Next Article in Journal
Is Revascularization the Treatment of Choice for Traumatized Necrotic Immature Teeth? A Systematic Review and Meta-Analysis
Previous Article in Journal
Neoadjuvant Therapy for Extrahepatic Biliary Tract Cancer: A Propensity Score-Matched Survival Analysis
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

The Modulation of Adipokines, Adipomyokines, and Sleep Disorders on Carcinogenesis

1
Department of Pulmonology and Lung Oncology, Wroclaw Medical University, Grabiszyńska 105, 53-439 Wroclaw, Poland
2
Department of Internal and Occupational Diseases, Hypertension and Clinical Oncology, Wroclaw Medical University, Borowska 213, 50-556 Wroclaw, Poland
3
Department of Thoracic Surgery, Wroclaw Medical University, Ludwika Pasteura 1, Grabiszyńska105, 53-439 Wroclaw, Poland
4
Department of Neurology, Wroclaw Medical University, Borowska 213, 50-556 Wroclaw, Poland
5
Department of Ophthalmology, Wroclaw Medical University, Borowska 213, 50-556 Wroclaw, Poland
6
Department of Biological Sciences, Salem University, 223 West Main Street, Salem, WV 26426, USA
*
Author to whom correspondence should be addressed.
J. Clin. Med. 2023, 12(7), 2655; https://doi.org/10.3390/jcm12072655
Submission received: 19 February 2023 / Revised: 24 March 2023 / Accepted: 31 March 2023 / Published: 2 April 2023
(This article belongs to the Section Oncology)

Abstract

:
Obesity and sarcopenia, i.e., decreased skeletal muscle mass and function, are global health challenges. Moreover, people with obesity and sedentary lifestyles often have sleep disorders. Despite the potential associations, metabolic disturbances linking obesity, sarcopenia, and sleep disorders with cancer are neither well-defined nor understood fully. Abnormal levels of adipokines and adipomyokines originating from both adipose tissue and skeletal muscles are observed in some patients with obesity, sarcopenia and sleep disorders, as well as in cancer patients. This warrants investigation with respect to carcinogenesis. Adipokines and adipomyokines may exert either pro-carcinogenic or anti-carcinogenic effects. These factors, acting independently or together, may significantly modulate the incidence and progression of cancer. This review indicates that one of the possible pathways influencing the development of cancer may be the mutual relationship between obesity and/or sarcopenia, sleep quantity and quality, and adipokines/adipomyokines excretion. Taking into account the high proportion of persons with obesity and sedentary lifestyles, as well as the associations of these conditions with sleep disturbances, more attention should be paid to the individual and combined effects on cancer pathophysiology.

Graphical Abstract

1. Introduction

Adipose tissue acts like an endocrine organ, participating in many physiological processes [1]. Proteins excreted solely by adipocytes or by adipocytes and other cells are called adipokines [2]. Moreover, skeletal muscles are a source of a large family of peptides that exhibit endocrine and paracrine/autocrine effects denominated as myokines [3]. Some of the proteins excreted by adipocytes and myocytes are called adipomyokines [4]. There is a close interaction between myokines and adipokines [5].
Obesity is associated with altered adipokine production because of processes accompanying adipocytes’ hypertrophy and hyperplasia, such as chronic low-grade inflammation, fibrosis, and matrix remodeling [6]. Sarcopenic obesity can be diagnosed in a person with a BMI ≥ 30 kg/m2 and decreased muscle mass and function [7]. Loss of muscle mass and function, i.e., sarcopenia, as well as sarcopenic obesity, change the levels of myokine and adipokines [8]. Sarcopenic obesity may be further differentiated into sarcopenic subcutaneous and sarcopenic visceral obesity [9]. Among cancer patients, sarcopenic obesity occurs in about 25% of obese patients (5.9–39.2%) [10]. As there are important influences of both obesity and sarcopenia on cancer, the concept of sarcopenic obesity has been proposed as a specific pathophysiological entity [11].
Sleep disorders are frequently associated with obesity and sarcopenia. Sleep duration and quality influence muscle strength [12]. In community-dwelling adults (≥65 years old), short sleep duration (<6 h) was associated with both an almost 3-fold increased risk of sarcopenia and an almost 2-fold increased risk of obesity [13]. There is a strong correlation between sleep disorders and obesity, as well as a bidirectional relation between these two factors because obesity, with its consequences, frequently impairs sleep quality [14].
Sarcopenia and/or obesity frequently accompany cancer. Sarcopenia is associated with the diagnosis and poorer prognosis of cancer [15,16]. Obesity is a risk factor for some types of cancer [17]. The prevalence of sarcopenic obesity in cancer patients is high, reaching up to one-fifth–one-third of cancer patients, and is associated with poor survival [18,19]. However, there is a lack of clear evidence of the influence of sarcopenic obesity on cancer occurrence [20].
The role of adipokines and adipomyokines in cancerogenesis is controversial and has not been fully understood. Thus, taking into account that obesity, sarcopenia and sarcopenic obesity are all associated with both sleep disorders and cancer, as well as with disturbed adipokines/myokines concentrations, we aimed to trace the relationship between adipokines and one of the myokine, i.e., irisin, with sleep disturbances and cancer.

2. The Association of Sarcopenia, Obesity, and Sarcopenic Obesity with Cancer

Sarcopenia existing before a cancer diagnosis is an independent risk factor for several cancers, such as lung cancer, colorectal cancer, breast cancer, head and neck cancer, pancreatic cancer, gastric cancer, esophageal cancer, ovarian cancer and hepatocellular cancer [21]. A meta-analysis of the results of 38 studies encompassing 7843 patients with various solid tumors revealed that sarcopenia–as diagnosed on the results of computed tomography studies–was associated with a 44% lower overall survival (HR = 1.44, 95% CI = 1.32–1.56, p < 0.001) and a 95% lower cancer-specific survival (HR = 1.93, 95% CI = 1.38–2.70, p < 0.001) [22]. A systematic review of the studies on sarcopenia diagnosed by bioelectrical impedance analysis indicated adverse clinical outcomes of sarcopenia in cancer patients [23]. Another meta-analysis found that sarcopenia, assessed by computed tomography, was associated with worse overall survival in cancer patients [24]. Among cancer patients receiving immunotherapy, those with sarcopenia alone, as well as those with sarcopenia and biochemical signs of systemic inflammation, obtained a shorter overall survival rate (HR 4.01, 90% CI 1.66–9.68, p = 0.002 and HR 8.46, 90% CI 2.65–27.01, p < 0.01, respectively) and shorter progression-free survival (HR 2.14, 90% CI 1.12–4.10, p = 0.22 and HR 12.29, 90% CI 5.15–29.32, p < 0.001, respectively) [25]. In contrast, another review reported that low muscle mass was not a factor that negatively affected survival rates in incurable cancer; but the studies included in this meta-analysis did not encompass assessments of either muscle strength or physical function [26].
An association between obesity (or simply being overweight) and several cancers has been established, including colorectal, gallbladder, pancreas, kidney, endometrial, breast cancer in postmenopausal women, ovarian, gastric, cardiac, thyroid, esophageal, adenocarcinoma, and multiple myeloma [27]. The strongest associations between being overweight and cancer were found for endometrial cancer, esophageal carcinoma, and kidney cancer [28]. In 2012, 3.6% of newly diagnosed adult cancers could be attributed to being overweight or obese [29]. About 14% of cancer deaths in men and around 20% of cancer deaths in women likewise can be attributed to obesity [30].
Obesity leads to chronic, low-grade inflammation by a disturbed balance between adipokines and cytokines, such as–among others–interleukins (IL-6, IL-8), monocyte chemoattractant protein-1 or tumor necrosis factor-α [31]. Chronic inflammation associated with obesity may promote cancerogenesis through its influence on hormonal balance with further impact on the immune system [32] by inducing tumor cell proliferation and angiogenesis, as well as by damaging genetic material [33].
Metabolic disturbances associated with obesity may strongly influence neoplastic disease [34]. However, at the molecular level, a precise link between increased adipose tissue and cancer has not been fully explained [35].
Considering the variety of possible links between obesity and cancer, the type of obesity should be taken into account [36]. Obesity can be regarded as metabolically healthy, metabolically unhealthy (or abnormal), or sarcopenic [37]. Metabolically abnormal obesity, contrary to metabolically healthy obesity, is associated with the co-occurrence of metabolic syndrome, type 2 diabetes, hypertension, or cardiovascular-cerebrovascular diseases. There is no clear cut-off point between healthy and unhealthy obesity [38]. Metabolically healthy obesity may describe up to 30% of obese people [39].
Sarcopenic obesity is associated with poor outcomes in most cancer patients, regardless of the localization and method of treatment [10]. This has been found by multiple studies and confirmed by meta-analyses [22,40,41]. Recent findings revealed a shortened survival in patients with sarcopenic obesity and head and neck cancer [42,43], gastric cancer [44,45], pancreatic cancer [46,47,48,49], urothelial cancer [50], lymphoma [51], colorectal cancer [52], as well as in women with colorectal cancer [53]. A meta-analysis of 14 studies of cancer patients revealed negative clinical outcomes associated with sarcopenic obesity, such as increased drug toxicity, more frequent surgical complications, and shortened survival [40]. In patients with hepatocellular carcinoma undergoing a hepatectomy, sarcopenic obesity was associated with worse median survival than non-sarcopenic non-obese patients [54]. Visceral adiposity with low muscularity was a risk factor for decreased survival [55].
In contrast, another study reported that sarcopenic obesity was not a risk factor for surgically treated hepatocellular carcinoma [56]. In patients with esophago-gastric cancer who were receiving palliative chemotherapy, sarcopenic obesity was associated with the occurrence of neurotoxicity from the chemotherapeutics but not with the progression of the disease or survival [57]. In patients with non-small-cell lung cancer undergoing chemoradiotherapy, sarcopenic obesity, diagnosed in 14% of patients, did not influence survival [58]. Sarcopenic obesity was not associated with overall survival in breast cancer patients [59] or colorectal metastatic cancer patients [60].
The role of adipokines in promoting genomic instability linking obesity and cancer has not been fully elucidated [61]. Altered microRNA secretion in adipose tissue may be implicated in oncogenesis [62], e.g., the exposure of prostate cancer cells to leptin downregulated the expression of micro-RNA-628 and led to increased cancer cell proliferation [63]. It has been shown that the development of renal cell carcinoma might be associated with obesity-associated alterations in gene expression, such as DNA methylation, single nucleotide polymorphisms, histone modification and microRNAs [64]. In renal cancer cells, the high methylation in leptin receptors predicted an increased risk of cancer progression and shorter recurrence-free survival of renal cancer patients [65]. The leptin receptor gene variant rs1137101 was proposed as a possible risk factor for renal cell carcinoma [66].
Possible associations between sleep disorders, obesity and sarcopenia with cancerogenesis are presented in Figure 1.

3. The Association of Sarcopenia, Obesity and Sarcopenic Obesity with Sleep Disorders

Age and inactivity [67,68] but also sleep disorders may influence progressive loss of muscle mass and function. Multiple sleep problems are associated with sarcopenia: long-sleep duration [69,70,71], disruption of sleep-wake rhythm by shift work [72], poor sleep quality [73], poor sleep quality in older patients with diabetes [74], increased sleep latency [75], later sleep timing [76], insomnia [77], poor sleep efficiency [78], complains of “problems sleeping” and taking sleeping pills [79]. As recently shown in the study encompassing 13,210 adults, long sleep duration (>9 h/day) in persons aged ≥ 65 years was significantly associated with sarcopenia, especially in women (odds ratio 2.19, 95% CI 1.26–3.81) [80].
Short- and long-sleep duration [81] and low sleep efficiency, especially in older men, were associated with obesity [82]. This association was reported by meta-analyses encompassing the result of studies of more than 5 million participants [83]. There is a bidirectional influence between sleep disorders increasing the probability of developing obesity and obesity increasing the chance of sleep disorders [84]. The most common sleep disorder associated with obesity is obstructive sleep apnea (OSA) [85].
Sarcopenic obesity may be associated with some sleep disorders, e.g., with OSA, that further worsens sleep quality [86].

4. The Association of Myokine Irisin with Sleep Disorders and Cancer

Irisin is the best-recognized myokine [87]; it is secreted by myocytes and adipocytes, and is also referred to as an adipomyokine [88]. Its levels depend on muscle mass and physical activity levels [89]. In obese patients, irisin levels are either decreased [8,90] or increased, indicating irisin resistance [91]. Irisin may be negatively influenced by sleep disturbances. Lowered irisin concentrations were associated with poor sleep quality, as shown in patients with rheumatoid arthritis [92]. OSA is often associated with obesity and generally leads to disordered sleep. Decreased irisin concentrations significantly and inversely correlated with OSA [93]. On the other side, however, elevated levels of irisin in OSA patients were associated with increased daytime sleepiness [94].
The role of irisin in carcinogenesis is not fully understood [95]. However, several recent findings indicate that irisin has a potent anticancerous action through different pathways [89]. Experimental studies have revealed that irisin suppressed the cell proliferation of many cancers, including pancreatic cancer cells [96], osteosarcoma cells [97], lung cancer cells [98], and breast cancer cells [99]. Irisin had no effect on the proliferation of cells linked to obesity-related cancers such as endometrial, colon, thyroid, and esophageal [100]. In pancreatic cancer cell lines, irisin activated adenosine monophosphate-activated protein kinase, downregulated the mTOR pathway and inhibited epithelial-to-mesenchymal transition leading to the suppression of the cell growth [96]. It was shown to inhibit endothelial-to-mesenchymal transition, a hallmark of cancer, via the STAT3/Snail signaling pathway in osteosarcoma [97]. In liver cancer cells, however, irisin was found to activate the PI3K/AKT pathway facilitating cancer progression [101].
Increased irisin serum levels correlated with a decreased risk of breast cancer [102] and colorectal cancer [103]. In one of the first clinical studies on the role of irisin in cancer patients, irisin levels in the serum of breast cancer patients were significantly lower than in the serum of healthy women and were associated with the tumor stage [102]. Additionally, irisin was found to play a protective role against spinal metastases in breast cancer patients. In patients with metastases to the spine, the concentrations of irisin in the serum were lower than in the patients without spinal metastases [104]. Decreased irisin serum levels were found in hepatocellular carcinoma patients [105]. A low irisin serum concentration may be regarded as a highly sensitive (80.5%) and specific (90%) biomarker of prostate cancer [106]. Low irisin serum concentrations in bladder cancer appeared to have high sensitivity (74.7%) and specificity (90.7%) as a diagnostic biomarker and predicted higher mortality rates in this type of cancer [107]. On the contrary, in renal cancer patients, irisin levels in the serum were higher than in the healthy controls [108]. Moreover, in patients with benign breast tumors or with breast cancer, irisin concentrations were elevated [109].

5. The Association of Adipokines with Sleep Disorders and Cancer

One adipokine that is negatively associated with obesity is adiponectin, an established anti-carcinogen. Another adipokine negatively associated with obesity is omentin-1, which has anti-inflammatory properties and is not clearly associated with carcinogenesis. Other adipokines that are positively correlated with obesity (leptin, resistin, vaspin, chemerin, nesfatin) do not exhibit an equivocal role in carcinogenesis. Some adipokines that are positively correlated with obesity (visfatin, osteopontin, apelin, retinol-binding protein 4, galectin) exert pro-carcinogenic effects. All these adipokines are influenced by sleep disorders.

5.1. Adiponectin

Adiponectin has an inverse correlation with BMI [110]. Adiponectin serum concentrations are significantly decreased in OSA [111,112,113,114]. Improved quality of sleep with the treatment of OSA resulted in increases in adiponectin serum concentrations [115]. In other situations of sleep loss, decreased levels of adiponectin were found [116], although this observation has not been confirmed in other studies [117,118].
Adiponectin is considered an adipokine with anti-tumor properties [119]. Hypoadiponectinemia is associated with an increased risk of various cancers, as shown by several meta-analyses [120,121,122].

5.2. Omentin-1

There is an inverse correlation between omentin-1 and obesity [123]. Omentin plasma levels were found to be significantly decreased in OSA patients [112] and correlated with sleep structure abnormalities [124].
In vitro studies of omentin-1 indicated both a potentially carcinogenetic role [125] and a protective role against cancer [126]. The meta-analysis revealed that increased levels of omentin-1 were strongly associated with an increased risk of colorectal, pancreas, and breast cancers [127]. The association between increased omentin plasma levels and colorectal cancer was found only in non-obese patients, with no relationship in obese colorectal cancer patients [128]. Increased concentrations of omentin-1 plasma levels were found in prostate cancer patients, and it has been postulated that its levels could serve as a diagnostic biomarker in this type of cancer [129]. A meta-analysis of case-control studies found downregulation of omentin in patients with lung cancer [130] and significantly decreased serum levels of omentin-1 in breast cancer patients [131] or it’s serum levels in renal cancer patients were found [132]. In breast cancer postmenopausal patients, omentin-1 serum levels were inversely associated with tumor markers and cancer stage [133].

5.3. Leptin

Leptin concentrations are positively correlated with obesity [1]. Leptin serum levels are increased in OSA syndrome patients, mostly because of obesity [112,134,135,136], and, in some studies, it correlated with the severity of this syndrome [137]. Moreover, poor sleep quality in overweight and obese subjects without OSA was associated with increased levels of leptin [138]. Short sleep duration led to an increase in leptin serum levels [139].
Leptin favors cancer cell proliferation and invasion, influencing cancer cell differentiation and migration, stimulating angiogenesis, and inhibiting cancer cell apoptosis [140]. The influence of leptin plasma levels on the development of cancer has been established in patients with breast cancer [141] and endometrial cancer [142]. In paraffin blocks taken from patients with colorectal cancer, an increased expression of leptin receptors was found [143]. In patients with glioma, the expression of leptin and leptin receptors in the specimens of resected tumors significantly correlated with the level of malignancy [144]. In patients with meningioma, the expression of leptin receptors in the tumor positively correlated with BMI [145]. In patients with ovarian cancer, leptin serum levels did not correlate with the stage of the disease [146].

5.4. Resistin

Resistin has a known relationship with obesity [147]. In one early study, resistin serum levels in OSA patients were found to be decreased [134]. Another study did not confirm the association between OSA and resistin levels [148]; however, recent reports indicated that OSA was associated with increased resistin serum concentrations [112].
Although some previous studies and meta-analyses have reported a positive correlation between increased resistin levels and cancer [149,150,151,152], another meta-analysis did not confirm a relationship between resistin and cancer risk [153].

5.5. Vaspin

Vaspin serum concentrations were found to be increased in obesity [154]. Vaspin plasma levels were increased in severe OSA patients compared to the controls [155].
Vaspin serum concentrations were significantly elevated in hepatocellular carcinoma, especially in obese patients [156]. However, decreased vaspin serum concentrations were associated with an increased risk of endometrial cancer in patients [157].

5.6. Chemerin

Chemerin is positively associated with obesity [158,159]. Chemerin’s serum levels are increased in OSA patients [112,155] and correlate with the severity of the disease; they were postulated to serve as a biomarker of the presence and severity of OSA syndrome [160].
Increased circulating levels of chemerin were found in non-small-cell lung cancer, tongue, esophageal, gastric, and colorectal cancers, as well as in neuroblastoma [140]. However, there are controversies regarding its role in cancer development, as its anti-tumoral and pro-tumoral actions have been described [161].

5.7. Nesfatin

Circulating nesfatin-1 levels positively correlate with BMI in humans [162]. The function of nesfatin-1 is related to energy homeostasis, behavior and sleep [163]. In patients with major depression, nesfatin-1 was found to be involved in the regulation of sleep patterns [164]. In OSA patients, nesfatin-1 serum levels were decreased, compared to healthy controls, and correlated with the severity of this syndrome [165,166], although not all studies confirm such associations [167].
Nesfatin was found to be elevated in colon cancer tissue [168]. In patients with gastric cancer, its plasma levels have been proposed as a novel biomarker [169]. Conversely, in lung cancer patients, nesfatin-1 serum levels depended on weight loss and either were not different from healthy controls or decreased based on the patients and weight loss [170].

5.8. Visfatin

Visfatin serum concentrations positively correlate with BMI [171]. Increased plasma levels of visfatin were found in OSA patients [172], although not all studies confirm this observation [124]. In one study, visfatin plasma levels in severe OSA, although not different than in the controls, correlated positively with disturbances in sleep architecture [173]. Interestingly, circulating visfatin levels were significantly increased in patients with narcolepsy: a disease associated with profound sleep architecture disturbances [174]. Similarly, short sleep duration and disturbed sleep architecture (short REM sleep duration) in patients without OSA were associated with increased visfatin serum levels [139].
A recent meta-analysis of 14 studies encompassing 1616 patients revealed increased expression of visfatin in various cancers and the association of this adipokine with poorer overall survival, as well as with tumor size, tumor stage, and the presence of lymph nodes or distant metastases [175].

5.9. Osteopontin

Osteopontin levels increase in obesity [176]. Osteopontin plasma levels were not different in patients with and without OSA but increased with OSA severity and daytime sleepiness [177].
Osteopontin plays an important role in tumor progression, promoting, among other things, tumor growth and tumor cell invasion [178]. Increased expression of osteopontin was found in human bladder cancer cell lines [179]. Increased plasma levels of osteopontin were found in melanoma patients, especially in patients with metastases [180]. It was found in the cell lines that cancer cell motility was regulated by osteopontin secreted by the cancer cells [181].

5.10. Apelin

Apelin levels increase in obese patients [182]. Plasma apelin levels in OSA patients were found to either be increased [183] or not influenced by sleep breathing disorders [184,185].
Apelin favors the proliferation and progression of various cancers [186]. In a group of patients with diverse cancers, including lung cancer, gastrointestinal, breast, ovarian, uterus, and prostate cancers, apelin was proposed as a strong biomarker of cancer progression [187].

5.11. Retinol Binding Protein 4

Retinol binding protein 4 (RBP-4) is positively associated with obesity [188]. In OSA patients, increased plasma levels of RBP-4 were observed [189]. In another study, RBP-4 serum levels in OSA patients were not influenced by the severity of the disease and did not correlate with sleep disturbances but were significantly decreased during treatment with continuous positive airway pressure [190]. Serum RBP-4 levels were associated with sleep quality in pregnant women [191]. Sleep duration does not influence RBP-4 [139].
In some cancer types (e.g., breast cancer), increased RBP-4 plasma levels were found when compared to healthy controls and non-metastatic breast cancer patients [192].

5.12. Galectin-3

The expression of galectin-3 is upregulated in obese patients [193]. Serum galectin-3 levels in OSA were increased and correlated with the severity of this syndrome [194]. In another study, plasma galectin-3 levels were increased only in women with OSA [195]. In a study of patients with moderate to severe OSA, galectin-3 serum levels were not found to be a useful biomarker of the severity of the disease [196].
Galectin-3 contributes to the proliferation and progression of some cancers [197,198,199].
The summarization of the associations of adipokines and their pro- or anti-inflammatory properties with obesity, sleep disorders and cancer is presented in Table 1.

6. Final Remarks and Conclusions

Some of the peptides excreted by adipocytes and/or myocytes have pro-inflammatory proprieties (leptin, resistin, chemerin, vaspin, RBP-4 [202]) or anti-inflammatory proprieties (adiponectin, omentin, vaspin and irisin [202,214]). Cancer development and evolution may depend on chronic inflammation [215]; thus, an imbalance in the pro-inflammatory and anti-inflammatory effects of adipokines/adipomyokines may also be one of the factors influencing cancer development.
The presented review indicates that one of the possible pathways influencing the development of cancer may be the mutual relationship between obesity and/or sarcopenia, sleep quantity and quality, and adipokines/adipomyokines excretion. This indicates a tempting field for further studies on the associations of adipose tissue and skeletal muscles’ paracrine/endocrine function with sleep disturbances in cancer patients. Considering the high proportion of persons with obesity and sedentary lifestyles, as well as the associations of these conditions with sleep disturbances, more attention should be paid to the individual and combined effects on cancer pathophysiology.

Author Contributions

Conceptualization, A.B., C.E.K. and S.G.S.; data curation A.B., M.M. and M.E.; writing—original draft preparation A.B., C.E.K.; writing—review and editing A.B., H.M., S.G.S., C.E.K. and M.K.; visualization, C.D.; supervision, C.E.K., S.G.S., M.M.-H. and H.M.; project administration, M.K. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

This review is based on the PubMed publications.

Acknowledgments

The study is a part of research project number SUBZ.C110.23.061 at Wroclaw Medical University.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Ragino, Y.I.; Stakhneva, E.M.; Polonskaya, Y.V.; Kashtanova, E.V. The Role of Secretory Activity Molecules of Visceral Adipocytes in Abdominal Obesity in the Development of Cardiovascular Disease: A Review. Biomolecules. 2020, 10, 374. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  2. Hogas, S.; Bilha, S.C.; Branisteanu, D.; Hogas, M.; Gaipov, A.; Kanbay, M.; Covic, A. Potential novel biomarkers of cardiovascular dysfunction and disease: Cardiotrophin-1, adipokines and galectin-3. Arch. Med. Sci. 2017, 13, 897–913. [Google Scholar] [CrossRef] [PubMed]
  3. Laurens, C.; Bergouignan, A.; Moro, C. Exercise-Released Myokines in the Control of Energy Metabolism. Front Physiol. 2020, 11, 91. [Google Scholar] [CrossRef]
  4. Campolo, J.; Corradi, E.; Parolini, M.; Di Guglielmo, M.L.; Rizzardi, A.; Dellanoce, C.; Tarlarini, P.; Cattaneo, M.; Scioscioli, E.; Trivella, M.G.; et al. Gender-Specific Behaviour in Obesity Stages I-II: Imbalance of Aminothiol Status and Adipomyokine Profile in Subjects with Different Insulin Resistance Severity. Oxid. Med. Cell Longev. 2021, 2021, 9713582. [Google Scholar] [CrossRef] [PubMed]
  5. Raschke, S.; Eckel, J. Adipo-myokines: Two sides of the same coin-mediators of inflammation and mediators of exercise. Mediators Inflamm. 2013, 2013, 320724. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  6. Landecho, M.F.; Tuero, C.; Valentí, V.; Bilbao, I.; de la Higuera, M.; Frühbeck, G. Relevance of Leptin and Other Adipokines in Obesity-Associated Cardiovascular Risk. Nutrients 2019, 11, 2664. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  7. Batsis, J.A.; Villareal, D.T. Sarcopenic obesity in older adults: Aetiology, epidemiology and treatment strategies. Nat. Rev. Endocrinol. 2018, 14, 513–537. [Google Scholar] [CrossRef]
  8. Alizadeh Pahlavani, H. Exercise Therapy for People with Sarcopenic Obesity: Myokines and Adipokines as Effective Actors. Front. Endocrinol. 2022, 13, 811751. [Google Scholar] [CrossRef] [PubMed]
  9. Perna, S.; Spadaccini, D.; Rondanelli, M. Sarcopenic obesity: Time to target the phenotypes. J. Cachexia Sarcopenia Muscle 2019, 10, 710–711. [Google Scholar] [CrossRef] [Green Version]
  10. Baracos, V.E.; Arribas, L. Sarcopenic obesity: Hidden muscle wasting and its impact for survival and complications of cancer therapy. Ann. Oncol. 2018, 29, ii1–ii9. [Google Scholar] [CrossRef]
  11. Hilmi, M.; Jouinot, A.; Burns, R.; Pigneur, F.; Mounier, R.; Gondin, J.; Neuzillet, C.; Goldwasser, F. Body composition and sarcopenia: The next-generation of personalized oncology and pharmacology? Pharmacol. Ther. 2019, 196, 135–159. [Google Scholar] [CrossRef] [PubMed]
  12. Pana, A.; Sourtzi, P.; Kalokairinou, A.; Pastroudis, A.; Chatzopoulos, S.T.; Velonaki, V.S. Association between muscle strength and sleep quality and duration among middle-aged and older adults: A systematic review. Eur. Geriatr. Med. 2021, 12, 27–44. [Google Scholar] [CrossRef]
  13. Chien, M.Y.; Wang, L.Y.; Chen, H.C. The Relationship of Sleep Duration with Obesity and Sarcopenia in Community-Dwelling Older Adults. Gerontology 2015, 61, 399–406. [Google Scholar] [CrossRef]
  14. Lee, J.H.; Cho, J. Sleep and Obesity. Sleep Med. Clin. 2022, 17, 111–116. [Google Scholar] [CrossRef]
  15. Zhang, G.; Li, X.; Sui, C.; Zhao, H.; Zhao, J.; Hou, Y.; Du, Y. Incidence and risk factor analysis for sarcopenia in patients with cancer. Oncol. Lett. 2016, 11, 1230–1234. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  16. Williams, G.R.; Dunne, R.F.; Giri, S.; Shachar, S.S.; Caan, B.J. Sarcopenia in the Older Adult with Cancer. J. Clin. Oncol. 2021, 39, 2068–2078. [Google Scholar] [CrossRef] [PubMed]
  17. Karra, P.; Winn, M.; Pauleck, S.; Bulsiewicz-Jacobsen, A.; Peterson, L.; Coletta, A.; Doherty, J.; Ulrich, C.M.; Summers, S.A.; Gunter, M.; et al. Metabolic dysfunction and obesity-related cancer: Beyond obesity and metabolic syndrome. Obesity 2022, 30, 1323–1334. [Google Scholar] [CrossRef]
  18. Gao, Q.; Hu, K.; Gao, J.; Shang, Y.; Mei, F.; Zhao, L.; Chen, F.; Ma, B. Prevalence and prognostic value of sarcopenic obesity in patients with cancer: A systematic review and meta-analysis. Nutrition 2022, 101, 111704. [Google Scholar] [CrossRef]
  19. Gortan Cappellari, G.; Brasacchio, C.; Laudisio, D.; Lubrano, C.; Pivari, F.; Barrea, L.; Muscogiuri, G.; Savastano, S.; Colao, A. Obesity Programs of Nutrition, Education, Research and Assessment (OPERA) group. Sarcopenic obesity: What about in the cancer setting? Nutrition 2022, 98, 111624. [Google Scholar] [CrossRef]
  20. Silveira, E.A.; da Silva Filho, R.R.; Spexoto, M.C.B.; Haghighatdoost, F.; Sarrafzadegan, N.; de Oliveira, C. The Role of Sarcopenic Obesity in Cancer and Cardiovascular Disease: A Synthesis of the Evidence on Pathophysiological Aspects and Clinical Implications. Int. J. Mol. Sci. 2021, 22, 4339. [Google Scholar] [CrossRef]
  21. Sun, M.Y.; Chang, C.L.; Lu, C.Y.; Wu, S.Y.; Zhang, J.Q. Sarcopenia as an Independent Risk Factor for Specific Cancers: A Propensity Score-Matched Asian Population-Based Cohort Study. Nutrients 2022, 14, 1910. [Google Scholar] [CrossRef]
  22. Shachar, S.S.; Williams, G.R.; Muss, H.B.; Nishijima, T.F. Prognostic value of sarcopenia in adults with solid tumours: A meta-analysis and systematic review. Eur. J. Cancer 2016, 57, 58–67. [Google Scholar] [CrossRef]
  23. Aleixo, G.F.P.; Shachar, S.S.; Nyrop, K.A.; Muss, H.B.; Battaglini, C.L.; Williams, G.R. Bioelectrical Impedance Analysis for the Assessment of Sarcopenia in Patients with Cancer: A Systematic Review. Oncologist 2020, 25, 170–182. [Google Scholar] [CrossRef]
  24. Brown, J.C.; Cespedes Feliciano, E.M.; Caan, B.J. The evolution of body composition in oncology-epidemiology, clinical trials, and the future of patient care: Facts and numbers. J. Cachexia Sarcopenia Muscle 2018, 9, 1200–1208. [Google Scholar] [CrossRef] [Green Version]
  25. Bilen, M.A.; Martini, D.J.; Liu, Y.; Shabto, J.M.; Brown, J.T.; Williams, M.; Khan, A.I.; Speak, A.; Lewis, C.; Collins, H.; et al. Combined Effect of Sarcopenia and Systemic Inflammation on Survival in Patients with Advanced Stage Cancer Treated with Immunotherapy. Oncologist 2020, 25, e528–e535. [Google Scholar] [CrossRef] [Green Version]
  26. Wiegert, E.V.M.; de Oliveira, L.C.; Calixto-Lima, L.; Borges, N.A.; Rodrigues, J.; da Mota Esilva Lopes, M.S.; Peres, W.A.F. Association between low muscle mass and survival in incurable cancer patients: A systematic review. Nutrition 2020, 72, 110695. [Google Scholar] [CrossRef] [PubMed]
  27. Pearson-Stuttard, J.; Zhou, B.; Kontis, V.; Bentham, J.; Gunter, M.J.; Ezzati, M. Worldwide burden of cancer attributable to diabetes and high body-mass index: A comparative risk assessment. Lancet Diabetes Endocrinol. 2018, 6, e6–e15. [Google Scholar] [CrossRef] [PubMed]
  28. Fang, X.; Wei, J.; He, X.; Lian, J.; Han, D.; An, P.; Zhou, T.; Liu, S.; Wang, F.; Min, J. Quantitative association between body mass index and the risk of cancer: A global Meta-analysis of prospective cohort studies. Int. J. Cancer 2018, 143, 1595–1603. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  29. Arnold, M.; Pandeya, N.; Byrnes, G.; Renehan, P.A.G.; Stevens, G.A.; Ezzati, P.M.; Ferlay, J.; Miranda, J.J.; Romieu, I.; Dikshit, R.; et al. Global burden of cancer attributable to high body-mass index in 2012: A population-based study. Lancet Oncol. 2015, 16, 36–46. [Google Scholar] [CrossRef] [PubMed]
  30. Calle, E.E.; Rodriguez, C.; Walker-Thurmond, K.; Thun, M.J. Overweight, obesity, and mortality from cancer in a prospectively studied cohort of U.S. adults. N. Engl. J. Med. 2003, 348, 1625–1638. [Google Scholar] [CrossRef] [Green Version]
  31. Gustafson, B. Adipose tissue, inflammation and atherosclerosis. J. Atheroscler. Thromb. 2010, 17, 332–341. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  32. Budek, M.; Nuszkiewicz, J.; Piórkowska, A.; Czuczejko, J.; Szewczyk-Golec, K. Inflammation Related to Obesity in the Etiopathogenesis of Gastroenteropancreatic Neuroendocrine Neoplasms. Biomedicines 2022, 10, 2660. [Google Scholar] [CrossRef]
  33. Kolb, R.; Sutterwala, F.S.; Zhang, W. Obesity and cancer: Inflammation bridges the two. Curr. Opin. Pharmacol. 2016, 29, 77–89. [Google Scholar] [CrossRef] [Green Version]
  34. Le, A.; Udupa, S.; Zhang, C. The Metabolic Interplay between Cancer and Other Diseases. Trends Cancer 2019, 5, 809–821. [Google Scholar] [CrossRef] [Green Version]
  35. Donohoe, C.L.; Lysaght, J.; O’Sullivan, J.; Reynolds, J.V. Emerging Concepts Linking Obesity with the Hallmarks of Cancer. Trends Endocrinol. Metab. 2017, 28, 46–62. [Google Scholar] [CrossRef] [PubMed]
  36. Donohoe, F.; Wilkinson, M.; Baxter, E.; Brennan, D.J. Mitogen-Activated Protein Kinase (MAPK) and Obesity-Related Cancer. Int. J. Mol. Sci. 2020, 21, 1241. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  37. Mayoral, L.P.; Andrade, G.M.; Mayoral, E.P.; Huerta, T.H.; Canseco, S.P.; Rodal Canales, F.J.; Cabrera-Fuentes, H.A.; Martinez Cruz, M.; Perez Santiago, A.D.; Alpuche, J.J.; et al. Obesity subtypes, related biomarkers &heterogeneity. Indian J. Med. Res. 2020, 151, 11–21. [Google Scholar] [CrossRef]
  38. Engin, A. The Definition and Prevalence of Obesity and Metabolic Syndrome. Adv. Exp. Med. Biol. 2017, 960, 1–17. [Google Scholar] [CrossRef]
  39. Ferrer, R.; Pardina, E.; Rossell, J.; Oller, L.; Viñas, A.; Baena-Fustegueras, J.A.; Lecube, A.; Vargas, V.; Balibrea, J.M.; Caubet, E.; et al. Morbidly “Healthy” Obese Are Not Metabolically Healthy but Less Metabolically Imbalanced Than Those with Type 2 Diabetes or Dyslipidemia. Obes. Surg. 2015, 25, 1380–1391. [Google Scholar] [CrossRef]
  40. Carneiro, I.P.; Mazurak, V.C.; Prado, C.M. Clinical Implications of Sarcopenic Obesity in Cancer. Curr. Oncol. Rep. 2016, 18, 62. [Google Scholar] [CrossRef]
  41. Mintziras, I.; Miligkos, M.; Wächter, S.; Manoharan, J.; Maurer, E.; Bartsch, D.K. Sarcopenia and sarcopenic obesity are significantly associated with poorer overall survival in patients with pancreatic cancer: Systematic review and meta-analysis. Int. J. Surg. 2018, 59, 19–26. [Google Scholar] [CrossRef] [PubMed]
  42. Fattouh, M.; Chang, G.Y.; Ow, T.J.; Shifteh, K.; Rosenblatt, G.; Patel, V.M.; Smith, R.V.; Prystowsky, M.B.; Schlecht, N.F. Association between pretreatment obesity, sarcopenia, and survival in patients with head and neck cancer. Head Neck 2019, 41, 707–714. [Google Scholar] [CrossRef]
  43. Chargi, N.; Bril, S.I.; Swartz, J.E.; Wegner, I.; Willems, S.M.; de Bree, R. Skeletal muscle mass is an imaging biomarker for decreased survival in patients with oropharyngeal squamous cell carcinoma. Oral. Oncol. 2020, 101, 104519. [Google Scholar] [CrossRef]
  44. Palmela, C.; Velho, S.; Agostinho, L.; Branco, F.; Santos, M.; Santos, M.P.; Oliveira, M.H.; Strecht, J.; Maio, R.; Cravo, M.; et al. Body Composition as a Prognostic Factor of Neoadjuvant Chemotherapy Toxicity and Outcome in Patients with Locally Advanced Gastric Cancer. J. Gastric Cancer 2017, 17, 74–87. [Google Scholar] [CrossRef] [Green Version]
  45. Kim, Y.M.; Kim, J.H.; Baik, S.J.; Chun, J.; Youn, Y.H.; Park, H. Sarcopenia and Sarcopenic Obesity as Novel Risk Factors for Gastric Carcinogenesis: A Health Checkup Cohort Study. Front. Oncol. 2019, 9, 1249. [Google Scholar] [CrossRef]
  46. Okumura, S.; Kaido, T.; Hamaguchi, Y.; Kobayashi, A.; Shirai, H.; Yao, S.; Yagi, S.; Kamo, N.; Hatano, E.; Okajima, H.; et al. Visceral Adiposity and Sarcopenic Visceral Obesity are Associated with Poor Prognosis After Resection of Pancreatic Cancer. Ann. Surg. Oncol. 2017, 24, 3732–3740. [Google Scholar] [CrossRef] [PubMed]
  47. Gruber, E.S.; Jomrich, G.; Tamandl, D.; Gnant, M.; Schindl, M.; Sahora, K. Sarcopenia and sarcopenic obesity are independent adverse prognostic factors in resectable pancreatic ductal adenocarcinoma. PLoS ONE 2019, 14, e0215915. [Google Scholar] [CrossRef]
  48. Chan, M.Y.; Chok, K.S.H. Sarcopenia in pancreatic cancer—Effects on surgical outcomes and chemotherapy. World J. Gastrointest. Oncol. 2019, 11, 527–537. [Google Scholar] [CrossRef]
  49. Tan, B.H.; Birdsell, L.A.; Martin, L.; Baracos, V.E.; Fearon, K.C. Sarcopenia in an overweight or obese patient is an adverse prognostic factor in pancreatic cancer. Clin. Cancer Res. 2009, 15, 6973–6979. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  50. Stangl-Kremser, J.; D’Andrea, D.; Vartolomei, M.; Abufaraj, M.; Goldner, G.; Baltzer, P.; Shariat, S.F.; Tamandl, D. Prognostic value of nutritional indices and body composition parameters including sarcopenia in patients treated with radiotherapy for urothelial carcinoma of the bladder. Urol. Oncol. 2019, 37, 372–379. [Google Scholar] [CrossRef]
  51. Jabbour, J.; Manana, B.; Zahreddine, A.; Saade, C.; Charafeddine, M.; Bazarbachi, A.; Blaise, D.; El-Cheikh, J. Sarcopenic obesity derived from PET/CT predicts mortality in lymphoma patients undergoing hematopoietic stem cell transplantation. Curr. Res. Transl. Med. 2019, 67, 93–99. [Google Scholar] [CrossRef]
  52. Chen, W.Z.; Chen, X.D.; Ma, L.L.; Zhang, F.M.; Lin, J.; Zhuang, C.L.; Yu, Z.; Chen, X.L.; Chen, X.X. Impact of Visceral Obesity and Sarcopenia on Short-Term Outcomes After Colorectal Cancer Surgery. Dig. Dis. Sci. 2018, 63, 1620–1630. [Google Scholar] [CrossRef] [PubMed]
  53. Caan, B.J.; Meyerhardt, J.A.; Kroenke, C.H.; Alexeeff, S.; Xiao, J.; Weltzien, E.; Feliciano, E.C.; Castillo, A.L.; Quesenberry, C.P.; Kwan, M.L.; et al. Explaining the Obesity Paradox: The Association between Body Composition and Colorectal Cancer Survival (C-SCANS Study). Cancer Epidemiol. Biomarkers Prev. 2017, 26, 1008–1015. [Google Scholar] [CrossRef] [Green Version]
  54. Kobayashi, A.; Kaido, T.; Hamaguchi, Y.; Okumura, S.; Shirai, H.; Yao, S.; Kamo, N.; Yagi, S.; Taura, K.; Okajima, H.; et al. Impact of Sarcopenic Obesity on Outcomes in Patients Undergoing Hepatectomy for Hepatocellular Carcinoma. Ann. Surg. 2019, 269, 924–931. [Google Scholar] [CrossRef]
  55. Hamaguchi, Y.; Kaido, T.; Okumura, S.; Kobayashi, A.; Shirai, H.; Yao, S.; Yagi, S.; Kamo, N.; Seo, S.; Taura, K.; et al. Preoperative Visceral Adiposity and Muscularity Predict Poor Outcomes after Hepatectomy for Hepatocellular Carcinoma. Liver Cancer 2019, 8, 92–109. [Google Scholar] [CrossRef] [PubMed]
  56. Kroh, A.; Uschner, D.; Lodewick, T.; Eickhoff, R.M.; Schöning, W.; Ulmer, F.T.; Neumann, U.P.; Binnebösel, M. Impact of body composition on survival and morbidity after liver resection in hepatocellular carcinoma patients. Hepatobiliary Pancreat. Dis. Int. 2019, 18, 28–37. [Google Scholar] [CrossRef] [PubMed]
  57. Dijksterhuis, W.P.M.; Pruijt, M.J.; van der Woude, S.O.; Klaassen, R.; Kurk, S.A.; van Oijen, M.G.H.; van Laarhoven, H.W.M. Association between body composition, survival, and toxicity in advanced esophagogastric cancer patients receiving palliative chemotherapy. J. Cachexia Sarcopenia Muscle 2019, 10, 199–206. [Google Scholar] [CrossRef] [Green Version]
  58. Kiss, N.; Beraldo, J.; Everitt, S. Early Skeletal Muscle Loss in Non-Small Cell Lung Cancer Patients Receiving Chemoradiation and Relationship to Survival. Support Care Cancer 2019, 27, 2657–2664. [Google Scholar] [CrossRef]
  59. Rier, H.N.; Jager, A.; Sleijfer, S.; van Rosmalen, J.; Kock, M.C.J.M.; Levin, M.D. Low muscle attenuation is a prognostic factor for survival in metastatic breast cancer patients treated with first line palliative chemotherapy. Breast 2017, 31, 9–15. [Google Scholar] [CrossRef] [PubMed]
  60. Lodewick, T.M.; van Nijnatten, T.J.; van Dam, R.M.; van Mierlo, K.; Dello, S.A.; Neumann, U.P.; Olde Damink, S.W.; Dejong, C.H. Are sarcopenia, obesity and sarcopenic obesity predictive of outcome in patients with colorectal liver metastases? HPB 2015, 17, 438–446. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  61. An, C.; Pipia, I.; Ruiz, A.S.; Argüelles, I.; An, M.; Wase, S.; Peng, G. The molecular link between obesity and genomic instability in cancer development. Cancer Lett. 2023, 555, 216035. [Google Scholar] [CrossRef] [PubMed]
  62. Fodor, A.; Lazar, A.L.; Buchman, C.; Tiperciuc, B.; Orasan, O.H.; Cozma, A. MicroRNAs: The Link between the Metabolic Syndrome and Oncogenesis. Int. J. Mol. Sci. 2021, 22, 6337. [Google Scholar] [CrossRef] [PubMed]
  63. Rios-Colon, L.; Chijioke, J.; Niture, S.; Afzal, Z.; Qi, Q.; Srivastava, A.; Ramalinga, M.; Kedir, H.; Cagle, P.; Arthur, E.; et al. Leptin modulated microRNA-628-5p targets Jagged-1 and inhibits prostate cancer hallmarks. Sci. Rep. 2022, 12, 10073. [Google Scholar] [CrossRef]
  64. Gluba-Brzózka, A.; Rysz, J.; Ławiński, J.; Franczyk, B. Renal Cell Cancer and Obesity. Int. J. Mol. Sci. 2022, 23, 3404. [Google Scholar] [CrossRef]
  65. Mendoza-Pérez, J.; Gu, J.; Herrera, L.A.; Tannir, N.M.; Zhang, S.; Matin, S.; Karam, J.A.; Wood, C.G.; Wu, X. Prognostic significance of promoter CpG island methylation of obesity-related genes in patients with nonmetastatic renal cell carcinoma. Cancer 2017, 123, 3617–3627. [Google Scholar] [CrossRef] [Green Version]
  66. Abdu Allah, A.M.; El-Hefnway, S.M.; Alhanafy, A.M.; Zahran, A.M.; Kasem, H.E. Leptin receptor gene (A/G) polymorphism rs1137101 and renal cell carcinoma. Mol. Cell. Biochem. 2018, 448, 137–144. [Google Scholar] [CrossRef]
  67. Cruz-Jentoft, A.J.; Baeyens, J.P.; Bauer, J.M.; Boirie, Y.; Cederholm, T.; Landi FMartin, F.C.; Michel, J.P.; Rolland, Y.; Schneider, S.M.; Topinková, E.; et al. Sarcopenia: European consensus on definition and diagnosis: Report of the European Working Group on Sarcopenia in Older People. Age Ageing 2010, 39, 412–423. [Google Scholar] [CrossRef] [Green Version]
  68. Fielding, R.A.; Vellas, B.; Evans, W.J.; Bhasin, S.; Morley, J.E.; Newman, A.B.; Abellan van Kan, G.; Andrieu, S.; Bauer, J.; Breuille, D.; et al. Sarcopenia: An undiagnosed condition in older adults. Current consensus definition: Prevalence, etiology, and consequences. International working group on sarcopenia. J. Am. Med. Dir. Assoc. 2011, 12, 249–256. [Google Scholar] [CrossRef] [Green Version]
  69. Fex, A.; Barbat-Artigas, S.; Dupontgand, S.; Filion, M.E.; Karelis, A.D.; Aubertin-Leheudre, M. Relationship between long sleep duration and functional capacities in postmenopausal women. J. Clin. Sleep Med. 2012, 8, 309–313. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  70. Kwon, Y.J.; Jang, S.Y.; Park, E.C.; Cho, A.R.; Shim, J.Y.; Linton, J.A. Long Sleep Duration is Associated with Sarcopenia in Korean Adults Based on Data from the 2008–2011 KNHANES. J. Clin. Sleep Med. 2017, 13, 1097–1104. [Google Scholar] [CrossRef] [PubMed]
  71. Kim, R.H.; Kim, K.I.; Kim, J.H.; Park, Y.S. Association between Sleep Duration and Body Composition Measures in Korean Adults: The Korea National Health and Nutrition Examination Survey 2010. Korean J. Fam. Med. 2018, 39, 219–224. [Google Scholar] [CrossRef]
  72. Choi, Y.I.; Park, D.K.; Chung, J.W.; Kim, K.O.; Kwon, K.A.; Kim, Y.J. Circadian rhythm disruption is associated with an increased risk of sarcopenia: A nationwide population-based study in Korea. Sci. Rep. 2019, 9, 12015. [Google Scholar] [CrossRef] [Green Version]
  73. Vasconcelos, K.S.; Dias, J.M.; Bastone Ade, C.; Vieira, R.A.; Andrade, A.C.; Perracini, M.R.; Guerra, R.O.; Dias, R.C. Handgrip Strength Cutoff Points to Identify Mobility Limitation in Community-dwelling Older People and Associated Factors. J. Nutr. Health Aging 2016, 20, 306–315. [Google Scholar] [CrossRef]
  74. Ida, S.; Kaneko, R.; Nagata, H.; Noguchi, Y.; Araki, Y.; Nakai, M.; Ito, S.; Ishihara, Y.; Imataka, K.; Murata, K. Association between sarcopenia and sleep disorder in older patients with diabetes. Geriatr. Gerontol. Int. 2019, 19, 399–403. [Google Scholar] [CrossRef]
  75. Locquet, M.; Beaudart, C.; Delandsheere, L.; Reginster, J.Y.; Bruyère, O. Subjective Sleep Quality among Sarcopenic and Non-Sarcopenic Older Adults: Results from the SarcoPhAge Cohort. J. Frailty Aging 2018, 7, 176–181. [Google Scholar] [CrossRef] [PubMed]
  76. Lucassen, E.A.; de Mutsert, R.; le Cessie, S.; Appelman-Dijkstra, N.M.; Rosendaal, F.R.; van Heemst, D.; den Heijer, M.; Biermasz, N.R.; NEO Study Group. 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 2017, 12, e0176685. [Google Scholar] [CrossRef] [Green Version]
  77. Auyeung, T.W.; Kwok, T.; Leung, J.; Lee, J.S.; Ohlsson, C.; Vandenput, L.; Wing, Y.K.; Woo, J. Sleep Duration and Disturbances Were Associated With Testosterone Level, Muscle Mass, and Muscle Strength—A Cross-Sectional Study in 1274 Older Men. J. Am. Med. Dir. Assoc. 2015, 16, 630.e1–630.e6. [Google Scholar] [CrossRef]
  78. Buchmann, N.; Spira, D.; Norman, K.; Demuth, I.; Eckardt, R.; Steinhagen-Thiessen, E. Sleep, Muscle Mass and Muscle Function in Older People. Dtsch. Arztebl. Int. 2016, 113, 253–260. [Google Scholar] [CrossRef] [Green Version]
  79. Yang, C.W.; Li, C.I.; Li, T.C.; Liu, C.S.; Lin, C.H.; Lin, W.Y.; Lin, C.C. Combined Effects of Having Sleep Problems and Taking Sleeping Pills on the Skeletal Muscle Mass and Performance of Community-Dwelling Elders. Sci. Rep. 2019, 9, 13760. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  80. Smith, L.; Shin, J.I.; Veronese, N.; Soysal, P.; López Sánchez, G.F.; Pizzol, D.; Demurtas, J.; Tully, M.A.; Barnett, Y.; Butler, L.; et al. Sleep duration and sarcopenia in adults aged ≥ 65 years from low and middle-income countries. Aging Clin. Exp. Res. 2022, 34, 1573–1581. [Google Scholar] [CrossRef] [PubMed]
  81. Tan, X.; Titova, O.E.; Lindberg, E.; Elmståhl, S.; Lind, L.; Schiöth, H.B.; Benedict, C. Association Between Self-Reported Sleep Duration and Body Composition in Middle-Aged and Older Adults. J. Clin. Sleep Med. 2019, 15, 431–435. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  82. Rubio-Arias, J.Á.; Rodríguez-Fernández, R.; Andreu, L.; Martínez-Aranda, L.M.; Martínez-Rodriguez, A.; Ramos-Campo, D.J. Effect of Sleep Quality on the Prevalence of Sarcopenia in Older Adults: A Systematic Review with Meta-Analysis. J. Clin. Med. 2019, 8, 2156. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  83. Jike, M.; Itani, O.; Watanabe, N.; Buysse, D.J.; Kaneita, Y. Long sleep duration and health outcomes: A systematic review, meta-analysis and meta-regression. Sleep Med. Rev. 2018, 39, 25–36. [Google Scholar] [CrossRef] [PubMed]
  84. Muscogiuri, G.; Barrea, L.; Annunziata, G.; Di Somma, C.; Laudisio, D.; Colao, A.; Savastano, S. Obesity and sleep disturbance: The chicken or the egg? Crit. Rev. Food Sci. Nutr. 2019, 59, 2158–2165. [Google Scholar] [CrossRef]
  85. Ryan, S.; Arnaud, C.; Fitzpatrick, S.F.; Gaucher, J.; Tamisier, R.; Pépin, J.L. Adipose tissue as a key player in obstructive sleep apnoea. Eur. Respir. Rev. 2019, 28, 190006. [Google Scholar] [CrossRef]
  86. Piovezan, R.D.; Hirotsu, C.; Moizinho, R.; de Sá Souza, H.; D’Almeida, V.; Tufik, S.; Poyares, D. Associations between sleep conditions and body composition states: Results of the EPISONO study. J. Cachexia Sarcopenia Muscle 2019, 10, 962–973. [Google Scholar] [CrossRef] [PubMed]
  87. Liu, J.; Qi, B.; Gan, L.; Shen, Y.; Zou, Y. A Bibliometric Analysis of the Literature on Irisin from 2012–2021. Int. J. Environ. Res. Public Health 2022, 19, 6153. [Google Scholar] [CrossRef]
  88. De Meneck, F.; de Souza, L.V.; Brioschi, M.L.; Franco, M.D.C. Emerging evidence for the opposite role of circulating irisin levels and brown adipose tissue activity measured by infrared thermography in anthropometric and metabolic profile during childhood. J. Therm. Biol. 2021, 99, 103010. [Google Scholar] [CrossRef]
  89. Sumsuzzman, D.M.; Jin, Y.; Choi, J.; Yu, J.H.; Lee, T.H.; Hong, Y. Pathophysiological role of endogenous irisin against tumorigenesis and metastasis: Is it a potential biomarker and therapeutic? Tumour. Biol. 2019, 41, 1010428319892790. [Google Scholar] [CrossRef] [Green Version]
  90. Ulualan, G.; Kiraz, Z.K.; Kırel, B. Relation of serum irisin levels to obesity and non-alcoholic fatty liver disease. Turk. J. Pediatr. 2022, 64, 246–254. [Google Scholar] [CrossRef]
  91. Shantavasinkul, P.C.; Omotosho, P.; Corsino, L.; Muehlbauer, M.J.; Chattranukulchai, P.; Torquati, A. Changes of Circulating Irisin and High-Sensitivity C-Reactive Protein Levels in Morbidly Obese Individuals with Type 2 Diabetes After Roux-en-Y Gastric Bypass. J. Laparoendosc. Adv. Surg. Tech A 2022, 32, 817–822. [Google Scholar] [CrossRef] [PubMed]
  92. Gamal, R.M.; Mohamed, M.E.; Hammam, N.; El Fetoh, N.A.; Rashed, A.M.; Furst, D.E. Preliminary study of the association of serum irisin levels with poor sleep quality in rheumatoid arthritis patients. Sleep Med. 2019, 67, 71–76. [Google Scholar] [CrossRef]
  93. Li, Y.; Li, X.; Sun, D.; Cai, S. Association of serum irisin concentrations with the presence and severity of obstructive sleep apnea syndrome. J. Clin. Lab. Anal. 2017, 31, e22077. [Google Scholar] [CrossRef]
  94. More, C.E.; Papp, C.; Harsanyi, S.; Gesztelyi, R.; Mikaczo, A.; Tajti, G.; Kardos, L.; Seres, I.; Lorincz, H.; Csapo, K.; et al. Altered irisin/BDNF axis parallels excessive daytime sleepiness in obstructive sleep apnea patients. Respir. Res. 2019, 20, 67. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  95. Korta, P.; Pocheć, E.; Mazur-Biały, A. Irisin as a Multifunctional Protein: Implications for Health and Certain Diseases. Medicina 2019, 55, 485. [Google Scholar] [CrossRef] [Green Version]
  96. Liu, J.; Song, N.; Huang, Y.; Chen, Y. Irisin inhibits pancreatic cancer cell growth via the AMPK-mTOR pathway. Sci. Rep. 2018, 8, 15247. [Google Scholar] [CrossRef] [Green Version]
  97. Kong, G.; Jiang, Y.; Sun, X.; Cao, Z.; Zhang, G.; Zhao, Z.; Zhao, Y.; Yu, Q.; Cheng, G. Irisin reverses the IL-6 induced epithelial-mesenchymal transition in osteosarcoma cell migration and invasion through the STAT3/Snail signaling pathway. Oncol. Rep. 2017, 38, 2647–2656. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  98. Shao, L.; Li, H.; Chen, J.; Song, H.; Zhang, Y.; Wu, F.; Wang, W.; Zhang, W.; Wang, F.; Li, H.; et al. Irisin suppresses the migration, proliferation, and invasion of lung cancer cells via inhibition of epithelial-to-mesenchymal transition. Biochem. Biophys. Res. Commun. 2017, 485, 598–605. [Google Scholar] [CrossRef] [PubMed]
  99. Gannon, N.P.; Vaughan, R.A.; Garcia-Smith, R.; Bisoffi, M.; Trujillo, K.A. Effects of the exercise-inducible myokine irisin on malignant and non-malignant breast epithelial cell behavior in vitro. Int. J. Cancer 2015, 136, E197–E202. [Google Scholar] [CrossRef] [PubMed]
  100. Moon, H.S.; Mantzoros, C.S. Regulation of cell proliferation and malignant potential by irisin in endometrial, colon, thyroid and esophageal cancer cell lines. Metabolism 2014, 63, 188–193. [Google Scholar] [CrossRef] [PubMed]
  101. Shi, G.; Tang, N.; Qiu, J.; Zhang, D.; Huang, F.; Cheng, Y.; Ding, K.; Li, W.; Zhang, P.; Tan, X. Irisin stimulates cell proliferation and invasion by targeting the PI3K/AKT pathway in human hepatocellular carcinoma. Biochem. Biophys. Res. Commun. 2017, 493, 585–591. [Google Scholar] [CrossRef] [PubMed]
  102. Provatopoulou, X.; Georgiou, G.P.; Kalogera, E.; Kalles, V.; Matiatou, M.A.; Papapanagiotou, I.; Sagkriotis, A.; Zografos, G.C.; Gounaris, A. Serum irisin levels are lower in patients with breast cancer: Association with disease diagnosis and tumor characteristics. BMC Cancer 2015, 15, 898. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  103. Zhu, H.; Liu, M.; Zhang, N.; Pan, H.; Lin, G.; Li, N.; Wang, L.; Yang, H.; Yan, K.; Gong, F. Serum and Adipose Tissue mRNA Levels of ATF3 and FNDC5/Irisin in Colorectal Cancer Patients with or Without Obesity. Front. Physiol. 2018, 9, 1125. [Google Scholar] [CrossRef] [PubMed]
  104. Zhang, Z.P.; Zhang, X.F.; Li, H.; Liu, T.J.; Zhao, Q.P.; Huang, L.H.; Cao, Z.J.; He, L.M.; Hao, D.J. Serum irisin associates with breast cancer to spinal metastasis. Medicine 2018, 97, e0524. [Google Scholar] [CrossRef]
  105. Zhang, J.; Ke, M.; Ren, Y.; Bi, J.; Du, Z.; Zhang, M.; Wang, Y.; Zhang, L.; Wu, Z.; Lv, Y.; et al. Serum Irisin Predicts Posthepatectomy Complications in Patients with Hepatocellular Carcinoma. Dis. Markers. 2019, 2019, 9850191. [Google Scholar] [CrossRef] [Green Version]
  106. Aslan, R.; Alp, H.H.; Eryılmaz, R.; Huyut, Z.; Sevim, M.; Araz, Ş.; Ertas, K.; Taken, K. Can the Irisin be a Biomarker for Prostate Cancer? A Case Control Study. Asian Pac. J. Cancer Prev. 2020, 21, 505–509. [Google Scholar] [CrossRef]
  107. Esawy, M.M.; Abdel-Samd, K.M. The diagnostic and prognostic roles of serum irisin in bladder cancer. Curr. Probl. Cancer 2019, 44, 100529. [Google Scholar] [CrossRef]
  108. Us Altay, D.U.; Keha, E.E.; Karagüzel, E.; Menteşe, A.; Yaman, S.O.; Alver, A. The Diagnostic Value of FNDC5/Irisin in Renal Cell Cancer. Int. Braz. J. Urol. 2018, 44, 734–739. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  109. Panagiotou, G.; Triantafyllidou, S.; Tarlatzis, B.C.; Papakonstantinou, E. Serum Levels of Irisin and Omentin-1 in Breast Neoplasms and Their Association with Tumor Histology. Int. J. Endocrinol. 2021, 2021, 6656671. [Google Scholar] [CrossRef]
  110. Varda, N.M.; Medved, M.; Ojsteršek, L. The associations between some biological markers, obesity, and cardiovascular risk in Slovenian children and adolescents. BMC Pediatr. 2020, 20, 81. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  111. Lu, M.; Fang, F.; Wang, Z.; Wei, P.; Hu, C.; Wei, Y. Association between serum/plasma levels of adiponectin and obstructive sleep apnea hypopnea syndrome: A meta-analysis. Lipids Health Dis. 2019, 18, 30. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  112. Xu, X.; Xu, J. Effects of different obesity-related adipokines on the occurrence of obstructive sleep apnea. Endocr. J. 2020, 67, 485–500. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  113. Celikhisar, H.; Ilkhan, G.D. Alterations in Serum Adropin, Adiponectin, and Proinflammatory Cytokine Levels in OSAS. Can. Respir. J. 2020, 2020, 2571283. [Google Scholar] [CrossRef] [PubMed]
  114. Silva, W.A.A.; Almeida-Pititto, B.; Santos, R.B.; Aielo, A.N.; Giatti, S.; Parise, B.K.; Souza, S.P.; Vivolo, S.F.; Lotufo, P.A.; Bensenor, I.M.; et al. Obstructive sleep apnea is associated with lower adiponectin and higher cholesterol levels independently of traditional factors and other sleep disorders in middle-aged adults: The ELSA-Brasil cohort. Sleep Breath. 2021, 25, 1935–1944. [Google Scholar] [CrossRef] [PubMed]
  115. Magnusdottir, S.; Thomas, R.J.; Hilmisson, H. Can improvements in sleep quality positively affect serum adiponectin-levels in patients with obstructive sleep apnea? Sleep Med. 2021, 84, 324–333. [Google Scholar] [CrossRef]
  116. Padilha, H.G.; Crispim, C.A.; Zimberg, I.Z.; De-Souza, D.A.; Waterhouse, J.; Tufik, S.; de-Mello, M.T. A link between sleep loss, glucose metabolism and adipokines. Braz. J. Med. Biol. Res. 2011, 44, 992–999. [Google Scholar] [CrossRef] [Green Version]
  117. Oliveira, R.F.; Daniele, T.M.D.C.; Façanha, C.F.S.; Forti, A.C.E.; Bruin, P.F.C.; Bruin, V.M.S. Adiponectin levels and sleep deprivation in patients with endocrine metabolic disorders. Rev. Assoc. Med. Bras. 2018, 64, 1122–1128. [Google Scholar] [CrossRef]
  118. Cha, E.; Talman, M.S.; Massey, A.H.; Yan, F.; Rogers, A.E. Sleep, Lifestyle Behaviors, and Cardiometabolic Health Markers in Overweight/Obese Young Adults: A Pilot Study Using the SenseWear® Armband. Biol. Res. Nurs. 2018, 20, 541–548. [Google Scholar] [CrossRef]
  119. Chu, D.T.; Phuong, T.N.T.; Tien, N.L.B.; Tran, D.K.; Nguyen, T.T.; Thanh, V.V.; Quang, T.L.; Minh, L.B.; Pham, V.H.; Ngoc, V.T.N.; et al. The Effects of Adipocytes on the Regulation of Breast Cancer in the Tumor Microenvironment: An Update. Cells 2019, 8, 857. [Google Scholar] [CrossRef] [Green Version]
  120. Wei, T.; Ye, P.; Peng, X.; Wu, L.L.; Yu, G.Y. Circulating adiponectin levels in various malignancies: An updated meta-analysis of 107 studies. Oncotarget 2016, 7, 48671–48691. [Google Scholar] [CrossRef] [Green Version]
  121. Yu, Z.; Tang, S.; Ma, H.; Duan, H.; Zeng, Y. Association of serum adiponectin with breast cancer: A meta-analysis of 27 case-control studies. Medicine 2019, 98, e14359. [Google Scholar] [CrossRef] [PubMed]
  122. Yap, N.Y.; Yap, F.N.; Perumal, K.; Rajandram, R. Circulating adiponectin as a biomarker in renal cell carcinoma: A systematic review and meta-analysis. Biomarkers 2019, 24, 607–614. [Google Scholar] [CrossRef] [PubMed]
  123. de Souza Batista, C.M.; Yang, R.Z.; Lee, M.J.; Glynn, N.M.; Yu, D.Z.; Pray, J.; Ndubuizu, K.; Patil, S.; Schwartz, A.; Kligman, M.; et al. Omentin plasma levels and gene expression are decreased in obesity. Diabetes 2007, 56, 1655–1661. [Google Scholar] [CrossRef] [Green Version]
  124. Zhang, D.M.; Pang, X.L.; Huang, R.; Gong, F.Y.; Zhong, X.; Xiao, Y. Adiponectin, Omentin, Ghrelin, and Visfatin Levels in Obese Patients with Severe Obstructive Sleep Apnea. Biomed. Res. Int. 2018, 2018, 3410135. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  125. Zhang, Y.; Zhao, X.; Chen, M. Autocrine action of adipokine omentin-1 in the SW480 colon cancer cell line. Oncol. Lett. 2020, 19, 892–898. [Google Scholar] [CrossRef] [Green Version]
  126. Ji, H.; Wan, L.; Zhang, Q.; Chen, M.; Zhao, X. The effect of omentin-1 on the proliferation and apoptosis of colon cancer stem cells and the potential mechanism. J. BUON 2019, 24, 91–98. [Google Scholar]
  127. Arjmand, M.H.; Moradi, A.; Akbari, A.; Mehrad-Majd, H. Clinical significance of circulating omentin levels in various malignant tumors: Evidence from a systematic review and meta-analysis. Cytokine 2020, 125, 154869. [Google Scholar] [CrossRef]
  128. Aleksandrova, K.; di Giuseppe, R.; Isermann, B.; Biemann, R.; Schulze, M.; Wittenbecher, C.; Fritsche, A.; Lehmann, R.; Menzel, J.; Weikert, C.; et al. Circulating Omentin as a Novel Biomarker for Colorectal Cancer Risk: Data from the EPIC-Potsdam Cohort Study. Cancer Res. 2016, 76, 3862–3871. [Google Scholar] [CrossRef] [Green Version]
  129. Zhou, L.; He, W.; Wang, W.; Zhou, D. Altered circulating levels of adipokine omentin-1 in patients with prostate cancer. Onco Targets Ther. 2019, 12, 3313–3319. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  130. Parida, S.; Siddharth, S.; Sharma, D. Role of Omentin in Obesity Paradox in Lung Cancer. Cancers 2021, 13, 275. [Google Scholar] [CrossRef]
  131. Tahmasebpour, N.; Hosseinpour Feizi, M.A.; Ziamajidi, N.; Pouladi, N.; Montazeri, V.; Farhadian, M.; Abbasalipourkabir, R. Association of Omentin-1 with Oxidative Stress and Clinical Significances in Patients with Breast Cancer. Adv. Pharm. Bull. 2020, 10, 106–113. [Google Scholar] [CrossRef]
  132. Shen, X.D.; Zhang, L.; Che, H.; Zhang, Y.Y.; Yang, C.; Zhou, J.; Liang, C.Z. Circulating levels of adipocytokine omentin-1 in patients with renal cell cancer. Cytokine 2016, 77, 50–55. [Google Scholar] [CrossRef]
  133. Christodoulatos, G.S.; Antonakos, G.; Karampela, I.; Psallida, S.; Stratigou, T.; Vallianou, N.; Lekka, A.; Marinou, I.; Vogiatzakis, E.; Kokoris, S.; et al. Circulating Omentin-1 as a Biomarker at the Intersection of Postmenopausal Breast Cancer Occurrence and Cardiometabolic Risk: An Observational Cross-Sectional Study. Biomolecules 2021, 11, 1609. [Google Scholar] [CrossRef]
  134. Wysocka, E.; Cofta, S.; Dzięgielewska, S.; Goździk, J.; Torliński, L.; Batura-Gabryel, H. Adipocytokines in sleep apnea syndrome. Eur. J. Med. Res. 2009, 14, 255–258. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  135. Sánchez-de-la-Torre, M.; Barceló, A.; Piérola, J.; de la Peña, M.; Valls, J.; Barbé, F. Impact of obstructive sleep apnea on the 24-h metabolic hormone profile. Sleep Med. 2014, 15, 625–630. [Google Scholar] [CrossRef] [PubMed]
  136. Pamuk, A.E.; Süslü, A.E.; Yalçınkaya, A.; Öztaş, Y.E.; Pamuk, G.; Özer, S.; Önerci, M. The serum leptin level in non-obese patients with obstructive sleep apnea. Auris Nasus Larynx 2018, 45, 796–800. [Google Scholar] [CrossRef] [PubMed]
  137. Li, X.; He, J. The Association Between Serum/Plasma Leptin Levels and Obstructive Sleep Apnea Syndrome: A Meta-Analysis and Meta-Regression. Front. Endocrinol. 2021, 12, 696418. [Google Scholar] [CrossRef]
  138. Sweatt, S.K.; Gower, B.A.; Chieh, A.Y.; Liu, Y.; Li, L. Sleep quality is differentially related to adiposity in adults. Psychoneuroendocrinology 2018, 98, 46–51. [Google Scholar] [CrossRef]
  139. Hayes, A.L.; Xu, F.; Babineau, D.; Patel, S.R. Sleep duration and circulating adipokine levels. Sleep 2011, 34, 147–152. [Google Scholar] [CrossRef] [Green Version]
  140. Modzelewska, P.; Chludzińska, S.; Lewko, J.; Reszeć, J. The influence of leptin on the process of carcinogenesis. Contemp. Oncol. 2019, 23, 63–68. [Google Scholar] [CrossRef]
  141. Wu, M.H.; Chou, Y.C.; Chou, W.Y.; Hsu, G.C.; Chu, C.H.; Yu, C.P.; Yu, J.C.; Sun, C.A. Circulating levels of leptin, adiposity and breast cancer risk. Br. J. Cancer 2009, 100, 578–582. [Google Scholar] [CrossRef] [Green Version]
  142. Wang, P.P.; He, X.Y.; Wang, R.; Wang, Z.; Wang, Y.G. High leptin level is an independent risk factor of endometrial cancer: A meta-analysis. Cell Physiol. Biochem. 2014, 34, 1477–1484. [Google Scholar] [CrossRef] [Green Version]
  143. Vuletic, M.S.; Milosevic, V.S.; Jancic, S.A.; Zujovic, J.T.; Krstic, M.S.; Vukmirovic, F.C. Clinical significance of Leptin receptor (LEPR) and Endoglin (CD105) expressions in colorectal adenocarcinoma. J. BUON 2019, 24, 2448–2457. [Google Scholar]
  144. Vokuda, R.S.; BH, S.; Madhugiri, V.S.; Velusamy, S.K.; Verma, S.K. The Expression of Leptin and Its Receptor During Tumorigenesis of Diffuse Gliomas such as Astrocytoma and Oligodendroglioma- Grade II, III and IV (NOS). Asian Pac. J. Cancer Prev. 2019, 20, 479–485. [Google Scholar] [CrossRef]
  145. Rutkowski, R.; Reszec, J.; Hermanowicz, A.; Chrzanowski, R.; Lyson, T.; Mariak, Z.; Chyczewski, L. Correlation of leptin receptor expression with BMI in differential grades of human meningiomas. Oncol. Lett. 2016, 11, 2515–2519. [Google Scholar] [CrossRef] [PubMed]
  146. Słomian, G.J.; Nowak, D.; Buczkowska, M.; Głogowska-Gruszka, A.; Słomian, S.P.; Roczniak, W.; Janyga, S.; Nowak, P. The role of adiponectin and leptin in the treatment of ovarian cancer patients. Endokrynol. Pol. 2019, 70, 57–63. [Google Scholar] [CrossRef] [Green Version]
  147. Zurita-Cruz, J.; Villasis-Keever, M.; Manuel-Apolinar, L.; Damasio-Santana, L.; Wakida-Kusunoki, G.H.; Padilla-Rojas, M.; Maldonado-Rivera, C. Resistin/Uric Acid Index as a Prognostic Factor in Adolescents with Obesity after Lifestyle Intervention. J. Pediatr. 2020, 219, 38–42.e1. [Google Scholar] [CrossRef]
  148. Mashaqi, S.; Badr, M.S. The Impact of Obstructive Sleep Apnea and Positive Airway Pressure Therapy on Metabolic Peptides Regulating Appetite, Food Intake, Energy Homeostasis, and Systemic Inflammation: A Literature Review. J. Clin. Sleep Med. 2019, 15, 1037–1050. [Google Scholar] [CrossRef]
  149. Dalamaga, M. Resistin as a biomarker linking obesity and inflammation to cancer: Potential clinical perspectives. Biomark. Med. 2014, 8, 107–118. [Google Scholar] [CrossRef]
  150. Cabia, B.; Andrade, S.; Carreira, M.C.; Casanueva, F.F.; Crujeiras, A.B. A role for novel adipose tissue-secreted factors in obesity-related carcinogenesis. Obes. Rev. 2016, 17, 361–376. [Google Scholar] [CrossRef] [PubMed]
  151. Gong, W.J.; Zheng, W.; Xiao, L.; Tan, L.M.; Song, J.; Li, X.P.; Xiao, D.; Cui, J.J.; Li, X.; Zhou, H.H.; et al. Circulating resistin levels and obesity-related cancer risk: A meta-analysis. Oncotarget 2016, 7, 57694–57704. [Google Scholar] [CrossRef] [Green Version]
  152. Mihajlovic, M.; Ninic, A.; Sopic, M.; Miljkovic, M.; Stefanovic, A.; Vekic, J.; Spasojevic-Kalimanovska, V.; Zeljkovic, D.; Trifunovic, B.; Stjepanovic, Z.; et al. Association among resistin, adenylate cyclase-associated protein 1 and high-density lipoprotein cholesterol in patients with colorectal cancer: A multi-marker approach, as a hallmark of innovative predictive, preventive, and personalized medicine. EPMA J. 2019, 10, 307–316. [Google Scholar] [CrossRef] [Green Version]
  153. Yoon, Y.S.; Kwon, A.R.; Lee, Y.K.; Oh, S.W. Circulating adipokines and risk of obesity related cancers: A systematic review and meta-analysis. Obes. Res. Clin. Pract. 2019, 13, 329–339. [Google Scholar] [CrossRef] [PubMed]
  154. Li, J.; Li, Q.; Zhu, Y.C.; Wang, Y.K.; Gao, C.P.; Li, X.Y.; Ji, T.; Bai, S.J. Association of vaspin rs2236242 gene variants with type 2 diabetes and obesity in a Chinese population: A prospective, single-center study. J. Cell Physiol. 2019, 234, 16097–16101. [Google Scholar] [CrossRef] [PubMed]
  155. Xu, T.; Lin, Y.; Sun, S.; Zhang, Q. Changes in four plasma adipokines before and after sleep in OSAS patients. Clin. Respir. J. 2017, 11, 968–974. [Google Scholar] [CrossRef]
  156. Pazgan-Simon, M.; Kukla, M.; Zuwała-Jagiełło, J.; Derra, A.; Bator, M.; Menżyk, T.; Lekstan, A.; Grzebyk, E.; Simon, K. Serum visfatin and vaspin levels in hepatocellular carcinoma (HCC). PLoS ONE 2020, 15, e0227459. [Google Scholar] [CrossRef] [Green Version]
  157. Erdogan, S.; Sezer, S.; Baser, E.; Gun-Eryilmaz, O.; Gungor, T.; Uysal, S.; Yilmaz, F.M. Evaluating vaspin and adiponectin in postmenopausal women with endometrial cancer. Endocr. Relat. Cancer. 2013, 20, 669–675. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  158. Siegrist, M.; Heitkamp, M.; Braun, I.; Vogg, N.; Haller, B.; Langhof, H.; Koenig, W.; Halle, M. Changes of omentin-1 and chemerin during 4 weeks of lifestyle intervention and 1 year follow-up in children with obesity. Clin. Nutr. 2021, 40, 5648–5654. [Google Scholar] [CrossRef]
  159. Würfel, M.; Breitfeld, J.; Gebhard, C.; Scholz, M.; Baber, R.; Riedel-Heller, S.G.; Blüher, M.; Stumvoll, M.; Kovacs, P.; Tönjes, A. Interplay between adipose tissue secreted proteins, eating behavior and obesity. Eur. J. Nutr. 2022, 61, 885–899. [Google Scholar] [CrossRef]
  160. Feng, X.; Li, P.; Zhou, C.; Jia, X.; Kang, J. Elevated levels of serum chemerin in patients with obstructive sleep apnea syndrome. Biomarkers 2012, 17, 248–253. [Google Scholar] [CrossRef]
  161. Treeck, O.; Buechler, C.; Ortmann, O. Chemerin and Cancer. Int. J. Mol. Sci. 2019, 20, 3750. [Google Scholar] [CrossRef] [Green Version]
  162. Ramanjaneya, M.; Chen, J.; Brown, J.E.; Tripathi, G.; Hallschmid, M.; Patel, S.; Kern, W.; Hillhouse, E.W.; Lehnert, H.; Tan, B.K.; et al. Identification of nesfatin-1 in human and murine adipose tissue: A novel depot-specific adipokine with increased levels in obesity. Endocrinology 2010, 151, 3169–3180. [Google Scholar] [CrossRef] [Green Version]
  163. Dore, R.; Levata, L.; Lehnert, H.; Schulz, C. Nesfatin-1: Functions and physiology of a novel regulatory peptide. J. Endocrinol. 2017, 232, R45–R65. [Google Scholar] [CrossRef] [Green Version]
  164. Caroleo, M.; Carbone, E.A.; Primerano, A.; Foti, D.; Brunetti, A.; Segura-Garcia, C. The role of hormonal, metabolic and inflammatory biomarkers on sleep and appetite in drug free patients with major depression: A systematic review. J. Affect. Disord. 2019, 250, 249–259. [Google Scholar] [CrossRef] [PubMed]
  165. Araz, O.; Yilmazel Ucar, E.; Dorman, E.; Bayraktutan, Z.; Yayla, M.; Yilmaz, N.; Acemoglu, H.; Halici, Z.; Akgun, M. Is There a Relationship between Obstructive Sleep Apnea Syndrome Severity and Nesfatin-1? Respiration 2015, 90, 105–110. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  166. Shen, P.; Han, Y.; Cai, B.; Wang, Y. Decreased levels of serum nesfatin-1in patients with obstructive sleep apnea syndrome. Sleep Breath. 2015, 19, 515–522. [Google Scholar] [CrossRef]
  167. Batura-Gabryel, H.; Bromińska, B.; Sawicka-Gutaj, N.; Cyrańska-Chyrek, E.; Kuźnar-Kamińska, B.; Winiarska, H.; Kostrzewska, M.; Zybek-Kocik, A.; Hernik, A.; Wrotkowska, E.; et al. Does nesfatin-1 influence the hypothalamic-pituitary-gonadal axis in adult males with obstructive sleep apnoea? Sci. Rep. 2019, 9, 11289. [Google Scholar] [CrossRef] [Green Version]
  168. Kan, J.Y.; Yen, M.C.; Wang, J.Y.; Wu, D.C.; Chiu, Y.J.; Ho, Y.W.; Kuo, P.L. Nesfatin-1/Nucleobindin-2 enhances cell migration, invasion, and epithelial-mesenchymal transition via LKB1/AMPK/TORC1/ZEB1 pathways in colon cancer. Oncotarget 2016, 7, 31336–31349. [Google Scholar] [CrossRef] [Green Version]
  169. Wang, X.Q.; Zheng, Y.; Fang, P.F.; Song, X.B. Nesfatin-1 is a potential diagnostic biomarker for gastric cancer. Oncol. Lett. 2020, 19, 1577–1583. [Google Scholar] [CrossRef] [Green Version]
  170. Cetinkaya, H.; Karagöz, B.; Bilgi, O.; Ozgün, A.; Tunçel, T.; Emirzeoğlu, L.; Top, C.; Kandemir, E.G. Nesfatin-1 in advanced lung cancer patients with weight loss. Regul. Pept. 2013, 181, 1–3. [Google Scholar] [CrossRef] [PubMed]
  171. Berndt, J.; Klöting, N.; Kralisch, S.; Kovacs, P.; Fasshauer, M.; Schön, M.R.; Stumvoll, M.; Blüher, M. Plasma visfatin concentrations and fat depot-specific mRNA expression in humans. Diabetes 2005, 54, 2911–2916. [Google Scholar] [CrossRef] [Green Version]
  172. Szymanska, A.; Platek, A.E.; Sierdzinski, J.; Szymanski, F.M. Visfatin as a predictor of obstructive sleep apnea in atrial fibrillation patients. Sleep Breath. 2020, 24, 1215–1218. [Google Scholar] [CrossRef] [Green Version]
  173. Trakada, G.; Steiropoulos, P.; Nena, E.; Gkioka, T.; Kouliatsis, G.; Pataka, A.; Sotiriou, I.; Anevlavis, S.; Papanas, N.; Bouros, D. Plasma visfatin levels in severe obstructive sleep apnea-hypopnea syndrome. Sleep Breath. 2009, 13, 349–355. [Google Scholar] [CrossRef]
  174. Dahmen, N.; Manderscheid, N.; Helfrich, J.; Musholt, P.B.; Forst, T.; Pfützner, A.; Engel, A. Elevated peripheral visfatin levels in narcoleptic patients. PLoS ONE 2008, 3, e2980. [Google Scholar] [CrossRef] [Green Version]
  175. Mohammadi, M.; Moradi, A.; Farhadi, J.; Akbari, A.; Pourmandi, S.; Mehrad-Majd, H. Prognostic value of visfatin in various human malignancies: A systematic review and meta-analysis. Cytokine 2020, 127, 154964. [Google Scholar] [CrossRef] [PubMed]
  176. Montecucco, F.; Liberale, L.; Carbone, F. Novel cardiovascular risk biomarkers in metabolic syndrome. Biomark. Med. 2019, 13, 1331–1334. [Google Scholar] [CrossRef] [Green Version]
  177. Sarac, F.; Basoglu, O.K.; Gunduz, C.; Bayrak, H.; Biray Avci, C.; Akcicek, F. Association of osteopontin and tumor necrosis factor-α levels with insulin resistance in obese patients with obstructive sleep apnea syndrome. J. Endocrinol. Invest. 2011, 34, 528–533. [Google Scholar] [CrossRef] [PubMed]
  178. Kariya, Y.; Kariya, Y. Osteopontin in Cancer: Mechanisms and Therapeutic Targets. Int. J. Transl. Med. 2022, 2, 419–447. [Google Scholar] [CrossRef]
  179. Zhang, N.; Li, F.; Gao, J.; Zhang, S.; Wang, Q. Osteopontin accelerates the development and metastasis of bladder cancer via activating JAK1/STAT1 pathway. Genes Genomics 2020, 42, 467–475. [Google Scholar] [CrossRef]
  180. Lin, Z.; Süsskind, D. Evaluation of a Three-Marker Panel for the Detection of Uveal Melanoma Metastases: A Single-Center Retrospective Analysis. Cancers 2021, 13, 2464. [Google Scholar] [CrossRef] [PubMed]
  181. Kariya, Y.; Oyama, M.; Kariya, Y.; Hashimoto, Y. Phosphorylated Osteopontin Secreted from Cancer Cells Induces Cancer Cell Motility. Biomolecules 2021, 11, 1323. [Google Scholar] [CrossRef] [PubMed]
  182. Wysocka, M.B.; Pietraszek-Gremplewicz, K.; Nowak, D. The Role of Apelin in Cardiovascular Diseases, Obesity and Cancer. Front. Physiol. 2018, 9, 557. [Google Scholar] [CrossRef]
  183. Henley, D.E.; Buchanan, F.; Gibson, R.; Douthwaite, J.A.; Wood, S.A.; Woltersdorf, W.W.; Catterall, J.R.; Lightman, S.L. Plasma apelin levels in obstructive sleep apnea and the effect of continuous positive airway pressure therapy. J. Endocrinol. 2009, 203, 181–188. [Google Scholar] [CrossRef] [PubMed]
  184. Zirlik, S.; Hauck, T.; Fuchs, F.S.; Neurath, M.F.; Konturek, P.C.; Harsch, I.A. Leptin, obestatin and apelin levels in patients with obstructive sleep apnoea syndrome. Med. Sci. Monit. 2011, 17, CR159–CR164. [Google Scholar] [CrossRef] [Green Version]
  185. Song, F.; Zou, J.; Song, Z.; Xu, H.; Qian, Y.; Zhu, H.; Liu, S.; Guan, J.; Chen, J.; Yi, H. Association of Adipocytokines with Carotid Intima Media Thickness and Arterial Stiffness in Obstructive Sleep Apnea Patients. Front. Endocrinol. 2020, 11, 177. [Google Scholar] [CrossRef]
  186. Masoumi, J.; Jafarzadeh, A.; Khorramdelazad, H.; Abbasloui, M.; Abdolalizadeh, J.; Jamali, N. Role of Apelin/APJ axis in cancer development and progression. Adv. Med. Sci. 2020, 65, 202–213. [Google Scholar] [CrossRef] [PubMed]
  187. Lacquaniti, A.; Altavilla, G.; Picone, A.; Donato, V.; Chirico, V.; Mondello, P.; Aloisi, C.; Marabello, G.; Loddo, S.; Buemi, A.; et al. Apelin beyond kidney failure and hyponatremia: A useful biomarker for cancer disease progression evaluation. Clin. Exp. Med. 2015, 15, 97–105. [Google Scholar] [CrossRef]
  188. Olsen, T.; Blomhoff, R. Retinol, Retinoic Acid, and Retinol-Binding Protein 4 are Differentially Associated with Cardiovascular Disease, Type 2 Diabetes, and Obesity: An Overview of Human Studies. Adv. Nutr. 2020, 11, 644–666. [Google Scholar] [CrossRef]
  189. Makino, S.; Fujiwara, M.; Suzukawa, K.; Handa, H.; Fujie, T.; Ohtaka, Y.; Komatsu, Y.; Aoki, Y.; Maruyama, H.; Terada, Y.; et al. Visceral obesity is associated with the metabolic syndrome and elevated plasma retinol binding protein-4 level in obstructive sleep apnea syndrome. Horm. Metab. Res. 2009, 41, 221–226. [Google Scholar] [CrossRef]
  190. Nena, E.; Steiropoulos, P.; Tzouvelekis, A.; Tsara, V.; Hatzizisi, O.; Kyriazis, G.; Froudarakis, M.; Trakada, G.; Papanas, N.; Bouros, D. Reduction of serum retinol-binding protein-4 levels in nondiabetic obstructive sleep apnea patients under continuous positive airway pressure treatment. Respiration 2010, 80, 517–523. [Google Scholar] [CrossRef]
  191. Du, M.; Liu, J.; Han, N.; Zhao, Z.; Luo, S.; Wang, H. Exploring the mediating role of serum retinol-binding protein 4 in the relationship between sleep quality and insulin resistance in pregnant women. Diabetes Res. Clin. Pract. 2021, 176, 108866. [Google Scholar] [CrossRef] [PubMed]
  192. Papiernik, D.; Urbaniak, A.; Kłopotowska, D.; Nasulewicz-Goldeman, A.; Ekiert, M.; Nowak, M.; Jarosz, J.; Cuprych, M.; Strzykalska, A.; Ugorski, M.; et al. Retinol-Binding Protein 4 Accelerates Metastatic Spread and Increases Impairment of Blood Flow in Mouse Mammary Gland Tumors. Cancers 2020, 12, 623. [Google Scholar] [CrossRef] [Green Version]
  193. Weigert, J.; Neumeier, M.; Wanninger, J.; Bauer, S.; Farkas, S.; Scherer, M.N.; Schnitzbauer, A.; Schäffler, A.; Aslanidis, C.; Schölmerich, J.; et al. Serum galectin-3 is elevated in obesity and negatively correlates with glycosylated hemoglobin in type 2 diabetes. J. Clin. Endocrinol. Metab. 2010, 95, 1404–1411. [Google Scholar] [CrossRef] [Green Version]
  194. Pusuroglu, H.; Somuncu, U.; Bolat, I.; Akgul, O.; Ornek, V.; Yıldırım, H.A.; Akkaya, E.; Karakurt, H.; Yıldırım, A.; Savaş, A.U. Galectin-3 is associated with coronary plaque burden and obstructive sleep apnoea syndrome severity. Kardiol. Pol. 2017, 75, 351–359. [Google Scholar] [CrossRef] [PubMed]
  195. Singh, M.; Hanis, C.L.; Redline, S.; Ballantyne, C.M.; Hamzeh, I.; Aguilar, D. Sleep apnea and galectin-3: Possible sex-specific relationship. Sleep Breath. 2019, 23, 1107–1114. [Google Scholar] [CrossRef]
  196. Slouka, D.; Kucera, R.; Gal, B.; Betka, J.; Skalova, A. Biomarkers—A possibility for monitoring of obstructive sleep apnea syndrome. Neuro Endocrinol. Lett. 2019, 40, 85–92. [Google Scholar]
  197. Souza, D.S.; Macheroni, C.; Pereira, G.J.S.; Vicente, C.M.; Porto, C.S. Molecular regulation of prostate cancer by Galectin-3 and estrogen receptor. Front. Endocrinol. 2023, 14, 1124111. [Google Scholar] [CrossRef]
  198. Mohammed, N.B.B.; Antonopoulos, A.; Dell, A.; Haslam, S.M.; Dimitroff, C.J. The pleiotropic role of galectin-3 in melanoma progression: Unraveling the enigma. Adv. Cancer Res. 2023, 157, 157–193. [Google Scholar] [CrossRef]
  199. Aureli, A.; Del Cornò, M.; Marziani, B.; Gessani, S.; Conti, L. Highlights on the Role of Galectin-3 in Colorectal Cancer and the Preventive/Therapeutic Potential of Food-Derived Inhibitors. Cancers 2022, 15, 52. [Google Scholar] [CrossRef] [PubMed]
  200. Fontvieille, E.; His, M.; Biessy, C.; Navionis, A.S.; Torres-Mejía, G.; Ángeles-Llerenas, A.; Alvarado-Cabrero, I.; Sánchez, G.I.; Navarro, E.; Cortes, Y.R.; et al. Inflammatory biomarkers and risk of breast cancer among young women in Latin America: A case-control study. BMC Cancer 2022, 22, 877. [Google Scholar] [CrossRef]
  201. Aljafary, M.A.; Al-Suhaimi, E.A. Adiponectin System (Rescue Hormone): The Missing Link between Metabolic and Cardiovascular Diseases. Pharmaceutics 2022, 14, 1430. [Google Scholar] [CrossRef] [PubMed]
  202. Ren, Y.; Zhao, H.; Yin, C.; Lan, X.; Wu, L.; Du, X.; Griffiths, H.R.; Gao, D. Adipokines, Hepatokines and Myokines: Focus on Their Role and Molecular Mechanisms in Adipose Tissue Inflammation. Front. Endocrinol. 2022, 13, 873699. [Google Scholar] [CrossRef]
  203. Spyrou, N.; Avgerinos, K.I.; Mantzoros, C.S.; Dalamaga, M. Classic and Novel Adipocytokines at the Intersection of Obesity and Cancer: Diagnostic and Therapeutic Strategies. Curr. Obes. Rep. 2018, 7, 260–275. [Google Scholar] [CrossRef] [PubMed]
  204. Yasir, M.; Senthilkumar, G.P.; Jayashree, K.; Ramesh Babu, K.; Vadivelan, M.; Palanivel, C. Association of serum omentin-1, apelin and chemerin concentrations with the presence and severity of diabetic retinopathy in type 2 diabetes mellitus patients. Arch. Physiol. Biochem. 2022, 128, 313–320. [Google Scholar] [CrossRef]
  205. Tezcan, N.; Özdemir-Kumral, Z.N.; Özkan Yenal, N.; Çilingir-Kaya, Ö.T.; Virlan, A.T.; Özbeyli, D.; Çetinel, Ş.; Yeğen, B.Ç.; Koç, M. Nesfatin-1 treatment preserves antioxidant status and attenuates renal fibrosis in rats with unilateral ureteral obstruction. Nephrol. Dial. Transplant. 2022, 37, 1238–1248. [Google Scholar] [CrossRef]
  206. Chen, K.J.; Hsieh, M.H.; Lin, Y.Y.; Chen, M.Y.; Lien, M.Y.; Yang, S.F.; Tang, C.H. Visfatin Polymorphisms, Lifestyle Risk Factors and Risk of Oral Squamous Cell Carcinoma in a Cohort of Taiwanese Males. Int. J. Med. Sci. 2022, 19, 762–768. [Google Scholar] [CrossRef] [PubMed]
  207. Yang, H.W.; Park, J.H.; Jo, M.S.; Shin, J.M.; Kim, D.W.; Park, I.H. Eosinophil-Derived Osteopontin Induces the Expression of Pro-Inflammatory Mediators and Stimulates Extracellular Matrix Production in Nasal Fibroblasts: The Role of Osteopontin in Eosinophilic Chronic Rhinosinusitis. Front. Immunol. 2022, 13, 777928. [Google Scholar] [CrossRef]
  208. Kadoglou, N.P.E.; Velidakis, N.; Khattab, E.; Kassimis, G.; Patsourakos, N. The interplay between statins and adipokines. Is this another explanation of statins’ ‘pleiotropic’ effects? Cytokine 2021, 148, 155698. [Google Scholar] [CrossRef]
  209. Chauhan, A.; Singhal, A.; Goyal, P.; Taneja, A. Serum Retinol Binding Protein-4 Levels in Prediabetics—Novel Biomarker of Insulin Resistance and Atherosclerosis. J. Assoc. Physicians India 2021, 69, 11–12. [Google Scholar]
  210. Li, M.; Tian, M.; Jiang, X.; Liu, Y.; Wang, Y.; Li, Y. Inhibition of galectin-3 ameliorates high-glucose-induced oxidative stress and inflammation in ARPE-19 cells. Cutan. Ocul. Toxicol. 2022, 41, 179–186. [Google Scholar] [CrossRef]
  211. Martínez-Bosch, N.; Rodriguez-Vida, A.; Juanpere, N.; Lloreta, J.; Rovira, A.; Albanell, J.; Bellmunt, J.; Navarro, P. Galectins in prostate and bladder cancer: Tumorigenic roles and clinical opportunities. Nat. Rev. Urol. 2019, 16, 433–445. [Google Scholar] [CrossRef] [PubMed]
  212. Manero-Rupérez, N.; Martínez-Bosch, N.; Barranco, L.E.; Visa, L.; Navarro, P. The Galectin Family as Molecular Targets: Hopes for Defeating Pancreatic Cancer. Cells 2020, 9, 689. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  213. Alves, H.R.; Lomba, G.S.B.; Gonçalves-de-Albuquerque, C.F.; Burth, P. Irisin, Exercise, and COVID-19. Front. Endocrinol. 2022, 13, 879066. [Google Scholar] [CrossRef]
  214. Mazur-Bialy, A.I.; Pocheć, E.; Zarawski, M. Anti-Inflammatory Properties of Irisin, Mediator of Physical Activity, Are Connected with TLR4/MyD88 Signaling Pathway Activation. Int. J. Mol. Sci. 2017, 18, 701. [Google Scholar] [CrossRef] [Green Version]
  215. Liu, W.; Deng, Y.; Li, Z.; Chen, Y.; Zhu, X.; Tan, X.; Cao, G. Cancer Evo-Dev: A Theory of Inflammation-Induced Oncogenesis. Front. Immunol. 2021, 12, 768098. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Possible association of sleep disorders, obesity and sarcopenia with cancerogenesis.
Figure 1. Possible association of sleep disorders, obesity and sarcopenia with cancerogenesis.
Jcm 12 02655 g001
Table 1. The associations between adipokines/adipomyokines and their pro-inflammatory properties with obesity, sleep disorders and cancer.
Table 1. The associations between adipokines/adipomyokines and their pro-inflammatory properties with obesity, sleep disorders and cancer.
Adipokines/
Adipomyokines
Level in ObesityLevel in Sleep DisordersPro-Inflammatory PropertiesRole in Cancerogenesis
adiponectinYes [200] and no [201]Anti-cancerous [119], decreased in many cancers [120,121,122].
omentinNo [202]Anti-cancerous or pro-cancerous [122,126], increased in some cancers [127,129], decreased in some cancers [130,131,132]
leptinYes [202]Pro-cancerous [140], increased in some cancers [141,142,143,144,145], controversial in some cancers [146,203]
resistin or or = Yes [202]Increased in some cancers [149,150,151,152] or not changed in some cancers [153]
vaspinNo [202]Increased in some cancers [156], decreased in some cancers [157]
chemerinYes [202] and no [204]Anti-cancerous or pro-cancerous [161], increased in various cancers [203]
nesfatin or =No [205]Increased in some cancers [168,169], decreased or not changed in some cancers [170]
visfatin Yes [206]Increased in various cancers [175]
osteopontinYes [207]Increased in various cancers [179,203]
apelin or =No [208]Pro-cancerous [186], increased in various cancers [187]
RBP-4 or =Yes [209]Increased in various cancers [192]
galectin-3 or or =Yes [210]Increased in various cancers [211,212]
irisin or or No [213]Anti-cancerous [89,96,97,98,99] or without pro-/anti-cancerous action [100], increased in some cancers [108,109], decreased in some cancers [102,105,106,107]
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Brzecka, A.; Martynowicz, H.; Daroszewski, C.; Majchrzak, M.; Ejma, M.; Misiuk-Hojło, M.; Somasundaram, S.G.; Kirkland, C.E.; Kosacka, M. The Modulation of Adipokines, Adipomyokines, and Sleep Disorders on Carcinogenesis. J. Clin. Med. 2023, 12, 2655. https://doi.org/10.3390/jcm12072655

AMA Style

Brzecka A, Martynowicz H, Daroszewski C, Majchrzak M, Ejma M, Misiuk-Hojło M, Somasundaram SG, Kirkland CE, Kosacka M. The Modulation of Adipokines, Adipomyokines, and Sleep Disorders on Carcinogenesis. Journal of Clinical Medicine. 2023; 12(7):2655. https://doi.org/10.3390/jcm12072655

Chicago/Turabian Style

Brzecka, Anna, Helena Martynowicz, Cyryl Daroszewski, Maciej Majchrzak, Maria Ejma, Marta Misiuk-Hojło, Siva G. Somasundaram, Cecil E. Kirkland, and Monika Kosacka. 2023. "The Modulation of Adipokines, Adipomyokines, and Sleep Disorders on Carcinogenesis" Journal of Clinical Medicine 12, no. 7: 2655. https://doi.org/10.3390/jcm12072655

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop