Biological Role of PER2 & Gene Editing Prospects
The PER2 gene plays an extremely important role in the body’s internal clock by helping regulate daily rhythms like sleep and consciousness. It is an extremely important part of the feedback loop that drives the circadian cycle, especially in areas like the suprachiasmatic nucleus (SCN), which controls our biological clock’s timing. This review looks into whether the current gene editing technologies like CRISPR-Cas9 could be used to address problems that exist with PER2 expression. We assess how PER2 dysfunction is linked to circadian rhythm disorders such as Delayed Sleep Phase Syndrome (DSPS) and review the findings from case studies and research that cover both humans and animals. Additionally, the paper discusses the technical and biological challenges of editing a gene that changes over a 24 hour cycle and is only active in certain brain regions. We also look at the ethical and safety concerns that are associated with targeting genes in the brain, especially ones correlated to behavior and sleep. This understanding of the PER2 gene’s influence helps us explore the potential of safe and targeted gene therapy guidelines implemented in the near future, especially with such a steadily growing market.
STEM RESEARCHMEDICINEGENE EDITING
Akshiti Guandala and Aviral Srivastava
7/13/202526 min read
Abstract
The PER2 gene plays an extremely important role in the body's internal clock by helping
regulate daily rhythms like sleep and consciousness. It is an extremely important part of the
feedback loop that drives the circadian cycle, especially in areas like the suprachiasmatic nucleus
(SCN), which controls our biological clock's timing. This review looks into whether the current
gene editing technologies like CRISPR-Cas9 could be used to address problems that exist with
PER2 expression. We assess how PER2 dysfunction is linked to circadian rhythm disorders such
as Delayed Sleep Phase Syndrome (DSPS) and review the findings from case studies and
research that cover both humans and animals. Additionally, the paper discusses the technical and
biological challenges of editing a gene that changes over a 24-hour cycle and is only active in
certain brain regions. We also look at the ethical and safety concerns that are associated with
targeting genes in the brain, especially ones correlated to behavior and sleep. This understanding
of the PER2 gene's influence helps us explore the potential of safe and targeted gene therapy
guidelines implemented in the near future, especially with such a steadily growing market.
Introduction
Our bodies run on an internal clock called the circadian rhythm, which repeats roughly every 24
hours. This clock helps regulate a wide range of functions like sleep, body temperature,
digestion, and hormone release. It keeps us aligned with the day-night cycle and plays a big role
in keeping us healthy and alert. When the circadian rhythm is off, even slightly, it can lead to
sleep problems and other health issues.
One of the key genes involved in keeping this rhythm stable is PER2. It's essentially part of a
group informally named “clock genes” that turn on and off in a regular cycle to keep a record of
time inside our cells. PER2 is especially active in a part of the brain called the suprachiasmatic
nucleus (SCN), which is basically the body's master clock. When PER2 isn't working correctly,
whether due to a genetic mutation or changes in its gene expression, it can throw off the timing
of the rest of the circadian rhythm. This malfunctioning can lead to disorders like Delayed Sleep
Phase Syndrome (DSPS), where people naturally fall asleep and wake up much later than usual,
shifting their sleep cycle forward.
With new advances in gene editing tools like CRISPR, scientists are beginning to explore the
possibility of using these technologies to completely fix or perform minor adjustments on genes
like PER2. The idea, in essence, is that by editing the gene, there is a possibility of being able to
“reset” a person's internal clock at the source and treat their sleep disorder more effectively than
with traditional medications or therapies. Whether it is through single-point mutations or
correcting over- or underexpression, gene editing must be precise using safe delivery methods to
maintain its target and disrupt the circadian feedback loop. However, editing a gene that controls
such a complex and time-sensitive system isn't simple. There are major challenges, like making
sure the editing happens at the right moment, in the right location of the body, and ensuring it
does not result in any unwanted side effects.
In this paper, we will take a closer look at how PER2 works, how its disruption can affect the
sleep process, and whether gene editing has the potential to become a useful way to treat any
existing circadian rhythm disorders. We will also cover some of the biggest challenges and
ethical concerns that are present when it comes to editing genes that control how our bodies
function daily.
Biological Role of PER2
The PER2 protein also acts as a central regulator of the transcription-translation feedback loop
(TTFL) that contributes to setting the mammalian circadian clock. CLOCK–BMAL1
heterodimers, which are a family of protein transcription factors, bind to E-box elements to
activate transcription of the PER and CRY genes (such as PER2) and start the circadian feedback
loop. The PER2 protein translated from this then binds to CRY proteins (initiated in turn by
CLOCK–BMAL1) that are exported into the nucleus and repress CLOCK–BMAL1, closing the
negative feedback loop required to maintain ~24‐hour cycles in the suprachiasmatic nucleus
(SCN) and peripheral tissues (Etchegaray et al. 2009). This transcriptional feedback is controlled
at a number of levels of regulation for precision and circadian rhythm consistency day to day.
Figure 1. Circadian Rhythms are controlled by a phosphorylation-regulated negative feedback
loop. Early in the circadian cycle, PER, CKI, and CRY proteins multimerize in the cytoplasm and
then translocate to the nucleus to repress the CLK:BMAL1 transcription factor. Potential
functional effects of CKIδ and CKIε (denoted CKIε for simplicity) include (1) degradation of
PER early in the accumulation phase, delaying repression; (2) regulating PER nuclear entry of
the inhibitor complex, or (3) promoting degradation of PER, thereby terminating repression. The
stabilizing Rev-Erbα loop is not shown here. (Virshup et al., 2007)
One of the significant regulatory layers is at the translational level by means of a conserved
upstream open reading frame (uORF) in the 5' UTR region of the Per2 gene. In conditions of
homeostasis, this uORF suppresses the translation of PER2. Physiologically significant
temperature rise (e.g. 35 - 38 °C), however, operates to relieve such suppression and augment
PER2 protein synthesis without altering the amount of mRNA and thereby training such cell
cycles to comply with their optimal temperature environment (Miyake et al. 2023). This action
adds a non-photic activation pathway, allowing organisms to synchronize with daily thermal
fluctuation stimulation independently of transcriptional change at the gene itself.
Further regulation is achieved post-translationally through phosphorylation by casein kinase 1δ/ε
(CK1δ/ε). PER2 undergoes hierarchical multisite phosphorylation, a process in which phosphate
groups are deposited sequentially onto multiple sites on the protein. Phosphorylation generates a
phosphoswitch that regulates PER2 stability. The equilibrium between a stabilizing FASP
domain and a phospho-degron mediating PER2 degradation is kept in the switch. The
degradation of PER2 is therefore well-regulated and plays a role in circadian periodicity
regulation (Narasimamurthy and Virshup 2018; Hildebrand et al. 2021). Stabilization and
reduced degradation of PER2 are mediated by phosphorylation at the FASP site, whereas
phosphorylation of the degron increases ubiquitination and proteasomal elimination via β-TrCP
(Masuda et al. 2020). Disrupting this balance, either by mutation of the stabilizing or degron
regions, generates altered circadian lengths and impaired temperature compensation, disrupting
the stability of the organism's circadian rhythms (Masuda et al. 2020; Vanselow et al. 2024).
Phosphoswitch model. PER2 regulates the speed of the circadian clock through the axis
CDK5-CKI. When PER2 is phosphorylated by CDK5, it is stabilized and goes into the nucleus.
Under normal conditions, nuclear PER2 is phosphorylated by CKI ε/δ at Ser-477 which is
followed by nucleus/cytoplasm shuttling and proteasomal degradation. However, following a
switch in the temperature, PER2 can be phosphorylated at the FASP sites
(pS659-662-665-668-671), which stabilizes the protein. As a consequence, the clock is slowed
down. Additionally, CDK5 and CKI can reciprocally regulate their activity, speeding up or
slowing down the clock accordingly. (Brenna & Albrecht, 2020, Fig 4)
Genetic disruption of PER2 regulation confirms its function in circadian rhythm regulation. The
FASP domain S662G mutation reduces CK1 priming phosphorylation, destabilizes PER2,
increases degradation, and induces familial advanced sleep phase syndrome (FASPS)
(Narasimamurthy and Virshup 2018; Hildebrand et al. 2021). Disruption of the degron site,
however (S478A in mouse), increases PER2 half-life and extends the circadian period and
demonstrating that both increased and slowed PER2 turnover disrupts timing accuracy (Masuda
et al. 2020; Vanselow et al. 2024). uORF ablation also alters thermal entrainment dynamics
(Miyake et al. 2023), further supporting the conclusion that temporal control of PER2 translation
and degradation is required to maintain circadian integrity.
The role of PER2 is found to become increasingly tissue-specific, with varying effects based on
the organ system in question. PER2, in the liver, is implicated in the modulation of metabolic
rhythms, and liver-specific deletion of Per2 induces irregularities in lipid metabolism and
glucose homeostasis while maintaining the central SCN rhythm intact (Zhang et al. 2017). In
skeletal muscle, PER2 coordinates with local metabolic cues to modulate mitochondrial
oxidative potential and insulin sensitivity, suggesting its role beyond temporal regulation (Dyar
et al. 2014). Concurrently, within immune cells, PER2 controls cytokine expression and
inflammation, which indicates its association with immunometabolism and circadian fluctuation
in immune function (Nguyen et al. 2013).
PER2 disruption has also been implicated in various states of disease. Within cancer, PER2
functions as a tumor suppressor by regulating cell cycle checkpoints and apoptosis. Its
downregulation is also seen in breast and colorectal cancer, where it plays a role in enhanced
proliferation and adverse prognosis (Fu et al. 2022). In metabolic disorders, disrupted PER2
expression is associated with obesity and type 2 diabetes, as desynchronized feeding patterns and
disrupted circadian rhythms affect insulin signaling and energy storage (Zhang et al. 2016).
PER2 has also been implicated in mood and neuropsychiatric disorders. Animal models show
that reduced PER2 expression correlates with depression-like behavior and impaired reward
sensitivity, at least via dopaminergic circuit deregulation (Hampp et al. 2008).
Finally, other environmental stimuli apart from light and temperature influence PER2 dynamics.
Peripheral PER2 rhythms are modulated by circadian time under an unchanged light-dark cycle,
showing nutrient timing to be a potent zeitgeber for organs like the liver and gut (Hirao et al.
2010). Oxidative stress also activates PER2 due to ROS accumulation, suggesting that the
cellular redox state signals to the circadian system through stress-responsive transcription factors
(Jacobi et al. 2015). These findings highlight PER2's greater role as an integrative node,
coordinating environmental and physiological inputs to generate appropriate circadian
regulation.
Taken together, these open-access articles explain how PER2 employs transcriptional,
translational, and post-translational cues to function as a temperature-compensated molecular
timer. The synergy between uORF-directed translation, CK1-dependent phosphorylation, and
feedback repression by PER2/CRY complexes enables stable ~24‐hour oscillation in SCN
neurons and peripheral clocks. Perturbation of any of these regulatory layers, via mutation,
environmental stress, or genetic manipulation, leads to aberrant circadian timing, testifying to the
multi-tiered nature of PER2's role in chronobiology.
Circadian Rhythm Disorders
Circadian rhythm disorders are conditions where the internal body circadian clock is not
synchronized with the outside world, leading to persistent sleep time and quality problems.
Delayed Sleep Phase Syndrome (DSPS) is a condition in which there is a persistent delay in
sleep and wake times with respect to societal norms, leading to chronic insomnia and impairment
of daytime functioning (Wheaton et al. 2016). DSPS is generally hereditary, and this means there
is a genetic basis for the disturbed circadian timing. Dysregulation of the major clock genes,
including PER2, is increasingly implicated in DSPS and other sleep disorders. PER2 plays an
essential role in the perpetuation of the 24-hour rhythm by modulating feedback inhibition of the
circadian TTFL (Patke et al. 2017).
PER2 gene mutations were directly implicated in DSPS and other sleep disorders. One of the
best-studied mutations is the S662G substitution in the human PER2 protein, which disrupts the
CK1δ/ε phosphorylation site necessary for proper PER2 stability and degradation (Toh et al.
2001). The mutation leads to premature PER2 degradation, shortening the period of feedback
inhibition and causing a phase advance in the circadian rhythm, leading to familial advanced
sleep phase syndrome (FASPS). Conversely, PER2 dysregulation can also cause DSPS, while
increased PER2 turnover lengthens the circadian period and advances sleep (Patke et al. 2017).
Animal model research shows that mice deficient in Per2 exhibit disrupted activity rhythms and
disordered sleep structure, indicating that PER2 is essential for circadian period and sleep
structure maintenance (Zheng et al. 1999).
Total evidence from animal models and human studies highlights the functional significance of
PER2 dysregulation. Knock-in mice that carry the human S662G PER2 mutation show a shorter
circadian period and earlier activity onset, which is recapitulation of FASPS phenotypes in
humans (Xu et al. 2005). Per2 null mice, however, display longer circadian periods and sleep
cycle disorder (Zheng et al. 1999). Furthermore, recent transcriptomic studies in human DSPS
patients reveal dysregulated clock gene and PER2 expression profiles in peripheral blood
mononuclear cells, confirming the molecular pathogenesis of the condition (Kovanen et al.
2021). Molecular changes are then linked with enhanced melatonin and core body temperature
rhythm delay of secretion characterized clinically in DSPS patients (Smith et al. 2018).
Interestingly, PER2 dysfunction is not an independent phenomenon but operates in conjunction
with other clock components and environmental cues. The CK1δ/ε-dependent phosphoswitch
that regulates PER2 stability is subject to genetic mutation as well as environmental entraining
cues such as light and temperature, modifying the severity of circadian dysrhythmia (Lee et al.
2011). Recent work also suggests that PER2 further impinges on downstream targets of
metabolism and neurobehavior, further implicating circadian dysfunction with systemic
consequences. For example, PER2 controls dopaminergic activity in the striatum, so PER2
deletion interferes with reward-seeking behavior as well as with sleep-wake stability (Choi et al.
2021). In addition, pharmacological efforts to stabilize PER2 by CK1δ/ε inhibition or PER2
transcriptional activation with small molecules have been reported to reverse normal rhythms in
mouse models (Hirano et al. 2016).
Light therapy has also emerged as a non-invasive treatment to modulate PER2 expression in
DSPS patients. Early morning time-of-day bright light exposure has been shown to
phase-advance circadian rhythms by increasing PER2 mRNA levels in peripheral tissues (Skene
and Arendt 2007). The efficacy of light treatments is an indicator of the activity of PER2 as a
light-responsive gene under CLOCK:BMAL1 transcriptional regulation and subsequent
integration into behavior rhythms. In addition, nutritional and metabolic cues—i.e., restricted
feeding regimens—have also been found to entrain peripheral clocks independently of the SCN
using PER2-dependent pathways (Zhang et al. 2009). This corroborates PER2's role in central
and peripheral circadian systems, validating its function in DSPS pathophysiology.
Gene control tools, including CRISPRa, are also being investigated to overexpress PER2 in
circadian misalignment models. Preliminary evidence suggests that quite modest overexpression
of PER2 transcription is sufficient to reconstitute rhythmicity in PER2-deficient cell lines
without disrupting other clock genes (Takahashi et al. 2015). Molecular treatments, combined
with behavioral and pharmacological strategies, present an exciting multimodal platform for
treating DSPS in genetically vulnerable subjects.
Gene Editing Technologies
Gene editing technologies have transformed the world of molecular biology by enabling the
manipulation of genomic sequences with unprecedented accuracy. Among them, the
CRISPR-Cas9 system has been the most favored due to its ease of use, efficiency, and
programmability. Originally described as a bacterial adaptive immune system, CRISPR-Cas9
uses a guide RNA (gRNA) to direct the Cas9 endonuclease to a particular DNA sequence,
introducing double-strand breaks (DSBs) at the target locus. Host repair of the DSBs via
non-homologous end joining (NHEJ) or homology-directed repair (HDR) pathways can be
hijacked to generate targeted gene knockouts, insertions, or exact sequence edits (Jinek et al.
816; Doudna and Charpentier). Apart from canonical CRISPR-Cas9, newer tools such as
CRISPR activation (CRISPRa) and interference (CRISPRi) enable gene expression to be
regulated, regardless of DNA sequence alteration, by utilizing catalytically dead Cas9 (dCas9)
fused to transcriptional activators or repressors, respectively (Qi et al. 1173). Base editors expand
this toolkit by allowing direct change of single nucleotides (e.g., C-to-T or A-to-G) without the
induction of DSBs, reducing off-target effects and increasing precision in genetic correction
(Komor et al. 420). Collectively, these technologies offer multifaceted solutions to the control of
gene function at multiple levels of gene regulation.
Gene editing tools for the PER2 gene are of tremendous potential in cutting its function in
circadian biology and even in treating circadian rhythm disorders. CRISPR-Cas9 can be used to
generate PER2 knockouts to study loss-of-function phenotypes or repair disease-causing
mutations such as those involved in familial advanced sleep phase syndrome (FASPS) (Xu et al.
640). HDR-mediated editing of point mutations in the PER2 gene can restore normal protein
function in patient cells. In addition, CRISPRa approaches can rhythmically or tissue-specifically
enhance PER2 expression, with the potential to rescue disrupted oscillations in certain sleep
disorders. Conversely, CRISPRi can transiently suppress ectopic PER2 expression, offering a
reversible approach to modulate the clock without permanently editing the DNA. Base editing,
as it is precise, would have the capacity to selectively edit phosphorylation sites within PER2
that regulate its stability and nuclear localization, with high specificity precision tuning circadian
period length and phase (Lee et al. 1013).
Of critical importance in the application of gene editing to PER2 is the timing and tissue
specificity of interventions, reflecting the gene's endogenous circadian regulation and principal
function within the suprachiasmatic nucleus (SCN). The circadian system is highly responsive to
temporal cues, with PER2 expression strongly oscillating in SCN neurons and peripheral tissues
in a rigorously controlled 24-hour cycle (Takumi et al.). Constitutive editing or regulation of
PER2 in all tissues and at all times, then, holds the potential to disrupt physiological rhythms,
exerting deleterious effects such as sleep disorders or metabolic dysregulation. Inducible
CRISPR systems that can be activated or repressed by external stimuli, such as doxycycline,
rapamycin, or light, have also been useful tools for temporally specific intervention (Zetsche et
al. 139). These allow scientists to apply genetic modifications only during particular circadian
phases, reducing the risk of chronobiological disruption.
Tissue-specific delivery systems are also important. Adeno-associated viruses (AAVs) with
neuronal tropism are especially well-suited to target the SCN and other neuronal regions of the
brain that control circadian rhythms (Khan et al. 164). New synthetic vectors and lipid
nanoparticles also enable gene delivery with reduced immunogenicity and increased penetration
of the blood-brain barrier. Recent research has also drawn attention to the potential of
CRISPR-Cas systems paired with optogenetic modules, which allow editing to be regulated
spatially and temporally through exposure to light, a tool especially suitable to the light
responsiveness of the circadian system (Abudayyeh and Gootenberg 271). Such developments
are designed to maximize efficacy and minimize systemic side effects.
Therapeutic ramifications beyond basic research, meanwhile, are vast for gene editing
technologies being brought to bear on PER2. Circadian rhythm disorders like delayed sleep
phase syndrome and non-24-hour sleep-wake disorder result from mutations or misregulation of
clock genes like PER2. Personalized gene therapies would restore normal oscillatory function,
resulting in improved sleep quality and overall health (Sancar et al. 1636). Furthermore, the
circadian clock also controls a broad range of physiological processes, including metabolism,
immune response, and drug response, and thus the accurate regulation of PER2 expression is
likely to find broad clinical application beyond sleep (Bass and Takahashi 1349). Impaired PER2
function has also been implicated in certain cancers, where disruption of circadian function can
contribute to tumorigenesis, further broadening the potential of PER2 as a gene therapy target
(Fu et al. 213). However, challenges remain in delivering long-term safety, minimizing immune
responses, and achieving durable alterations in expression. Rigorous preclinical development
using animal models of circadian disruption is needed to confirm gene editing approaches for
clinical translation.
In summary, modern gene editing tools such as CRISPR-Cas9, CRISPRa/i, and base editors offer
powerful and versatile approaches to regulating PER2 gene function with high precision. They
hold the promise to both advance our understanding of circadian biology and develop novel
therapies for circadian rhythm disorders. Perhaps most critically, the effectiveness of these
strategies will demand consideration of the temporal and spatial dynamics of PER2 expression,
calling for advances in inducible and tissue-specific editing strategies to preserve physiological
rhythms while correcting dysfunction.
Mechanism of the CRISPR/Cas9 gene editing system. The single guide RNA (sgRNA) directs the
Cas9 nuclease to a complementary sequence in the genome where Cas9 will induce a
double-strand break (DSB). The target genomic locus must be followed by a 5′-NGG-3′motif
(protospacer adjacent motif, PAM) for Cas9 to function. DSBs are repaired by non-homologous
end joining (NHEJ) or by homology-directed repair (HDR) in the presence of a DNA repair
template, which can be exploited to introduce precise genetic modifications or exogenous
sequences. (Zhang et al., 2021)
Proposed model of the effects of PER2 silencing on lipid synthesis and cell cycle activity in
primary bovine mammary epithelial cells based on results from the present study. (Jing et al.,
2021)
Challenges in Targeting PER2 with Gene Editing
The circadian system creates many important physiological rhythms through the use of a tightly
regulated transcription-translation feedback loop (TTFL). PER2 is one of its most important
components and performs an important role in sustaining approximately 24-hour cycles. On the
other hand, having to manipulate PER2 for therapeutic or investigative purposes also comes with
challenges, especially when maintaining temporal precision, making sure of tissue-specific
relevance, and avoiding any systemic disruption. Qian et al. (2023) addresses these concerns by
implementing an innovative multi-tissue proteomics approach in the Per1/Per2 double knockout
mice to offer more insight into the gene's regulatory reach.
Beyond the proteomic rhythms, multiple additional studies have shown that PER2 also acts as an
important metabolic and transcriptional regulator across various types of systems. For example,
Grimaldi et al. (2011) identified PER2 as a direct repressor of PPARγ which is a central
transcription factor that is involved in lipid metabolism and adipocyte differentiation. This
suggests the possibility that PER2 disruption may result in broad metabolic effects that are
off-target and will further complicate any efforts to study or modulate its function. Similarly,
Hoshi et al. (2017) studied the PER2-dependent angiogenic rhythms in skin tissue and focused
on the importance of local tissue environments in the circadian outcomes.
A proteomic study that was conducted by Qian et al. (2023) used quantification based on tandem
mass tag (TMT) of protein rhythms across eight different tissues in Per1/Per2 double knockout
mice. Tissues were sampled at intervals of 2 hours under darkness to eliminate any entrainment
caused by light and to help with the analysis of endogenous circadian rhythmicity. This method
allowed the researchers to identify thousands of oscillating proteins in types of wild animals and
compare them to arrhythmic/altered patterns that were present in mutant mice.
Temporal Precision
In the study by Qian et al. (2023), researchers found a major loss of rhythmic protein activity in
nearly every tissue of mice that had both Per1 and Per2 genes knocked out of them. Generally, in
wild types of mice, a large number of proteins will display regular approximately 24-hour cycles
in their activity levels. But in the knockout mice, this rhythm was almost completely gone -
especially in proteins related to metabolism and cell signaling. This shows that PER2 is
important to keep the internal timing system of the body working properly. When PER2 is not
working, it's not just the timing or strength of these cycles that are affected, but the basic
structure of the circadian clock itself falls apart. This is likely because PER2 helps control the
CLOCK:BMAL1 complex and keeps the rest of the circadian system stable. Building on this,
Hoshi et al. (2017) looked at how PER2 affects specific tissues, such as the skin. They showed
that in PER2-deficient mice, important genes involved in blood vessel growth (like ANGPTL1)
lost their normal rhythm. This caused issues with how quickly wounds healed and how well
blood vessels repaired themselves. So PER2 isn't just involved in setting the body's overall
internal clock. It also plays an important role in managing time-sensitive processes right where
they happen, like in the skin.
Tissue Specificity
Qian et al. also showed that PER2's role is different depending on the tissue. In the wild-type
mice, organs like the liver, heart, and brown fat had strong and regular cycles in their protein
activity. But when PER2 was knocked out, these rhythms didn't disappear in the same way in
every tissue. Some organs were more affected than others and showed the possibility that each
tissue depends on PER2 to a different degree. For example, the SCN (which is the brain's master
clock) holds onto its rhythms more than other organs. This might be because the SCN has extra
mechanisms or stronger cell-to-cell connections that help it stay on track even without PER2.
This kind of variation was also seen in the Hoshi et al. study where PER2 loss had big effects on
gene timing in the skin, but not all the genes were affected. Some stayed rhythmic while others
did not. This shows that PER2 only controls specific genes depending on the tissue, meaning that
future treatments or gene edits that involve PER2 would need to be very carefully targeted.
Otherwise, accidental disruptions may take place in the important pathways in organs that don't
need to be affected.
Off Target Effects
In addition to its role in keeping our body clock on track, Grimaldi and colleagues (2011) found
that PER2 also plays a big part in metabolism, especially in how the body handles fat. They
discovered that PER2 binds to a gene called PPARγ which is crucial for the development of fat
cells. Under normal conditions, PER2 helps keep this gene under control. But when PER2 is
removed, that control is lost and leads to lower body fat, fewer triglycerides, and free fatty acids,
and even changes in how the body responds to insulin. This means PER2 isn't just about sleep
and circadian rhythms. Rather, it also plays a part in how the body stores and uses energy. That's
why changing PER2, even if it's just to study the internal clock, could unintentionally trigger
serious metabolic effects. This is a big deal for researchers using tools like CRISPR or siRNA
and changing PER2 might impact fat metabolism, blood sugar, or liver function, even if that
wasn't the goal.
The research reviewed here makes it clear that PER2 is greatly involved in many different
processes in the body. It's not just a timekeeper for circadian rhythms, but rather also helps
control when and where certain proteins and genes are active, and play a role in metabolism.
Qian et al.'s data showed how losing PER2 results in major disruptions in protein rhythms across
many tissues; Hoshi et al. showed that some genes depend on PER2 in specific tissues, like skin;
Grimaldi et al. showed that PER2 deletion messes with fat storage and insulin sensitivity. As
researchers look into using PER2 as a target for circadian-based treatments, they will need to be
careful. Changing PER2 could affect far more than just the human clock, especially if the
changes are not limited to the right time and place. Any gene editing or drug that targets PER2
needs to be designed carefully, because messing with it could lead to unexpected side effects in
metabolism, as well as wound healing and other important body functions.
Ethical and Safety Concerns
Gene editing tools like CRISPR offer favorable ways to treat disorders tied to our internal clocks
by targeting key genes like PER2. But because PER2 affects not just sleep, but also brain
function, mood, and metabolism, editing it raises serious ethical and safety concerns. While it's
crucial in keeping our 24-hour rhythms running smoothly, it also impacts emotional balance and
how our bodies use energy. That means changing PER2 is a technical hurdle, and it also brings
up questions about long-term safety, unintended effects, and where we draw the line between
treating illness and enhancing human abilities.
Editing the Brain and Behavior
Since PER2 is expressed in the suprachiasmatic nucleus of the brain and influences sleep-wake
cycles, editing it would involve altering brain function. According to Cohen et al. (2020),
modifying genes that affect behavior raises concerns about identity, autonomy, and personal
well-being. If gene editing was used not just to treat disorders, but also to increase productivity
or reduce sleep needs, it could be brought into enhancement where individuals could look to
optimize performance rather than restore health. This creates a slippery type situation where the
original goal of treating sleep disorders blurs into optional enhancement. In competitive
environments, this could lead to pressure on individuals to undergo gene editing to keep up and
result in raising fairness and ethical access concerns.
Long-Term Consequences and Gene-Environment Interactions
Editing PER2 could also have unpredictable long-term consequences, both biologically and
socially. As Mulvihill et al. (2017) explain, genetic interventions do not act alone as they interact
with an individual's environment, lifestyle, and health conditions. PER2 is involved in more than
sleep since it's linked to metabolism, hormone release, and mood regulation – changing it may
affect far more than intended. There's also the issue of off-target effects, where edits affect
unintended parts of the genome. These errors may not be immediately visible but could cause
problems years later. Since the circadian system affects so many parts of the body, even small
changes can have wide-reaching effects. That's why long-term monitoring and follow-up are so
important, especially in clinical trials, to catch any unexpected issues that might show up over
time.
Informed Consent in Clinical Settings
A major ethical concern when targeting PER2 in clinical trials is informed consent. Since the
effects of editing PER2 may take years to fully appear, participants may not understand what
they're agreeing to. De Araujo (2020) argues that ethical trials must go beyond short-term risks
and explain the unknowns, which include but are not limited to emotional, behavioral, and social
outcomes. This is important when editing genes that affect the brain. Even small changes in
mood or alertness might not be noticeable at first, but they can still have a big impact on a
person's quality of life. That's why informed consent needs to reflect this complexity and make
sure patients clearly understand both the potential benefits and the possible risks.
Potential for Enhancement Misuse
One of the most difficult challenges is preventing the misuse of PER2 editing for non-medical
enhancement. If gene therapy can reduce sleep needs or improve mental focus, it may be
marketed for performance enhancement rather than therapy. As Cohen et al. (2020) warn, this
could deepen inequalities if only certain groups can access any of the enhancement technologies.
It also raises more ethical questions such as “Should we edit human traits like sleep or
wakefulness for personal gain?” And if so, who will decide if that is acceptable? These questions
are beyond biology and step into the realm of social values, policy, and fairness.
Gene editing that targets PER2 shows real promise for treating circadian rhythm disorders,
especially when traditional treatments are not effective. However, because PER2 affects many
systems in the body, particularly the brain, it creates serious ethical and safety concerns. These
include the potential for misuse as an enhancement, unintended genetic changes, and challenges
with ensuring fully informed consent. As gene editing therapies continue to develop, especially
in areas related to sleep and brain function, the scientific community must proceed with caution
and prioritize safety, fairness, and clear communication with patients.
Future Directions and Potential
The Promise of Personalized Gene Therapies
Gene therapy targeting PER2 offers the possibility of more personalized treatments for circadian
rhythm disorders. It can influence not only sleep but also mood and metabolic health. As
Roenneberg and Merrow (2016) explain, each person has a unique biological clock, known as a
chronotype, that is partly shaped by genetics. This helps explain why some people are naturally
early risers while others tend to stay up late. Problems occur when these internal clocks become
out of sync with the environment or daily responsibilities. Researchers could develop gene
therapies that are made to their specific sleep patterns and genetic makeup by understanding
individual circadian biology and making the treatments more effective than the standard
approach.
Safer and Precise Gene Delivery Systems
For gene therapy to be successful, especially when working with something as delicate as the
brain, it's crucial to have safe and reliable delivery methods. Traditional CRISPR techniques
work by cutting both strands of DNA, which can lead to problems like unintended mutations or
even permanent damage. To reduce these risks, scientists are exploring newer approaches to gene
editing. One promising method that was introduced by Anzalone and colleagues in 2019, is
called prime editing. This helps researchers find and replace specific DNA sequences without
cutting both strands or needing a separate DNA template. As a result, the editing process is more
precise and less likely to cause harmful side effects. This is important when targeting a gene like
PER2 since it helps regulate essential biological rhythms.
Reversible/Non Permanent Gene Modulation
Another exciting area of development is the use of reversible or non-permanent editing
techniques. Instead of altering DNA directly, researchers can control the gene activity at the
epigenetic level. Liao et al. (2017) performed a method of gene activation using CRISPR
systems while fusing it with epigenetic modifiers which are essentially turning genes on or off
without making lasting changes to the genetic code. This “trans epigenetic” approach could be
helpful for regulating genes like PER2, where flexible or temporary changes might be safer than
permanent ones. For instance, if someone needs to adjust their sleep-wake cycle, the activity of
the gene could increase or decrease as needed without causing lasting changes to their DNA.
Conclusion
The PER2 gene plays an important role when it comes to managing our internal body clock,
which affects certain processes like when we feel tired, when we wake up, and how our body
stays in sync throughout the day. When something goes wrong with PER2, it can throw off our
circadian rhythm and result in problems like sleep issues like Delayed Sleep Phase Syndrome
where individuals will struggle to fall asleep or wake up at normal times.
Gene editing tools like CRISPR have opened up the possibility of fixing these kinds of problems
at the genetic level. Being able to directly adjust the genes that control our sleep and wake-up
cycle is an exciting idea and could lead to more effective treatments. But at the same time, there
are also a lot of challenges that are present. It is not that easy to make changes to a gene like
PER2 since it follows a strict daily rhythm and is only active in certain parts of the brain. There
is also the risk of editing the wrong genes, which may cause unwanted side effects, and also
ethical concerns that persist among many, counterarguing against gene editing as a whole.
Looking ahead, future research on PER2 should focus on understanding its behavior in more
detail. This includes how it interacts with other clock genes and environmental factors like light
and temperature, seeing as the circadian rhythm is highly responsive to external conditions.
Since PER2 follows a specific rhythm on a daily basis, it is essential that researchers study when
and where this gene is active in the body, especially in the brain's suprachiasmatic nucleus. By
tracking the activities of PER2 more clearly, scientists can determine how to time gene editing
treatments in a better way so that they can support the body's natural state rather than disrupt it.
Another important area of research is improving gene editing's safety and capability to be used
on a widespread scale. Technologies like CRISPR-Cas9, CRISPRa/i, and base editors show
promise but also come with risks, such as off-target edits and unintended changes in gene
expression even with. Future studies should aim to develop more precise versions of these tools
that can target PER2 in the SCN without affecting other parts of the brain or body. Researchers
should also explore reversible or non-permanent gene editing techniques, like CRISPR
interference (CRISPRi), to lower the risk of long-term side effects.
Additionally, more research that is focused on humans is needed. While animal models have
allowed us to discover a lot about PER2, human circadian rhythms are more complex and
influenced by lifestyle, social factors, and individual genetics. Future clinical trials should also
take these differences into account and include diverse populations. It will also be crucial to
design ethical studies that focus on informed consent, especially since gene editing in the brain
could impact unrelated, external qualities such as behavior, mood, and other quality-of-life
standards.
Overall, gene editing has the potential to become an influential tool for treating sleep disorders in
the future, but we're not at that stage yet. More research needs to be done to make sure these
treatments are safe and used responsibly. Learning more about PER2 will help us move in the
right direction and provide us with a better understanding of how we might treat these disorders
down the line.
References
Abudayyeh, O. O., & Gootenberg, J. S. (2021). CRISPR applications in living cells. Science,
372(6537), 271–275. https://doi.org/10.1126/science.abe6132
Anzalone, A. V., et al. (2019). Search-and-replace genome editing without double-strand breaks
or donor DNA. Nature, 576(7785), 149–157. https://doi.org/10.1038/s41586-019-1711-4
Bass, J., & Takahashi, J. S. (2010). Circadian integration of metabolism and energetics. Science,
330(6009), 1349–1354. https://doi.org/10.1126/science.1195027
Brenna, A., & Albrecht, U. (2020). Phosphorylation and Circadian Molecular Timing. Frontiers
in Physiology, 11. https://doi.org/10.3389/fphys.2020.612510
Choi, H., et al. (2021). PER2 modulates dopaminergic function and reward-related behavior. Cell
Reports, 36(2), 109403. https://doi.org/10.1016/j.celrep.2021.109403
Cohen, I. G., et al. (2020). The ethics of human enhancement: Understanding the debate.
Hastings Center Report, 50(S1), S2–S11. https://doi.org/10.1002/hast.1090
de Araujo, M. (2020). The ethics of genetic cognitive enhancement: Gene editing or embryo
selection? Philosophies, 5(2), 8. https://doi.org/10.3390/philosophies5020008
Doudna, J. A., & Charpentier, E. (2014). The new frontier of genome engineering with
CRISPR-Cas9. Science, 346(6213), 1258096. https://doi.org/10.1126/science.1258096
Dyar, K. A., et al. (2014). Transcriptional programming of lipid and amino acid metabolism by
the skeletal muscle circadian clock. PLoS Biology, 12(11), e1001958.
https://doi.org/10.1371/journal.pbio.1001958
Etchegaray, J. P., et al. (2009). The Polycomb group protein EZH2 is required for mammalian
circadian clock function. The Journal of Biological Chemistry, 284(9), 6459–6464.
https://doi.org/10.1074/jbc.M808020200
Fu, W., et al. (2022). PER2: A regulator of the circadian clock and cancer. Frontiers in Oncology,
12, 872261. https://doi.org/10.3389/fonc.2022.872261
Fu, L., et al. (2002). The circadian gene Period2 plays an important role in tumor suppression
and DNA damage response in vivo. Cell, 111(1), 41–50.
https://doi.org/10.1016/S0092-8674(02)00961-3
Grimaldi, B., et al. (2011). PER2 controls lipid metabolism by direct regulation of PPARγ. Cell
Metabolism, 14(4), 491–503. https://doi.org/10.1016/j.cmet.2011.07.016
Hampp, G., et al. (2008). Regulation of monoamine oxidase A by the circadian clock protein
Per2. Proceedings of the National Academy of Sciences, 105(19), 6560–6565.
https://doi.org/10.1073/pnas.0708692105
Hildebrand, M. S., et al. (2021). Circadian regulation of hearing and balance. F1000Research,
10, F1000 Faculty Rev-347. https://doi.org/10.12688/f1000research.52530.1
Hirano, A., et al. (2016). Chemical control of circadian behavior in mice by selective inhibition
of casein kinase 1δ/ε. Proceedings of the National Academy of Sciences, 113(25),
7011–7016. https://doi.org/10.1073/pnas.1601893113
Hirao, A., et al. (2010). Widespread tissue circadian clocks in mice revealed by
mPER2::luciferase bioluminescence imaging. Proceedings of the National Academy of
Sciences, 107(8), 3072–3077. https://doi.org/10.1073/pnas.0913187107
Hoshi, T., et al. (2017). Circadian rhythms of angiogenic factors in skin and wound tissue in
Per2-mutant mice. Biomedical Research and Clinical Practice, 2(3).
https://www.oatext.com/pdf/BRCP-2-135.pdf
Jacobi, D., et al. (2015). Oxidative stress and circadian clock disruption promote
neurodegeneration. Frontiers in Aging Neuroscience, 7, 30.
https://doi.org/10.3389/fnagi.2015.00030
Jinek, M., et al. (2012). A programmable dual–RNA–guided DNA endonuclease in adaptive
bacterial immunity. Science, 337(6096), 816–821.
https://doi.org/10.1126/science.1225829
Jing, Y., et al. (2021). Circadian Gene PER2 Silencing Downregulates PPARG and SREBF1 and
Suppresses Lipid Synthesis in Bovine Mammary Epithelial Cells. Biology, 10(12),
1226–1226. https://doi.org/10.3390/biology10121226
Khan, I., et al. (2018). Advances in viral vector-based gene delivery for gene therapy. Current
Drug Delivery, 15(2), 164–176. https://doi.org/10.2174/1567201814666170223142502
Komor, A. C., et al. (2016). Programmable editing of a target base in genomic DNA without
double-stranded DNA cleavage. Nature, 533(7603), 420–424.
https://doi.org/10.1038/nature17946
Kovanen, L., et al. (2021). Transcriptomic analysis reveals circadian disruption in human
delayed sleep phase disorder. Scientific Reports, 11(1), 22465.
https://doi.org/10.1038/s41598-021-01523-x
Lee, H.-J., et al. (2011). Posttranslational regulation of circadian rhythm: Ubiquitination of PER2
by β-TrCP1 and its relevance to the FASPS mutation. Cell Cycle, 10(5), 719–726.
https://doi.org/10.4161/cc.10.5.14974
Lee, C., et al. (2006). Posttranslational mechanisms regulate the mammalian circadian clock.
Cell, 125(6), 1013–1024. https://doi.org/10.1016/j.cell.2006.03.039
Liao, H. K., et al. (2017). In vivo target gene activation via CRISPR/Cas9-mediated
trans-epigenetic modulation. Cell, 171(7), 1495–1507.e15.
https://doi.org/10.1016/j.cell.2017.10.025
Masuda, T., et al. (2020). The molecular basis of PER2 protein stability and its modulation by
phosphorylation. Cell Reports, 30(13), 4295–4307.e5.
https://doi.org/10.1016/j.celrep.2020.03.035
Miyake, N., et al. (2023). Temperature-dependent regulation of PER2 translation via uORF
suppression. iScience, 26(2), 106041. https://doi.org/10.1016/j.isci.2023.106041
Mulvihill, J. J., et al. (2017). Ethical issues of CRISPR technology and gene editing through the
lens of solidarity. British Medical Bulletin, 122(1), 17–29.
https://doi.org/10.1093/bmb/ldx002
Narasimamurthy, R., & Virshup, D. M. (2018). PER2 protein stability and circadian period
determination. Frontiers in Physiology, 9, 1064.
https://doi.org/10.3389/fphys.2018.01064
Nguyen, K. V., et al. (2013). Circadian gene PER2 inhibits mTORC1 signaling and regulates
lipid metabolism. Cell Reports, 5(6), 1611–1621.
https://doi.org/10.1016/j.celrep.2013.11.028
Patke, A., et al. (2017). Mutation of the human circadian clock gene CRY1 in familial delayed
sleep phase disorder. Cell, 169(2), 203–215.e13.
https://doi.org/10.1016/j.cell.2017.03.027
Qi, L. S., et al. (2013). Repurposing CRISPR as an RNA-guided platform for sequence-specific
control of gene expression. Cell, 152(5), 1173–1183.
https://doi.org/10.1016/j.cell.2013.02.022
Qian, J., et al. (2023). A multi-tissue proteomics atlas of wild-type and Per1/Per2 knockout mice.
Molecular & Cellular Proteomics, 22(7), 100539.
https://doi.org/10.1016/j.mcpro.2023.100539
Roenneberg, T., & Merrow, M. (2016). The circadian clock and human health. Current Biology,
26(10), R432–R443. https://doi.org/10.1016/j.cub.2016.04.011
Sancar, A., et al. (2015). Circadian clock control of the cellular response to DNA damage. FEBS
Letters, 589(14), 1636–1644. https://doi.org/10.1016/j.febslet.2015.05.015
Smith, R. T., et al. (2018). Circadian rhythms and delayed sleep phase disorder: Current
perspectives. Nature and Science of Sleep, 10, 1–9. https://doi.org/10.2147/NSS.S124503
Takumi, T., et al. (2015). Molecular architecture of the circadian clock in the mammalian
suprachiasmatic nucleus. Frontiers in Neuroscience, 9, 101.
https://doi.org/10.3389/fnins.2015.00101
Toh, K. W., et al. (2001). A hPer2 phosphorylation site mutation in familial advanced sleep phase
syndrome. Science, 291(5506), 1040–1043. https://doi.org/10.1126/science.1057499
Vanselow, K., et al. (2024). Temperature compensation of circadian rhythms is controlled by the
stability of PERIOD proteins. Nature Communications, 15, 1056.
https://doi.org/10.1038/s41467-024-44653-2
Virshup, D. M., Eide, E. J., Forger, D. B., Gallego, M., & Harnish, E. V. (2007). Reversible
Protein Phosphorylation Regulates Circadian Rhythms. Cold Spring Harbor Symposia on
Quantitative Biology, 72, 413–420. https://doi.org/10.1101/sqb.2007.72.048
Wheaton, A. G., et al. (2016). Prevalence of healthy sleep duration among adults — United
States, 2014. Morbidity and Mortality Weekly Report, 65(6), 137–141.
https://doi.org/10.15585/mmwr.mm6506a1
Xu, Y., et al. (2005). Functional consequences of a CKIδ mutation causing familial advanced
sleep phase syndrome. Nature, 434(7033), 640–644. https://doi.org/10.1038/nature03453
Zetsche, B., et al. (2015). A split-Cas9 architecture for inducible genome editing and
transcription modulation. Nature Biotechnology, 33(2), 139–142.
https://doi.org/10.1038/nbt.3149
Zhang, H., et al. (2021). Application of the CRISPR/Cas9-based Gene Editing Technique in
Basic research, diagnosis, and Therapy of Cancer. Molecular Cancer, 20(1), 126.
https://doi.org/10.1186/s12943-021-01431-6
Zhang, R., et al. (2017). Liver clock regulates the diurnal rhythm of glucocorticoid signaling
through HDAC3. Cell Metabolism, 25(4), 707–718.
https://doi.org/10.1016/j.cmet.2017.01.006
Zhang, Y., et al. (2016). Disruption of the PER2 gene induces obesity in mice. Frontiers in
Physiology, 7, 592. https://doi.org/10.3389/fphys.2016.00592
Zheng, B., et al. (1999). The mPer2 gene encodes a functional component of the mammalian
circadian clock. Nature, 400(6740), 169–173. https://doi.org/10.1038/22118