Skip to main content
Aging and Disease logoLink to Aging and Disease
. 2023 Aug 1;14(4):1038–1069. doi: 10.14336/AD.2022.1203

Clinical Spectrum and Trajectory of Innovative Therapeutic Interventions for Insomnia: A Perspective

Yun-Jo Lo 1, Viraj Krishna Mishra 2, Hung-Yao Lo 1, Navneet Kumar Dubey 3,4,*, Wen-Cheng Lo 5,6,7,*
PMCID: PMC10389812  PMID: 37163444

Abstract

Increasing incidences of insomnia in adults, as well as the aging population, have been reported for their negative impact on the quality of life. Insomnia episodes may be associated with neurocognitive, musculoskeletal, cardiovascular, gastrointestinal, renal, hepatic, and metabolic disorders. Epidemiological evidence also revealed the association of insomnia with oncologic and asthmatic complications, which has been indicated as bidirectional. Two therapeutic approaches including cognitive behavioral therapy (CBT) and drugs-based therapies are being practiced for a long time. However, the adverse events associated with drugs limit their wide and long-term application. Further, Traditional Chinese medicine, acupressure, and pulsed magnetic field therapy may also provide therapeutic relief. Notably, the recently introduced cryotherapy has been demonstrated as a potential candidate for insomnia which could reduce pain, by suppressing oxidative stress and inflammation. It seems that the synergistic therapeutic approach of cryotherapy and the above-mentioned approaches might offer promising prospects to further improve efficacy and safety. Considering these facts, this perspective presents a comprehensive summary of recent advances in pathological aetiologies of insomnia including COVID-19, and its therapeutic management with a greater emphasis on cryotherapy.

Keywords: insomnia, cognitive behavioral therapy, whole-body cryotherapy, traditional chinese medicine, pulsed magnetic field therapy, acupressure


Insomnia is one of the archaic but prevalent disorders causing sleep deprivation and impacting the quality of life, health, and economy throughout the world [1]. The prevalence of insomnia is more prominent in the elderly population and its progression depends on factors such as age, gender, behavior, environment, health conditions, the status of disorders, psychological and social conditions, etc. [2]. Additionally, the risk of insomnia may escalate up to three folds among cancer patients [3]. Incomplete sleep or recurring awakening during sleep are primary indicators of insomnia [4, 5]. The classical approach subdivides insomnia into three categories: i) Initial or pre-dormitional insomnia (delayed sleep), ii) Middle insomnia (fragmented sleep) and iii) Terminal or post-dormitional/ late insomnia-incomplete sleep [6]. However, various insomnia-related sleep characteristics have been grouped into the following four symptoms: (i) difficulty in initiating sleep (DIS), (ii) difficulty in maintaining sleep (DMS), (iii) early morning awakening (EMA), and non-restorative sleep (NRS) [7]. Acute insomnia mostly exerts short-term adverse effects, whereas chronic insomnia could potentially increase the risk of infection, depression, cardiovascular and respiratory disorders due to the impact of sleep disturbance on both innate and adaptive immunity [8-10]. Additionally, conditions such as comorbid insomnia and sleep apnea (COMISA), render difficulty in determining the exact etiology of insomnia [11]. Lack of sleep or disturbed sleep also deteriorates the condition of seizures due to neuroinflammation [12]. Insomnia disrupts the balance between sleep, the immune, and the central nervous system leading to inflammation and alteration in antiviral response [13]. Recently, the COVID-19 outbreak and related stress have been demonstrated to increase the risk of insomnia [14]. Therefore, sufficient sleep is highly crucial for hormonal balance and homeostatic immune functions for the defense of inflammatory activities, which is advantageous for long-term insomnia triggering systemic inflammatory disorders such as neurodegenerative disorders, diabetes, and atherosclerosis [15]. To address insomnia, current therapeutic interventions such as psychological, yoga, and meditation, traditional Chinese medicines and acupressure, as well as drugs have been explored [16]. However, these alternatives remain inadequate in overcoming insomnia [17]. Hence the novel electrostimulation-based approach such as cranial electrical stimulation (CES) is considered an effective candidate which mediates its therapeutic effect by regulating hormonal, neural, and behavioral responses [6]. Further, recent progress has shown that cryotherapy, in particular, whole-body cryotherapy (WBC) may exert a positive impact on disorders such as asthma, osteoarthritis, rheumatoid arthritis, pain, etc. [18]. WBC has also been evident in improving sleep quality, which opens the way to explore its therapeutic applicability for insomnia [19]. Based on the above-mentioned evidence, we have extensively reviewed pathophysiological aetiologies, incidence, and therapeutic progress in the treatment of insomnia.

Aetiology, epidemiology, and pathophysiology of insomnia

Insomnia is variably widespread throughout the world among all gender, age groups, and geographical locations which depends on factors such as stress, lifestyles, working culture, disease status, environment, sleeping conditions, etc. (Fig. 1)[20, 21]. The cases of insomnia have increased considerably and reached over 200 million in recent years [22]. The overall prevalence varies from 5% to 50%; however, the prevalence rate largely depends on the diagnostic criteria applied and the type or characteristics of insomnia [23]. The incidence of insomnia varies from 5.8 to 20% among the adult population in wider prospects [24], whereas, the prevalence of insomnia varies from 30% to 48% among the aging population [2]. This data could be under-reported due to lesser visits to seek medical assistance [25, 26]. Moreover, the recent change in lifestyle and excessive use of electronic devices have affected sleep quality and efficiency, thereby elevating the cases of sleep disturbances and insomnia [27-29]. The increasing dependency on mobile or computer screens for study, profession, social contact, and entertainment among youth, adolescents, and children is impacting their sleep quality [30-32]. Further, a cross-sectional study observed an association between insufficient physical activity and sleep problems [33]. The prevalence of insomnia is reported as 15.4%-33%, 15%, 24.6%-32.1%, 37%, 7%, 10.1%, 23.8%, 15%-29%, 3.0%-5.5%, 20.8%, 17%-23%, and 30-40% in the population of India [34-36], China [37], Sweden [38, 39], UK [40], Italy [41, 42], Europe [41], Canada, France [43, 44], Qatar [45], Spain, South Korea [46] and USA [47]. The factors which increase the prevalence of insomnia include response to social and economic factors, low income, and education level, susceptibility to chronic diseases such as osteoporosis, complications that arise due to menopause, etc. [48-51]. On a gender basis, the incidence of insomnia is higher among the female and aged population in Saudi Arabia, which may be associated with being elderly, widowed/divorced, females, or housewives, lack of education, and excessive tea consumption [52]. Moreover, ethnicity has a substantial and varying effect on insomnia prevalence and is considered a prominent factor in diagnosis [53, 54] This difference could be attributed to socioeconomic and lifestyle patterns of ethnic groups without any significant relation to genetic traits [55]. Furthermore, the COVID-19 pandemic has increased the prevalence of insomnia due to associated stress, anxiety, depression, fear, post-COVID-19 complications [56, 57].

Figure 1.

Figure 1.

Etiolopathological factors of insomnia, which mainly involve heavy food, coffee, daily habits (smoking, alcohol, blue light), headache, chronic pain, aging, gender, and geographical location.

Understanding the pathophysiology of insomnia is essential to developing effective therapeutic approaches. Insomnia is the result of an intricate communication of behavioral, physiological, and genetic factors [58] (Fig. 2). Multiple attempts have been made to develop models to interpret and explain the initiation and progression of insomnia [4]; however, are insufficient to express a comprehensive understanding. According to the neurocognitive model, the central character of insomnia is associated with hyperarousal, enhanced cognitive, cortical, and somatic activation [59-61]. The present models of hyperarousal and stress dysregulation explain the insomnia mechanism specifically by stimulating neurobiological and psychological activities triggering sleeping disturbance Intellectualtual or emotional cognitive arousal is associated with ruminative thinking leading to nocturnal awakening and hyperarousal [62]. Nocturnal arousal also causes depression and pre-sleep somatic arousal associated with insomnia. Additional activities such as computer gaming, internet surfing, and television watching impact sleep patterns resulting in a detrimental effect on the learning and memory of an adult [63-65]. Hence, even a half-hour of cognitive activity before sleep may induce emotional and mental flow resulting in sleep arousal and an increase in sleep period indicating early insomnia symptoms [23]. Further, the irregularity in sleep patterns such as prolongation of sleep latency, decrease in sleep efficiency, and sleep duration engender nocturnal cognitive arousal leading to insomnia and depression. However, cognitive arousal is more frequent during the night and could lead to physiologic hyperarousal [66]. The report evidenced that emotions either negative or positive, internal sleep locus control, and sleep belief are linked to varying impacts on sleep arousal in both genders [67]. It has also been noted that though hyperarousal is associated with positive and negative emotions in all gender, pre-sleep arousal is more inclined towards negative emotions among women.

Figure 2.

Figure 2.

Physiological factors contributing to insomnia. Hyperarousal as a major contributor to insomnia may occur due to an increase in cognitive, cortical, and somatic activation, induction of RBFOX3, and 5-HTTLPR genes. Other factors include downregulation of sleep promoters (Adenosine, GABA, Melatonin, Serotonin, Prostaglandin D2) and upregulation of sleep suppressors (Histamine, orexin, catecholamines, and norepinephrine) which instigate sleep disturbance resulting in insomnia. The genetic factors (apoE gene, SNP, and CLOCK genes), cognitive, and behavioral factors (Anxiety, depression, psychosis, cognitive distortion, unemployment, and alcohol/tobacco consumption) also contribute to insomnia. Disturbance in sleep regulatory switch owing to molecular alterations associated with sleep (VLPO) and wake (Orexins, TMN, DR, LC) also results in insomnia symptoms. RBFOX3: RNA binding fox-1 homolog 3, 5-HTTLPR: Serotonin (5-HT) transporter polymorphism, GABA: Gamma-Aminobutyric Acid, apo E: apolipoprotein E, SNP: Single nucleotide polymorphisms, CLOCK: Circadian locomotor output cycles kaput protein, VLPO: Ventrolateral preoptic nucleus, TMN: Tuberomammillary nucleus, DR: dorsal raphe nucleus, LC: noradrenergic locus coeruleus.

In addition, physiologic hyperarousal has been reported in half of the chronic insomnia cases with an increased risk of diabetes, hypertension, and neurocognitive impairment leading to increased mortality [68, 69]. Moreover, severe physiological hyperarousal has been associated with underweight conditions. Furthermore, physiological hyperarousal-mediated insomnia triggers hypertension which has been confirmed by longer multiple sleep latency test (MSLT) values [70]. Physiologic hyperarousal also negatively impacts cognitive activity leading to an increase in error rate in different tasks [71]. Cortical hyperarousal is also an indicator of insomnia which is mainly triggered through pre-sleep cortical activity and causes delayed sleep onset [72]. Hyperarousal is indicated by the presence of symptoms such as increased heart rate, anxiety, anger or irritability, startle response, and struggle with relaxation [73, 74]. The higher brain activity and glucose metabolism in the wake center are recorded as evidence of hyperarousal among insomnia patients [75]. Failure of the arousal mechanism to trigger wake to sleep state could be linked to the incapacity to fall asleep. Low resilience triggers emotion, stress, and arousal dysregulation [76]. In summary, cognitive, physiological, and cortical hyperarousal has been reported as causative factors for insomnia, though extensive studies are required to decipher an exact mechanism underlying hyperarousal.

Recent research developments have paved the way to understanding the molecular mechanism and genes involved in the regulation of sleep, however, the exact pathways involved in insomnia have not yet been explored [77]. The predisposing, precipitating, and perpetuating factors representing the 3P model of insomnia indicate that inheritance and genetic factors are associated with insomnia and change in sleep pattern in response to stress. Sleep is primarily regulated through independent molecular mechanisms including sleep homeostasis and circadian rhythm. These two cellular pathways regulate the sleep/wake cycle under environmental conditions in association with light patterns [78]. Accumulation of adenosine and melatonin in the daytime primarily regulates sleep homeostasis and the circadian system [79].

Besides, the common methodologies including genome-wide association study (GWAS), family and twin studies along with candidate gene studies are being used to understand the role of genes in insomnia progression [80]. As per reports, the circadian gene such as PER1, 2, 3, and CRY1 may be the genetic determinant of insomnia [81, 82]. The variable number of tandem-repeat (VNTR) polymorphism in PER3 triggers sleep deficiency through the expression of the sleep homeostasis gene, therefore impacting sleep regulation and circadian insomnia-associated pathway [82, 83]. PER 3 causes a change in light and cognitive responses due to altered brain activity and hypothalamic gene expression. The SNP in various genes has been studied to establish a correlation with insomnia; however, no strong association exists between them [84, 85]. Notwithstanding, SNPs such as rs10493596, rs2302729, rs12927162, rs324957/rs324981, and rs1823125 in genes AK5, CACNA1C, TOX3, neuropeptide S receptor (NPSR1), and PAX8, respectively contribute to insomnia symptoms [86]. In an integrative genome analysis, the abnormal expression level of genes including DALRD3, LDHA, HEBP2, TEX264, and FGFR3 have been identified as candidate genes contributing to insomnia, however, extensive studies are needed to establish their functional pathway [87]. The decreased levels of brain-derived neurotrophic factor (BDNF) during stress also trigger depression and sleep dysregulation leading to sleep deprivation or insomnia [88]. In response to stress, RBFOX3, a gene involved in the overexpression of neurotransmitter GABA, the ABCC9 gene in KATP channel, and the 5-HTTLPR gene have been attributed to hyperarousal and wake promotion triggering insomnia [80]. Additionally, apolipoprotein E4 (Apo E4), HLA-DQB1*0602, homozygous Clock gene 3111C/C Clock, and short (s-) allele of the 5-HTTLPR has also been associated [89].

Biomolecules such as Orexins-A and Orexin-B regulate sleep-wake cycles. In presence of light, the orexin causes slow wave sleep and rapid eye movement and is responsible for the change of sleep to wake state [90, 91]. Orexins stimulate wake periods through activating orexin, cholinergic and monoaminergic neurons present in the hypothalamus/brainstem regions and specifically those expressed in the lateral hypothalamic area (LHA) [92, 93]. Notably, orexin neurons are distributed densely in the histaminergic tuberomammillary nucleus (TMN), serotonergic dorsal raphe nucleus (DR), and noradrenergic locus coeruleus (LC) which are involved in keeping individuals awake [92-95]. TMN and ventrolateral preoptic nucleus (VLPO) is the prominent region in the anterior and posterior hypothalamus which regulates the sleep-wakefulness cycle [96]. VLPO neurons are co-expressed with galanin and γ-aminobutyric acid (GABA) which triggers sleep, whereas TMN releases histamine involved in sustaining the wake stage [96, 97]. It has been demonstrated that synaptic suppression of the VLPO region disrupts the normal function of sleep-promoting neurons leading to increased wakefulness time [98]. VLPO seems to play a crucial role in sleep promotion, however, a recent study in VLPO lesion rat models demonstrated that VLPO might be involved in both sleep and arousal cycles [99]. Similarly, an increase in the level of catecholamines triggers disturbance and loss of nocturnal sleep [100]. The increase in the nocturnal level of norepinephrine is correlated with insomnia, indicating the significance of sleep in maintaining the function of the immune and sympathetic nervous systems [101]. Histamine also plays a crucial role in the control of the sleep/wake cycle. Specifically, the histamine receptors H1 and H3 play a crucial role in wake and sleep respectively, and the activation of the H3 receptor promotes sleep by downregulating the level of histamine [102].

γ-Aminobutyric acid (GABA) is a neurotransmitter inhibitor of the central nervous system (CNS) and regulates sleep by inhibiting neurotransmitter activity related to the wake cycle [103, 104]. Further, the increase in GABA level is considered an indicator of hyperarousal and its decreased levels in the anterior cingulate cortex (ACC) and medial prefrontal cortex (mPFC) have been associated with shorter sleep duration resulting in insomnia and impaired memory function [105]. Adenosine is a neuromodulator that accumulates during the wake cycle [106] and induces sleep by inhibiting GABAergic neurons in the ventrolateral preoptic area [107]. Further, chronic consumption of ethanol results in sleep disruption and remains sustained even after its termination by lowering ENT1 expression, adenosine, and glutamate transporter 1 resulting in the progression of insomnia [108]. Serotonin (5HT) plays a crucial role in sleep-wake regulation and its impact depends on receptors (5HT1A, 5HT1B, 5HT2, and 5HT3) and brain regions [109, 110]. Specifically, a decrease in 5HT may promote depression and insomnia among healthy individuals, which could be restored through therapeutic drugs [111]. Further, aerobic exercise exerts a positive impact on 5HT activities in the mid-brain and bloodstream and its release in the diencephalon and cerebrum promotes normal sleep [112]. Pineal gland-secreted melatonin during darkness maintains the circadian clock resulting in sleep stimulation and its regulation [113-115]. Reportedly, disruption in melatonin expression could also result in reduced immune response, sleep disturbance, and cancer [116]. Advancing age-dependent reduction of melatonin levels occurs mainly due to calcification of the pineal gland [117, 118]. Further, the prostaglandin D2 (PGD2) a lipid mediator which after being synthesized in the choroid plexus, leptomeninges, and oligodendrocytes region of the brain is secreted in cerebrospinal fluid, thereby inducing the adenosine accumulation in the hypothalamus leading to sleep induction [119]. The PGD2 in cerebrospinal fluid impacts D-type prostanoid receptors resulting in sleep induction, though the pathway by which PGD2 is cleared from the cerebrospinal fluid is unclear [120]. In addition to psychiatric disorders such as anxiety, depression, psychosis, and cognitive distortion, the factors which promote an abnormal increase in aminergic response, central aminergic state, and reduce the inhibitory neurotransmission could also be aetiologies of insomnia. [74, 121]. Thus, molecules such as orexins, GABA, 5-HT, melatonin, and PGD2 play a crucial role in sleep regulation and insomnia and have also been evaluated for their therapeutic candidacy for sleep disorders and insomnia. Besides these molecular factors, sociodemographic variables like higher education, unemployment, alcohol/tobacco consumption, and generalized anxiety disorders have also been significantly correlated with insomnia among the Indian population [122].

Common Complications of Insomnia

Insomnia adversely impacts various organs (Fig. 3), resulting in aberrant sleep patterns, cognitive function, and emotional responses [123]. It also triggers neuropsychiatric disorders such as depression, dementia, mania, schizophrenia, and anxiety disorders [124], in addition to participating in the pathologic progression of the immune, endocrine and cardiovascular systems. It has been further reported that insomnia enhances the risk of hypertension, diabetes mellitus, arthritis, stomach ulcers, migraine, depression, obesity, heart attack/stroke, asthma, menstrual problem, obesity, and infection [123, 125, 126]. These disorders and complications have a cumulative impact on insomnia. Objective short sleep duration (OSSD) is a severe variant of insomnia and causes hyperarousal, hypertension, diabetes, variation in heart rate, neurocognitive disorder, induction of stress system, and increase in mortality [127]. It enhances the incidence of hypertension three times and is more prevalent than subjective short sleep [128, 129]. OSSD increases the risk of persistent insomnia though behavioral factors such as alcohol, caffeine, and cigarettes consumptions have not been significantly correlated [130]. Additionally, severe insomnia occurs with the deterioration of mental conditions, and OSSD of less than 6h is responsive to cognitive behavioral therapy [131]. Insomnia impacts telomere length; however, it varies according to gender, age, and ethnicity [132].

Figure 3.

Figure 3.

Extent and health consequences of insomnia. Possible mono- or bidirectional association between insomnia and other pathophysiologies including neurological (epilepsy, neuropathic pain, Alzheimer’s disease, Parkinson’s disease, cognitive and learning, Huntington’s disease, Multiple sclerosis), musculoskeletal (osteoarthritis, Rheumatoid arthritis, osteoporosis), metabolic (Diabetes mellitus, obesity, GERD), renal, and cancer (Glioma, thyroid, cervical, colorectal and lung). GERD: Gastroesophageal reflux disease.

Insomnia and neuropsychiatric disorders

Anxiety could be associated with fear/phobia, panic perception, separation, social discomfort, or attachments [121]. Multiple studies have indicated that anxiety with worry and extreme anxiety could be used as diagnostic indicators of insomnia [133-135]. A cross-sectional study confirms the relationship between anxiety and poor sleep quality among the Indonesian population [136]. Depression and insomnia are bidirectionally correlated, and both enhance the risk of each other [137, 138]. Major depression episodes are associated with symptoms of insomnia and hyper-insomnia, which increases the risk of recurrence of depression and suicidal attempts [139, 140]. It has been observed that regular insomnia could promote the late onset of depression and the promotion of major recurrent depressive disorders [141]. The directional association of insomnia, anxiety disorder, and depression has been studied and it was evident that prior anxiety is significantly associated with insomnia [142]. In a similar pattern, prior depression is not significantly associated with insomnia; however, prior insomnia could lead to severe depression. Nonetheless, more studies are required to establish the directional relationship between anxiety, insomnia, and depression.

Insomnia and neurocognitive disorders

Prevalence of sleep disorders/insomnia has been observed among neurocognitive disorder patients. In particular, chronic insomnia, sleep apnoea and disrupted circadian rhythm has been correlated with Alzheimer’s, Parkinson’s disease, and Lewy body dementia [143]. The objective short sleep impedes cognitive functions and related cardiometabolic conditions [144, 145]. Sleep disturbance disrupts the balance between attention, memory, and the execution of cognitive controls [146]. Disturbed sleep might exacerbate neuropathic pain; however, a causal bidirectional association between them has also been implied [147, 148]. Neuropathic pain when associated with herpetic/post-traumatic trigeminal nerve dysfunction, or diabetes negatively impacts sleep quality resulting in characteristics of insomnia [147, 149-151]. Reports also indicate the association between sleep/awake state and epilepsy syndromes which has led to an acceptance of polysomnographic testing, a common diagnostic technique for monitoring epilepsy as well as comprehensive sleep [152]. Epilepsy patients also suffer from frequent insomnia with obviously increased morbidity and reduced quality of life (QoL) [153-156]. Poor sleep quality is strongly associated with the female gender due to overstress and gender discrimination [157, 158]. However, contrary studies also exist showing no significant difference in the frequency of insomnia on a gender basis [159]. Other factors such as social security and employment conditions negatively impact cognitive functions and the development of a sense of insecurity resulting in poor sleep [157]. Molecular studies have revealed that the dysregulated miRNAs in a patient with sleep-related epilepsy syndromes include miRNA-211, miRNA-155, miRNA-328, miR 194-5p, and miR 106b associated with CHRNA4, CHRNA2 CHRNB2 SCN1A, PAXNEB, GRIN2A, EFHC1, ClCN2, KCNQ3, KCNMB3, GABRA1, BRD2, KCNJ10, and CACNA1A [143, 151-159].

Besides, the stroke also increases the frequency of insomnia (one-third of stroke survivors) and is considered a prevalent complication that adversely impacts QoL, and mortality and enhances the risk of other secondary complications [160-163]. The association of post-stroke insomnia with depression, fatigue, and disability disrupts recovery and normal working life [163]. The incidence of insomnia is more prevalent among multiple sclerosis (MS) female patients than in normal populations and has been associated with MS-related fatigue [164, 165]. Sleep disturbance among MS patients could also be associated with depression, immunomodulatory therapy, body pain, and daytime sleep [166], and could contribute to memory loss [167]. Further, nearly 87.8% of Huntington's disease (HD) patients suffer from insomnia and other sleep disorders [168, 169]. The elevated motor function during sleep decreases rapid eye movement which results in short sleep duration and leg movement during sleep [168]. Nonetheless, sleep interference could be related to arousal associated with irregular movement rather than rapid eye movement [170]. Sleep disorders progressively worsen HD patients’ efficiency to perform daily routine/essential activities resulting in poor QoL [171]. The aberrant structural change and atrophy in brain regions such as the hypothalamus, locus coeruleus, and striatum could be associated with sleep disturbance among HD patients [171-175]. In the HD mice model, it has been demonstrated that rather than any direct impact from mutated huntingtin protein, the change in melatonin, cortisol, or disruption in circadian rhythms negatively impacts sleep [169, 176].

Sleep disturbance is also prevalent among glioma patients and the etiology of which could be multifactorial [177]. Insomnia could be associated with not only parenchyma damage, chemotherapy, and radiotherapy, but also with corticosteroids, antiepileptics, and psychoactive drugs administered to HD patients. Scientific evidence supports the role of glial cells in sleep homeostasis, determination of sleep period, regulation of damaged axons, and efflux of toxic content [178, 179]. These glial cell-mediated activities could be disrupted by insomnia [178]. A pre-clinical model suggests that chronic sleep loss accelerates the activation of microglia leading to astrocytes phagocytosis and risk of brain damage [180]. Chronic insomnia results in the increase of S100 calcium binding protein B and glial fibrillary acidic protein, whereas a decrease in BDNF and glial cell line-derived neurotrophic factor in serum is associated with pathological alteration in astrocytes leading to cognitive function loss [181]. These pieces of evidence indicate the adverse impact of insomnia on brain structure and function in terms of impaired cognitive function.

Insomnia and Immune disorders

Sleep loss results in immune-associated disorders such as cardiovascular, autoimmune, and neurodegenerative diseases by negatively impacting both innate and adaptive immune systems by inducing inflammation [182]. It also reduces the immune response and therefore increases the risk of infection [183]. During chronic insomnia, the immune cell population of CD3+, CD4+, and CD8+ lymphocytes decreases, however without significantly affecting white blood cells, natural killer cells, and cytokine levels [184]. Further, sleep loss exacerbates the release of pro-inflammatory molecules C-reactive protein (CRP), tumor necrosis factor (TNF), IL-1β, IL-6, and IL-17, and promotes the progression of inflammatory disorders [182]. This occurs via induction of vascular endothelial molecules such as sICAM-1 and E-selectin along with activation of the nuclear factor kappa B (NF-κB) pathway [185-188]. Sleep deprivation induces the expression of signal transducer and activator of transcription (STAT) proteins (STAT1, STAT2, STAT3) in peripheral blood mononuclear cells (PBMC), monocyte, and lymphocyte populations resulting in activated innate immune arm and inflammation [189]. Sleep duration also plays a significant role in maintaining immune system balance; however, the associated pathways remain to be completely understood.

Insomnia and carcinogenesis

In recent years, the association between insomnia/short sleep duration and cancer has also been investigated. A systematic review and meta-analysis by Shi et al. reported a 24% increase in cancer incidence with insomnia [190]. Specifically, the carcinogenic impact of nocturnal work and regular sleep disturbance has been evidenced in the terms of enhanced risk of advanced breast cancer, nasal neoplasm, lung cancer, colorectal adenoma, cervical carcinoma, and thyroid cancer [191-193]. This increased risk of malignancies might be associated with an immunocompromised state. While inducing carcinogenic events, insomnia initially inhibits melatonin synthesis, and disrupts immune function, circadian rhythm, and metabolism, leading to increased reactive oxygen species (ROS) [194]. Melatonin inhibits the uptake of tumor precursors-contained saturated and polyunsaturated fatty acids responsible for the synthesis of 13-hydroxyoctadecadienoic acid, a mitogen for linoleic acid-dependent tumor progression [194, 195]. Moreover, sleep deprivation also enhances DNA damage through the generation of ROS, thus elevating the risk of cancer. The disruption in circadian rhythm impacts angiogenesis, epithelial-mesenchymal transition, and KRAS signaling pathways and introduces the risk of bladder cancer and other malignant tumors [196]. Insomnia and fatigue could be indicators of cancer initiation and progression and need to be considered as a pre-diagnostic factor for screening cancer [193]. A Mendelian randomization study indicates the increased risk of lung adenocarcinoma and squamous cell lung cancer among insomnia patients [197]. A systematic review indicates that frequencies of insomnia are variably prevalent among different types of cancer patients in the range of 30%-93.1% [198, 199]. Insomnia also prevails in survivors of oral/head-neck cancer, and lung cancer [198, 200, 201], which might be attributed to stress, anxiety, depression, and pain, particularly among patients treated with chemotherapy [202]. It is of note that though scientific evidence is accumulating on the correlation between insomnia to cancer initiation and progression, the pathologic flow in the terms of mono-or bidirectional nature still needs to be investigated.

Insomnia and musculoskeletal pathologies

It has been established that sleep loss also negatively affects bone mineral density (BMD) and its metabolism by altering the levels of sex/growth hormone and steroids, thereby inducing inflammation, another aberrant metabolism, and physical inactivity [203-206]. It has been evident that insomnia increases the risk of osteoporosis (7-fold) among aged women (age > 50 years) with a sleep duration of less than 5hrs/day [207]. Prominent bone loss may occur in postmenopausal women due to a lack of estrogen and the resultant increase in systemic inflammation [207-210]. A study among women indicates that short sleep, insomnia, or sleep disorders enlarge the scope of recurrent falls and body fractures except for the hip, which could be associated with a higher risk of osteoporosis [211, 212]. A population-based study asserts that short sleep duration significantly reduces the trabecular bone score (TBS) among obese adults and non-Hispanic women by adversely impacting the bone microarchitecture [213]. Thus, a combined low BMD and TBS representing the high frequency of osteoporotic fractures may be associated with insomnia [214]. The underlying possible mechanisms may include insomnia-mediated dysregulation of the hypothalamo-pituitary-adrenal (HPA) axis due to increased secretion of cortisol, altered levels of hormones (orexins, ghrelin, serotonin, melatonin), and stress-related metabolic alterations with its resultant impact on the sympathetic system [215].

Insomnia is also commonly found in osteoarthritis (OA) patients, possibly due to an increase in levels of systemic inflammatory cytokines [216]. Higher pain due to pro-inflammatory response in older OA patients has been shown to reduce with improvement in insomnia [217]. Another type of musculoskeletal disorder, known as rheumatoid arthritis (RA) has also been found significantly associated with insomnia, along with other factors such as depression, poor subjective health, and retirement [218]. A population-based survey indicates that pain and the urge to use the washroom may be the major cause of sleep fragmentation or disturbance among OA and RA patients [219]. Insomnia-mediated fatigue and emotional response such as anxiety and depression have an additive effect on pain under RA conditions [220]. The insomnia progression causes depression among RA patients due to an increase in inflammatory cytokines such as TNF-α, and cyclic citrullinated peptide, a rheumatoid factor [221].

Insomnia and cardiovascular disorders (CVDs)

The current scientific studies have evidenced that insomnia exacerbates CVDs and increases the mortality and morbidity rate [222]. It enhances the risk of stroke and heart attack at a considerable level [223]. In a community-based cohort study, the risk of CVDs has been associated with poor sleep efficiency and a possible underlying mechanism could be linked to the activation of the HPA axis and nervous system resulting in increased blood pressure, urinary cortisol, and catecholamines levels, reduction/variation in heart rate which causes endothelial dysfunction and atherosclerosis [224, 225]. Besides, insomnia-mediated hypertension and diabetes could be associated with the induction of CVDs.

Insomnia and asthma

Besides musculoskeletal deformities, insomnia severely worsens asthma characteristics along with sleep quality [226]. Thirty-seven percent of insomnia cases among asthmatic patients have shown increasing anxiety and depression and therefore a reduced quality of life [226]. The bronchial hyper-responsiveness increased bronchial resistance, and intense response to allergens escalate sleep disturbance among asthma patients [227]. In contrast to reports of bidirectional relationships, a positive unidirectional correlation between insomnia and asthma has been established using a genome-wide association study (GWAS) [228]. The study found that three shared genes i.e ITPR3, HEXIM1, and IQCH were involved between insomnia and asthma, while ITPR3 and HEXIM1 seem to be associated with an inflammatory response.

Insomnia and gastrointestinal disorders

Alteration in cytokines (IL-1, IL-6, and TNF) levels also triggers gastrointestinal disorders such as gastroesophageal reflux disease (GERD), and inflammatory bowel disorder [229, 230]. A population-based study by Jansson et al. indicates that besides age, gender, obesity, tobacco/alcohol consumption, and social status, sleep-associated difficulty plays a vital role in the progression of GERD [231]. This may result from insomnia and sleep disorders' ability to lower salivary volume, the number of swallows, and frequencies of peristalsis [229]. The disruption in circadian rest-activity rhythms contributes to the progression of non-alcoholic fatty acid disorders, gastrointestinal, inflammatory, and irritable bowel disorders [232, 233].

Insomnia and renal deficiencies

Insomnia also influences the sleep of kidney-transplanted patients by increasing stress, anxiety, and depression [234]. In the case of chronic insomnia, the risk of a high estimated glomerular filtration rate significantly increases with accelerated damage to kidney function; however, does not significantly contribute to end-stage renal stage disease (ESRD) [235]. However, the symptom of insomnia is also prominent among ESRD patients undergoing hemodialysis [236]. Hemodialysis is commonly associated with depression and its proper management can improve sleep quality. The increase in chronic kidney disease has also been associated with a sudden break in night sleep [237].

Insomnia and liver disorders

Sleep disorders increase the risk of chronic renal failure and liver cirrhosis, and sleep abnormalities which is a general symptom of cirrhotic patients [238, 239]. Insomnia is a common symptom associated with liver cirrhosis [240], particularly in the presence of hepatic encephalopathy (HE) along with dysregulation of glucose and melatonin metabolism, thermoregulation, and ghrelin secretion [240]. Insomnia results in excessive daytime sleep (EDS) which enhances the risk of non-alcoholic fatty liver disease (NAFLD), a leading causative agent for chronic HE and liver cirrhosis [241]. The insomnia-mediated insulin resistance and metabolic syndrome could be correlated with the progression of NAFLD [241-243], possibly due to increased secretion of inflammatory cytokines such as IL-6 and TNF-α, leading to an abnormal increase in hepatic free fatty acids owing to adipocyte lipolysis [244-246]. Change in appetite behavior due to an increase in ghrelin and a decrease in leptin level promotes obesity, metabolic syndrome, lesser physical activity, and fatigue which significantly contribute to the progression of NAFLD [247-249]. Insomnia is also prevalent among hepatitis B virus-related liver disorder Chinese patients [250]. Based on the above-mentioned evidence, the condition of insomnia should be properly addressed to overcome its adverse impacts.

Insomnia and metabolic syndrome

Sleep disorders disrupt the synergy of metabolism resulting in abnormal weight gain and obesity [251] through altering metabolic pathways triggered by the change in hormonal concentration such as leptin/ghrelin, appetite pattern, ANS response, HPA axis, and circadian rhythm which causes glucose tolerance and fat deposition. Sleep disturbance and circadian rhythm dysregulation may increase insulin resistance, glucose intolerance, and dysfunction of pancreatic β-cells leading to T2DM [252]. A cohort study indicates that insomnia, and short and long sleep duration is associated with obesity and central obesity [253]. However, though these recent studies have indicated the role of insomnia in abnormal weight gain, more in-depth studies are required to reach a consensus.

A population-based study documented that chronic insomnia plays a significant role in the progression of diabetes, while objective short-sleep duration might perpetuate the risk of cardiometabolic morbidity [254]. A bidirectional relationship between insomnia/short sleep and glucose metabolism, obesity, along with insulin resistance has also been indicated [255]. Sleep deficiency triggers deviation in metabolism pathways, food habits, and behavioral changes leading to metabolic complications [256]. Short sleep duration with slow wave sleep induces insulin resistance which diminishes glucose tolerance leading to type 2 diabetes mellitus (T2DM) development [257]. Reduced leptins and increased ghrelin levels, and the resultant inflammation occurring due to short sleep also play a crucial role in instigating T2DM [255-258]. The additional impact of sleep loss on food patterns includes an alteration in food sensitivity, and an increase in hunger and food consumption which promotes obesity and T2DM [259, 260]. Current studies indicate that insomnia has a significant role in the progression of T2DM; however, Green et al. pointed out that compared to short sleep duration, other factors such as smoking, lack of physical activity, alcohol consumption, obesity, and stress could also contribute to the diabetic condition [261]. Hence, the association between insomnia and diabetes needs to be extensively explored.

These aforementioned studies imply that pathological abnormalities associated with cancer, musculoskeletal, CVDs, asthma, gastrointestinal, renal, liver, and metabolic syndrome have been uni-or bi-directionally associated with insomnia and sleep disturbances. However, extensive studies are required to comprehend the molecular and biochemical pathways involved. While addressing therapeutic needs, it is essential to inhibit the progression of mild to chronic insomnia. Nowadays, cognitive behavioral therapy (CBT), pharmacological intervention with lifestyle changes, yoga, and exercise are commonly recommended.

Insomnia: The Current therapeutic approaches

CBT: A psychological intervention

CBT targets cognitive factors which contribute to the development of psychological abnormalities [262]. The primary objectives of CBT include a decrease in symptoms, functional recovery, and respite care. As an additive therapeutic approach, CBT is cognition, emotion, and behavior-specific [262, 263], and its ameliorative impact on depression, anxiety, stress, phobia, panic, and eating disorders has also been demonstrated [264, 265]. Based on the reported efficacy and safety of CBT in providing relief from various psychological disorders, it has also become an important non-pharmacological intervention for insomnia. CBT addresses insomnia-contributory factors such as regular diurnal napping, disturbed sleep-wake cycle, overconcern for poor sleep, anxiety, and overthinking [266]. Sleep restriction, stimulus control, and cognitive ordering through education are three basic elements of CBT [266, 267], which have shown a significant effect on sleep onset latency, sleep duration, sleep quality, sleep efficiency, and depression; however, its impact on pain has been found comparatively low [267]. CBT provides a relatively long-term effect lasting up to one year even after therapy [268]. A complex bidirectional relationship between insomnia and depression indicates that vestigial symptoms of depression after recovery could lead to the progression of insomnia [269-271]. The sequential treatment of insomnia and depression with CBT shows short-term efficacy, though long-term evaluation needed is needed to corroborate the therapeutic outcome [270]. CBT has also been found effective in controlling insomnia in OA pain and post-breast cancer survivors [272, 273]. In addition, compared to acupuncture therapy, CBT has been found as an effective candidate for primary care due to greater and long-term efficacy [274, 275].

Insomnia is associated with inflammation, as it impacts neurotransmitter levels, the HPA axis, oxidative stress, and mitochondrial activity [276-278]. The effect of CBT on suppressing inflammation has been demonstrated in depression [276] by lowering at least one of the inflammatory markers out of CRP, IL-6, and TNF-α [276, 279, 280]. A 12-week session of CBT effectively lowers the level of inflammatory mediators including IL-1Ra, IL-5, IL-6, IL-8, IL-10, granulocyte-colony stimulating factor, interferon-γ (IFN-γ), toll-like receptor 4 (TLR4), nuclear factor-κβ (NF- κβ) and TNF-α [281, 282]. Thus, CBT is promising in controlling inflammation and overcoming insomnia, however, treatment duration needs to be optimized. The therapeutic impact of CBT is mediated through its recruiting ability of emotion-regulatory circuits involving the ventromedial prefrontal cortex, anterior cingulate cortex, and amygdala leading to inhibited fear [283]. Thus, CBT has been established as a crucial therapeutic approach for insomnia and related disorders and should be extensively evaluated to recommend it as a primary treatment before prescribing medication.

Pharmacological interventions

Therapeutic intervention mainly focuses on elevating sleep quality and duration along with improving daytime impairments [284]. If the initial psychological intervention does not respond sufficiently, the pharmacological approach is the immediate approach for the management. CBT has been associated with various limitations such as the unavailability of trained professionals, time-intensive and heavy cost, which lowers its wide application. Recent studies have also indicated its lesser efficacy on objective sleep parameters [285-287]. Thus, to provide immediate relief, easily accessible pharmaceuticals at lower cost with improved efficacy are preferred [284, 288, 289]. Short-term pharmacological treatment could also be supplemented with CBT [289]. Common drugs which are recommended for the treatment of insomnia are benzodiazepine receptor agonists (BZRAs) such as benzodiazepines (eszopiclone, zaleplon, and zolpidem), dual orexin receptor antagonists (suvorexant and lemborexant), sedative antidepressants (doxepin and trazodone), melatonin and melatonin receptor [285, 290-293]. BZRAs and Z-drugs are specifically preferred among older populations, however, their effect is weaker, short-termed, and is also associated with adverse effects such as the increased risk of falls and hip fractures cognitive impairment, daytime confusion, tolerance, and dependence [294, 295]. Further, though many insomnia patients are responsive to BZRAs, it has been evidenced that few patients are not remitted particularly in a comorbid condition that can further cause hypertension, and major depressive disorders [296]. To avoid BZRAs and Z-drug-associated adverse responses, new target sites such as orexin receptors are being explored [297]. Loss of orexinergic neurons has been associated with the condition of excessive sleepiness and narcolepsy [297]. Suvorexant has been reported as the first dual orexin receptor antagonist (DORA) which suppresses activation of the arousal system through reversible binding to orexin receptors. Not many neurophysiological disturbances or any other adverse effects of suvorexant exist except in a drowsy state [297, 298]. Though the associated adverse outcomes such as diurnal somnolence, unconscious nocturnal activity, suicidal ideation, and motor driving impairment have been reported at doses over 20 mg [297, 299]. Suvorexant treatment can improve total sleep time, sleep efficiency, insulin sensitivity, glycemic control, and inhibition of sympathetic activity among type 2 diabetes mellitus [300]. DORA is not being considered a replacement for BZRAs, however, these drugs have the potential to become a considerable alternative to BZRAs [301].

Insomnia Treatment: Progress and novel approaches

Recent progress in insomnia treatments includes traditional and novel therapeutic approaches such as traditional Chinese herbs/medicine (TCH/TCM), acupuncture, pulsed magnetic field therapy (PMFT), and cryotherapy. In further sections, we have comprehensively summarized the developments of the above-mentioned approaches with a greater emphasis on cryotherapy in ameliorating insomnia.

Traditional Chinese herbs (TCH)

TCH has been used for health promotion and prevention of diseases for a long time [302]. TCH alone or as an adjunctive therapy could be an effective tool to provide therapeutic relief among insomnia patients [303]. Wen dan tang, Suan Zao ren tang, Ban xia shu mi tang, suanzaoren, Fuling, Gancao, and Gui pi tang are the most recommended TCH formulation for insomnia [304, 305]. These herbs impart a sedative effect and improve insomnia through GABA or induction of the GABA receptor, suppression of the 5-hydroxytryptamine 1A receptor, and increase in orexin-A, orexin receptor-1, leptin, and leptin receptor level in the brain [305]. Suanzaoren decoction is one of the preferred choices for traditional treatment and its therapeutic potential could be associated with the regulation of Orexin-A, homeostasis of the HPA axis, and neurotransmitters [306]. Insomnia increases two-fold in depression which pushes mortality upward [307-309]. TCH and its formulations such as banxia houpo, chaihu shugansan, Ge Gen, Huang Qin, Dan Shen, Bei Mu, Da Huang Ge-gen-tang, ganmaidazao, Kai-Xin-San, Ping-wei-san, sinisan, She Gan, shuganjieyu, Shu-jing-huo-xue-tang, Shao-yao-gan-cao-tang, wuling, xiaoyaosan, and yueju are effective in the control of depression [309, 310]. Though these herbs might be effective, the herbal formulations are the preferred choice due to the additive effect and detoxication potential which increases the efficacy and safety of TCH [310]. The efficacy of TCH to address the therapeutic needs of insomnia and depression has been evident and could be an essential component of adjunctive therapy. A cohort study demonstrates that the use of TCH lowers the incidence rate of depression from 37.97 to 17.24 per 1000 among insomnia patients [307]. In the various randomized controlled trial (RCT), Suanzaoren decoction has been reported as effective and safe, though the incoherency in methods and fewer studies are limiting factors to establish any conclusion [311]. A meta-analysis found more efficacy of Chaihu Longgu Muli decoction (CLMD) than common drugs in reducing Pittsburgh Sleep Quality Index (PSQI) and improving total sleep time [312]. CLMD regulates the HPA axis, level of neurotransmitters such as norepinephrine, dopamine, and 5-hydroxytryptamine, suppression of adrenocorticotropic hormone and corticosterone, and inhibition of MEK/ERK pathways. Thus, TCH is considered an effective and safe alternative therapy for the treatment of insomnia and related disorders; however, its efficacy might be increased by supplementing other alternatives.

Acupuncture

Acupuncture has been recognized as an effective nonpharmacological traditional intervention for neuroendocrinological disorders such as depression, menopause, and insomnia [313-315]. Short-term clinical impacts of acupuncture in lowering the Pittsburgh sleep quality index (PSQI), Zung self-rating anxiety scale, and Zung self-rating depression scale, indicates improvement in sleep quality, anxiety, and depression [316]. Acupuncture affects the neuroendocrine and immune systems by regulating neurotransmitters like serotonin, norepinephrine, dopamine, endorphins, and glucocorticoids [316, 317]. Traditional acupuncture improves sleep duration for a shorter duration [318], no significant effect impact has also been demonstrated in a case of residual insomnia associated with major depressive disorder [319]. Similarly, evidence shows that auricular acupuncture could be used for symptomatic treatment [320]. Of various acupuncture, scalp acupuncture and electroacupuncture (EA) have been more effective followed by warm and conventional acupuncture [321].

Approved by Food and drug administration (FDA), acupuncture therapy may overcome postoperative and chemotherapy adverse effects such as nausea, vomiting, myofascial pain, headache, low back pain, postoperative dental pain, asthma, fibromyalgia, and osteoarthritis [322-324]. Cancer-related insomnia, anxiety, and depression could be controlled through acupuncture; however, extensive studies are required to reach a consensus [322]. The underlying possible mechanism may be associated with a reduction in mitochondrial oxidative stress, HPA axis, and sympathetic nervous activity along with an increase in the level of ATP, gamma-aminobutyric acid (GABA), GABA(A) receptor, and melatonin [325-329]. Electroacupuncture (EA) inhibits glucose metabolism in the hypothalamus which decreases sympathetic stimulation [330, 331]. A pre-clinical study indicates that acupuncture lowers hippocampal neuronal loss, oxidative and inflammatory stress in vascular dementia by downregulating the expression of a thioredoxin-interacting protein involved in the activation of NOD-like receptor protein 3 (NLRP3) inflammasome [332]. Thus, acupuncture and EA have been reported for their potential impact on insomnia; however, the exact mechanism and concrete supportive evidence need to be further explored.

Electro and pulsed magnetic field therapy (PMFT)

PMFT affects cellular water content, membrane structure and permeability, uptake of oxygen, and nutrients, mitochondrial function, and macrophage migration [333]. The application of alternating current (AC) affects the body’s electrolytes resulting in reversing of functional impairment via increasing blood circulation and cellular oxygen uptake. The magnetic field improves melatonin secretion from the pineal gland which restores circadian rhythm and sleep latency with the increase in sleep duration [333-335]. The impulse magnetic field has also been effective in providing relief from migraine and headaches [336]. In a double-blind controlled study, impulse magnetic field therapy has also demonstrated substantial or even complete relief from insomnia [333].

Further, transcranial magnetic stimulation (TMS), a non-invasive procedure that employs magnetic fields to stimulate nerve cells may depolarize neurons and modulate neuronal activity by generating an electric current [337, 338]. Repetitive TMS (rTMS) has also been found effective in neurological and psychological disorders. In a systematic review, its impact on sleep disorders has been documented; however, with inconsistent results [337].

Notably, BioBoosti, a device emitting a pulse of an electromagnetic field might invigorate endothelial and red blood cells by altering cell membrane charge and increasing microcirculation and perfusion [339]. Two weeks of treatment with BioBoosti significantly enhances slow-wave sleep and decreases night sleep disturbance resulting in improved sleep quality and insomnia symptoms. Interestingly, the magnetic stimulation at acupoints such as Neiguan (PC6), Shenmen (HT7), and Sanyinjiao (SP6) has also improved brain functional networking indicating improvement in sleep quality [340]. Thus, the treatment with a magnetic field seems to be a promising therapy for insomnia. However, extensive studies are needed to understand the comprehensive potential of magnetic therapy.

Electrotherapy is another perspective non-pharmacological therapeutic intervention for insomnia. In cranial electrotherapy stimulation (CES), the microcurrents of 1-1.5 mA affect specific nerve cells, and increase levels of neurotransmitters such as acetylcholine, adrenocorticotrophic hormone, serotonin, dopamine, β-endorphin, cholinesterase, and norepinephrine, and decreases cortisol, leading to increased flow of blood and cerebrospinal fluid [341-344]. These neurochemical alterations reduce pain, stress response, and spasticity, maintain homeostasis, and enhance mood, relaxation, immune function, pain tolerance, and pleasure resulting in improved sleep quality [344]. CES enhances alpha while reducing beta and delta activity associated with relaxation, ruminative thoughts, and fatigue, respectively. In addition, CSE also improves focus, concentration, and mental awareness by influencing beta regions [345]. CES decreases sleep latency and enhances sleep improvement with sleep quality through its implication in the treatment of depression and anxiety needs to be carefully evaluated [346]. CES shows satisfactory improvement in ESAS, HADS, BPI, and PSQI scores among cancer patients indicating relief from anxiety, depression, pain, and insomnia [347]. Sleep hygiene improves the efficacy of CES treatment and both approaches could be recommended together as adjunctive therapy [348]. Consequently, the CES, transcutaneous trigeminal electrical neuromodulation (TTEN) inhibits noradrenergic activity and could be enveloped as a novel treatment option [349]. CES is approved by US FDA and has been effective to control anxiety, depression, and insomnia, however, this treatment is considered the last option and has been associated with mild and limited adverse events such as vertigo, skin irritation, and headache.

Cryotherapy

Cryotherapy was first used successfully to treat rheumatism in the 1970s [350, 351]. In recent years, cryotherapy has been extended to mental disorders, depression, anxiety, inflammatory disorders, pain, and muscle fatigue [352-357]. Cryotherapy has been classified into partial-body (PBC) and whole-body cryotherapy (WBC), and both methods expose the body partial or whole) at a very low temperature of -10°C to -170°C for durations between 20 s to 3 min in cryo-compartments such as cryo-chamber or cryo-cabin [358]. PBC focuses on a single individual and nitrogen is applied directly on body parts excluding the head and neck, whereas WBC involves exposure to extremely cold dry air (usually between -100°C and -140°C) for short durations generally between 2 to 5 minutes [358-360]. Taking this into consideration, even a few global companies including Euda wellness, a subsidiary company of Victory Biotechnology Co., Ltd. Taipei, Taiwan, are offering WBC for insomnia. The therapeutic potential of cryotherapy in pain has been associated with its potential to suppress oxidative stress, inflammation, edema, and neurotransmission in pain fibers [359, 361, 362], by lowering the level of cytokines such as IL-1, IL-6, IL-10, and TNF-α [363]. The WBC further improves the efficacy of kinesiotherapy two-folds resulting in improved pain and spinal movement among ankylosing spondylitis (AS) patients [364] by lowering CRP, fibrinogen, TNF- α, ICAM-1 as well as a considerable reduction in lipid profile, atherosclerosis plaque instability, oxidative stress, [365, 366]. The WBC reduces paraoxonase-1 activity, total oxidative status, inflammatory endothelium parameters, serum CD40L, and serum amyloid A resulting in decreased oxidative stress and a positive effect on human endothelial homeostasis [367]. Computerized cognitive training when applied synergically with WBC could improve cognitive and depressive functions [368, 369]. After cryotherapy, the blood flow in the skin and extremities improves and becomes richer in oxygen, and nutrients [370]. Cryotherapy also stimulates the production of endorphins and anti-inflammatory biomacromolecules [371]. First exposure to cryotherapy may be unpleasant, however, repeated exposure improves the treatment experience [370]. The effectiveness of cryotherapy in regulating cytokines, oxidative, stress inflammatory response, blood flow, oxygen level, fatigue, pain, depression, and anxiety provides an opportunity to improve sleep quality. In a small scale clinical trial, cryo-stimulation (partial body cryo-stimulation at -180 0C for 180 seconds) among professional soccer players indicated a controlled movement in sleep and better sleep quality [372]. On the other hand, two weeks of regular cryo-stimulation exposure at -900C improve somnolence and psychological behavior among the aged population with sleep disturbance and restless leg syndrome [373]. After exhausting training and tournament, the pro-inflammatory cytokines such as TNF-α, and IL-1β, increases which causes systemic inflammation leading to the development of the overreaching syndrome and poor performance [374-376]. A five-day WBC (-120 0C) treatment twice a day can also significantly lowers TNF-α and increase IL-6, indicating the efficacy of cryotherapy in improving the performance of tennis players [376]. The 3-minute WBC improves sleep quality after exercise/training and improves pain relief by ameliorating parasympathetic nervous activity during the slow-wave sleep period [377]. Cryotherapy provides relief from depression and anxiety due to its positive impact on the HPA axis and endogenous opioids [356]. WBC enhances the therapeutic impact of anti-depressant and their response time [378]. In a randomized clinical trial, WBC significantly improved depressive symptoms, quality of life, and self-assessed mood which indicates its additive impact on pharmaceutical treatment [379]. Cryotherapy has also been found effective in tackling tension-type headaches (TTH) associated with stress and mental pressure among students [380]. The needle pain causes anxiety among children and impacts satisfaction during blood collection, which could be suppressed by cryotherapy resulting in increased satisfaction [381]. In addition, the combination of cryotherapy with vibration lowers the fear, anxiety, and anguish among children [382].

The potential of cryotherapy to reduce inflammation and pain has also been explored to assess its impact on OA and other types of arthritis. Cryotherapy with physical exercise further improves pain relief among OA patients [383]. A systematic review evidenced that cryotherapy improves knee pain and motion in OA; however without any significant impact on blood loss during knee replacement surgery [384]. In the rat model, cryotherapy has been demonstrated to decrease synovial fluid leukocyte count and inflammatory cytokine level resulting in improved footprint patterns [385]. Notably, no significant impact of short-term cryotherapy on pain, function, and quality of life improvement among knee OA patients has also been reported [386]. Cryotherapy improves pain in the anterior knee infusion model, though with no significant impact on vastus medialis motoneuron pool excitability [387]. A 10 cycle of WBC significantly lowers the frequency and pain level resulting in a decreased use of analgesics and improved physical activity and well-being [388]. Cryotherapy has been observed to lower synovial power-Doppler ultrasonographic (PDUS) activity and pain among OA patients [389]. The encouraging outcome of WBC has also been observed among RA patients by reducing the level of pro-inflammatory molecules such as IL-6 and TNF-α [390], possibly by decreasing the level of total peroxyl radical trapping antioxidant capacity of plasma [391]. WBC ceases inflammation, and pain along with functional recovery among RA patients which could be associated with its detrimental impact on histamine secretion and degradation [392]. The therapeutic efficacy of cryotherapy could also be correlated with its impact on improving microcirculation, muscle tension, hormonal, and immune response along with analgesic, anti-oedematous and anti-inflammatory effects [388, 393-396]. Oxidative stress is a characteristic phenomenon in neurodegenerative disorders and could be associated with inflammatory reactions [360, 397, 398]. WBC possesses the potential to modulate systemic oxidative stress among MS patients by increasing total antioxidative status [398, 399]. Fatigue in MS could also be a causative agent for insomnia which can be overcome with combined therapy of WBC and kinesiotherapy [400].

The cognitive dysfunction due to insomnia could be addressed through cryotherapy which has become a potent tool for early stage of mild cognitive impairment (MCI) [401]. The progression of MCI to dementia is well-established and up to 64% of patients suffer from dementia [402]. The efficacy of the pharmacological intervention in improving mental health/disorders such as depression and QoL could be further improved by add-on cryotherapy [403, 404]. Decreased level of nitric oxide indicates endothelial dysfunction and oxidative stress, WBC improves inducible nitric oxide synthase though no significant effect has been observed on nitric-oxidative stress or inflammation markers [405]. Cryotherapy possesses the potential to alter hormonal and lipid profiles contributing to its anti-inflammatory and oxidative role which hinders the etiopathogenesis of dementia [390]. Systemic cryotherapy lowers rheumatic activity and related pain; however, no significant change has been observed in the levels of pro-inflammatory and anti-inflammatory cytokines [406]. It has also been observed that cryotherapy significantly improves locomotor activity resulting in encouragement to work and positive self-assessment [407].

Cryosurgery is a minimally invasive approach that is frequently used in the treatment of metastatic liver and prostate cancer [408]. Though the therapy is normally followed by chemo- or radiotherapy to improve its efficacy, it is associated with freeze injury, coagulative necrosis, cell membrane damage, local blood vessel thrombosis, and apoptosis [408, 409]. Cryotherapy is promising in the treatment of liver cancer without any significant bile leakage or mortality, [410]. in combination with low-level laser therapy has been found effective in cancer patients with oral mucositis [411] or bladder cancer [412]. Further, percutaneous cryoablation provides progression-free survival without any major adverse events and a significant reduction in hematuria, urinary irritation, hypogastralgia, and lumbago [413]. The cryotherapy intervention improves insomnia and associated disorders resulting in ameliorated QoL; These scientific reports are evident in exploring the therapeutic role of cryotherapy; however, extensive studies are required to establish the underlying mechanistic insight.

Biomaterials, stem cells and insomnia

Traditional drug delivery restricts the therapeutic outcomes which necessitate the development of novel, effective and safer alternatives [414]. Biomaterials such as biopolymers and synthetic biopolymers have been widely explored in the controlled transdermal, intranasal, and transbuccal delivery of drugs [414, 415]. The oral uptake of melatonin and other insomnia drugs results in low bioavailability, and limited efficacy [416, 417]. To overcome the low bioavailability, various attempts have been made to develop drug delivery systems. A single-walled carbon nanotube (SWCN) improved bio-absorption of zaleplon (sedative-hypnotic) via nasal delivery, by increasing carbon nanotube-mediated blood-brain crossing ability [417], leading to prolonged neuroprotective effect. Melatonin is another suitable therapeutic candidate for insomnia which could be transdermally delivered due to its low molecular weight [415, 418-420]. A microneedle patch synthesized from proline, melatonin and silk fibroin has been found effective in maintaining drug concentration in blood and demonstrates physiological resemblance in drug release in Sprague-Dawley rat models of insomnia [416]. Moreover, soya phosphatidylcholine and sodium deoxycholate-based elastic liposomes may facilitate melatonin release by enhancing transdermal flux and reducing lag time; therefore, could be helpful in improving insomnia [415]. Thus, biomaterials-based drug delivery seems an effective approach to better the therapeutic outcomes of insomnia drugs. However, more extensive clinical and scientific studies are required to select the most effective and safe delivery systems.

Another regenerative biomaterial therapies include stem cells, which have become a prominent choice for treatment of various degenerative disorders. Sleep fragmentation has also been correlated with stem cells. Specifically, it impacts the immunity and functioning of hematopoietic stem cells (HSPC) through epigenetic modification, leading to aging of hematopoietic system and collapse of clonal diversity and homogenization of myeloid pool [421]. Notably, insomnia-related genes such as MADD, CASP9, PLEKHM2 have been found significantly enriched in neural stem cells (NSCs) [422]. The NSC also plays a key role in genesis of sleep-waking cycle switch-associated neurons. Though no direct studies exist, this above-mentioned evidence indicates that stem cells therapies may be helpful in generation of sleep-related neurons. Consequently, this may also assist in the improvement of insomnia associated complications.

Prospects of Combinatorial cryotherapy: Future directions for augmenting therapeutic impacts

Cryotherapy in combination with other therapeutic approaches implies the potential to improve insomnia symptoms (Fig. 4). It has been applied as an add-on therapy to pharmacological treatment for the treatment of depression [379] by regulating pain, inflammation, oxidative stress, immune response, and the circulation system [356]. WBC's role in enhancing the level of pro-inflammatory cytokines and lowering the circulation of pro-inflammatory molecules along with its potential to regulate the hypothalamic-pituitary-adrenal (HPA) axis could support pharmacological outcomes of drugs in insomnia treatment. Moreover, the adverse effect of pharmaceutics such as cognitive impairment, and daytime confusion could be addressed by cryotherapy [401, 423]. Insomnia related to multiple sclerosis, osteoarthritis, chronic pain, rheumatoid arthritis, gastrointestinal disorders, cardiovascular disorders, and fibromyalgia could also be managed by cryotherapy to avoid contraindications of pharmaceutical treatments for co-morbid insomnia [397, 424, 425]. Cryotherapy could augment antidepressant efficacy in the treatment of depression and present evidence to assess the efficacy of cryotherapy in the synergistic treatment of insomnia [378]. The addition of cryotherapy could enhance recovery during CBT as cryotherapy provided cognitive relief, regulates the HPA axis, and reduce the severity of stress, anxiety, and depression. The synergistic effect of cryotherapy on CBT not only improves cognitive function, memory, and focus but also provides mild relief from depression [426]. Cryotherapy at Hoku point lowered pain during hemodialysis which indicates that a combination of acupressure and cryotherapy has the potential to address the therapeutic need of pain-associated insomnia [427]. TCH efficacy in controlling pain, oxidative stress, inflammation, and circulation of inflammatory molecules could be supported by using cryotherapy resulting in improved outcomes for insomnia treatment. Though the additive /synergistic impact of cryotherapy is promising, more studies are required to establish a comprehensive procedure and conclusion to implicate the efficacy and safety of combinatorial therapy.

Figure 4.

Figure 4.

Possible synergistic application of whole-body therapy and other alternatives including PMFT, TCM, CBT, and acupressure for insomnia. This combination therapy may decrease the level of inflammatory molecules (IL-1, IL-6, IL-10, TNF-α, and CRP), oxidative stress, edema, pain neurotransmission, and lipid profile. The limitations of cryotherapy include transient erythema, edema, cold urticaria, mild pain, swelling and redness, blistering, crusting, color alteration, scar, and viral infection in rare cases, which might be recovered through other combinational therapeutic alternatives. TCM: Traditional Chinese Medicine, CBT: Cognitive behavioral therapy, PMFT: Electro- and Pulsed Magnetic Field therapy. IL-Interleukin, TNF: Tumour necrosis factor, CRP: C-reactive protein.

Limitation of Cryotherapy in Insomnia

Cryotherapy is emerging as a novel non-invasive therapeutic option and studies have reported safe and effective if recommended treatment guidelines are being followed. After cryotherapy, the significant increase in WBC count has been associated with mild side effects such as pain, swelling, redness, blistering, crusting, color alteration, scars, and viral infection in rare cases [428]. Common adverse effects do not require any specific medical attention; however, increased pain at the treatment site, excessive redness, and swelling need medical attention. Cryosurgery could cause transient erythema, edema, and cold urticaria [429], but antihistamines such as cetirizine, loratadine, desloratadine, ebastine, leukotriene receptor antagonists, sulfasalazine, immunosuppressive agents like methotrexate, cyclosporine, omalizumab, and systemic glucocorticoids like prednisolone could be assistive to overcome cold urticaria [430, 431]. Rupatadine, a second-generation antagonist of histamine receptor and platelet-activating factor, at 20mg/day has been found effective and tolerated during acquired cold urticaria [432]. The procedural coherence, availability of expertise, cryo-chamber, and short-term relief are limiting factors for wide acceptability. Though there have been many positive outcomes of cryotherapy as well as its synergistic administration with other alternatives for insomnia, more studies are required to understand the mechanism and establish a uniform therapeutic procedure for the treatment of sleep disorders.

Key challenges for the treatment of insomnia

Insomnia is one of the most overlooked conditions and is not addressed until it poses serious health risk. There is a wide spectrum of treatment approaches available; however, their selection and effectiveness depend on different factors. The pros and cons of available and prospective therapies are summarized in Table 1. The complex association between insomnia and other comorbidities is not well-understood which poses a challenge in establishing proper treatment [433].

Table 1.

List of pros and cons of various treatment alternatives of insomnia.

Therapy Pros Cons References
CBT Economical, safe, and highly effective Lack of trained CBT professionals,
Need awareness among acceptance of therapy.
[434]
Pharmacological
interventions
Easily accessible, effects are visible in a short time, widely accepted routine therapy Tolerance, abuse, associated adverse events, and adverse psychological impact [435, 436]
TCH Effective in the treatment of chronic and incurable diseases. Lack of clinical trials and pre-clinical studies [305, 437, 438]
Acupuncture Improves clinical efficacy, Proven traditional therapeutic approach Lack of consistent data in clinical studies and pre-clinical multiple centers randomized clinical studies [439, 440]
PMFT Effective in alleviating insomnia, quick reaction time Safer approach compared to drug therapy [441]
Cryotherapy Non-invasive, safe, and effective Adverse events such as erythema, edema, and cold urticaria. availability of expertise, cryo-chamber, and short-term relief [442, 443]
Biomaterials and stem cell Regenerative therapy, improvement in the delivery of therapeutic agents Lack of pre-clinical, and clinical studies, [414, 415, 421, 422]

CBT: Cognitive-Behavioral Therapy, TCH: Traditional Chinese herbs, PMFT: Electro and pulsed magnetic field therapy.

Conclusions

Recent adverse changes in lifestyle, work culture, and social behavior along with the COVID-19 pandemic situation have exponentially increased stress and other etiological factors which have resulted in disrupted circadian rhythm. The increase in insomnia incidence is moving towards being epidemic which will further deteriorate individual physical and psychological health. Considering the urgency to address this situation, current interventions such as CBT and pharmaceutical therapies supported by lifestyle changes have made considerable progress. In particular, cryotherapy has been reported to reduce oxidative stress, pain, and inflammation resulting in improved sleep efficiency. Novel approaches such as the synergy of cryotherapy and pharmacological/ traditional Chinese medicine/ bioboosti/ electromagnetic and acupressure could provide a more effective way to address the therapeutic needs of insomnia, by suppressing inflammation, oxidative stress, edema, lipid profile, and pain neurotransmission; however, extensive studies are required to establish its broader applicability.

Footnotes

Disclosure statement

The authors have no conflicts of interest to declare.

References

  • [1].Léger D, Bayon V (2010). Societal costs of insomnia. Sleep Medicine Reviews, 14:379-389. [DOI] [PubMed] [Google Scholar]
  • [2].Patel D, Steinberg J, Patel P (2018). Insomnia in the Elderly: A Review. Journal of clinical sleep medicine : JCSM : official publication of the American Academy of Sleep Medicine, 14:1017-1024. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [3].Palesh O, Scheiber C, Kesler S, Janelsins MC, Guido JJ, Heckler C, et al. (2018). Feasibility and acceptability of brief behavioral therapy for cancer-related insomnia: effects on insomnia and circadian rhythm during chemotherapy: a phase II randomised multicentre controlled trial. British Journal of Cancer, 119:274-281. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [4].Levenson JC, Kay DB, Buysse DJ (2015). The pathophysiology of insomnia. Chest, 147:1179-1192. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [5].Kandeger A, Selvi Y, Tanyer DK (2019). The effects of individual circadian rhythm differences on insomnia, impulsivity, and food addiction. Eat Weight Disord, 24:47-55. [DOI] [PubMed] [Google Scholar]
  • [6].Perlis M, Gehrman P (2013). Types of insomnia. Encyclopedia of Sleep: 199-202. [Google Scholar]
  • [7].Walsh JK, Coulouvrat C, Hajak G, Lakoma MD, Petukhova M, Roth T, et al. (2011). Nighttime insomnia symptoms and perceived health in the America Insomnia Survey (AIS). Sleep, 34:997-1011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [8].Besedovsky L, Lange T, Haack M (2019). The Sleep-Immune Crosstalk in Health and Disease. Physiol Rev, 99:1325-1380. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [9].Garbarino S, Lanteri P, Bragazzi NL, Magnavita N, Scoditti E (2021). Role of sleep deprivation in immune-related disease risk and outcomes. Communications biology, 4:1304-1304. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [10].Irwin MR (2015). Why Sleep Is Important for Health: A Psychoneuroimmunology Perspective. Annual Review of Psychology, 66:143-172. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [11].Bjorvatn B, Jernelöv S, Pallesen S (2021). Insomnia - A Heterogenic Disorder Often Comorbid With Psychological and Somatic Disorders and Diseases: A Narrative Review With Focus on Diagnostic and Treatment Challenges. Frontiers in Psychology, 12:289. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [12].Bonilla-Jaime H, Zeleke H, Rojas A, Espinosa-Garcia C (2021). Sleep Disruption Worsens Seizures: Neuroinflammation as a Potential Mechanistic Link. International journal of molecular sciences, 22:12531. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [13].Irwin MR (2019). Sleep and inflammation: partners in sickness and in health. Nat Rev Immunol, 19:702-715. [DOI] [PubMed] [Google Scholar]
  • [14].Cheng Z, Mendolia S, Paloyo AR, Savage DA, Tani M (2021). Working parents, financial insecurity, and childcare: mental health in the time of COVID-19 in the UK. Rev Econ Househ: 1-22. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [15].Besedovsky L, Lange T, Haack M (2019). The Sleep-Immune Crosstalk in Health and Disease. Physiological reviews, 99:1325-1380. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [16].Attele AS, Xie J-T, Yuan C-S (2000). Treatment of insomnia: an alternative approach. Alternative Medicine Review, 5:249-259. [PubMed] [Google Scholar]
  • [17].Attele AS, Xie JT, Yuan CS (2000). Treatment of insomnia: an alternative approach. Altern Med Rev, 5:249-259. [PubMed] [Google Scholar]
  • [18].Baranov A, Pakhomov O, Fedorov A, Ivanov V, Zaitsev A, Polyakov R (2019). Technique and Technology of Whole-Body Cryotherapy (WBC). In Morosuk T and Sultan M (Eds.), Low-temperature Technologies. IntechOpen. [Google Scholar]
  • [19].Hawkins J, Reeder M, Heumann K (2017). Cryotherapy - A call for further research. Medical Research Archives, 5. [Google Scholar]
  • [20].Grandner MA, Smith TE, Jackson N, Jackson T, Burgard S, Branas C (2015). Geographic distribution of insufficient sleep across the United States: a county-level hotspot analysis. Sleep Health, 1:158-165. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [21].Tang J, Liao Y, Kelly BC, Xie L, Xiang YT, Qi C, et al. (2017). Gender and Regional Differences in Sleep Quality and Insomnia: A General Population-based Study in Hunan Province of China. Sci Rep, 7:43690. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [22].Ford ES, Wheaton AG, Cunningham TJ, Giles WH, Chapman DP, Croft JB (2014). Trends in outpatient visits for insomnia, sleep apnea, and prescriptions for sleep medications among US adults: findings from the National Ambulatory Medical Care survey 1999-2010. Sleep, 37:1283-1293. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [23].Morin CM, LeBlanc M, Daley M, Gregoire JP, Mérette C (2006). Epidemiology of insomnia: prevalence, self-help treatments, consultations, and determinants of help-seeking behaviors. Sleep Med, 7:123-130. [DOI] [PubMed] [Google Scholar]
  • [24].Bjorøy I, Jørgensen VA, Pallesen S, Bjorvatn B (2020). The Prevalence of Insomnia Subtypes in Relation to Demographic Characteristics, Anxiety, Depression, Alcohol Consumption and Use of Hypnotics. Frontiers in Psychology, 11. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [25].Wade AG (2010). The societal costs of insomnia. Neuropsychiatric disease and treatment, 7:1-18. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [26].Dündar Y, Boland A, Strobl J, Dodd S, Haycox A, Bagust A, et al. (2004). Newer hypnotic drugs for the short-term management of insomnia: a systematic review and economic evaluation. Health Technol Assess, 8:iii-x, 1-125. [DOI] [PubMed] [Google Scholar]
  • [27].Fossum IN, Nordnes LT, Storemark SS, Bjorvatn B, Pallesen S (2014). The Association Between Use of Electronic Media in Bed Before Going to Sleep and Insomnia Symptoms, Daytime Sleepiness, Morningness, and Chronotype. Behavioral Sleep Medicine, 12:343-357. [DOI] [PubMed] [Google Scholar]
  • [28].Janson C, Lindberg E, Gislason T, Elmasry A, Boman G (2001). Insomnia in men-a 10-year prospective population based study. Sleep, 24:425-430. [DOI] [PubMed] [Google Scholar]
  • [29].Flo E, Pallesen S, Åkerstedt T, Magerøy N, Moen BE, Grønli J, et al. (2013). Shift-related sleep problems vary according to work schedule. Occup Environ Med, 70:238-245. [DOI] [PubMed] [Google Scholar]
  • [30].Hale L, Guan S (2015). Screen time and sleep among school-aged children and adolescents: a systematic literature review. Sleep medicine reviews, 21:50-58. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [31].Hjetland GJ, Skogen JC, Hysing M, Sivertsen B (2021). The Association Between Self-Reported Screen Time, Social Media Addiction, and Sleep Among Norwegian University Students. Frontiers in public health, 9:794307-794307. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [32].Royant-Parola S, Londe V, Tréhout S, Hartley S (2018). [The use of social media modifies teenagers' sleep-related behavior]. Encephale, 44:321-328. [DOI] [PubMed] [Google Scholar]
  • [33].Bláfoss R, Sundstrup E, Jakobsen MD, Bay H, Garde AH, Andersen LL (2019). Are Insomnia Type Sleep Problems Associated With a Less Physically Active Lifestyle? A Cross-Sectional Study Among 7,700 Adults From the General Working Population. Frontiers in Public Health, 7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [34].Roy SK, Bhattacharjee AK, Chakraborti C, Singh R (2015). Prevalence of insomnia in urban population of West Bengal: A community-based cross-sectional study. International Journal of Medicine and Public Health, 5. [Google Scholar]
  • [35].Panda S, Taly AB, Sinha S, Gururaj G, Girish N, Nagaraja D (2012). Sleep-related disorders among a healthy population in South India. Neurol India, 60:68-74. [DOI] [PubMed] [Google Scholar]
  • [36].Bhaskar S, Hemavathy D, Prasad S (2016). Prevalence of chronic insomnia in adult patients and its correlation with medical comorbidities. Journal of family medicine and primary care, 5:780-784. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [37].Cao XL, Wang SB, Zhong BL, Zhang L, Ungvari GS, Ng CH, et al. (2017). The prevalence of insomnia in the general population in China: A meta-analysis. PLoS One, 12:e0170772. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [38].Mallon L, Broman J-E, Åkerstedt T, Hetta J (2014). Insomnia in Sweden: A Population-Based Survey. Sleep Disorders, 2014:843126. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [39].Ohayon MM, Bader G (2010). Prevalence and correlates of insomnia in the Swedish population aged 19-75 years. Sleep Medicine, 11:980-986. [DOI] [PubMed] [Google Scholar]
  • [40].Morphy H, Dunn KM, Lewis M, Boardman HF, Croft PR (2007). Epidemiology of insomnia: a longitudinal study in a UK population. Sleep, 30:274-280. [PubMed] [Google Scholar]
  • [41].Baglioni C, Altena E, Bjorvatn B, Blom K, Bothelius K, Devoto A, et al. (2020). The European Academy for Cognitive Behavioural Therapy for Insomnia: An initiative of the European Insomnia Network to promote implementation and dissemination of treatment. Journal of sleep research, 29:e12967. [DOI] [PubMed] [Google Scholar]
  • [42].Ohayon MM, Smirne S (2002). Prevalence and consequences of insomnia disorders in the general population of Italy. Sleep medicine, 3:115-120. [DOI] [PubMed] [Google Scholar]
  • [43].Leger D, Guilleminault C, Dreyfus JP, Delahaye C, Paillard M (2000). Prevalence of insomnia in a survey of 12,778 adults in France. J Sleep Res, 9:35-42. [DOI] [PubMed] [Google Scholar]
  • [44].Beck F, Richard JB, Léger D (2013). [Insomnia and total sleep time in France: prevalence and associated socio-demographic factors in a general population survey]. Revue neurologique, 169:956-964. [DOI] [PubMed] [Google Scholar]
  • [45].Khaled SM, Petcu C, Al-Thani MA, Al-Hamadi AMHA, Daher-Nashif S, Zolezzi M, et al. (2021). Prevalence and associated factors of DSM-5 insomnia disorder in the general population of Qatar. BMC Psychiatry, 21:84. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [46].Chung S, Cho SW, Jo M-W, Youn S, Lee J, Sim CS (2020). The Prevalence and Incidence of Insomnia in Korea during 2005 to 2013. Psychiatry investigation, 17:533-540. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [47].Julie A Dopheide PBF (2020). Insomnia Overview: Epidemiology, Pathophysiology, Diagnosis and Monitoring, and Nonpharmacologic Therapy. Supplements and Featured Publications, 26. [DOI] [PubMed] [Google Scholar]
  • [48].Lallukka T, Sares-Jäske L, Kronholm E, Sääksjärvi K, Lundqvist A, Partonen T, et al. (2012). Sociodemographic and socioeconomic differences in sleep duration and insomnia-related symptoms in Finnish adults. BMC public Health, 12:1-22. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [49].Murtagh KN, Hubert HB (2004). Gender differences in physical disability among an elderly cohort. American journal of public health, 94:1406-1411. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [50].Zeng L-N, Zong Q-Q, Yang Y, Zhang L, Xiang Y-F, Ng CH, et al. (2020). Gender Difference in the Prevalence of Insomnia: A Meta-Analysis of Observational Studies. Frontiers in Psychiatry, 11. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [51].Baker FC, de Zambotti M, Colrain IM, Bei B (2018). Sleep problems during the menopausal transition: prevalence, impact, and management challenges. Nature and science of sleep, 10:73-95. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [52].Ahmed AE, Al-Jahdali H, Fatani A, Al-Rouqi K, Al-Jahdali F, Al-Harbi A, et al. (2017). The effects of age and gender on the prevalence of insomnia in a sample of the Saudi population. Ethn Health, 22:285-294. [DOI] [PubMed] [Google Scholar]
  • [53].Jehan S, Myers AK, Zizi F, Pandi-Perumal SR, Jean-Louis G, Singh N, et al. (2018). Sleep health disparity: the putative role of race, ethnicity and socioeconomic status. Sleep medicine and disorders : international journal, 2:127-133. [PMC free article] [PubMed] [Google Scholar]
  • [54].Martinez SA, Beebe LA, Thompson DM, Wagener TL, Terrell DR, Campbell JE (2018). A structural equation modeling approach to understanding pathways that connect socioeconomic status and smoking. PLoS One, 13:e0192451. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [55].Grandner MA, Williams NJ, Knutson KL, Roberts D, Jean-Louis G (2016). Sleep disparity, race/ethnicity, and socioeconomic position. Sleep medicine, 18:7-18. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [56].Morin CM, Carrier J (2021). The acute effects of the COVID-19 pandemic on insomnia and psychological symptoms. Sleep medicine, 77:346-347. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [57].Alimoradi Z, Broström A, Tsang HWH, Griffiths MD, Haghayegh S, Ohayon MM, et al. (2021). Sleep problems during COVID-19 pandemic and its’ association to psychological distress: A systematic review and meta-analysis. EClinicalMedicine, 36. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [58].Tahmasian M, Samea F, Khazaie H, Zarei M, Kharabian Masouleh S, Hoffstaedter F, et al. (2020). The interrelation of sleep and mental and physical health is anchored in grey-matter neuroanatomy and under genetic control. Communications Biology, 3:171. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [59].Bonnet M, Arand D (1997). Hyperarousal and insomnia. Sleep medicine reviews, 1:97-108. [DOI] [PubMed] [Google Scholar]
  • [60].Riemann D, Spiegelhalder K, Feige B, Voderholzer U, Berger M, Perlis M, et al. (2010). The hyperarousal model of insomnia: a review of the concept and its evidence. Sleep medicine reviews, 14:19-31. [DOI] [PubMed] [Google Scholar]
  • [61].Perlis M, Giles D, Mendelson W, Bootzin R, Wyatt J (1997). Psychophysiological insomnia: the behavioural model and a neurocognitive perspective. Journal of sleep research, 6:179-188. [DOI] [PubMed] [Google Scholar]
  • [62].Kalmbach DA, Buysse DJ, Cheng P, Roth T, Yang A, Drake CL (2020). Nocturnal cognitive arousal is associated with objective sleep disturbance and indicators of physiologic hyperarousal in good sleepers and individuals with insomnia disorder. Sleep medicine, 71:151-160. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [63].Dworak M, Schierl T, Bruns T, Strüder H (2007). Impact of Singular Excessive Computer Game and Television Exposure on Sleep Patterns and Memory Performance of School-aged Children. Pediatrics, 120:978-985. [DOI] [PubMed] [Google Scholar]
  • [64].Higuchi S, Motohashi Y, Liu Y, Maeda A (2005). Effects of playing a computer game using a bright display on presleep physiological variables, sleep latency, slow wave sleep and REM sleep. Journal of sleep research, 14:267-273. [DOI] [PubMed] [Google Scholar]
  • [65].Van den Bulck J (2004). Television Viewing, Computer Game Playing, and Internet Use and Self-Reported Time to Bed and Time out of Bed in Secondary-School Children. Sleep, 27:101-104. [DOI] [PubMed] [Google Scholar]
  • [66].Wuyts J, De Valck E, Vandekerckhove M, Pattyn N, Bulckaert A, Berckmans D, et al. (2012). The influence of pre-sleep cognitive arousal on sleep onset processes. Int J Psychophysiol, 83:8-15. [DOI] [PubMed] [Google Scholar]
  • [67].Hantsoo L, Khou CS, White CN, Ong JC (2013). Gender and cognitive-emotional factors as predictors of pre-sleep arousal and trait hyperarousal in insomnia. Journal of psychosomatic research, 74:283-289. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [68].Ren R, Zhang Y, Yang L, Sanford LD, Tang X (2021). Insomnia with physiological hyperarousal is associated with lower weight: a novel finding and its clinical implications. Translational Psychiatry, 11:604. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [69].Vgontzas AN, Fernandez-Mendoza J, Liao D, Bixler EO (2013). Insomnia with objective short sleep duration: the most biologically severe phenotype of the disorder. Sleep medicine reviews, 17:241-254. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [70].Li Y, Vgontzas AN, Fernandez-Mendoza J, Bixler EO, Sun Y, Zhou J, et al. (2015). Insomnia With Physiological Hyperarousal Is Associated With Hypertension. Hypertension, 65:644-650. [DOI] [PubMed] [Google Scholar]
  • [71].Edinger JD, Means MK, Krystal AD (2013). Does physiological hyperarousal enhance error rates among insomnia sufferers? Sleep, 36:1179-1186. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [72].Kalmbach DA, Cuamatzi-Castelan AS, Tonnu CV, Tran KM, Anderson JR, Roth T, et al. (2018). Hyperarousal and sleep reactivity in insomnia: current insights. Nat Sci Sleep, 10:193-201. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [73].Bruno A, Rizzo A, Muscatello MRA, Celebre L, Silvestri MC, Zoccali RA, et al. (2020). Hyperarousal Scale: Italian Cultural Validation, Age and Gender Differences in a Nonclinical Population. Int J Environ Res Public Health, 17. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [74].Riemann D, Spiegelhalder K, Feige B, Voderholzer U, Berger M, Perlis M, et al. (2010). The hyperarousal model of insomnia: a review of the concept and its evidence. Sleep Med Rev, 14:19-31. [DOI] [PubMed] [Google Scholar]
  • [75].Nofzinger EA, Buysse DJ, Germain A, Price JC, Miewald JM, Kupfer DJ (2004). Functional neuroimaging evidence for hyperarousal in insomnia. Am J Psychiatry, 161:2126-2128. [DOI] [PubMed] [Google Scholar]
  • [76].Palagini L, Moretto U, Novi M, Masci I, Caruso D, Drake CL, et al. (2018). Lack of Resilience Is Related to Stress-Related Sleep Reactivity, Hyperarousal, and Emotion Dysregulation in Insomnia Disorder. Journal of clinical sleep medicine : JCSM : official publication of the American Academy of Sleep Medicine, 14:759-766. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [77].Palagini L, Biber K, Riemann D (2014). The genetics of insomnia--evidence for epigenetic mechanisms? Sleep Med Rev, 18:225-235. [DOI] [PubMed] [Google Scholar]
  • [78].Grima M, Hunter T, Zhang Y (2017) Molecular mechanisms of the sleep wake cycle : therapeutic applications to insomnia. [Google Scholar]
  • [79].Moore RY (2007). Suprachiasmatic nucleus in sleep-wake regulation. Sleep Med, 8 Suppl 3:27-33. [DOI] [PubMed] [Google Scholar]
  • [80].Lind MJ, Gehrman PR (2016). Genetic Pathways to Insomnia. Brain sciences, 6:64. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [81].Bragantini D, Sivertsen B, Gehrman P, Lydersen S, Güzey IC (2019). Variations in circadian genes and individual nocturnal symptoms of insomnia. The HUNT study. Chronobiol Int, 36:681-688. [DOI] [PubMed] [Google Scholar]
  • [82].Dijk DJ, Archer SN (2010). PERIOD3, circadian phenotypes, and sleep homeostasis. Sleep Med Rev, 14:151-160. [DOI] [PubMed] [Google Scholar]
  • [83].Archer SN, Schmidt C, Vandewalle G, Dijk DJ (2018). Phenotyping of PER3 variants reveals widespread effects on circadian preference, sleep regulation, and health. Sleep Med Rev, 40:109-126. [DOI] [PubMed] [Google Scholar]
  • [84].Byrne EM, Gehrman PR, Medland SE, Nyholt DR, Heath AC, Madden PA, et al. (2013). A genome-wide association study of sleep habits and insomnia. Am J Med Genet B Neuropsychiatr Genet, 162b:439-451. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [85].Bragantini D, Sivertsen B, Gehrman P, Lydersen S, Güzey IC (2019). Genetic polymorphisms associated with sleep-related phenotypes; relationships with individual nocturnal symptoms of insomnia in the HUNT study. BMC medical genetics, 20:179-179. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [86].Xie Y, Zhao Y, Zhou L, Zhao L, Wang J, Ma W, et al. (2020). Gene polymorphisms (rs324957, rs324981) in NPSR1 are associated with increased risk of primary insomnia: A cross-sectional study. Medicine, 99:e21598-e21598. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [87].Sun H, Zhang J, Ma Y, Liu J (2020). Integrative genomics analysis identifies five promising genes implicated in insomnia risk based on multiple omics datasets. Bioscience reports, 40:BSR20201084. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [88].Schmitt K, Holsboer-Trachsler E, Eckert A (2016). BDNF in sleep, insomnia, and sleep deprivation. Ann Med, 48:42-51. [DOI] [PubMed] [Google Scholar]
  • [89].Kaur H, Spurling BC, Bollu PC (2021). Chronic insomnia. StatPearls [Internet]. [PubMed] [Google Scholar]
  • [90].Li F, Li S, Liu Y, Cao K, Yang M (2016). Effect of Heweianshen Decoction on Orexin-A and Cholecystokinin-8 Expression in Rat Models of Insomnia. Evidence-Based Complementary and Alternative Medicine, 2016:8034263. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [91].Adamantidis AR, Zhang F, Aravanis AM, Deisseroth K, De Lecea L (2007). Neural substrates of awakening probed with optogenetic control of hypocretin neurons. Nature, 450:420-424. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [92].Inutsuka A, Yamanaka A (2013). The physiological role of orexin/hypocretin neurons in the regulation of sleep/wakefulness and neuroendocrine functions. Front Endocrinol (Lausanne), 4:18. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [93].Inutsuka A, Yamanaka A (2013). The regulation of sleep and wakefulness by the hypothalamic neuropeptide orexin/hypocretin. Nagoya J Med Sci, 75:29-36. [PMC free article] [PubMed] [Google Scholar]
  • [94].Saper CB, Scammell TE, Lu J (2005). Hypothalamic regulation of sleep and circadian rhythms. Nature, 437:1257-1263. [DOI] [PubMed] [Google Scholar]
  • [95].Sakurai T, Nagata R, Yamanaka A, Kawamura H, Tsujino N, Muraki Y, et al. (2005). Input of Orexin/Hypocretin Neurons Revealed by a Genetically Encoded Tracer in Mice. Neuron, 46:297-308. [DOI] [PubMed] [Google Scholar]
  • [96].Cheng J, Wu F, Zhang M, Ding D, Fan S, Chen G, et al. (2020). The Interaction Between the Ventrolateral Preoptic Nucleus and the Tuberomammillary Nucleus in Regulating the Sleep-Wakefulness Cycle. Front Neurosci, 14:615854. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [97].Kroeger D, Absi G, Gagliardi C, Bandaru SS, Madara JC, Ferrari LL, et al. (2018). Galanin neurons in the ventrolateral preoptic area promote sleep and heat loss in mice. Nature Communications, 9:4129. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [98].Venner A, De Luca R, Sohn LT, Bandaru SS, Verstegen AMJ, Arrigoni E, et al. (2019). An Inhibitory Lateral Hypothalamic-Preoptic Circuit Mediates Rapid Arousals from Sleep. Current Biology, 29:4155-4168.e4155. [DOI] [PubMed] [Google Scholar]
  • [99].Lombardi F, Gómez-Extremera M, Bernaola-Galván P, Vetrivelan R, Saper CB, Scammell TE, et al. (2020). Critical Dynamics and Coupling in Bursts of Cortical Rhythms Indicate Non-Homeostatic Mechanism for Sleep-Stage Transitions and Dual Role of VLPO Neurons in Both Sleep and Wake. The Journal of Neuroscience, 40:171. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [100].Irwin M, Thompson J, Miller C, Gillin JC, Ziegler M (1999). Effects of sleep and sleep deprivation on catecholamine and interleukin-2 levels in humans: clinical implications. J Clin Endocrinol Metab, 84:1979-1985. [DOI] [PubMed] [Google Scholar]
  • [101].Irwin M, Clark C, Kennedy B, Christian Gillin J, Ziegler M (2003). Nocturnal catecholamines and immune function in insomniacs, depressed patients, and control subjects. Brain Behav Immun, 17:365-372. [DOI] [PubMed] [Google Scholar]
  • [102].Thakkar MM (2011). Histamine in the regulation of wakefulness. Sleep medicine reviews, 15:65-74. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [103].Morgan PT, Pace-Schott EF, Mason GF, Forselius E, Fasula M, Valentine GW, et al. (2012). Cortical GABA Levels in Primary Insomnia. Sleep, 35:807-814. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [104].Plante DT, Jensen JE, Winkelman JW (2012). The role of GABA in primary insomnia. Sleep, 35:741-742. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [105].Park S, Kang I, Edden RAE, Namgung E, Kim J, Kim J (2020). Shorter sleep duration is associated with lower GABA levels in the anterior cingulate cortex. Sleep Medicine, 71:1-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [106].Porkka-Heiskanen T, Strecker RE, Thakkar M, Bjorkum AA, Greene RW, McCarley RW (1997). Adenosine: a mediator of the sleep-inducing effects of prolonged wakefulness. Science, 276:1265-1268. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [107].Ren C-Y, Rao J-X, Zhang X-X, Zhang M, Xia L, Chen G-H (2021). Changed signals of blood adenosine and cytokines are associated with parameters of sleep and/or cognition in the patients with chronic insomnia disorder. Sleep Medicine, 81:42-51. [DOI] [PubMed] [Google Scholar]
  • [108].Nam HW, McIver SR, Hinton DJ, Thakkar MM, Sari Y, Parkinson FE, et al. (2012). Adenosine and glutamate signaling in neuron-glial interactions: implications in alcoholism and sleep disorders. Alcoholism, clinical and experimental research, 36:1117-1125. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [109].Ursin R (2002). Serotonin and sleep. Sleep Med Rev, 6:55-69. [DOI] [PubMed] [Google Scholar]
  • [110].Portas CM, Bjorvatn B, Ursin R (2000). Serotonin and the sleep/wake cycle: special emphasis on microdialysis studies. Prog Neurobiol, 60:13-35. [DOI] [PubMed] [Google Scholar]
  • [111].Vashadze S (2007). [Insomnia, serotonin and depression]. Georgian medical news:22-24. [PubMed] [Google Scholar]
  • [112].Melancon MO, Lorrain D, Dionne IJ (2014). Exercise and sleep in aging: emphasis on serotonin. Pathol Biol (Paris), 62:276-283. [DOI] [PubMed] [Google Scholar]
  • [113].Lemoine P, Garfinkel D, Laudon M, Nir T, Zisapel N (2011). Prolonged-release melatonin for insomnia - an open-label long-term study of efficacy, safety, and withdrawal. Therapeutics and clinical risk management, 7:301-311. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [114].Zisapel N (2007). Sleep and sleep disturbances: biological basis and clinical implications. Cell Mol Life Sci, 64:1174-1186. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [115].Tan DX, Manchester LC, Reiter RJ (2016). CSF generation by pineal gland results in a robust melatonin circadian rhythm in the third ventricle as an unique light/dark signal. Med Hypotheses, 86:3-9. [DOI] [PubMed] [Google Scholar]
  • [116].Blask DE (2009). Melatonin, sleep disturbance and cancer risk. Sleep Med Rev, 13:257-264. [DOI] [PubMed] [Google Scholar]
  • [117].Vasey C, McBride J, Penta K (2021). Circadian Rhythm Dysregulation and Restoration: The Role of Melatonin. Nutrients, 13:3480. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [118].Rogers NL, Dinges DF, Kennaway DJ, Dawson D (2003). Potential action of melatonin in insomnia. SLEEP-NEW YORK THEN WESTCHESTER-, 26:1058-1059. [PubMed] [Google Scholar]
  • [119].Yoshihiro Urade OH (2010). Crucial role of prostaglandin D2 and adenosine in sleep regulation: experimental evidence from pharmacological approaches to gene-knockout mice. Future Neurology, 5:363-376. [Google Scholar]
  • [120].Tachikawa M, Tsuji K, Yokoyama R, Higuchi T, Ozeki G, Yashiki A, et al. (2012). A Clearance System for Prostaglandin D<sub>2</sub>, a Sleep-Promoting Factor, in Cerebrospinal Fluid: Role of the Blood-Cerebrospinal Barrier Transporters. Journal of Pharmacology and Experimental Therapeutics, 343:608. [DOI] [PubMed] [Google Scholar]
  • [121].Khurshid KA (2018). Comorbid Insomnia and Psychiatric Disorders: An Update. Innovations in clinical neuroscience, 15:28-32. [PMC free article] [PubMed] [Google Scholar]
  • [122].Khan IW, Juyal R, Shikha D, Gupta R (2018). Generalized Anxiety disorder but not depression is associated with insomnia: a population based study. Sleep science (Sao Paulo, Brazil), 11:166-173. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [123].Yun S, Jo S (2021). Understanding insomnia as systemic disease. Yeungnam University journal of medicine, 38:267-274. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [124].Reddy MS, Chakrabarty A (2011). "Comorbid" insomnia. Indian journal of psychological medicine, 33:1-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [125].Budhiraja R, Roth T, Hudgel DW, Budhiraja P, Drake CL (2011). Prevalence and polysomnographic correlates of insomnia comorbid with medical disorders. Sleep, 34:859-867. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [126].from: EaHCoCSLaSDA. 2006. Extent and Health Consequences of Chronic Sleep Loss and Sleep Disorders. In Sleep Disorders and Sleep Deprivation: An Unmet Public Health Problem. AB Colten HR, editor. Institute of Medicine (US) Committee on Sleep Medicine and Research, : National Academies Press (US), Washington (DC). [PubMed] [Google Scholar]
  • [127].Vgontzas AN, Fernandez-Mendoza J, Liao D, Bixler EO (2013). Insomnia with objective short sleep duration: the most biologically severe phenotype of the disorder. Sleep Med Rev, 17:241-254. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [128].Bathgate CJ, Edinger JD, Wyatt JK, Krystal AD (2016). Objective but Not Subjective Short Sleep Duration Associated with Increased Risk for Hypertension in Individuals with Insomnia. Sleep, 39:1037-1045. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [129].Vgontzas AN, Liao D, Bixler EO, Chrousos GP, Vela-Bueno A (2009). Insomnia with objective short sleep duration is associated with a high risk for hypertension. Sleep, 32:491-497. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [130].Vgontzas AN, Fernandez-Mendoza J, Bixler EO, Singareddy R, Shaffer ML, Calhoun SL, et al. (2012). Persistent insomnia: the role of objective short sleep duration and mental health. Sleep, 35:61-68. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [131].Bathgate CJ, Edinger JD, Krystal AD (2017). Insomnia Patients With Objective Short Sleep Duration Have a Blunted Response to Cognitive Behavioral Therapy for Insomnia. Sleep, 40:zsw012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [132].Barragán R, Ortega-Azorín C, Sorlí JV, Asensio EM, Coltell O, St-Onge M-P, et al. (2021). Effect of Physical Activity, Smoking, and Sleep on Telomere Length: A Systematic Review of Observational and Intervention Studies. Journal of clinical medicine, 11:76. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [133].Ohayon MM, Lemoine P (2002). [A connection between insomnia and psychiatric disorders in the French general population]. Encephale, 28:420-428. [PubMed] [Google Scholar]
  • [134].Alfano CA, Reynolds K, Scott N, Dahl RE, Mellman TA (2013). Polysomnographic sleep patterns of non-depressed, non-medicated children with generalized anxiety disorder. J Affect Disord, 147:379-384. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [135].Monti JM, Monti D (2000). Sleep disturbance in generalized anxiety disorder and its treatment. Sleep Med Rev, 4:263-276. [DOI] [PubMed] [Google Scholar]
  • [136].Chang M, Hasan S, Suwangto EG, Widjaja NT (2020). Relationship between anxiety and sleep quality in patients attending Kecamatan Penjaringan Public Health Center, North Jakarta. 2020, 8:6. [Google Scholar]
  • [137].Sivertsen B, Salo P, Mykletun A, Hysing M, Pallesen S, Krokstad S, et al. (2012). The bidirectional association between depression and insomnia: the HUNT study. Psychosom Med, 74:758-765. [DOI] [PubMed] [Google Scholar]
  • [138].Buysse DJ, Angst J, Gamma A, Ajdacic V, Eich D, Rössler W (2008). Prevalence, course, and comorbidity of insomnia and depression in young adults. Sleep, 31:473-480. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [139].Soehner AM, Kaplan KA, Harvey AG (2014). Prevalence and clinical correlates of co-occurring insomnia and hypersomnia symptoms in depression. J Affect Disord, 167:93-97. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [140].Zhao J, Liu H, Wu Z, Wang Y, Cao T, Lyu D, et al. (2021). Clinical features of the patients with major depressive disorder co-occurring insomnia and hypersomnia symptoms: a report of NSSD study. Sleep Med, 81:375-381. [DOI] [PubMed] [Google Scholar]
  • [141].Pigeon WR, Hegel M, Unützer J, Fan MY, Sateia MJ, Lyness JM, et al. (2008). Is insomnia a perpetuating factor for late-life depression in the IMPACT cohort? Sleep, 31:481-488. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [142].Johnson EO, Roth T, Breslau N (2006). The association of insomnia with anxiety disorders and depression: Exploration of the direction of risk. Journal of Psychiatric Research, 40:700-708. [DOI] [PubMed] [Google Scholar]
  • [143].Moderie C, Carrier J, Dang-Vu TT (2021). [Sleep disorders in patients with a neurocognitive disorder]. L'Encephale. [DOI] [PubMed] [Google Scholar]
  • [144].Fernandez-Mendoza J, He F, Puzino K, Amatrudo G, Calhoun S, Liao D, et al. (2021). Insomnia with objective short sleep duration is associated with cognitive impairment: a first look at cardiometabolic contributors to brain health. Sleep, 44:zsaa150. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [145].Fernandez-Mendoza J, He F, Calhoun SL, Vgontzas AN, Liao D, Bixler EO (2020). Objective short sleep duration increases the risk of all-cause mortality associated with possible vascular cognitive impairment. Sleep Health, 6:71-78. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [146].Banks S, DF D.2011. Kryger M, Roth T, Dement WC. Principles and practice of sleep medicine. Philadelphia: Elsevier Saunders. [Google Scholar]
  • [147].Vinik A (2010). The approach to the management of the patient with neuropathic pain. J Clin Endocrinol Metab, 95:4802-4811. [DOI] [PubMed] [Google Scholar]
  • [148].Ferini-Strambi L (2017). Neuropathic Pain and Sleep: A Review. Pain and Therapy, 6:19-23. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [149].Ferini-Strambi L (2017). Neuropathic Pain and Sleep: A Review. Pain and therapy, 6:19-23. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [150].Roth T, van Seventer R, Murphy TK (2010). The effect of pregabalin on pain-related sleep interference in diabetic peripheral neuropathy or postherpetic neuralgia: a review of nine clinical trials. Curr Med Res Opin, 26:2411-2419. [DOI] [PubMed] [Google Scholar]
  • [151].Benoliel R, Eliav E, Sharav Y (2009). Self-reports of pain-related awakenings in persistent orofacial pain patients. J Orofac Pain, 23:330-338. [PubMed] [Google Scholar]
  • [152].Dziadkowiak E, Chojdak-Łukasiewicz J, Olejniczak P, Paradowski B (2021). Regulation of microRNA Expression in Sleep Disorders in Patients with Epilepsy. International journal of molecular sciences, 22:7370. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [153].Vendrame M, Yang B, Jackson S, Auerbach SH (2013). Insomnia and epilepsy: a questionnaire-based study. Journal of clinical sleep medicine : JCSM : official publication of the American Academy of Sleep Medicine, 9:141-146. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [154].Quigg M, Gharai S, Ruland J, Schroeder C, Hodges M, Ingersoll KS, et al. (2016). Insomnia in epilepsy is associated with continuing seizures and worse quality of life. Epilepsy Research, 122:91-96. [DOI] [PubMed] [Google Scholar]
  • [155].Macêdo PJOM, Oliveira PSd, Foldvary-Schaefer N, Gomes MdM (2017). Insomnia in people with epilepsy: A review of insomnia prevalence, risk factors and associations with epilepsy-related factors. Epilepsy Research, 135:158-167. [DOI] [PubMed] [Google Scholar]
  • [156].Planas-Ballvé A, Grau-López L, Jiménez M, Ciurans J, Fumanal A, Becerra JL (2021). Insomnia and poor sleep quality are associated with poor seizure control in patients with epilepsy. Neurología (English Edition). [DOI] [PubMed] [Google Scholar]
  • [157].Adem K, Kassew T, Birhanu A, Abate A (2020). Sleep Quality and Associated Factors among Peoples with Epilepsy Who Have a Follow-Up at Amanuel Mental Specialized Hospital, Addis Ababa, Ethiopia, 2019: An Institutional Based Cross-Sectional Study. Psychiatry journal, 2020:1402712-1402712. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [158].Simie Tsega S, Yazew BG, Mekonnen K (2021). Sleep quality and associated factors among adult patients with epilepsy attending follow-up care at referral hospitals in Amhara region, Ethiopia. PloS one, 16:e0261386-e0261386. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [159].Neves GSL, Noé RA, da Mota Gomes M (2016). Sleep quality and quality of life in patients with epilepsy in a public teaching hospital in Rio de Janeiro, Brazil. Revista Brasileira de Neurologia, 51. [Google Scholar]
  • [160].Kim S-H, Lim J-H (2021). Herbal medicine for post-stroke insomnia: A protocol for systematic review and meta-analysis. Medicine, 100:e26223-e26223. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [161].Li LJ, Yang Y, Guan BY, Chen Q, Wang AX, Wang YJ, et al. (2018). Insomnia is associated with increased mortality in patients with first-ever stroke: a 6-year follow-up in a Chinese cohort study. Stroke Vasc Neurol, 3:197-202. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [162].Tang W-K, Grace Lau C, Mok V, Ungvari GS, Wong K-S (2015). Insomnia and health-related quality of life in stroke. Topics in stroke rehabilitation, 22:201-207. [DOI] [PubMed] [Google Scholar]
  • [163].Baylan S, Griffiths S, Grant N, Broomfield NM, Evans JJ, Gardani M (2020). Incidence and prevalence of post-stroke insomnia: A systematic review and meta-analysis. Sleep Med Rev, 49:101222. [DOI] [PubMed] [Google Scholar]
  • [164].Bamer AM, Johnson KL, Amtmann D, Kraft GH (2008). Prevalence of sleep problems in individuals with multiple sclerosis. Multiple Sclerosis Journal, 14:1127-1130. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [165].Kaminska M, Kimoff RJ, Schwartzman K, Trojan DA (2011). Sleep disorders and fatigue in multiple sclerosis: evidence for association and interaction. J Neurol Sci, 302:7-13. [DOI] [PubMed] [Google Scholar]
  • [166].Akın YA, Polat BSA (2021). Sleep disorders in multiple sclerosis. Gülhane Tip Dergisi, 63:141. [Google Scholar]
  • [167].Sumowski JF, Horng S, Brandstadter R, Krieger S, Leavitt VM, Katz Sand I, et al. (2021). Sleep disturbance and memory dysfunction in early multiple sclerosis. Ann Clin Transl Neurol, 8:1172-1182. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [168].Arnulf I, Nielsen J, Lohmann E, Schiefer J, Wild E, Jennum P, et al. (2008). Rapid eye movement sleep disturbances in Huntington disease. Arch Neurol, 65:482-488. [DOI] [PubMed] [Google Scholar]
  • [169].Voysey Z, Fazal SV, Lazar AS, Barker RA (2021). The sleep and circadian problems of Huntington’s disease: when, why and their importance. Journal of Neurology, 268:2275-2283. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [170].Neutel D, Tchikviladzé M, Charles P, Leu-Semenescu S, Roze E, Durr A, et al. (2015). Nocturnal agitation in Huntington disease is caused by arousal-related abnormal movements rather than by rapid eye movement sleep behavior disorder. Sleep Medicine, 16:754-759. [DOI] [PubMed] [Google Scholar]
  • [171].Goodman AOG, Morton AJ, Barker RA.2010. Identifying sleep disturbances in Huntington's disease using a simple disease-focused questionnaire. In PLoS currents. RRN1189. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [172].Politis M, Pavese N, Tai YF, Tabrizi SJ, Barker RA, Piccini P (2008). Hypothalamic involvement in Huntington's disease: an in vivo PET study. Brain, 131:2860-2869. [DOI] [PubMed] [Google Scholar]
  • [173].Petersén Å, Gabery S (2012). Hypothalamic and limbic system changes in Huntington's disease. Journal of Huntington's disease, 1:5-16. [DOI] [PubMed] [Google Scholar]
  • [174].Pini L, Jacquemot C, Cagnin A, Meneghello F, Semenza C, Mantini D, et al. (2020). Aberrant brain network connectivity in presymptomatic and manifest Huntington's disease: a systematic review. Human Brain Mapping, 41:256-269. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [175].Herzog-Krzywoszanska R, Krzywoszanski L (2019). Sleep Disorders in Huntington’s Disease. Frontiers in Psychiatry, 10. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [176].Maywood ES, Fraenkel E, McAllister CJ, Wood N, Reddy AB, Hastings MH, et al. (2010). Disruption of peripheral circadian timekeeping in a mouse model of Huntington's disease and its restoration by temporally scheduled feeding. Journal of neuroscience, 30:10199-10204. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [177].Robertson ME, McSherry F, Herndon JE, Peters KB (2016). Insomnia and its associations in patients with recurrent glial neoplasms. SpringerPlus, 5:823. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [178].Stanhope BA, Jaggard JB, Gratton M, Brown EB, Keene AC (2020). Sleep Regulates Glial Plasticity and Expression of the Engulfment Receptor Draper Following Neural Injury. Current Biology, 30:1092-1101.e1093. [DOI] [PubMed] [Google Scholar]
  • [179].Lim MM, Gerstner JR, Holtzman DM (2014). The sleep-wake cycle and Alzheimer's disease: what do we know? Neurodegenerative disease management, 4:351-362. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [180].Bellesi M, de Vivo L, Chini M, Gilli F, Tononi G, Cirelli C (2017). Sleep Loss Promotes Astrocytic Phagocytosis and Microglial Activation in Mouse Cerebral Cortex. J Neurosci, 37:5263-5273. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [181].Zhang P, Li Y-X, Zhang Z-Z, Yang Y, Rao J-X, Xia L, et al. (2020). Astroglial Mechanisms Underlying Chronic Insomnia Disorder: A Clinical Study. Nature and science of sleep, 12:693-704. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [182].Garbarino S, Lanteri P, Bragazzi NL, Magnavita N, Scoditti E (2021). Role of sleep deprivation in immune-related disease risk and outcomes. Communications Biology, 4:1304. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [183].Besedovsky L, Lange T, Born J (2012). Sleep and immune function. Pflügers Archiv - European Journal of Physiology, 463:121-137. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [184].Savard J, Laroche L, Simard S, Ivers H, Morin CM (2003). Chronic insomnia and immune functioning. Psychosom Med, 65:211-221. [DOI] [PubMed] [Google Scholar]
  • [185].van Leeuwen WM, Lehto M, Karisola P, Lindholm H, Luukkonen R, Sallinen M, et al. (2009). Sleep restriction increases the risk of developing cardiovascular diseases by augmenting proinflammatory responses through IL-17 and CRP. PloS one, 4:e4589. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [186].Haack M, Sanchez E, Mullington JM (2007). Elevated inflammatory markers in response to prolonged sleep restriction are associated with increased pain experience in healthy volunteers. Sleep, 30:1145-1152. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [187].Sauvet F, Leftheriotis G, Gomez-Merino D, Langrume C, Drogou C, Van Beers P, et al. (2010). Effect of acute sleep deprivation on vascular function in healthy subjects. Journal of applied physiology, 108:68-75. [DOI] [PubMed] [Google Scholar]
  • [188].Irwin MR, Opp MR (2017). Sleep Health: Reciprocal Regulation of Sleep and Innate Immunity. Neuropsychopharmacology : official publication of the American College of Neuropsychopharmacology, 42:129-155. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [189].Irwin MR, Witarama T, Caudill M, Olmstead R, Breen EC (2015). Sleep loss activates cellular inflammation and signal transducer and activator of transcription (STAT) family proteins in humans. Brain, Behavior, and Immunity, 47:86-92. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [190].Shi T, Min M, Sun C, Zhang Y, Liang M, Sun Y (2020). Does insomnia predict a high risk of cancer? A systematic review and meta-analysis of cohort studies. J Sleep Res, 29:e12876. [DOI] [PubMed] [Google Scholar]
  • [191].Fang H-F, Miao N-F, Chen C-D, Sithole T, Chung M-H (2015). Risk of Cancer in Patients with Insomnia, Parasomnia, and Obstructive Sleep Apnea: A Nationwide Nested Case-Control Study. Journal of Cancer, 6:1140-1147. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [192].Shi T, Min M, Sun C, Zhang Y, Liang M, Sun Y (2020). Does insomnia predict a high risk of cancer? A systematic review and meta-analysis of cohort studies. Journal of Sleep Research, 29:e12876. [DOI] [PubMed] [Google Scholar]
  • [193].Garland SN, Irwin MR, Posner D, Perlis ML (2018). Are sleep continuity disturbance and fatigue prodromal symptoms of cancer development? Medical hypotheses, 120:72-75. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [194].Haus EL, Smolensky MH (2013). Shift work and cancer risk: potential mechanistic roles of circadian disruption, light at night, and sleep deprivation. Sleep Med Rev, 17:273-284. [DOI] [PubMed] [Google Scholar]
  • [195].Sauer LA, Blask DE, Dauchy RT (2007). Dietary factors and growth and metabolism in experimental tumors. J Nutr Biochem, 18:637-649. [DOI] [PubMed] [Google Scholar]
  • [196].Zhou R, Chen X, Liang J, Chen Q, Tian H, Yang C, et al. (2021). A circadian rhythm-related gene signature associated with tumor immunity, cisplatin efficacy, and prognosis in bladder cancer. Aging, 13:25153-25179. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [197].Huo Z, Ge F, Li C, Cheng H, Lu Y, Wang R, et al. (2021). Genetically predicted insomnia and lung cancer risk: a Mendelian randomization study. Sleep Medicine, 87:183-190. [DOI] [PubMed] [Google Scholar]
  • [198].Gasparro R, Calabria E, Coppola N, Marenzi G, Sammartino G, Aria M, et al. (2021). Sleep Disorders and Psychological Profile in Oral Cancer Survivors: A Case-Control Clinical Study. Cancers, 13:1855. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [199].Slade AN, Waters MR, Serrano NA (2020). Long-term sleep disturbance and prescription sleep aid use among cancer survivors in the United States. Support Care Cancer, 28:551-560. [DOI] [PubMed] [Google Scholar]
  • [200].Santoso AMM, Jansen F, de Vries R, Leemans CR, van Straten A, Verdonck-de Leeuw IM (2019). Prevalence of sleep disturbances among head and neck cancer patients: A systematic review and meta-analysis. Sleep Med Rev, 47:62-73. [DOI] [PubMed] [Google Scholar]
  • [201].Gooneratne NS, Dean GE, Rogers AE, Nkwuo JE, Coyne JC, Kaiser LR (2007). Sleep and quality of life in long-term lung cancer survivors. Lung cancer (Amsterdam, Netherlands), 58:403-410. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [202].George M, Elias A, Shafiei M (2015). Insomnia in Cancer--Associations and Implications. Asian Pac J Cancer Prev, 16:6711-6714. [DOI] [PubMed] [Google Scholar]
  • [203].Fuggle N, Westbury L, Syddall H, Duggal N, Shaw S, Maslin K, et al. (2018). Relationships between markers of inflammation and bone density: findings from the Hertfordshire Cohort Study. Osteoporosis International, 29:1581-1589. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [204].Seixas AA, Vallon J, Barnes-Grant A, Butler M, Langford AT, Grandner MA, et al. (2018). Mediating effects of body mass index, physical activity, and emotional distress on the relationship between short sleep and cardiovascular disease. Medicine, 97. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [205].Swanson CM, Kohrt WM, Buxton OM, Everson CA, Wright KP Jr, Orwoll ES, et al. (2018). The importance of the circadian system & sleep for bone health. Metabolism, 84:28-43. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [206].Tang Y, Wang S, Yi Q, Xia Y, Geng B (2021). Sleep pattern and bone mineral density: a cross-sectional study of National Health and Nutrition Examination Survey (NHANES) 2017-2018. Arch Osteoporos, 16:157. [DOI] [PubMed] [Google Scholar]
  • [207].Lee C-L, Tzeng H-E, Liu W-J, Tsai C-H (2021). A cross-sectional analysis of the association between sleep duration and osteoporosis risk in adults using 2005-2010 NHANES. Scientific reports, 11:9090-9090. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [208].Dolsen MR, Crosswell AD, Prather AA (2019). Links between stress, sleep, and inflammation: are there sex differences? Current psychiatry reports, 21:1-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [209].Lucassen EA, De Mutsert R, Le Cessie S, Appelman-Dijkstra NM, Rosendaal FR, Van Heemst D, et al. (2017). Poor sleep quality and later sleep timing are risk factors for osteopenia and sarcopenia in middle-aged men and women: The NEO study. PloS one, 12:e0176685. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [210].Blake J, Cosman FA, Lewiecki EM, McClung MR, Pinkerton J, Shapiro M (2021). Management of osteoporosis in postmenopausal women: the 2021 position statement of The North American Menopause Society. MENOPAUSE-THE JOURNAL OF THE NORTH AMERICAN MENOPAUSE SOCIETY, 28:973-997. [DOI] [PubMed] [Google Scholar]
  • [211].Cauley JA, Hovey KM, Stone KL, Andrews CA, Barbour KE, Hale L, et al. (2019). Characteristics of Self-Reported Sleep and the Risk of Falls and Fractures: The Women's Health Initiative (WHI). Journal of bone and mineral research : the official journal of the American Society for Bone and Mineral Research, 34:464-474. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [212].Ochs-Balcom HM, Hovey KM, Andrews C, Cauley JA, Hale L, Li W, et al. (2020). Short Sleep Is Associated With Low Bone Mineral Density and Osteoporosis in the Women's Health Initiative. J Bone Miner Res, 35:261-268. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [213].Shiao Y-C, Chen W-T, Chen W-L (2021). Association of short sleep duration and trabecular bone score. Scientific reports, 11:19821-19821. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [214].Su Y, Leung J, Hans D, Aubry-Rozier B, Kwok T (2017). Added clinical use of trabecular bone score to BMD for major osteoporotic fracture prediction in older Chinese people: the Mr. OS and Ms. OS cohort study in Hong Kong. Osteoporosis international, 28:151-160. [DOI] [PubMed] [Google Scholar]
  • [215].Yi SS, Chung S-H, Kim PS (2018). Sharing Pathological Mechanisms of Insomnia and Osteoporosis, and a New Perspective on Safe Drug Choice. Journal of menopausal medicine, 24:143-149. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [216].Quartana PJ, Finan PH, Page GG, Smith MT (2015). Effects of insomnia disorder and knee osteoarthritis on resting and pain-evoked inflammatory markers. Brain, behavior, and immunity, 47:228-237. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [217].Heffner KL, France CR, Ashrafioun L, Quiñones M, Walsh P, Maloney MD, et al. (2018). Clinical Pain-related Outcomes and Inflammatory Cytokine Response to Pain Following Insomnia Improvement in Adults With Knee Osteoarthritis. Clin J Pain, 34:1133-1140. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [218].Freitas DC, Schlosser TC, dos Santos AA, Neri AL, Ceolim MF (2013). [Association between insomnia and rheumatoid arthritis in elderly]. Rev Esc Enferm USP, 47:869-875. [DOI] [PubMed] [Google Scholar]
  • [219].Taylor-Gjevre RM, Gjevre JA, Nair B, Skomro R, Lim HJ (2011). Components of sleep quality and sleep fragmentation in rheumatoid arthritis and osteoarthritis. Musculoskeletal Care, 9:152-159. [DOI] [PubMed] [Google Scholar]
  • [220].ZHENG S, HOU D (2021). Effects of pain on fatigue in patients with rheumatoid arthritis: multiple mediating effects of negative emotions and insomnia. Chinese Journal of Practical Nursing:103-108. [Google Scholar]
  • [221].Fawzy RM, Abdel-Monem SM, El-Brashi A-WS, Mohamed AA (2022). A comparative study between rheumatoid arthritis and osteoarthritis regarding association of insomnia with disease status. Egyptian Rheumatology and Rehabilitation, 49:4. [Google Scholar]
  • [222].Sofi F, Cesari F, Casini A, Macchi C, Abbate R, Gensini GF (2014). Insomnia and risk of cardiovascular disease: a meta-analysis. Eur J Prev Cardiol, 21:57-64. [DOI] [PubMed] [Google Scholar]
  • [223].Bertisch SM, Pollock BD, Mittleman MA, Buysse DJ, Bazzano LA, Gottlieb DJ, et al. (2018). Insomnia with objective short sleep duration and risk of incident cardiovascular disease and all-cause mortality: Sleep Heart Health Study. Sleep, 41:zsy047. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [224].Yan B, Yang J, Zhao B, Fan Y, Wang W, Ma X (2021). Objective Sleep Efficiency Predicts Cardiovascular Disease in a Community Population: The Sleep Heart Health Study. Journal of the American Heart Association, 10:e016201-e016201. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [225].Lao XQ, Liu X, Deng H-B, Chan T-C, Ho KF, Wang F, et al. (2018). Sleep quality, sleep duration, and the risk of coronary heart disease: a prospective cohort study with 60,586 adults. Journal of Clinical Sleep Medicine, 14:109-117. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [226].Luyster FS, Strollo PJ Jr., Holguin F, Castro M, Dunican EM, Fahy J, et al. (2016). Association Between Insomnia and Asthma Burden in the Severe Asthma Research Program (SARP) III. Chest, 150:1242-1250. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [227].Kavanagh J, Jackson DJ, Kent BD (2018). Sleep and asthma. Curr Opin Pulm Med, 24:569-573. [DOI] [PubMed] [Google Scholar]
  • [228].Kim DJ, Ha T-W, Jung HU, Baek EJ, Lee WJ, Kim HK, et al. (2021). Characterisation of insomnia as an environmental risk factor for asthma via Mendelian randomization and gene environment interaction. Scientific Reports, 11:21813. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [229].Khanijow V, Prakash P, Emsellem HA, Borum ML, Doman DB (2015). Sleep Dysfunction and Gastrointestinal Diseases. Gastroenterology & hepatology, 11:817-825. [PMC free article] [PubMed] [Google Scholar]
  • [230].Plotogea OM, Gheorghe G, Stan-Ilie M, Constantinescu G, Bacalbasa N, Bungau S, et al. (2021). Assessment of Sleep among Patients with Chronic Liver Disease: Association with Quality of Life. J Pers Med, 11. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [231].Jansson C, Nordenstedt H, Wallander MA, Johansson S, Johnsen R, Hveem K, et al. (2009). A population-based study showing an association between gastroesophageal reflux disease and sleep problems. Clin Gastroenterol Hepatol, 7:960-965. [DOI] [PubMed] [Google Scholar]
  • [232].Conley S, Jeon S, Lehner V, Proctor DD, Redeker NS (2021). Sleep Characteristics and Rest-Activity Rhythms Are Associated with Gastrointestinal Symptoms Among Adults with Inflammatory Bowel Disease. Dig Dis Sci, 66:181-189. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [233].Orr WC, Fass R, Sundaram SS, Scheimann AO (2020). The effect of sleep on gastrointestinal functioning in common digestive diseases. Lancet Gastroenterol Hepatol, 5:616-624. [DOI] [PubMed] [Google Scholar]
  • [234].Barutcu Atas D, Aydin Sunbul E, Velioglu A, Tuglular S (2021). The association between perceived stress with sleep quality, insomnia, anxiety and depression in kidney transplant recipients during Covid-19 pandemic. PLoS One, 16:e0248117. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [235].Lu JL, Freire AX, Molnar MZ, Kalantar-Zadeh K, Kovesdy CP (2018). Association of Chronic Insomnia With Mortality and Adverse Renal Outcomes. Mayo Clinic Proceedings, 93:1563-1570. [DOI] [PubMed] [Google Scholar]
  • [236].Lufiyani I, Noorana Zahra A, Yona S (2019). Factors related to insomnia among end-stage renal disease patients on hemodialysis in Jakarta, Indonesia. Enfermería Clínica, 29 Suppl 2. [Google Scholar]
  • [237].Sasaki S, Yoshioka E, Saijo Y, Bannai A, Kita T, Tamakoshi A, et al. (2018). A prospective cohort study of insomnia and chronic kidney disease in Japanese workers. Sleep and Breathing, 22:257-265. [DOI] [PubMed] [Google Scholar]
  • [238].Kumar M, Kainth S, Kumar S, Bhardwaj A, KumarAggarwal H, Maiwall R, et al. (2021). Prevalence of and Factors Associated with Sleep-Wake Abnormalities in Patients with Cirrhosis. J Clin Exp Hepatol, 11:453-465. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [239].Formentin C, Garrido M, Montagnese S (2018). Assessment and Management of Sleep Disturbance in Cirrhosis. Curr Hepatol Rep, 17:52-69. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [240].Bruyneel M, Sersté T (2018). Sleep disturbances in patients with liver cirrhosis: prevalence, impact, and management challenges. Nature and science of sleep, 10:369-375. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [241].Wijarnpreecha K, Thongprayoon C, Panjawatanan P, Ungprasert P (2017). Insomnia and risk of nonalcoholic fatty liver disease: A systematic review and meta-analysis. Journal of postgraduate medicine, 63:226-231. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [242].Saponaro C, Gaggini M, Gastaldelli A (2015). Nonalcoholic fatty liver disease and type 2 diabetes: common pathophysiologic mechanisms. Current diabetes reports, 15:1-13. [DOI] [PubMed] [Google Scholar]
  • [243].Wijarnpreecha K, Thongprayoon C, Edmonds P, Cheungpasitporn W (2016). Associations of sugar-and artificially sweetened soda with nonalcoholic fatty liver disease: a systematic review and meta-analysis. QJM: An International Journal of Medicine, 109:461-466. [DOI] [PubMed] [Google Scholar]
  • [244].Jarrar MH, Baranova A, Collantes R, Ranard B, Stepanova M, Bennett C, et al. (2008). Adipokines and cytokines in non-alcoholic fatty liver disease. Aliment Pharmacol Ther, 27:412-421. [DOI] [PubMed] [Google Scholar]
  • [245].Prather AA, Marsland AL, Hall M, Neumann SA, Muldoon MF, Manuck SB (2009). Normative variation in self-reported sleep quality and sleep debt is associated with stimulated pro-inflammatory cytokine production. Biol Psychol, 82:12-17. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [246].Langin D, Arner P (2006). Importance of TNFalpha and neutral lipases in human adipose tissue lipolysis. Trends Endocrinol Metab, 17:314-320. [DOI] [PubMed] [Google Scholar]
  • [247].Bernsmeier C, Weisskopf DM, Pflueger MO, Mosimann J, Campana B, Terracciano L, et al. (2015). Sleep Disruption and Daytime Sleepiness Correlating with Disease Severity and Insulin Resistance in Non-Alcoholic Fatty Liver Disease: A Comparison with Healthy Controls. PLoS One, 10:e0143293. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [248].Spiegel K, Leproult R, L'Hermite-Balériaux M, Copinschi G, Penev PD, Van Cauter E (2004). Leptin levels are dependent on sleep duration: relationships with sympathovagal balance, carbohydrate regulation, cortisol, and thyrotropin. J Clin Endocrinol Metab, 89:5762-5771. [DOI] [PubMed] [Google Scholar]
  • [249].Spiegel K, Tasali E, Leproult R, Scherberg N, Van Cauter E (2011). Twenty-four-hour profiles of acylated and total ghrelin: relationship with glucose levels and impact of time of day and sleep. J Clin Endocrinol Metab, 96:486-493. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [250].Zhao J, Liu M, Ungvari GS, Ng CH, Chow IHI, Wang T, et al. (2021). Prevalence and clinical correlates of insomnia symptoms and their association with quality of life in Chinese patients with HBV-related liver disease. PeerJ, 9:e10956. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [251].Ding C, Lim LL, Xu L, Kong APS (2018). Sleep and Obesity. Journal of obesity & metabolic syndrome, 27:4-24. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [252].Briançon-Marjollet A, Weiszenstein M, Henri M, Thomas A, Godin-Ribuot D, Polak J (2015). The impact of sleep disorders on glucose metabolism: endocrine and molecular mechanisms. Diabetology & metabolic syndrome, 7:25-25. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [253].Cai G-H, Theorell-Haglöw J, Janson C, Svartengren M, Elmståhl S, Lind L, et al. (2018). Insomnia symptoms and sleep duration and their combined effects in relation to associations with obesity and central obesity. Sleep Medicine, 46:81-87. [DOI] [PubMed] [Google Scholar]
  • [254].Vgontzas AN, Liao D, Pejovic S, Calhoun S, Karataraki M, Bixler EO (2009). Insomnia with objective short sleep duration is associated with type 2 diabetes: A population-based study. Diabetes Care, 32:1980-1985. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [255].Lucassen EA, Rother KI, Cizza G (2012). Interacting epidemics? Sleep curtailment, insulin resistance, and obesity. Annals of the New York Academy of Sciences, 1264:110-134. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [256].Grandner MA, Seixas A, Shetty S, Shenoy S (2016). Sleep Duration and Diabetes Risk: Population Trends and Potential Mechanisms. Current diabetes reports, 16:106-106. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [257].Copinschi G, Leproult R, Spiegel K (2014). The important role of sleep in metabolism. Front Horm Res, 42:59-72. [DOI] [PubMed] [Google Scholar]
  • [258].Reutrakul S, Van Cauter E (2018). Sleep influences on obesity, insulin resistance, and risk of type 2 diabetes. Metabolism, 84:56-66. [DOI] [PubMed] [Google Scholar]
  • [259].Antza C, Kostopoulos G, Mostafa S, Nirantharakumar K, Tahrani A (2021). The links between sleep duration, obesity and type 2 diabetes mellitus. The Journal of endocrinology, 252:125-141. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [260].Shoji S, Shoji Y (2009). [Insomnia in diabetes]. Nihon rinsho. Japanese journal of clinical medicine, 67:1525-1531. [PubMed] [Google Scholar]
  • [261].Green MJ, Espie CA, Popham F, Robertson T, Benzeval M (2017). Insomnia symptoms as a cause of type 2 diabetes Incidence: a 20 year cohort study. BMC Psychiatry, 17:94. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [262].Hofmann SG, Asnaani A, Vonk IJJ, Sawyer AT, Fang A (2012). The Efficacy of Cognitive Behavioral Therapy: A Review of Meta-analyses. Cognitive therapy and research, 36:427-440. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [263].Hofmann SG.2011. An introduction to modern CBT: Psychological solutions to mental health problems: John Wiley & Sons. [Google Scholar]
  • [264].Butler AC, Chapman JE, Forman EM, Beck AT (2006). The empirical status of cognitive-behavioral therapy: a review of meta-analyses. Clin Psychol Rev, 26:17-31. [DOI] [PubMed] [Google Scholar]
  • [265].Moberg LT, Solvang B, Sæle RG, Myrvang AD (2021). Effects of cognitive-behavioral and psychodynamic-interpersonal treatments for eating disorders: a meta-analytic inquiry into the role of patient characteristics and change in eating disorder-specific and general psychopathology in remission. J Eat Disord, 9:74. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [266].Morin CM (2004). Cognitive-behavioral approaches to the treatment of insomnia. J Clin Psychiatry, 65:33-40. [PubMed] [Google Scholar]
  • [267].Koffel EA, Koffel JB, Gehrman PR (2015). A meta-analysis of group cognitive behavioral therapy for insomnia. Sleep medicine reviews, 19:6-16. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [268].van der Zweerde T, Bisdounis L, Kyle SD, Lancee J, van Straten A (2019). Cognitive behavioral therapy for insomnia: A meta-analysis of long-term effects in controlled studies. Sleep Medicine Reviews, 48:101208. [DOI] [PubMed] [Google Scholar]
  • [269].Dombrovski AY, Cyranowski JM, Mulsant BH, Houck PR, Buysse DJ, Andreescu C, et al. (2008). Which symptoms predict recurrence of depression in women treated with maintenance interpersonal psychotherapy? Depress Anxiety, 25:1060-1066. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [270].Asarnow LD, Manber R (2019). Cognitive Behavioral Therapy for Insomnia in Depression. Sleep medicine clinics, 14:177-184. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [271].Taylor DJ, Walters HM, Vittengl JR, Krebaum S, Jarrett RB (2010). Which depressive symptoms remain after response to cognitive therapy of depression and predict relapse and recurrence? J Affect Disord, 123:181-187. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [272].McCurry SM, Zhu W, Von Korff M, Wellman R, Morin CM, Thakral M, et al. (2021). Effect of Telephone Cognitive Behavioral Therapy for Insomnia in Older Adults With Osteoarthritis Pain: A Randomized Clinical Trial. JAMA Intern Med, 181:530-538. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [273].Zachariae R, Amidi A, Damholdt MF, Clausen CDR, Dahlgaard J, Lord H, et al. (2018). Internet-Delivered Cognitive-Behavioral Therapy for Insomnia in Breast Cancer Survivors: A Randomized Controlled Trial. JNCI: Journal of the National Cancer Institute, 110:880-887. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [274].Garland SN, Xie SX, DuHamel K, Bao T, Li Q, Barg FK, et al. (2019). Acupuncture Versus Cognitive Behavioral Therapy for Insomnia in Cancer Survivors: A Randomized Clinical Trial. Journal of the National Cancer Institute, 111:1323-1331. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [275].Mitchell MD, Gehrman P, Perlis M, Umscheid CA (2012). Comparative effectiveness of cognitive behavioral therapy for insomnia: a systematic review. BMC Family Practice, 13:40. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [276].Lopresti AL (2017). Cognitive behaviour therapy and inflammation: A systematic review of its relationship and the potential implications for the treatment of depression. Aust N Z J Psychiatry, 51:565-582. [DOI] [PubMed] [Google Scholar]
  • [277].Maes M, Galecki P, Chang YS, Berk M (2011). A review on the oxidative and nitrosative stress (O&NS) pathways in major depression and their possible contribution to the (neuro)degenerative processes in that illness. Prog Neuropsychopharmacol Biol Psychiatry, 35:676-692. [DOI] [PubMed] [Google Scholar]
  • [278].Moylan S, Maes M, Wray NR, Berk M (2013). The neuroprogressive nature of major depressive disorder: pathways to disease evolution and resistance, and therapeutic implications. Mol Psychiatry, 18:595-606. [DOI] [PubMed] [Google Scholar]
  • [279].Gazal M, Souza LD, Fucolo BA, Wiener CD, Silva RA, Pinheiro RT, et al. (2013). The impact of cognitive behavioral therapy on IL-6 levels in unmedicated women experiencing the first episode of depression: a pilot study. Psychiatry Res, 209:742-745. [DOI] [PubMed] [Google Scholar]
  • [280].Moreira FP, Cardoso Tde A, Mondin TC, Souza LD, Silva R, Jansen K, et al. (2015). The effect of proinflammatory cytokines in Cognitive Behavioral Therapy. J Neuroimmunol, 285:143-146. [DOI] [PubMed] [Google Scholar]
  • [281].Dahl J, Ormstad H, Aass HC, Sandvik L, Malt UF, Andreassen OA (2016). Recovery from major depressive disorder episode after non-pharmacological treatment is associated with normalized cytokine levels. Acta Psychiatr Scand, 134:40-47. [DOI] [PubMed] [Google Scholar]
  • [282].Kéri S, Szabó C, Kelemen O (2014). Expression of Toll-Like Receptors in peripheral blood mononuclear cells and response to cognitive-behavioral therapy in major depressive disorder. Brain Behav Immun, 40:235-243. [DOI] [PubMed] [Google Scholar]
  • [283].Månsson KNT, Lueken U, Frick A (2021). Enriching CBT by Neuroscience: Novel Avenues to Achieve Personalized Treatments. International Journal of Cognitive Therapy, 14:182-195. [Google Scholar]
  • [284].Lie JD, Tu KN, Shen DD, Wong BM (2015). Pharmacological Treatment of Insomnia. P & T : a peer-reviewed journal for formulary management, 40:759-771. [PMC free article] [PubMed] [Google Scholar]
  • [285].Madari S, Golebiowski R, Mansukhani MP, Kolla BP (2021). Pharmacological Management of Insomnia. Neurotherapeutics : the journal of the American Society for Experimental NeuroTherapeutics, 18:44-52. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [286].Haynes J, Talbert M, Fox S, Close E (2018). Cognitive Behavioral Therapy in the Treatment of Insomnia. South Med J, 111:75-80. [DOI] [PubMed] [Google Scholar]
  • [287].Mitchell LJ, Bisdounis L, Ballesio A, Omlin X, Kyle SD (2019). The impact of cognitive behavioural therapy for insomnia on objective sleep parameters: A meta-analysis and systematic review. Sleep Med Rev, 47:90-102. [DOI] [PubMed] [Google Scholar]
  • [288].Schutte-Rodin S, Broch L, Buysse D, Dorsey C, Sateia M (2008). Clinical guideline for the evaluation and management of chronic insomnia in adults. J Clin Sleep Med, 4:487-504. [PMC free article] [PubMed] [Google Scholar]
  • [289].Schutte-Rodin S, Broch L, Buysse D, Dorsey C, Sateia M (2008). Clinical guideline for the evaluation and management of chronic insomnia in adults. Journal of clinical sleep medicine : JCSM : official publication of the American Academy of Sleep Medicine, 4:487-504. [PMC free article] [PubMed] [Google Scholar]
  • [290].Liang L, Huang Y, Xu R, Wei Y, Xiao L, Wang G (2019). Eszopiclone for the treatment of primary insomnia: a systematic review and meta-analysis of double-blind, randomized, placebo-controlled trials. Sleep Med, 62:6-13. [DOI] [PubMed] [Google Scholar]
  • [291].Clark JW, Brian ML, Drummond SPA, Hoyer D, Jacobson LH (2020). Effects of orexin receptor antagonism on human sleep architecture: A systematic review. Sleep Med Rev, 53:101332. [DOI] [PubMed] [Google Scholar]
  • [292].Goforth HW (2009). Low-dose doxepin for the treatment of insomnia: emerging data. Expert Opin Pharmacother, 10:1649-1655. [DOI] [PubMed] [Google Scholar]
  • [293].Stahl SM (2009). Mechanism of action of trazodone: a multifunctional drug. CNS Spectr, 14:536-546. [DOI] [PubMed] [Google Scholar]
  • [294].Pétein C, Spinewine A, Henrard S (2021). Trends in benzodiazepine receptor agonists use and associated factors in the Belgian general older population: analysis of the Belgian health interview survey data. Ther Adv Psychopharmacol, 11:20451253211011874. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [295].Lee JY, Farrell B, Holbrook AM (2019). Deprescribing benzodiazepine receptor agonists taken for insomnia: a review and key messages from practice guidelines. Pol Arch Intern Med, 129:43-49. [DOI] [PubMed] [Google Scholar]
  • [296].Pillai V, Roth T, Roehrs T, Moss K, Peterson EL, Drake CL (2016). Effectiveness of Benzodiazepine Receptor Agonists in the Treatment of Insomnia: An Examination of Response and Remission Rates. Sleep, 40. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [297].Dubey AK, Handu SS, Mediratta PK (2015). Suvorexant: The first orexin receptor antagonist to treat insomnia. Journal of pharmacology & pharmacotherapeutics, 6:118-121. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [298].Michelson D, Snyder E, Paradis E, Chengan-Liu M, Snavely DB, Hutzelmann J, et al. (2014). Safety and efficacy of suvorexant during 1-year treatment of insomnia with subsequent abrupt treatment discontinuation: a phase 3 randomised, double-blind, placebo-controlled trial. Lancet Neurol, 13:461-471. [DOI] [PubMed] [Google Scholar]
  • [299].Callander GE, Olorunda M, Monna D, Schuepbach E, Langenegger D, Betschart C, et al. (2013). Kinetic properties of "dual" orexin receptor antagonists at OX1R and OX2R orexin receptors. Front Neurosci, 7:230. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [300].Toi N, Inaba M, Kurajoh M, Morioka T, Hayashi N, Hirota T, et al. (2018). Improvement of glycemic control by treatment for insomnia with suvorexant in type 2 diabetes mellitus. Journal of clinical & translational endocrinology, 15:37-44. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [301].Norman JL, Anderson SL (2016). Novel class of medications, orexin receptor antagonists, in the treatment of insomnia - critical appraisal of suvorexant. Nat Sci Sleep, 8:239-247. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [302].Tsai W-H, Yang CC, Li PC, Chen WC, Chien CT (2013). Therapeutic Potential of Traditional Chinese Medicine on Inflammatory Diseases. Journal of Traditional and Complementary Medicine, 3:142-151. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [303].Ni X, Shergis JL, Guo X, Zhang AL, Li Y, Lu C, et al. (2015). Updated clinical evidence of Chinese herbal medicine for insomnia: a systematic review and meta-analysis of randomized controlled trials. Sleep Medicine, 16:1462-1481. [DOI] [PubMed] [Google Scholar]
  • [304].Ni X, Shergis JL, Zhang AL, Guo X, Lu C, Li Y, et al. (2018). Traditional Use of Chinese Herbal Medicine for Insomnia and Priorities Setting of Future Clinical Research. The Journal of Alternative and Complementary Medicine, 25:8-15. [DOI] [PubMed] [Google Scholar]
  • [305].Singh A, Zhao K (2017). Treatment of Insomnia With Traditional Chinese Herbal Medicine. Int Rev Neurobiol, 135:97-115. [DOI] [PubMed] [Google Scholar]
  • [306].Dong YJ, Jiang NH, Zhan LH, Teng X, Fang X, Lin MQ, et al. (2021). Soporific effect of modified Suanzaoren Decoction on mice models of insomnia by regulating Orexin-A and HPA axis homeostasis. Biomed Pharmacother, 143:112141. [DOI] [PubMed] [Google Scholar]
  • [307].Chiao Y-W, Livneh H, Guo H-R, Chen W-J, Lu M-C, Lin M-C, et al. (2021). Use of Chinese Herbal Medicines Is Related to a Reduction in Depression Risk Among Patients With Insomnia: A Matched Cohort Study. Frontiers in Neurology, 11. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [308].Baglioni C, Battagliese G, Feige B, Spiegelhalder K, Nissen C, Voderholzer U, et al. (2011). Insomnia as a predictor of depression: a meta-analytic evaluation of longitudinal epidemiological studies. Journal of affective disorders, 135:10-19. [DOI] [PubMed] [Google Scholar]
  • [309].Cuijpers P, Vogelzangs N, Twisk J, Kleiboer A, Li J, Penninx BW (2014). Comprehensive meta-analysis of excess mortality in depression in the general community versus patients with specific illnesses. American journal of psychiatry, 171:453-462. [DOI] [PubMed] [Google Scholar]
  • [310].Li C, Huang J, Cheng Y-C, Zhang Y-W (2020). Traditional Chinese Medicine in Depression Treatment: From Molecules to Systems. Frontiers in Pharmacology, 11. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [311].Xie C-l, Gu Y, Wang W-W, Lu L, Fu D-l, Liu A-j, et al. (2013). Efficacy and safety of Suanzaoren decoction for primary insomnia: a systematic review of randomized controlled trials. BMC complementary and alternative medicine, 13:18-18. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [312].Wang X, Ju J, Li J, Fan Y, Xu H (2020). Chaihu Longgu Muli decoction, a Chinese herbal formula, for the treatment of insomnia: A systematic review and meta-analysis. Medicine, 99:e22462-e22462. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [313].Lee SH, Lim SM (2016). Acupuncture for insomnia after stroke: a systematic review and meta-analysis. BMC Complement Altern Med, 16:228. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [314].Zhao K (2013). Acupuncture for the treatment of insomnia. Int Rev Neurobiol, 111:217-234. [DOI] [PubMed] [Google Scholar]
  • [315].Zhang M, Zhao J, Li X, Chen X, Xie J, Meng L, et al. (2019). Effectiveness and safety of acupuncture for insomnia: Protocol for a systematic review. Medicine (Baltimore), 98:e17842. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [316].Zhang L, Tang Y, Hui R, Zheng H, Deng Y, Shi Y, et al. (2020). The effects of active acupuncture and placebo acupuncture on insomnia patients: a randomized controlled trial. Psychol Health Med, 25:1201-1215. [DOI] [PubMed] [Google Scholar]
  • [317].Li S, Zhong W, Peng W, Jiang G (2018). Effectiveness of acupuncture in postpartum depression: a systematic review and meta-analysis. Acupunct Med, 36:295-301. [DOI] [PubMed] [Google Scholar]
  • [318].Yeung W-F, Yu BY-M, Yuen JW-M, Ho JYS, Chung K-F, Zhang Z-J, et al. (2021). Semi-Individualized Acupuncture for Insomnia Disorder and Oxidative Stress: A Randomized, Double-Blind, Sham-Controlled Trial. Nature and science of sleep, 13:1195-1207. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [319].Chung KF, Yeung WF, Yu YM, Yung KP, Zhang SP, Zhang ZJ, et al. (2015). Acupuncture for residual insomnia associated with major depressive disorder: a placebo- and sham-controlled, subject- and assessor-blind, randomized trial. J Clin Psychiatry, 76:e752-760. [DOI] [PubMed] [Google Scholar]
  • [320].Lee M, Shin BC, Suen L, Park TY, Ernst E (2008). Auricular acupuncture for insomnia: a systematic review. International Journal of Clinical Practice, 62:1744-1752. [DOI] [PubMed] [Google Scholar]
  • [321].Xu H, Shi Y, Xiao Y, Liu P, Wu S, Pang P, et al. (2019). Efficacy Comparison of Different Acupuncture Treatments for Primary Insomnia: A Bayesian Analysis. Evidence-Based Complementary and Alternative Medicine, 2019:8961748. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [322].Han Q-Q, Fu Y, Le J-M, Ma Y-J, Wei X-D, Ji H-L, et al. (2021). The Therapeutic Effects of Acupuncture and Electroacupuncture on Cancer-related Symptoms and Side-Effects. Journal of Cancer, 12:7003-7009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [323].Morey SS (1998). NIH issues consensus statement on acupuncture. Am Fam Physician, 57:2545-2546. [PubMed] [Google Scholar]
  • [324].Li XM (2009). Complementary and alternative medicine in pediatric allergic disorders. Curr Opin Allergy Clin Immunol, 9:161-167. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [325].Zhou YL, Gao XY, Wang PY, Ren S (2012). [Effect of acupuncture at different acupoints on expression of hypothalamic GABA and GABA(A) receptor proteins in insomnia rats]. Zhen Ci Yan Jiu, 37:302-307. [PubMed] [Google Scholar]
  • [326].Park HJ, Park HJ, Chae Y, Kim JW, Lee H, Chung JH (2011). Effect of acupuncture on hypothalamic-pituitary-adrenal system in maternal separation rats. Cell Mol Neurobiol, 31:1123-1127. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [327].You W, Shi YJ, Han YJ, Jia BH, Tu Y (2010). [Effect of electroacupuncture of "Baihui" (GV 20)-"Yintang" (EX-HN 3) on the expression of glucocorticoid and glucocorticoid receptor mRNA of the chronic stress model rats]. Zhen Ci Yan Jiu, 35:261-266. [PubMed] [Google Scholar]
  • [328].Dong JZ, Wei YT, Xu HY, Zhang Y, Yong RL, Xue YN, et al. (2018). [Electroacupuncture of "Zusanli" (ST 36) Raises Muscular Force by Adjusting AMPK/PGC-1 α Signaling in Rats with Chronic Fatigue Syndrome]. Zhen Ci Yan Jiu, 43:335-340. [DOI] [PubMed] [Google Scholar]
  • [329].Spence DW, Kayumov L, Chen A, Lowe A, Jain U, Katzman MA, et al. (2004). Acupuncture increases nocturnal melatonin secretion and reduces insomnia and anxiety: a preliminary report. J Neuropsychiatry Clin Neurosci, 16:19-28. [DOI] [PubMed] [Google Scholar]
  • [330].Li S, Wang Z, Wu H, Yue H, Yin P, Zhang W, et al. (2020). Electroacupuncture versus Sham Acupuncture for Perimenopausal Insomnia: A Randomized Controlled Clinical Trial. Nature and science of sleep, 12:1201-1213. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [331].Li J, Peng C, Lai D, He K, Wang Y, Zhang G, et al. (2019). Changes in cerebral glucose metabolism after acupuncture at KI3 in spontaneously hypertensive rats: a positron emission tomography study. Acupunct Med, 37:107-115. [DOI] [PubMed] [Google Scholar]
  • [332].Du SQ, Wang XR, Zhu W, Ye Y, Yang JW, Ma SM, et al. (2018). Acupuncture inhibits TXNIP-associated oxidative stress and inflammation to attenuate cognitive impairment in vascular dementia rats. CNS Neurosci Ther, 24:39-46. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [333].Pelka RB, Jaenicke C, Gruenwald J (2001). Impulse magnetic-field therapy for insomnia: a double-blind, placebo-controlled study. Adv Ther, 18:174-180. [DOI] [PubMed] [Google Scholar]
  • [334].Pasche B, Barbault A (2004). Low-energy emission therapy: current status and future directions. Bioelectromagnetic Medicine:324-330. [Google Scholar]
  • [335].Ohayon MM, Stolc V, Freund FT, Milesi C, Sullivan SS (2019). The potential for impact of man-made super low and extremely low frequency electromagnetic fields on sleep. Sleep medicine reviews, 47:28-38. [DOI] [PubMed] [Google Scholar]
  • [336].Pelka RB, Jaenicke C, Gruenwald J (2001). Impulse magnetic-field therapy for migraine and other headaches: a double-blind, placebo-controlled study. Adv Ther, 18:101-109. [DOI] [PubMed] [Google Scholar]
  • [337].Oroz R, Kung S, Croarkin PE, Cheung J (2021). Transcranial magnetic stimulation therapeutic applications on sleep and insomnia: a review. Sleep Science and Practice, 5:3. [Google Scholar]
  • [338].Valero-Cabré A, Amengual JL, Stengel C, Pascual-Leone A, Coubard OA (2017). Transcranial magnetic stimulation in basic and clinical neuroscience: a comprehensive review of fundamental principles and novel insights. Neuroscience & Biobehavioral Reviews, 83:381-404. [DOI] [PubMed] [Google Scholar]
  • [339].Pavlova MK, Latreille V, Puri N, Johnsen J, Batool-Anwar S, Javaheri S, et al. (2019). Novel non-pharmacological insomnia treatment - a pilot study. Nature and science of sleep, 11:189-195. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [340].Zheng W, Yu H, Ding W, Guo L, Xu G, Yin N (2019). Changes in Brain Functional Networks of Insomniacs Induced by Magnetic Stimulation at Acupoints. IEEE Transactions on Applied Superconductivity, 29:1-4. [Google Scholar]
  • [341].Haley H, Gorman K, Tademaru P, Stephenson G Anxiety, Depression, or Insomnia in College Students: An Evaluation of Cranial Electrotherapy Stimulation (CES) and Mindfulness. [Google Scholar]
  • [342].Rose KM, Taylor AG, Bourguignon C (2009). Effects of cranial electrical stimulation on sleep disturbances, depressive symptoms, and caregiving appraisal in spousal caregivers of persons with Alzheimer's disease. Appl Nurs Res, 22:119-125. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [343].Kavirajan HC, Lueck K, Chuang K (2014). Alternating current cranial electrotherapy stimulation (CES) for depression. Cochrane Database Syst Rev:Cd010521. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [344].Kirsch DL, Nichols F (2013). Cranial electrotherapy stimulation for treatment of anxiety, depression, and insomnia. Psychiatr Clin North Am, 36:169-176. [DOI] [PubMed] [Google Scholar]
  • [345].Lande RG, Gragnani CT (2018). Prospective study of brain wave changes associated with cranial electrotherapy stimulation. The primary care companion for CNS disorders, 20:26714. [DOI] [PubMed] [Google Scholar]
  • [346].Brunyé TT, Patterson JE, Wooten T, Hussey EK (2021). A Critical Review of Cranial Electrotherapy Stimulation for Neuromodulation in Clinical and Non-clinical Samples. Frontiers in Human Neuroscience, 15. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [347].Yennurajalingam S, Kang D-H, Hwu W-J, Padhye NS, Masino C, Dibaj SS, et al. (2018). Cranial Electrotherapy Stimulation for the Management of Depression, Anxiety, Sleep Disturbance, and Pain in Patients With Advanced Cancer: A Preliminary Study. Journal of Pain and Symptom Management, 55:198-206. [DOI] [PubMed] [Google Scholar]
  • [348].Kwon D, Park J, Lee Y, Kwak J, Do J, Kim JE (2019). Synergic Effects of Cranial Electrotherapy Stimulation with Sleep Hygiene in Patients with Chronic Insomnia. Journal of Sleep Medicine, 16:36-40. [Google Scholar]
  • [349].Um YH, Wang S-M, Kang DW, Kim N-Y, Lim HK (2021). Impact of transdermal trigeminal electrical neuromodulation on subjective and objective sleep parameters in patients with insomnia: a pilot study. Sleep and Breathing. [DOI] [PubMed] [Google Scholar]
  • [350].Rymaszewska J, Ramsey D (2008). Whole body cryotherapy as a novel adjuvant therapy for depression and anxiety. Archives of Psychiatry & Psychotherapy, 10. [Google Scholar]
  • [351].Brosseau L, Yonge KA, Robinson V, Marchand S, Judd M, Wells G, et al. (2003). Thermotherapy for treatment of osteoarthritis. Cochrane Database Syst Rev, 2003:Cd004522. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [352].Doets JJ, Topper M, Nugter AM (2021). A systematic review and meta-analysis of the effect of whole body cryotherapy on mental health problems. Complementary therapies in medicine, 63:102783. [DOI] [PubMed] [Google Scholar]
  • [353].He Y, Fekete G (2021). The Effect of Cryotherapy on Balance Recovery at Different Moments after Lower Extremity Muscle Fatigue. Physical Activity and Health, 5. [Google Scholar]
  • [354].Moosazadeh M, Nesami MB, Goudarzian AH (2020). Effect of cryotherapy on arteriovenous fistula puncture-related pain in hemodialysis patients: A systematic review and meta-analysis. Complementary therapies in medicine, 49:102326. [DOI] [PubMed] [Google Scholar]
  • [355].Abdo S, Nashed A, Hasanin M, Yassin R (2020). Effect of interferential current therapy versus cryotherapy on knee pain in osteoporotic postmenopausal women: a single-blind randomized controlled trial. Physiotherapy Quarterly, 28:30-34. [Google Scholar]
  • [356].Rymaszewska J, Ramsey D, Chładzińska-Kiejna S (2008). Whole-body cryotherapy as adjunct treatment of depressive and anxiety disorders. Archivum Immunologiae et Therapiae Experimentalis, 56:63-68. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [357].Hirvonen HE, Mikkelsson MK, Kautiainen H, Pohjolainen TH, Leirisalo-Repo M (2006). Effectiveness of different cryotherapies on pain and disease activity in active rheumatoid arthritis. A randomised single blinded controlled trial. Clin Exp Rheumatol, 24:295-301. [PubMed] [Google Scholar]
  • [358].Bouzigon R, Grappe F, Ravier G, Dugue B (2016). Whole- and partial-body cryostimulation/cryotherapy: Current technologies and practical applications. J Therm Biol, 61:67-81. [DOI] [PubMed] [Google Scholar]
  • [359].Garcia C, Karri J, Zacharias NA, Abd-Elsayed A (2021). Use of Cryotherapy for Managing Chronic Pain: An Evidence-Based Narrative. Pain and therapy, 10:81-100. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [360].Stanek A, Sieroń-Stołtny K, Romuk E, Cholewka A, Wielkoszyński T, Cieślar G, et al. (2016). Whole-Body Cryostimulation as an Effective Method of Reducing Oxidative Stress in Healthy Men. Adv Clin Exp Med, 25:1281-1291. [DOI] [PubMed] [Google Scholar]
  • [361].Rivera J, Tercero MJ, Salas JS, Gimeno JH, Alejo JS (2018). The effect of cryotherapy on fibromyalgia: a randomised clinical trial carried out in a cryosauna cabin. Rheumatol Int, 38:2243-2250. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [362].Wright B, Kronen PW, Lascelles D, Monteiro B, Murrell JC, Robertson S, et al. (2020). Ice therapy: cool, current and complicated. J Small Anim Pract, 61:267-271. [DOI] [PubMed] [Google Scholar]
  • [363].Klemm P, Becker J, Aykara I, Asendorf T, Dischereit G, Neumann E, et al. (2021). Serial whole-body cryotherapy in fibromyalgia is effective and alters cytokine profiles. Adv Rheumatol, 61:3. [DOI] [PubMed] [Google Scholar]
  • [364].Stanek A, Cholewka A, Gadula J, Drzazga Z, Sieron A, Sieron-Stoltny K (2015). Can Whole-Body Cryotherapy with Subsequent Kinesiotherapy Procedures in Closed Type Cryogenic Chamber Improve BASDAI, BASFI, and Some Spine Mobility Parameters and Decrease Pain Intensity in Patients with Ankylosing Spondylitis? BioMed Research International, 2015:404259. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [365].Stanek A, Cholewka A, Wielkoszyński T, Romuk E, Sieroń A (2018). Whole-Body Cryotherapy Decreases the Levels of Inflammatory, Oxidative Stress, and Atherosclerosis Plaque Markers in Male Patients with Active-Phase Ankylosing Spondylitis in the Absence of Classical Cardiovascular Risk Factors. Mediators of Inflammation, 2018:8592532. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [366].Stanek A, Romuk E, Wielkoszyński T, Bartuś S, Cieślar G, Cholewka A (2019). Decreased Lipid Profile and Oxidative Stress in Healthy Subjects Who Underwent Whole-Body Cryotherapy in Closed Cryochamber with Subsequent Kinesiotherapy. Oxid Med Cell Longev, 2019:7524878. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [367].Stanek A, Wielkoszyński T, Bartuś S, Cholewka A (2020). Whole-Body Cryostimulation Improves Inflammatory Endothelium Parameters and Decreases Oxidative Stress in Healthy Subjects. Antioxidants (Basel, Switzerland), 9:1308. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [368].Senczyszyn A, Wallner R, Szczesniak DM, Łuc M, Rymaszewska J (2021). The Effectiveness of Computerized Cognitive Training Combined With Whole Body Cryotherapy in Improving Cognitive Functions in Older Adults. A Case Control Study. Frontiers in Psychiatry, 12. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [369].Doets JJR, Topper M, Nugter AM (2021). A systematic review and meta-analysis of the effect of whole body cryotherapy on mental health problems. Complementary Therapies in Medicine, 63:102783. [DOI] [PubMed] [Google Scholar]
  • [370].Legrand FD, Bogard F, Beaumont F, Bouchet B, Blancheteau Y, Polidori G (2020). Affective response to whole-body cryotherapy: Influence of sex, body mass index, age, time of day, and past experience. Complementary Therapies in Medicine, 55:102539. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [371].Hammond L, Mitchell K, Cuttell S (2020). Ventilatory responses to whole body cryotherapy. Journal of Thermal Biology, 91:102633. [DOI] [PubMed] [Google Scholar]
  • [372].Douzi W, Dupuy O, Theurot D, Boucard G, Dugué B (2019). Partial-body cryostimulation after training improves sleep quality in professional soccer players. BMC Research Notes, 12:141. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [373].Kasmi S, Filliard J-R, Polidori G, Bouchet B, Blancheteau Y, Legrand FD (2020). Effects of Whole-Body Cryostimulation (-90°C) on Somnolence and Psychological Well-Being in an Older Patient with Restless Legs Syndrome. Applied Psychology: Health and Well-Being, 12:259-267. [DOI] [PubMed] [Google Scholar]
  • [374].SMITH LL (2000). Cytokine hypothesis of overtraining: a physiological adaptation to excessive stress? Medicine & Science in Sports & Exercise, 32:317. [DOI] [PubMed] [Google Scholar]
  • [375].Main LC, Dawson B, Grove JR, Landers GJ, Goodman C (2009). Impact of training on changes in perceived stress and cytokine production. Research in Sports Medicine, 17:112-123. [DOI] [PubMed] [Google Scholar]
  • [376].Ziemann E, Olek RA, Kujach S, Grzywacz T, Antosiewicz J, Garsztka T, et al. (2012). Five-day whole-body cryostimulation, blood inflammatory markers, and performance in high-ranking professional tennis players. Journal of athletic training, 47:664-672. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [377].Douzi W, Dupuy O, Tanneau M, Boucard G, Bouzigon R, Dugué B (2019). 3-min whole body cryotherapy/cryostimulation after training in the evening improves sleep quality in physically active men. European Journal of Sport Science, 19:860-867. [DOI] [PubMed] [Google Scholar]
  • [378].Krzystanek M, Romańczyk M, Surma S, Koźmin-Burzyńska A (2021). Whole Body Cryotherapy and Hyperbaric Oxygen Treatment: New Biological Treatment of Depression? A Systematic Review. Pharmaceuticals (Basel), 14. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [379].Rymaszewska J, Lion KM, Pawlik-Sobecka L, Pawłowski T, Szcześniak D, Trypka E, et al. (2020). Efficacy of the Whole-Body Cryotherapy as Add-on Therapy to Pharmacological Treatment of Depression—A Randomized Controlled Trial. Frontiers in Psychiatry, 11. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [380].Hassan M, Asaad T (2020). Tension-type headache, its relation to stress, and how to relieve it by cryotherapy among academic students. Middle East Current Psychiatry, 27:20. [Google Scholar]
  • [381].Redfern RE, Chen JT, Sibrel S (2018). Effects of Thermomechanical Stimulation during Vaccination on Anxiety, Pain, and Satisfaction in Pediatric Patients: A Randomized Controlled Trial. J Pediatr Nurs, 38:1-7. [DOI] [PubMed] [Google Scholar]
  • [382].Mendes-Neto JM, Santos SL (2020). Vibration associated with cryotherapy to relieve pain in children. BrJP, 3:53-57. [Google Scholar]
  • [383].Silva ALP, Imoto DM, Croci AT (2007). Comparison of cryotherapy, exercise and short waves in knee osteoarthritis treatment. Acta ortopedica brasileira, 15:204-209. [Google Scholar]
  • [384].Adie S, Kwan A, Naylor JM, Harris IA, Mittal R (2012). Cryotherapy following total knee replacement. Cochrane Database Syst Rev:Cd007911. [DOI] [PubMed] [Google Scholar]
  • [385].Barbosa GM, Cunha JE, Cunha TM, Martinho LB, Castro PATS, Oliveira FFB, et al. (2019). Clinical-like cryotherapy improves footprint patterns and reduces synovial inflammation in a rat model of post-traumatic knee osteoarthritis. Scientific Reports, 9:14518. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [386].Dantas LO, Breda CC, da Silva Serrao PRM, Aburquerque-Sendín F, Serafim Jorge AE, Cunha JE, et al. (2019). Short-term cryotherapy did not substantially reduce pain and had unclear effects on physical function and quality of life in people with knee osteoarthritis: a randomised trial. Journal of Physiotherapy, 65:215-221. [DOI] [PubMed] [Google Scholar]
  • [387].Park J, Ty Hopkins J (2012). Immediate effects of acupuncture and cryotherapy on quadriceps motoneuron pool excitability: randomised trial using anterior knee infusion model. Acupunct Med, 30:195-202. [DOI] [PubMed] [Google Scholar]
  • [388].Chruściak T (2016). Subjective evaluation of the effectiveness of whole-body cryotherapy in patients with osteoarthritis. Reumatologia, 54:291-295. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [389].Guillot X, Tordi N, Prati C, Verhoeven F, Pazart L, Wendling D (2017). Cryotherapy decreases synovial Doppler activity and pain in knee arthritis: A randomized-controlled trial. Joint Bone Spine, 84:477-483. [DOI] [PubMed] [Google Scholar]
  • [390].Stanczykiewicz B, Szczesniak D, Rymaszewska J (2015). Whole-body Cryotherapy in Dementia. European Psychiatry, 30:1-1.25169445 [Google Scholar]
  • [391].Hirvonen H, Kautiainen H, Moilanen E, Mikkelsson M, Leirisalo-Repo M (2017). The effect of cryotherapy on total antioxidative capacity in patients with active seropositive rheumatoid arthritis. Rheumatology International, 37:1481-1487. [DOI] [PubMed] [Google Scholar]
  • [392].Wojtecka-Lukasik E, Ksiezopolska-Orlowska K, Gaszewska E, Krasowicz-Towalska O, Rzodkiewicz P, Maslinska D, et al. (2010). Cryotherapy decreases histamine levels in the blood of patients with rheumatoid arthritis. Inflamm Res, 59 Suppl 2:S253-255. [DOI] [PubMed] [Google Scholar]
  • [393].Sieroń A, Cieślar G (2007). Krioterapia-leczenie zimnem. Wydawnictwo Alfa Medica Press, Bielsko-Biała, 15:149. [Google Scholar]
  • [394].Suszko R (2003). Krioterapia ogólnoustrojowa. Rehabilitacja Medyczna, 7:63-71. [Google Scholar]
  • [395].Sieroń A, Pasek J, Mucha R (2007). Krioterapia w rehabilitacji. Rehabil Prakt, 3:34-36. [Google Scholar]
  • [396].Stanek A, Cieślar G, Matyszkiewicz B, Rozmus-Kuczia I, Sieroń-Stołtny K, Koczy B, et al. (2005). Subiektywna ocena skuteczności terapeutycznej krioterapii ogólnoustrojowej u pacjentów z zesztywniającym zapaleniem stawów kręgosłupa. Balneol Pol, 47:24-32. [Google Scholar]
  • [397].Miller E, Mrowicka M, Malinowska K, Mrowicki J, Saluk-Juszczak J, Kędziora J (2010). The effects of whole-body cryotherapy on oxidative stress in multiple sclerosis patients. Journal of Thermal Biology, 35:406-410. [Google Scholar]
  • [398].Miller E, Mrowicka M, Malinowska K, Mrowicki J, Saluk-Juszczak J, Kędziora J (2011). Effects of whole-body cryotherapy on a total antioxidative status and activities of antioxidative enzymes in blood of depressive multiple sclerosis patients. World J Biol Psychiatry, 12:223-227. [DOI] [PubMed] [Google Scholar]
  • [399].Bryczkowska I, Radecka A, Knyszyńska A, Łuczak J, Lubkowska A (2018). Effect of whole body cryotherapy treatments on antioxidant enzyme activity and biochemical parameters in patients with multiple sclerosis. Family Medicine & Primary Care Review, 20:214-217. [Google Scholar]
  • [400].Miller E (2010). [Cryostimulation factor supporting rehabilitation patients with multiple sclerosis and fatigue syndrome]. Wiad Lek, 63:41-45. [PubMed] [Google Scholar]
  • [401].Rymaszewska J, Lion KM, Stańczykiewicz B, Rymaszewska JE, Trypka E, Pawlik-Sobecka L, et al. (2021). The improvement of cognitive deficits after whole-body cryotherapy - A randomised controlled trial. Exp Gerontol, 146:111237. [DOI] [PubMed] [Google Scholar]
  • [402].Gauthier S, Reisberg B, Zaudig M, Petersen RC, Ritchie K, Broich K, et al. (2006). Mild cognitive impairment. Lancet, 367:1262-1270. [DOI] [PubMed] [Google Scholar]
  • [403].Rymaszewska J, Lion KM, Pawlik-Sobecka L, Pawłowski T, Szcześniak D, Trypka E, et al. (2020). Efficacy of the Whole-Body Cryotherapy as Add-on Therapy to Pharmacological Treatment of Depression-A Randomized Controlled Trial. Front Psychiatry, 11:522. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [404].Rymaszewska J, Urbanska K, Szcześniak D, Pawłowski T, Pieniawska-Śmiech K, Kokot I, et al. (2019). Whole-body cryotherapy - promising add-on treatment of depressive disorders. Psychiatr Pol, 53:1053-1067. [DOI] [PubMed] [Google Scholar]
  • [405].Wiecek M, Szygula Z, Gradek J, Kusmierczyk J, Szymura J (2021). Whole-Body Cryotherapy Increases the Activity of Nitric Oxide Synthase in Older Men. Biomolecules, 11. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [406].Sadura-Sieklucka T, Sołtysiuk B, Karlicka A, Sokołowska B, Kontny E, Księżopolska-Orłowska K (2019). Effects of whole body cryotherapy in patients with rheumatoid arthritis considering immune parameters. Reumatologia, 57:320-325. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [407].Księżopolska-Orłowska K, Pacholec A, Jędryka-Góral A, Bugajska J, Sadura-Sieklucka T, Kowalik K, et al. (2016). Complex rehabilitation and the clinical condition of working rheumatoid arthritis patients: does cryotherapy always overtop traditional rehabilitation? Disability and Rehabilitation, 38:1034-1040. [DOI] [PubMed] [Google Scholar]
  • [408].Baust JG, Gage AA, Clarke D, Baust JM, Van Buskirk R (2004). Cryosurgery--a putative approach to molecular-based optimization. Cryobiology, 48:190-204. [DOI] [PubMed] [Google Scholar]
  • [409].Dhaliwal A, Saghir SM, Mashiana HS, Braseth A, Dhindsa BS, Ramai D, et al. (2022). Endoscopic cryotherapy: Indications, techniques, and outcomes involving the gastrointestinal tract. World journal of gastrointestinal endoscopy, 14:17-28. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [410].Bala MM, Riemsma RP, Wolff R, Pedziwiatr M, Mitus JW, Storman D, et al. (2019). Cryotherapy for liver metastases. The Cochrane database of systematic reviews, 7:CD009058-CD009058. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [411].Lai C-C, Chen S-Y, Tu Y-K, Ding Y-W, Lin J-J (2021). Effectiveness of low level laser therapy versus cryotherapy in cancer patients with oral mucositis: Systematic review and network meta-analysis. Critical Reviews in Oncology/Hematology, 160:103276. [DOI] [PubMed] [Google Scholar]
  • [412].Kubrak T, Karakuła M, Czop M, Kawczyk-Krupka A, Aebisher D (2022). Advances in Management of Bladder Cancer—The Role of Photodynamic Therapy. Molecules (Basel, Switzerland), 27. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [413].Liang Z, Fei Y, Lizhi N, Jianying Z, Zhikai Z, Jibing C, et al. (2014). Percutaneous cryotherapy for metastatic bladder cancer: Experience with 23 patients. Cryobiology, 68:79-83. [DOI] [PubMed] [Google Scholar]
  • [414].Santos LF, Correia IJ, Silva AS, Mano JF (2018). Biomaterials for drug delivery patches. European Journal of Pharmaceutical Sciences, 118:49-66. [DOI] [PubMed] [Google Scholar]
  • [415].Dubey V, Mishra D, Asthana A, Jain NK (2006). Transdermal delivery of a pineal hormone: Melatonin via elastic liposomes. Biomaterials, 27:3491-3496. [DOI] [PubMed] [Google Scholar]
  • [416].Qi Z, Cao J, Tao X, Wu X, Kundu SC, Lu S (2021). Silk Fibroin Microneedle Patches for the Treatment of Insomnia. Pharmaceutics, 13:2198. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [417].Yu P, Zhang X, Cheng G, Guo H, Shi X, Zhang X, et al. (2022). Construction of a new multifunctional insomnia drug delivery system. Chemical Engineering Journal, 430:132633. [Google Scholar]
  • [418].Mirza-Aghazadeh-Attari M, Mihanfar A, Yousefi B, Majidinia M (2022). Nanotechnology-based advances in the efficient delivery of melatonin. Cancer Cell International, 22:43. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [419].Cardinali DP, Srinivasan V, Brzezinski A, Brown GM (2012). Melatonin and its analogs in insomnia and depression. J Pineal Res, 52:365-375. [DOI] [PubMed] [Google Scholar]
  • [420].Marilena V, Angeliki S.2018. Melatonin Modified Release Formulations Designed for Sleep Disorders. In Manuela D Cristina, and Crenguţa N Alina,Melatonin editors. Rijeka: IntechOpen. Ch. 9. [Google Scholar]
  • [421].McAlpine CS, Kiss MG, Zuraikat FM, Cheek D, Schiroli G, Amatullah H, et al. (2022). Sleep exerts lasting effects on hematopoietic stem cell function and diversity. Journal of Experimental Medicine, 219. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [422].Ding M, Li P, Wen Y, Zhao Y, Cheng B, Zhang L, et al. (2018). Integrative analysis of genome-wide association study and brain region related enhancer maps identifies biological pathways for insomnia. Progress in Neuro-Psychopharmacology and Biological Psychiatry, 86:180-185. [DOI] [PubMed] [Google Scholar]
  • [423].Rymaszewska J, Urbanska KM, Szczesniak D, Stanczykiewicz B, Trypka E, Zablocka A (2018). The Improvement of Memory Deficits after Whole-Body Cryotherapy - The First Report. Cryo Letters, 39:190-195. [PubMed] [Google Scholar]
  • [424].Vitiello MV, Rybarczyk B, Von Korff M, Stepanski EJ (2009). Cognitive behavioral therapy for insomnia improves sleep and decreases pain in older adults with co-morbid insomnia and osteoarthritis. Journal of Clinical Sleep Medicine, 5:355-362. [PMC free article] [PubMed] [Google Scholar]
  • [425].Thomas Roth P (2009). Comorbid Insomnia: Current Directions and Future Challenges. Supplements and Featured Publications, 15. [PubMed] [Google Scholar]
  • [426].Senczyszyn A, Wallner R, Szczesniak DM, Łuc M, Rymaszewska J (2021). The Effectiveness of Computerized Cognitive Training Combined With Whole Body Cryotherapy in Improving Cognitive Functions in Older Adults. A Case Control Study. Frontiers in psychiatry, 12:649066-649066. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [427].Porramezani N, Imani Goghary Z, Firouzabadi M, Balvardi M, Irannejad-Parizi F (2019). The Effect of Cryotherapy in the Hoku point on the Severity of Pain of Fistula Catheterization in Patients on Hemodialysis. Journal of Advances in Medical and Biomedical Research, 27:37-42. [Google Scholar]
  • [428].Clebak KT, Mendez-Miller M, Croad JR (2020). Cutaneous cryosurgery for common skin conditions. Am Fam physician, 101: 399-406 [PubMed] [Google Scholar]
  • [429].Giménez-Arnau A, Ali Al-Haqan E, Sacrista M, Curto L, Pujol RM (2013). Ultra-freeze induced cold contact wheals during cryosurgery: an uncommon subset of acquired cold contact urticaria. Eur J Dermatol, 23:694-699. [DOI] [PubMed] [Google Scholar]
  • [430].Cordes F, Ellermann C, Ehrchen J, Ullerich H, Eckardt L (2019). Diagnosis and management of cold urticaria in cryoablation of atrial fibrillation: a case report. European Heart Journal - Case Reports, 3:1-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [431].Godse KV (2009). Chronic urticaria and treatment options. Indian journal of dermatology, 54:310-312. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [432].Metz M, Scholz E, Ferrán M, Izquierdo I, Giménez-Arnau A, Maurer M (2010). Rupatadine and its effects on symptom control, stimulation time, and temperature thresholds in patients with acquired cold urticaria. Ann Allergy Asthma Immunol, 104:86-92. [DOI] [PubMed] [Google Scholar]
  • [433].Roach M, Juday T, Tuly R, Chou JW, Jena AB, Doghramji PP (2021). Challenges and opportunities in insomnia disorder. Int J Neurosci, 131:1058-1065. [DOI] [PubMed] [Google Scholar]
  • [434].Rossman J (2019). Cognitive-Behavioral Therapy for Insomnia: An Effective and Underutilized Treatment for Insomnia. Am J Lifestyle Med, 13:544-547. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [435].Barbera J, Shapiro C (2005). Benefit-Risk Assessment of Zaleplon in the Treatment of Insomnia. Drug Safety, 28:301-318. [DOI] [PubMed] [Google Scholar]
  • [436].Morin CM, Benca R (2012). Chronic insomnia. The Lancet, 379:1129-1141. [DOI] [PubMed] [Google Scholar]
  • [437].Tang H, Huang W, Ma J, Liu L (2018). SWOT analysis and revelation in traditional Chinese medicine internationalization. Chinese Medicine, 13:5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [438].Yeung W-F, Chung K-F, Poon MM-K, Ho FY-Y, Zhang S-P, Zhang Z-J, et al. (2012). Prescription of Chinese Herbal Medicine and Selection of Acupoints in Pattern-Based Traditional Chinese Medicine Treatment for Insomnia: A Systematic Review. Evidence-Based Complementary and Alternative Medicine, 2012:902578. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [439].Jing R, Feng K (2021). Efficacy of intradermal acupuncture for insomnia: a meta-analysis. Sleep Medicine, 85:66-74. [DOI] [PubMed] [Google Scholar]
  • [440].Liu F-g, Tan A-h, Peng C-q, Tan Y-x, Yao M-c (2021). Efficacy and Safety of Scalp Acupuncture for Insomnia: A Systematic Review and Meta-Analysis. Evidence-Based Complementary and Alternative Medicine, 2021:6621993. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [441].Azizi E, Ayoobi F, Shamsizadeh A, Moghadam-Ahmadi A, Shafiei SA, Khoshdel A, et al. (2022). Effect of Short-time Exposure of Local Extremely Low-Frequency Magnetic Fields on Sleepiness in Male Rats. Basic Clin Neurosci J, 13:519-530. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [442].Gründer W, Goldammer A, Schober R, Vitzthum H-E (2003). Cryotherapy of the brain--a new methodologic approach. Zeitschrift fur medizinische Physik, 13:203-207. [DOI] [PubMed] [Google Scholar]
  • [443].Collins N (2008). Is ice right? Does cryotherapy improve outcome for acute soft tissue injury? Emergency Medicine Journal, 25:65-68. [DOI] [PubMed] [Google Scholar]

Articles from Aging and Disease are provided here courtesy of JKL International LLC

RESOURCES