Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 31
Filter
1.
Antioxidants (Basel) ; 11(8)2022 Aug 10.
Article in English | MEDLINE | ID: mdl-36009264

ABSTRACT

Senescent cells accumulate in the organs of aged animals and exacerbate organ dysfunction, resulting in age-related diseases. Oxidative stress accelerates cellular senescence. Placental extract, used in the alleviation of menopausal symptoms and promotion of wound healing and liver regeneration, reportedly protects against oxidative stress. In this study, we investigated the effects of human placental extract (HPE) on cellular senescence in normal human dermal fibroblasts (NHDFs) under oxidative stress conditions. We demonstrated that HPE delays the onset of cellular senescence. Next-generation sequencing analysis revealed that under oxidative stress conditions, HPE treatment enhanced the expression of the antioxidant genes CYGB, APOE, NQO1, and PTGS1. Further, HPE treatment under oxidative stress conditions increased the protein level of nuclear factor-erythroid factor 2-related factor 2 (NRF2)-a vital molecule in the antioxidant pathway-via post-transcriptional and/or post-translational regulations. These findings indicate that HPE treatment in NHDFs, under chronic oxidative stress, delays cellular senescence by mitigating oxidative stress via upregulation of the NRF2-mediated antioxidant pathway, and HPE treatment could potentially ameliorate skin-aging-associated damage, in vivo.

2.
Sci Rep ; 12(1): 11031, 2022 06 30.
Article in English | MEDLINE | ID: mdl-35773304

ABSTRACT

As skin aging is one of the most common dermatological concerns in recent years, scientific research has promoted treatment strategies aimed at preventing or reversing skin aging. Breakdown of the extracellular matrix (ECM), such as collagen and elastin fibers, in the skin results in decreased skin elasticity and tension. Cutaneous cells, especially fibroblasts in the dermis layer of the skin, mainly produce ECM proteins. Although clinical studies have demonstrated that placental extract (PE) has positive effects on skin health, the molecular mechanisms by which PE acts against skin aging are still largely unknown. In this study, we performed RNA-sequence analysis to investigate whether human PE (HPE) alters ECM-related gene expression in normal human dermal fibroblast (NHDF) cells. Gene ontology analysis showed that genes related to extracellular matrix/structure organization, such as COL1A1, COL5A3, ELN, and HAS2 were highly enriched, and most of these genes were upregulated. We further confirmed that the HPE increased the type I collagen, proteoglycan versican, elastin, and hyaluronan levels in NHDF cells. Our results demonstrate that HPE activates global ECM-related gene expression in NHDF cells, which accounts for the clinical evidence that the HPE affects skin aging.


Subject(s)
Placental Extracts , Skin Aging , Skin , Cells, Cultured , Elastin/genetics , Elastin/metabolism , Extracellular Matrix/metabolism , Female , Fibroblasts/drug effects , Fibroblasts/metabolism , Gene Expression/drug effects , Humans , Placenta/chemistry , Placenta/metabolism , Placental Extracts/pharmacology , Pregnancy , Skin/drug effects , Skin/metabolism , Skin Aging/drug effects , Versicans/metabolism
3.
J Biochem ; 171(5): 473-476, 2022 May 11.
Article in English | MEDLINE | ID: mdl-35383844

ABSTRACT

For decades, considerable efforts have been expended for solving the molecular mechanisms of disease progression. An important clue to tackle this question is the circadian clock. Recent findings have uncovered previously unknown molecular connections between circadian clock and disease incidence, consequently causing the ageing process. Furthermore, 'chronotherapy' is emerging as a new concept of optimizing the time of the day for drug administration according to target gene expressions in order to maximize therapeutic efficacy and minimize the side effects. This concept will help cure patients and prevent them from suffering evitable pain and side effects. This JB special issue 'Molecular connections between circadian clock and health/aging' discusses how the circadian clocks link to health and ageing from molecular to organismal levels.


Subject(s)
Circadian Clocks , Aging/genetics , Chronotherapy , Circadian Clocks/genetics , Humans
4.
J Biochem ; 171(5): 493-500, 2022 May 11.
Article in English | MEDLINE | ID: mdl-34668549

ABSTRACT

Ageing is one of the greatest risk factors for chronic non-communicable diseases, and cellular senescence is one of the major causes of ageing and age-related diseases. The persistent presence of senescent cells in late life seems to cause disarray in a tissue-specific manner. Ageing disrupts the circadian clock system, which results in the development of many age-related diseases such as metabolic syndrome, cancer, cardiac diseases and sleep disorders and an increased susceptibility to infections. In this review, we first discuss cellular senescence and some of its basic characteristics and detrimental roles. Then, we discuss a relatively unexplored topic on the link between cellular senescence and the circadian clock and attempt to determine whether cellular senescence could be the underlying factor for circadian clock disruption.


Subject(s)
Circadian Clocks , Cellular Senescence
5.
Front Neurosci ; 15: 647589, 2021.
Article in English | MEDLINE | ID: mdl-34108855

ABSTRACT

The circadian clock possesses robust systems to maintain the rhythm approximately 24 h, from cellular to organismal levels, whereas aging is known to be one of the risk factors linked to the alternation of circadian physiology and behavior. The amount of many metabolites in the cells/body is altered with the aging process, and the most prominent metabolite among them is the oxidized form of nicotinamide adenine dinucleotide (NAD+), which is associated with posttranslational modifications of acetylation and poly-ADP-ribosylation status of circadian clock proteins and decreases with aging. However, how low NAD+ condition in cells, which mimics aged or pathophysiological conditions, affects the circadian clock is largely unknown. Here, we show that low NAD+ in cultured cells promotes PER2 to be retained in the cytoplasm through the NAD+/SIRT1 axis, which leads to the attenuated amplitude of Bmal1 promoter-driven luciferase oscillation. We found that, among the core clock proteins, PER2 is mainly affected in its subcellular localization by NAD+ amount, and a higher cytoplasmic PER2 localization was observed under low NAD+ condition. We further found that NAD+-dependent deacetylase SIRT1 is the regulator of PER2 subcellular localization. Thus, we anticipate that the altered PER2 subcellular localization by low NAD+ is one of the complex changes that occurs in the aged circadian clock.

6.
Front Neurosci ; 15: 659883, 2021.
Article in English | MEDLINE | ID: mdl-33935640

ABSTRACT

The mammalian circadian clock systems regulate the day-night variation of several physiological functions such as the sleep/wake cycle and core body temperature. Disturbance in the circadian clock due to shiftwork and chronic jetlag is related to the risk of several disorders such as metabolic syndrome and cancer. Recently, it has been thought that shiftwork increases the risk of sarcopenia which is characterized by age-related decline of muscle mass and its dysfunctions including muscle strength and/or physical performance. First, we summarize the association between circadian rhythm and the occurrence of sarcopenia and discuss its mechanistic insight by focusing on the muscle function and molecular clock gene in knockout or mutant mice. The clock gene knockout or mutant mice showed early aging phenotypes, including low survival rate and muscle loss. It suggests that improvement in the disturbance of the circadian clock plays an important role in the aging process of healthy muscles. Nutritional intake has the potential to augment muscle growth and entrain the peripheral clock. Second, we discuss the potential of chrono-nutrition in preventing aging-related muscle loss and dysfunction. We also focus on the effects of time-restricted feeding (TRF) and the distribution of protein intake across three meals.

7.
Int J Mol Sci ; 22(7)2021 Apr 02.
Article in English | MEDLINE | ID: mdl-33918226

ABSTRACT

Aging is a phenomenon underlined by complex molecular and biochemical changes that occur over time. One of the metabolites that is gaining strong research interest is nicotinamide adenine dinucleotide, NAD+, whose cellular level has been shown to decrease with age in various tissues of model animals and humans. Administration of NAD+ precursors, nicotinamide mononucleotide (NMN) and nicotinamide riboside (NR), to supplement NAD+ production through the NAD+ salvage pathway has been demonstrated to slow down aging processes in mice. Therefore, NAD+ is a critical metabolite now understood to mitigate age-related tissue function decline and prevent age-related diseases in aging animals. In human clinical trials, administration of NAD+ precursors to the elderly is being used to address systemic age-associated physiological decline. Among NAD+ biosynthesis pathways in mammals, the NAD+ salvage pathway is the dominant pathway in most of tissues, and NAMPT is the rate limiting enzyme of this pathway. However, only a few activators of NAMPT, which are supposed to increase NAD+, have been developed so far. In this review, we will focus on the importance of NAD+ and the possible application of an activator of NAMPT to promote successive aging.


Subject(s)
Aging/metabolism , NAD/metabolism , Nicotinamide Phosphoribosyltransferase/metabolism , Animals , Humans
8.
Front Neurosci ; 15: 638122, 2021.
Article in English | MEDLINE | ID: mdl-33568972

ABSTRACT

Senescent cells, which show the permanent growth arrest in response to various forms of stress, accumulate in the body with the progression of age, and are associated with aging and age-associated diseases. Although the senescent cells are growth arrested, they still demonstrate high metabolic rate and altered gene expressions, indicating that senescent cells are still active. We recently showed that the circadian clock properties, namely phase and period of the cells, are altered with the establishment of replicative senescence. However, whether cellular senescence triggers the alteration of circadian clock properties in the cells is still unknown. In this study we show that the oxidative stress-induced premature senescence induces the alterations of the circadian clock, similar to the phenotypes of the replicative senescent cells. We found that the oxidative stress-induced premature senescent cells display the prolonged period and delayed phases. In addition, the magnitude of these changes intensified over time, indicating that cellular senescence changes the circadian clock properties. Our current results corroborate with our previous findings and further confirm that cellular senescence induces altered circadian clock properties, irrespective of the replicative senescence or the stress-induced premature senescence.

9.
Genes Cells ; 25(8): 593-602, 2020 Aug.
Article in English | MEDLINE | ID: mdl-32533606

ABSTRACT

A main feature of aged organisms is the accumulation of senescent cells. Accumulated senescent cells, especially stress-induced premature senescent cells, in aged organisms lead to the decline of the regenerative potential and function of tissues. We recently reported that the over-expression of NAMPT, which is the rate-limiting enzyme in mammalian NAD+ salvage pathway, delays replicative senescence in vitro. However, whether Nampt-overexpressing cells are tolerant of stress-induced premature senescence remains unknown. Here, we show that primary mouse embryonic fibroblasts derived from Nampt-overexpressing transgenic mice (Nampt Tg-MEF cells) possess resistance against stress-induced premature senescence in vitro. We found that higher oxidative or endoplasmic reticulum (ER) stress is required to induce premature senescence in Nampt Tg-MEF cells compared to wild-type cells. Moreover, we found that Nampt Tg-MEF cells show acute expression of unfolded protein response (UPR)-related genes, which in turn would have helped to restore proteostasis and avoid cellular senescence. Our results demonstrate that NAMPT/NAD+ axis functions to protect cells not only from replicative senescence, but also from stress-induced premature senescence in vitro. We anticipate that in vivo activation of NAMPT activity or increment of NAD+ would protect tissues from the accumulation of premature senescent cells, thereby maintaining healthy aging.


Subject(s)
Cellular Senescence/physiology , Nicotinamide Phosphoribosyltransferase/genetics , Animals , Antioxidants/physiology , Cell Proliferation/physiology , Cells, Cultured , Cellular Senescence/genetics , Endoplasmic Reticulum Stress/physiology , Fibroblasts , Gene Expression/genetics , Gene Expression Regulation/genetics , Mice , Nicotinamide Phosphoribosyltransferase/metabolism , Oxidation-Reduction , Oxidative Stress/physiology
10.
Aging (Albany NY) ; 11(3): 950-973, 2019 02 09.
Article in English | MEDLINE | ID: mdl-30738414

ABSTRACT

Over the last decade, a wide array of evidence has been accumulated that disruption of circadian clock is prone to cause age-related diseases and premature aging. On the other hand, aging has been identified as one of the risk factors linked to the alteration of circadian clock. These evidences suggest that the processes of aging and circadian clock feedback on each other at the animal level. However, at the cellular level, we recently revealed that the primary fibroblast cells derived from Bmal1-/- mouse embryo, in which circadian clock is completely disrupted, do not demonstrate the acceleration of cellular aging, i.e., cellular senescence. In addition, little is known about the impact of cellular senescence on circadian clock. In this study, we show for the first time that senescent cells possess a longer circadian period with delayed peak-time and that the variability in peak-time is wider in the senescent cells compared to their proliferative counterparts, indicating that senescent cells show alterations of circadian clock. We, furthermore, propose that investigation at cellular level is a powerful and useful approach to dissect molecular mechanisms of aging in the circadian clock.


Subject(s)
Cellular Senescence , Circadian Clocks , Cell Line , Circadian Rhythm , Humans , Period Circadian Proteins/metabolism
11.
J Biol Chem ; 293(31): 12167-12176, 2018 08 03.
Article in English | MEDLINE | ID: mdl-29895619

ABSTRACT

Somites are a pair of epithelial spheres beside a neural tube and are formed with an accurate periodicity during embryogenesis in vertebrates. It has been known that Hes7 is one of the core clock genes for somitogenesis, and its expression domain is restricted in the presomitic mesoderm (PSM). However, the molecular mechanism of how Hes7 transcription is regulated is not clear. Here, using transgenic mice and luciferase-based reporter assays and in vitro binding assays, we unravel the mechanism by which Hes7 is expressed exclusively in the PSM. We identified a Hes7 essential region residing -1.5 to -1.1 kb from the transcription start site of mouse Hes7, and this region was indispensable for PSM-specific Hes7 expression. We also present detailed analyses of cis-regulatory elements within the Hes7 essential region that directs Hes7 expression in the PSM. Hes7 expression in the PSM was up-regulated through the E-box, T-box, and RBPj-binding element in the Hes7 essential region, presumably through synergistic signaling involving mesogenin1, T-box6 (Tbx6), and Notch. Furthermore, we demonstrate that Tbx18, Ripply2, and Hes7 repress the activation of the Hes7 essential region by the aforementioned transcription factors. Our findings reveal that a unified transcriptional regulatory network involving a Hes7 essential region confers robust PSM-specific Hes7 gene expression.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/chemistry , Basic Helix-Loop-Helix Transcription Factors/genetics , Mesoderm/metabolism , Receptor, Notch1/metabolism , Somites/metabolism , T-Box Domain Proteins/metabolism , Transcription Factors/metabolism , Animals , Base Sequence , Basic Helix-Loop-Helix Transcription Factors/metabolism , E-Box Elements , Gene Expression Regulation , Gene Expression Regulation, Developmental , Mesoderm/chemistry , Mesoderm/embryology , Mice , Receptor, Notch1/genetics , Repressor Proteins/genetics , Repressor Proteins/metabolism , Signal Transduction , Somites/embryology , T-Box Domain Proteins/genetics , Transcription Factors/genetics
12.
Chronobiol Int ; 35(5): 730-738, 2018 05.
Article in English | MEDLINE | ID: mdl-29372841

ABSTRACT

Bmal1 is a core circadian clock gene. Bmal1-/- mice show disruption of the clock and premature aging phenotypes with a short lifespan. However, little is known whether disruption of Bmal1 leads to premature aging at cellular level. Here, we established primary mouse embryonic fibroblast (MEF) cells derived from Bmal1-/- mice and investigated its effects on cellular senescence. Unexpectedly, Bmal1-/- primary MEFs that showed disrupted circadian oscillation underwent neither premature replicative nor stress-induced cellular senescence. Our results therefore uncover that Bmal1 is not required for in vitro cellular senescence, suggesting that circadian clock does not control in vitro cellular senescence.


Subject(s)
ARNTL Transcription Factors/deficiency , Cellular Senescence , Circadian Rhythm , Fibroblasts/metabolism , ARNTL Transcription Factors/genetics , Animals , Cell Proliferation , Cells, Cultured , Endoplasmic Reticulum Stress , Fibroblasts/pathology , Gene Expression Regulation , Mice, Knockout , Oxidative Stress , Time Factors
13.
Genes Cells ; 22(12): 982-992, 2017 Dec.
Article in English | MEDLINE | ID: mdl-29178516

ABSTRACT

Senescent cells accumulate in tissues of aged animals and deteriorate tissue functions. The elimination of senescent cells from aged mice not only attenuates progression of already established age-related disorders, but also extends median lifespan. Nicotinamide phosphoribosyltransferase (NAMPT), the rate-limiting enzyme in mammalian NAD+ salvage pathway, has shown a protective effect on cellular senescence of human primary cells. However, it still remains unclear how NAMPT has a protective impact on aging in vitro and in vivo. In this study, we found that primary mouse embryonic fibroblast (MEF) cells undergo progressive decline of NAMPT and NAD+ contents during serial passaging before becoming senescent. Furthermore, we showed that constitutive Nampt over-expression increases cellular NAD+ content and delays cellular senescence of MEF cells in vitro. We further found that constitutive Nampt over-expression increases SIRT1 activity, increases the expression of antioxidant genes, superoxide dismutase 2 and catalase and promotes resistance against oxidative stress. These findings suggest that Nampt over-expression in MEF cells delays cellular senescence by the mitigation of oxidative stress via the upregulation of superoxide dismutase 2 and catalase gene expressions by SIRT1 activation.


Subject(s)
Antioxidants/metabolism , Cellular Senescence , Cytokines/metabolism , Gene Expression Regulation , Nicotinamide Phosphoribosyltransferase/metabolism , Sirtuin 1/metabolism , Animals , Cell Proliferation , Cells, Cultured , Cytokines/genetics , Embryo, Mammalian/cytology , Embryo, Mammalian/metabolism , Female , Fibroblasts/cytology , Fibroblasts/metabolism , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Nicotinamide Phosphoribosyltransferase/genetics , Oxidative Stress , Sirtuin 1/genetics
14.
Biomed Res Int ; 2016: 3208429, 2016.
Article in English | MEDLINE | ID: mdl-28050554

ABSTRACT

Gene expression is known to be a stochastic phenomenon. The stochastic gene expression rate is thought to be altered by topological change of chromosome and/or by chromatin modifications such as acetylation and methylation. Changes in mechanical properties of chromosome/chromatin by soluble factors, mechanical stresses from the environment, or metabolites determine cell fate, regulate cellular functions, or maintain cellular homeostasis. Circadian clock, which drives the expression of thousands of genes with 24-hour rhythmicity, has been known to be indispensable for maintaining cellular functions/homeostasis. During the last decade, it has been demonstrated that chromatin also undergoes modifications with 24-hour rhythmicity and facilitates the fine-tuning of circadian gene expression patterns. In this review, we cover data which suggests that chromatin structure changes in a circadian manner and that NAD+ is the key metabolite for circadian chromatin remodeling. Furthermore, we discuss the relationship among circadian clock, NAD+ metabolism, and aging/age-related diseases. In addition, the interventions of NAD+ metabolism for the prevention and treatment of aging and age-related diseases are also discussed.


Subject(s)
Aging/metabolism , Chromatin Assembly and Disassembly , Circadian Rhythm , NAD/metabolism , Animals , Circadian Clocks , Humans , Metabolome
15.
PLoS Biol ; 13(11): e1002293, 2015.
Article in English | MEDLINE | ID: mdl-26562092

ABSTRACT

Intracellular circadian clocks, composed of clock genes that act in transcription-translation feedback loops, drive global rhythmic expression of the mammalian transcriptome and allow an organism to anticipate to the momentum of the day. Using a novel clock-perturbing peptide, we established a pivotal role for casein kinase (CK)-2-mediated circadian BMAL1-Ser90 phosphorylation (BMAL1-P) in regulating central and peripheral core clocks. Subsequent analysis of the underlying mechanism showed a novel role of CRY as a repressor for protein kinase. Co-immunoprecipitation experiments and real-time monitoring of protein-protein interactions revealed that CRY-mediated periodic binding of CK2ß to BMAL1 inhibits BMAL1-Ser90 phosphorylation by CK2α. The FAD binding domain of CRY1, two C-terminal BMAL1 domains, and particularly BMAL1-Lys537 acetylation/deacetylation by CLOCK/SIRT1, were shown to be critical for CRY-mediated BMAL1-CK2ß binding. Reciprocally, BMAL1-Ser90 phosphorylation is prerequisite for BMAL1-Lys537 acetylation. We propose a dual negative-feedback model in which a CRY-dependent CK2-driven posttranslational BMAL1-P-BMAL1 loop is an integral part of the core clock oscillator.


Subject(s)
ARNTL Transcription Factors/metabolism , Casein Kinase II/metabolism , Circadian Clocks , Cryptochromes/metabolism , Protein Processing, Post-Translational , ARNTL Transcription Factors/chemistry , ARNTL Transcription Factors/genetics , Animals , Casein Kinase II/chemistry , Casein Kinase II/genetics , Cell Line , Cells, Cultured , Cryptochromes/chemistry , Cryptochromes/genetics , Embryo, Mammalian/cytology , Humans , Mice , Mice, Knockout , Mice, Transgenic , Mutation , Phosphorylation , Protein Interaction Domains and Motifs , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/metabolism , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism
16.
Cell Rep ; 12(11): 1887-901, 2015 Sep 22.
Article in English | MEDLINE | ID: mdl-26344767

ABSTRACT

Rett syndrome (RTT) is a neurodevelopmental disorder caused by MECP2 mutations. Although emerging evidence suggests that MeCP2 deficiency is associated with dysregulation of mechanistic target of rapamycin (mTOR), which functions as a hub for various signaling pathways, the mechanism underlying this association and the molecular pathophysiology of RTT remain elusive. We show here that MeCP2 promotes the posttranscriptional processing of particular microRNAs (miRNAs) as a component of the microprocessor Drosha complex. Among the MeCP2-regulated miRNAs, we found that miR-199a positively controls mTOR signaling by targeting inhibitors for mTOR signaling. miR-199a and its targets have opposite effects on mTOR activity, ameliorating and inducing RTT neuronal phenotypes, respectively. Furthermore, genetic deletion of miR-199a-2 led to a reduction of mTOR activity in the brain and recapitulated numerous RTT phenotypes in mice. Together, these findings establish miR-199a as a critical downstream target of MeCP2 in RTT pathogenesis by linking MeCP2 with mTOR signaling.


Subject(s)
Methyl-CpG-Binding Protein 2/metabolism , MicroRNAs/metabolism , Rett Syndrome/metabolism , TOR Serine-Threonine Kinases/metabolism , Animals , Disease Models, Animal , Methyl-CpG-Binding Protein 2/antagonists & inhibitors , Methyl-CpG-Binding Protein 2/genetics , Mice , Mice, Knockout , MicroRNAs/antagonists & inhibitors , MicroRNAs/genetics , Phenotype , Rett Syndrome/genetics , Ribonuclease III/genetics , Ribonuclease III/metabolism , Signal Transduction , TOR Serine-Threonine Kinases/genetics , Up-Regulation
17.
Sci Rep ; 4: 6462, 2014 Sep 24.
Article in English | MEDLINE | ID: mdl-25248974

ABSTRACT

A set of genes in the posterior end of developing mouse embryos shows oscillatory expression, thereby regulating periodic somite segmentation. Although the mechanism for generating oscillation has extensively been clarified, what regulates the oscillation period is still unclear. We attempted to elongate the oscillation period by increasing the time to transcribe Hes7 in this research. We generated knock-in mice, in which a large intron was inserted into Hes7 3'UTR. The exogenous intron was unexpectedly not properly spliced out and the transcripts were prematurely terminated. Consequently, Hes7 mRNA lost its 3'UTR, thereby reducing the amount of Hes7 protein. Oscillation was damped in the knock-in embryos and periodic somite segmentation does not occur properly. Thus, we demonstrated that Hes7 3'UTR is essential to accumulate adequate amounts of Hes7 protein for the somite segmentation clock that orchestrates periodic somite formation.


Subject(s)
3' Untranslated Regions/genetics , Basic Helix-Loop-Helix Transcription Factors/genetics , Body Patterning/genetics , Embryo, Mammalian/cytology , Somites/embryology , Somites/metabolism , Animals , Base Sequence , Cells, Cultured , Embryo, Mammalian/metabolism , Embryonic Development , In Situ Hybridization , Mice , Molecular Sequence Data , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Sequence Homology, Nucleic Acid , Somites/cytology
18.
FEBS J ; 281(1): 146-56, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24165510

ABSTRACT

Somite segmentation, a prominent periodic event in the development of vertebrates, is instructed by cyclic expression of several genes, including Hes7 and Lunatic fringe (Lfng). Transcriptional regulation accounts for the cyclic expression. In addition, because the expression patterns vary in a cycle, rapid turnover of mRNAs should be involved in the cyclic expression, although its contribution remains unclear. Here, we demonstrate that 3'-UTR-dependent rapid turnover of Lfng and Hes7 plays a critical role in their dynamic expression patterns. The regions active in the transcription of Lfng and Hes7 are wholly overlapped in the posterior presomitic mesoderm (PSM) of the mouse embryo. However, their distribution patterns are slightly different; Hes7 mRNA shows a broader distribution pattern than Lfng mRNA in the posterior PSM. Lfng mRNA is less stable than Hes7 mRNA, where their 3'-UTRs are responsible for the different stability. Using transgenic mice expressing Venus under the control of the Hes7 promoter, which leads to cyclic transcription in the PSM, we reveal that the Lfng 3'-UTR provides the narrow distribution pattern of Lfng mRNA, whereas the Hes7 3'-UTR contributes the relatively broad distribution pattern of Hes7 mRNA. Thus, we conclude that 3'-UTR-dependent mRNA stability accounts for the differential distribution patterns of Lfng and Hes7 mRNA. Our findings suggest that 3'-UTR-dependent regulation of mRNA turnover plays a crucial role in the diverse patterns of mRNA distribution during development.


Subject(s)
3' Untranslated Regions/genetics , Basic Helix-Loop-Helix Transcription Factors/physiology , Gene Expression Regulation, Developmental , Glycosyltransferases/physiology , RNA, Messenger/metabolism , Animals , Blotting, Western , Body Patterning , Female , HEK293 Cells , Humans , In Situ Hybridization , Male , Mesoderm/cytology , Mesoderm/metabolism , Mice , Mice, Transgenic , Promoter Regions, Genetic/genetics , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction , Somites/cytology , Somites/metabolism
19.
Proc Natl Acad Sci U S A ; 110(9): 3333-8, 2013 Feb 26.
Article in English | MEDLINE | ID: mdl-23341587

ABSTRACT

Circadian rhythms govern a wide variety of physiological and metabolic functions in many organisms, from prokaryotes to humans. We previously reported that silent information regulator 1 (SIRT1), a NAD(+)-dependent deacetylase, contributes to circadian control. In addition, SIRT1 activity is regulated in a cyclic manner in virtue of the circadian oscillation of the coenzyme NAD(+). Here we used specific SIRT1 activator compounds both in vitro and in vivo. We tested a variety of compounds to show that the activation of SIRT1 alters CLOCK:BMAL1-driven transcription in different systems. Activation of SIRT1 induces repression of circadian gene expression and decreases H3 K9/K14 acetylation at corresponding promoters in a time-specific manner. Specific activation of SIRT1 was demonstrated in vivo using liver-specific SIRT1-deficient mice, where the effect of SIRT1 activator compounds was shown to be dependent on SIRT1. Our findings demonstrate that SIRT1 can fine-tune circadian rhythm and pave the way to the development of pharmacological strategies to address a broad range of therapeutic indications.


Subject(s)
Circadian Rhythm/genetics , Enzyme Activators/pharmacology , Sirtuin 1/metabolism , ARNTL Transcription Factors/metabolism , Animals , CLOCK Proteins/genetics , CLOCK Proteins/metabolism , Cell Line , Chromatin/metabolism , Circadian Rhythm/drug effects , Gene Expression Regulation/drug effects , Heterocyclic Compounds, 4 or More Rings/pharmacology , Humans , Liver/drug effects , Liver/metabolism , Mice , Mice, Knockout , NAD/metabolism , Transcription, Genetic/drug effects
20.
Science ; 324(5927): 654-7, 2009 May 01.
Article in English | MEDLINE | ID: mdl-19286518

ABSTRACT

Many metabolic and physiological processes display circadian oscillations. We have shown that the core circadian regulator, CLOCK, is a histone acetyltransferase whose activity is counterbalanced by the nicotinamide adenine dinucleotide (NAD+)-dependent histone deacetylase SIRT1. Here we show that intracellular NAD+ levels cycle with a 24-hour rhythm, an oscillation driven by the circadian clock. CLOCK:BMAL1 regulates the circadian expression of NAMPT (nicotinamide phosphoribosyltransferase), an enzyme that provides a rate-limiting step in the NAD+ salvage pathway. SIRT1 is recruited to the Nampt promoter and contributes to the circadian synthesis of its own coenzyme. Using the specific inhibitor FK866, we demonstrated that NAMPT is required to modulate circadian gene expression. Our findings in mouse embryo fibroblasts reveal an interlocked transcriptional-enzymatic feedback loop that governs the molecular interplay between cellular metabolism and circadian rhythms.


Subject(s)
Circadian Rhythm , Cytokines/metabolism , Feedback, Physiological , Gene Expression Regulation , NAD/metabolism , Nicotinamide Phosphoribosyltransferase/metabolism , Sirtuins/metabolism , Trans-Activators/metabolism , ARNTL Transcription Factors , Acrylamides/pharmacology , Animals , Basic Helix-Loop-Helix Transcription Factors/metabolism , Biological Clocks , CLOCK Proteins , Cell Line , Chromatin Assembly and Disassembly , Cytokines/antagonists & inhibitors , Cytokines/genetics , Enzyme Inhibitors/pharmacology , Male , Mice , Mice, Inbred BALB C , Mice, Knockout , Niacinamide/metabolism , Nicotinamide Phosphoribosyltransferase/antagonists & inhibitors , Nicotinamide Phosphoribosyltransferase/genetics , Piperidines/pharmacology , Promoter Regions, Genetic , Sirtuin 1 , Trans-Activators/genetics , Transcription, Genetic
SELECTION OF CITATIONS
SEARCH DETAIL