Article
To read the full-text of this research, you can request a copy directly from the authors.

Abstract

Nuclear receptors (NRs) control cell fate and regulate tissue function. Some of the NRs are expressed in a circadian and tissue-specific manner. Clock genes are part of the circadian network and fine-tune gene expression in adipose and skeletal tissues. Pparg, a master transcription factor that determines adipogenesis, exhibits a circadian expression pattern in white adipose tissue and liver. Here we report the finding that the message and protein for a peripheral clock gene, nocturnin, is markedly upregulated with Pparg activation in adipocytes and bone marrow stromal cells. Nocturnin is also expressed in relatively high amounts in other tissues that may have physiologic relevance for bone, including the brain and hypothalamus. Of importance, we found polymorphic strain differences in bone marrow nocturnin expression that relate to phenotypic determinants of skeletal acquisition. Defining the function of nocturnin in peripheral tissues should provide new insights into lineage allocation and the intimate relationship between nuclear receptors and physiologic timekeeping.

No full-text available

Request Full-text Paper PDF

To read the full-text of this research,
you can request a copy directly from the authors.

... Consistent with this, there is evidence that mammalian NOCT transcription can be controlled by other transcription factors. Sequence analysis indicates the presence of binding sites for CRE [55], RevERBα [51], NFκB [10,56], and PPARγ [57]; and functional data support regulation by FoxO [58], PPARγ [57,59], STAT3 [60] and Nanog [60][61][62]. Furthermore, CLOCK-independent control of NOCT transcription is known in Xenopus photoreceptor cells and is mediated in part by CREB through a cis-regulatory element termed the Nocturnin Element [63,64]; however, this activity has not been extensively characterized. ...
... Consistent with this, there is evidence that mammalian NOCT transcription can be controlled by other transcription factors. Sequence analysis indicates the presence of binding sites for CRE [55], RevERBα [51], NFκB [10,56], and PPARγ [57]; and functional data support regulation by FoxO [58], PPARγ [57,59], STAT3 [60] and Nanog [60][61][62]. Furthermore, CLOCK-independent control of NOCT transcription is known in Xenopus photoreceptor cells and is mediated in part by CREB through a cis-regulatory element termed the Nocturnin Element [63,64]; however, this activity has not been extensively characterized. ...
... The observation that the NOCT E195 mutant retains RNA decay activity also is significant because mutation of the analogous residue in mouse NOCT was previously used as a tool for analyzing exoribonuclease-independent activity [18]. In light of human NOCT E195A activity, it is uncertain whether the mouse E193A mutation can accurately discriminate exoribonuclease-dependent and -independent activities, confounding the interpretation of data suggesting that NOCT performs a deadenylase-independent function [57]. ...
Article
Full-text available
Post-transcriptional control of messenger RNAs (mRNA) is an important layer of gene regulation that modulates mRNA decay, translation, and localization. Eukaryotic mRNA decay begins with the catalytic removal of the 3′ poly-adenosine tail by deadenylase enzymes. Multiple deadenylases have been identified in vertebrates and are known to have distinct biological roles; among these proteins is Nocturnin, which has been linked to circadian biology, adipogenesis, osteogenesis, and obesity. Multiple studies have investigated Nocturnin’s involvement in these processes; however, a full understanding of its molecular function remains elusive. Recent studies have provided new insights by identifying putative Nocturnin-regulated mRNAs in mice and by determining the structure and regulatory activities of human Nocturnin. This review seeks to integrate these new discoveries into our understanding of Nocturnin’s regulatory functions and highlight the important remaining unanswered questions surrounding its regulation, biochemical activities, protein partners, and target mRNAs.
... Furthermore, pesticide ligands of NRs and AChR disrupt AHR mRNA/HES1 osciltations and the MASH-1/ Notch signaling pathway (Akahoshi et al., 2006). Organotin, for example, activates RXR-PPAR heterodimers that exert their mutagenic and carcinogenic capabilities (Prival et al., 1977), and deregulate the behavioral and circadian rhythms (Ema et al., 1991), resulting in circadian clock disruption, mood and CNS disorders (Logan and McClung, 2019), and global prevalence of obesity by both HFD-induced disruption of metabolic rhythms (Eckel-Mahan et al., 2012) and ER/FFAs/PPARγ signaling activation (Liu et al., 2013), which suppresses the Per1 gene expression (Killilea et al., 2017;Kawai et al., 2010;Kawai et al., 2010;Killilea et al., 2017). ...
... Furthermore, pesticide ligands of NRs and AChR disrupt AHR mRNA/HES1 osciltations and the MASH-1/ Notch signaling pathway (Akahoshi et al., 2006). Organotin, for example, activates RXR-PPAR heterodimers that exert their mutagenic and carcinogenic capabilities (Prival et al., 1977), and deregulate the behavioral and circadian rhythms (Ema et al., 1991), resulting in circadian clock disruption, mood and CNS disorders (Logan and McClung, 2019), and global prevalence of obesity by both HFD-induced disruption of metabolic rhythms (Eckel-Mahan et al., 2012) and ER/FFAs/PPARγ signaling activation (Liu et al., 2013), which suppresses the Per1 gene expression (Killilea et al., 2017;Kawai et al., 2010;Kawai et al., 2010;Killilea et al., 2017). ...
Article
Pesticides are widely-used chemicals commonly applied in agriculture for the protection of crops from pests. Depending on the class of pesticides, the specific substances may have a specific set of adverse effects on humans, especially in cases of acute poisoning. In past years, evidence regarding sequelae of chronic, low-level exposure has been accumulating. Cognitive impairment and dementia heavily affect a person’s quality of life and scientific data has been hinting towards an association between them and antecedent chronic pesticide exposure. Here, we reviewed animal and human studies exploring the association between pesticide exposure, cognition and dementia. Additionally, we present potential mechanisms through which pesticides may act neurotoxically and lead to neurodegeneration. Study designs rarely presented homogeneity and the estimation of the exposure to pesticides has been most frequently performed without measuring the synergic effects and the possible interactions between the toxicants within mixtures, and also overlooking low exposures to environmental toxicants. It is possible that a Real-Life Risk Simulation approach would represent a robust alternative for future studies, so that the safe exposure limits and the net risk that pesticides confer to impaired cognitive function can be examined. Previous studies that evaluated the effect of low dose chronic exposure to mixtures of pesticides and other chemicals intending to simulate real life exposure scenarios showed that hermetic neurobehavioral effects can appear after mixture exposure at doses considered safe for individual compounds and these effects can be exacerbated by a coexistence with specific conditions such as vitamin deficiency. However, there is an overall indication, derived from both epidemiologic and laboratory evidence, supporting an association between exposure to neurotoxic pesticides and cognitive dysfunction, dementia and Alzheimer’s disease.
... As shown in Fig. 2b, the majority of differentially regulated lncRNAs were unique per tissue type. Among the enterohepatic tissues, jejunum had the most uniquely differentially regulated lncRNAs by lack of gut microbiota (576), followed by colon (173), liver (165), duodenum (133), and ileum (39). There were three lncRNAs that were differentially regulated in all five organs, namely NONMMUG019446.2, NONMMUG0 26539.1, and NONMMUG041315.2 (Fig. 2c). ...
... Both NONMMUG026539.1 and Ccrn4l are transcribed from the Watson strand and were down-regulated in the absence of gut microbiota. Ccrn4l knockout mice develop lipid droplets in BAT, suggesting that down-regulation of Ccrn4l may regulate lipid metabolism, and the co-regulated NONMMUG026539.1 may also participate in this pathway [38][39][40]. The lncRNA NONMMUG043919.1 on chromosome 9 consists of two isoforms both transcribed from the 3'-UTR (Fig. 11b) of the sodium coupled neutral amino acid transporter Slc38a3. ...
Article
Full-text available
Background Long non-coding RNAs (lncRNAs) are increasingly recognized as regulators of tissue-specific cellular functions and have been shown to regulate transcriptional and translational processes, acting as signals, decoys, guides, and scaffolds. It has been suggested that some lncRNAs act in cis to regulate the expression of neighboring protein-coding genes (PCGs) in a mechanism that fine-tunes gene expression. Gut microbiome is increasingly recognized as a regulator of development, inflammation, host metabolic processes, and xenobiotic metabolism. However, there is little known regarding whether the gut microbiome modulates lncRNA gene expression in various host metabolic organs. The goals of this study were to 1) characterize the tissue-specific expression of lncRNAs and 2) identify and annotate lncRNAs differentially regulated in the absence of gut microbiome. Results Total RNA was isolated from various tissues (liver, duodenum, jejunum, ileum, colon, brown adipose tissue, white adipose tissue, and skeletal muscle) from adult male conventional and germ-free mice (n = 3 per group). RNA-Seq was conducted and reads were mapped to the mouse reference genome (mm10) using HISAT. Transcript abundance and differential expression was determined with Cufflinks using the reference databases NONCODE 2016 for lncRNAs and UCSC mm10 for PCGs. Although the constitutive expression of lncRNAs was ubiquitous within the enterohepatic (liver and intestine) and the peripheral metabolic tissues (fat and muscle) in conventional mice, differential expression of lncRNAs by lack of gut microbiota was highly tissue specific. Interestingly, the majority of gut microbiota-regulated lncRNAs were in jejunum. Most lncRNAs were co-regulated with neighboring PCGs. STRING analysis showed that differentially expressed PCGs in proximity to lncRNAs form tissue-specific networks, suggesting that lncRNAs may interact with gut microbiota/microbial metabolites to regulate tissue-specific functions. Conclusions This study is among the first to demonstrate that gut microbiota critically regulates the expression of lncRNAs not only locally in intestine but also remotely in other metabolic organs, suggesting that common transcriptional machinery may be shared to transcribe lncRNA-PCG pairs, and lncRNAs may interact with PCGs to regulate tissue-specific pathways. Electronic supplementary material The online version of this article (10.1186/s12864-018-5235-3) contains supplementary material, which is available to authorized users.
... This results in a need to process and store these extra metabolites, and expansion of adipose tissue is one mechanism facilitating lipid storage. Kawai and colleagues found that Noc is highly upregulated in 3T3-L1 cells undergoing adipogenesis [38,39], and similarly, mice chronically exposed to a HFD exhibit increased expression of Noc in epididymal white adipose tissue (eWAT) [40]. Under ad lib feeding conditions on a standard chow diet, Noc expression is not rhythmic in eWAT [5]. ...
... Noc is also significantly induced in mesenchymal stromal cells (MSCs) transfected with Pparg2 (one of two major forms of the PPARγ protein expressed mainly in adipogenic cells) and treated with the PPARγ agonist rosiglitazone [4]. Together, these data suggest that Nocturnin is a transcriptional target of PPARγ [38]. As previously mentioned, a pro-adipogenic function has been associated with Noc as its expression is highly upregulated in cells undergoing adipogenesis and Noc expression is gradually increased in eWAT of mice fed a HFD over 4 generations [39,40]. ...
Article
Many aspects of metabolism exhibit daily rhythmicity under the control of endogenous circadian clocks, and disruptions in circadian timing result in dysfunctions associated with the metabolic syndrome. Nocturnin (Noc) is a robustly rhythmic gene that encodes a deadenylase thought to be involved in the removal of polyA tails from mRNAs. Mice lacking the Noc gene display resistance to diet-induced obesity and hepatic steatosis, due in part to reduced lipid trafficking in the small intestine. In addition, Noc appears to play important roles in other tissues and has been implicated in lipid metabolism, adipogenesis, glucose homeostasis, inflammation and osteogenesis. Therefore, Noc is a potential key post-transcriptional mediator in the circadian control of many metabolic processes.
... Recently our laboratories defined an important link between PPARγ and nocturnin (Noc), a circadian regulated gene that evolved millions of years ago from the yeast family of transcription factors regulating cellular responses to external cues. [12][13][14] The role of this network in the bone micro environment during aging and its relationship to IGF-I informs us about both physiologic and pathologic processes that become operational as the mammalian organism ages. brown fat, the context specific nature and unique location of these cells indicate these cells are likely to have a more specialized function. ...
... Based on these observations, we next performed micro-array analysis to detect genes affected by PPARγ2 in U-33/γ2 cells and found Noc to be one of the most highly upregulated genes with PPARγ2 activation by rosiglitazone ( fig. 5). 6,12 As mentioned earlier, Noc functions as a deadenylase that degrades mRNA from poly-adenylation site of 3'UTR, thus adding a post-transcriptional regulation to gene expression. Importantly, RNA-binding proteins are required for Noc recruitment to RNA and function. ...
Article
Full-text available
Aging is associated with profound changes in bone mass and body composition. Emerging evidence supports the hypothesis that alterations in mesenchymal stromal cell fate are a critical etiologic factor. In addition, time-keeping at the cellular level is affected as aging progresses, particularly in the adipocyte. In this Extra View we discuss the interactive role of three molecules, PPARγ, nocturnin and IGF-I in regulating stem cell fate in the marrow and the potential implications of this network for understanding cellular aging.
... In addition to the molecular circadian clock components, genes regulated by the circadian machinery are shown to affect adipogenesis. Nocturnin, which is a deadenylase, is shown to be upregulated in differentiated adipocytes [38]. A further study, which reported that nocturnin stimulates PPARγ translocation, provides a mechanistic link between nocturnin and adipogenesis [39]. ...
Article
Full-text available
Essential for survival and reproduction, the circadian timing system (CTS) regulates adaptation to cyclical changes such as the light/dark cycle, temperature change, and food availability. The regulation of energy homeostasis possesses rhythmic properties that correspond to constantly fluctuating needs for energy production and consumption. Adipose tissue is mainly responsible for energy storage and, thus, operates as one of the principal components of energy homeostasis regulation. In accordance with its roles in energy homeostasis, alterations in adipose tissue’s physiological processes are associated with numerous pathologies, such as obesity and type 2 diabetes. These alterations also include changes in circadian rhythm. In the current review, we aim to summarize the current knowledge regarding the circadian rhythmicity of adipogenesis, lipolysis, adipokine secretion, browning, and non-shivering thermogenesis in adipose tissue and to evaluate possible links between those alterations and metabolic diseases. Based on this evaluation, potential therapeutic approaches, as well as clock genes as potential therapeutic targets, are also discussed in the context of chronotherapy.
... The temca brains displayed a 52% decrease in the number of p-ERK + neurons compared with controls (Fig. 4 F and G), indicative of impaired neuronal activity. Bulk RNA-seq analysis of whole-brain tissues dissected from control and temca fish placed at 33°C for 1 d identified 649 genes (n = 369 and 280 for up and down-regulated genes, respectively) with significantly changed expression levels (adjusted P [P-adj] < 0.05; fold change > 1.5) (Fig. 4H and Dataset S1), including genes influencing brain function, such as npas4a and fosab (38,39), circadian clock genes (nr1d1, nocta, and ciarta) (40)(41)(42), and steroid biosynthesis genes (hsd11b2, cyp51, and srd5a2b) (43). Gene Ontology analysis of up-regulated genes indicated enrichment in responses to cadmium ions, whereas the down-regulated genes were enriched for steroid/sterol/cholesterol biosynthesis/metabolic processes, and circadian regulation terms (Fig. 4I). ...
Article
Full-text available
Teleost fishes and urodele amphibians can regenerate amputated appendages, whereas this ability is restricted to digit tips in adult mammals. One key component of appendage regeneration is reinnervation of the wound area. However, how innervation is regulated in injured appendages of adult vertebrates has seen limited research attention. From a forward genetics screen for temperature-sensitive defects in zebrafish fin regeneration, we identified a mutation that disrupted regeneration while also inducing paralysis at the restrictive temperature. Genetic mapping and complementation tests identify a mutation in the major neuronal voltage-gated sodium channel (VGSC) gene scn8ab. Conditional disruption of scn8ab impairs early regenerative events, including blastema formation, but does not affect morphogenesis of established regenerates. Whereas scn8ab mutations reduced neural activity as expected, they also disrupted axon regrowth and patterning in fin regenerates, resulting in hypoinnervation. Our findings indicate that the activity of VGSCs plays a proregenerative role by promoting innervation of appendage stumps.
... Transcription factor enrichment analysis revealed peroxisome proliferator-activated receptor gamma (PPARg) as the top hit for both cluster 3 and 4. PPARg senses the energy status of the cell to regulate lipid uptake and storage, and in fact has often been described as an important circadian transcription factor in adipose tissue [26,27]. Its expression is regulated by CCAAT/enhancer-binding protein alpha (CEBPA), which we identified as first hit following PPARg in cluster 4 ( Figure 1B), and its activity is enhanced upon interaction with the circadian protein nocturnin (NOCT) [26,28]. In our dataset, Cebpa expression by itself was found oscillating within cluster 3, while expression of Pparg itself and Noct were not oscillating (data not shown), likely because PPARg is broadly regulated at the posttranscriptional level [26]. ...
Article
Full-text available
Objective: Brown adipose tissue (BAT) burns fatty acids (FAs) to produce heat, and shows diurnal oscillation in glucose and triglyceride (TG)-derived FA-uptake, peaking around wakening. Here we aimed to gain insight in the diurnal regulation of metabolic BAT activity. Methods: RNA-sequencing, chromatin immunoprecipitation (ChIP)-sequencing, and lipidomics analyses were performed on BAT samples of wild type C57BL/6J mice collected at 3-hour intervals throughout the day. Knockout and overexpression models were used to study causal relationships in diurnal lipid handling by BAT. Results: We identified pronounced enrichment of oscillating genes involved in extracellular lipolysis in BAT, accompanied by oscillations of FA and monoacylglycerol content. This coincided with peak lipoprotein lipase (Lpl) expression, and was predicted to be driven by peroxisome proliferator-activated receptor gamma (PPARγ) activity. ChIP-sequencing for PPARγ confirmed oscillation in binding of PPARγ to Lpl. Of the known LPL-modulators, angiopoietin-like 4 (Angptl4) showed the largest diurnal amplitude opposite to Lpl, and both Angptl4 knockout and overexpression attenuated oscillations of LPL activity and TG-derived FA-uptake by BAT. Conclusions: Our findings highlight involvement of PPARγ and a crucial role of ANGPTL4 in mediating the diurnal oscillation of TG-derived FA-uptake by BAT, and imply that time of day is essential when targeting LPL activity in BAT to improve metabolic health.
... NOCT was also reported to negatively regulate osteoblastogenesis and knockout of NOCT resulted in an increase in bone density (10,41,51). Interestingly, we identified RORB mRNA as a NOCT-repressed target. ...
Preprint
Full-text available
Nocturnin (NOCT) is a eukaryotic enzyme that belongs to a superfamily of exoribonucleases, endonucleases, and phosphatases. In this study, we analyze the expression, processing, localization, and cellular functions of human NOCT. We demonstrate that the NOCT protein is differentially expressed and processed in a cell and tissue type specific manner as a means to control its localization to the cytoplasm or mitochondria. Our studies also show that the N-terminus of NOCT is necessary and sufficient to confer mitochondrial localization. We then measured the impact of cytoplasmic NOCT on the transcriptome and report that it regulates the levels of hundreds of mRNAs that are enriched for components of signaling pathways, neurological functions, and regulators of osteoblast differentiation. Recent biochemical data indicate that NOCT dephosphorylates nicotinamide adenine dinucleotide (NAD) metabolites, and thus we measured the effect of NOCT on these cofactors in cells. We find that NOCT increases NAD(H) and decreases NADP(H) levels in a manner dependent on its intracellular localization. Collectively, our data indicate that NOCT can regulate levels of both mRNAs and NADP(H) cofactors in manner specified by its intracellular localization.
... Some studies suggest that this may be related to abnormal functioning of hypothalamus-regulated timekeeping and clock genes (e.g., nocturnin) that are found to be involved in shifting differentiation of mesenchymal stem cells towards adipogenesis in aging. (41)(42)(43)(44) Additionally, reports on association between leptin levels and cognitive function or AD are highly contradictory and this association needs to be further explored. (45,46) Nevertheless, the model of independent neural and neurohumoral arms that contribute to bone loss in individuals with AD is perhaps oversimplified. ...
Article
Full-text available
Background: Animal studies provide strong evidence that the CNS directly regulates bone remodeling through the actions of the hypothalamus via two distinct pathways, the neural (mediated by leptin) arm and neurohumoral (mediated by neurohormones and growth factors) arm. The impact of AD on central regulatory mechanisms of bone mass is not known. Objectives: To test a model that assesses the relationship between hypothalamic atrophy and bone loss in Alzheimer's disease (AD) and potential mediation through neural (leptin) and neurohumoral (insulin-like growth factor -1, IGF-1) mechanisms. Hypotheses: AD-related hypothalamic structural change alters neural and neurohumoral regulatory systems of bone remodeling and contributes to bone loss in early AD. Design: A secondary data analysis of data obtained in a two-year longitudinal study with path analysis and longitudinal mediation modeling. Participants: The data were collected as a part of the University of Kansas Brain Aging Project, a two-year observational study of 71 older adults with early stage AD and 69 non-demented controls. Measurements: Demographic characteristics and measures of bone density, body composition, and hypothalamic volume, serum levels of leptin, growth hormone, and IGF-1 were collected. Results: Hypothalamic atrophy and bone loss were observed in AD group and were associated. Data modeling suggests that bone loss may precede measurable changes in the brain. Leptin increased over two years in AD and the increase in leptin was associated with hypothalamic atrophy. However, changes in leptin or IGF-1 levels did not mediate the relationship between hypothalamic atrophy and bone loss. Conclusions: This study extends previous findings by suggesting that bone loss in AD may be related to neurodegenerative changes (atrophy) in the hypothalamus. Further studies are needed to explore the role of brain atrophy and mediating mechanisms in bone loss. Further exploring temporal relationship between bone loss and AD may have an important diagnostic value.
... Masanobu Kawai et al. reported that NOC-/-mice have reduced bone marrow adiposity in vitro and that in vivo observations were the same [59]. Carla B Green et al. reported that NOC is highly expressed in differentiating adipocytes and is regulated by the activation of PPAR- [61]. Moreover, NOC-mediated nuclear translocation of PPAR-can be blocked by a short peptide fragment of NOC that inhibits its interaction with PPAR- [58]. ...
Article
Full-text available
Background: Under the transcriptional control of numerous factors and intracellular signals, mesenchymal stem cells (MSCs) can differentiate into various cell types, including adipocytes and osteoblasts. However, the precise cellular signaling factors that determine the cell fate of MSCs in bone marrow remain largely unknown. Objective: In this review, we focus on the ties of PPAR-γ and Wnt signaling in MSC differentiation into adipocytes and osteoblasts. Results: Peroxisome proliferator-activated receptor-γ (PPAR-γ) is well established as a prime inducer of adipogenesis, while the Wnt pathway is regarded as the master moderator of osteogenesis. A theoretical inverse relationship exists between adipogenic and osteogenic lineage commitment and differentiation: the differentiation toward an osteoblast phenotype occurs at the expense of an adipocyte phenotype. Conclusion: It has been proposed that the balance between osteogenic and adipogeneic MSC differentiation is disrupted in diverse areas of human health. Therefore, understanding the ties between PPAR- γand Wnt signaling in MSC differentiation has significant implications in diverse areas of human health, from obesity to osteoporosis to regenerative medicine.
... Accumulating evidence indicates that mNoc plays an important role in the interplay between circadian clocks and metabolism (discussed in Douris and Green, 2008;Kawai and Rosen, 2010). mNoc has been implicated in adipogenesis (Kawai et al., 2010b;Kawai et al., 2010c) and bone metabolism (Kawai et al., 2010a). In flies, the Noc homologue gene is required at pupal stage for proper wing morphogenesis (Gronke et al., 2009) and is involved in circadian light responses (Nagoshi et al., 2010). ...
Article
Full-text available
Circadian rhythms are biological variables that oscillate cyclically with a periodclose to 24 h. These oscillations are generated endogenously by mechanisms driven bybiological clocks. The clock at the molecular level is composed of interconnectedfeedback loops in which a set of genes -clock genes- regulates each other, and also, theexpression of an important number of genes -clock controlled genes-, in order toregulate a significant number of rhythmic cellular, metabolic, and physiologicalprocesses. Importantly, about 3-25% of the transcripts expressed in a given tissue/organexhibit a circadian oscillation in their levels, explaining how clocks regulate biochemicaland cellular processes.The current clock model in Eukaryotes involves at least two interconnectedtranscriptional feedback loops. Additionally, the posttranslational modificationsregulating translocations, interactions, and stability of clock proteins, have been alsoshown to be an important component of the clock mechanism. Just in recent years it hasbecome increasingly clear that posttranscriptional processes are also important forunderstanding the clock functioning and kinetics. Presently, we know that severalposttranscriptional events participate in the clock mechanism itself, and in the control ofcircadian gene expression. These events include the regulation of splicing,polyadenylation, mRNA stability, and translation. RNA-binding proteins (RBPs) andmicroRNAs (miRNAs) have been reported to be implicated in most of these processes,and have emerged as important players in controlling gene expression. Importantly, theperturbation of some rhythmic posttranscriptional events has been shown to causemetabolic syndromes and other diseases. The aim of this chapter is to shed light on therole that posttranscriptional regulation plays in the molecular clock mechanism, as wellas in the modulation of circadian gene expression.
... Interestingly, the subcellular localization of nuclear receptor PPARc is under the tight control of Nocturnin (NOC). NOC is a direct downstream target of PPARc [57], and its expression follows a circadian oscillatory pattern with maximum levels observed at night. Further studies revealed that the interactions between PPARc and NOC induce the nuclear translocation of PPARc to enhance PPARc-mediated transcriptional activity [58] (Fig 4). ...
Article
Full-text available
Circadian rhythms characterize almost every aspect of human physiology, endocrinology, xenobiotic detoxification, cell growth, and behavior. Modern lifestyles that disrupt our normal circadian rhythms are increasingly thought to contribute to various disease conditions ranging from depression and metabolic disorders to cancer. This self-sustained time-keeping system is generated and maintained by an endogenous molecular machine, the circadian clock, which is a transcriptional mechanism composed of the transcription factors CLOCK and BMAL and their co-repressors, PER and CRY. Nuclear receptors (NRs) represent a large family of hormone-sensitive transcriptional regulators involved in a myriad of biological processes such as development, energy metabolism, reproduction, inflammation, and tissue homeostasis. Recent studies point not only to NR regulation by the clock, but also to NR regulation of the clock itself. Here, we discuss recent studies that functionally and mechanistically implicate NRs as key components of both the universal and adaptive circadian clock mechanisms. As proven pharmacological targets, nuclear receptors are promising targets for therapeutic control of many pathological conditions associated with the disruption of circadian rhythm.
... Noc was suggested as a linking factor between external stimuli and metabolic output as Noc-deficient mice manifested resistance to diet-induced obesity and hepatic steatosis, together with dampened expression of genes related to lipid metabolism (19). Recently, Noc has been reported to be activated by peroxisome proliferator-activated receptor γ (PPARγ) (20), which in turn stimulates PPARγ nuclear translocation and thus promoting adipogenesis (13). ...
Article
The deadenylase nocturnin (Noc, Ccrn4l) has been recently found to regulate lipid metabolism and to control preadipocyte differentiation. Here, we showed that among the five deadenylases tested, Noc and Pan2 exhibited a biphasic expression which is out of phase to each other during adipocyte differentiation of 3T3-L1 cells. The expression levels of other deadenylases, including Parn, Ccr4, and Caf1, were relatively unchanged or reduced. The immediate early expressed Noc during 3T3-L1 adipogenesis was involved in regulating mitotic clonal expansion (MCE) and cyclin D1 expression, as demonstrated in Noc-silenced 3T3-L1 cells and Noc(-/-) primary mouse embryonic fibroblasts (MEFs). Transcriptional profiling of Noc-depleted 3T3-L1 adipocytes revealed that most of the differentially expressed genes were related to cell growth and proliferation. In human adipose tissue, NOC mRNA level negatively associated with both fasting serum insulin and homeostasis model assessment of insulin resistance, and positively associated with both adiponectin mRNA levels and circulating adiponectin levels. Taken together, these results suggest the role of Noc in the modulation of early adipogenesis as well as systemic insulin sensitivity.
... Although the underlying mechanisms remain to be fully elucidated, this is a striking example There are also recent examples of metabolically important proteins regulated at the level of mRNA stability. The rhythmically expressed mammalian deadenylase Nocturnin is sensitive to acute extracellular stimuli, such as induction by insulin in 3T3-L1 preadipocytes (79,80) and fasting in white adipose tissue (62). Nocturnin Ϫ/Ϫ mice are resistant to dietinduced weight gain and hepatosteatosis, although the direct mRNA targets of this deadenylase that confer this phenotype are not yet known (64). ...
Article
Full-text available
Regulated cell metabolism involves acute and chronic regulation of gene expression by various nutritional and endocrine stimuli. To respond effectively to endogenous and exogenous signals, cells require rapid response mechanisms to modulate transcript expression and protein synthesis and cannot, in most cases, rely on control of transcriptional initiation that requires hours to take effect. Thus, co- and posttranslational mechanisms have been increasingly recognized as key modulators of metabolic function. This review highlights the critical role of mRNA translational control in modulation of global protein synthesis as well as specific protein factors that regulate metabolic function. First, the complex lifecycle of eukaryotic mRNAs will be reviewed, including our current understanding of translational control mechanisms, regulation by RNA binding proteins and microRNAs, and the role of RNA granules, including processing bodies and stress granules. Second, the current evidence linking regulation of mRNA translation with normal physiological and metabolic pathways and the associated disease states are reviewed. A growing body of evidence supports a key role of translational control in metabolic regulation and implicates translational mechanisms in the pathogenesis of metabolic disorders such as type 2 diabetes. The review also highlights translational control of apolipoprotein B (apoB) mRNA by insulin as a clear example of endocrine modulation of mRNA translation to bring about changes in specific metabolic pathways. Recent findings made on the role of 5'-untranslated regions (5'-UTR), 3'-UTR, RNA binding proteins, and RNA granules in mediating insulin regulation of apoB mRNA translation, apoB protein synthesis, and hepatic lipoprotein production are discussed.
... 95 Importantly, Pparγ activation in bone marrow stromal cells induced Noc gene expression nearly 30-fold. 18,96 Offspring from mice fed a western-like fat diet develop obesity over four generations, and Noc is one of the most upregulated genes in the stromal vascular fractions of adipose tissue from these mice. 97 Thus, Noc appears to be important in lipid homeostasis. ...
Article
Peroxisome proliferator-activated receptor γ (PPARγ) is a critical factor for adipogenesis and glucose metabolism, but accumulating evidence demonstrates the involvement of PPARγ in skeletal metabolism as well. PPARγ agonists, the thiazolidinediones, have been widely used for the treatment of type 2 diabetes mellitus owing to their effectiveness in lowering blood glucose levels. However, the use of thiazolidinediones has been associated with bone loss and fractures. Thiazolidinedione-induced alterations in the bone marrow milieu-that is, increased bone marrow adiposity with suppression of osteogenesis-could partially explain the pathogenesis of drug-induced bone loss. Furthermore, several lines of evidence place PPARγ at the center of a regulatory loop between circadian networks and metabolic output. PPARγ exhibits a circadian expression pattern that is magnified by consumption of a high-fat diet. One gene with circadian regulation in peripheral tissues, nocturnin, has been shown to enhance PPARγ activity. Importantly, mice deficient in nocturnin are protected from diet-induced obesity, exhibit impaired circadian expression of PPARγ and have increased bone mass. This Review focuses on new findings regarding the role of PPARγ in adipose tissue and skeletal metabolism and summarizes the emerging role of PPARγ as an integral part of a complex circadian regulatory system that modulates food storage, energy consumption and skeletal metabolism.
... cotransfected with WT-Noc-Flag and a Pparg2-expression vector and treated with 5 μM of rosiglitazone. Noc and Pparg expression were visualized by immunofluorescence using anti-Flag antibody and anti-PPAR-γ antibody. Figures shown are the representative of at least three independent experiments. Values are expressed as the mean ± SEM. *, P < 0.05. Kawai et al. PNAS | June 8, 2010 | vol. 107 | no. 23 | 10509 CELL BIOLOGY Pparg2 transcriptional activation by rosiglitazone was impaired in Noc-deficient mouse embryonic fibroblasts (MEFs) compared with WT MEFs, although basal transcriptional activity was comparable between WT and Noc −/− MEFs (Fig. 3A). To examine whether physical binding of NOC to PPAR-γ2 caused any ...
Article
Full-text available
Nocturnin (NOC) is a circadian-regulated protein related to the yeast family of transcription factors involved in the cellular response to nutrient status. In mammals, NOC functions as a deadenylase but lacks a transcriptional activation domain. It is highly expressed in bone-marrow stromal cells (BMSCs), hepatocytes, and adipocytes. In BMSCs exposed to the PPAR-gamma (peroxisome proliferator-activated receptor-gamma) agonist rosiglitazone, Noc expression was enhanced 30-fold. Previously, we reported that Noc(-/-) mice had low body temperature, were protected from diet-induced obesity, and most importantly exhibited absence of Pparg circadian rhythmicity on a high-fat diet. Consistent with its role in influencing BMSCs allocation, Noc(-/-) mice have reduced bone marrow adiposity and high bone mass. In that same vein, NOC overexpression enhances adipogenesis in 3T3-L1 cells but negatively regulates osteogenesis in MC3T3-E1 cells. NOC and a mutated form, which lacks deadenylase activity, bind to PPAR-gamma and markedly enhance PPAR-gamma transcriptional activity. Both WT and mutant NOC facilitate nuclear translocation of PPAR-gamma. Importantly, NOC-mediated nuclear translocation of PPAR-gamma is blocked by a short peptide fragment of NOC that inhibits its physical interaction with PPAR-gamma. The inhibitory effect of this NOC-peptide was partially reversed by rosiglitazone, suggesting that effect of NOC on PPAR-gamma nuclear translocation may be independent of ligand-mediated PPAR-gamma activation. In sum, Noc plays a unique role in the regulation of mesenchymal stem-cell lineage allocation by modulating PPAR-gamma activity through nuclear translocation. These data illustrate a unique mechanism whereby a nutrient-responsive gene influences BMSCs differentiation, adipogenesis, and ultimately body composition.
Article
Full-text available
Circadian rhythms are generated by intrinsic 24-h oscillations that anticipate the extrinsic changes associated with solar day. A conserved transcriptional-translational feedback loop generates these molecular oscillations of clock genes at the organismal and the cellular levels. One of the recently discovered outputs of circadian clock is Nocturnin (Noct) or Ccrn4l. In mice, Noct mRNA is broadly expressed in cells throughout the body, with a particularly high-amplitude rhythm in liver. NOCT belongs to the EEP family of proteins with the closest similarity to the CCR4 family of deadenylases. Multiple studies have investigated the role of Nocturnin in development, adipogenesis, lipid metabolism, inflammation, osteogenesis, and obesity. Further, mice lacking Noct (Noct KO or Noct −/−) are protected from high-fat diet-induced obesity and hepatic steatosis. Recent studies had provided new insights by investigating various aspects of Nocturnin, ranging from its sub-cellular localization to identification of its target transcripts. However, a profound understanding of its molecular function remains elusive. This review article seeks to integrate the available literature into our current understanding of the functions of Nocturnin, their regulatory roles in key tissues and to throw light on the existing scientific lacunae.
Preprint
Full-text available
Impairment of the central nervous system (CNS) functions in astronauts is a major health risk for long-duration space missions. Here, for the first time, we combine single-cell multiomics (transcriptomics and chromatin accessibility) and spatial transcriptomics analyses to discover spaceflight-mediated changes in the mouse brain. By comparing ground control and spaceflight animals, we found that the main processes affected by spaceflight include neurogenesis, synaptogenesis and synaptic transmission in cortex, hippocampus, striatum and neuroendocrine structures as well as astrocyte activation and immune dysfunction. At the pathway level, spaceflight resembles neurodegenerative diseases with oxidative stress and protein misfolding components, such as in Parkinson’s disease. Our integrated spatial multiomics approach reveals both widespread and localized brain impairments and suggests key structures and mechanisms to be targeted for countermeasure development. All datasets can be mined through an interactive data portal as well as the National Aeronautics and Space Administration (NASA) GeneLab repositories.
Thesis
Full-text available
La découverte récente d'adipocytes bruns fonctionnels chez les humains adultes a conduit à envisager leur utilisation afin d’augmenter la dépense énergétique dans de potentiels traitements contre l'obésité et les maladies associées. Par ailleurs, des ilots d’adipocytes bruns, appelés adipocytes "brite" (brown in white), émergent dans le tissu adipeux blanc après une exposition au froid ou une stimulation des récepteurs β3-adrénergiques. En utilisant les cellules hMADS, nous avons identifié plusieurs miARNs régulés pendant le « britening ». miR-125b et let-7i ont des niveaux d’expression plus bas dans les adipocytes « brites ». Des analyses fonctionnelles utilisant un « mimic » de miR-125b ou un inhibiteur ont révélé que miR-125b agit comme un frein sur le « brunissage » des cellules hMADS en altérant leur respiration ainsi que leur contenu mitochondrial. In vivo, nous avons montré que miR-125b et let-7i sont moins exprimés dans le tissu adipeux brun par rapport au tissu adipeux blanc. La stimulation des récepteurs β3-adrénergiques ou l'exposition au froid induit une diminution d’expression des miARNs dans les deux tissus et est associée à l'activation du tissu adipeux brun et au recrutement des adipocytes « brites ». Nous avons constaté que l’injection de miR-125b ou let-7i dans le tissu adipeux blanc sous-cutané inhibait l’expression de gènes du « brunissage » induite par la stimulation de la voie β3-adrénergique. En conclusion, nos observations ont montré que miR-125b et let-7i jouaient un rôle important dans la modulation des adipocytes « brites » et des adipocytes « bruns » en ciblant l’expression de gènes mitochondriaux et en diminuant la biogenèse mitochondriale.
Article
Intrauterine growth restriction leads to the development of adult onset obesity/metabolic syndrome, diabetes mellitus, cardiovascular disease, hypertension, stroke, dyslipidemia, and non-alcoholic fatty liver disease/steatohepatitis. Continued postnatal growth restriction has been shown to ameliorate many of these sequelae. To further our understanding of the mechanism of how intrauterine and early postnatal growth affects adult health we have employed Affymetrix microarray-based expression profiling to characterize hepatic gene expression of male offspring in a rat model of maternal nutrient restriction in early and late life. At day 21 of life (p21) combined intrauterine and postnatal calorie restriction treatment led to expression changes in circadian, metabolic, and insulin-like growth factor genes as part of a larger transcriptional response that encompasses 144 genes. Independent and controlled experiments at p21 confirm the early life circadian, metabolic, and growth factor perturbations. In contrast to the p21 transcriptional response, at day 450 of life (d450) only seven genes, largely uncharacterized, were differentially expressed. This lack of a transcriptional response identifies non-transcriptional mechanisms mediating the adult sequelae of intrauterine growth restriction. Independent experiments at d450 identify a circadian defect as well as validate expression changes to four of the genes identified by the microarray screen which have a novel association with growth restriction. Emerging from this rich dataset is a portrait of how the liver responds to growth restriction through circadian dysregulation, energy/substrate management, and growth factor modulation. J. Cell. Biochem. © 2014 Wiley Periodicals, Inc.
Article
The development of obesity is the consequence of a multitude of complex interactions between both genetic and environmental factors. It has been suggested that the dramatic increase in the prevalence of obesity over the past 30 years has been the result of environmental changes that have enabled the full realization of genetic susceptibility present in the population. Among the many environmental alterations that have occurred in our recent history is the ever-increasing dyssynchrony between natural cycles of light/dark and altered patterns of sleep/wake and eating behavior associated with our "24-hour" lifestyle. An extensive research literature has established clear links between increased risk for obesity and both sleep deprivation and shift work, and our understanding of the consequences of such dyssynchrony at the molecular level is beginning to emerge. Studies linking alterations in cellular circadian clocks to metabolic dysfunction point to the increasing importance of chronobiology in obesity etiology.
Article
The role of circadian proteins in regulating whole-body metabolism and bone turnover has been studied in detail and has led to the discovery of an elemental system for timekeeping involving the core genes Clock, Bmal1, Per, and Cry. Nocturnin (Noc; Ccrn4l), a peripheral circadian-regulated gene has been shown to play a very important role in regulating adipogenesis by deadenylation of key mRNAs and intracytoplasmic transport of PPARγ. The role that it plays in osteogenesis has previously not been studied in detail. In this report we examined in vitro and in vivo osteogenesis in the presence and absence of Noc and show that loss of Noc enhances bone formation and can rescue rosiglitazone-induced bone loss in mice. The circadian rhythm of Noc is likely to be an essential element of marrow stromal cell fate.
Article
During endochondral ossification, the cartilage is surrounded by a layer of cells that constitute the perichondrium. Communication between osteoblasts in the perichondrium via N-cadherin adherens junctions is essential for endochondral bone growth. We observed that adherens junction molecule N-cadherin and its interacting partners p120, β-catenin and PTEN are expressed by cells present in the perichondrium. To study if N-cadherin mediated adherens junctions play a role in mediating signal transduction events during bone development, we utilized MC3T3E1 preosteoblasts plated at sub confluent (low) and confluent (high) densities to mimic adherens junction formation. When MC3T3E1 cells were plated at high density we observed an increase in phosphorylation of AKTSer473 and its downstream target GSK3Ser9, which coincided with an increase in Osterix, Osteomodulin and Osteoglycin gene expression. Using immunofluorescence, we identified N-cadherin, p120 and β-catenin localized at the membrane of MC3T3E1 cells. Treatment of confluent MC3T3E1 cells with an N-cadherin junction inhibitor-EGTA and a PI3K inhibitor LY294002 resulted in reduction of phosphorylation levels of AKT and GSK3 and expression of Osterix, Osteomodulin and Osteoglycin. Furthermore, utilizing an N-cadherin blocking antibody resulted in reduced AKT signaling and Osterix gene expression, suggesting that osteoblast junction formation is linked to activation of PI3K signaling, which leads to osteoblast differentiation. To further explore the strength of this linkage, we utilized a conditional knockout approach using Dermo1cre to delete β-catenin and PTEN, two important proteins known to be essential for adherens junctions and PI3K signaling, respectively. In the absence of β-catenin, we observed a decrease in adherens junctions and AKT signaling in the perichondrium. PTEN deletion, on the other hand, increased the number of cells expressing N-cadherin in the perichondrium. These observations show that N-cadherin mediated junctions between osteoblasts are needed for osteoblast gene transcription.
Article
IGF-I is an anabolic factor that mediates GH and PTH actions in bone. Expression of skeletal Igf1 differs for inbred strains of mice, and Igf expression levels correlate directly with bone mass. Previously we reported that peroxisome proliferator-activated receptor-γ2 activation in bone marrow suppressed Igf1 expression and that peroxisome proliferator-activated receptor-γ2 activation-induced Nocturnin (Noc) expression, a circadian gene with peak expression at light offset, which functions as a deadenylase. In 24-h studies we found that Igf1 mRNA exhibited a circadian rhythm in femur with the lowest Igf1 transcript levels at night when Noc transcripts were highest. Immunoprecipitation/RT-PCR analysis revealed a physical interaction between Noc protein and Igf1 transcripts. To clarify which portions of the Igf1 3' untranslated region (UTR) were necessary for regulation by Noc, we generated luciferase constructs containing various lengths of the Igf1 3'UTR. Noc did not affect the 170-bp short-form 3'UTR, but suppressed luciferase activity in constructs bearing the longer-form 3'UTR, which contains a number of potential regulatory motifs involved in mRNA degradation. C57BL/6J mice have low skeletal Igf1 mRNA compared with C3H/HeJ mice, and the Igf1 3' UTR is polymorphic between these strains. Interestingly, the activity of luciferase constructs bearing the long-form 3'UTR from C57BL/6J mice were repressed by Noc overexpression, whereas those bearing the corresponding region from C3H/HeJ were not. In summary, Noc interacts with Igf1 in a strain- and tissue-specific manner and reduces Igf1 expression by targeting the longer form of the Igf1 3'UTR. Posttranscriptional regulation of Igf1 may be critically important during skeletal acquisition and maintenance.
Article
Full-text available
Stress induces secretion of corticosterone through activation of the hypothalamic-pituitary-adrenal axis. This corticosterone secretion is thought to be controlled by a circadian clock in the suprachiasmatic nucleus (SCN). The hypothalamic paraventricular nucleus (PVN) receives convergent information from both stress and the circadian clock. Recent reports demonstrate that mammalian orthologs (Per1, Per2, and Per3) of the Drosophila clock gene Period are expressed in the SCN, PVN, and peripheral tissues. In this experiment, we examined the effect of physical and inflammatory stressors on mPer gene expression in the SCN, PVN, and liver. Forced swimming, immobilization, and lipopolysaccharide injection elevated mPer1 gene expression in the PVN but not in the SCN or liver. A stress-induced increase in mPer1 expression was observed in the corticotropin-releasing factor-positive cells of the PVN; however, the stressors used in this study did not affect mPer2 expression in the PVN, SCN, or liver. The present study suggests that a stress-induced disturbance of circadian corticosterone secretion may be associated with the stress-induced expression of mPer1 mRNA in the PVN.
Article
Full-text available
Circadian rhythms result from feedback loops involving clock genes and their protein products. In mammals, 2 orphan nuclear receptors, REV-ERBalpha and RORalpha, play important roles in the transcription of the clock gene Bmal1. The authors now considerably extend these findings with the demonstration that all members of the REV-ERB (alpha and beta) and ROR (alpha, beta, and gamma) families repress and activate Bmal1 transcription, respectively. The authors further show that transcription of Bmal1 is the result of competition between REV-ERBs and RORs at their specific response elements (RORE). Moreover, they demonstrate that Reverb genes are similarly expressed in the thymus, skeletal muscle, and kidney, whereas Ror genes present distinct expression patterns. Thus, the results indicate that all members of the REV-ERB and ROR families are crucial components of the molecular circadian clock. Furthermore, their strikingly different patterns of expression in nervous and peripheral tissues provide important insights into functional differences between circadian clocks within the organism.
Article
Full-text available
An analysis of transcriptional variation in the liver using a panel of B.A chromosome substitution strains identified 4209 transcripts that are differentially expressed relative to the C57BL/6J background and 1010 transcripts that are differentially expressed between C57BL/6J and A/J strains. A subset of these strains (substituting Chromosomes 1, 6, and 15) was used to identify 386 additional differentially expressed transcripts in the kidney. Approximately 15% of differentially expressed transcripts are located on the substituted chromosome. These cis-QTL are codirectionally expressed with the donor strain A/J. By comparison, trans-regulated loci comprise 85% of differentially expressed transcripts, often show opposite direction of change compared with A/J, and can be regulated by multiple chromosome substitutions. Gene expression differences in this study provide evidence for transgressive segregation: Only 438 of 4209 QTL in liver were inside the parental range. By combining QTL data with known biological functions, we were able to identify physiologic pathways altered in multiple strains. In many cases the same pathways were altered by multiple distinct chromosome substitutions. Taken together, these results suggest that widespread epistatic background effects may result in complex and overlapping transcriptional relationships among different chromosome substitution strains. Transcriptional profiling of chromosome substitution strains reveals a complex genetic architecture of transcriptional regulation.
Article
Full-text available
As sensors for fat-soluble hormones and dietary lipids, oscillations in nuclear receptor (NR) expression in key metabolic tissues may contribute to circadian entrainment of nutrient and energy metabolism. Surveying the diurnal expression profiles of all 49 mouse nuclear receptors in white and brown adipose tissue, liver, and skeletal muscle revealed that of the 45 NRs expressed, 25 are in a rhythmic cycle and 3 exhibit a single transient pulse of expression 4 hr into the light cycle. While thyroid hormones are generally constant, we find that TRalpha and beta dramatically cycle, suggesting that fundamental concepts such as "basal metabolism" may require reexamination. The dynamic but coordinated changes in nuclear receptor expression, along with their key target genes, offers a logical explanation for known cyclic behavior of lipid and glucose metabolism and suggests novel roles for endocrine and orphan receptors in coupling the peripheral circadian clock to divergent metabolic outputs.
Article
Full-text available
An increase in adipocyte number is a major contributor to the increase in adipose tissue mass that is characteristic of obesity. The identity and regulation of the adipocyte precursor cell (or preadipocyte) and the preadipocyte precursor cell (or progenitor cell) have been intensely studied for many years. In this issue of the JCI, Crossno et al. report that progenitor cells originating from outside the adipose tissue, in particular the bone marrow, can contribute to an increase in adipocyte number (see the related article beginning on page 3220). Their study in mice reveals that treatment with the thiazolidinedione rosiglitazone or exposure to a high-fat diet promotes the trafficking of circulating bone marrow-derived progenitor cells into adipose tissue, where they become multilocular adipocytes. This adds a new and unexpected dimension to this research arena.
Article
Full-text available
The mammalian circadian system consists of a central oscillator in the suprachiasmatic nucleus of the hypothalamus, which coordinates peripheral clocks in organs throughout the body. Although circadian clocks control the rhythmic expression of a large number of genes involved in metabolism and other aspects of circadian physiology, the consequences of genetic disruption of circadian-controlled pathways remain poorly defined. Here we report that the targeted disruption of Nocturnin (Ccrn4l) in mice, a gene that encodes a circadian deadenylase, confers resistance to diet-induced obesity. Mice lacking Nocturnin remain lean on high-fat diets, with lower body weight and reduced visceral fat. However, unlike lean lipodystrophic mouse models, these mice do not have fatty livers and do not exhibit increased activity or reduced food intake. Gene expression data suggest that Nocturnin knockout mice have deficits in lipid metabolism or uptake, in addition to changes in glucose and insulin sensitivity. Our data support a pivotal role for Nocturnin downstream of the circadian clockwork in the posttranscriptional regulation of genes necessary for nutrient uptake, metabolism, and storage. • mRNA • clock • diabetes • posttranscriptional • lipid
Article
Full-text available
A core group of regulatory factors control circadian rhythms in mammalian cells. While the suprachiasmatic nucleus in the brain serves as the central core circadian oscillator, circadian clocks also exist within peripheral tissues and cells. A growing body of evidence has demonstrated that >20% of expressed mRNAs in bone and adipose tissues oscillate in a circadian manner. The current manuscript reports evidence of the core circadian transcriptional apparatus within primary cultures of murine and human bone marrow-derived mesenchymal stem cells (BMSCs). Exposure of confluent, quiescent BMSCs to dexamethasone synchronized the oscillating expression of the mRNAs encoding the albumin D binding protein (dbp), brain-muscle arnt-like 1 (bmal1), period 3 (per3), rev-erb alpha (Rev A), and rev-erb beta (Rev B). The genes displayed a mean oscillatory period of 22.2 to 24.3 h. The acrophase or peak expression of mRNAs encoding "positive" (bmal1) and "negative" (per3) components of the circadian regulatory apparatus were out of phase with each other by approximately 8-12 h, consistent with in vivo observations. In vivo, phosphyrylation by glycogen synthase kinase 3beta (GSK3beta) is known to regulate the turnover of per3 and components of the core circadian regulatory apparatus. In vitro addition of lithium chloride, a GSK3beta inhibitor, significantly shifted the acrophase of all genes by 4.2-4.7 h oscillation in BMSCs; however, only the male murine BMSCs displayed a significant increase in the length of the period of oscillation. We conclude that human and murine BMSCs represent a valid in vitro model for the analysis of circadian mechanisms in bone metabolism and stem cell biology.
Article
Full-text available
There is compelling evidence to suggest that both the development of bone to peak bone mass at maturity and subsequent loss depend on the interaction between genetic, hormonal, environmental and nutritional factors. The major part (< or = 80%) of the age-specific variation in bone turnover and bone density is genetically determined. However, the notion of genetic determinant is of little value unless the specific genes that are involved can be identified. Most work in this area of osteoporosis research has focused on the candidate gene approach, which has identified several candidate genes for osteoporosis, including genes encoding the vitamin D receptor (VDR), oestrogen receptors (alpha and beta), apolipoprotein E, collagen type I alpha 1 and methylenetetrahydrofolate reductase, amongst many others. However, in general, findings from numerous studies of the association between such genes and various bone variables have been inconsistent. In addition to possible gene-gene interactions it is likely that there are interactions between these genes and certain environmental factors, especially nutrition, that may mediate expression of bone-related phenotypes. While these potential interactions add a level of complexity to our understanding of these apparent genetic effects on bone, identification of a role for genetic factors without knowledge of their interaction with nutrients can do little to advance prevention and treatment of osteoporosis. This information is especially important because, unlike genotype, diet and nutrition can be modified. The aim of the present review is to critically evaluate current knowledge relating to candidate genes for osteoporosis, with particular emphasis on their interaction with nutrients and dietary factors in determining bone health.
Article
Stress induces secretion of corticosterone through activation of the hypothalamic-pituitary-adrenal axis. This corticosterone secretion is thought to be controlled by a circadian clock in the suprachiasmatic nucleus (SCN). The hypothalamic paraventricular nucleus (PVN) receives convergent information from both stress and the circadian clock. Recent reports demonstrate that mammalian orthologs (Per1, Per2, and Per3) of the Drosophila clock gene Period are expressed in the SCN, PVN, and peripheral tissues. In this experiment, we examined the effect of physical and inflammatory stressors on mPer gene expression in the SCN, PVN, and liver. Forced swimming, immobilization, and lipopolysaccharide injection elevated mPer1 gene expression in the PVN but not in the SCN or liver. A stress-induced increase in mPer1 expression was observed in the corticotropin-releasing factor–positive cells of the PVN; however, the stressors used in this study did not affect mPer2 expression in the PVN, SCN, or liver. The present study suggests that a stress-induced disturbance of circadian corticosterone secretion may be associated with the stress-induced expression of mPer1 mRNA in the PVN.
Article
Emerging evidence points to a critical role for the skeleton in several homeostatic processes, including energy balance. The connection between fuel utilization and skeletal remodeling begins in the bone marrow with lineage allocation of mesenchymal stem cells to adipocytes or osteoblasts. Mature bone cells secrete factors that influence insulin sensitivity, and fat cells synthesize cytokines that regulate osteoblast differentiation; thus, these two pathways are closely linked. The emerging importance of the bone-fat interaction suggests that novel molecules could be used as targets to enhance bone formation and possibly prevent fractures. In this article, we discuss three pathways that could be pharmacologically targeted for the ultimate goal of enhancing bone mass and reducing osteoporotic fracture risk: the leptin, peroxisome proliferator-activated receptor gamma and osteocalcin pathways. Not surprisingly, because of the complex interactions across homeostatic networks, other pathways will probably be activated by this targeting, which could prove to be beneficial or detrimental for the organism. Hence, a more complete picture of energy utilization and skeletal remodeling will be required to bring any potential agents into the future clinical armamentarium.
Article
Rosiglitazone (Rosi), a member of the thiazolidinedione class of drugs used to treat type 2 diabetes, activates the adipocyte-specific transcription factor peroxisome proliferator-activated receptor gamma (PPARgamma). This activation causes bone loss in animals and humans, at least in part due to suppression of osteoblast differentiation from marrow mesenchymal stem cells (MSC). In order to identify mechanisms by which PPARgamma2 suppresses osteoblastogenesis and promotes adipogenesis in MSC, we have analyzed the PPARgamma2 transcriptome in response to Rosi. A total of 4,252 transcriptional changes resulted when Rosi (1 microM) was applied to the U-33 marrow stromal cell line stably transfected with PPARgamma2 (U-33/gamma2) as compared to non-induced U-33/gamma2 cells. Differences between U-33/gamma2 and U-33 cells stably transfected with empty vector (U-33/c) comprised 7,928 transcriptional changes, independent of Rosi. Cell type-, time- and treatment-specific gene clustering uncovered distinct patterns of PPARgamma2 transcriptional control of MSC lineage commitment. The earliest changes accompanying Rosi activation of PPARgamma2 included effects on Wnt, TGFbeta/BMP and G-protein signaling activities, as well as sustained induction of adipocyte-specific gene expression and lipid metabolism. While suppression of osteoblast phenotype is initiated by a diminished expression of osteoblast-specific signaling pathways, induction of the adipocyte phenotype is initiated by adipocyte-specific transcriptional regulators. This indicates that distinct mechanisms govern the repression of osteogenesis and the stimulation of adipogenesis. The co-expression patterns found here indicate that PPARgamma2 has a dominant role in controlling osteoblast differentiation and suggests numerous gene-gene interactions that could lead to the identification of a "master" regulatory scheme directing this process.
Article
Adult BMD, an important risk factor for fracture, is the result of genetic and environmental interactions. A quantitative trait locus (QTL) for the phenotype of volumetric BMD (vBMD), named Bmd8, was found on mid-distal chromosome (Chr) 6 in mice. This region is homologous to human Chr 3p25. The B6.C3H-6T (6T) congenic mouse was previously created to study this QTL. Using block haplotyping of the 6T congenic region, expression analysis in the mouse, and examination of nonsynonymous SNPs, peroxisome proliferator activated receptor gamma (Pparg) was determined to be the most likely candidate gene for the Bmd8 QTL of the 630 genes located in the congenic region. Furthermore, in the C3H/HeJ (C3H) strain, which is the donor strain for the 6T congenic, several polymorphisms were found in the Pparg gene. On challenge with a high-fat diet, we found that the 6T mouse has a lower areal BMD (aBMD) and volume fraction of trabecular bone (BV/TV%) of the distal femur compared with B6 mice. Interactions between SNPs in the PPARG gene and dietary fat for the phenotype of BMD were examined in the Framingham Offspring Cohort. This analysis showed that there was a similar interaction of the PPARG gene and diet (fat intake) on aBMD in both men and women. These findings suggest that dietary fat has a significant influence on BMD that is dependent on the alleles present for the PPARG gene.
Article
More than 70% of the variability in human bone density has been attributed to genetic factors as a result of studies with twins, osteoporotic families, and individuals with rare heritable bone disorders. We have applied the Stratec XCT 960M pQCT, specifically modified for small skeletal specimens, to analyses of bones from 11 inbred strains (AKR/J, BALB/cByJ, C3H/HeJ, C57BL/6J, C57L/J, DBA/2J, NZB/B1NJ, SM/J, SJL/BmJ, SWR/BmJ, and 129/J) of female mice to determine the extent of heritable differences in peak bone density, pQCT scans were taken of femurs from (a) 12-month-old inbred strain females and (b) a subset of four strains (C3H/HeJ, DBA/2J, BALB/cByJ, C57BL/6J) at 2, 4, and 8 months. In addition, pQCT scans were also obtained from L5-L6 vertebrae and proximal phalanges from the same subset of four inbred strains at 12 months of age. Comparison of bone parameters among inbred strains revealed significant differences at each of the three sites investigated. Femoral and phalangeal bones differed among strains with respect to total and cortical density, mineral, and volume. Only cortical bone parameters were significantly different among strains at the vertebral site. With respect to strain differences, the highest value for any given bone parameter was found in the C3H/HeJ strain, whereas C57BL/6J values were absolutely, or statistically, the lowest. Similarly, with respect to bone sites, cortical bone density was significantly correlated among strains. On the other hand, we found that none of the femur, vertebral, or phalangeal parameters correlated with body weight, even though body weight varied by 86% among those inbred strains. The developmental studies of femurs conducted at 2, 4, and 8 months of age with C3H/HeJ, DBA/2J, BALB/cByJ, and C57BL/6J females showed differences in total density among strains at 2 months and thereafter. Adult peak bone density was typically achieved by 4 months, whereas femurs continued to lengthen for 4 to 8 months thereafter. We conclude that (1) major genetic effects on femoral, vertebral, and phalangeal bone density are detectable among inbred strains of mice; (2) cortical bone density shares common genetic regulation at the three measured sites; and (3) within the femur, genes that regulate length and density are different.
Article
Significant differences in vertebral (9%) and femoral (50%) adult bone mineral density (BMD) between the C57BL/6J (B6) and C3H/HeJ (C3H) inbred strains of mice have been subjected to genetic analyses for quantitative trait loci (QTL). Nine hundred eighty-six B6C3F2 females were analyzed to gain insight into the number of genes that regulate peak BMD and their locations. Femurs and lumbar vertebrae were isolated from 4-month-old B6C3F2 females at skeletal maturity and then BMD was determined by peripheral quantitative computed tomography (pQCT). Estimates of BMD heritability were 83% for femurs and 72% for vertebrae. Genomic DNA from F2 progeny was screened for 107 polymerase chain reaction (PCR)-based markers discriminating B6 and C3H alleles on all 19 autosomes. The regression analyses of markers on BMD revealed ten chromosomes (1, 2, 4, 6, 11, 12, 13, 14, 16, and 18) carrying QTLs for femurs and seven chromosomes (1, 4, 7, 9, 11, 14, and 18) carrying QTLs for vertebrae, each with log10 of the odds ratio (LOD) scores of 2.8 or better. The QTLs on chromosomes (Chrs) 2, 6, 12, 13, and 16 were unique to femurs, whereas the QTLs on Chrs 7 and 9 were unique to vertebrae. When the two bone sites had a QTL on the same chromosome, the same marker had the highest, although different, LOD score. A pairwise comparison by analysis of variance (ANOVA) did not reveal significant gene x gene interactions between QTLs for either bone site. BMD variance accounted for by individual QTLs ranged from 1% to 10%. Collectively, the BMD QTLs for femurs accounted for 35.1% and for vertebrae accounted for 23.7 % of the F2 population variances in these bones. When mice were homozygous c3/c3 in the QTL region, 8 of the 10 QTLs increased, while the remaining two QTLs on Chrs 6 and 12 decreased, femoral BMD. Similarly, when mice were homozygous c3/c3 in the QTL region for the vertebrae, five of the seven QTLs increased, while two QTLs on Chrs 7 and 9 decreased, BMD. These findings show the genetic complexity of BMD with multiple genes participating in its regulation. Although 5 of the 12 QTLs are considered to be skeleton-wide loci and commonly affect both femurs and vertebrae, each of the bone sites also exhibited unique QTLs. Thus, the BMD phenotype can be partitioned into its genetic components and the effects of these loci on normal bone biology can be determined. Importantly, the BMD QTLs that we have identified are in regions of the mouse genome that have known human homology, and the QTLs will become useful experimental tools for mechanistic and therapeutic analyses of bone regulatory genes.
Article
Insulin-like growth factor (IGF) I is a critical peptide for skeletal growth and consolidation. However, its regulation is complex and, in part, heritable. We previously indicated that changes in both serum and skeletal IGF-I were related to strain-specific differences in total femoral bone mineral density (BMD) in mice. In addition, we defined four quantitative trait loci (QTLs) that contribute to the heritable determinants of the serum IGF-I phenotype in F2 mice derived from progenitor crosses between C3H/HeJ (C3H; high total femoral BMD and high IGF-I) and C57BL/6J (B6; low total femoral BMD and low IGF-I) strains. The strongest QTL, IGF-I serum level 1 (Igflsl-1; log10 of the odds ratio [LOD] score, approximately 9.0), is located on the middle portion of chromosome (Chr) 6. For this locus, C3H alleles are associated with a significant reduction in serum IGF-I. To test the effect of this QTL in vivo, we generated a new congenic strain (B6.C3H-6T [6T]) by placing the Chr 6 QTL region (D6Mit93 to D6Mit150) from C3H onto the B6 background. We then compared serum and skeletal IGF-I levels, body weight, and several skeletal phenotypes from the N9 generation of 6T congenic mice against B6 control mice. Female 6T congenic mice had 11-21% lower serum IGF-I levels at 6, 8, and 16 weeks of age compared with B6 (p < 0.05 for all). In males, serum IGF-I levels were similar in 6T congenics and B6 controls at 6 weeks and 8 weeks but were lower in 6T congenic mice at 16 weeks (p < 0.02). In vitro, there was a 40% reduction in secreted IGF-I in the conditioned media (CMs) from 6T calvaria osteoblasts compared with B6 cells (p < 0.01). Total femoral BMD as measured by peripheral quantitative computed tomography (pQCT) was lower in both 6T male (-4.8%, p < 0.01) and 6T female (-2.3%, p = 0.06) congenic mice. Geometric features of middiaphyseal cortical bone were reduced in 6T congenic mice compared with control mice. Femoral cancellous bone volume (BV) density and trabecular number (Tb.N) were 50% lower, whereas trabecular separation (Tb.Sp) was 90% higher in 8-week-old female 6T congenic mice compared with B6 control mice (p < 0.01 for all). Similarly, vertebral cancellous BV density and Tb.N were lower (-29% and -19%, respectively), whereas Tb.Sp was higher (+29%) in 16-week-old female 6T congenic mice compared with B6 control mice (p < 0.001 for all). Histomorphometric evaluation of the proximal tibia indicated that 6T congenics had reduced BV fraction, labeled surface, and bone formation rates compared with B6 congenic mice. In summary, we have developed a new congenic mouse strain that confirms the Chr 6 QTL as a major genetic regulatory determinant for serum IGF-I. This locus also influences bone density and morphology, with more dramatic effects in cancellous bone than in cortical bone.
Article
The mammalian circadian timing system is composed of almost as many individual clocks as there are cells. These countless oscillators have to be synchronized by a central pacemaker to coordinate temporal physiology and behavior. Recently, there has been some progress in understanding the relationship and communication mechanisms between central and peripheral clocks.
Article
Targeted gene studies have demonstrated the importance of insulin-like growth factor-I (IGF-I) for osteoblast (OB) differentiation and the acquisition of peak bone mineral density (BMD). The skeletal response to allelic differences in IGF-I expression can also be measured in vivo, using congenic mice. We created a congenic strain with reduced (approximately 20%) circulating IGF-I (C3H.B6-6T [6T]) by backcrossing a small genomic region (30 cM) of Chromosome 6 (Chr6) from C3H/HeJ (C3H) onto a C57Bl/6J (B6) background. 6T female mice have lower serum IGF-I (P<0.001 vs. B6) but similar growth hormone (GH) and serum IGF binding protein (IGFBP) concentrations as B6. At 16 weeks of age, congenics have greater body fat (P<0.02 vs. B6) despite less total body weight, and exhibit smaller femoral cross-sectional size (P=0.001), reduced cortical thickness (P<0.001) and lower trabecular BV/TV (P<0.05) than B6. 6T mice also have suppressed serum leptin (P<0.01), but compared to B6 have similar markers of bone resorption (i.e., urine CTx and serum TRAP 5B). At 8 weeks of age, skeletal IGF-I mRNA from long bones was reduced by 40% (P<0.05) as were liver mRNA transcripts (i.e., 50%, P<0.01). Osteoblast progenitors from the bone marrow of 6T mice formed less colony forming unit fibroblasts by crystal violet staining than B6 (P<0.007) and had significantly reduced alkaline phosphatase-positive colonies than B6(P<0.0001). In addition, staining of bone marrow with oil red O revealed greater numbers of adipocytes in 6T than B6. Several candidate genes in the Chr6 QTL were excluded by lack of strain-related expression differences in bone, but genes positively regulating adipocyte differentiation including Alox 5 and PPAR-gamma require further study as either "pathway" or candidate genes. In summary, allelic differences in a QTL on Chr6 result in altered IGF-I gene expression, changes in OB lineage allocation, and reduced peak bone mass. Congenic mice are useful models not only for mapping genes related to bone mass but also for elucidating the biology underlying various skeletal phenotypes associated with more subtle manipulation of the mouse genome.
Article
Insulin-like growth factor-I (IGF-I) is critical for optimal skeletal growth and maintenance. Knockout and transgenic models have provided significant insights into the role of IGF-I in bone modeling and remodeling. Congenic mice demonstrate allelic differences in particular quantitative trait loci (QTL). One such model is congenic 6T, which contains a QTL for reduced serum IGF-I donated from C3H/HeJ on a pure C57Bl/6 J (B6) background. In this study we found a 30%-50% reduction in IGF-I expression in bone, liver, and fat of the congenic 6T mouse, as well as lower circulating IGF-I compared with control B6. 6T mice also had a greater percentage body fat, but reduced serum leptin. These changes were associated with reduced cortical and trabecular bone mineral density, impaired bone formation but no change in bone resorption. Moreover, the anabolic skeletal response to intermittent parathyroid hormone (PTH) therapy was blunted in 6T compared with B6, potentially in response to greater programmed cell death in osteocytes and osteoblasts of 6T. In summary, allelic differences in IGF-I expression impact peak bone acquisition and body composition, as well as the skeletal response to PTH. Lifelong changes in circulating and skeletal IGF-I may be relevant for the pathophysiology of several diseases, including chronic renal failure.
Article
Within the retina there is a circadian clock that controls the 24-h timing of processes such as hormone release, cell movement, and gene transcription. In an effort to better understand the molecular nature of this retinal clock, a differential display (DD) screen was performed to isolate a gene with high amplitude circadian rhythmicity in the Xenopus retina. A novel gene expressed in the early evening in photoreceptor cells was isolated and named nocturnin for night factor. This article outlines the steps we took to study a protein of unknown function, particularly highlighting the analyses one can perform when little more than the primary sequence of a gene is known. In addition, we describe the results of sequence analysis that assisted in predicting the function of nocturnin. We have shown that nocturnin acts as a deadenylase in vitro, removing the poly(A) tail from a mature messenger RNA in a process that either leads to degradation or translational silencing of a message. Although the role of nocturnin in the retina is unknown, future studies to identify target mRNAs that are deadenylated by nocturnin will assist in elucidating its physiological role in this tissue.
Article
Nuclear hormone receptors (NRs) function as ligand dependent DNA binding proteins that translate physiological/nutritional signals into gene regulation. Dysfunctional NR signaling leads to many disorders in reproduction, inflammation, and metabolism. The opportunity to identify novel regulatory pathways in the context of human health and disease drives the challenge to unravel the biological function of the "orphan nuclear hormone receptors". For example, the Rev-erb (NR1D) subgroup (Rev-erbalpha/NR1D1 and Rev-erbbeta/NR1D2) of orphan NRs are transcriptional silencers and negative regulators of RORalpha mediated trans-activation. The NR1D subgroup is highly enriched in peripheral tissues with onerous energy demands including skeletal muscle, brown and white adipose, brain, liver and kidney. This alludes to the involvement of this subgroup in metabolism. In this context, Rev-erbalpha-/- mice have a dyslipidemic phenotype. Recent studies in vascular smooth and skeletal muscle cells also suggest that the NR1D subgroup modulates inflammation by regulating IkappaBalpha/NFkappaB dependent gene expression. Rev-erbalpha has been identified as a critical regulator (and target) of circadian rhythm, a factor in blood pressure control and inflammation. Finally, two recent reports have demonstrated: (i) lithium mediated regulation of Rev-erbalpha stability and (ii) E75 (the Drosophila orthologue of human Rev-erbalpha) is tightly bound by heme, and functions as a "gas sensor" through interaction with CO/NO and interferes with the repression of DHR3 (the Drosophila orthologue of human RORalpha). In conclusion, the role of these receptors at the cross-roads of metabolism, inflammation, and circadian cycling underscores the importance of understanding the organ-specific function of the NR1D subgroup in homeostasis.
Article
Osteoporosis and obesity, two disorders of body composition, are growing in prevalence. Interestingly, these diseases share several features including a genetic predisposition and a common progenitor cell. With aging, the composition of bone marrow shifts to favor the presence of adipocytes, osteoclast activity increases, and osteoblast function declines, resulting in osteoporosis. Secondary causes of osteoporosis, including diabetes mellitus, glucocorticoids and immobility, are associated with bone-marrow adiposity. In this review, we ask a provocative question: does fat infiltration in the bone marrow cause low bone mass or is it a result of bone loss? Unraveling the interface between bone and fat at a molecular and cellular level is likely to lead to a better understanding of several diseases, and to the development of drugs for both osteoporosis and obesity.
Article
The genes encoding the core circadian transcription factors display an oscillating expression profile in murine calvarial bone. More than 26% of the calvarial bone transcriptome exhibits a circadian rhythm, comparable with that observed in brown and white adipose tissues and liver. Thus, circadian mechanisms may directly modulate oxidative phosphorylation and multiple metabolic pathways in bone homeostasis. Although circadian rhythms have been associated historically with central regulatory mechanisms, there is emerging evidence that the circadian transcriptional apparatus exists in peripheral tissues. The aim of this study was to determine the presence and extent of circadian oscillation in the transcriptome of murine calvarial bone. Cohorts of 8-week-old male AKR/J mice were maintained in a controlled 12-h light:12-h dark cycle on an ad libitum diet for 2 weeks. Groups of three mice were killed every 4 h over a 48-h period. The level of gene expression at successive times-points was determined by quantitative RT-PCR and Affymetrix microarray. Data were analyzed using multiple statistical time series algorithms, including Cosinor, Fisher g-test, and the permutation time test. Both the positive (Bmal1, Npas2) and negative (Cry1, Cry2, Per1, Per2, Per3) elements of the circadian transcriptional apparatus and their immediate downstream targets and mediators (Dbp, Rev-erbalpha, Rev-erbbeta) exhibited oscillatory expression profiles. Consistent with findings in other tissues, the positive and negative elements were in antiphase relative to each other. More than 26% of the genes present on the microarray displayed an oscillatory profile in calvarial bone, comparable with the levels observed in brown and white adipose tissues and liver; however, only a subset of 174 oscillating genes were shared among all four tissues. Our findings show that the components of the circadian transcriptional apparatus are represented in calvarial bone and display coordinated oscillatory behavior. However, these are not the only genes to display an oscillatory expression profile, which is seen in multiple pathways involving oxidative phosphorylation and lipid, protein, and carbohydrate metabolism.
Article
Time-dependent variations in clock gene expression have recently been observed in mouse hematopoietic cells, but the activity of these genes in human bone marrow (BM) has so far not been investigated. Since such data can be of considerable clinical interest for monitoring the dynamics in stem/progenitor cells, the authors have studied mRNA expression of the clock genes hPer1 , hPer2, hCry1, hCry2, hBmal1, hRev-erb alpha, and hClock in human hematopoietic CD34-positive (CD34( +)) cells. CD34(+) cells were isolated from the BM samples obtained from 10 healthy men at 6 times over 24 h. In addition, clock gene mRNA expression was analyzed in the whole BM in 3 subjects. Rhythms in serum cortisol, growth hormone, testosterone, and leukocyte counts documented that subjects exhibited standardized circadian patterns. All 7 clock genes were expressed both in CD34(+) cells and the whole BM, with some differences in magnitude between the 2 cell populations. A clear circadian rhythm was shown for hPer1, hPer2, and hCry2 expression in CD34(+) cells and for hPer1 in the whole BM, with maxima from early morning to midday. Similar to mouse hematopoietic cells, h Bmal1 was not oscillating rhythmically. The study demonstrates that clock gene expression in human BM stem/progenitor cells may be developmentally regulated, with strong or weaker circadian profiles as compared to those reported in other mature tissues.
Article
The adult skeleton is constantly renewed through bone remodeling. Four recent papers (Baldock et al., 2007; Lee et al., 2007; Lundberg et al., 2007; Sato et al., 2007) provide new insights into central and peripheral control of this remodeling sequence. Two of the studies add to our knowledge of the complex hypothalamic modulation of bone turnover mediated by NMU and NPY via the sympathetic nervous system, while the other two focus on the peripheral neural target, the osteoblast, and its regulation by neuropeptides and osteocalcin. These findings support a new paradigm concerning the regulation of bone remodeling and provide a foundation for novel approaches to preventing osteoporosis.
Article
Nocturnin has been identified as a clock-controlled gene based on its rhythmic expression and night-time peak of transcript level in Xenopus retina. Further studies show that the widespread expression and rhythmicity of nocturnin mRNA level parallel the expression of clock genes. In Xenopus, nocturnin transcription is regulated by cAMP response element-binding protein (CREB) binding the nocturnin element (NE). However, mechanism(s) underlying the regulation of nocturnin transcription in human cells is unknown at present. In this study, we demonstrated that the transcription of human nocturnin gene displayed circadian oscillations in Huh7 cells (a human hepatoma cell line) and was regulated by CLOCK/BMAL1 heterodimer via the E-box of nocturnin promoter. In addition, E-box2 is more efficient than E-box1 in the regulation of CLOCK/BMAL1 on nocturnin transcription in vitro.
Article
The circadian clock is a conserved internal timekeeping mechanism that controls many aspects of physiology and behavior via the rhythmic expression of many genes. One of these rhythmic genes, Nocturnin, encodes a deadenylase--a ribonuclease that specifically removes the poly(A) tails from mRNAs. This enzyme is expressed at high levels during the night in a number of tissues in mammals and has recently been implicated in circadian control of metabolism. Targeted ablation of this gene in mice results in resistance to hepatic steatosis and diet-induced obesity. Nocturnin appears to exert rhythmic posttranscriptional control of genes necessary for metabolic functions including nutrient absorption, glucose/insulin sensitivity, and lipid storage. In the Western world and many developing countries, overnutrition--the 'obesity epidemic' suggests that the ability to sequester fat stores in times of plenty is no longer advantageous to our survival. Understanding the role that the circadian clock plays in controlling these metabolic processes is important in treatment and eventual eradication of this public health crisis.
Fat targets for skeletal health [review]
  • Kawai