ArticlePDF AvailableLiterature Review

A ticking clock links metabolic pathways and organ systems function in health and disease

Authors:

Abstract and Figures

Rhythmic variations with 24-h periodicity hallmark homeostatic regulation, metabolic processes and organ systems function, driven by a circadian timing system composed of central and peripheral oscillators. Recent reports suggest that disrupted circadian rhythmicity of physiology and behavior severely alters body homeostasis. Nuclear receptors and transcriptional regulators sense hormonal and metabolic cues and manage the rhythmic patterns of chromatin remodelling and gene expression, playing a key role in the cross talk between the circadian clock circuitry, the metabolic pathways and the organ systems. The alteration of this cross talk contributes to the pathophysiology of metabolic, degenerative, immune-related and neoplastic diseases.
Content may be subject to copyright.
REVIEW ARTICLE
A ticking clock links metabolic pathways and organ systems
function in health and disease
Manlio Vinciguerra Maria Florencia Tevy
Gianluigi Mazzoccoli
Received: 16 January 2013 / Accepted: 19 March 2013
ÓSpringer-Verlag Italia 2013
Abstract Rhythmic variations with 24-h periodicity
hallmark homeostatic regulation, metabolic processes and
organ systems function, driven by a circadian timing sys-
tem composed of central and peripheral oscillators. Recent
reports suggest that disrupted circadian rhythmicity of
physiology and behavior severely alters body homeostasis.
Nuclear receptors and transcriptional regulators sense
hormonal and metabolic cues and manage the rhythmic
patterns of chromatin remodelling and gene expression,
playing a key role in the cross talk between the circadian
clock circuitry, the metabolic pathways and the organ
systems. The alteration of this cross talk contributes to the
pathophysiology of metabolic, degenerative, immune-
related and neoplastic diseases.
Keywords Circadian rhythm Clock gene
Neuroendocrine-immune system Metabolism
Introduction
Living organisms on Earth including humans may be
deemed as complex hierarchical multi-component systems
facing environmental conditions that change over time,
exposing internal equilibrium to failures of different types
and physiological challenges that may be episodic/unex-
pected or periodic/predictable. The biological processes
underlying body homeostasis maintenance must afford
adaptive responses to sporadic events, and anticipatory
adjustments to expected fluctuations, providing selective
advantage [1]. In this context, organismal regulatory
phenomena are characterized by time-related variations
realizing a multifrequency/multiphase array of rhythms,
prevalently with 24-h periodicity and defined circadian
(from the Latin circa, about and dies, a day) [1]. They are
synchronized by a timing system comprising central oscil-
lators in the hypothalamic suprachiasmatic nuclei (SCN)
and subsidiary oscillators in peripheral tissues, functioning
as cellular biological clocks, and driven by the SCN through
the anatomic hardwire of neural circuits and the diffuse
modulation of humoral mediators, but also entrainable by
local signals, such as metabolite flux related to feeding [1].
The master pacemaker is timed by photic cues conveyed by
the retinohyopthalamic tract, entraining the circadian
rhythmicity of body physiology and behavior to the daily
geophysical cycles of alternating light and darkness [2].
A proper temporal organization of homeostatic processes
along with neuroendocrine-immune and organ systems
function is critical for the preservation of health, and recent
reports have put in evidence that alteration of the biological
clocks and/or desynchronization related to altered behav-
ioral cycles of sleep/wake, rest/activity and fasting/feeding,
and caused by exposure to electric light at night with
intentional sleep restriction, may underlie the elevated and
increasing risk of cancer worldwide [3]. Besides, alteration
of the circadian timing system contributes to metabolic
derangement [4], and the epidemic of metabolic syndrome,
obesity and diabetes mellitus is attaining outstanding
relevance, impacting on cardiovascular risk in the general
M. Vinciguerra
Division of Medicine, Institute of Liver and Digestive Health,
University College London, Royal Free Campus, London, UK
M. F. Tevy G. Mazzoccoli (&)
Division of Internal Medicine and Chronobiology Unit,
Department of Medical Sciences, IRCCS Scientific Institute and
Regional General Hospital ‘Casa Sollievo della Sofferenza,’
Opera di Padre Pio da Pietrelcina, Cappuccini Avenue,
71013 San Giovanni Rotondo, FG, Italy
e-mail: g.mazzoccoli@operapadrepio.it
123
Clin Exp Med
DOI 10.1007/s10238-013-0235-8
population and representing an important challenge for the
health-care systems. The recognition of the relevance of
circadian rhythmicity in human physiology calls for new
therapeutic strategies addressing the role played by
disruption of the circadian clock circuitry in the patho-
physiology of metabolic, as well as degenerative, immune-
related and neoplastic diseases [5,6].
The molecular clockwork
A molecular clockwork goes ticking in every single cell,
functioning through a feedback loop of transcription/
translation involving a set of genes, called clock genes,
encoding transcription factors (CLOCK/NPAS2 and
BMAL1-2) that heterodimerize and activate expression of
genes encoding circadian proteins (PER 1-3 and CRY 1-2),
which in turn inhibit clock gene transcription [7]. The
heterodimeric transcriptional complex also activates the
expression of the genes encoding the nuclear receptors
REVERBa/band RORa/c, which play negative and posi-
tive transcriptional roles, respectively, and steer amplitude
and robustness of oscillation in this autoregulatory tran-
scriptional feedback mechanism that completes a cycle in
approximately 24 h [8,9]. The heterodimers, the nuclear
receptors and the circadian proteins also drive the expres-
sion of clock-controlled genes encoding transcription fac-
tors (HLF, TEF, DBP, E4BP4, DEC1-2), which entrain
transcriptional processes to environmental cues and syn-
chronize gene expression and epigenetic modification in
metabolic pathways with energy balance, redox state and
fasting/feeding-rest/activity cycles through interaction with
nutrient sensors [1,10]. A critical and somehow essential
role in the circadian oscillatory functioning of the molecular
clock is played by rhythmic post-translational modifications
operated predominantly by processes of phosphorylation,
catalyzed by casein kinases, AMP-activated protein kinase
and glycogen synthase kinase-3b, and through sumoylation
and ubiquitylation [1]. An additional mechanism of circa-
dian transcriptional regulation is represented by epigenetic
modifications represented by chromatin remodelling
through acetylation/deacetylation processes. CLOCK
directly acetylates and the type III nicotinamide adenine
dinucleotide (NAD?)-dependent histone/protein deacetyl-
ase SIRT1 deacetylates BMAL1, counteracting the acety-
lating action exerted by CLOCK. SIRT1 links metabolism
and circadian rhythmicity gauging cellular redox and met-
abolic status: NAD?biosynthesis from tryptophan is cat-
alyzed by the nicotinamide phosphoribosyltransferase
(NAMPT/visfatin), whose expression is controlled by the
biological clock and drives circadian oscillations of NAD?,
which operates as a metabolic oscillator, and influencing
SIRT1 deacetylating activity regulates the circadian
synthesis of its own rate-limiting enzyme [11]. The clock
gene machinery controls basic cell processes, such as cell
cycle, proliferation, differentiation, DNA damage repair,
apoptosis, whose deregulation is involved in carcinogenesis.
A circadian pattern of expression characterizes approxi-
mately 5–20 % of the transcriptome, diverging greatly
among tissues and reaching 80 % in the liver, and the clock-
controlled transcription factors drive the expression of
tissue-specific output genes involved in the regulation of
transcriptional networks that direct tissue/organ functions in
liver, kidney, adipose tissue, immune and hematopoietic
system, cardio-vascular apparatus [10] (Fig. 1).
Circadian rhythms in the neuroendocrine regulation
of immune function
Circadian rhythmicity characterizes oscillations of neuro-
endocrine function and innate/adaptive immune response,
driven by neural and humoral outputs from the SCN, which
regulate hormone secretion and well-timed trafficking of
leukocytes [12], and self-sustained biological clocks tick-
ing in the immune effectors, which organize their tasks.
Several transcription factors encoded by clock-controlled
genes are engaged in the control of the innate immune
response [10], the molecular oscillators drive the produc-
tion of cytolytic factors in natural killer cells [13], and gate
the response timing to immune challenges such as bacterial
lipopolysaccharide stimulation in macrophages [14]. In
turn, immunological factors such as TNF-aand IL-1b
feedback on the circadian clock circuitry influencing the
cross talk between the circadian and the neuroendocrine-
immune system pathways [15]. Circadian variations have
been found in immune system variables, including daily
rhythms in circulating leukocytes and serum cytokines
concentration [16], and circadian control of the antigen-
specific immune response and T cell proliferation regula-
tory mechanisms has been put in evidence [13]. Circadian
T cell immune responses are timed by a self-sustained
oscillator, and the transcriptional processes are connected
to rhythmicity of cell processes driven by the molecular
clockwork through the nuclear factor jB (NF-jB) pathway
[16]. On the other hand, experiments performed in animals
with clock gene mutations have highlighted different
immune phenotypes and evidenced altered expression of
immune-related genes and immune cells function, with
reduced leukocyte levels and altered circadian rhythmicity
of variation in the peripheral blood [13,17,18]. Mouse
lymph nodes exhibit rhythmic clock gene expression, and T
cells from lymph nodes collected over 24 h show a circa-
dian variation in proliferation after stimulation via the T
cell receptor, which is dampened in mice with Clock gene
mutation [13]. In humans, the total number of lymphocytes
Clin Exp Med
123
in the peripheral blood changes rhythmically over 24 h
with a nocturnal zenith, and opposing rhythms characterize
the nycthemeral oscillations of specific lymphocyte sub-
sets, with effector cytotoxic function peaking during the
day and memory function peaking at night [13]. The cir-
cadian timing system drives immune system function also
controlling the oscillatory secretion of neuroendocrine
hormones with immunomodulatory action (cortisol peaking
in the early morning, melatonin, growth hormone, prolactin
and thyroid-stimulating hormone peaking at night), and
directing the sleep/wake cycle. Sleep at night shifts
monocyte cytokine production to type 1 proinflammatory
pattern inducing predominance of T helper (Th1)-mediated
adaptive response, respect to anti-inflammatory type 2
cytokines that stimulate Th2-mediated immunity, and type
1–type 2 cytokine equilibrium is modulated by cross-
inhibition and regulated by the neuroendocrine system
[13].
The dynamic process of Th cell differentiation is
specifically and progressively influenced by fine tuning
of transcriptional control and gene expression patterns
[19], and several transcription factors encoded by
clock-controlled genes are engaged in the control of the
adaptive immune response [10]. For instance, the tran-
scription factors DEC1 and DEC2 oscillate with circadian
rhythmicity in peripheral blood mononuclear cells [20],
playing a key role in Th cell function and fate: DEC1
influences T lymphocyte activation, whereas DEC2 is
expressed principally during Th2 differentiation [21]. Th2
responses are also regulated by the highly expressed tran-
scription factor E4BP4, which controls cytokine production
and effector function in Th2 cells [22].
The biological clock and the connection
between immunity and metabolism
A crucial role in the multidirectional communication
among the circadian clock circuitry, the metabolic path-
ways and the neuroendocrine-immune system is played by
the nuclear receptors, comprising a superfamily of ligand
activated proteins expressed with different rhythmicity in
many tissues, capable to bind a number of molecules, such
as metabolites and hormones, and to influence gene
Fig. 1 The molecular clockwork. The biological oscillator is oper-
ated by an intracellular mechanism composed of a set of interlocking
transcription–translation feedback loops in which the expression of
clock genes is suppressed periodically by their protein products and
complete one cycle in approximately 24 h. The period genes (Per1-3)
and cryptochrome genes (Cry1-2) represent the core of the negative
limb in the feedback loop and are activated by the transcription
factors CLOCK and BMAL1, which heterodimerize and bind to
E-box enhancer elements in their promoters. The Per and Cry mRNAs
then give rise to PER and CRY proteins that are phosphorylated (Ph)
by casein kinase (CK), form a repression complex that translocates
back into the nucleus, interact directly with CLOCK and BMAL1 and
hinder their transcriptional activity. Otherwise, PER and CRY are,
respectively, targeted by CK and AMP kinase (AMPK) for degrada-
tion. CLOCK and BMAL1 also activate the expression of REV-
ERBs, which activate E4BP4 transcription and negatively control the
rhythmic transcription of Bmal1 impeding RORs binding at the
response elements, the nuclear receptor PPAR, the transcription
factors DBP, HLF, TEF, DEC and the clock-controlled genes, which
drive cell, tissue and organ functions
Clin Exp Med
123
expression through the modification of their transcriptional
activity after ligand binding [23]. The core clock genes
drive expression of nuclear receptors for several molecules,
such as fatty acids, cholesterol, oxysterols, androgens,
estrogens, glucocorticoids [10], and the circadian protein
PER2 is able to bind rhythmically at the promoters of
nuclear receptors target genes, co-regulating their tran-
scription and coordinating the biological oscillator with
metabolite/sensing, hormone/binding and transcriptional
processes [23]. The clock gene machinery also drives daily
variations in levels and responsiveness of pattern recogni-
tion receptors for pathogen-associated molecules such as
toll-like receptor 9, whose expression is driven through
binding of the transcriptional heterodimer CLOCK/
BMAL1 on TLR9 gene promoter, inducing circadian
rhythmic functional changes of TLR9-dependent cytokine
response engaged in the pathophysiological mechanisms
involved in septic shock, influencing clinical severity and
prognostic outcome [24].
The core clock genes also drive a number of transcrip-
tion factors, such as IRF-1, STAT 1-4, GATA-3, NF-AT,
NF-jB and AP 1-2 [10], implicated in the control of
inflammatory gene expression and differentiation/function
of immune cells, through regulation of cytokine production
and recruitment of costimulatory molecules influencing
immune responses. The rapid reprogramming of tran-
scriptional circuits upon inflammatory cytokine signaling
drives the acute phase response, a manifestation of innate
immunity involved in tissue homeostasis and defence.
Elevated levels of acute phase proteins together with lipid
and bile acid sensing nuclear receptors impinge on meta-
bolic and inflammatory/anti-inflammatory pathways
involved in derangements of cholesterol metabolism and
lipoprotein properties, atherogenesis and metabolic disor-
ders [25]. Liver X receptor (LXR, binding oxysterols),
peroxisome proliferator-activated receptors (PPARs, bind-
ing fatty acids), liver receptor homolog-1 (LRH-1, binding
phosphatidylinositols) and farnesoid X receptor (FXR,
binding bile acids) operate transcriptional interference with
the signal-dependent activation of proinflammatory tran-
scription factors, such as NF-jB, STATs and AP1, inhib-
iting the activities of other classes of transcription factors
in a ligand-dependent manner, and repressing inflammatory
gene expression by a mechanism defined transrepression
[25]. This mechanism does not require DNA binding by the
nuclear receptor and results in the elimination from geno-
mic binding sites of the nuclear receptor–proinflammatory
transcription factor complexes [25]. Animal studies have
shown that the inflammatory response is influenced by
REV-ERBaand RORapresent in vascular smooth muscle
cells, where IL-6 and cyclooxygenase-2 expression
levels are up-regulated by REV-ERBa. Besides, in
primary human macrophages present in the vascular wall,
REV-ERBaexpression is transcriptionally activated by
LXR, which controls cholesterol homeostasis, inflamma-
tion and the immune response through activation of TLR4
expression. TLR4 in turn is negatively regulated by binding
of REV-ERBato a response element site in its promoter
overlapping with the LXR response element site [11].
Furthermore, the clock gene machinery and PPARs are
strictly interconnected: PER2 negatively regulates expres-
sion of PPARcand inhibits PPARcrecruitment to target
promoters, PPARcpositively regulates Bmal1 transcrip-
tion, a reciprocal positive regulation of expression between
BMAL1 and PPARahas been put in evidence, and
CLOCK directly controls the circadian expression of Ppara
[11]. Interestingly, PPARs influence T lymphocyte differ-
entiation, proliferation, function and cytokine secretion,
independently from ligand binding [26], and self-sustained
biological clocks have been evidenced in human peripheral
blood mononuclear cells, with circadian rhythmicity of
expression of the core clock genes [20]. On the other hand,
RORaand RORct are involved in the differentiation pro-
cess of Th17 cells, which intervene in immune responses
against extracellular bacteria and fungi and contribute to
the initiation of organ-specific autoimmune diseases [27].
Cholesterol sulfate and derivatives are potential ligands of
RORa, which has also been proposed as the nuclear
mediator of melatonin signalling, entailing the pineal
monoamine as a key regulator of the molecular switch
leading to the reciprocally restricted generation of Th17
and T regulatory cells, essential to maintain self-tolerance
[28]. RORais also important for the differentiation and
expansion of the nuocytes, cytokine-secreting cells that
share a developmental lineage with natural helper cells and
innate helper cells and are strongly involved in type 2
immune responses mediated diseases, such as asthma and
allergy [29]. Furthermore, RORais expressed by IgA(?)
memory B cells and is involved in the preservation of
subset integrity and function in class-specific memory B
cells [30]. Interestingly, chronodisruption, represented by
desynchronization with respect to the daily light/dark cycle
of humoral and behavioral rhythms, is accompanied by
severe alteration of melatonin and cortisol secretion pat-
terns, inadequate pancreatic insulin secretion and increased
post-prandial plasma glucose concentrations, as evidenced
by experiments of forced desynchronization and prolonged
sleep restriction in humans [4,31,32]. The differentiation
of Th17 cells and Foxp3(?) T regulatory cells is also
modulated by the transcription factor aryl hydrocarbon
receptor (AHR), which manages biochemical effects of
exogenous and endogenous toxic molecules [33]. AHR
displays circadian variation of expression levels, driving
time-related pharmacological and immunomodulatory
effects of AHR agonists, represented by environmental
pollutants and metabolic by products [11].
Clin Exp Med
123
The circadian cross talk between the homeostatic
processes
The circadian timing system, the metabolic network and
the neuroendocrine-immune system interact on a complex
level integrating mutually environmental cues and physi-
ological signals originated from body tissues. The nuclear
receptors provide reciprocal communications among sev-
eral biological phenomena through nutrient and hormone
sensing, recruitment of coactivator/corepressor complexes
and modulation of numerous transcriptional circuits. These
interconnections may influence the cross talk among cir-
cadian, metabolic and neuroendocrine-immune pathways
in different tissues and may modulate the well-timed
functioning of homeostatic processes [1,11,13].
The circadian clock circuitry and lipid metabolism
The circadian clock circuitry drives the metabolic path-
ways controlling chromatin remodelling and transcriptional
circuits in metabolically active tissues, such as liver,
muscle, adipose tissue, and clock gene mutations and/or
hystone modification disorders in animal models determine
different metabolic phenotypes. In fact, in mouse liver, it
has been evidenced that histone deacetylation is charac-
terized by circadian rhythmicity related to histone deace-
tylase 3 (HDAC3) recruitment to the genome and
colocalization with REV-ERBanear genes regulating lipid
metabolism, and when HDAC3 and REV-ERBs are not
present the 24-h periodicity of gene expression disappears,
causing remarkable consequences on hepatic lipid metab-
olism and liver steatosis [34,35]. Furthermore, considering
that the expression of several genes encoding enzymes
involved in lipid metabolism is driven by the clock gene
machinery, the dysregulation of the circadian clock cir-
cuitry and/or incoordination with feeding times cause
improper expression patterns with fatty acid flux pertur-
bation, lipotoxicity, non-alcoholic fatty liver disease,
obesity, metabolic syndrome, diabetes mellitus, cardio-
vascular disease and atherothrombosis [11].
In this respect, studies performed in both rodents and
humans have shown that the hepatic clock gene machinery
controls both bile acid and lipid biosynthesis, driving
the circadian variation of expression of genes encoding
among the others 3-hydroxy-3-methylglutaryl coenzyme A
reductase, fatty acid and acyl-CoA synthase, fatty acid
transport proteins, lipolytic enzymes, apolipoproteins,
lipoprotein receptors [11]. In particular, RORaand REV-
ERBacontrol plasma triglyceride turnover driving, with
positive and negative roles, respectively, the expression of
the gene encoding apolipoprotein C-III. Synthesis, catab-
olism and export of cholesterol are tightly regulated to
maintain steady levels of this indispensible, but potentially
harmful molecule, transcriptionally regulated by sterol
regulatory element-binding proteins (SREBPs) that induce
cholesterol biosynthesis and uptake when intracellular
levels fall, and LXR that promotes cholesterol export and
elimination when intracellular cholesterol is excessive [11].
REV-ERBadrives bile acid synthesis interacting with
SREBPs and controlling oscillations of their quantity and
activity. In turn, expression of Cyp7a1, encoding the rate-
limiting enzyme in bile acid synthesis, is driven positively
by LXR binding with oxysterols and negatively by FXR
binding with bile acids, oscillates with circadian rhyth-
micity, and participates in the feeding-related synchroni-
zation of peripheral clocks and metabolic oscillators via
nutritional and hormonal cues [11]. Adipose tissue pro-
duces adipokines such as leptin and adiponectin that in
humans are expressed with circadian rhythmicity, modulate
insulin sensitivity and regulate energy balance influencing
feeding/fasting behavior. Obesity is characterized by dis-
ruption of adipokines 24-h profiles, accompanied by an
important component of the inflammatory response, the
endoplasmic reticulum stress within adipose tissue. A
characteristic of obesity is represented by decreased SIRT1
expression in adipose tissue together with macrophage
recruitment. SIRT1 down-regulation induces histone
hyperacetylation and ectopic inflammatory gene expres-
sion, playing a key role in macrophage influx into adipose
tissue during overnutrition and high-fat diet in rodents and
humans, and fostering the progression to metabolic dys-
function [36,37]. The expanded visceral adipose tissue
releases free fatty acids, TNF-a, IL-6, CC chemokine
ligand 2, which are implicated in the development of
insulin resistance and atherosclerosis [11], and as stated
above immune mediators and cytokines feedback with a
negative action on core clock components [15]. Lipids,
cytokines and metabolic stress pathways trigger a low-
grade chronic inflammatory condition defined ‘‘metaflam-
mation’ that impinges on cell, tissue and organ function,
including adipocytes and macrophages, and leads to unfa-
vorable metabolic and inflammatory derangement influ-
encing lipoprotein properties, cholesterol metabolism and
atherogenesis [25].
The circadian clock circuitry and glucose metabolism
The circadian clock circuitry also controls glucose
metabolism by means of circadian genes and proteins and
through hormones (cortisol, melatonin) and autonomic
nerves from the SCN, which modulate nycthemeral chan-
ges in insulin sensitivity and insulin secretory capacity of
endocrine pancreas. A number of studies performed in
animal models have shown that insulin secretion is
Clin Exp Med
123
coordinated with the sleep/wake cycle by self-sustained
oscillators in the Langerhans islets bcells, and the clock
gene machinery drives the circadian rhythmicity of
expression changes of genes encoding enzymes and
transporters involved in glucose homeostasis [11]. CLOCK
controls hepatic glycogen synthesis, CRY 1 and CRY2
regulate hepatic gluconeogenesis, and clock gene muta-
tions settle on disparate metabolic phenotypes: liver-spe-
cific Bmal1 disruption causes hypoglycemia specifically
during the inactivity phase, and impaired glucose tolerance,
reduced insulin secretion, and defects in size and prolif-
eration of pancreatic islets worsening with age characterize
Clock
D19/D19
and pancreas-specific Bmal1
/
mutant mice
[11]. Besides, in humans, single nucleotide polymorphisms
within the genes encoding CRY2 and the MT
2
melatonin
receptor (MTNR1B) have been found capable to influence
fasting blood glucose concentrations in genome-wide
association studies [11].
The circadian timing system from health to disease
The breakdown of the array of rhythms or a change of their
functional interactions may lead to distortion of body
rhythmicity with tainted communication among circadian
and metabolic pathways, immune cell functions, cytokine/
chemokine production and neuroendocrine secretion
influencing metabolism and immunomodulation, fostering
illness-related or aging-related pathological processes.
Circadian rhythmicity characterizes fluctuations of
symptomatology in acute and chronic inflammatory dis-
eases, as exemplified by recrudescence in the early morn-
ing of joint pain, morning stiffness, and functional
disability in patients with rheumatoid arthritis. The daily
oscillation of symptom intensity is related to nycthemeral
variation of cytokine levels (in particular TNF-aand
IFN-c), endogenous l-opiatergic agonists (b-endorphin),
hormones (ACTH, cortisol, melatonin), neurotransmitters
and neuropeptides released by peripheral and autonomic
nervous system fibers, immunologic parameters (immuno-
globulins and immune-competent cells) [38,39].
Among the age-related diseases, Alzheimer’s dementia
is one of the most costly degenerative diseases in devel-
oped countries and is characterized by amyloid-b(Ab)
accumulation in the brain extracellular space, a process
related to level changes in brain interstitial fluid, neuronal
activity and the sleep-wake cycle, and preceding clinical
onset by several years. Animal studies have put in evidence
that sleep restriction significantly increases Abplaque
formation [40], and in turn, plaque formation alters the
sleep-wake cycle and diurnal fluctuation of Abin mice and
humans, suggesting that the sleep-wake cycle may play a
role in the pathogenesis of Alzheimer’s diseases [41].
Summary
Chronodisruption and desynchronization of the internal
clocks contribute to the alteration of the organismal time
structure causing functional disorders and modifications of
the anatomic integrity, as demonstrated by epidemiological
studies reporting an increased risk of metabolic, degenera-
tive, immune-related and neoplastic diseases in relationship
to protracted exposure to light at night or long-term rotating
night shifts/work schedules associated with sleep curtail-
ment and/or mistiming. The increasing interest in circadian
physiology and genetics opens new frontiers in the study of
disease mechanisms and compels accurate knowledge of
the fourth dimension of the biological processes in the
evaluation of new therapeutic approaches. As perspective
endeavor, it will be worth considering the alteration of the
temporal structure of body homeostasis among the patho-
physiological bases of disease, with the aim to (1) develop
pharmacological agents influencing the circadian timing
system and the molecular clockwork to recover disarray; (2)
redirect and reinforce the circadian clock circuitry by means
of natural entraining factors such as feeding time and light
exposure; (3) set up suitable chronomodulated drug delivery
systems to improve treatment of symptoms and therapy of
diseases related to chronodisruption.
Acknowledgments We apologize for not comment on all the rele-
vant studies and not citing all pertinent references due to space lim-
itations. This work was supported by ‘Italian Ministry of Health’’
grant RC1203ME46 through Department of Medical Sciences, Divi-
sion of Internal Medicine and Chronobiology Unit, IRCCS Scientific
Institute and Regional General Hospital ‘‘Casa Sollievo della Sof-
ferenza,’ Opera di Padre Pio da Pietrelcina, San Giovanni Rotondo
(FG), Italy.
Conflict of interest The authors declare that there is no conflict of
interest with respect to the authorship and/or publication of this
article.
References
1. Bass J (2012) Circadian topology of metabolism. Nature 491:
348–356
2. Gooley JJ, Rajaratnam SM, Brainard GC, Kronauer RE, Czeisler
CA, Lockley SW (2010) Spectral responses of the human circa-
dian system depend on the irradiance and duration of exposure to
light. Sci Transl Med 2(31):31ra33
3. Stevens RG, Hansen J, Costa G, Haus E, Kauppinen T, Aronson
KJ, Castan
˜o-Vinyals G, Davis S, Frings-Dresen MH, Fritschi L,
Kogevinas M, Kogi K, Lie JA, Lowden A, Peplonska B, Pesch B,
Pukkala E, Schernhammer E, Travis RC, Vermeulen R, Zheng T,
Cogliano V, Straif K (2011) Considerations of circadian impact
for defining ‘shift work’ in cancer studies: IARC working group
report. Occup Environ Med 68(2):154–162
4. Buxton OM, Cain SW, O’Connor SP, Porter JH, Duffy JF,
Wang W, Czeisler CA, Shea SA (2012) Adverse metabolic
Clin Exp Med
123
consequences in humans of prolonged sleep restriction combined
with circadian disruption. Sci Transl Med 4(129):129ra143
5. Li J, Lu WQ, Beesley S, Loudon AS, Meng QJ (2012) Lithium
impacts on the amplitude and period of the molecular circadian
clockwork. PLoS ONE 7(3):e33292
6. Solt LA, Wang Y, Banerjee S, Hughes T, Kojetin DJ, Lundasen
T, Shin Y, Liu J, Cameron MD, Noel R, Yoo SH, Takahashi JS,
Butler AA, Kamenecka TM, Burris TP (2012) Regulation of
circadian behaviour and metabolism by synthetic REV-ERB
agonists. Nature 485(7396):62–68
7. Koike N, Yoo SH, Huang HC, Kumar V, Lee C, Kim TK,
Takahashi JS (2012) Transcriptional architecture and chromatin
landscape of the core circadian clock in mammals. Science
338(6105):349–354
8. Cho H, Zhao X, Hatori M, Yu RT, Barish GD, Lam MT, Chong
LW, DiTacchio L, Atkins AR, Glass CK, Liddle C, Auwerx J,
Downes M, Panda S, Evans RM (2012) Regulation of circadian
behaviour and metabolism by REV-ERB-aand REV-ERB-b.
Nature 485(7396):123–127
9. Takeda Y, Jothi R, Birault V, Jetten AM (2012) RORcdirectly
regulates the circadian expression of clock genes and downstream
targets in vivo. Nucleic Acids Res 40(17):8519–8535
10. Bozek K, Relo
´gio A, Kielbasa SM, Heine M, Dame C, Kramer A,
Herzel H (2009) Regulation of clock-controlled genes in mam-
mals. PLoS ONE 4(3):e4882
11. Mazzoccoli G, Pazienza V, Vinciguerra M (2012) Clock genes
and clock-controlled genes in the regulation of metabolic
rhythms. Chronobiol Int 29(3):227–251
12. Scheiermann C, Kunisaki Y, Lucas D, Chow A, Jang JE, Zhang
D, Hashimoto D, Merad M, Frenette PS (2012) Adrenergic nerves
govern circadian leukocyte recruitment to tissues. Immunity
37(2):290–301
13. Cermakian N, Lange T, Golombek D, Sarkar D, Nakao A,
Shibata S, Mazzoccoli G (2013) Cross-talk between the circa-
dian clock circuitry and the immune system. Chronobiol Int
(in Press)
14. Keller M, Mazuch J, Abraham U, Eom GD, Herzog ED, Volk
HD, Kramer A, Maier B (2009) A circadian clock in macro-
phages controls inflammatory immune responses. Proc Natl Acad
Sci USA 106:21407–21412
15. Cavadini G, Petrzilka S, Kohler P, Jud C, Tobler I, Birchler T,
Fontana A (2007) TNF-alpha suppresses the expression of clock
genes by interfering with E-box-mediated transcription. Proc Natl
Acad Sci USA 104(31):12843–12848
16. Bollinger T, Leutz A, Leliavski A, Skrum L, Kovac J, Bonacina
L, Benedict C, Lange T, Westermann J, Oster H, Solbach W
(2011) Circadian clocks in mouse and human CD4 ?T cells.
PLoS ONE 6:e29801
17. Gibbs JE, Blaikley J, Beesley S, Matthews L, Simpson KD,
Boyce SH, Farrow SN, Else KJ, Singh D, Ray DW, Loudon AS
(2012) The nuclear receptor REV-ERBamediates circadian
regulation of innate immunity through selective regulation of
inflammatory cytokines. Proc Natl Acad Sci USA 109:582–587
18. Narasimamurthy R, Hatori M, Nayak SK, Liu F, Panda S, Verma
IM (2012) Circadian clock protein cryptochrome regulates the
expression of proinflammatory cytokines. Proc Natl Acad Sci
USA 109(31):12662–12667
19. Kaech SM, Cui W (2012) Transcriptional control of effector and
memory CD8(?) T cell differentiation. Nat Rev Immunol. doi:
10.1038/nri3307. [Epub ahead of print]
20. Ebisawa T, Numazawa K, Shimada H, Izutsu H, Sasaki T, Kato
N, Tokunaga K, Mori A, Honma K, Honma S, Shibata S (2010)
Self-sustained circadian rhythm in cultured human mononuclear
cells isolated from peripheral blood. Neurosci Res 66:223–227
21. Liu Z, Li Z, Mao K, Zou J, Wang Y, Tao Z, Lin G, Tian L, Ji Y,
Wu X, Zhu X, Sun S, Chen W, Xiang C, Sun B (2009) Dec2
promotes Th2 cell differentiation by enhancing IL-2R signaling.
J Immunol 183:6320–6329
22. Kashiwada M, Cassel SL, Colgan JD, Rothman PB (2011)
NFIL3/E4BP4 controls type 2 T helper cell cytokine expression.
EMBO J 30:2071–2082
23. Schmutz I, Ripperger JA, Baeriswyl-Aebischer S, Albrecht U
(2010) The mammalian clock component PERIOD2 coordinates
circadian output by interaction with nuclear receptors. Genes Dev
24:345–357
24. Silver AC, Arjona A, Walker WE, Fikrig E (2012) The circadian
clock controls toll-like receptor 9-mediated innate and adaptive
immunity. Immunity 36(2):251–261
25. Venteclef N, Jakobsson T, Steffensen KR, Treuter E (2011)
Metabolic nuclear receptor signaling and the inflammatory acute
phase response. Trends Endocrinol Metab 22:333–343
26. Klotz L, Burgdorf S, Dani I, Saijo K, Flossdorf J, Hucke S,
Alferink J, Nowak N, Beyer M, Mayer G, Langhans B, Klock-
gether T, Waisman A, Eberl G, Schultze J, Famulok M, Kolanus
W, Glass C, Kurts C, Knolle PA (2009) The nuclear receptor
PPAR gamma selectively inhibits Th17 differentiation in a T cell-
intrinsic fashion and suppresses CNS autoimmunity. J Exp Med
206:2079–2089
27. Rauen T, Juang YT, Hedrich CM, Kis-Toth K, Tsokos GC (2012)
A novel isoform of the orphan receptor RORct suppresses IL-17
production in human T cells. Genes Immun 13:346–350
28. Lardone PJ, Guerrero JM, Ferna
´ndez-Santos JM, Rubio A,
Martı
´n-Lacave I, Carrillo-Vico A (2011) Melatonin synthesized
by T lymphocytes as a ligand of the retinoic acid-related orphan
receptor. J Pineal Res 51:454–462
29. Wong SH, Walker JA, Jolin HE, Drynan LF, Hams E, Camelo A,
Barlow JL, Neill DR, Panova V, Koch U, Radtke F, Hardman CS,
Hwang YY, Fallon PG, McKenzie AN (2012) Transcription
factor RORais critical for nuocyte development. Nat Immunol
13:229–236
30. Wang NS, McHeyzer-Williams LJ, Okitsu SL, Burris TP, Reiner
SL, McHeyzer-Williams MG (2012) Divergent transcriptional
programming of class-specific B cell memory by T-bet and
RORa. Nat Immunol 13(6):604–611
31. Scheer FA, Hilton MF, Mantzoros CS, Shea SA (2009) Adverse
metabolic and cardiovascular consequences of circadian mis-
alignment. Proc Natl Acad Sci USA 106(11):4453–4458
32. Dijk DJ, Duffy JF, Silva EJ, Shanahan TL, Boivin DB, Czeisler
CA (2012) Amplitude reduction and phase shifts of melatonin,
cortisol and other circadian rhythms after a gradual advance of
sleep and light exposure in humans. PLoS ONE 7(2):e30037
33. Singh NP, Singh UP, Singh B, Price RL, Nagarkatti M,
Nagarkatti PS (2011) Activation of aryl hydrocarbon receptor
(AhR) leads to reciprocal epigenetic regulation of FoxP3 and
IL-17 expression and amelioration of experimental colitis. PLoS
ONE 6(8):e23522
34. Feng D, Liu T, Sun Z, Bugge A, Mullican SE, Alenghat T, Liu
XS, Lazar MA (2011) A circadian rhythm orchestrated by histone
deacetylase 3 controls hepatic lipid metabolism. Science
331(6022):1315–1319
35. Bugge A, Feng D, Everett LJ, Briggs ER, Mullican SE, Wang F,
Jager J, Lazar MA (2012) Rev-erbaand Rev-erbbcoordinately
protect the circadian clock and normal metabolic function. Genes
Dev 26(7):657–667
36. Gillum MP, Kotas ME, Erion DM, Kursawe R, Chatterjee P,
Nead KT, Muise ES, Hsiao JJ, Frederick DW, Yonemitsu S,
Banks AS, Qiang L, Bhanot S, Olefsky JM, Sears DD, Caprio S,
Shulman GI (2011) SirT1 regulates adipose tissue inflammation.
Diabetes 60(12):3235–3245
37. Chalkiadaki A, Guarente L (2012) High-fat diet triggers inflam-
mation-induced cleavage of SIRT1 in adipose tissue to promote
metabolic dysfunction. Cell Metab 16(2):180–188
Clin Exp Med
123
38. Straub RH, Cutolo M (2007) Circadian rhythms in rheumatoid
arthritis: implications for pathophysiology and therapeutic man-
agement. Arthritis Rheum 56(2):399–408
39. Pongratz G, Straub RH (2013) Role of peripheral nerve fibres in
acute and chronic inflammation in arthritis. Nat Rev Rheumatol
9(2):117–126
40. Kang JE, Lim MM, Bateman RJ, Lee JJ, Smyth LP, Cirrito JR,
Fujiki N, Nishino S, Holtzman DM (2009) Amyloid-beta
dynamics are regulated by orexin and the sleep-wake cycle.
Science 326(5955):1005–1007
41. Roh JH, Huang Y, Bero AW, Kasten T, Stewart FR, Bateman RJ,
Holtzman DM (2012) Disruption of the sleep-wake cycle and
diurnal fluctuation of b-amyloid in mice with Alzheimer’s disease
pathology. Sci Transl Med 4(150):150ra122
Clin Exp Med
123
... The recurrence of biological processes cycling at different frequency ranges enables healthy organisms to maintain homeostasis, that is the body's ability to regulate stability in its inner environment in response to changing conditions in the outside environment. Rhythmic processes hallmarked by the same frequency may have the same phase or different phases, and usually show well-defined time-relationships to each other: functional disturbances and alteration of anatomic integrity may be caused by loss of this multifrequency time structure leading to internal desynchronization [2]. ...
... Tissue functions and cellular processes in living beings show 24-h rhythms regulated by the biological clocks, present in every tissue and cell, and ticking through a molecular clockwork operated by a transcriptional/translational feedback loop hard-wired by core circadian genes and proteins (ARNTL-1/2, CLOCK/NPAS2, CRY1-2, PER1-3, NR1D1-2, RORA) driving the circadian oscillation of thousands of clock-controlled genes [2]. Circadian rhythmicity of the body is driven by environmental lighting conditions via a neural pathway including the retina and a specific retinohypothalamic tract projecting to the biological clocks of the suprachiasmatic nuclei, which in turn drive adrenal and pineal gland secretion of cortisol in the early morning and melatonin at nighttime, respectively [2]. ...
... Tissue functions and cellular processes in living beings show 24-h rhythms regulated by the biological clocks, present in every tissue and cell, and ticking through a molecular clockwork operated by a transcriptional/translational feedback loop hard-wired by core circadian genes and proteins (ARNTL-1/2, CLOCK/NPAS2, CRY1-2, PER1-3, NR1D1-2, RORA) driving the circadian oscillation of thousands of clock-controlled genes [2]. Circadian rhythmicity of the body is driven by environmental lighting conditions via a neural pathway including the retina and a specific retinohypothalamic tract projecting to the biological clocks of the suprachiasmatic nuclei, which in turn drive adrenal and pineal gland secretion of cortisol in the early morning and melatonin at nighttime, respectively [2]. The pineal gland modulates hypothalamus-pituitary-adrenal axis function, melatonin modulates the transcriptional activity of glucocorticoid receptors, and in turn adrenal corticosteroids modulate pineal melatonin synthesis [7]. ...
... The harmonized regulation of sleep/ wake, rest/activity, fasting/feeding cycles with biochemical processes and metabolic pathways is crucial to maintain body homeostasis and preserve health. Coordinating biological rhythms and anticipated changes of spatio/ temporal niches impacts organism and species survival of free-living animals, influencing behavioral cycles of feeding, predation, competition, mating and providing fitness advantage as opposed to natural selection pressure [11][12][13]. Loss of resonance of body rhythmicity with external synchronizers or a change of oscillation phase may alter the physiological array of rhythms with onset of internal desynchronization, also known as chronodisruption, which promotes neoplastic, metabolic, inflammatory and neurodegenerative diseases [11][12][13]. Daily timekeeping is driven in mammals and humans by the circadian timing system, comprising a central pace-maker and master oscillators located in the suprachiasmatic nuclei (SCN) of the hypothalamus and autonomous self-sustained oscillators in every cell of peripheral tissues. ...
... Coordinating biological rhythms and anticipated changes of spatio/ temporal niches impacts organism and species survival of free-living animals, influencing behavioral cycles of feeding, predation, competition, mating and providing fitness advantage as opposed to natural selection pressure [11][12][13]. Loss of resonance of body rhythmicity with external synchronizers or a change of oscillation phase may alter the physiological array of rhythms with onset of internal desynchronization, also known as chronodisruption, which promotes neoplastic, metabolic, inflammatory and neurodegenerative diseases [11][12][13]. Daily timekeeping is driven in mammals and humans by the circadian timing system, comprising a central pace-maker and master oscillators located in the suprachiasmatic nuclei (SCN) of the hypothalamus and autonomous self-sustained oscillators in every cell of peripheral tissues. Ambient natural or artificial photic cues are perceived by non-image forming intrinsically photosensitive retinal ganglion cells containing melanopsin, whose output is transferred to the SCN by the retino-hypothalamic tract [14][15][16]. ...
Article
Full-text available
Several different signaling pathways that regulate cell proliferation and differentiation are initiated by binding of ligands to cell-surface and membrane-bound enzyme-linked receptors, such as receptor tyrosine kinases and serine-threonine kinases. They prompt phosphorylation of tyrosine and serine-threonine residues and initiate downstream signaling pathways and priming of intracellular molecules that convey the signal in the cytoplasm and nucleus, with transcriptional activation of specific genes enriching cell growth and survival-related cascades. These cell processes are rhythmically driven by molecular clockworks endowed in every cell type and when deregulated play a crucial role in cancer onset and progression. Growth factors and their matching receptor-dependent signaling are frequently overexpressed and/or dysregulated in many cancer types. In this review we focus on the interplay between biological clocks and Growth Factor Receptor-dependent signaling in the context of carcinogenesis.
... Circadian rhythmicity characterizes the function of body systems that allow homeostasis maintenance, and among these, the metabolic pathways are preeminent. Derangement of customary circadian rhythmicity underlies pathophysiological mechanisms in several metabolic [14][15][16][17][18][19][20][21][22]. Circadian rhythms are endogenous cycles driven by molecular clockworks ticking in mammals through transcriptional/translational feedback loops operated by a positive limb, working through the transcriptional activators ARNTL/ARTL2 and CLOCK (or NPAS2) which are complex and activate a negative limb, with Period (PER1, PER2, PER3) and Cryptochrome (CRY1, CRY2) proteins that in turn inhibit the transactivation complex and close the loop [23,24]. ...
Article
Full-text available
Heme is a member of the porphyrins family of cyclic tetrapyrroles and influences various cell processes and signalling pathways. Enzyme deficiencies in the heme biosynthetic pathway provoke rare human inherited metabolic diseases called porphyrias. Protein levels and activity of enzymes involved in the heme biosynthetic pathway and especially 5′-Aminolevulinate Synthase 1 are featured by 24-h rhythmic oscillations driven by the biological clock. Heme biosynthesis and circadian pathways intermingle with mutual modulatory roles. Notably, heme is a ligand of important cogs of the molecular clockwork, which upon heme binding recruit co-repressors and inhibit the transcription of numerous genes enriching metabolic pathways and encoding functional proteins bringing on crucial cell processes. Herein, we assessed mRNA levels of circadian genes in patients suffering from porphyrias and found several modifications of core clock genes and clock-controlled genes expression, associated with metabolic and electrolytic changes. Overall, our results show an altered expression of circadian genes accompanying heme biosynthesis disorders and confirm the need to deepen the knowledge of the mechanisms through which the alteration of the circadian clock circuitry could take part in determining signs and symptoms of porphyria patients and then again could represent a target for innovative therapeutic strategies.
... In summary, VLCADD, as an inborn error of fatty acid oxidation, illustrates the biological theme about the adverse effects of alteration of metabolic pathways in health and disease. 68,69 In addition to direct mitochondrial dysfunction, the negative effects of VLCADD on cell/organ function including its direct link to the clinical manifestation of rhabdomyolysis are well characterised. [70][71][72] The present data support the idea that inflammation, both humoral and cellular, is a novel physiologic dimension of VLCADD. ...
Article
Full-text available
Objectives Very-long-chain acyl-CoA dehydrogenase deficiency (VLCADD) is a disorder of fatty acid oxidation. Symptoms are managed by dietary supplementation with medium-chain fatty acids that bypass the metabolic block. However, patients remain vulnerable to hospitalisations because of rhabdomyolysis, suggesting pathologic processes other than energy deficit. Since rhabdomyolysis is a self-destructive process that can signal inflammatory/immune cascades, we tested the hypothesis that inflammation is a physiologic dimension of VLCADD. Methods All subjects (n = 18) underwent informed consent/assent. Plasma cytokine and cytometry analyses were performed. A prospective case analysis was carried out on a patient with recurrent hospitalisation. Health data were extracted from patient medical records. Results Patients showed systemic upregulation of nine inflammatory mediators during symptomatic and asymptomatic periods. There was also overall abundance of immune cells with high intracellular expression of IFNγ, IL-6, MIP-1β (CCL4) and TNFα, and the transcription factors p65-NFκB and STAT1 linked to inflammatory pathways. A case analysis of a patient exhibited already elevated plasma cytokine levels during diagnosis in early infancy, evolving into sustained high systemic levels during recurrent rhabdomyolysis-related hospitalisations. There were corresponding activated leukocytes, with higher intracellular stores of inflammatory molecules in monocytes compared to T cells. Exposure of monocytes to long-chain free fatty acids recapitulated the cytokine signature of patients. Conclusion Pervasive plasma cytokine upregulation and pre-activated immune cells indicate chronic inflammatory state in VLCADD. Thus, there is rationale for practical implementation of clinical assessment of inflammation and/or translational testing, or adoption, of anti-inflammatory intervention(s) for personalised disease management.
... Since decades, it has been shown that light-dark alternation influences the synchronization of circadian rhythms of most human systems [1]. Cellular processes show 24-h rhythms regulated by the biological clocks, which are ticking through a molecular clockwork, operated by a complex transcriptional/translational feed-back loop, hard-wired by core circadian genes and proteins [2,3]. The regulation of this complex machinery is extremely delicate, so that changing the time can disrupts body clocks and cause pathophysiological consequences far beyond the regulation of sleep [4]. ...
Article
Full-text available
Background: The available evidence on the effects of daylight saving time (DST) transitions on major cardiovascular diseases is limited and conflicting. We carried out the first meta-analysis aimed at evaluating the risk of acute myocardial infarction (AMI) following DST transitions. Methods: We searched cohort or case-control studies evaluating the incidence of AMI, among adults (≥18 y), during the weeks following spring and/or autumn DST shifts, versus control periods. The search was made in MedLine and Scopus, up to 31 December 2018, with no language restriction. A summary odds ratio of AMI was computed after: (1) spring, (2) autumn or (3) both transitions considered together. Meta-analyses were also stratified by gender and age. Data were combined using a generic inverse-variance approach. Results: Seven studies (>115,000 subjects) were included in the analyses. A significantly higher risk of AMI (Odds Ratio: 1.03; 95% CI: 1.01⁻1.06) was observed during the two weeks following spring or autumn DST transitions. However, although AMI risk increased significantly after the spring shift (OR: 1.05; 1.02⁻1.07), the incidence of AMI during the week after winter DST transition was comparable with control periods (OR 1.01; 0.98⁻1.04). No substantial differences were observed when the analyses were stratified by age or gender. Conclusion: The risk of AMI increases modestly but significantly after DST transitions, supporting the proposal of DST shifts discontinuation. Additional studies that fully adjust for potential confounders are required to confirm the present findings.
... Since decades, it has been shown that light-dark alternation influences the synchronization of circadian rhythms of most human systems [1]. Cellular processes show 24-h rhythms regulated by the biological clocks, which are ticking through a molecular clockwork, operated by a complex transcriptional/translational feed-back loop, hard-wired by core circadian genes and proteins [2,3]. The regulation of this complex machinery is extremely delicate, so that changing the time can disrupts body clocks and cause pathophysiological consequences far beyond the regulation of sleep [4]. ...
Article
Background The current evidence on the effects of daylight saving time (DST) transitions on major cardiovascular diseases is limited, and available results are conflicting. We carried out the first meta-analysis aimed at evaluating the risk of acute myocardial infarction (AMI) following DST transitions. Methods We searched MedLine and Scopus up to December 31, 2018, with no language restriction, to retrieve cohort or case-control studies evaluating AMI incidence among adults (≥18y) in the week following spring and/or autumn DST shifts versus control periods. A summary relative risk of AMI was computed after: (1) spring, (2) autumn, (3) both transitions considered together versus control weeks. Stratified analyses were performed by gender and age. Data were combined using a generic inverse-variance approach. Results Seven studies (>115,000 subjects) were included in the analyses. A significantly higher risk of AMI (Odds Ratio: 1.03; 95% CI: 1.01-1.06) was observed in the two weeks following spring or winter DST transitions. The risk increase was however significant only after the spring shift (OR: 1.05; 1.02-1.07), while AMI incidence in the week after winter DST transition was comparable to control periods (OR 1.01; 0.98-1.04). No substantial differences by age or gender emerged. Conclusions The risk of AMI increases modestly but significantly following DST transitions, supporting the proposal of DST shifts discontinuation. Additional studies fully adjusting for potential confounders are required to confirm the present findings. Key messages The risk of acute myocardial infarction increases modestly but significantly following DST transitions. Although preliminary, our findings support the proposal of DST shifts discontinuation.
... In mice, NOCTURNIN is the sole deadenylase affecting the high-amplitude rhythms and shows nocturnal peaks, whereas most other deadenylases are arrhythmic or have very low-amplitude rhythms that peak during the day [2]. Several metabolic pathways are related to the circadian clock [4]. The NOCTURNIN gene regulates the circadian cycles and binds them to the metabolic rhythms; its involvement in lipid metabolism, adipogenesis, glycaemic homeostasis, and inflammation entails that it plays a role in promoting metabolic alterations [5][6][7]. ...
Article
Full-text available
Objective: The NOCTURNIN gene links nutrient absorption and metabolism to the circadian clock. Shift workers are at a heightened risk of overweight and of developing obesity and metabolic syndrome. This study investigates the diurnal variation of NOCTURNIN in healthy volunteers and its expression levels in rotational shift and daytime workers. Methods: NOCTURNIN expression levels were evaluated in peripheral blood lymphocytes from 15 healthy volunteers at 4-hour intervals for 24 h. Metabolic parameters and NOCTURNIN expression were measured in workers engaged in shift and daytime work. Results: In the group of volunteers NOCTURNIN expression showed diurnal variation, with a peak at 8:00 AM. NOCTURNIN expression was higher in shift workers than in daytime workers. Multivariate analysis confirmed the role of shift work as an independent factor affecting NOCTURNIN expression. Notably, its level correlated directly with body mass index and inversely with total energy expenditure. Conclusions: Measuring NOCTURNIN expression levels in human peripheral blood lymphocytes can improve investigations on the relationship between changes in circadian rhythm and metabolic disorders. Shift workers show higher NOCTURNIN levels than daytime workers.
... In our population, participants who skipped lunch show higher diastolic pressure compared to non-skippers. The synchronization between blood pressure and other physiological processes with the external M A N U S C R I P T A C C E P T E D ACCEPTED MANUSCRIPT 13 environment is driven by the circadian clocks pacing self-sustained and cell-autonomous molecular oscillators in peripheral tissues [18][19][20][21][22]. Breakfast skipping may adversely affect the circadian clock and correlates with increased postprandial glycemic response [23]. ...
Article
Full-text available
Background and Aims The frequency and timing of meals may affect cardiovascular health (CVH) outcomes, but large-scale epidemiological studies are lacking. To understand the relationship between eating time interval and frequency, and measures of ideal CVH in the Kardiovize Brno cohort study, a random urban sample population in Central Europe. Methods and Results 1659 members of the Kardiovize Brno 2030 cohort were included in a cross-sectional study (mean age=46.86 years; 44.6% male). Exposure variables were eating time interval and frequency, and skipping meals. Primary outcomes were indices of CVH, including body mass index, diet, physical activity, smoking, blood pressure, glucose and cholesterol, and the composite CVH score. Cluster analysis and binary logistic regression analysis were used to evaluate eating habits and the association between variables. After adjustment for well-known risk factors, subjects who skipped breakfast or the afternoon snack had a higher risk of poor CVH (OR=1.613; 95%CI= 1.121-2.320; p=0.010; OR=1.409; 95%CI=1.110-1.788; p=0.005, respectively). Moreover, we identified three clusters of individuals based on eating habits; from cluster 1 to cluster 3, eating time interval and frequency increased and this was associated with increases in CVH score from 8.70 (SEM=0.10) in cluster 1, and 9.06 (SEM=0.08) in cluster 2 to 9.42 (SEM=0.09) in cluster 3 (p-trend=0.019). Conclusions Our findings suggest that skipping breakfast or the afternoon snack are risk factors for poor CVH, while higher eating time interval and frequency may promote ideal CVH.
... Besides, gut microbiota impact the biological clock 113 ; appropriate timing of circadian patterns of hormone secretion, metabolism, bile acid turnover, autophagy, and inflammation with behavioral cycles is necessary to avoid hepatic dysfunction and metabolic disorders. [114][115][116][117][118][119][120][121][122][123] Furthermore, experiments performed in aryl hydrocarbon receptor (AHR, a xenobiotic receptor for exogenous toxicants) liver-specific and inducible transgenic mice fed an obesogenic diet showed that AHR signaling pathway activation worsens liver steatosis and in contradiction avoids obesity and systemic insulin resistance interplaying with the biological clock. 124,125 Interestingly, fibroblast growth factor 21, which interacting with b-Klotho coordinates a change to oxidative metabolism during fasting and starvation and has been involved as a mediator joining nutrition, growth, reproduction, and longevity, 126 was recognized as a direct AHR target in the liver and as the inducer of the systemic metabolic benefits in addition to liver steatosis observed in AHR transgenic mice. ...
Article
The molecular clockwork drives rhythmic oscillations of signaling pathways managing intermediate metabolism; the circadian timing system synchronizes behavioral cycles and anabolic/catabolic processes with environmental cues, mainly represented by light/darkness alternation. Metabolic pathways, bile acid synthesis, and autophagic and immune/inflammatory processes are driven by the biological clock. Proper timing of hormone secretion, metabolism, bile acid turnover, autophagy, and inflammation with behavioral cycles is necessary to avoid dysmetabolism. Disruption of the biological clock and mistiming of body rhythmicity with respect to environmental cues provoke loss of internal synchronization and metabolic derangements, causing liver steatosis, obesity, metabolic syndrome, and diabetes mellitus.
Article
Full-text available
Cardiovascular disease (CVD), which is the leading cause of death worldwide, is strongly affected by diet. Diet can affect CVD directly by modulating the composition of vascular plaques, and indirectly by affecting the rate of aging. This review summarizes research on the relationships of fasting, meal timing, and meal frequency with CVD incidence and progression. Relevant basic research studies, epidemiological studies, and clinical studies are highlighted. In particular, we discuss both intermittent and periodic fasting interventions with the potential to prevent and treat CVD.
Article
Full-text available
Synchronizing rhythms of behaviour and metabolic processes is important for cardiovascular health and preventing metabolic diseases. The nuclear receptors REV-ERB-α and REV-ERB-β have an integral role in regulating the expression of core clock proteins driving rhythms in activity and metabolism. Here we describe the identification of potent synthetic REV-ERB agonists with in vivo activity. Administration of synthetic REV-ERB ligands alters circadian behaviour and the circadian pattern of core clock gene expression in the hypothalami of mice. The circadian pattern of expression of an array of metabolic genes in the liver, skeletal muscle and adipose tissue was also altered, resulting in increased energy expenditure. Treatment of diet-induced obese mice with a REV-ERB agonist decreased obesity by reducing fat mass and markedly improving dyslipidaemia and hyperglycaemia. These results indicate that synthetic REV-ERB ligands that pharmacologically target the circadian rhythm may be beneficial in the treatment of sleep disorders as well as metabolic diseases.
Article
Full-text available
Various features, components, and functions of the immune system present daily variations. Immunocompetent cell counts and cytokine levels present variations according to the time of day and the sleep-wake cycle. Moreover, different immune cell types, such as macrophages, natural killer cells, and lymphocytes, contain a circadian molecular clockwork. The biological clocks intrinsic to immune cells and lymphoid organs, together with inputs from the central pacemaker of the suprachiasmatic nuclei via humoral and neural pathways, regulate the function of cells of the immune system, including their response to signals and their effector functions. Consequences of this include, for example, the daily variation in the response to an immune challenge (e.g., bacterial endotoxin injection) and the circadian control of allergic reactions. The circadian-immune connection is bidirectional, because in addition to this circadian control of immune functions, immune challenges and immune mediators (e.g., cytokines) were shown to have strong effects on circadian rhythms at the molecular, cellular, and behavioral levels. This tight crosstalk between the circadian and immune systems has wide-ranging implications for disease, as shown by the higher incidence of cancer and the exacerbation of autoimmune symptoms upon circadian disruption. (Author correspondence: g.mazzoccoli@operapadrepio.it).
Article
Full-text available
Biological clocks are genetically encoded oscillators that allow organisms to anticipate changes in the light-dark environment that are tied to the rotation of Earth. Clocks enhance fitness and growth in prokaryotes, and they are expressed throughout the central nervous system and peripheral tissues of multicelled organisms in which they influence sleep, arousal, feeding and metabolism. Biological clocks capture the imagination because of their tie to geophysical time, and tools are now in hand to analyse their function in health and disease at the cellular and molecular level.
Article
Full-text available
Aggregation of β-amyloid (Aβ) in the brain begins to occur years before the clinical onset of Alzheimer's disease (AD). Before Aβ aggregation, concentrations of extracellular soluble Aβ in the interstitial fluid (ISF) space of the brain, which are regulated by neuronal activity and the sleep-wake cycle, correlate with the amount of Aβ deposition in the brain seen later. The amount and quality of sleep decline with normal aging and to a greater extent in AD patients. How sleep quality as well as the diurnal fluctuation in Aβ change with age and Aβ aggregation is not well understood. We report a normal sleep-wake cycle and diurnal fluctuation in ISF Aβ in the brain of the APPswe/PS1δE9 mouse model of AD before Aβ plaque formation. After plaque formation, the sleep-wake cycle markedly deteriorated and diurnal fluctuation of ISF Aβ dissipated. As in mice, diurnal fluctuation of cerebrospinal fluid Aβ in young adult humans with presenilin mutations was also markedly attenuated after Aβ plaque formation. Virtual elimination of Aβ deposits in the mouse brain by active immunization with Aβ(42) normalized the sleep-wake cycle and the diurnal fluctuation of ISF Aβ. These data suggest that Aβ aggregation disrupts the sleep-wake cycle and diurnal fluctuation of Aβ. Sleep-wake behavior and diurnal fluctuation of Aβ in the central nervous system may be functional and biochemical indicators, respectively, of Aβ-associated pathology.
Article
Full-text available
The mammalian circadian clock involves a transcriptional feed back loop in which CLOCK and BMAL1 activate the Period and Cryptochrome genes, which then feedback and repress their own transcription. We have interrogated the transcriptional architecture of the circadian transcriptional regulatory loop on a genome scale in mouse liver and find a stereotyped, time-dependent pattern of transcription factor binding, RNA polymerase II (RNAPII) recruitment, RNA expression, and chromatin states. We find that the circadian transcriptional cycle of the clock consists of three distinct phases: a poised state, a coordinated de novo transcriptional activation state, and a repressed state. Only 22% of messenger RNA (mRNA) cycling genes are driven by de novo transcription, suggesting that both transcriptional and posttranscriptional mechanisms underlie the mammalian circadian clock. We also find that circadian modulation of RNAPII recruitment and chromatin remodeling occurs on a genome-wide scale far greater than that seen previously by gene expression profiling.
Article
The peripheral nervous system takes an active part in inflammatory processes by regulating effector cell function and reallocation of energy to the immune system. During acute inflammation, rapid neuronal reorganization and change of activity takes place. The hallmarks of this process are an increase in systemic sympathetic activity, a decrease in systemic parasympathetic activity and loss of sympathetic nerve fibres from sites of inflammation concomitant with increased innervation with sensory nerve fibres and increased sensory nerve fibre activity. On a systemic level, the increase in sympathetic activity (and decrease in parasympathetic activity) is necessary to provide enough energy to nourish the activated immune system. In locally inflamed tissue, the decrease in sympathetic nerve fibre density results in reduced anti-inflammatory signalling and, together with neuropeptides released from sensory nerve fibres, promotes local inflammation. In acute inflammation, this 'inflammatory configuration' of the peripheral nervous system favours the rapid clearance of antigenic threats. However, in chronic autoimmune inflammation, these changes of the peripheral nervous system lead to an unfavourable situation with ongoing energy reallocation and continuous local destruction. As an example of a chronic inflammatory condition, we discuss evidence for neuroimmune regulation in autoimmune arthritis with a focus on the sympathetic nervous system.
Article
During an infection, T cells can differentiate into multiple types of effector and memory T cells, which help to mediate pathogen clearance and provide long-term protective immunity. These cells can vary in their phenotype, function and location, and in their long-term fate in terms of their ability to populate the memory T cell pool. Over the past decade, the signalling pathways and transcriptional programmes that regulate the formation of heterogeneous populations of effector and memory CD8(+) T cells have started to be characterized, and this Review discusses the major advances in these areas.
Article
Adipose tissue plays an important role in storing excess nutrients and preventing ectopic lipid accumulation in other organs. Obesity leads to excess lipid storage in adipocytes, resulting in the generation of stress signals and the derangement of metabolic functions. SIRT1 is an important regulatory sensor of nutrient availability in many metabolic tissues. Here we report that SIRT1 functions in adipose tissue to protect from inflammation and obesity under normal feeding conditions, and to forestall the progression to metabolic dysfunction under dietary stress and aging. Genetic ablation of SIRT1 in adipose tissue leads to gene expression changes that highly overlap with changes induced by high-fat diet in wild-type mice, suggesting that dietary stress signals inhibit the activity of SIRT1. Indeed, we show that high-fat diet induces the cleavage of SIRT1 protein in adipose tissue by the inflammation-activated caspase-1, providing a link between dietary stress and predisposition to metabolic dysfunction.