLD. We also performed immunohistochemical, immunofluorescence, immunoblot, and quantitative reverse-transcription polymerase chain reaction analyses (qRT-PCR) in liver specimens from NAFLD subjects, from a murine dietary NAFLD model, and in FFA-treated hepatic spheroids and hepatocytes. RESULTS: YAP-target gene expression correlated with increasing fibrosis stage in NAFLD patients and was associated with fibrosis in mice fed a NAFLD-inducing diet. Hepatocyte-specific YAP deletion in the murine NAFLD model attenuated diet-induced fibrosis, suggesting a causative role of YAP in NAFLD-related fibrosis. Likewise, in hepatic spheroids composed of Huh7 hepatoma cells and primary human hepatic stellate cells, Huh7 YAP silencing reduced FFA-induced fibrogenic gene expression. Notably, inhibition of p38 mitogen-activated protein kinase could block YAP activation in FFA-treated Huh7 cells. CONCLUSIONS: These studies provide further evidence for the pathological role of YAP in NAFLD-associated fibrosis and that YAP activation in NAFLD may be driven by FFA-induced p38 MAPK activation.


Subject(s)
Fatty Acids/metabolism , Liver Cirrhosis/etiology , Liver Cirrhosis/metabolism , YAP-Signaling Proteins/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism , Animals , Biomarkers , Computational Biology/methods , Disease Models, Animal , Disease Progression , Disease Susceptibility , Female , Gene Expression Profiling , Gene Expression Regulation , Hepatocytes/metabolism , Humans , Immunohistochemistry , Liver Cirrhosis/pathology , Liver Function Tests , Male , Mice , Models, Biological , Non-alcoholic Fatty Liver Disease/etiology , Non-alcoholic Fatty Liver Disease/metabolism , Non-alcoholic Fatty Liver Disease/pathology
2.
Nat Struct Mol Biol ; 22(6): 476-484, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25961797

ABSTRACT

The molecular circadian clock in mammals is generated from transcriptional activation by the bHLH-PAS transcription factor CLOCK-BMAL1 and subsequent repression by PERIOD and CRYPTOCHROME (CRY). The mechanism by which CRYs repress CLOCK-BMAL1 to close the negative feedback loop and generate 24-h timing is not known. Here we show that, in mouse fibroblasts, CRY1 competes for binding with coactivators to the intrinsically unstructured C-terminal transactivation domain (TAD) of BMAL1 to establish a functional switch between activation and repression of CLOCK-BMAL1. TAD mutations that alter affinities for co-regulators affect the balance of repression and activation to consequently change the intrinsic circadian period or eliminate cycling altogether. Our results suggest that CRY1 fulfills its role as an essential circadian repressor by sequestering the TAD from coactivators, and they highlight regulation of the BMAL1 TAD as a critical mechanism for establishing circadian timing.


Subject(s)
ARNTL Transcription Factors/metabolism , Circadian Clocks , Cryptochromes/metabolism , Animals , Cells, Cultured , Fibroblasts/physiology , Humans , Mice , Mice, Knockout
3.
PLoS Genet ; 10(4): e1004244, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24699442

ABSTRACT

In animals, circadian rhythms in physiology and behavior result from coherent rhythmic interactions between clocks in the brain and those throughout the body. Despite the many tissue specific clocks, most understanding of the molecular core clock mechanism comes from studies of the suprachiasmatic nuclei (SCN) of the hypothalamus and a few other cell types. Here we report establishment and genetic characterization of three cell-autonomous mouse clock models: 3T3 fibroblasts, 3T3-L1 adipocytes, and MMH-D3 hepatocytes. Each model is genetically tractable and has an integrated luciferase reporter that allows for longitudinal luminescence recording of rhythmic clock gene expression using an inexpensive off-the-shelf microplate reader. To test these cellular models, we generated a library of short hairpin RNAs (shRNAs) against a panel of known clock genes and evaluated their impact on circadian rhythms. Knockdown of Bmal1, Clock, Cry1, and Cry2 each resulted in similar phenotypes in all three models, consistent with previous studies. However, we observed cell type-specific knockdown phenotypes for the Period and Rev-Erb families of clock genes. In particular, Per1 and Per2, which have strong behavioral effects in knockout mice, appear to play different roles in regulating period length and amplitude in these peripheral systems. Per3, which has relatively modest behavioral effects in knockout mice, substantially affects period length in the three cellular models and in dissociated SCN neurons. In summary, this study establishes new cell-autonomous clock models that are of particular relevance to metabolism and suitable for screening for clock modifiers, and reveals previously under-appreciated cell type-specific functions of clock genes.


Subject(s)
Adipocytes/metabolism , Circadian Clocks/genetics , Circadian Rhythm/genetics , Hepatocytes/metabolism , Animals , Cell Line , Luciferases/genetics , Mice , Mice, Knockout , NIH 3T3 Cells , Neurons/metabolism , Period Circadian Proteins/genetics , Period Circadian Proteins/metabolism
4.
J Vis Exp ; (67)2012 Sep 27.
Article in English | MEDLINE | ID: mdl-23052244

ABSTRACT

In mammals, many aspects of behavior and physiology such as sleep-wake cycles and liver metabolism are regulated by endogenous circadian clocks (reviewed). The circadian time-keeping system is a hierarchical multi-oscillator network, with the central clock located in the suprachiasmatic nucleus (SCN) synchronizing and coordinating extra-SCN and peripheral clocks elsewhere. Individual cells are the functional units for generation and maintenance of circadian rhythms, and these oscillators of different tissue types in the organism share a remarkably similar biochemical negative feedback mechanism. However, due to interactions at the neuronal network level in the SCN and through rhythmic, systemic cues at the organismal level, circadian rhythms at the organismal level are not necessarily cell-autonomous. Compared to traditional studies of locomotor activity in vivo and SCN explants ex vivo, cell-based in vitro assays allow for discovery of cell-autonomous circadian defects. Strategically, cell-based models are more experimentally tractable for phenotypic characterization and rapid discovery of basic clock mechanisms. Because circadian rhythms are dynamic, longitudinal measurements with high temporal resolution are needed to assess clock function. In recent years, real-time bioluminescence recording using firefly luciferase as a reporter has become a common technique for studying circadian rhythms in mammals, as it allows for examination of the persistence and dynamics of molecular rhythms. To monitor cell-autonomous circadian rhythms of gene expression, luciferase reporters can be introduced into cells via transient transfection or stable transduction. Here we describe a stable transduction protocol using lentivirus-mediated gene delivery. The lentiviral vector system is superior to traditional methods such as transient transfection and germline transmission because of its efficiency and versatility: it permits efficient delivery and stable integration into the host genome of both dividing and non-dividing cells. Once a reporter cell line is established, the dynamics of clock function can be examined through bioluminescence recording. We first describe the generation of P(Per2)-dLuc reporter lines, and then present data from this and other circadian reporters. In these assays, 3T3 mouse fibroblasts and U2OS human osteosarcoma cells are used as cellular models. We also discuss various ways of using these clock models in circadian studies. Methods described here can be applied to a great variety of cell types to study the cellular and molecular basis of circadian clocks, and may prove useful in tackling problems in other biological systems.


Subject(s)
Circadian Clocks/genetics , Gene Expression Profiling/methods , Luciferases/genetics , Luminescent Measurements/methods , 3T3 Cells , Animals , Cell Line, Tumor , HEK293 Cells , Humans , Lentivirus/genetics , Luciferases/biosynthesis , Luciferases/chemistry , Mice , Promoter Regions, Genetic
5.
J Biol Chem ; 287(31): 25917-26, 2012 Jul 27.
Article in English | MEDLINE | ID: mdl-22692217

ABSTRACT

Circadian clocks in mammals are based on a negative feedback loop in which transcriptional repression by the cryptochromes, CRY1 and CRY2, lies at the heart of the mechanism. Despite similarities in sequence, domain structure, and biochemical activity, they play distinct roles in clock function. However, detailed biochemical studies have not been straightforward and Cry function has not been examined in real clock cells using kinetic measurements. In this study, we demonstrate, through cell-based genetic complementation and real-time molecular recording, that Cry1 alone is able to maintain cell-autonomous circadian rhythms, whereas Cry2 cannot. Using this novel functional assay, we identify a cryptochrome differentiating α-helical domain within the photolyase homology region (PHR) of CRY1, designated as CRY1-PHR(313-426), that is required for clock function and distinguishes CRY1 from CRY2. Contrary to speculation, the divergent carboxyl-terminal tail domain (CTD) is dispensable, but serves to modulate rhythm amplitude and period length. Finally, we identify the biochemical basis of their distinct function; CRY1 is a much more potent transcriptional repressor than CRY2, and the strength of repression by various forms of CRY proteins significantly correlates with rhythm amplitude. Taken together, our results demonstrate that CRY1-PHR(313-426), not the divergent CTD, is critical for clock function. These findings provide novel insights into the evolution of the diverse functions of the photolyase/cryptochrome family of flavoproteins and offer new opportunities for mechanistic studies of CRY function.


Subject(s)
Circadian Clocks , Cryptochromes/metabolism , Feedback, Physiological , Amino Acid Motifs , Cryptochromes/chemistry , Cryptochromes/genetics , Fibroblasts/metabolism , Fibroblasts/physiology , Gene Expression Regulation , HEK293 Cells , Humans , Models, Molecular , Protein Structure, Tertiary/physiology , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Sequence Deletion , Structural Homology, Protein , Transcription, Genetic
6.
Bioorg Med Chem Lett ; 21(16): 4913-8, 2011 Aug 15.
Article in English | MEDLINE | ID: mdl-21741835

ABSTRACT

The synthesis and biological evaluation of novel pyrazole and imidazole carboxamides as CB1 antagonists are described. As a part of eastern amide SAR, various chemically diverse motifs were introduced on rimonabant template. The central pyrazole core was also replaced with its conformationally constrained motif and imidazole moieties. In general, a range of modifications were well tolerated. Several molecules with low- and sub-nanomolar potencies were identified as potent CB1 receptor antagonists. The in vivo proof of principle for weight loss is demonstrated with a lead compound in DIO mice model.


Subject(s)
Aminoimidazole Carboxamide/pharmacology , Pyrazoles/pharmacology , Receptor, Cannabinoid, CB1/antagonists & inhibitors , Aminoimidazole Carboxamide/analogs & derivatives , Aminoimidazole Carboxamide/chemistry , Animals , Body Weight/drug effects , Dose-Response Relationship, Drug , Humans , Mice , Mice, Inbred C57BL , Molecular Structure , Pyrazoles/chemical synthesis , Pyrazoles/chemistry , Stereoisomerism , Structure-Activity Relationship
7.
J Neurosci ; 31(23): 8342-50, 2011 Jun 08.
Article in English | MEDLINE | ID: mdl-21653839

ABSTRACT

Circadian clocks sustain daily oscillations in gene expression, physiology, and behavior, relying on transcription-translation feedback loops of clock genes for rhythm generation. Cultured astrocytes display daily oscillations of extracellular ATP, suggesting that ATP release is a circadian output. We hypothesized that the circadian clock modulates ATP release via mechanisms that regulate acute ATP release from glia. To test the molecular basis for circadian ATP release, we developed methods to measure in real-time ATP release and Bmal1::dLuc circadian reporter expression in cortical astrocyte cultures from mice of different genotypes. Daily rhythms of gene expression required functional Clock and Bmal1, both Per1 and Per2, and both Cry1 and Cry2 genes. Similarly, high-level, circadian ATP release also required a functional clock mechanism. Whereas blocking IP(3) signaling significantly disrupted ATP rhythms with no effect on Bmal1::dLuc cycling, blocking vesicular release did not alter circadian ATP release or gene expression. We conclude that astrocytes depend on circadian clock genes and IP(3) signaling to express daily rhythms in ATP release.


Subject(s)
Adenosine Triphosphate/metabolism , Astrocytes/metabolism , Cerebral Cortex/metabolism , Circadian Rhythm/physiology , Analysis of Variance , Animals , Astrocytes/cytology , CLOCK Proteins/genetics , CLOCK Proteins/metabolism , Cells, Cultured , Cerebral Cortex/cytology , Cryptochromes/genetics , Cryptochromes/metabolism , Immunohistochemistry , Mice , Period Circadian Proteins/genetics , Period Circadian Proteins/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism
8.
Bioorg Med Chem Lett ; 21(1): 562-8, 2011 Jan 01.
Article in English | MEDLINE | ID: mdl-21075633

ABSTRACT

The synthesis and biological evaluation of novel pyrazole-3-carboxamide derivatives as CB1 antagonists are described. As a part of eastern amide SAR, various chemically diverse motifs were introduced. In general, a range of modifications were well tolerated. Several molecules with high polar surface area were also identified as potent CB1 receptor antagonists. The in vivo proof of principle for weight loss is exemplified with a lead compound from this series.


Subject(s)
Amides/chemistry , Pyrazoles/chemistry , Receptor, Cannabinoid, CB1/antagonists & inhibitors , Tetrazoles/chemistry , Administration, Oral , Amides/chemical synthesis , Amides/pharmacology , Animals , Mice , Pyrazoles/chemical synthesis , Pyrazoles/pharmacology , Rats , Receptor, Cannabinoid, CB1/metabolism , Structure-Activity Relationship , Tetrazoles/chemical synthesis , Tetrazoles/pharmacology , Weight Loss/drug effects
9.
Thromb Res ; 126(4): 299-305, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20728920

ABSTRACT

BACKGROUND: Cardiovascular disease remains the leading cause of morbidity and premature mortality in most industrialized countries as well as in developing nations. A pro-oxidative state appears to promote and/or exacerbate vascular disease complications. Furthermore, a state of low-grade chronic inflammation can promote increased oxidative stress and lead to endothelial cell and platelet dysfunction ultimately contributing to thrombogenesis. OBJECTIVES: In this study, the effect of a proprietary astaxanthin prodrug (CDX-085) on thrombus formation was investigated using a mouse model of arterial thrombosis. The influence of free astaxanthin, the active drug of CDX-085, on human endothelial cells and rat platelets was evaluated to investigate potential mechanisms of action. METHODS AND RESULTS: Oral administration of CDX-085 (0.4% in chow, approximately 500 mg/kg/day) to 6-8 week old C57BL/6 male mice for 14 days resulted in significant levels of free astaxanthin in the plasma, liver, heart and platelets. When compared to control mice, the CDX-085 fed group exhibited significant increases in basal arterial blood flow and significant delays in occlusive thrombus formation following the onset of vascular endothelial injury. Primary human umbilical vein endothelial cells (HUVECs) and platelets isolated from Wistar-Kyoto rats treated with free astaxanthin demonstrated significantly increased levels of released nitric oxide (NO) and significantly decreased peroxynitrite (ONOO-) levels. CONCLUSION: Observations of increased NO and decreased ONOO- levels in endothelial cells and platelets support a potential mechanism of action for astaxanthin (CDX-085 active drug). These studies support the potential of CDX-085 and its metabolite astaxanthin in the treatment or prevention of thrombotic cardiovascular complications.


Subject(s)
Fibrinolytic Agents/therapeutic use , Prodrugs/therapeutic use , Thrombosis/drug therapy , Administration, Oral , Animals , Blood Flow Velocity/drug effects , Blood Platelets/drug effects , Blood Platelets/metabolism , Cells, Cultured , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Endothelium, Vascular/cytology , Fibrinolytic Agents/administration & dosage , Fibrinolytic Agents/pharmacokinetics , Humans , Male , Mice , Mice, Inbred C57BL , Nitric Oxide/metabolism , Peroxynitrous Acid/metabolism , Prodrugs/administration & dosage , Prodrugs/pharmacokinetics , Rats , Rats, Wistar , Thrombosis/physiopathology , Vasodilator Agents/administration & dosage , Vasodilator Agents/pharmacokinetics , Vasodilator Agents/therapeutic use , Xanthophylls/administration & dosage , Xanthophylls/pharmacokinetics , Xanthophylls/therapeutic use
SELECTION OF CITATIONS
SEARCH DETAIL
...