Elsevier

Experimental Gerontology

Volume 55, July 2014, Pages 70-79
Experimental Gerontology

Differential modulation of clock gene expression in the suprachiasmatic nucleus, liver and heart of aged mice

https://doi.org/10.1016/j.exger.2014.03.011Get rights and content

Highlights

  • Advanced age affects the clock gene network at central and peripheral levels.

  • In SCN the decline of the rhythmic function of CCGS impairs circadian output.

  • At the peripheral level a deterioration of the rhythm of core clock genes prevails.

Abstract

Studies on the molecular clockwork during aging have been hitherto addressed to core clock genes. These previous investigations indicate that circadian profiles of core clock gene expression at an advanced age are relatively preserved in the master circadian pacemaker and the hypothalamic suprachiasmatic nucleus (SCN), and relatively impaired in peripheral tissues. It remains to be clarified whether the effects of aging are confined to the primary loop of core clock genes, or also involve secondary clock loop components, including Rev-erbα and the clock-controlled genes Dbp and Dec1. Using quantitative real-time RT-PCR, we here report a comparative analysis of the circadian expression of canonical core clock genes (Per1, Per2, Cry1, Cry2, Clock and Bmal1) and non-core clock genes (Rev-erbα, Dbp and Dec1) in the SCN, liver, and heart of 3 month-old vs 22 month-old mice. The results indicate that circadian clock gene expression is significantly modified in the SCN and peripheral oscillators of aged mice. These changes are not only highly tissue-specific, but also involve different clock gene loops. In particular, we here report changes of secondary clock loop components in the SCN, changes of the primary clock loop in the liver, and minor changes of clock gene expression in the heart of aged mice. The present findings outline a track to further understanding of the role of primary and secondary clock loop components and their crosstalk in the impairment of circadian output which characterizes aging.

Introduction

The mammalian circadian system consists of multiple oscillators distributed throughout the organism and a central pacemaker in the suprachiasmatic nucleus (SCN) of the hypothalamus which coordinates the entire system making it coherent. The molecular mechanisms underlying circadian function are similar in central and peripheral oscillators and consist of highly conserved genes, the so-called clock genes. Together with their protein products, clock genes are organized in transcription–translation feedback loops able to complete a cycle in ~ 24 h (Ko and Takahashi, 2006).

The primary feedback loop, or core loop, consists of different components. The positive transcriptional activators CLOCK and BMAL1 (or its paralog NPAS2) heterodimerize and promote transcription of the Period genes (Per1 and Per2) and Cryptochrome genes (Cry1 and Cry2). The negative components include PER and CRY which heterodimerize, translocate back to the nucleus and repress their own transcription by acting on the CLOCK–BMAL1 complex (Sato et al., 2006). The CLOCK–BMAL1 complex is involved in an additional loop, interlocked with the core loop, that induces the expression of the nuclear hormone receptors Rev-erbα and Rorα. Their products, REV-ERBα and RORα, bind competitively to Bmal1 and determine the cycling of Bmal1 expression by suppressing and activating, respectively, its transcription (Guillaumond et al., 2005, Preitner et al., 2002).

A key function of the molecular clock machinery is represented by the circadian output, whose efficacy depends on the coordinated circadian behavior and physiology of the entire organism (see for reviews Takahashi et al., 2008, Albrecht, 2012). The transcriptional regulation of this function is achieved not only directly via core loop transcription factors, but also indirectly via auxiliary loops interlocking with the core loop, which include the transcription factors Dbp and Dec1. These interlocking loops and their constituents, the so-called first order “clock-controlled genes” (CCGS) (Reppert and Weaver, 2002) play a crucial intermediate role in conveying circadian signals to downstream local clock-controlled genes, specific for each tissue (Liu et al., 2008).

The cyclic transcription of Dbp is under the control of CLOCK–BMAL1, and is repressed by PER/CRY heterodimers. In turn, DBP can regulate the core clock machinery activating Per1 transcription. Dec1 rhythmic transcription is positively regulated by CLOCK–BMAL1, while DEC1, interacting with BMAL1 for E-box occupation, negatively acts on CLOCK/BMAL1 activity (Honma et al., 2002, Kawamoto et al., 2004; see also for review Bonaconsa et al., 2013).

Evidence in experimental animals and in humans indicates that the circadian timing system is progressively modified with advancing age, as shown by the reduced amplitude, phase and period length of circadian rhythms, increased tendency towards internal desynchronization and deficient responsiveness to phase-shifting stimuli with loss of synchronization to the environment (Gibson et al., 2009, Hofman and Swaab, 2006). Circadian rhythms of locomotor activity, core body temperature, blood pressure, and the sleep–wake cycle are especially affected (Reppert and Weaver, 2002). The possibility that these aging-related changes are due to impairment of the molecular clockwork in the SCN and/or in peripheral tissues has been repeatedly investigated.

In the SCN, Per1 and Cry1 circadian rhythms have been reported to be preserved at an advanced age in mice (Weinert et al., 2001), rats (Asai et al., 2001), and hamsters (Kolker et al., 2003). A decrease of Per2 circadian oscillation has been reported in aged mice (Weinert et al., 2001), but not in rats (Asai et al., 2001) and hamsters (Kolker et al., 2003). An aging-related decline in Clock and Bmal1 circadian rhythm has been reported in hamsters (Kolker et al., 2003).

Studies exploring the impact of aging on clock gene expression in peripheral tissues have indicated that Per1 expression is preserved in aged rats (Asai et al., 2001, Yamazaki et al., 2002). Moderate aging-related changes of Per2 and Bmal1 circadian rhythms have been reported in the macaque pituitary gland (Sitzmann et al., 2010). Analyses conducted in rats at only two time points for 24 h have indicated a significant age-related decrease of Per1–3 mRNAs and unchanged Cry1–2, Clock and Bmal1 mRNAs in the liver, associated with Per1–3 decrease and Bmal1 increase in the heart (Claustrat et al., 2005).

It remains to be clarified whether a definite dysfunction of the molecular clockwork occurs in the circadian impairment that characterizes aging. The matter is still open not only because the available data are limited and in some instances controversial, but also because they mainly focus on clock genes of the core loop, while information on the clock output function mediated by CCGS is limited.

On this basis, using quantitative real-time RT-PCR, we here assessed in young and aged mice the circadian expression of canonical core clock genes (Per1–2, Cry1–2, Clock and Bmal1), of the transcription factor Rev-erbα which, as mentioned above, forms with Bmal1 a central regulatory loop, and of the first-order CCGS Dec1 and Dbp. In order to compare the clock properties of central and peripheral oscillators, we here investigated the SCN, liver and heart.

Section snippets

Animals and tissue sampling

Two groups of male Balb/c mice of different ages, 3 and 22 month-old, respectively (n = 24 per age group), were housed in the animal facility at a standard room temperature (21 ± 2 °C), under a 12 h/12 h light/dark (LD) cycle, and fed ad libitum. Life span of Balb/c mice is about 23 months (Jucker and Ingram, 1997). All efforts were made to avoid animal suffering and minimize the number of animals used. The experiments received approval by the Animal Care and Use Committee of the University of Verona

Results

To analyze the mechanism underlying age-related impairment of circadian clock function, we examined the 24 h patterns of mRNA expression of nine clock genes, including the six canonical genes of the core clock feedback loop (Per1, Per2, Cry1, Cry2, Clock, Bmal1) and three clock genes representative of the CCGS (Rev-erbα, Dbp and Dec1). As mentioned previously, the latter genes, components of clock gene secondary loops interlocking with the core loop, act as intermediaries in circadian signaling.

Discussion

The present study shows that aging is associated with changes of the clock gene network which show distinct features at central and peripheral levels. The findings obtained in the different analyzed tissues are discussed below.

Conflict of interest

The authors declare no conflicts of interest.

Acknowledgments

The authors are grateful to E. Moccheggiani (INRCCA, Ancona, Italy) for his help in obtaining aged animals, and to anonymous Experimental Gerontology reviewers for their helpful criticisms. The support of intramural funds of the University of Verona is gratefully acknowledged. Thanks are due to Valeria Colavito for her help in statistical analyses.

References (75)

  • F. Kawakami et al.

    Loss of day–night differences in VIP mRNA levels in the suprachiasmatic nucleus of aged rats

    Neurosci. Lett.

    (1997)
  • T. Kawamoto et al.

    A novel autofeedback loop of Dec1 transcription involved in circadian rhythm regulation

    Biochem. Biophys. Res. Commun.

    (2004)
  • K.J. Livak et al.

    Analysis of relative gene expression data using real-time quantitative PCR and the 2(− Delta Delta C(T)) method

    Methods

    (2001)
  • K. Oishi et al.

    Antiphase circadian expression between Bmal1 and period homologue mRNA in the suprachiasmatic nucleus and peripheral tissues of rats

    Biochem. Biophys. Res. Commun.

    (1998)
  • K. Oishi et al.

    Genome-wide expression analysis of mouse liver reveals CLOCK-regulated circadian output genes

    J. Biol. Chem.

    (2003)
  • K. Oishi et al.

    Circadian mRNA expression of coagulation and fibrinolytic factors is organ-dependently disrupted in aged mice

    Exp. Gerontol.

    (2011)
  • N. Preitner et al.

    The orphan nuclear receptor REV-ERB alpha controls circadian transcription within the positive limb of the mammalian circadian oscillator

    Cell

    (2002)
  • B. Roozendaal et al.

    Changes in vasopressin cells of the rat suprachiasmatic nucleus with aging

    Brain Res.

    (1987)
  • A. Sadki et al.

    Suppressors, receptors and effects of cytokines on the aging mouse biological clock

    Neurobiol. Aging

    (2007)
  • I. Schmutz et al.

    The role of clock genes and rhythmicity in the liver

    Mol. Cell. Endocrinol.

    (2012)
  • B.D. Sitzmann et al.

    Effects of age on clock gene expression in the rhesus macaque pituitary gland

    Neurobiol. Aging

    (2010)
  • C.A. Wyse et al.

    Impact of aging on diurnal expression patterns of CLOCK and BMAL1 in the mouse brain

    Brain Res.

    (2010)
  • L. Yan et al.

    Per1 and Per2 gene expression in the rat suprachiasmatic nucleus: circadian profile and the compartment-specific response to light

    Neuroscience

    (1999)
  • B. Zheng et al.

    Nonredundant roles of the mPer1 and mPer2 genes in the mammalian circadian clock

    Cell

    (2001)
  • M.J. Zylka et al.

    Three period homologs in transcripts outside of brain

    Neuron

    (1998)
  • M. Asai et al.

    Circadian profile of Per gene mRNA expression in the suprachiasmatic nucleus, paraventricular nucleus, and pineal body of aged rats

    J. Neurosci. Res.

    (2001)
  • M. Bonaconsa et al.

    Cell clocks and neuronal networks: neuron ticking and synchronization in aging and aging-related neurodegenerative disease

    Curr. Alzheimer. Res.

    (2013)
  • M.S. Bray et al.

    Disruption of the circadian clock within the cardiomyocyte influences myocardial contractile function, metabolism, and gene expression

    Am. J. Physiol. Heart Circ. Physiol.

    (2008)
  • G. Cavadini et al.

    TNF-α suppresses the expression of clock genes by interfering with E-box-mediated transcription

    Proc. Natl. Acad. Sci. U. S. A.

    (2007)
  • C.S. Colwell

    Linking neural activity and molecular oscillations in the SCN

    Nat. Rev. Neurosci.

    (2011)
  • A.M. Curtis et al.

    Circadian variation of blood pressure and the vascular response to asynchronous stress

    Proc. Natl. Acad. Sci. U. S. A.

    (2007)
  • X.-H. Deng et al.

    Glial transcripts and immune-challenged glia in the suprachiasmatic nucleus of young and aged mice

    Chronobiol. Int.

    (2010)
  • C. Dibner et al.

    The mammalian circadian timing system: organization and coordination of central and peripheral clocks

    Annu. Rev. Physiol.

    (2010)
  • H. Duez et al.

    Rev-erb-α: an integrator of circadian rhythms and metabolism

    J. Appl. Physiol.

    (2009)
  • D.J. Durgan et al.

    The intrinsic circadian clock within the cardiomyocyte

    Am. J. Physiol. Heart Circ. Physiol.

    (2005)
  • D.J. Durgan et al.

    Evidence suggesting that the cardiomyocyte circadian clock modulates responsiveness of the heart to hypertrophic stimuli in mice

    Chronobiol. Int.

    (2011)
  • S. Farajnia et al.

    Evidence for neuronal desynchrony in the aged suprachiasmatic nucleus clock

    J. Neurosci.

    (2012)
  • Cited by (79)

    • Autophagy, Clock Genes, and Cardiovascular Disease

      2023, Canadian Journal of Cardiology
    • Age-related changes in circadian rhythms and non-visual responses to light during adulthood

      2023, Encyclopedia of Sleep and Circadian Rhythms: Volume 1-6, Second Edition
    • Deciphering clock genes as emerging targets against aging

      2022, Ageing Research Reviews
      Citation Excerpt :

      Tahara et al. (2017) also pointed out that the peripheral clocks including kidney, liver, and submandibular gland of aged mice are almost normal whether the light-dark information presents or not, even if SCN-driven behavioral rhythms are impaired. In addition, the diurnal rhythm of the expression of BMAL1, PER1, PER2, and CRY1 preserves in the aged mice heart, though with severe amplitude dampening of BMAL1, PER1, and PER2 expression (Bonaconsa et al., 2014). These may imply that the expression patterns of clock genes in different organs are not exactly parallel or/and the organ sensitivity of clock genes expression is different towards age-related attritions, highlighting the necessity for cell and organ type-specific approaches to extensively study the roles of clock genes in aging.

    • The core circadian component, Bmal1, is maintained in the pineal gland of old killifish brain

      2021, iScience
      Citation Excerpt :

      However, the mechanisms underlying age-dependent changes of circadian clock system and physiological rhythms have yet to be clearly defined. For example, It is still unclear which accessory and/or core clock components undergo age-dependent changes in function and which are responsible for the age-dependent changes in physiological rhythms (Banks et al., 2016; Bonaconsa et al., 2014; Weinert et al., 2001). Conversely, it is known that defects in circadian components can affect longevity and age-related phenotypes (Kondratov et al., 2006, 2009; Sun et al., 2006).

    View all citing articles on Scopus
    View full text