• Open Access
    Review

    An intricate relationship between circadian rhythm dysfunction and psychiatric diseases

    Saptadip Samanta 1*
    Debasis Bagchi 2,3

    Explor Neurosci. 2024;3:321–351 DOI: https://doi.org/10.37349/en.2024.00053

    Received: March 20, 2024 Accepted: June 11, 2024 Published: August 23, 2024

    Academic Editor: Ertugrul Kilic, Istanbul Medipol University, Turkey

    This article belongs to the special issue Circadian Rhythm and Melatonin

    Abstract

    There is a complex relationship between circadian rhythm dysfunctions and various psychiatric disorders. Circadian (~24 h) rhythms indicate the rhythmic change of different physiological activities in relation to the environmental light-dark cycle. Shift work, light exposure at night, and chronic and acute jet lag affect circadian rhythm that have a negative impact on psychological functions, and behaviors. Additionally, professional stress, mental instability, and social disintegration influence psychiatric disorders. PubMed/MEDLINE, Springer Nature, Science Direct (Elsevier), Wiley Online, ResearchGate, and Google Scholar databases were searched to collect relevant articles. Circadian rhythm disruption causes impaired neurotransmitter release, impaired melatonin and cortisol rhythm, metabolic dysfunctions, neuroinflammation, and neural apoptosis; collectively these factors influence the development of psychiatric disorders. Circadian dysfunction also alters the expression of several clock control genes in the mesolimbic areas that are associated with pathologies of psychiatric disorders. Additionally, chronotherapy and applications of anti-psychotic medicine can improve psychiatric diseases. This review focuses on the effects of circadian clock dysfunction on the vulnerability of psychiatric disorders and the implications of chronotherapy.

    Different factors affect the circadian rhythm and also subucortical areas. Suprachiasmatic nucleus (SCN) acts as a master clock and sends output in different areas of the brain for the regulation of various physiological functions. Impairment of SCN activities and rhythmic gene expression promote psychiatric disorders. ALAN: artificial light at night; CD: circadian; ENV: environmental; MT: melatonin; NT: neurotransmitters; RHT: retinohypothalamic tract

    Keywords

    Suprachiasmatic nucleus, circadian rhythm dysfunction, psychiatric disorders, melatonin, cortisol, antipsychotic treatment

    Introduction

    Psychiatric disorders are serious problems throughout the world. The global scenario indicates that the mental illness burden is about 13.0% of disability-adjusted life-years (DALY) and 32.4% of years lived with disability (YLD) [1]. Psychiatric disorders affect cognition, emotional, and behavioral activities. Psychiatric disorders begin in early life and often appear as chronic recurrent diseases. There are different types of psychiatric diseases, including schizophrenia, attention deficit/hyperactivity disorder (ADHD), autism, major depressive disorder (MDD), bipolar disorder (BD), anxiety disorders, and seasonal affective disorder (SAD). Mental illness is detrimental to cognitive behavior, social activities, and economic performance. Psychiatric disorders have complex etiologies, and genetic defects, altered gene expression, mitochondrial dysfunction, and poor antioxidant activity are the influencing factors. Other factors, like environmental factors (antenatal maternal viral infection, fetal hypoxia due to obstetric complications, mother’s stress during fetal neural development, migration, urbanicity, social stress, and exposure to lead) and neuroinflammation are also associated with psychiatric diseases. Inflammatory mediators, particularly different cytokines alter neuroendocrine activity, synaptic plasticity, release of neurotransmitters (NTs) release, and monoamine metabolism, leading to the progression of mental illness [2, 3]. Some diseases may be linked with gender variation. The risk of autism is higher in males, while females have a higher rate of MDD, and anxiety disorders [4]. According to Patel and Kondratov [5], adverse circumstances, early childhood abuse, violence, poverty, and stress (physical, mental, and social) can promote psychiatric disorders. Currently, different psychotropic drugs are used for the treatment of schizophrenia, BD, major depression, and anxiety disorders. However, chronotherapy can improve psychiatric disorders.

    Circadian rhythms are the rhythmic physiological manifestations along the solar day. These ~24-hour rhythms are controlled by the suprachiasmatic nucleus (SCN) of the hypothalamus. A complex relationship exists between circadian rhythms and mental health. Jet lag, shift work, and artificial light exposure at night promote circadian rhythm disruption. This dysfunction disturbs clock-controlled responses, such as the sleep-wake cycle cortisol and melatonin secretion, and core clock gene expression, resulting in mood disorders, depression, mania, and schizophrenia [6, 7]. The present review has focused on the relationship between circadian rhythm dysfunction and psychiatric disease, particularly schizophrenia, anxiety, depression, BD, MDD, autism, and SAD.

    Methodology

    PubMed/MEDLINE, Springer Nature, Science Direct (Elsevier), Wiley Online, ResearchGate, Google Scholar databases, and others were thoroughly reviewed to search for relevant articles on SCN activity, circadian dysfunction, and psychiatric disorders. Different keywords like structure and functions of SCN, circadian rhythm dysfunction/chronodisruption, psychiatric disorders/mental illness, and circadian dysfunction and psychiatric diseases were used during the literature survey. The final version of this review has been prepared based on the search content.

    Background of psychiatric diseases

    Psychiatric disorders are characterized by significant disturbances in cognition, emotion, and behavior. Different cytokines, chemokines, and growth factors are prime components of neuroinflammation and these agents act as the pro-inflammatory and anti-inflammatory subsets. An imbalance in the expression of pro-inflammatory and anti-inflammatory agents potentially influences the development of various neuropsychiatric diseases [8]. Multiple sclerosis (MS) is an inflammation-mediated neurodegenerative disease. The risk of depression and anxiety is higher in patients with MS [9]. MS patients showing a relapsing stage exhibited elevated levels of inflammatory cytokines TNF-α, IL-1β, and IL-2 in the cerebrospinal fluid (CSF) [10]. Several areas of the brain are affected by various psychiatric diseases, and multiple factors are associated with these disorders (Table 1).

    Primary symptoms, cause, and fundamental treatment strategies of common psychiatric diseases

    Psychiatric disordersSymptomsAffected brain area        CausesTreatment
    SchizophreniaHallucinations, delusions, disorganized thinking, avolition, diminished emotional expression, cognitive impairmentsFrontal and temporal lobes, hippocampus, amygdala, and thalamus
    • Improper secretion of dopamine and serotonin

    • Mutation in DTNBP1, NRG1, COMT, RGS4, GRM3, DISC1, and DAOA genes

    • Oxidative stress and neuroinflammation

    • Use of clozapine

    • Application of nitric oxide synthase 1 adaptor protein

    • Nonsteroidal anti-inflammatory drugs

    Anxiety disordersChest pain, palpitations, respiratory difficulty, headache fear, tachycardia, shortness of breath, sweatingAmygdala, hypothalamus, locus coeruleus
    • Impaired activity of GABAergic neurons

    • Abnormal secretion of cortisol and norepinephrine

    • Use of escitalopram, venlafaxine, benzodiazepines, and buspirone

    • Cognitive behavioral therapy

    DepressionPoor mental health, high suicide risk, decreased quality of life, drug addictionPrefrontal cortex, hippocampus, amygdala, and Brodmann Area 25
    • Insufficient concentration of norepinephrine and serotonin

    • Abnormal secretion of cortisol

    • Focal ischemia and neuroinflammation

    • Use of monoamine oxidase inhibitors (clorgyline and deprenyline)

    • Use of monoamine reuptake inhibitors (imipramine and amitryptiline)

    • Application of flavonoids like antioxidants

    Major depressive disorderDepressed mood, sadness, poor pleasure feeling, insomnia, excessive guilty feeling, hopelessness, problem to concentrate on work, suicidal thinkingHypothalamus, limbic system, basal ganglia, and cerebellum
    • Neuroinflammation

    • Abnormal activities of serotoninergic, and GABAergic neurons

    • Cognitive-behavioral therapy

    • Use of perazine, clomipramine, and fluoxetine as psychotic drugs

    Display full size

    DTNBP1: dystrobrevin binding protein 1; NRG1: neuregulin 1; COMT: catechol-O-methyl transferase; RGS4: regulator of G-protein signaling 4; GRM3: glutamate metabotropic receptor 3; DISC1: disrupted in schizophrenia 1; DAOA: D-amino acid oxidase activator; GABA: γ-aminobutyric acid

    Schizophrenia

    Schizophrenia is a chronic mental illness. The superior temporal gyrus and dorsolateral prefrontal cortex (PFC) are the most affected areas [11, 12]. This disease is characterized by hallucinations, delusions, disorganized thinking, avolition, diminished emotional expression, and cognitive impairments. In extreme cases, schizophrenia affects the patients’ livelihoods [13]. Schizophrenia patients show hallucinations through sensory modalities, particularly by hearing voices or noises. Delusions depend on beliefs and negative thoughts that are not associated with the patient’s culture. The patients exhibit social deficits, disorganized thoughts, speech-impaired emotional responses, and lack of motivation. There are learning restrictions and poor memory capacity among the sufferers [14]. Different cognitive tests like Stroop tests, Wisconsin card sorting tests, and verbal learning tests exhibit poor results [15]. Abnormalities occur in the frontal and temporal lobes, hippocampus, amygdala, and thalamus. Moreover, impaired secretion of dopamine and serotonin is also associated with this disease. Many genes are associated with various mental diseases. More than a dozen genes increase the risk of schizophrenia. Among these, mutation in the dystrobrevin binding protein 1 (DTNBP1), neuregulin 1 (NRG1), catechol-O-methyl transferase (COMT), regulator of G-protein signaling 4 (RGS4), glutamate metabotropic receptor 3 (GRM3), disrupted in schizophrenia 1 (DISC1), and D-amino acid oxidase activator (DAOA) genes increase the susceptibility of schizophrenia [16]. Neuroinflammation plays a vital role in schizophrenia pathology [17]. Results of a meta-analysis study indicated that patients with schizophrenia have increased levels of IL-17, IL-23, IL-6, TNF-α, and soluble IL-2 receptor (sIL-2R) [18, 19].

    Microglia are activated by damage-associated molecular patterns, ionized calcium-binding adaptor molecule 1, and heat shock protein 70. Activated microglia produces pro-inflammatory cytokines (IL-1β, IL-6, TNF-α) and reactive oxygen species (ROS). Microglia also induce the expression of inducible nitric oxide synthase (iNOS) for the synthesis of nitric oxide (NO). Expressions of advanced glycation end-product receptors in the neuronal microenvironment also occur during pathogenesis. Collectively, all these effects promote atrophy of the astrocytes and apoptosis of the neural cells, resulting in behavioral and cognitive deficits in schizophrenic patients [20]. Antipsychotic medication like clozapine is used for the treatment of schizophrenia. Moreover, counseling and rehabilitation can give positive results [21]. Cognitive therapy is applicable for the improvement of schizophrenia.

    Anxiety and stress disorders

    Anxiety disorders have complex features. Most of the patients show somatic complications, such as chest pain, palpitations, respiratory difficulty, and headache. The impaired activity of γ-aminobutyric acid (GABA)ergic neurons and functional defects in the amygdala-hypothalamus-central grey matter-locus coeruleus circuit are associated with anxiety disorders [22]. Panic disorder is a class of anxiety disorders, which is accompanied by fear, tachycardia, shortness of breath, and excessive sweating [23, 24]. People with panic disorder may restrict their activities to avoid situations where they feel panic. They sometimes avoid crowds, traveling, and elevators. Decreased levels of cortisol and prolactin may be the causative factors [25]. Excessive worry and fear have been observed in generalized anxiety disorder, which stimulates the sympathetic nervous system [26].

    Social anxiety disorder is a chronic mental condition where fear and anxiety appear when an individual is surrounded by people. It is a common type of anxiety disorder. Affected individuals are feeling shyness, discomfort, and embarrassment in certain situations. They commonly try to avoid public addresses. They feel nervous and worried about teasing, rejection, and humiliation. Their voice becomes shaky. Therefore, they have poor social skills, difficulty in making social relationships, and low academic and employment achievements. Victim persons are feeling fear when they are scrutinized, evaluated, or judged by others. They have difficulties during answering a question in a class, job interview, and speaking in a public interview. Primarily, social anxiety disorder has a familial history and appears as an inherited trait. In this case, hyperactivity occurs in the amygdala. Cognitive behavioral therapy (CBT), antidepressants like selective serotonin reuptake inhibitors (SSRIs), beta-blockers, and anxiolytic drugs like benzodiazepines give positive results against social anxiety disorder [27].

    Phobias are a type of anxiety disorder that causes an irrational fear from a situation, living system, or object. Specific phobia arises from the realization of a specific object, an event, or a situation. Social phobia is a fear of public humiliation. Agoraphobia creates a fear of a situation from which it would be difficult to escape. Phobias are also divided into different subtypes like personalized (blood, injection, injury), animals (spiders, insects, snakes, dogs), natural (flood, storm, earthquake, height), and situations (airplanes, elevators, enclosed places). Affected individuals show sweating, abnormal breathing, tachycardia, headache, trembling, chest pain, and dry mouth. Antidepressants like SSRIs, beta-blockers, benzodiazepines, and monoamine oxidase (MAO) inhibitors can be used for treatment [28].

    Posttraumatic stress disorder (PTSD) shows traumatic episodes and emotional numbness after any exposure to environmental situations that remind the trauma and affect sleep quality [29]. Various causes, like accidents, disasters, physical or sexual assault, and terror attacks can initiate PTSD. The individuals show unnecessary fear, nightmares, severe anxiety, feeling tense, and depressing thoughts. In anxiety and PTSD, functional defects occur in the hypothalamus and amygdala [30, 31]. Abnormal secretions of cortisol and norepinephrine are also involved in these disorders. Moreover, different psychological factors like negative appraisals, fatalistic beliefs, early childhood traumas, lack of social support, limited coping skills, and poor self-capacities affect traumatic disorders. Hayes et al. [32] reported that CBT can improve the conditions of post-traumatic disorder. Social support, education, and guidance can improve the symptoms. The application of SSRIs may give positive results.

    Mood disorders

    Mood disorders predominantly affect the emotional state of the person, which is indicated by depression, and irritation. Moreover, abnormalities in emotional behavior cause physiological changes, such as disturbance in the sleep-wake cycle, loss of appetite, poor energy output, and cognitive impairment. MDD and BD are the most common mood disorders.

    Depression is considered a psychological symptom of stress, which is a highly comorbid disorder. This condition causes upsetting of mental health that affects the individual’s life. Depression is known as one of the most common psychiatric conditions where the risk of suicide, drug consumption, and healthcare costs, are high, and their quality of life is poor. Affected persons are facing social and economic problems [33]. Depression commonly occurs when insufficient concentrations of NTs are present in monoaminergic synapses. Impaired secretion of noradrenaline (NA) and 5-hydroxytryptamine (5-HT) also known as serotonin can promote depression. The exogenous depressive agent like reserpine depletes monoamine NT and tends to increase depression. Tyrosine hydroxylase is an essential enzyme for NA synthesis. Mutation in chromosome 11 causes defective expression of tyrosine hydroxylase, resulting in low levels of NA. On the other hand, dysfunction in the serotoninergic neurons in different areas, including the PFC, hippocampus, amygdala, and Brodmann area 25 causes exaggeration of depression. Neuroinflammation also decreases serotonin levels by activating the kynurenine pathway. This activity modulates the function of the N-methyl-D-aspartate (NMDA) receptors and diminishes serotonin levels, leading to the progression of depressive disorder [34]. Moreover, the hypothalamic-pituitary-adrenal (HPA) axis is also involved in pathophysiology of depression [35].

    Stress response influences focal ischemia and glial activation that induces the expression of cytokines (TNF-α, IL-1β, and IL-6) for the advancement of depression [36]. The stress response also activates NMDA receptors that transiently increase the expression of iNOS for the synthesis of NO. The subsequent effect is the induction of cytokine release. Experimentally, lipopolysaccharide (LPS) is the potent inducer of TNF-α expression. The inflammatory response also affects NT metabolism, neuroendocrine function, and neural plasticity. Synthesis of dopamine and serotonin is notably decreased in depressed individuals [36]. Pharmacotherapy and CBT are the common treatments for depression and anxiety [37]. MAO inhibitors such as clorgyline and deprenyline decrease the breakdown of monoamines and act as the antidepressants. The reuptake of monoamine from the synaptic terminal is inhibited by reuptake inhibitors (imipramine and amitryptiline) that can improve depressive effects. Moreover, antidepressants downregulate the expression of presynaptic monoamine receptors. Natural products are being used as alternatives to pharmacotherapy for the management of various diseases, including depression and anxiety [38]. Among them, natural compounds flavonoids are effective in decreasing depressive symptoms in experimental models, possibly through the brain-derived neurotrophic factor (BDNF) expression, activation of monoaminergic systems as well as antioxidant effects [39].

    Major depressive disorder

    It is characterized by continuously depressed mood, sadness, psychomotor agitation, poor pleasure feelings, insomnia or hypersomnia, excessive guilty feelings, hopelessness, decreased ability to concentrate on work, and feelings of worthlessness and thoughts of death. Some physiological changes also occur; these include sleep disturbance, poor memory, loss of appetite, weight loss, decreased energy output, and slowed motor movements [40]. Neuroinflammation progresses MDD. High levels of cytokines, such as IL-17, IL-23, IL-6, TNF-α, sIL-2R, and IL-1 receptor antagonist (IL-1RA) were observed in patients with MDD and BD [18, 41]. However, macrophage migration inhibitory factor (MIF) exerts protective measures against MDD [41].

    Bipolar disorder

    BD shows extreme mood swings behavior. Episodic mania and depression along with relative periods of healthy mood (euthymia) are the common features of BD that appear cyclically with a period of interval. BD is divided into four categories on the basis of symptoms. Bipolar I disorder shows severe manic symptoms that last for at least 7 days. Depressive episodes also occur that continue for 2 weeks. Bipolar II disorder exhibits a pattern of depressive episodes and hypomanic episodes. The third type is cyclothymic disorder, where recurring hypomanic and depressive symptoms appear but do not last for a long time. If the symptoms do not match with these three categories then the disorder is specified as other or bipolar related disorders.

    Sometimes BD shows both features of bipolar I and II. This is also called mixed affective BD. In this type, depression and mania or hypomania occur at the same time or very quickly after each other. The mixed affective states are more severe and very difficult to treat. Imbalance occurs in both the catecholaminergic and cholinergic systems. The mixed affective disorder is also associated with circadian disruption. Circadian rhythm irregularities cause stressful life, sleep problems, arrhythmic melatonin, and cortisol secretion. Thus, circadian rhythm dysfunction is a major contributor to affective symptoms [42].

    Mania is a typical feature of BD, which is characterized by insomnia, and irritability. Mania often exhibits intrusive, impulsive, disinhibited behaviors, and poor judgment. Social jet lag promotes mania, but not depression in BD patients [43]. BD patients show sleep problems. The patients may suffer from thought difficulties and jump rapidly from one idea to another. BD seems to be a genetic disorder and exhibits heritability characteristics [44]. The affected areas are the basal ganglia and cerebellum. Abnormal activities of noradrenergic, serotoninergic, and GABAergic neurons are linked with BD. Mice model study revealed that low expression of dopamine transporter exhibits mania-like behaviors; thus, dopaminergic signaling is associated with BD [45].

    Unipolar disorder

    Unipolar disorder shows one or more episodes of moderate to severe depression. Mania is a common phenomenon in BD. However, in unipolar disorder, there is a lack of alternative episodes of mania and depression. Affected individuals show sleep problems, feelings of sadness, inability to experience pleasure, lack of concentration in activity, change in appetite, body weight, and suicidal thoughts. Commonly, social, familial, and neurological factors are involved in this disease. Unipolar disorder is qualitatively different in etiology from BD. BD has a stronger genetic basis than unipolar disorder. An imaging study revealed that reduced blood flow occurs into the cerebral cortex, particularly in the PFC, and corpus callosum. Impaired metabolism was observed in the PFC, temporal cortex, amygdala, and basal ganglia. Low levels of dopamine and NA decrease the activity of the mesolimbic area. Decreased level of serotonergic regulation is also involved in the pathogenesis. CBT, SSRIs, tricyclic anti-depressants, and MAO inhibitors can be used for treatment purposes [46].

    Autism spectrum disorder and ADHD

    Autism spectrum disorder (ASD) develops in early life, mostly in childhood. Affected individuals show intellectual and adaptive deficits. They feel difficulties in social communication and social interaction. Moreover, they have poor interest in activities and weak verbal and non-verbal communication. There is no definite cause of ASD. It is mostly idiopathic and multiple factors are associated with this disease. In some cases, genetic factors are involved. Single nucleotide variants and copy number variants are considered as the genetic problems [47]. Epigenetic changes increase the rate of abnormal DNA methylation, which promotes ASD. Older fathers carry multiple imprinted gene loci that are susceptible to epigenetic changes [48]. Animal studies revealed that abnormal nutrition, stress, and transplacental psychiatric drugs affect GABAergic, dopaminergic, serotonergic, and glutamatergic neurotransmission and hamper neurodevelopment in fetal life. Collectively, these factors promote ASD [49]. Singh et al. [50] reported that perinatal brain injury, especially cerebellar injury, is a contributing factor in ASD development. There is no curative treatment for ASD. However, behavior and communication therapy, speech therapy, and sensory integration therapy are effective for the treatment of ASD. These therapies increase social activity, and language skills and decrease behavioral problems, aggressiveness, and agitation. The goal of the treatment is to make the children functional and independent and to improve their quality of life. There is no specific medication. However, some medicines can improve certain symptoms. Neuroleptics (pimozide and haloperidol) are an effective choice against behavioral problems. Risperidone decreases aggressiveness and agitation. Antidepressant drugs and psychotic drugs can improve behavior. SSRIs act as the mood stabilizers. Melatonin gives promising results in combating sleep problems [51].

    ADHD is a specific type of autism. Significant loss of attention and impulsive behavior are the primary symptoms. Failure to self-identity, impaired regulatory systems and mind development, high levels of stress, and family problems can influence ADHD. Prenatal difficulties have a negative impact on this disease. Different areas are mostly affected; these include the frontal and temporal lobes, striatum, limbic system, cerebellum, and brain stem nuclei. Improper secretion of dopamine occurs in this condition. There is a genetic predisposition in the contributing list. ADHD has a high risk of inheritability. Genetic variation may contribute to ADHD. Single nucleotide polymorphism (SNP) has a role in the development of ADHD. Several genes such as DRD4 (dopamine receptor), DRD5, DAT1 (dopamine transporter), SNAP25 (gene for synaptosomal associated protein 25 kd), and COMT (catechol O-methyl transferase) are associated with this disorder. Chromosomal abnormalities (sex chromosome, specifically, X chromosome) may also be involved. Various extrinsic factors like smoking, alcoholism, poor nutrition, and exposure to carbon monoxide (CO), lead, and polychlorinated biphenyl (PCB) during pregnancy, excessive mental stress and obesity of the mother, and low birth weight of the child have a role in the development of ADHD [52]. Different therapies can improve the symptoms of ADHD. CBT (training for social skills, function, and cognitive development) is a common psychotherapy, which is effective against ADHD without any medication. Pharmacotherapies are used for symptomatic treatment. There are three types of medication. These include stimulants (methylphenidate, dexamethylphenidate amphetamine, norepinephrine, and dopamine), non-stimulant (α-2 adrenergic receptor agonist, selective norepinephrine reuptake inhibitor), and antidepressants (bupropion, venlafaxine, desipromine). Additionally, supplementation of polyunsaturated fatty acids has some effective role [53].

    Seasonal affective disorder

    SAD also called seasonal depression is a specific type of depression that is associated with seasonal changes. In most cases, SAD appears during early winter and goes away during the sunnier days of spring and summer. Some people show the opposite effects. Symptoms start during spring or summer. The common symptoms are feeling sad, depression, anxiety, restlessness, sleep problems, appetite changes, weight gain/loss, loss of interest in activities and enjoyment, difficulty to concentrating on work, feeling hopeless, worthless, or guilty, and suicidal thinking. Misalignment of circadian rhythm due to changes in the duration of the light period alters the melatonin output and sleep properties, resulting in the progression of SAD [54]. In addition to sleep disorders, abnormalities in the diurnal rhythm of core body temperature, impaired cortisol, and melatonin secretion are the contributing factors [55]. Moreover, genetic variations in the endogenous circadian clock are also linked with SAD [56, 57].

    Impaired serotonergic activity with seasonal variation intensifies SAD. Animals and normal humans exhibit altered serotonin activity across the seasons. Sunlight regulates the serotonergic pathway. A lack of sunlight during winter decreases serotonin levels, leading to further amplification of depression. Human post-mortem samples of the hypothalamus showed considerable seasonal variations in serotonin levels. The minimum levels were found during the winter months of December and January. This effect probably alters eating behavior, carbohydrate craving, and body weight [58].

    Tryptophan is the precursor of serotonin. There is a seasonal variation in tryptophan metabolism. 5-hydroxyindole acetic acid (5-HIAA) appears during serotonin synthesis. High levels of 5-HIAA indicate weak serotonergic activity in the brain. Low levels of 5-HIAA occur in the spring season. The highest level of tryptophan occurs during April and May. Moreover, the tryptophan depletion rate is low during the summer season [58]. Johansson et al. [59] reported that serotonin-related polymorphisms cause SAD. Morning bright light therapy (BLT) is the primary treatment choice for SAD. Serotonin supplementation along with LT exhibits promising results against SAD. Serotonin receptor 5-HT2C agonists and SSRIs are also effective against SAD. Moreover, negative air ionization liberates charged particles in the sleep area, which is effective for the treatment of SAD [58].

    Brain areas and networks impairments in psychiatric disorders

    The cortical and subcortical areas of the human brain make the networks for synchronization of neural activity. These networks are responsible for the operation of different tasks and cognitive control. Aberration in the brain networks is the neurological basis of psychiatric disorders [60]. The neuroimaging study revealed that affected areas in most of the psychiatric disorders are the dorsal anterior cingulate cortex (dACC) and anterior insula. These areas are responses to motivational demands and environmental constraints. The medial PFC, posterior cingulate cortex, precuneus, and lateral parietal cortex are also associated with psychiatric illness. Another important network is frontoparietal network, which comprises lateral prefrontal and intraparietal areas. This network is associated with cognitive tasks, attention, and behavioral activity [61]. Cognitive control is regulated by three distinct brain networks, including the salience network (SN), the fronto-parietal network (FPN: “central-executive”), and the default mode network (DMN). It is suggested that these three networks are associated with psychiatric disorders: the SN is linked with the anterior insula, dorsal anterior cingulate cortex, and subcortical nodes. This network is associated with reward processing regions. The dorsolateral PFC and posterior parietal cortex are the part of FPN. DMN is formed with the medial posterior cingulate cortex, ventromedial PFC, medial temporal lobe, and angular gyrus [60]. Segal et al. [62] studied the heterogeneity of gray matter volume (GMV) on 1294 cases of different psychiatric disorders (ADHD, ASD, BD, schizophrenia, and others). They concluded that person-specific deviations were possible and regional GMV were highly heterogeneous, affecting rate is < 7% of people. PFC dysfunction is implicated with MDD, BD, ADHD, and schizophrenia. Right and left lateral PFC, and dorsolateral PFC are mostly affected areas, and these regions are overlapped in different psychiatric disorders.

    In humans, the limbic system is involved in the regulation of emotion and behavior. It consists of a group of nuclei. These include the cingulate gyrus of the cerebral cortex, the hypothalamus, the fornix, the hippocampus, and the amygdala. Different fiber tracts make the input and output connection with the limbic system. The limbic system is connected with the PFC and association cortex. There are reciprocal and interneuronal communications among the hypothalamus, prefrontal and association cortex, mamillary bodies, anterior thalamic nuclei, cingulate gyrus, entorhinal cortex, fornix, hippocampus, and amygdala. The cingulate gyrus is connected with the parahippocampal gyrus, and it runs along with the corpus callosum. The cingulate gyrus is interconnected with other areas of the limbic system. The main efferent projections move to the anterior thalamic nuclei. Its response regulates autonomic, somatic, or behavioral activities. The hippocampus is another important part, which receives inputs from the entorhinal area of the hippocampal gyrus, and septal nuclei. The outputs project to the entorhinal area, subiculum, and septal nuclei. The septal nuclei receive reciprocal inputs from the hippocampus via the fornix, amygdala, ventral tegmental nuclei, cingulate gyrus, hypothalamus, and preoptic region via the medial forebrain bundle. The efferent connections go to the hippocampus, ventral tegmental nuclei, habenular nucleus, lateral hypothalamus (LH), and preoptic region. Stimulation of the septal area is associated with pleasure feelings. Dopamine is the main NT in this area, which helps in pleasurable sensations. The amygdala receives inputs from sensory areas of the cerebral cortex, temporal cortex, insular parts of the cortex, thalamic nuclei, and ventromedial hypothalamic nuclei. The efferents move to the stria terminalis, anterior hypothalamus, lateral preoptic nucleus, LH, thalamic nuclei, basal nuclei, reticular formation, and parasympathetic cranial nerve nuclei. The amygdala regulates behavioral and autonomic functions. The insula is a part of the cerebral cortex and is associated with the frontal, parietal, and temporal cortex. The rostral end of the insula is part of the limbic system. The hypothalamus is a crucial part for emotional behavior and regulates visceral and somatomotor activity. Different types of stresses such as physical, physiological, mental, and social stresses are important factors in psychiatric disorders. The hypothalamus, amygdala, hippocampus, and PFC are implicated in the stress response and also associated with psychiatric disorders. Increased and sustained activity in the amygdala is related to depression and mental health.

    Shift work and jet lag disrupt circadian clock functions in the SCN and hamper the functions of the HPA axis. Li and Androulakis [63] studied the effect of shift work and jet lag on the circadian clock and the activity of the HPA axis. They observed that light sensitivity varies from individual to individual. Male subjects and younger individuals have good light sensitivity, while female and elderly people are less sensitive to light. They also predicted that diversity and flexibility are varied along with altered light schedules among the individuals. Transient shift work and jet lag affect the functions of the HPA axis in all individuals. However, some individuals are well tolerated and resynchronize the SCN (core and shell)-HPA axis activity; others are less tolerable and show a negative impact. Stress response stimulates the HPA axis and increases plasma cortisol levels. The paraventricular nucleus (PVN) of the hypothalamus releases corticotropin-releasing hormone (CRH), which stimulates the adrenal cortex through pituitary ACTH. The stress response also activates the hippocampus and PFC. Glucocorticoid receptors are present in the hippocampus and induce stress-related activity. Kalsbeek and Buijs [64] reported that SCN also controls the activity of thyrotropin-releasing hormone (TRH), and gonadotropin-releasing hormone (GnRH) secreting neurons. These hormones regulate thyroid and gonadal functions.

    Circadian rhythm: an overview

    On our planet, all organisms are adapted to the 24-hour light-dark cycle, which is familiar as the circadian (circa = about; dies = day) rhythm. Many physiological and behavioral activities are maintained by the circadian rhythm (Figure 1). Endogenous physiological events are rhythmically coordinated by the internal clock. Hypothalamic SCN acts as the master clock that synchronizes with the environmental light-dark cycle. SCN receives impulse (photic signal) from the non-visual intrinsically photoreceptive retinal ganglion cells (ipRGCs) through the retinohypothalamic tract (RHT) (Figure 1). The ipRGCs express blue light (~480 nm) sensitive photopigment, known as melanopsin [6567]. On exposure to light, ipRGCs propagate signals to the SCN through the RHT, which releases glutamate as an excitatory NT. Another NT is pituitary adenylate cyclase-activating polypeptide (PACAP). Glutamatergic and PACAPergic neurons are co-localized with GABAergic inhibitory neurons. Light-induced activation of the SCN increases Ca2+ influx and activates the intracellular signaling cascade [68].

    The relationship between circadian disruption and promotion of psychiatric disorders. The upper portion of the figure represents the regular functions of circadian rhythm and the lower portion of the figure shows how circadian dysfunction is associated with psychiatric diseases. CRSDs: circadian rhythm sleep disorders; RGC: retinal ganglion cells; RHT: retinohypothalamic tract; SCN: suprachiasmatic nucleus; Vipr2: vasoactive intestinal peptide receptor 2

    SCN also receives non-retinal inputs. These projections come from the median raphe nucleus and intergeniculate leaflet (IGL) of the thalamus via the geniculohypothalamic tract. IGL neurons convey both photic and non-photic information to the SCN. These connections release GABA, neuropeptide Y (NPY), enkephalin, and neurotensin as the NT. The neurons from the median raphe nucleus are exclusively serotonergic. These inputs reset the SCN clock in response to behavioral and locomotor activity [69]. Ni et al. [70] reported that SCN receives non-photic inputs from locus coeruleus and periaqueductal gray in tree shrews. The SCN also receives projections from the ventromedial hypothalamus (VMH), arcuate nucleus (ARC), medial preoptic area (MPOA), paraventricular thalamus (PVT), and PVN of the hypothalamus [71]. The neurons of this projection release cholecystokinin (CCK) as a NT [72]. The vasoactive intestinal peptide (VIP)ergic and CCKergic secondary projections may be associated with motivational and behavioral activity. Dopaminergic neurons from the ventral tegmental area (VTA) send the impulse to the SCN and regulate photoentrainment and feeding-related activity [69].

    Another important factor is steroid hormones. Different steroid hormones (testosterone, dihydrotestosterone, and estradiol) can cross the blood-brain barrier. The SCN expresses androgen receptor (AR), and estrogen receptors (ER) α and β [69]. ARs are expressed in the ventral SCN core. Testosterone/dihydrotestosterone interacts with ARs and then binds with androgen response elements on several core clock genes. This association also occurs on the negative regulator of Per1. Regulation of androgen-mediated transcriptional activity occurs in to dose-dependent manner [73]. ERs appear in the SCN shell and ERβ is predominant. Estrogen-ER complexes bind with estrogen response elements on the core clock genes, particularly Per2, and circadian locomotor output cycle kaput (Clock) [74, 75]. Thus, humoral factors appear in the SCN and regulate the expression of core clock genes.

    SCN sends output signals to the hypothalamus, brainstem, pituitary gland, pineal gland, locus coeruleus, thalamic PVN, and other areas of the brain. SCN secretes a variety of NTs, including acetylcholine (Ach), glutamate, NPY, serotonin, norepinephrine, VIP, arginine vasopressin (AVP), GABA, and gastrin-releasing peptide (GRP). These NTs regulate the sleep-wake cycle, neuro-endocrine functions, melatonin release, and others. The rhythmic expression of clock genes controls the SCN-mediated functions. Arrhythmic expression of clock genes is associated with a variety of pathologies, including cancers, neurodegenerative diseases, and psychiatric disorders [68, 76].

    The SCN sends the VIP, AVP, and prokineticin 2 (PK2)-expressing neurons to the adjacent hypothalamic and thalamic nuclei. The projections move to the subparaventricular zone, PVN of the hypothalamus, dorsomedial hypothalamus, VMH, anterior hypothalamus, posterior hypothalamus, ARC, ventromedial preoptic nucleus, ventrolateral preoptic nucleus, MPOA, periventricular nucleus, lateral habenula, lateral septum, stria terminalis, organum vasculosum of the lamina terminalis [69].

    GABAergic neurons from the SCN to the dorsomedial hypothalamus inhibit the activity of sleep-promoting ventrolateral preoptic neurons. Glutamatergic projections to the dorsomedial hypothalamus activate wake-promoting hypocretin neurons in the LH. Hypothalamic regions directly or indirectly activate the reticular activating system and cerebral cortex to sustain wakefulness. Thus SCN regulates the sleep-wake cycle. Impairments in SCN activity promote sleep problems. Hypophagic or hyperphagic responses sometimes appear in psychiatric disorders. There is a relationship between the SCN and feeding activity. The SCN-ARC circuit may regulate NPY and agouti-related peptide release. The SCN projections also regulate leptin receptor expression. These activities have an impact on feeding behavior [77]. Light-dark cycle has a role in aggressive behavior. The SCN-VIP neurons send impulses to the GABAergic neurons of the subparaventricular zone that is connected with VMH. The circadian active phase increases aggressiveness. However, the inactive phase reduces the activity of the vesicular GABA transporter and decreases aggressive behavior [78]. Thus, circadian disruption may increase aggressive behavior. The SCN is indirectly connected with different regions of the brain that regulate arousal, motivation, mood, and behavior. Projections to the VTA, locus coeruleus, dorsal raphe nucleus, and nucleus accumbens via the dorsomedial hypothalamus, lateral habenula, MPOA, and thalamus regulate different neurological activities [7981]. These connections may be associated with behavioral activity.

    The SCN secretes different factors that can pass through the mesh capsule and act on the surrounding regions. These factors also enter into the CSF through the third ventricle. These diffusible factors include VIP, AVP, GRP, PK2, transforming growth factor-α (TGF-α), and cardiotrophin-like cytokine (CLC). The SCN can also release these factors to the portal system that is directly connected to the vascular bed in the organum vasculosum of the lamina terminalis. CLC receptors are located on neurons adjacent to the third ventricle and passes to the CSF. TGF-α receptors are present on the PVN that is associated with neuroendocrine functions. These factors show paracrine effects to the target regions of the brain [69]. These diffusible factors are rhythmically appeared and regulate the functions of the hypothalamus and other brain regions. Circadian dysfunction alters the activity of the SCN and may hamper the secretion of these factors. Thus, there is a chance of disorganized brain functions.

    The molecular activity of the circadian clock is operated through transcriptional-translational feedback loops (TTFL). Different gene products of the circadian clock operate the TTFL process. Two proteins, CLOCK and brain and muscle ARNT-like protein 1 (BMAL1) form heterodimers. CLOCK-BMAL1 complex interacts with DNA at the E-boxes and upregulates the transcriptional activity of Period (Per1, Per2, and Per3) and Cryptochrome (Cry1 and Cry 2) genes. PER and CRY proteins accumulate in the cytoplasm and translocate into the nucleus followed by the formation of heterodimer. PER-CRY heterodimer represses CLOCK-BMAL1-mediated transcriptional activity. Takahashi suggested that in mice, CLOCK-BMAL1-induced activation appears in the early morning but the PER-CRY complex exerts repressive effects in the evening/night [82]. On the other hand, reverse erythroblastosis viral oncogene homolog α (REV-ERB α) and RORα show contrastive effects. REV-ERB α promotes Bmal1 expression, whereas RORα represses Bmal1 transcription [83].

    The risk of artificial light exposure at night is high in shift work, night work, social and acute jet lag. Light exposure at night causes phase shifting and alters the expression of core clock genes. Aberrant light exposure causes circadian clock dysfunction that disrupts different physiological rhythms, such as the sleep-wake cycle, cortisol, and melatonin secretion [84]. SCN regulates melatonin secretion from the pineal gland during normal photo-periodic activity. Exposure to light at night suppresses melatonin release. In the absence or low concentration of melatonin at night, cells switch on their activities in “day-mode” instead of “night mode”. The results are deregulation of gene expression, metabolic dysfunction, poor endogenous protective activity, and impaired behavior of the cells. Secretion of glucocorticoid occurs in the circadian rhythm by regulating the activity of the HPA axis. Glucocorticoid peak appears in the morning just before awakening and decreases throughout the day [85]. Circadian rhythm dysfunction alters the pattern of glucocorticoid release. Glucocorticoid regulates several physiological functions and is also regarded as a stress hormone. Dedovic and Ngiam [86] reported that impaired glucocorticoid secretion exaggerates MDD. Another report suggested that light-induced dysfunction of the HPA axis causes sleep disorders and increases the prevalence of cortisol-associated mood disorders [87]. Exposure to light at night disturbs the functions of ipRGC projections to the brain and may affect mood [88]. Melanopsin knockout mice have no impact on depressive responses on exposure to light at night [89].

    Circadian rhythm disruption and psychiatric diseases: a complex relationship

    Shift work, night work, acute and chronic jet lag, and exposure to artificial light at night are the intensive factors for circadian rhythm disruption (Figure 1). Light pollution is a serious issue in different metro cities of various countries. In recent years, many people are involved in shift work and night work. They are also facing jet lag problems. These people are in the risk zone for the development of psychiatric disorders. There is a close relationship between circadian rhythm dysfunction and psycho-behavioral disorders. The expression of core clock genes is under the control of circadian rhythm, while circadian rhythm dysfunction causes physiological, metabolic, and behavioral problems, resulting in the progression of psychiatric disorders (Figure 1). Circadian rhythm dysfunction promotes sleep disorders and alters the biomarkers of different physiological rhythms. Sleep problems appear as an early symptom of psychiatric disorders. Circadian rhythm disruption deregulates melatonin rhythm and sleep-wake cycle, which are associated with different psychiatric disorders, including SAD, BD, unipolar disorder, mood problems, schizophrenia, ASD, and ADHD (Figure 1) [9092].

    Schizophrenia is a well-known mental disorder with some symptomatic features, including delusions, hallucinations, movement disorders, anhedonia, alogia, and cognitive failure. Circadian rhythm sleep disorders (CRSDs) can promote schizophrenia [93, 94]. A report is also available on the arrhythmic expression of genes in post-mortem brain tissues of schizophrenic patients [95]. Monteleone et al. [96] reported that disruption of melatonin rhythm is associated with schizophrenia. Circadian rhythm disruption and phase shifting can uncouple the melatonin rhythm with the environmental light-dark cycle, leading to sleep disorder and the progression of schizophrenia [97]. Elevated cortisol level was observed in schizophrenic patients, which occurs due to hyperactivity of the HPA axis and also indicates the physiological stress during schizophrenia [98]. A specific gene, early gene growth response 3 (Egr3) is associated with circadian disruption and schizophrenia [99]. Egr3-deficient mice showed the characteristics of schizophrenia [100]. Mutation or altered expression of RORβ, Per2, Per3, Npas2, Clock, and Cry1 genes can develop schizophrenia [101, 102]. VIP is an essential NT in the CNS, particularly in the cortical interneurons. Abnormalities in VIP levels or its functions promote schizophrenia. Reduction in VIP levels in the prefrontal and orbitofrontal cortices occurs in schizophrenia. Despite VIP, the activity of VIP receptors is also important. Vasoactive intestinal peptide receptor 2 (VIPR2) knockout mice showed impaired hippocampal-associative memory deficit, irregular activity of the HPA axis, and cortisol secretion [103]. Alternatively, Duplication of the Vipr2 gene increases the risk of schizophrenia [104]. This duplication increases the expression of VIPR2; the result is the exaggeration of VIPR2 signaling and increased cAMP levels at the neuronal cells. Thus, VIPR2 signaling has a role in the pathogenesis of schizophrenia. These effects can progress schizophrenia and VIPR2 can be the potential target for the development of anti-psychiatric drugs.

    Shift work influences sleep problems, anxiety, and mood changes. Imposed of night shift work to the day shift workers promotes sleep problems and anxiety [7]. Jet lag can influence the appearance of sleep disruption, anxiety, and dysphoric mood [105]. Nurses are facing psychiatric problems as they follow rotating shift schedules. They showed higher scores in anxiety studies when measured on the Hospital Anxiety and Depression Scale [106]. Animal experiments support the link between circadian disruption and anxiety. Anxiety-like behavior appeared when the adult rats were kept in constant light [107]. Genetic study indicates that improper expression of core clock genes has a relation to anxiety-like disorders. Overexpression of the Clock gene increases anxiety symptoms and ClockΔ19 mutation in mice decrease anxiety. Alternatively, anxiety-related problems are more common in Per1 and Per2 deficient mice. Suppression of Per1 and Per2 expression in the nucleus accumbens causes anxiety [108]. Disruption of circadian rhythms by exposure to constant light (20 h light-dark cycle) causes shortening of dendritic length and poor complexity of neurons in the prelimbic PFC in mice; the result is the exaggeration of anxiety [109].

    Deregulation of the circadian rhythm increases the risk of MDD, which is characterized by feelings of sadness and irritability, sleep deprivation (SD), loss of sexual desire, poor appetite, slowing of speech, and suicidal thoughts [110]. Several studies indicated a clear relationship between shift work and depression [111113]. A meta-analysis study was done by Lee et al. [112], and they reported that night shift work increases a 40% risk of depression compared to daytime workers. Ohayon and Hong [114] reported that the prevalence of MDD is more common in shift workers. A cohort study on night shift workers in Brazil indicated that females are mostly affected by MDD in comparison to males [115]. SD has an impact on MDD. The phase of rapid eye movement sleep increases due to decreased latency, whereas there is a marked deprivation in slow wave sleep. SD, particularly slow-wave sleep disrupts the secretory pattern of melatonin and cortisol, and also alters (increases) nocturnal body temperature; these factors can advance MDD [116]. A marked reduction in the expression pattern (amplitude and phase) of core clock genes had been observed in the post-mortem brain sample of MDD patients [117].

    Several experimental studies on animal models indicate the association between circadian rhythm disruption and depression. Bmal1-knockdown mice showed depressive behavior during the impaired light-dark cycle [118, 119]. Nighttime light (dim light) exposure induces depression. Exposure to light at night in female Siberian hamsters alters the expression of clock proteins and cortisol rhythm [120]. Female hamsters kept in experimental jet lag condition exhibit neuroinflammation, decreased density of the dendritic spine in the hippocampus, and symptoms of depression [121]. The application of TNF-α inhibitor is protective against depressive-like behavior [122]. Experimental studies on male and female mice established that circadian dysfunction is associated with depressive-like disorders [123]. In this experiment, light exposure at night decreased vascular endothelial growth factor A (VEGF-A) in the hippocampus in both male and female mice. On the other hand, VEGF receptor 1 (VEGFR1) and IL-1β expression increased in female mice, while BDNF expression decreased in male mice. Moreover, acute exposure to dim light at night caused circadian dysfunction and altered the expression of clock genes [123]. Constant light exposure to rats for 8 weeks showed an arrhythmic pattern of melatonin and cortisol secretion, and depressive behavior [107]. Application of melatonin receptors’ agonist (agomelatine) on rats restored melatonin and cortisol rhythms and decreased depressive symptoms after exposure to chronic constant light [124, 125].

    Walker et al. [123] indicated that LT could improve MDD. Applications of LT, wake therapy (WT), and antidepressants, such as SSRIs, are effective for the treatment of depression [126]. Treatment with LT, SSRIs, and melatonin receptors agonists make the phase advancement of body temperature, melatonin, and cortisol rhythms that improve depressive symptoms [127129]. WT increases the amount of slow-wave sleep and decreases REM sleep, which gives potential benefits against MDD [130].

    Another psychiatric problem is BD, where mania and depressive mood have cyclically appeared with a definite interval. Jet lag-related circadian rhythm dysfunction is associated with BD [131]. The transmeridian journey from east to west causes depression, while west to east develops mania [132]. Meta-analysis of clinical studies indicated that circadian rhythm disruption promotes BD [133]. Bellivier et al. [134] reported that clock genes are associated with BD. Polymorphisms in clock genes may increase the possibility of BD development. Le-Niculescu et al. [135] had shown the relationship between polymorphisms in core clock genes and the development of BD. Mutation in the Clock gene promotes mania-like symptoms. ClockΔ19 mutant mice showed bipolar mania, and administration of dopamine improved mania-like behaviors [136]. Moon et al. [137] suggested that the determination of clock gene expression and cortisol rhythm can be considered as the biomarkers in BD patients. Stabilization of sleep and circadian rhythm effectively improve BD [134]. Like MDD, LT is also effective in BD [138]. Therapeutic administration of lithium counteracts the fast-running circadian clock in BD patients and stabilizes circadian rhythmicity, leading to improvement in BD symptoms [139]. A report from different literature surveys revealed that impaired protein kinase C activity alters the neuronal signaling in the frontal and limbic area of the brain, leading to BD [140]. Cry and Npas2 genes are associated with unipolar disorder. The delayed sleep phase is common in patients with unipolar disorder. Affected individuals show higher sleep latency, late sleep off, and longer duration of sleep time [141].

    Children with ASD exhibit sleep problems and circadian rhythm dysfunction. Moreover, circadian dysfunction also promotes sleep disorders. So, there is a close relationship between sleep problems and ASD [142]. Prolonged sleep latency, frequent waking at night, alterations in sleep pattern (low levels of REM sleep and higher percentage of slow-wave sleep), unusual morning arousal, and reduction in total sleep time are common features of ASD [143, 144]. Abnormal cortisol levels, higher serotonin levels, and high levels of melatonin in the daytime are associated with ASD [145]. Melatonin has a positive role in the sleep cycle and also regulates the clock gene expression in the SCN. Circadian misalignment deregulates melatonin rhythm, which also affects SCN functions. Melke et al. [146] reported that low levels of serum and urinary melatonin were observed in ASD individuals. The PVN secretes CRH, which regulates the activity of the HPA axis. The PVN receives neural input from the hippocampus, amygdala, PFC, and SCN. Circadian dysfunction alters SCN activities and CRH release from PVN, resulting in abnormalities in cortisol rhythm [147]. Serotonin is essential for the development of the forebrain in the fetus. Synthesis of serotonin depends on the sufficient concentrations of tryptophan during the pregnancy period. Abnormal serotonin concentration is detrimental for the development of ASD. There is a relationship between disturbance in the serotonergic system and circadian dysfunction; the result is the progression of ASD [148]. Abnormalities in clock gene expression cause ASD. The timing deficit in ASD may be associated with the structural and functional activity of the clock-related genes [149]. Experiments on mice model systems showed that NPAS2 is involved in the regulation of REM sleep and total sleep time. Impairment in NPAS2 increases the risk of ASD [150]. Mutation in NPAS2 (cytosine/thymine), and PER1 (cytosine/guanine SNP rs885747; cytosine/adenine SNP rs6416892) have a role in ASD development. Substitutions of proline/alanine at amino acid 1228 in PER2 and arginine/glutamine at amino acid 366 in PER3 are related to the pathogenesis of ASD. The impaired function of RORα alters the expression and function of Bmal1, which might be associated with ASD [145, 151].

    Circadian dysfunction and sleep disorders (short sleep period and evening chronotype) are associated with ADHD. Huang et al. [152] reported that circadian dysfunction affects dopamine levels and the development of dopaminergic neurons, leading to the progression of ADHD. Melatonin and cortisol are the most important factors for the regulation of neurophysiological activities. Arrhythmic secretion of melatonin and cortisol occurs in ADHD individuals. The peak of melatonin and cortisol decreases in ADHD [153]. SNP in CLOCK locus causes ADHD. Animal experiments revealed that the CLOCK gene is involved in circadian sleep disorder and ADHD [154]. Genetic variation in the 3՛ untranslated region (UTR) of CLOCK gene may increase the susceptibility to ADHD development [155]. Baird et al. [156] observed arrhythmic expression of BMAL1 and PER2 genes in the ADHD group.

    SAD/winter depression is a recurring depressive behavior that occurs annually in the winter and spontaneously disappears in the spring/summer [54]. Seasonal change alters the natural light-dark cycle that causes SAD. In the North Pole region, short day lengths in winter can initiate dysthymia. Oppositely, prolonged daylight in the equator region during summer starts euthymia. SAD patients showed advanced sleep phase syndrome (ASPS) and delayed sleep phase disorder/syndrome (DSPS) [54]. In ASPS, the sleep episode starts earlier than the desired clock time, leading to early sleep onset and early awakening. DSPS occurs when the onset of sleep and awakening is delayed than the desired clock time. Seasonal changes affect the natural light-dark cycle, causing a phase shift of the circadian clock that alters NT release (particularly serotonin), functions of neural circuits, melatonin, and cortisol rhythm. Excess melatonin in winter increases sleepiness. The collective effects are mood-related problems. LT and antipsychotic drugs can improve SAD [157]. In humans, SAD can develop after the formation of the variant protein NPAS2 471 Leu/Ser [158]. Polymorphism of PER3 (P415A/H417R) is associated with advanced sleep phase and SAD [159].

    Regulation of sleep-wake cycle is a very complex system. During the daytime, high levels of NTs decrease sleep pressure and maintain awaking state. At night, sleep pressure reaches maximum. Orexigenic peptide orexin and its receptors are involved in the regulation of the sleep cycle. The neurons of LH release orexin A and orexin B. These peptides stimulate cholinergic and monoamine (serotonin, dopamine, NA, and histamine) secreting neurons in the hypothalamus and the brain stem for arousal response and awakening activities [160]. During daytime SCN continuously activates the LH and maintains wakefulness. Monoaminergic pathways also inhibit the ventrolateral preoptic area (VLPO) for suppressing the sleep cycle. At night, VLPO secrets sleep-inducing agents, which inhibit monoaminergic pathways in the LH for inducing sleep. Circadian disruption affects these pathways and disturbs the sleep-wake cycle, leading to the progression of psychiatric disorders.

    The SCN controls the activity of the pineal gland and regulates melatonin synthesis and secretion. Melatonin receptors are present in the SCN, hypothalamus, substentia nigra, hippocampus, cerebellum, VTA, and nucleus accumbens [161]. Melatonin has a crucial role in brain functions and shows a sleep-inducing effect. Circadian rhythm disruption hampers melatonin rhythm and also causes SD. SD and circadian rhythm disruption occur in schizophrenic patients. They showed impaired melatonin rhythm [162]. Similarly, Naismith et al. [163] reported that patients with MDD have low salivary melatonin. Thus, there must be a link among circadian rhythm disruption, impaired melatonin rhythm, sleep disorders, and psychiatric disorders. Patients with BD and MDD also showed sleep disruption that may intensify the psychiatric disorders. Impaired BDNF level is associated with psychiatric disorders. BDNF can induce spontaneous wakefulness, leading to impaired neural functions and the progression of BD and MDD [164]. Glucose is an essential metabolite for neural cells. SD also affects glucose metabolism in the PFC [165]. Thus, sleep problems and psychiatric disorders have a bidirectional relationship.

    Shank proteins (plasticity-associated synaptic proteins) are involved in synaptic plasticity. These proteins follow a circadian rhythm. Light affects the expression of Shank proteins. Sarowar et al. [166] suggested that schizophrenia is associated with impaired expression of Shank3. Cell adhesion molecule neurexin promotes synaptogenesis, synaptic transmission, and regulation of sleep quality [167]. Defects in neurexin gene expression in circadian dysfunction cause ASD and schizophrenia.

    Circadian rhythm critically regulates the HPA axis activity. In the gestational period, glucocorticoid crosses the placenta and regulates neural development in the fetus. Circadian dysfunction acts as a stressor, and prenatal stress disturbs the expression of core clock genes in the SCN. Animal experiments also revealed that stress response in mothers causes impaired expression of Clock and Rev-erb α genes in the peripheral tissues (adrenal and liver). Additionally, hyperactivity of the HPA axis reduces the sensitivity of negative feedback of glucocorticoids in adult mice. Thus, stress response or circadian dysfunction affects the HPA axis and alters glucocorticoid rhythm. These effects on mother may increase the risk of development of ASD in the child during the postnatal period [165]. Disruption of HPA axis activity advances MDD, BD, and schizophrenia [168].

    Chronotype is a factor in psychiatric disorders. MDD, BD, SAD, and anxiety disorders are associated with evening chronotype [169]. Shift work, jet lag, and night work have detrimental effects on the evening chronotype. These events are also the cause of circadian misalignment. A diffusion tensor imaging study indicated that the integrity of white matter in the frontal and temporal lobe, cingulate gyrus, and corpus callosum in the evening chronotype has some differences compared to the morning chronotype [170]. These differences are probably associated with psychiatric disorders in the evening chronotype. Thus, there is a complex relationship among chronotype, circadian dysfunction, and psychiatric disorders.

    NTs like serotonin, dopamine, and NA are involved in pathologies of psychiatric disorders [171]. Serotonin is essential for the regulation of circadian rhythm, mood, anxiety, and cognitive functions [172]. The serotonergic system also regulates the HPA axis and cortisol release. Disruption of circadian rhythm causes disintegration in serotonergic activity and neural functions, as well as melatonin and cortisol secretion, resulting in an increased risk of MDD [173]. Depression is also associated with impaired functions of GABAergic and glutamatergic systems [174]. Moreover, low levels of serotonin, dopamine, and norepinephrine are correlated with depression [175]. In schizophrenia, impaired working memory and cognitive functions occur due to a deficit of glutamatergic NMDA receptors in the pyramidal neurons of the PFC [176]. Inhibitory GABA signaling in the pyramidal interneuron network is essential for working memory. Impaired GABAergic transmission in this area causes working memory deficits in schizophrenic subjects [177]. Kumar et al. [178] reported that low levels of GABAergic activity increase dopamine levels, which deregulate the activity of the dopaminergic system, causing behavioral problems in schizophrenia [179]. Moreover, excess dopamine secretion causes the progression of schizophrenia. In ASD, an imbalance occurs between inhibitory NT GABA and excitatory NT glutamate [180]. The glutamatergic projections from the frontal area to the striatum of basal ganglia may increase motor activities in ASD [181]. Mutation in glutamatergic receptor genes GRIN2A and GRIN2B promotes ASD [180]. Abnormalities in dopamine, norepinephrine, and serotonin secretion are linked with ASD and are correlated with sleep disturbance, mood disorders, and behavioral problems [182, 183]. Marotta et al. [180] reported that ASD patients had reduced dopamine levels in the PFC and nucleus accumbens. Impaired dopamine secretion in the nigrostriatal pathway and mesocorticolimbic area causes motor dysfunction and behavioral problems in ASD subjects, respectively. Thus, evidence-based studies revealed that there is a complex relationship among circadian disruption, neural activities, neuroendocrine dysfunction, and psychiatric disorders.

    Treatment strategy

    Treatments of psychiatric diseases decrease symptoms and provide better quality of life. Applications of antipsychotic, antidepressant, and mood-stabilizing drugs, such as haloperidol, perazine, lithium, valproate acid, clomipramine, fluoxetine, and SSRIs give positive results against psychiatric diseases. Escitalopram, venlafaxine, benzodiazepines, and buspirone are the prescribed drugs for anxiety disorder [31].

    SSRIs are the first-line drug choice for the treatment of depression, anxiety, and other psychiatric disorders. SSRIs block the reuptake of serotonin, resulting in slow clearance of this monoamine NT. SSRIs increase serotonin levels at the synaptic cleft and promote serotonergic activities in the brain. Elevation of extraneural serotonin concentrations facilitates the binding of serotonin with somatodendritic autoreceptors (5-HT1A) and the autoreceptors of the presynaptic end (5-HT1B). This binding effectively maintains serotonergic neurotransmission. The common SSRIs are paroxetine, mirtazapine, nefazodone, and venlafaxine [184]. MAO inhibitors and SSRIs are used for the treatment of depression. Sometimes, prolonged use of SSRIs increases the risk of obesity [184].

    Köhler et al. [185] reported that anti-inflammatory agents give relief against depressive symptoms. SSRIs decrease IL-1β, IL-6, and TNF-α in the peripheral circulation [186]. Baumeister et al. [187] indicated that clomipramine and fluoxetine reduce IL-6, TNF-α, and IFN-γ levels. Application of TNF inhibitor in female Siberian hamsters prevents dendritic spine formation in the hippocampus and ameliorates depressive-like behavior [120]. However, anti-inflammatory drugs can be used as an adjuvant in association with antipsychotic drugs for the better treatment of psychiatric diseases [188]. SSRIs, serotonin and NA reuptake inhibitors (SNRIs), agomelatine (MT1 and MT2 agonists), and 5-HT2C antagonists improve MDD. They help in phase advancement that is effective in the treatment process.

    The pharmacological applications of lithium chloride for the treatment of circadian disruption show a significant effect on the resynchronization of circadian rhythm by altering the activity of intracellular kinases, including glycogen synthase kinase-3beta (GSK-3beta). The regulation of the circadian clock at the molecular level reduces the symptoms of mania, and depression and improves behavioral activity [6]. Lithium-induced normalization of circadian rhythms is essential for the treatment of BD. Lithium slows down the circadian dysfunction and ameliorates the symptoms of BD [189].

    Sleep problem is associated with psychiatric disorders. The application of melatonin improves sleep problems and increases sleep quality. FDA also approved melatonin agonists for the treatment of insomnia. The melatonin agonist agomelatine acts as a zeitgeber and helps to establish regular sleep cycle. Agomelatine also shows antidepressant properties [190]. Administration of agomelatine to rats prevents depressive-like behavior even after exposure to constant light [124, 125]. The application of agomelatine on human subjects improves the rest-activity cycle and sleep quality, leading to the improvement of depressive symptoms [191].

    Other pharmacological agents, such as NO synthase 1 adaptor protein (NOS1AP), a specific anti-psychiatric drug, are applicable for the treatment of BD and schizophrenia [192]. The allelic variants of FK506-binding protein 51 (FKBP51) reduce depression and anxiety disorders [193].

    Psychotherapy such as CBT can improve panic disorder, phobias, and major depression. This treatment regulates attitudes and behaviors and increases intellectual activity and cognitive response [194, 195].

    Chronotherapy is based on the regulation of circadian rhythms. It refers to non-pharmaceutical treatment that depends on exposure to external environmental stimuli to regulate the biological rhythms [196]. Chronotherapy includes SD or WT, sleep phase advance (SPA) therapy, and LT or BLT [197]. Exposure to white light (intensity 2,000–10,000 lux) for 30–120 minutes per day is given in BLT. Most of the time, treatment duration is 2–4 weeks. LT is based on neurobiological principles, and it is the effective treatment choice for SAD [198]. Khalifeh [199] reported that BLT is the promising treatment choice for SAD. LT is also effective for the treatment of non-seasonal depression and mood disorders [200, 201]. LT is effective for the treatment of SAD and non-seasonal depression. Tao et al. [202] conducted a meta-analysis on randomized controlled trials (RCTs) of depression study. They analyzed 23 RCTs with 1120 participants. The result indicated that LT has significant effects to reduce non-seasonal depressive symptoms. Another study on 89 patients (age 60 years or older) with MDD showed improvements in mood, sleep quality, and melatonin levels after BLT [203]. Hizli et al. [204] compared the effects between BLT and BLT with SSRI (fluoxetine). They reported that the application of BLT with SSRI had no extra benefit on depressive symptoms and sleep quality. LT along with antidepressants in unipolar disorder patients is a better choice for the treatment. The application of LT with lithium is also effective in BD patients [200, 201]. BLT in the morning also promotes phase advancement and improves depressive symptoms. Randomized, placebo-controlled clinical trials revealed that BLT in the midday prevents the episode of depression in BD [138]. LT is an alternative choice for patients who refuse, resist, or cannot tolerate any antidepressant drugs [205].

    SD/WT is a process where individuals remain awake for long periods (up to 36 h) to reduce depressive symptoms [206, 207]. WT gives quick response and better efficacy in reducing depressive symptoms [196]. The effect of WT is short lasting and there is a chance of relapse. Casher et al. [208] suggested that WT increases serotonin, dopamine, and norepinephrine levels in the CNS for the improvement of psychiatric problems. Applications of BLT, and WT therapy improve MDD. WT increases the levels of slow wave sleep and decreases the latency of REM sleep, which are effective for the treatment of MDD [7]. A comparative study on MDD patients where one group is taking a combined treatment of WT, LT, and sleep time stabilization and another group is performing exercise therapy; the result indicated that WT, LT, and sleep time stabilization give better results than exercise therapy [209]. Benedetti et al. [210] reported that WT in patients with BD with or without drug resistance history has a remarkable potentiality of therapeutic efficacy.

    Acombined use of BLT, WT, and SPA is used in triple chronotherapy. This therapy can give rapid improvements in depressive symptoms [211]. Moscovici and Kotler [212] applied chronobiologic multistage intervention (CMI) by using LT, SD, and SPA on 12 patients with moderate to severe depression and observed significant improvements in depressive symptoms. Several authors also reported that triple chronotherapy shows promising results for the treatment of depression [211, 213]. Wu et al. [214] also reported that triple chronotherapy along with the application of sertraline and lithium is effective for the treatment of BD.

    Another type of chronotherapy is dark therapy (DT), where patients are kept in a complete dark room at night. Sometimes, blue-blocking sunglasses are used to induce “circadian darkness” [215]. DT is effective for the patient with BD [216]. A clinical trial reported that DT and blocking of blue light exposure prevent bipolar mania [217].

    Recently, gene therapy and stem cell therapy have been applied as newer approaches for the treatment of psychiatric diseases. Both these processes show significant success in mouse models for the treatment of different forms of mental illness. In humans, low levels of the p11 protein expression are associated with depression. p11 protein knockout mice also exhibit depression. Administration of an adeno-associated virus carrying the gene of the p11 protein into the nucleus accumbens restores the expression of p11 protein and reduces the symptoms of depression [218].

    Conclusions

    In recent years, circadian rhythm dysfunction is a common phenomenon in our modern society due to shift work, night work, exposure to bright light at night, and jet lag. Circadian rhythm misalignment adversely affects human health, particularly neural functions, neuroendocrine activity, and metabolic functions. Alterations of circadian rhythms are also associated with neurodegenerative diseases and psychiatric disorders, including schizophrenia, anxiety, depression, MDD, and BD. Circadian disruption deregulates the expression of core clock genes, melatonin and cortisol rhythm, activity of NTs, and function of different areas of the brain. Moreover, physical and physiological stress, neuroinflammation, and neurodegeneration also affect neuronal activity. Collectively, these effects promote psychiatric diseases. Resynchronization of circadian rhythms can improve the symptoms of psychiatric illness. Anti-psychotic drugs, CBT, LT, WT, and family support are beneficial for the treatment of psychiatric disorders. Finally, it can be concluded that public awareness about mental illness and circadian dysfunction is the forward step for combating psychiatric disorders. Activities at the personal level to maintain the circadian rhythm can give fruitful results in the near future.

    Abbreviations

    5-HIAA:

    5-hydroxyindole acetic acid

    5-HT:

    5-hydroxytryptamine

    ADHD:

    attention deficit/hyperactivity disorder

    AR:

    androgen receptor

    ARC:

    arcuate nucleus

    ASD:

    autism spectrum disorder

    BD:

    bipolar disorder

    BDNF:

    brain-derived neurotrophic factor

    BLT:

    bright light therap

    BMAL1:

    muscle ARNT-like protein 1

    CBT:

    cognitive behavioral therap

    CLOCK:

    circadian locomotor output cycle kaput

    CRH:

    corticotropin-releasing hormone

    CSF:

    cerebrospinal fluid

    DT:

    dark therapy

    ER:

    estrogen receptors

    GABA:

    γ-aminobutyric acid

    HPA:

    hypothalamic-pituitary-adrenal

    iNOS:

    inducible nitric oxide synthase

    ipRGCs:

    intrinsically photoreceptive retinal ganglion cells

    LH:

    lateral hypothalamus

    LT:

    light therapy

    MAO:

    monoamine oxidase

    MDD:

    major depressive disorder

    MPOA:

    medial preoptic area

    MS:

    multiple sclerosis

    NA:

    noradrenaline

    NMDA:

    N-methyl-D-aspartate

    NO:

    nitric oxide

    NPY:

    neuropeptide Y

    NTs:

    neurotransmitters

    PFC:

    prefrontal cortex

    PTSD:

    posttraumatic stress disorder

    PVN:

    paraventricular nucleus

    REV-ERB α:

    reverse erythroblastosis viral oncogene homolog α

    SAD:

    seasonal affective disorder

    SCN:

    suprachiasmatic nucleus

    SD:

    sleep deprivation

    SNP:

    single nucleotide polymorphism

    SPA:

    sleep phase advance

    SSRIs:

    selective serotonin reuptake inhibitors

    VIP:

    vasoactive intestinal peptide

    VIPR2:

    vasoactive intestinal peptide receptor 2

    VMH:

    ventromedial hypothalamus

    VTA:

    ventral tegmental area

    WT:

    wake therapy

    Declarations

    Author contributions

    SS: Conceptualization, Data curation, Writing—original draft, Writing—review & editing. DB: Supervision. The authors have agreed to the published version of the manuscript.

    Conflicts of interest

    Dr. Blum is the inventor of the Genetic Addiction Risk Severity (GARS), USA and foreign patents and KB220 Patented products. The other author declare that there is no conflicts of interest.

    Ethical approval

    Not applicable.

    Consent to participate

    Not applicable.

    Consent to publication

    Not applicable.

    Availability of data and materials

    Not applicable.

    Funding

    Not applicable.

    Copyright

    © The Author(s) 2024.

    References

    Vigo D, Thornicroft G, Atun R. Estimating the true global burden of mental illness. Lancet Psychiatry. 2016;3:1718. [DOI] [PubMed]
    Beurel E, Toups M, Nemeroff CB. The Bidirectional Relationship of Depression and Inflammation: Double Trouble. Neuron. 2020;107:234256. [DOI] [PubMed] [PMC]
    Haroon E, Raison CL, Miller AH. Psychoneuroimmunology meets neuropsychopharmacology: translational implications of the impact of inflammation on behavior. Neuropsychopharmacology. 2012;37:13762. [DOI] [PubMed] [PMC]
    Vahia VN. Diagnostic and statistical manual of mental disorders 5: A quick glance. Indian J Psychiatry. 2013;55:2203. [DOI] [PubMed] [PMC]
    Patel SA, Kondratov RV. Clock at the Core of Cancer Development. Biology (Basel). 2021;10:150. [DOI] [PubMed] [PMC]
    Karatsoreos IN. Links between Circadian Rhythms and Psychiatric Disease. Front Behav Neurosci. 2014;8:162. [DOI] [PubMed] [PMC]
    Walker WH 2nd, Walton JC, DeVries AC, Nelson RJ. Circadian rhythm disruption and mental health. Transl Psychiatry. 2020;10:28. [DOI] [PubMed] [PMC]
    Yuan N, Chen Y, Xia Y, Dai J, Liu C. Inflammation-related biomarkers in major psychiatric disorders: a cross-disorder assessment of reproducibility and specificity in 43 meta-analyses. Transl Psychiatry. 2019;9:233. [DOI] [PubMed] [PMC]
    Boeschoten RE, Braamse AMJ, Beekman ATF, Cuijpers P, Oppen Pv, Dekker J, et al. Prevalence of depression and anxiety in Multiple Sclerosis: A systematic review and meta-analysis. J Neurol Sci. 2017;372:331341. [DOI] [PubMed]
    Rossi S, Studer V, Motta C, Polidoro S, Perugini J, Macchiarulo G, et al. Neuroinflammation drives anxiety and depression in relapsing-remitting multiple sclerosis. Neurology. 2017;89:133847. [DOI] [PubMed]
    Matsumoto H, Simmons A, Williams S, Hadjulis M, Pipe R, Murray R, et al. Superior temporal gyrus abnormalities in early-onset schizophrenia: similarities and differences with adult-onset schizophrenia. Am J Psychiatry. 2001;158:1299304. [DOI] [PubMed]
    Gould RA, Mueser KT, Bolton E, Mays V, Goff D. Cognitive therapy for psychosis in schizophrenia: an effect size analysis. Schizophr Res. 2001;48:33542. [DOI] [PubMed]
    Lehman AF, Lieberman JA, Dixon LB, McGlashan TH, Miller AL, Perkins DO, et al.; Steering Committee on Practice Guidelines. Practice guideline for the treatment of patients with schizophrenia, second edition. Am J Psychiatry. 2004;161:156. [PubMed]
    Ibanez-Casas I, Portugal ED, Gonzalez N, McKenney KA, Haro JM, Usall J, et al. Deficits in executive and memory processes in delusional disorder: a case-control study. PLoS One. 2013;8:e67341. [DOI] [PubMed] [PMC]
    Ukai K, Kimura H, Arao M, Aleksic B, Yamauchi A, Ishihara R, et al. Effectiveness of low-dose milnacipran for a patient suffering from pain disorder with delusional disorder (somatic type) in the orofacial region. Psychogeriatrics. 2013;13:99102. [DOI] [PubMed]
    Straub RE, Weinberger DR. Schizophrenia genes - famine to feast. Biol Psychiatry. 2006;60:813. [DOI] [PubMed]
    Khandaker GM, Cousins L, Deakin J, Lennox BR, Yolken R, Jones PB. Inflammation and immunity in schizophrenia: implications for pathophysiology and treatment. Lancet Psychiatry. 2015;2:25870. [DOI] [PubMed] [PMC]
    Goldsmith DR, Rapaport MH, Miller BJ. A meta-analysis of blood cytokine network alterations in psychiatric patients: comparisons between schizophrenia, bipolar disorder and depression. Mol Psychiatry. 2016;21:1696709. [DOI] [PubMed] [PMC]
    Angerhofer CR. A biological review of mental illness: an overview of genetics and pathophysiology of schizophrenia, major depression, and addiction. Biology and microbiology graduate students plan B research projects. 2018;5:20.
    Réus GZ, Fries GR, Stertz L, Badawy M, Passos IC, Barichello T, et al. The role of inflammation and microglial activation in the pathophysiology of psychiatric disorders. Neuroscience. 2015;300:14154. [DOI] [PubMed]
    Pfammatter M, Junghan UM, Brenner HD. Efficacy of psychological therapy in schizophrenia: conclusions from meta-analyses. Schizophr Bull. 2006;32:S6480. [DOI] [PubMed] [PMC]
    Comer RJ. Abnormal psychology. 8th ed. New York: Worth Publishers; 2013.
    Machado S, Lattari E, Kahn JP. Possible mechanisms linking panic disorder and cardiac syndromes. In: Nardi A, Freire R, editors. Panic disorder. Cham: Springer; 2016. pp. 185–202. [DOI]
    Schenberg LC. A neural system approach to the study of the respiratory type panic disorder. In: Nardi AE, Freire RCR, editors. Panic disorder: neurobiological and treatment aspects. Cham: Springer; 2016. pp. 9–76.
    Cosci F. Staging of panic disorder: implications for neurobiology and treatment. In: Nardi A, Freire R, editors. Panic disorder. Cham: Springer; 2016. pp. 113–2. [DOI]
    Eagleson C, Hayes S, Mathews A, Perman G, Hirsch CR. The power of positive thinking: Pathological worry is reduced by thought replacement in Generalized Anxiety Disorder. Behav Res Ther. 2016;78:138. [DOI] [PubMed] [PMC]
    Alomari NA, Bedaiwi SK, Ghasib AM, Kabbarah AJ, Alnefaie SA, Hariri N, et al. Social Anxiety Disorder: Associated Conditions and Therapeutic Approaches. Cureus. 2022;14:e32687. [DOI] [PubMed] [PMC]
    Eaton WW, Bienvenu OJ, Miloyan B. Specific phobias. Lancet Psychiatry. 2018;5:67886. [DOI] [PubMed] [PMC]
    Park JE, Ahn H-N, Jung Y-E. Prevention and Treatment of Trauma- and Stressor-Related Disorders: Focusing on Psychosocial Interventions for Adult Patients. J Korean Neuropsychiatr Assoc. 2016;55:8996. [DOI]
    Schwartz CE, Kunwar PS, Hirshfeld-Becker DR, Henin A, Vangel MG, Rauch SL, et al. Behavioral inhibition in childhood predicts smaller hippocampal volume in adolescent offspring of parents with panic disorder. Transl Psychiatry. 2015;5:e605. [DOI] [PubMed] [PMC]
    More B. Psychiatric diseases and treatment – a review. DJ IJMR. 2016;1:2736. [DOI]
    Hayes AM, Yasinski C, Grasso D, Ready CB, Alpert E, McCauley T, et al. Constructive and Unproductive Processing of Traumatic Experiences in Trauma-Focused Cognitive-Behavioral Therapy for Youth. Behav Ther. 2017;48:16681. [DOI] [PubMed] [PMC]
    Whitney DG, Shapiro DN, Warschausky SA, Hurvitz EA, Peterson MD. The contribution of neurologic disorders to the national prevalence of depression and anxiety problems among children and adolescents. Ann Epidemiol. 2019;29:814.e2. [DOI] [PubMed] [PMC]
    Labrie V, Brundin L. Harbingers of Mental Disease-Infections Associated With an Increased Risk for Neuropsychiatric Illness in Children. JAMA Psychiatry. 2019;76:2378. [DOI] [PubMed]
    Ferrari F, Villa RF. The Neurobiology of Depression: an Integrated Overview from Biological Theories to Clinical Evidence. Mol Neurobiol. 2017;54:484765. [DOI] [PubMed]
    Miller AH, Maletic V, Raison CL. Inflammation and its discontents: the role of cytokines in the pathophysiology of major depression. Biol Psychiatry. 2009;65:73241. [DOI] [PubMed] [PMC]
    de Abreu Costa M, D’Alò de Oliveira GS, Tatton-Ramos T, Manfro GG, Salum GA. Anxiety and stress-related disorders and mindfulness-based interventions: A systematic review and multilevel meta-analysis and meta-regression of multiple outcomes. Mindfulness. 2019;10:9961005. [DOI]
    Ferber SG, Namdar D, Hen-Shoval D, Eger G, Koltai H, Shoval G, et al. The "Entourage Effect": Terpenes Coupled with Cannabinoids for the Treatment of Mood Disorders and Anxiety Disorders. Curr Neuropharmacol. 2020;18:8796. [DOI] [PubMed] [PMC]
    Hritcu L, Ionita R, Postu PA, Gupta GK, Turkez H, Lima TC, et al. Antidepressant Flavonoids and Their Relationship with Oxidative Stress. Oxid Med Cell Longev. 2017;2017:5762172. [DOI] [PubMed] [PMC]
    Rothschild AJ. Treatment for Major Depression With Psychotic Features (Psychotic Depression). Focus (Am Psychiatr Publ). 2016;14:2079. [DOI] [PubMed] [PMC]
    Petralia MC, Mazzon E, Fagone P, Basile MS, Lenzo V, Quattropani MC, et al. Pathogenic contribution of the Macrophage migration inhibitory factor family to major depressive disorder and emerging tailored therapeutic approaches. J Affect Disord. 2020;263:1524. [DOI] [PubMed]
    Muneer A. Mixed States in Bipolar Disorder: Etiology, Pathogenesis and Treatment. Chonnam Med J. 2017;53:113. [DOI]
    Malkoff-Schwartz S, Frank E, Anderson BP, Hlastala SA, Luther JF, Sherrill JT, et al. Social rhythm disruption and stressful life events in the onset of bipolar and unipolar episodes. Psychol Med. 2000;30:100516. [DOI] [PubMed]
    McGuffin P, Rijsdijk F, Andrew M, Sham P, Katz R, Cardno A. The heritability of bipolar affective disorder and the genetic relationship to unipolar depression. Arch Gen Psychiatry. 2003;60:497502. [DOI] [PubMed]
    Young JW, Cope ZA, Romoli B, Schrurs E, Joosen A, Enkhuizen Jv, et al. Mice with reduced DAT levels recreate seasonal-induced switching between states in bipolar disorder. Neuropsychopharmacology. 2018;43:172131. [DOI] [PubMed] [PMC]
    Cuellar AK, Johnson SL, Winters R. Distinctions between bipolar and unipolar depression. Clin Psychol Rev. 2005;25:30739. [DOI]
    Xiong J, Chen S, Pang N, Deng X, Yang L, He F, et al. Neurological Diseases With Autism Spectrum Disorder: Role of ASD Risk Genes. Front Neurosci. 2019;13:349. [DOI] [PubMed] [PMC]
    Smith RG, Reichenberg A, Kember RL, Buxbaum JD, Schalkwyk LC, Fernandes C, et al. Advanced paternal age is associated with altered DNA methylation at brain-expressed imprinted loci in inbred mice: implications for neuropsychiatric disease. Mol Psychiatry. 2013;18:6356. [DOI] [PubMed]
    Morgan CP, Bale TL. Early prenatal stress epigenetically programs dysmasculinization in second-generation offspring via the paternal lineage. J Neurosci. 2011;31:1174855. [DOI] [PubMed] [PMC]
    Singh R, Turner RC, Nguyen L, Motwani K, Swatek M, Lucke-Wold BP. Pediatric Traumatic Brain Injury and Autism: Elucidating Shared Mechanisms. Behav Neurol. 2016;2016:8781725. [DOI] [PubMed] [PMC]
    Shenoy MD, Indla V, Reddy H. Comprehensive Management of Autism: Current Evidence. Indian J Psychol Med. 2017;39:72731. [DOI] [PubMed] [PMC]
    Thapar A, Cooper M, Jefferies R, Stergiakouli E. What causes attention deficit hyperactivity disorder? Arch Dis Child. 2012;97:2605. [DOI] [PubMed] [PMC]
    Nazarova VA, Sokolov AV, Chubarev VN, Tarasov VV, Schiöth HB. Treatment of ADHD: Drugs, psychological therapies, devices, complementary and alternative methods as well as the trends in clinical trials. Front Pharmacol. 2022;13:1066988. [DOI] [PubMed] [PMC]
    Lewy AJ, Emens JS, Songer JB, Sims N, Laurie AL, Fiala SC, et al. Winter Depression: Integrating mood, circadian rhythms, and the sleep/wake and light/dark cycles into a bio-psycho-social-environmental model. Sleep Med Clin. 2009;4:28599. [DOI] [PubMed] [PMC]
    Lam RW, Levitan RD. Pathophysiology of seasonal affective disorder: a review. J Psychiatry Neurosci. 2000;25:46980. [PubMed] [PMC]
    Bunney WE, Bunney BG. Molecular clock genes in man and lower animals: possible implications for circadian abnormalities in depression. Neuropsychopharmacology. 2000;22:33545. [DOI] [PubMed]
    Desan PH, Oren DA, Malison R, Price LH, Rosenbaum J, Smoller J, et al. Genetic polymorphism at the CLOCK gene locus and major depression. Am J Med Genet. 2000;96:41821. [DOI] [PubMed]
    Gupta A, Sharma PK, Garg VK, Singh AK, Mondal SC. Role of serotonin in seasonal affective disorder. Eur Rev Med Pharmacol Sci. 2013;17:4955.
    Johansson C, Smedh C, Partonen T, Pekkarinen P, Paunio T, Ekholm J, et al. Seasonal affective disorder and serotonin-related polymorphisms. Neurobiol Dis. 2001;8:3517. [DOI] [PubMed]
    Menon V. Brain networks and cognitive impairment in psychiatric disorders. World Psychiatry. 2020;19:30910. [DOI] [PubMed] [PMC]
    Downar J, Blumberger DM, Daskalakis ZJ. The Neural Crossroads of Psychiatric Illness: An Emerging Target for Brain Stimulation. Trends Cogn Sci. 2016;20:10720. [DOI] [PubMed]
    Segal A, Parkes L, Aquino K, Kia SM, Wolfers T, Franke B, et al. Regional, circuit and network heterogeneity of brain abnormalities in psychiatric disorders. Nat Neurosci. 2023;26:161329. [DOI] [PubMed] [PMC]
    Li Y, Androulakis IP. Light entrainment of the SCN circadian clock and implications for personalized alterations of corticosterone rhythms in shift work and jet lag. Sci Rep. 2021;11:17929. [DOI] [PubMed] [PMC]
    Kalsbeek A, Buijs RM. Output pathways of the mammalian suprachiasmatic nucleus: coding circadian time by transmitter selection and specific targeting. Cell Tissue Res. 2002;309:10918. [DOI] [PubMed]
    Hannibal J, Hindersson P, Ostergaard J, Georg B, Heegaard S, Larsen PJ, et al. Melanopsin is expressed in PACAP-containing retinal ganglion cells of the human retinohypothalamic tract. Invest Ophthalmol Vis Sci. 2004;45:42029. [DOI] [PubMed]
    Schibler U, Gotic I, Saini C, Gos P, Curie T, Emmenegger Y, et al. Clock-Talk: Interactions between Central and Peripheral Circadian Oscillators in Mammals. Cold Spring Harb Symp Quant Biol. 2015;80:22332. [DOI] [PubMed]
    Hastings MH, Maywood ES, Brancaccio M. Generation of circadian rhythms in the suprachiasmatic nucleus. Nat Rev Neurosci. 2018;19:45369. [DOI] [PubMed]
    Samanta S. Physiological and pharmacological perspectives of melatonin. Arch Physiol Biochem. 2022;128:134667. [DOI] [PubMed]
    Starnes AN, Jones JR. Inputs and Outputs of the Mammalian Circadian Clock. Biology (Basel). 2023;12:508. [DOI] [PubMed] [PMC]
    Ni R, Shu Y, Luo P, Zhou J. Whole-brain mapping of afferent projections to the suprachiasmatic nucleus of the tree shrew. Tissue Cell. 2021;73:101620. [DOI] [PubMed]
    Todd WD, Venner A, Anaclet C, Broadhurst RY, Luca RD, Bandaru SS, et al. Suprachiasmatic VIP neurons are required for normal circadian rhythmicity and comprised of molecularly distinct subpopulations. Nat Commun. 2020;11:4410. [DOI] [PubMed] [PMC]
    Yuan X, Wei H, Xu W, Wang L, Qu W, Li R, et al. Whole-Brain Monosynaptic Afferent Projections to the Cholecystokinin Neurons of the Suprachiasmatic Nucleus. Front Neurosci. 2018;12:807. [DOI] [PubMed] [PMC]
    Karatsoreos IN, Wang A, Sasanian J, Silver R. A role for androgens in regulating circadian behavior and the suprachiasmatic nucleus. Endocrinology. 2007;148:548795. [DOI] [PubMed] [PMC]
    Gery S, Virk RK, Chumakov K, Yu A, Koeffler HP. The clock gene Per2 links the circadian system to the estrogen receptor. Oncogene. 2007;26:791620. [DOI] [PubMed]
    Xiao L, Chang AK, Zang M, Bi H, Li S, Wang M, et al. Induction of the CLOCK gene by E2-ERα signaling promotes the proliferation of breast cancer cells. PLoS One. 2014;9:e95878. [DOI] [PubMed] [PMC]
    Samanta S. A Profound Relationship between Circadian Rhythm Dysfunction and Cancer Progression: An Approach to Exploration. Crit Rev Oncog. 2021;26:141. [DOI] [PubMed]
    Padilla SL, Perez JG, Ben-Hamo M, Johnson CW, Sanchez REA, Bussi IL, et al. Kisspeptin Neurons in the Arcuate Nucleus of the Hypothalamus Orchestrate Circadian Rhythms and Metabolism. Curr Biol. 2019;29:592604.e4. [DOI] [PubMed] [PMC]
    Todd WD, Fenselau H, Wang JL, Zhang R, Machado NL, Venner A, et al. A hypothalamic circuit for the circadian control of aggression. Nat Neurosci. 2018;21:71724. [DOI] [PubMed] [PMC]
    Deurveilher S, Semba K. Indirect projections from the suprachiasmatic nucleus to major arousal-promoting cell groups in rat: implications for the circadian control of behavioural state. Neuroscience. 2005;130:16583. [DOI] [PubMed]
    Luo AH, Aston-Jones G. Circuit projection from suprachiasmatic nucleus to ventral tegmental area: a novel circadian output pathway. Eur J Neurosci. 2009;29:74860. [DOI] [PubMed] [PMC]
    Legoratti-Sánchez MO, Guevara-Guzmán R, Solano-Flores LP. Electrophysiological evidences of a bidirectional communication between the locus coeruleus and the suprachiasmatic nucleus. Brain Res Bull. 1989;23:2838. [DOI] [PubMed]
    Takahashi JS. Molecular components of the circadian clock in mammals. Diabetes Obes Metab. 2015;17:611. [DOI] [PubMed] [PMC]
    Solt LA, Kojetin DJ, Burris TP. The REV-ERBs and RORs: molecular links between circadian rhythms and lipid homeostasis. Future Med Chem. 2011;3:62338. [DOI] [PubMed] [PMC]
    Samanta S, Ali SA. Impact of circadian clock dysfunction on human health. Explor Neurosci. 2022;1:430. [DOI]
    Son GH, Chung S, Kim K. The adrenal peripheral clock: glucocorticoid and the circadian timing system. Front Neuroendocrinol. 2011;32:45165. [DOI] [PubMed]
    Dedovic K, Ngiam J. The cortisol awakening response and major depression: examining the evidence. Neuropsychiatr Dis Treat. 2015;11:11819. [DOI] [PubMed] [PMC]
    Dijk D, Duffy JF, Silva EJ, Shanahan TL, Boivin DB, Czeisler CA. 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. 2012;7:e30037. [DOI] [PubMed] [PMC]
    LeGates TA, Altimus CM, Wang H, Lee H, Yang S, Zhao H, et al. Aberrant light directly impairs mood and learning through melanopsin-expressing neurons. Nature. 2012;491:5948. [DOI] [PubMed] [PMC]
    Hattar S, Kumar M, Park A, Tong P, Tung J, Yau K, et al. Central projections of melanopsin-expressing retinal ganglion cells in the mouse. J Comp Neurol. 2006;497:32649. [DOI] [PubMed] [PMC]
    Pacchierotti C, Iapichino S, Bossini L, Pieraccini F, Castrogiovanni P. Melatonin in psychiatric disorders: a review on the melatonin involvement in psychiatry. Front Neuroendocrinol. 2001;22:1832. [DOI] [PubMed]
    Tordjman S, Chokron S, Delorme R, Charrier A, Bellissant E, Jaafari N, et al. Melatonin: Pharmacology, Functions and Therapeutic Benefits. Curr Neuropharmacol. 2017;15:43443. [DOI] [PubMed] [PMC]
    Lai M, Lombardo MV, Baron-Cohen S. Autism. Lancet. 2014;383:896910. [DOI] [PubMed]
    Bromundt V, Köster M, Georgiev-Kill A, Opwis K, Wirz-Justice A, Stoppe G, et al. Sleep-wake cycles and cognitive functioning in schizophrenia. Br J Psychiatry. 2011;198:26976. [DOI] [PubMed]
    Benson KL. Sleep in Schizophrenia: Pathology and Treatment. Sleep Med Clin. 2015;10:4955. [DOI] [PubMed]
    Seney ML, Cahill K, 3rd JFE, Logan RW, Huo Z, Zong W, et al. Diurnal rhythms in gene expression in the prefrontal cortex in schizophrenia. Nat Commun. 2019;10:3355. [DOI] [PubMed] [PMC]
    Monteleone P, Maj M, Fusco M, Kemali D, Reiter RJ. Depressed nocturnal plasma melatonin levels in drug-free paranoid schizophrenics. Schizophr Res. 1992;7:7784. [DOI] [PubMed]
    Afonso P, Figueira ML, Paiva T. Sleep-promoting action of the endogenous melatonin in schizophrenia compared to healthy controls. Int J Psychiatry Clin Pract. 2011;15:3115. [DOI] [PubMed]
    Coulon N, Brailly-Tabard S, Walter M, Tordjman S. Altered circadian patterns of salivary cortisol in individuals with schizophrenia: A critical literature review. J Physiol Paris. 2016;110:43947. [DOI] [PubMed]
    Zhang R, Lu S, Meng L, Min Z, Tian J, Valenzuela RK, et al. Genetic evidence for the association between the early growth response 3 (EGR3) gene and schizophrenia. PLoS One. 2012;7:e30237. [DOI] [PubMed] [PMC]
    Maple AM, Rowe RK, Lifshitz J, Fernandez F, Gallitano AL. Influence of Schizophrenia-Associated Gene Egr3 on Sleep Behavior and Circadian Rhythms in Mice. J Biol Rhythms. 2018;33:66270. [DOI] [PubMed] [PMC]
    Mansour HA, Talkowski ME, Wood J, Chowdari KV, McClain L, Prasad K, et al. Association study of 21 circadian genes with bipolar I disorder, schizoaffective disorder, and schizophrenia. Bipolar Disord. 2009;11:70110. [DOI] [PubMed] [PMC]
    Johansson A, Owe-Larsson B, Hetta J, Lundkvist GB. Altered circadian clock gene expression in patients with schizophrenia. Schizophr Res. 2016;174:1723. [DOI] [PubMed]
    Fahrenkrug J, Georg B, Hannibal J, Jørgensen HL. Altered rhythm of adrenal clock genes, StAR and serum corticosterone in VIP receptor 2-deficient mice. J Mol Neurosci. 2012;48:58496. [DOI] [PubMed]
    Vacic V, McCarthy S, Malhotra D, Murray F, Chou H, Peoples A, et al. Duplications of the neuropeptide receptor gene VIPR2 confer significant risk for schizophrenia. Nature. 2011;471:499503. [DOI] [PubMed] [PMC]
    Sack RL. Clinical practice. Jet lag. N Engl J Med. 2010;362:4407. [DOI] [PubMed]
    Flo E, Pallesen S, Magerøy N, Moen BE, Grønli J, Nordhus IH, et al. Shift work disorder in nurses--assessment, prevalence and related health problems. PLoS One. 2012;7:e33981. [DOI] [PubMed] [PMC]
    Tapia-Osorio A, Salgado-Delgado R, Angeles-Castellanos M, Escobar C. Disruption of circadian rhythms due to chronic constant light leads to depressive and anxiety-like behaviors in the rat. Behav Brain Res. 2013;252:19. [DOI] [PubMed]
    Spencer S, Falcon E, Kumar J, Krishnan V, Mukherjee S, Birnbaum SG, et al. Circadian genes Period 1 and Period 2 in the nucleus accumbens regulate anxiety-related behavior. Eur J Neurosci. 2013;37:24250. [DOI] [PubMed] [PMC]
    Karatsoreos IN, Bhagat S, Bloss EB, Morrison JH, McEwen BS. Disruption of circadian clocks has ramifications for metabolism, brain, and behavior. Proc Natl Acad Sci U S A. 2011;108:165762. [DOI] [PubMed] [PMC]
    Belmaker RH, Agam G. Major depressive disorder. N Engl J Med. 2008;358:5568. [DOI] [PubMed]
    Lee HY, Kim MS, Kim O, Lee I, Kim H. Association between shift work and severity of depressive symptoms among female nurses: the Korea Nurses' Health Study. J Nurs Manag. 2016;24:192200. [DOI] [PubMed]
    Lee A, Myung SK, Cho JJ, Jung YJ, Yoon JL, Kim MY. Night Shift Work and Risk of Depression: Meta-analysis of Observational Studies. J Korean Med Sci. 2017;32:10916. [DOI] [PubMed] [PMC]
    Booker LA, Sletten TL, Alvaro PK, Barnes M, Collins A, Chai-Coetzer CL, et al. Exploring the associations between shift work disorder, depression, anxiety and sick leave taken amongst nurses. J Sleep Res. 2020;29:e12872. [DOI] [PubMed]
    Ohayon MM, Hong S. Prevalence of major depressive disorder in the general population of South Korea. J Psychiatr Res. 2006;40:306. [DOI] [PubMed]
    Oenning NSX, Ziegelmann PK, Goulart BNGd, Niedhammer I. Occupational factors associated with major depressive disorder: A Brazilian population-based study. J Affect Disord. 2018;240:4856. [DOI] [PubMed]
    Vadnie CA, McClung CA. Circadian Rhythm Disturbances in Mood Disorders: Insights into the Role of the Suprachiasmatic Nucleus. Neural Plast. 2017;2017:1504507. [DOI] [PubMed] [PMC]
    Li JZ, Bunney BG, Meng F, Hagenauer MH, Walsh DM, Vawter MP, et al. Circadian patterns of gene expression in the human brain and disruption in major depressive disorder. Proc Natl Acad Sci U S A. 2013;110:99505. [DOI] [PubMed] [PMC]
    Ben-Hamo M, Larson TA, Duge LS, Sikkema C, Wilkinson CW, Iglesia HOdl, et al. Circadian Forced Desynchrony of the Master Clock Leads to Phenotypic Manifestation of Depression in Rats. eNeuro. 2017;3:ENEURO.023716.2016. [DOI] [PubMed] [PMC]
    Landgraf D, Long JE, Proulx CD, Barandas R, Malinow R, Welsh DK. Genetic Disruption of Circadian Rhythms in the Suprachiasmatic Nucleus Causes Helplessness, Behavioral Despair, and Anxiety-like Behavior in Mice. Biol Psychiatry. 2016;80:82735. [DOI] [PubMed] [PMC]
    Bedrosian TA, Galan A, Vaughn CA, Weil ZM, Nelson RJ. Light at night alters daily patterns of cortisol and clock proteins in female Siberian hamsters. J Neuroendocrinol. 2013;25:5906. [DOI] [PubMed]
    Gibson EM, Wang C, Tjho S, Khattar N, Kriegsfeld LJ. Experimental 'jet lag' inhibits adult neurogenesis and produces long-term cognitive deficits in female hamsters. PLoS One. 2010;5:e15267. [DOI] [PubMed] [PMC]
    Bedrosian TA, Weil ZM, Nelson RJ. Chronic dim light at night provokes reversible depression-like phenotype: possible role for TNF. Mol Psychiatry. 2013;18:9306. [DOI] [PubMed]
    Walker WH 2nd, Borniger JC, Gaudier-Diaz MM, Meléndez-Fernández OH, Pascoe JL, DeVries AC, et al. Acute exposure to low-level light at night is sufficient to induce neurological changes and depressive-like behavior. Mol Psychiatry. 2020;25:108093. [DOI] [PubMed] [PMC]
    Tchekalarova J, Stoynova T, Ilieva K, Mitreva R, Atanasova M. Agomelatine treatment corrects symptoms of depression and anxiety by restoring the disrupted melatonin circadian rhythms of rats exposed to chronic constant light. Pharmacol Biochem Behav. 2018;171:19. [DOI] [PubMed]
    Tchekalarova J, Stoyanova T, Gesheva R, Atanasova M. Agomelatine treatment corrects depressive-like behaviour induced by chronic constant light exposure through modulation of circadian rhythm of corticosterone release. C R Acad Bulgare Sci. 2019;72:53946.
    Germain A, Kupfer DJ. Circadian rhythm disturbances in depression. Hum Psychopharmacol. 2008;23:57185. [DOI] [PubMed] [PMC]
    Leproult R, Onderbergen AV, L'hermite-Balériaux M, Cauter EV, Copinschi G. Phase-shifts of 24-h rhythms of hormonal release and body temperature following early evening administration of the melatonin agonist agomelatine in healthy older men. Clin Endocrinol (Oxf). 2005;63:298304. [DOI] [PubMed]
    Bodinat Cd, Guardiola-Lemaitre B, Mocaër E, Renard P, Muñoz C, Millan MJ. Agomelatine, the first melatonergic antidepressant: discovery, characterization and development. Nat Rev Drug Discov. 2010;9:62842. [DOI] [PubMed]
    Robillard R, Carpenter JS, Feilds K, Hermens DF, White D, Naismith SL, et al. Parallel Changes in Mood and Melatonin Rhythm Following an Adjunctive Multimodal Chronobiological Intervention With Agomelatine in People With Depression: A Proof of Concept Open Label Study. Front Psychiatry. 2018;9:624. [DOI] [PubMed] [PMC]
    Berger M, Calker Dv, Riemann D. Sleep and manipulations of the sleep-wake rhythm in depression. Acta Psychiatr Scand Suppl. 2003;108:8391. [DOI] [PubMed]
    Logan RW, McClung CA. Animal models of bipolar mania: The past, present and future. Neuroscience. 2016;321:16388. [DOI] [PubMed] [PMC]
    Katz G, Knobler HY, Laibel Z, Strauss Z, Durst R. Time zone change and major psychiatric morbidity: the results of a 6-year study in Jerusalem. Compr Psychiatry. 2002;43:3740. [DOI] [PubMed]
    Melo MCA, Abreu RLC, Neto VBL, Bruin PFCd, Bruin VMSd. Chronotype and circadian rhythm in bipolar disorder: A systematic review. Sleep Med Rev. 2017;34:4658. [DOI] [PubMed]
    Bellivier F, Geoffroy P, Etain B, Scott J. Sleep- and circadian rhythm–associated pathways as therapeutic targets in bipolar disorder. Expert Opin Ther Targets. 2015;19:74763. [DOI] [PubMed]
    Le-Niculescu H, Patel SD, Bhat M, Kuczenski R, Faraone SV, Tsuang MT, et al. Convergent functional genomics of genome-wide association data for bipolar disorder: comprehensive identification of candidate genes, pathways and mechanisms. Am J Med Genet B Neuropsychiatr Genet. 2009;150B:15581. [DOI] [PubMed]
    Roybal K, Theobold D, Graham A, DiNieri JA, Russo SJ, Krishnan V, et al. Mania-like behavior induced by disruption of CLOCK. Proc Natl Acad Sci U S A. 2007;104:640611. [DOI] [PubMed] [PMC]
    Moon J, Cho C, Son GH, Geum D, Chung S, Kim H, et al. Advanced Circadian Phase in Mania and Delayed Circadian Phase in Mixed Mania and Depression Returned to Normal after Treatment of Bipolar Disorder. EBioMedicine. 2016;11:28595. [DOI] [PubMed] [PMC]
    Sit DK, McGowan J, Wiltrout C, Diler RS, Dills JJ, Luther J, et al. Adjunctive Bright Light Therapy for Bipolar Depression: A Randomized Double-Blind Placebo-Controlled Trial. Am J Psychiatry. 2018;175:1319. [DOI] [PubMed]
    Gold AK, Kinrys G. Treating Circadian Rhythm Disruption in Bipolar Disorder. Curr Psychiatry Rep. 2019;21:14. [DOI] [PubMed] [PMC]
    Saxena A, Scaini G, Bavaresco DV, Leite C, Valvassori SS, Carvalho AF, et al. Role of Protein Kinase C in Bipolar Disorder: A Review of the Current Literature. Mol Neuropsychiatry. 2017;3:10824. [DOI] [PubMed] [PMC]
    Robillard R, Naismith SL, Rogers NL, Ip TKC, Hermens DF, Scott EM, et al. Delayed sleep phase in young people with unipolar or bipolar affective disorders. J Affect Disord. 2013;145:2603. [DOI] [PubMed]
    Johnson KP, Zarrinnegar P. Autism Spectrum Disorder and Sleep. Child Adolesc Psychiatr Clin N Am. 2021;30:195208. [DOI] [PubMed]
    Polimeni MA, Richdale AL, Francis AJP. A survey of sleep problems in autism, Asperger's disorder and typically developing children. J Intellect Disabil Res. 2005;49:2608. [DOI] [PubMed]
    Buckley AW, Rodriguez AJ, Jennison K, Buckley J, Thurm A, Sato S, et al. Rapid eye movement sleep percentage in children with autism compared with children with developmental delay and typical development. Arch Pediatr Adolesc Med. 2010;164:10327. [DOI] [PubMed] [PMC]
    Lorsung E, Karthikeyan R, Cao R. Biological Timing and Neurodevelopmental Disorders: A Role for Circadian Dysfunction in Autism Spectrum Disorders. Front Neurosci. 2021;15:642745. [DOI] [PubMed] [PMC]
    Melke J, Botros HG, Chaste P, Betancur C, Nygren G, Anckarsäter H, et al. Abnormal melatonin synthesis in autism spectrum disorders. Mol Psychiatry. 2008;13:908. [DOI] [PubMed] [PMC]
    Li S, Kirouac GJ. Sources of inputs to the anterior and posterior aspects of the paraventricular nucleus of the thalamus. Brain Struct Funct. 2012;217:25773. [DOI] [PubMed]
    Takumi T, Tamada K, Hatanaka F, Nakai N, Bolton PF. Behavioral neuroscience of autism. Neurosci Biobehav Rev. 2020;110:6076. [DOI] [PubMed]
    Wimpory D, Nicholas B, Nash S. Social timing, clock genes and autism: a new hypothesis. J Intellect Disabil Res. 2002;46:3528. [DOI] [PubMed]
    Franken P, Dudley CA, Estill SJ, Barakat M, Thomason R, O'Hara BF, et al. NPAS2 as a transcriptional regulator of non-rapid eye movement sleep: genotype and sex interactions. Proc Natl Acad Sci U S A. 2006;103:711823. [DOI] [PubMed] [PMC]
    Goto M, Mizuno M, Matsumoto A, Yang Z, Jimbo EF, Tabata H, et al. Role of a circadian-relevant gene NR1D1 in brain development: possible involvement in the pathophysiology of autism spectrum disorders. Sci Rep. 2017;7:43945. [DOI] [PubMed] [PMC]
    Huang J, Zhong Z, Wang M, Chen X, Tan Y, Zhang S, et al. Circadian modulation of dopamine levels and dopaminergic neuron development contributes to attention deficiency and hyperactive behavior. J Neurosci. 2015;35:257287. [DOI] [PubMed] [PMC]
    Dück A, Reis O, Wagner H, Wunsch K, Häßler F, Kölch M, et al. Clock Genes Profiles as Diagnostic Tool in (Childhood) ADHD—A Pilot Study. Brain Sci. 2022;12:1198. [DOI] [PubMed] [PMC]
    Schuch JB, Genro JP, Bastos CR, Ghisleni G, Tovo-Rodrigues L. The role of CLOCK gene in psychiatric disorders: Evidence from human and animal research. Am J Med Genet B Neuropsychiatr Genet. 2018;177:18198. [DOI] [PubMed]
    Xu X, Breen G, Chen C, Huang Y, Wu Y, Asherson P. Association study between a polymorphism at the 3'-untranslated region of CLOCK gene and attention deficit hyperactivity disorder. Behav Brain Funct. 2010;6:48. [DOI] [PubMed] [PMC]
    Baird AL, Coogan AN, Siddiqui A, Donev RM, Thome J. Adult attention-deficit hyperactivity disorder is associated with alterations in circadian rhythms at the behavioural, endocrine and molecular levels. Mol Psychiatry. 2012;17:98895. [DOI] [PubMed]
    Desan PH, Oren DA. Is seasonal affective disorder a disorder of circadian rhythms? CNS Spectr. 2001;6:487501. [DOI] [PubMed]
    Johansson C, Willeit M, Smedh C, Ekholm J, Paunio T, Kieseppä T, et al. Circadian clock-related polymorphisms in seasonal affective disorder and their relevance to diurnal preference. Neuropsychopharmacology. 2003;28:7349. [DOI] [PubMed]
    Zhang L, Hirano A, Hsu P, Jones CR, Sakai N, Okuro M, et al. A PERIOD3 variant causes a circadian phenotype and is associated with a seasonal mood trait. Proc Natl Acad Sci U S A. 2016;113:E153644. [DOI] [PubMed] [PMC]
    Scammell TE, Arrigoni E, Lipton JO. Neural Circuitry of Wakefulness and Sleep. Neuron. 2017;93:74765. [DOI] [PubMed] [PMC]
    Hickie IB, Rogers NL. Novel melatonin-based therapies: potential advances in the treatment of major depression. Lancet. 2011;378:62131. [DOI] [PubMed]
    Wulff K, Dijk D, Middleton B, Foster RG, Joyce EM. Sleep and circadian rhythm disruption in schizophrenia. Br J Psychiatry. 2012;200:30816. [DOI] [PubMed] [PMC]
    Naismith SL, Hermens DF, Ip TKC, Bolitho S, Scott E, Rogers NL, et al. Circadian profiles in young people during the early stages of affective disorder. Transl Psychiatry. 2012;2:e123. [DOI] [PubMed] [PMC]
    Robillard R, Oxley C, Hermens DF, White D, Wallis R, Naismith SL, et al. The relative contributions of psychiatric symptoms and psychotropic medications on the sleep-wake profile of young persons with anxiety, depression and bipolar disorders. Psychiatry Res. 2016;243:4036. [DOI] [PubMed]
    Alachkar A, Lee J, Asthana K, Monfared RV, Chen J, Alhassen S, et al. The hidden link between circadian entropy and mental health disorders. Transl Psychiatry. 2022;12:281. [DOI] [PubMed] [PMC]
    Sarowar T, Chhabra R, Vilella A, Boeckers TM, Zoli M, Grabrucker AM. Activity and circadian rhythm influence synaptic Shank3 protein levels in mice. J Neurochem. 2016;138:88795. [DOI] [PubMed]
    Tong H, Li Q, Zhang ZC, Li Y, Han J. Neurexin regulates nighttime sleep by modulating synaptic transmission. Sci Rep. 2016;6:38246. [DOI] [PubMed] [PMC]
    Menke A. Is the HPA Axis as Target for Depression Outdated, or Is There a New Hope? Front Psychiatry. 2019;10:101. [DOI] [PubMed] [PMC]
    Taylor BJ, Hasler BP. Chronotype and Mental Health: Recent Advances. Curr Psychiatry Rep. 2018;20:59. [DOI] [PubMed]
    Zou H, Zhou H, Yan R, Yao Z, Lu Q. Chronotype, circadian rhythm, and psychiatric disorders: Recent evidence and potential mechanisms. Front Neurosci. 2022;16:811771. [DOI] [PubMed] [PMC]
    Adan A. A chronobiological approach to addiction. J Subst Use. 2012;18:17183. [DOI]
    Bacqué-Cazenave J, Bharatiya R, Barrière G, Delbecque J, Bouguiyoud N, Giovanni GD, et al. Serotonin in Animal Cognition and Behavior. Int J Mol Sci. 2020;21:1649. [DOI] [PubMed] [PMC]
    Daut RA, Fonken LK. Circadian regulation of depression: A role for serotonin. Front Neuroendocrinol. 2019;54:100746. [DOI] [PubMed] [PMC]
    Duman RS, Sanacora G, Krystal JH. Altered Connectivity in Depression: GABA and Glutamate Neurotransmitter Deficits and Reversal by Novel Treatments. Neuron. 2019;102:7590. [DOI] [PubMed] [PMC]
    Teleanu RI, Niculescu A, Roza E, Vladâcenco O, Grumezescu AM, Teleanu DM. Neurotransmitters—Key Factors in Neurological and Neurodegenerative Disorders of the Central Nervous System. Int J Mol Sci. 2022;23:5954. [DOI] [PubMed] [PMC]
    Krystal JH, Anticevic A, Yang GJ, Dragoi G, Driesen NR, Wang X, et al. Impaired Tuning of Neural Ensembles and the Pathophysiology of Schizophrenia: A Translational and Computational Neuroscience Perspective. Biol Psychiatry. 2017;81:87485. [DOI] [PubMed] [PMC]
    Dienel SJ, Lewis DA. Alterations in cortical interneurons and cognitive function in schizophrenia. Neurobiol Dis. 2019;131:104208. [DOI] [PubMed] [PMC]
    Kumar V, Vajawat B, Rao NP. Frontal GABA in schizophrenia: A meta-analysis of 1H-MRS studies. World J Biol Psychiatry. 2021;22:113. [DOI] [PubMed]
    Cai Y, Xing L, Yang T, Chai R, Wang J, Bao J, et al. The neurodevelopmental role of dopaminergic signaling in neurological disorders. Neurosci Lett. 2021;741:135540. [DOI] [PubMed]
    Marotta R, Risoleo MC, Messina G, Parisi L, Carotenuto M, Vetri L, et al. The Neurochemistry of Autism. Brain Sci. 2020;10:163. [DOI] [PubMed] [PMC]
    Häge A, Banaschewski T, Buitelaar JK, Dijkhuizen RM, Franke B, Lythgoe DJ, et al. Glutamatergic medication in the treatment of obsessive compulsive disorder (OCD) and autism spectrum disorder (ASD) – study protocol for a randomised controlled trial. Trials. 2016;17:141. [DOI] [PubMed] [PMC]
    McCarty PJ, Pines AR, Sussman BL, Wyckoff SN, Jensen A, Bunch R, et al. Resting State Functional Magnetic Resonance Imaging Elucidates Neurotransmitter Deficiency in Autism Spectrum Disorder. J Pers Med. 2021;11:969. [DOI] [PubMed] [PMC]
    Abdul F, Sreenivas N, Kommu JVS, Banerjee M, Berk M, Maes M, et al. Disruption of circadian rhythm and risk of autism spectrum disorder: role of immune-inflammatory, oxidative stress, metabolic and neurotransmitter pathways. Rev Neurosci. 2021;33:93109. [DOI] [PubMed]
    Deshmukh R, Franco K. Managing weight gain as a side effect of antidepressant therapy. Cleve Clin J Med. 2003;70:614, 616, 618, passim. [PubMed]
    Köhler O, Benros ME, Nordentoft M, Farkouh ME, Iyengar RL, Mors O, et al. Effect of anti-inflammatory treatment on depression, depressive symptoms, and adverse effects: a systematic review and meta-analysis of randomized clinical trials. JAMA Psychiatry. 2014;71:138191. [DOI] [PubMed]
    Wang L, Wang R, Liu L, Qiao D, Baldwin DS, Hou R. Effects of SSRIs on peripheral inflammatory markers in patients with major depressive disorder: A systematic review and meta-analysis. Brain Behav Immun. 2019;79:2438. [DOI] [PubMed]
    Baumeister D, Ciufolini S, Mondelli V. Effects of psychotropic drugs on inflammation: consequence or mediator of therapeutic effects in psychiatric treatment? Psychopharmacology (Berl). 2016;233:157589. [DOI] [PubMed]
    Abdoli A, Taghipour A, Pirestani M, Jahromi MAM, Roustazadeh A, Mir H, et al. Infections, inflammation, and risk of neuropsychiatric disorders: the neglected role of "co-infection". Heliyon. 2020;6:e05645. [DOI] [PubMed] [PMC]
    Kripke DF, Mullaney DJ, Atkinson M, Wolf S. Circadian rhythm disorders in manic-depressives. Biol Psychiatry. 1978;13:33551. [PubMed]
    Wirz-Justice A. The implications of chronobiology for psychiatry. Psychiatric Times. 2011;56–61.
    Kasper S, Hajak G, Wulff K, Hoogendijk WJG, Montejo AL, Smeraldi E, et al. Efficacy of the novel antidepressant agomelatine on the circadian rest-activity cycle and depressive and anxiety symptoms in patients with major depressive disorder: a randomized, double-blind comparison with sertraline. J Clin Psychiatry. 2010;71:10920. [DOI] [PubMed]
    Wang J, Jin L, Zhu Y, Zhou X, Yu R, Gao S. Research progress in NOS1AP in neurological and psychiatric diseases. Brain Res Bull. 2016;125:99105. [DOI] [PubMed]
    Minelli A, Maffioletti E, Cloninger CR, Magri C, Sartori R, Bortolomasi M, et al. Role of allelic variants of FK506-binding protein 51 (FKBP5) gene in the development of anxiety disorders. Depress Anxiety. 2013;30:11706. [DOI] [PubMed]
    Chawathey K, Ford A. Cognitive behavioural therapy. InnovAiT. 2016;9:51823. [DOI]
    Aviram A, Westra HA, Constantino MJ, Antony MM. Responsive management of early resistance in cognitive–behavioral therapy for generalized anxiety disorder. J Consult Clin Psychol. 2016;84:78394. [DOI] [PubMed]
    Wirz-Justice A, Terman M. Chronotherapeutics (light and wake therapy) as a class of interventions for affective disorders. Handb Clin Neurol. 2012;106:697713. [DOI] [PubMed]
    Ohdo S. Chronotherapeutic strategy: Rhythm monitoring, manipulation and disruption. Adv Drug Deliv Rev. 2010;62:85975. [DOI] [PubMed]
    Terman M, Terman JS. Light therapy. In: Kryger MH, Roth T, Dement WC, editors. Principles and Practice of Sleep Medicine. 5th ed. St Louis: Elsevier/Saunders; 2010. pp.1682–92. [DOI]
    Khalifeh AH. The effect of chronotherapy on depressive symptoms. Evidence-based practice. Saudi Med J. 2017;38:45764. [DOI] [PubMed] [PMC]
    Even C, Schröder CM, Friedman S, Rouillon F. Efficacy of light therapy in nonseasonal depression: a systematic review. J Affect Disord. 2008;108:1123. [DOI] [PubMed]
    Golden RN, Gaynes BN, Ekstrom RD, Hamer RM, Jacobsen FM, Suppes T, et al. The efficacy of light therapy in the treatment of mood disorders: a review and meta-analysis of the evidence. Am J Psychiatry. 2005;162:65662. [DOI] [PubMed]
    Tao L, Jiang R, Zhang K, Qian Z, Chen P, Lv Y, et al. Light therapy in non-seasonal depression: An update meta-analysis. Psychiatry Res. 2020;291:113247. [DOI] [PubMed]
    Lieverse R, Someren EJWV, Nielen MMA, Uitdehaag BMJ, Smit JH, Hoogendijk WJG. Bright light treatment in elderly patients with nonseasonal major depressive disorder: a randomized placebo-controlled trial. Arch Gen Psychiatry. 2011;68:6170. [DOI] [PubMed]
    Hizli Sayar G, Agargun, Tan, Bulut. Comparison of effects of bright light therapy alone or combined with fluoxetine on severity of depression, circadian rhythms, mood disturbance, and sleep quality, in patients with non-seasonal depression. ChronoPhysiol Ther. 2013;3:539. [DOI]
    Wirz-Justice A, Bader A, Frisch U, Stieglitz R, Alder J, Bitzer J, et al. A randomized, double-blind, placebo-controlled study of light therapy for antepartum depression. J Clin Psychiatry. 2011;72:98693. [DOI] [PubMed]
    Benedetti F, Barbini B, Colombo C, Smeraldi E. Chronotherapeutics in a psychiatric ward. Sleep Med Rev. 2007;11:50922. [DOI] [PubMed]
    Ioannou M, Wartenberg C, Greenbrook JTV, Larson T, Magnusson K, Schmitz L, et al. Sleep deprivation as treatment for depression: Systematic review and meta-analysis. Acta Psychiatr Scand. 2021;143:2235. [DOI] [PubMed] [PMC]
    Casher M, Schuldt S, Haq A, Burkhead-Weiner D. Chronotherapy in Treatment-Resistant Depression. Psychiatr Ann. 2012;42:1669. [DOI]
    Martiny K, Refsgaard E, Lund V, Lunde M, Sørensen L, Thougaard B, et al. The day-to-day acute effect of wake therapy in patients with major depression using the HAM-D6 as primary outcome measure: results from a randomised controlled trial. PLoS One. 2013;8:e67264. [DOI] [PubMed] [PMC]
    Benedetti F, Barbini B, Fulgosi MC, Colombo C, Dallaspezia S, Pontiggia A, et al. Combined total sleep deprivation and light therapy in the treatment of drug-resistant bipolar depression: acute response and long-term remission rates. J Clin Psychiatry. 2005;66:153540. [DOI] [PubMed]
    Echizenya M, Suda H, Takeshima M, Inomata Y, Shimizu T. Total sleep deprivation followed by sleep phase advance and bright light therapy in drug-resistant mood disorders. J Affect Disord. 2013;144:2833. [DOI] [PubMed]
    Moscovici L, Kotler M. A multistage chronobiologic intervention for the treatment of depression: a pilot study. J Affect Disord. 2009;116:2017. [DOI] [PubMed]
    Sahlem GL, Kalivas B, Fox JB, Lamb K, Roper A, Williams EN, et al. Adjunctive triple chronotherapy (combined total sleep deprivation, sleep phase advance, and bright light therapy) rapidly improves mood and suicidality in suicidal depressed inpatients: an open label pilot study. J Psychiatr Res. 2014;59:1017. [DOI] [PubMed] [PMC]
    Wu JC, Kelsoe JR, Schachat C, Bunney BG, DeModena A, Golshan S, et al. Rapid and sustained antidepressant response with sleep deprivation and chronotherapy in bipolar disorder. Biol Psychiatry. 2009;66:298301. [DOI] [PubMed]
    Phelps J. Dark therapy for bipolar disorder using amber lenses for blue light blockade. Med Hypotheses. 2008;70:2249. [DOI] [PubMed]
    Barbini B, Benedetti F, Colombo C, Dotoli D, Bernasconi A, Cigala-Fulgosi M, et al. Dark therapy for mania: a pilot study. Bipolar Disord. 2005;7:98101. [DOI] [PubMed]
    Henriksen TE, Skrede S, Fasmer OB, Schoeyen H, Leskauskaite I, Bjørke-Bertheussen J, et al. Blue-blocking glasses as additive treatment for mania: a randomized placebo-controlled trial. Bipolar Disord. 2016;18:22132. [DOI] [PubMed] [PMC]
    Alexander B, Warner-Schmidt J, Eriksson T, Tamminga C, Arango-Lievano M, Ghose S, et al. Reversal of depressed behaviors in mice by p11 gene therapy in the nucleus accumbens. Sci Transl Med. 2010;2:54ra76. [DOI] [PubMed] [PMC]