Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 207
Filter
1.
Sci Adv ; 6(41)2020 10.
Article in English | MEDLINE | ID: mdl-33036982

ABSTRACT

Gene and protein expressions display circadian oscillations, which can be disrupted in diseases in most body organs. Whether these oscillations occur in the healthy hippocampus and whether they are altered in epilepsy are not known. We identified more than 1200 daily oscillating transcripts in the hippocampus of control mice and 1600 in experimental epilepsy, with only one-fourth oscillating in both conditions. Comparison of gene oscillations in control and epilepsy predicted time-dependent alterations in energy metabolism, which were verified experimentally. Although aerobic glycolysis remained constant from morning to afternoon in controls, it increased in epilepsy. In contrast, oxidative phosphorylation increased in control and decreased in epilepsy. Thus, the control hippocampus shows circadian molecular remapping, which is altered in epilepsy. We suggest that the hippocampus operates in a different functioning mode in epilepsy. These alterations need to be considered when studying epilepsy mechanisms, designing drug treatments, and timing their delivery.


Subject(s)
Epilepsy, Temporal Lobe , Epilepsy , Animals , Epilepsy, Temporal Lobe/genetics , Epilepsy, Temporal Lobe/metabolism , Hippocampus/metabolism , Mice , Proteome/metabolism , Transcriptome
2.
Diabetes Obes Metab ; 17 Suppl 1: 17-22, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26332964

ABSTRACT

The circadian clock controls a large variety of neuronal, endocrine, behavioural and physiological responses in mammals. This control is exerted in large part at the transcriptional level on genes expressed in a cyclic manner. A highly specialized transcriptional machinery based on clock regulatory factors organized in feedback autoregulatory loops governs a significant portion of the genome. These oscillations in gene expression are paralleled by critical events of chromatin remodelling that appear to provide plasticity to circadian regulation. Specifically, the nicotinamide adenine dinucleotide (NAD)(+) -dependent deacetylases SIRT1 and SIRT6 have been linked to circadian control of gene expression. This, and additional accumulating evidence, shows that the circadian epigenome appears to share intimate links with cellular metabolic processes and has remarkable plasticity showing reprogramming in response to nutritional challenges. In addition to SIRT1 and SIRT6, a number of chromatin remodellers have been implicated in clock control, including the histone H3K4 tri-methyltransferase MLL1. Deciphering the molecular mechanisms that link metabolism, epigenetic control and circadian responses will provide valuable insights towards innovative strategies of therapeutic intervention.


Subject(s)
CLOCK Proteins/genetics , Chromatin Assembly and Disassembly , Circadian Rhythm/genetics , Mammals/genetics , Transcription, Genetic , Animals , Cells/metabolism , Epigenomics , Gene Expression , Humans , NAD/genetics , NAD/metabolism , Nutritional Physiological Phenomena/genetics , Sirtuin 1/genetics , Sirtuins/genetics
3.
Trends Cell Biol ; 24(6): 329-31, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24794425

ABSTRACT

Circadian oscillations play a critical role in coordinating the physiology, homeostasis, and behavior of biological systems. Once thought to only be controlled by a master clock, recent high-throughput experiments suggest many genes and metabolites in a cell are potentially capable of circadian oscillations. Each cell can reprogram itself and select a relatively small fraction of this broad repertoire for circadian oscillations, as a result of genetic, environmental, and even diet changes.


Subject(s)
Circadian Rhythm , Animals , Biological Clocks , Circadian Rhythm Signaling Peptides and Proteins/genetics , Gene Expression Profiling , Gene Expression Regulation , Metabolome
4.
Neuroscience ; 264: 76-87, 2014 Apr 04.
Article in English | MEDLINE | ID: mdl-24486964

ABSTRACT

Experimental and epidemiological evidence reveal the profound influence that industrialized modern society has imposed on human social habits and physiology during the past 50 years. This drastic change in life-style is thought to be one of the main causes of modern diseases including obesity, type 2 diabetes, mental illness such as depression, sleep disorders, and certain types of cancer. These disorders have been associated to disruption of the circadian clock, an intrinsic time-keeper molecular system present in virtually all cells and tissues. The circadian clock is a key element in homeostatic regulation by controlling a large array of genes implicated in cellular metabolism. Importantly, intimate links between epigenetic regulation and the circadian clock exist and are likely to prominently contribute to the plasticity of the response to the environment. In this review, we summarize some experimental and epidemiological evidence showing how environmental factors such as stress, drugs of abuse and changes in circadian habits, interact through different brain areas to modulate the endogenous clock. Furthermore we point out the pivotal role of the deacetylase silent mating-type information regulation 2 homolog 1 (SIRT1) as a molecular effector of the environment in shaping the circadian epigenetic landscape.


Subject(s)
Brain/metabolism , Circadian Clocks/genetics , Epigenesis, Genetic , Animals , Brain/physiology , Brain/physiopathology , Circadian Clocks/physiology , Humans , Male , Sirtuin 1/physiology , Stress, Psychological/genetics , Stress, Psychological/physiopathology
5.
Article in English | MEDLINE | ID: mdl-22179986

ABSTRACT

The mammalian cell contains a molecular clock that contributes, within each organism, to circadian rhythms and variety of physiological and metabolic processes. The clock machinery is constituted by interwined transcriptional-translational feedback loops that, through the action of specific transcription factors, modulate the expression of clock-controlled genes. These oscillations in gene expression necessarily implicate events of chromatin remodeling on a relatively large, global scale, considering that as many 10% of cellular transcripts oscillate in a circadian manner. CLOCK, a transcription factor crucial for circadian function, has intrinsic histone acetyltransferase activity and operates within a large nuclear complex with other chromatin remodelers. CLOCK directs the cyclic acetylation of the histone H3 and of its own partner BMAL1. A search for the histone deacetylase (HDAC) that counterbalanced CLOCK activity revealed that SIRT1, a nicotinamide adenine dinucleotide (NAD(+))-dependent HDAC, functions in a circadian manner. Importantly, SIRT1 is a regulator of several metabolic processes and was found to interact with CLOCK and to be recruited to circadian promoters in a cyclic manner. As many transcripts that oscillate in mammalian peripheral tissues encode proteins that have central roles in metabolic processes, these findings establish a functional and molecular link among energy balance, chromatin remodeling, and circadian physiology.


Subject(s)
Circadian Clocks/physiology , Metabolism , NAD/metabolism , Sirtuin 1/metabolism , Animals , Chromatin Assembly and Disassembly , Humans , Models, Biological
8.
Horm Metab Res ; 39(8): 560-6, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17712720

ABSTRACT

The MC2-Receptor (melanocortin 2 receptor, MC2-R) is a Gs-protein coupled receptor that is upregulated by its own ligand ACTH and by forskolin. The mechanisms regulating MC2-R expression are still unclear. We therefore investigated the role of the stimulatory transcription factors CREB and CREM and the inhibitory factor ICER for regulation of human MC2-R expression. We cotransfected mouse adrenocortical Y1 cells with luciferase reporter gene vectors containing full length and deleted human MC2-R promoter constructs with expression plasmids for CREB, CREBS133A, CREMtau, CREMtauS117A, or ICER. Direct protein-DNA interaction was investigated by EMSA. Wild type CREB did not significantly affect promoter activity due to high endogenous CREB activity. However, CREBS133A decreased forskolin stimulated MC2-R promoter activity by 48+/-5% (mean+/-SEM) while unstimulated values remained unchanged. CREMtau moderately increased basal and forskolin stimulated luciferase activity in a dose-dependent manner (maximum effect 252+/-24% and 186+/-13% VS. control vector, respectively). While this effect required the full length promoter, cAMP stimulation was retained in shorter constructs. ICER reduced basal luciferase activity in Y1 cells by 17+/-28%, but completely abolished forskolin stimulation. Although 5'-deletion constructs mapped the minimum promoter region required for ICER effect to the shortest -64/+40 construct, direct protein DNA interaction in this promoter region could not be identified by EMSA. Moreover, mutation of the SF-1 binding sites, which retained ICER dependent inhibition, excluded SF-1 to be required for this effect. We conclude from these data that transcription factors of the CREB/CREM/ATF family have a moderate effect on human MC2-R promoter activity, but seem to play a minor role in transmitting stimulation of the cAMP pathway to increased MC2-R expression.


Subject(s)
Adrenal Cortex/metabolism , Cyclic AMP Response Element Modulator/physiology , Cyclic AMP Response Element-Binding Protein/physiology , Receptor, Melanocortin, Type 2/genetics , Animals , Cell Line , Colforsin/pharmacology , Cyclic AMP/pharmacology , Gene Expression Regulation , Humans , Mice , Promoter Regions, Genetic/drug effects , Transfection
9.
Article in English | MEDLINE | ID: mdl-18419267

ABSTRACT

The molecular machinery that governs circadian rhythmicity is based on clock gene products organized in regulatory feedback loops. Recently, we have shown that CLOCK, a master circadian regulator, has histone acetyltransferase activity essential for clock gene expression. The Lys-14 residue of histone H3 is a preferential target of CLOCK-mediated acetylation. As the role of chromatin remodeling in eukaryotic transcription is well recognized, this finding identified unforeseen links between histone acetylation and cellular physiology. Indeed, we have shown that the enzymatic function of CLOCK drives circadian control. We reasoned that CLOCK's acetyltransferase activity could also target nonhistone proteins, a feature displayed by other HATs. Indeed, CLOCK also acetylates a nonhistone substrate: its own partner, BMAL1. This protein undergoes rhythmic acetylation in the mouse liver, with a timing that parallels the down-regulation of circadian transcription of clock-controlled genes. BMAL1 is specifically acetylated on a unique, highly conserved Lys-537 residue. This acetylation facilitates recruitment of the repressor CRY1 to BMAL1, indicating that CLOCK may intervene in negative circadian regulation. Our findings reveal that the enzymatic interplay between two clock core components is crucial for the circadian machinery.


Subject(s)
Chromatin Assembly and Disassembly/physiology , Circadian Rhythm/physiology , Trans-Activators/physiology , ARNTL Transcription Factors , Acetylation , Amino Acid Sequence , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/physiology , CLOCK Proteins , Chromatin Assembly and Disassembly/genetics , Circadian Rhythm/genetics , Gene Expression , Histone Acetyltransferases/genetics , Histone Acetyltransferases/physiology , Humans , Models, Biological , Molecular Sequence Data , Sequence Homology, Amino Acid , Suprachiasmatic Nucleus/physiology , Trans-Activators/genetics
10.
J Neuroendocrinol ; 18(1): 42-9, 2006 Jan.
Article in English | MEDLINE | ID: mdl-16451219

ABSTRACT

We have shown recently that the rapid decline in corticotrophin-releasing hormone (CRH) transcription following activation by stress is associated with induction and binding to the CRH promoter of the repressor isoforms of cAMP responsive element modulator (CREM), inducible cAMP early repressor (ICER). The ability of ICER to inhibit CRH transcription was examined in the hypothalamic cell line, 4B, which expresses CRH. Co-transfection of the inhibitory isoforms of CREM, ICER I and II and CREMbeta, and CRH promoter-luciferase constructs in 4B cells blunted basal and forskolin-stimulated CRH promoter activity, an effect which was abolished by mutation of the CRE of the CRH promoter. Western blot analyses and electromobility gel-shift and super-shift showed increases in endogenous ICER after 3 h of incubation with forskolin. Consistent with an inhibitory effect of CREM on CRH transcription, chromatin immunoprecipitation assays in cells transfected with ICER I revealed recruitment of CREM by the CRH promoter in conjunction with decreases in Pol II association. The study shows that generation of ICER following prolonged stimulation with forskolin, or transfection of an ICER expression vector in hypothalamic cell lines expressing CRH, is associated with CREM binding to the CRH promoter and transcriptional repression. The data support the hypothesis that induction of repressor isoforms of CREM is part of an intracellular feedback mechanism contributing to the termination of CRH transcription during stimulation.


Subject(s)
Corticotropin-Releasing Hormone/biosynthesis , Cyclic AMP Response Element Modulator/metabolism , Gene Expression Regulation/physiology , Hypothalamus/metabolism , Neurons/metabolism , Analysis of Variance , Animals , Cell Line , Corticotropin-Releasing Hormone/genetics , Cyclic AMP Response Element Modulator/physiology , Down-Regulation , Hypothalamus/cytology , Neurons/cytology , Promoter Regions, Genetic/physiology , RNA, Messenger/metabolism , Rats , Statistics, Nonparametric , Transcriptional Activation/genetics , Transfection
12.
Mol Hum Reprod ; 11(8): 567-74, 2005 Aug.
Article in English | MEDLINE | ID: mdl-16143638

ABSTRACT

The cAMP-responsive element modulator (CREM) gene plays a pivotal role in the mouse spermatogenesis, but its role in the human infertility has not been fully established. We performed a mutation screening in 13 Slovenian men with round spermatid arrest and in six controls. Eleven genetic changes have been identified in the human CREM gene, three novel single-nucleotide polymorphisms [within the promoters P1, P3 and intervening sequence 1 (IVS1)], one insertion (IVS2) and one non-sense mutation (exon gamma). Some infertile patients seem to accumulate potentially harmful genetic changes. We identified a patient with no CREM immunoreactive protein that was homozygous for the nucleotide changes in all promoters, IVS 1, 2, 6, and was heterozygous for the mutation in exon gamma. Interestingly, insertion in IVS2 (IVS2-58_55insT) results in a four-fold decrease in binding of nuclear proteins. Computer predictions suggested the presence of a potential novel CREM promoter, however, random amplification of cDNA ends from the human testis cDNA library was not successful in confirming a novel transcription start site of the CREM gene. Screening of a larger number of patients and controls is required to elucidate whether the observed combinations of genetic changes in the CREM gene can explain some forms of male infertility.


Subject(s)
Cyclic AMP Response Element Modulator/genetics , Cyclic AMP/physiology , Infertility, Male/genetics , Response Elements/genetics , Adult , Base Sequence , Chromosomes, Artificial, Bacterial , Cyclic AMP Response Element Modulator/metabolism , Genetic Markers , Haplotypes , Humans , Male , Molecular Sequence Data , Mutation , Sequence Alignment , Sequence Analysis, DNA , Testis/metabolism
13.
J Mol Endocrinol ; 30(3): 381-97, 2003 Jun.
Article in English | MEDLINE | ID: mdl-12790807

ABSTRACT

Transcriptional induction by cAMP is mediated through the interaction of the cAMP response-element binding protein (CREB) with a cAMP response element (CRE) in the promoter of target genes. The steroidogenic acute regulatory (StAR) protein gene is regulated by cAMP-mediated signaling in steroidogenic cells even though its promoter lacks a consensus CRE. Previously, we have identified three highly conserved 5'-CRE half-sites within the -96/-67 bp region of the mouse StAR gene, and a member of the CREB family (CREB/CRE modulator (CREM)) was shown to be involved in its expression and regulation. Here we show that CREB and CREMtau (but not CREMalpha and CREMbeta) have qualitatively similar effects on StAR promoter activity in response to (Bu)(2)cAMP. Studies on the effects of the functional integrity of the CRE half-sites on CREB-dependent (Bu)(2)cAMP-mediated StAR gene transcription demonstrated the greater importance of the CRE2 site in comparison with the CRE1 and CRE3 sites. The CRE2 sequence was also found to bind specifically to recombinant CREB protein and nuclear extract from MA-10 mouse Leydig tumor cells. The cAMP and CREB/CREM responsive region (-151/-1 bp) of the mouse StAR promoter also contains three recognition motifs for steroidogenic factor 1 (SF-1). Electrophoretic mobility shift assays and reporter gene analyses demonstrated the involvement of different SF-1 elements in StAR gene expression with the order of importance being SF-1/3>SF-1/1>SF-1/2. Specific mutations that eliminated the binding sites of CRE and SF-1 elements, either alone or in combination, resulted in an attenuation of StAR promoter activity, indicating that CREB and SF-1 can regulate StAR gene transcription in a cooperative fashion. In addition, mammalian two-hybrid assays revealed a high affinity protein-protein interaction between CREB/CREMtau and SF-1 which appeared to be dependent upon CREB protein phosphorylation. These findings further demonstrate CREB's role in StAR gene transcription and also provide evidence that the combined action of CREB/CREMtau and SF-1 results in enhanced activation of the StAR promoter.


Subject(s)
Cyclic AMP Response Element-Binding Protein/physiology , DNA-Binding Proteins/physiology , Phosphoproteins/genetics , Transcription Factors/physiology , Transcription, Genetic/physiology , Animals , Base Sequence , DNA Primers , Electrophoretic Mobility Shift Assay , Fushi Tarazu Transcription Factors , Homeodomain Proteins , Mice , Receptors, Cytoplasmic and Nuclear , Steroidogenic Factor 1 , Tumor Cells, Cultured
14.
Hum Reprod ; 17(3): 710-7, 2002 Mar.
Article in English | MEDLINE | ID: mdl-11870125

ABSTRACT

BACKGROUND: It is now possible for infertile males to father their own genetic children through the technique of ICSI. This prospect has consequently prompted several investigations into the quality of sperm being retrieved from infertile males. One potential risk is the use of aneuploid sperm or spermatids, which might then be transferred to the fertilized oocyte. METHODS: In this investigation, aneuploidy of spermatids was assessed through immunocytochemistry using antibodies directed against chromosome centromeric regions and complexes. Three different types of infertile male mice with phenotypes closely resembling those described in human non-obstructive azoospermia [PP1cgamma-deficient mice, CREM-deficient mice and C57BL/6J.MAC-17(0--23) mice] were examined for chromosome numbers by counting the number of kinetochores in round spermatids using a CREST antiserum. RESULTS: PP1cgamma(-/-) and CREM(-/-) spermatids from infertile mice showed highly significant elevated levels in the rate of aneuploidy compared with wild-type animals (P < 0.0001). Thus infertile males with independent genetic mutations resulting in different histopathologies showed a high risk in the level of aneuploidy in their spermatids. CONCLUSIONS: These results suggest that impaired spermatogenesis may lead to production of aneuploid gametes. Analysis of aneuploidy in gametes from infertile men, coupled with appropriate genetic counselling, is recommended prior to ICSI.


Subject(s)
Aneuploidy , Infertility, Male/genetics , Repressor Proteins , Spermatids/physiology , Animals , Cell Size , Chromosome Aberrations , Cyclic AMP Response Element Modulator , DNA-Binding Proteins/deficiency , DNA-Binding Proteins/genetics , Isoenzymes/deficiency , Isoenzymes/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Knockout/genetics , Mice, Mutant Strains , Mutation/genetics , Phospholipase C gamma , Spermatids/pathology , Type C Phospholipases/deficiency , Type C Phospholipases/genetics
15.
Sci STKE ; 2001(107): re16, 2001 Nov 06.
Article in English | MEDLINE | ID: mdl-11698692

ABSTRACT

The mammalian circadian system is critical for the proper regulation of behavioral and physiological rhythms. The central oscillator, or master clock, is located in the hypothalamic suprachiasmatic nucleus (SCN). Additional circadian clocks are dispersed throughout most organs and tissues of an animal. The most prominent stimuli capable of synchronizing circadian oscillations to the environment is light. This occurs through daily photic signaling to the SCN, which ultimately results in the appropriate phasing of the various biological rhythms. Two critical aspects of circadian biology that will be discussed here are photic signaling and the communication between central and peripheral clocks. After 10 years of investigation, the primary mammalian circadian photoreceptor remains elusive. Recent findings suggest that multiple photoreceptive molecules may contribute to the perception of environmental light cycles. In addition, the relatively recent identification of cell-autonomous peripheral clocks has opened up an entirely new area of investigation. Deciphering the communication networks responsible for harmonious central and peripheral clock function is a critical step toward the development of effective therapies for circadian-related disorders.


Subject(s)
Biological Clocks/physiology , Circadian Rhythm/physiology , Photoreceptor Cells, Vertebrate/physiology , Signal Transduction/physiology , Animals , Biological Clocks/genetics , Circadian Rhythm/genetics , Humans , Light , Photic Stimulation , Photoreceptor Cells, Vertebrate/metabolism , Signal Transduction/genetics , Suprachiasmatic Nucleus/metabolism , Suprachiasmatic Nucleus/physiology
16.
Mol Cell Biol ; 21(20): 7089-96, 2001 Oct.
Article in English | MEDLINE | ID: mdl-11564891

ABSTRACT

The protein kinase ribosomal S6 kinase 2 (RSK2) has been implicated in phosphorylation of transcription factor CREB and histone H3 in response to mitogenic stimulation by epidermal growth factor. Binding of phospho-CREB to the coactivator CBP allows gene activation through recruitment of the basal transcriptional machinery. Acetylation of H3 by histone acetyltransferase (HAT) activities, such as the one carried by CBP, has been functionally coupled to H3 phosphorylation. While various lines of evidence indicate that coupled histone acetylation and phosphorylation may act in concert to induce chromatin remodeling events facilitating gene activation, little is known about the coupling of the two processes at the signaling level. Here we show that CBP and RSK2 are associated in a complex in quiescent cells and that they dissociate within a few minutes upon mitogenic stimulus. CBP preferentially interacts with unphosphorylated RSK2 in a complex where both RSK2 kinase activity and CBP acetylase activity are inhibited. Dissociation is dependent on phosphorylation of RSK2 on Ser227 and results in stimulation of both kinase and HAT activities. We propose a model in which dynamic formation and dissociation of the CBP-RSK2 complex in response to mitogenic stimulation allow regulated phosphorylation and acetylation of specific substrates, leading to coordinated modulation of gene expression.


Subject(s)
Acetylesterase/metabolism , Gene Expression Regulation, Enzymologic , Mitogens/metabolism , Nuclear Proteins/metabolism , Phosphotransferases/metabolism , Ribosomal Protein S6 Kinases/metabolism , Saccharomyces cerevisiae Proteins , Trans-Activators/metabolism , Acetyltransferases/metabolism , Animals , Blotting, Western , COS Cells , CREB-Binding Protein , Epidermal Growth Factor/pharmacology , Glutathione Transferase/metabolism , Histone Acetyltransferases , Humans , Models, Biological , Phorbol Esters/pharmacology , Phosphorylation , Precipitin Tests , Protein Binding , Recombinant Fusion Proteins/metabolism , Transcription, Genetic , Transcriptional Activation , Transfection , Ultraviolet Rays
18.
Proc Natl Acad Sci U S A ; 98(18): 10178-83, 2001 Aug 28.
Article in English | MEDLINE | ID: mdl-11517315

ABSTRACT

The primary hallmark of circadian clocks is their ability to entrain to environmental stimuli. The dominant, and therefore most physiologically important, entraining stimulus comes from environmental light cycles. Here we describe the establishment and characterization of a new cell line, designated Z3, which derives from zebrafish embryos and contains an independent, light-entrainable circadian oscillator. Using this system, we show distinct and differential light-dependent gene activation for several central clock components. In particular, activation of Per2 expression is shown to be strictly regulated and dependent on light. Furthermore, we demonstrate that Per1, Per2, and Per3 all have distinct responses to light-dark (LD) cycles and light-pulse treatments. We also show that Clock, Bmal1, and Bmal2 all oscillate under LD and dark-dark conditions with similar kinetics, but only Clock is significantly induced while initiating a light-induced circadian oscillation in Z3 cells that have never been exposed to a LD cycle. Finally, our results suggest that Per2 is responsible for establishing the phase of a circadian rhythm entraining to an alternate LD cycle. These findings not only underscore the complexity by which central clock genes are regulated, but also establishes the Z3 cells as an invaluable system for investigating the links between light-dependent gene activation and the signaling pathways responsible for vertebrate circadian rhythms.


Subject(s)
Circadian Rhythm/physiology , Circadian Rhythm/radiation effects , Animals , Cell Line , Circadian Rhythm/genetics , Darkness , Gene Expression/radiation effects , Kinetics , Light , Photoperiod , Zebrafish
19.
EMBO J ; 20(15): 3967-74, 2001 Aug 01.
Article in English | MEDLINE | ID: mdl-11483500

ABSTRACT

A group of specialized genes has been defined to govern the molecular mechanisms controlling the circadian clock in mammals. Their expression and the interactions among their products dictate circadian rhythmicity. Three genes homologous to Drosophila period exist in the mouse and are thought to be major players in the biological clock. Here we present the generation of mice in which the founding member of the family, Per1, has been inactivated by homologous recombination. These mice present rhythmicity in locomotor activity, but with a period almost 1 h shorter than wild-type littermates. Moreover, the expression of clock genes in peripheral tissues appears to be delayed in Per1 mutant animals. Importantly, light-induced phase shifting appears conserved. The oscillatory expression of clock genes and the induction of immediate-early genes in response to light in the master clock structure, the suprachiasmatic nucleus, are unaffected. Altogether, these data demonstrate that Per1 plays a distinct role within the Per family, as it may be involved predominantly in peripheral clocks and/or in the output pathways of the circadian clock.


Subject(s)
Biological Clocks/physiology , Circadian Rhythm/physiology , Nuclear Proteins/physiology , Animals , Base Sequence , Biological Clocks/genetics , Cell Cycle Proteins , Circadian Rhythm/genetics , DNA, Complementary , Female , Gene Expression , Gene Targeting , Light , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Molecular Sequence Data , Mutagenesis , Nuclear Proteins/genetics , Period Circadian Proteins , Proto-Oncogene Proteins c-fos/genetics , RNA, Messenger , Running
20.
Mol Endocrinol ; 15(8): 1255-63, 2001 Aug.
Article in English | MEDLINE | ID: mdl-11463851

ABSTRACT

A key regulatory point in fine tuning of steroidogenesis is the synthesis of steroidogenic acute regulatory protein, which transfers cholesterol into mitochondria. Heat shock and toxic insults reduce steroidogenic acute regulatory protein, severely compromising steroid synthesis. As the molecular mechanisms for this reduction remain elusive, we tested the hypothesis that heat shock directly interferes with transcription of the steroidogenic acute regulatory protein gene. We show that, in mouse MA-10 Leydig tumor cells, heat shock caused drastic declines in (Bu)(2)cAMP-induced progesterone accumulation and steroidogenic acute regulatory protein transcript abundance. A proximal steroidogenic acute regulatory protein promoter fragment (-85 to +39) is sufficient to direct both cAMP inducibility and heat shock inhibition. Nuclear extracts from MA-10 cells displayed binding to this proximal promoter fragment as a low mobility complex in gel shift experiments. This complex disappeared in nuclear extracts taken at 5 and 10 min after initiation of heat shock and reappeared in extracts taken at 2 and 8 h. Similar low- mobility complexes formed on oligonucleotides representing the overlapping subfragments of the minimal steroidogenic acute regulatory protein promoter fragment sensitive to the heat shock effect. Extracts from heat-shocked MA-10 cells displayed reduced complex formation to each of the subfragments. We conclude that heat shock reduces progesterone synthesis, steroidogenic acute regulatory protein mRNA abundance, and steroidogenic acute regulatory protein promoter activity and disrupts binding of nuclear proteins to the proximal region of the steroidogenic acute regulatory protein promoter. Together these observations provide strong evidence for a mechanism of transcriptional inhibition in the down-regulation of steroidogenic acute regulatory protein expression by heat shock.


Subject(s)
Hot Temperature , Phosphoproteins/genetics , Steroids/biosynthesis , Transcription, Genetic , Animals , Blotting, Northern , Blotting, Western , Bucladesine/pharmacology , Cholesterol/metabolism , DNA/metabolism , Leydig Cell Tumor/metabolism , Mice , Mitochondria/metabolism , Nuclear Proteins/metabolism , Progesterone/biosynthesis , Promoter Regions, Genetic , RNA, Messenger/analysis , Transfection , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL
...