Skip to main content

Excessive fat expenditure in cachexia is associated with dysregulated circadian rhythm: a review

Abstract

Cachexia is a progressive metabolic disorder characterized by the excessive depletion of adipose tissue. This hypermetabolic condition has catastrophic impacts on the survival and quality of life for patients suffering from critical illness. However, efficient therapies to prevent adipose expenditure have not been discovered. It has been established that the circadian clock plays an important role in modulating fat metabolic processes. Recently, an increasing number of studies had provided evidence showing that disrupted circadian rhythm leads to insulin resistance and obesity; however, studies analyzing the relationship between circadian misalignment and adipose tissue expenditure in cachexia are scarce. In the present review, we cover the involvement of the circadian clocks in the regulation of adipogenesis, lipid metabolism and thermogenesis as well as inflammation in white and brown adipose tissue. According to the present review, we conclude that circadian clock disruption is associated with lipid metabolism imbalance and elevated adipose tissue inflammation. Moreover, under cachexia conditions, lipid synthesis and storage processes lost rhythm and decreased, while lipolysis and thermogenesis activities remained high for 24 h. Therefore, disordered circadian clock may be responsible for fat expenditure in cachexia by adversely influencing lipid synthesis/ storage/lipolysis/utilization. Further study needs to be performed to explore the direct interaction between circadian clock and fat expenditure in cachexia, it will likely provide potential efficient drugs for the treatment of fat expenditure in cachexia.

Introduction

The circadian clock is a key homeostatic regulator that controls many physiological and behavioral responses [1]. The master clock located in the hypothalamus synchronizes multiple peripheral oscillators in most cells of the body to ensure the temporal coordination of behavior and metabolism. At the molecular level, the circadian clock involves transcriptional and translational feedback loops, in which the circadian locomotor output cycles kaput (CLOCK) and brain and muscle ARNT-like 1 protein (BMAL1) complex promotes the transcription of clock genes Period (Per1 and Per2) and cryptochrome (Cry1 and Cry2). The PER and CRY proteins accumulate in the cytoplasm, and then translocate to the nucleus in the evening to repress their own transcription by interacting with the CLOCK:BMAL1 heterodimer [2]. Additionally, the CLOCK:BMAL1 heterodimer promotes the expression of nuclear receptor reverse ERBα (REV-ERBα) and retinoic acid receptor-related orphan receptor isoforms (RORα and RORγ), which negatively and positively regulate Bmal1 expression, respectively, by binding to ROR response elements (ROREs) in the Bmal1 promoter [3,4,5].

Cachexia syndrome is a complex metabolic disorder that results in depletion of skeletal muscle associated, in some cases, with significant reduced fat mass [6, 7]. The hypermetabolic condition has catastrophic impacts on survival and quality of life for patients suffering from critical illness such as cancer, end-stage heart failure, chronic obstructive pulmonary diseases, and severe burn injury [8]. Currently, although augmenting protein intake, anti-inflammatory drugs are prescribed and select appetite stimulants and exercises are suggested as therapies for cachexia [9], the prevention and management of cachexia are still major challenges to clinicians and patients.

Study performed in gastrointestinal cancer patients have found that cachexia is characterized by preferential loss of adipose tissue and decreased fat cell volume, while muscle mass is less affected [10]. Moreover, adipose tissue expenditure may occur earlier than muscle loss [11, 12]. Cachexia manifested by adipose atrophy is a common and severe complication in critically ill patients admitted to the intensive care unit [13]. Available outcome data of critically ill patients have suggested that obese patients have improved outcomes as compared to the critically patients with healthy body weight, termed “obesity paradox” [14,15,16]. One explain for this phenomenon is that obese individuals have nutritional “reserves”, which provides substrate during critical illness [17]. The accelerated loss of adipose tissue predicts poorer survival, while maintaining adipose tissue mass may prevent muscle loss and improve patient quality of life and survival outcomes [18, 19].

Increased adipocyte lipolysis and activation of brown/beige adipocytes have been found in adipose tissue of patients with cancer cachexia [20]. Since adipose tissue plays an important role in maintaining energy homeostasis and secreting hormones and adipokines that modulate appetite and nutrient metabolism, alterations in adipose tissue mass can have significant effects on whole-body energy homeostasis [21]. It has been established that adipose tissue activity shows circadian rhythmicity and fat metabolic processes, including adipogenesis, lipid utilization/storage and thermogenesis are under controlled of circadian clock. Disrupted circadian rhythm leads to insulin resistance and metabolic inflammation, increases risk of obesity and type 2 diabetes [22]. These studies have led to increased interest in clock-related treatment strategies, such as “chronotherapeutics”, to mitigate metabolic disorders. Nevertheless, to date, studies analyzing the relationship between circadian misalignment and adipose tissue expenditure in cachexia have been lacking. The causal link between circadian misalignment and adipose wasting in cachexia is interesting to explore, which will provide potential treatment for cachexia.

Given the high number of excellent reviews summarizing impacts of circadian clock in background of obesity, the present review aimed to highlight understanding of the links between circadian clock disruption and adipose expenditure in condition of cachexia. In accordance with this purpose, studies published from 1980 to 2021 were reviewed. Basically, Google Academic, PubMed and Web of Science were used to scan scientific articles. Terms such as “circadian rhythm or circadian clock or biological clock or clock gene” and “lipolysis or lipogenic or energy metabolism or lipid metabolism or energy homeostasis” and “adipose tissue or fat tissue or brown adipose tissue or brown adipocyte” and “inflammation or cytokines” were used as key terms.

Impaired lipogenesis and storage and elevated lipolysis and lipid utilization contribute to fat expenditure in cachexia

Adipose tissue plays a vital role in whole-body energy homeostasis. In mammals, adipocyte presents in three distinct categories: white, brown and beige. White adipocytes have large unilocular lipid droplets and a small number of mitochondria. They synthesize and store triglycerides in fat droplets and break down triglycerides during periods of energy surplus or demand [23]. Additionally, they synthesize and secrete adipokines and hormones that modulate energy metabolism [24]. Compared with white adipocytes, brown adipocytes have more mitochondria and multilocular lipid droplets. Instead of storing lipids, they convert chemical energy into heat through activating uncoupling protein 1 (UCP-1) expressed in the internal membrane of mitochondria—a phenomenon phenotypically referred to as non-shivering thermogenesis [25]. Through this function, brown adipocytes play important roles in modulating body temperature. Additionally, a third type of inducible “brownlike”, UPC-1-expressing adipocytes have been identified within some white adipose depots. These adipocytes have been named beige. Although they are developmentally related to white adipose tissue, they exhibit functional plasticity in terms of their ability to function like brown adipocytes. Beige adipocytes are rich in mitochondria and express UCP-1. As their name suggests, although they are found in white adipose tissue, they have thermogenic capacity in responds to cold exposure, sympathetic stimuli and hormones to dissipate heat through UCP-1 [26]. In contrast, in the context of a high-fat diet, these beige adipocytes are converted into white adipocytes [27].

Exogenous free fatty acids (FFAs) derived from the diet and those synthesized via de novo lipogenesis in hepatocytes are transported into adipocytes by transport proteins such as cluster of differentiation 36 (CD36) or fatty acid transport protein1 (FATP1). To synthesize triglyceride, three FFAs are esterified to a glycerol backbone, and the triglyceride is processed through the endoplasmic reticulum for storage within the lipid droplet [28]. In response to nutritional conditions, adipocytes increase their size (hypertrophy) to store excessive triglyceride effectively in adipose tissue [29]. However, under conditions of cachexia, lipogenesis and lipid storage capacity are impaired [30]. Adipose tissue from tumor-bearing mice contained shrunken adipocytes that were heterogeneous in size [31]. Expression of key adipogenic transcription factors, such as CCAAT/enhancer binding protein α (C/EBPα), CCAAT/enhancer-binding protein beta (C/EBPβ), peroxisome proliferator-activated receptor γ (Pparγ), sterol regulatory element-binding protein-1c (SREBP-1c) and associated target genes involved in lipid synthesis pathways, including fatty acid synthase (Fas), acetyl-CoA carboxylase (Acc), stearoyl CoA desaturase 1 (Scd1), glycerol-3-phosphate acyl transferase (G3P) and diacylglycerol O-acyltransferase (Dgat2), was significantly decreased [32], leading to reduced lipid accumulation in adipocytes.

Cachexia caused by the hypermetabolic response is characterized by increased lipolytic activities in white adipocytes and thermogenesis in beige and brown adipocytes (Fig. 1). Catecholamines and inflammatory cytokines have been implicated as the primary mediators of this hypermetabolic response in diseases such as cancer and burns [33,34,35,36]. They activate their respective receptors on the surface of adipocytes, leading to cellular signaling that activates protein kinase A (PKA) and G (PKG). Then, adipose triglyceride lipase (ATGL), which is a PKA/PKG-activated lipase, initiates triacylglycerol (TAG) catabolism by selectively hydrolyzing the first ester bond, resulting in the formation of diacylglycerol (DAG). Hormone‐sensitive lipase (HSL) preferentially hydrolyzes DAG to monoacylglycerol (MAG), which is further converted to G by monoglyceride lipase (MGL) [37, 38]. This process produces large amounts of FFAs. Excessively increased lipolysis causes FFA efflux into non-adipose tissue, which leads to lipotoxicity in liver, skeleton muscle, pancreas and heart [39, 40]. In cancer patients, increased lipolysis was found to be related to elevated enzyme activities of ATGL and HSL [41]. In tumors-induced cachexia mice models, the increased expression of ATGL contributed to fat depletion and increased rates of lipolysis, while knockout of ATGL protected the tumor-bearing mice from cachexia and loss of fat mass [19]. Similarly, the activated PKA in early cancer induced lipolysis through up-regulation of HSL [42].

Fig. 1
figure 1

Increased lipolytic activities in white adipocytes and thermogenesis in beige and brown adipocytes contribute to excessive fat expenditure in cachexia. In cachexia, increased catecholamines, corticosteroids and inflammatory cytokines promote lipid lipolysis via active PKA/PKG signaling, causing the hydrolysis of lipid droplets as mediated by ATGL/HSL/Peprelin in white adipocytes. In addition, activated PKA/PKG signaling promotes the transcriptional regulation of white adipose tissue browning as characterized by the upregulated expression of Ucp-1, Pparγ and Pdrm16. This process produces large amounts of FFAs, which contribute to mitochondrial thermogenesis in brown and beige adipocytes, accelerating adipose expenditure. Additionally, excessive FFAs efflux into peripheral tissue or organs leads to ectopic fat deposition, such as that observed in atherosclerosis and hepatic steatosis, causing insulin resistance. PKA/PKG protein kinase A/G; ATGL lipases adipose triglyceride lipase; HSL hormone‐sensitive lipase; Ucp-1 uncoupling protein 1; Pparγ peroxisome proliferator-activated receptor gamma; Pdrm16 PR domain-containing 16; FFAs free fatty acids

Lipolysis-derived FFAs are imported into brown adipocytes and serve as substrate for mitochondrial thermogenesis. In addition, catecholamines and inflammatory cytokines also directly or indirectly, via the activation of PKA/PKG, promote the activity of transcriptional regulators of white adipose tissue browning, including PR domain-containing 16 (PDRM16), PPARγ, and PPARγ coactivator 1α (PGC1α), to induce UCP-1 expression and further induce thermogenesis [43] Recently, inducing “browning” within white adipose tissue has attracted considerable interest in the field of obesity since it shows beneficial metabolic effects by increasing energy expenditure and reducing adiposity. However, it seems to be deleterious in the context of hypermetabolic conditions such as severe burns, end-stage heart diseases and cancer [44,45,46]; under these conditions, excessive white adipose tissue browning accelerates lipid utilization, ultimately leading to the adipose wasting associated with cachexia [39]. In the colon-26 murine model of cachexia, genes of the fatty acid uptake enzyme lipoprotein lipase (Lpl) and the key lipid uptake and transporter protein CD36 and FFA oxidation Ppar were increased. Meanwhile, genes involving thermogenesis such as p38-MAPK, Pgc1α, Prdm16 and Ucp-1 were increased. It indicated that increased FFA oxidation and thermogenesis in brown adipose tissue in cancer-induced cachexia [42].

Taken together, as shown in Fig. 1, not only impaired lipogenesis and lipid-storage and increased lipolysis in white adipose tissue contribute to reduced adipose accumulation, but also enhanced thermogenesis induced by activating brown/beige adipose tissue aggravates excessive adipose expenditure in cachexia [47, 48].

Disrupted circadian rhythm is associated with impaired adipogenesis and lipid synthesis and enhanced lipolysis in white adipose tissue

Circadian clocks in adipose tissue are known to regulate adipogenesis and lipid synthesis. Dysfunctional circadian clocks are associated with abnormal adipogenesis and lipid synthesis. Circadian-disrupted mouse models and adipocyte cell models have been used to independently examine the effects of those aspects (Table 1). For example, ClockΔ19/Δ19 mice exhibited increased weight gain related to visceral fat accumulation, and developed hypercholesterolemia and hyperglycemia compared to wild-type littermate controls [49]. In 3T3-L1 preadipocytes, inhibiting Clock gene lead to advanced adipogenesis differentiation and lipid synthesis at a very early stage of differentiation [50]. It suggests that CLOCK inhibits adipogenesis and prevents fat accumulation. BMAL1 promotes adipose differentiation and lipogenesis. During the differentiation 3T3-L1 preadipocytes to adipocytes, BMAL1 is highly upregulated. Inhibiting Bmal1 expression in 3T3-L1 preadipocytes by introduction of siRNA prevented adipogenesis and decreased the expression of lipogenesis genes, such as PPARγ, adipocyte protein 2 (aP2) and C/EBP. Conversely, overexpression of Bmal1 in 3T3-L1 preadipocytes elevated lipid synthesis activity [51]. Similarly, Bmal1 overexpression in mouse liver elevated the mRNA levels of lipogenic enzymes and promoted lipogenesis, while Bmal1 deficiency in hepatocytes displayed decreased levels of de novo lipogenesis and lipogenic enzymes [52]. These findings suggest that BMAL1 regulates adipose differentiation and lipogenesis. The transcription of another clock gene, REV-ERBα has been demonstrated that promotes 3T3-L1 preadipocytes differentiation into mature adipocytes via activation of PPARγ [53]. Rev-erbα deficient mice exhibited lipid metabolism disorder, causing increased liver triglyceride and free fatty acid [54]. Treatment of diet-induced obese mice with a REV-ERB agonist reduced triglyceride and free fatty acid in mice [55]. Cry1 deficient mice (Cry − / −) were less likely to gain weight and displayed reduced fat accumulation on a high-lipid diet compared to wild-type littermates [56]. Moreover, knockdown of Cry1 significantly inhibited the expression of adipogenic markers and lipid droplet formation in 3T3-L1 preadipocytes under adipogenic induction [57]. The Per3 gene has been found to exhibit robust circadian oscillations in pre-adipocytes and deletion of Per3 gene significantly increased adipogenesis in vivo [58]. Taken together, it has been convincingly shown that clock genes are involved in adipogenesis by directly or indirectly regulating transcription factors involved in adipogenesis and lipogenesis processes, and disrupted clock genes expression lead to abnormal fat accumulation.

Table 1 Summary of chronodisruption-induced metabolic phenotype

In addition, circadian clock plays an important role in regulating the rate of lipolysis in white adipose tissue, where lipid is stored in large amounts in the form of triglycerides. To fulfill the day-dependent energy requirement in the human cycles of sleep/wake and fasting/feeding, lipolysis activity is partly mediated by circadian clock in adipoctye. Suzuki et al. [59] firstly observed a diurnal change in the lipolytic activity of isolated epididymal fat cells. Consistently, in humans under constant routine conditions, it has been demonstrated that blood levels of FFAs are directly under circadian control [60]. In obese and/or diabetic individuals, the diurnal rhythm of the lipolysis process is disrupted, as indicated by a study showing that, compared with healthy control individuals, expressions of core clock genes and genes of the canonical metabolic pathways involved in lipolysis lost rhythm in patients with type 2 diabetes [60]. At the molecular level, the key enzymes of lipolysis, including ATGL and HSL, are under the transcriptional control of CLOCK:BMAL1 [61]. In Clock∆19 and Bmal1−/− circadian mutant mice, the circadian variations of ATGL and HSL were disrupted; in addition, the circadian patterns of serum FFAs and glycerol concentrations and the lipolytic activity in the fat pads were abolished [61]. Bmal1−/− mice exhibited reduced capacity of fat storage in adipose tissue, resulting increased levels of circulating FFAs and triglycerides. The increased circulating FFAs level induced the formation of ectopic fat in the liver and skeletal muscle [62]. Therefore, circadian clocks are crucial factors in the regulation of balance between lipid storage and lipolysis.

As shown in Fig. 2, in the white adipose tissue of mice bearing C26 tumors, the expression of adipogenesis genes, including PPARγ and C/EBPα, and lipogenic genes Fas and Dgat decreased and lost rhythmicity, indicating decreased lipid accumulation in cachexia. Additionally, the protein levels of the lipolytic enzymes perilipin and ATGL were increased at two time points, indicating increased lipolysis during both the day and night cycles. In addition, cachexia resulted in loss of rhythmicity of Rev-erbα and Per2, and the expressions of Bmal1 and Cry1 increased [31]. However, the mechanistic interconnectivity between disrupted circadian clock and reduced lipid synthesis and increased lipolysis in cachexia warrants further investigation.

Fig. 2
figure 2

Dysregulated circadian rhythm is involved in lipid accumulation/lipolysis/utilization and thermogenesis in white and brown adipocytes by modulating enzymes involved in fat metabolism. The lipolysis and lipid storage of white adipocytes and thermogenesis of brown adipocytes are mediated by diurnal oscillations controlled by circadian rhythm, while dysregulated circadian rhythm may disrupt the circadian pattern in cachexia. Compared to that in the health condition, in the cachexia condition, the expression of adipogenesis genes, including Pparγ and C/EBPα, and the lipogenic genes Fas and Dgat decreased and lost rhythmicity in white adipocytes, indicating decreased lipid accumulation. In addition, the expression of lipases, such as ATGL, HSL and perilipin, and the thermogenesis gene Ucp-1 increased in white and brown adipocytes during both the day and night cycles, indicating increased lipolysis and lipid expenditure, compared with the expression in the health condition, in both the inactive and active phases. Pparγ transcriptional coregulator peroxisome proliferator-activated receptor gamma; C/EBPα CCAAT/enhancer-binding protein α; Fas fatty acid synthase; Dgat diacylglycerol O-acyltransferase; ATGL lipases adipose triglyceride lipase; HSL hormone‐sensitive lipase

Increased fat expenditure in brown adipose tissue is related to disrupted circadian rhythm

Brown adipose tissue is a metabolically active organ distinguished by its unique capacity for adaptive thermogenesis in response to cold and adrenergic stimuli [63]; moreover, its energy-dissipating capacity is necessary to maintain the whole-body energy balance [64, 65]. A study performed by van den Berg et al. [66] revealed that brown adipose tissue displayed a pronounced daily rhythm in FFAs uptake, which was synchronized with the light/dark cycle and was highest upon wakening. Additionally, the diurnal rhythmicity in brown adipose tissue activity determines the rate at which lipids are cleared from circulation, thereby imposing a daily rhythm on plasma lipid concentrations [66]. Indeed, an early study reported that the robust and coordinated expression of circadian oscillator genes, including Bmal1, Per1-3, Cry1-2 and Rev-erbα, in murine brown adipose tissue [67]; moreover, the oscillations of these clock genes were found to be involved in the modulation of lipid metabolism and thermogenesis processes in brown adipose tissue [68]. Disruption of circadian rhythm by prolonged light exposure decreased sympathetic input into brown adipose tissue and reduced β3-adrenergic intracellular signaling, which concomitantly caused increased adiposity, as indicated by reduced utilization of FFAs and glucose in brown adipose tissue [69]. Thus, impaired diurnal activity of brown adipose tissue contributes to disordered metabolism, while restoring the daily rhythm of brown adipose tissue may be a potential target for the treatment of metabolic disorders.

Like white adipocytes, circadian clock genes have also been demonstrated to be involved in brown adipose tissue activity (Table 1). Whole body Clock mutant mice is characterized by excessive lipid accumulation and loss of multilocular appearance in their brown adipocytes [70]. BMAL1 is required for normal lipid metabolism in brown fat. Mice lacking Bmal1 in brown adipocytes revealed increased lipid accumulation and larger lipid droplets accompanied by dysregulation of genes involved in lipid metabolism and adaptive thermogenesis [71]; moreover, the 24 h rhythmicity of fatty acid utilization was disrupted in brown adipose tissue, and these mice were more prone to diet-induced obesity [72]. Additionally, BMAL1 suppresses brown adipogenesis via direct transcriptional control of key components of the transforming growth factor beta (TGF-β) pathway. Global ablation of Bmal1 in mice increased brown fat mass and cold tolerance [73]. Conversely, REV-ERBα promotes brown adipogenesis by suppressing key components of the TGF-β, genetic ablation of Rev-erbα in mice severely impaired brown fat formation accompanied by loss of brown identity [74]. In regards of brown fat function, REV-ERBα controls circadian thermogenic in a manner that is adaptable to environmental demands through repressing UCP-1 expression in brown adipocyte. Genetic loss of Rev-erbα abolished normal rhythms of body temperature and brown fat activity [75]. ROR shares nuclear targets with REV-ERBα, and together with REV-ERBα, it generates the rhythmic expression of BMAL1 that drives the circadian clock cycle [5]. Enhanced UCP-1 and thermogenic gene expression/activity were observed in brown fat of Rorα-deficient mice, indicating repressive effective of RORα on brown fat thermogenesis [76]. PER2 in brown adipose tissue, as a coactivator of PPARα, increases the expression of UCP-1 and promotes the expression of fatty acid-binding protein 3 (Fabp3), a protein important for transporting free fatty acids from plasma to mitochondria to activate UCP-1. These two effects of PER2 were favorable for maintaining the normal body temperature of animals exposed to cold conditions. In contrast, Per2-mutant mice are cold sensitive because their adaptive thermogenesis system is less efficient [77]. Thus, the effect of each clock gene on brown adipose tissue is diverse, and the pathophysiological role of circadian clock in brown adipose tissue need to be further explored.

Effects of circadian clock on brown adipose tissue in the background of cachexia is less clear. In brown adipose tissue of C26 tumor-induced cachexia mice, diurnal expression profiling of key regulators of lipid accumulation and fatty acid β-oxidation and their corresponding target genes revealed dramatic molecular changes, indicating diminished de novo synthesis and storage of lipids and increased uptake of lipids for β-oxidation and energy expenditure, as shown in Fig. 2. Increased Ucp-1, Pbe, and Cpt1α expression at specific points coincided with higher brown adipose tissue temperatures during the dark cycle. This result suggested that perturbed diurnal expression patterns of lipid uptake/accumulation/utilization and thermogenesis pathways in brown adipose tissue likely contribute to the fat expenditure in cancer cachexia [78]. However, in the background of cachexia, the direct causal relationship between circadian clock and increased energy expenditure in brown adipose tissue has not yet been systematically explored.

The circadian clock modulates the expression of chemokines and cytokines involved in fat expenditure in cachexia

Inflammatory cytokines are intimately involved in the initiation and progression of cachexia, with the upregulation of circulating proinflammatory cytokines, including tumor necrosis factor α (TNF-α), interleukin 1 (IL-1), and interleukin 6 (IL-6), as well as decreased anti-inflammatory mediators, including IL-10 [79]. In cancer cachexia, tumor-derived factors such as IL-6 and TNF-α are key drivers of lipolysis and fat depletion [80, 81]. IL-6 is a key inflammatory mediator that upregulates the expression of UCP-1, which mediates white adipose tissue browning during the hypermetabolic response. Mice lacking the IL-6 gene exhibited reduced white adipose tissue browning in response to burn injury [80, 82]. Moreover, IL-6 suppresses lipid synthesis and accelerates lipid catabolism in adipocytes [18]. TNF-α, another critical inflammatory mediator, plays a significant role in cancer cachexia. It promotes lipolysis by increasing the activity of HSL in white adipocytes [83]. More importantly, TNF-α exacerbates macrophage infiltration, which is critical for increasing IL-6 and other proinflammatory cytokines and generating a vicious cycle of macrophage recruitment [84].

The circadian clock controls inflammation and immune functions by modulating the expression of chemokines and cytokines. Circadian clock dysfunction is associated with metabolic inflammation in adipose tissue [85]. CLOCK promotes inflammation by enhancing the genes expression of IL-6, IL-1β, TNF-α and chemokine (C–C motif) ligand 2 (Ccl2) [86]. In contrast, BMAL1 is the central mediator regulating the immune system by directly suppressing Ccl2 expression, leading to lower recruitment of inflammatory monocytes into inflamed tissues [87]. Additionally, overexpression of CLOCK leads to enhanced transcriptional activity of the nuclear factor kappa-B (NF-κB) complex, while BMAL1 attenuates the effect of CLOCK on NF-kB, most likely by sequestering CLOCK [88]. This evidence suggests that BMAL1 suppresses, while CLOCK potentiates, the inflammatory response. REV-ERBα prevents the release of IL-6 from macrophages and attenuates Ccl2 by directly binding to the promoter [89, 90]. Time-of-day gating of macrophages isolated from mice with global Rev-erbα deletion disappeared, and these mice had increased expression of inflammatory cytokines, including IL-6, Ccl2 and chemokine (C–C motif) ligand 5 (Ccl5), in macrophages [91]. In contrast, a REV-ERBα agonist suppressed the expression of IL-6 and Ccl2 in lipopolysaccharide-stimulated human monocyte-derived macrophages [89]. Two other clock proteins, PER/CRY, constitute another mechanism of clock-mediated inflammation modulation. The global disruption of the clock genes Per1 and Per2 exacerbates inflammation [92]. CRY also inhibits inflammation by suppressing the NF-κB pathway, and the absence of CRY results in constitutive elevation of proinflammatory cytokines [93].

Myeloid cells including monocytes and macrophages are the chief cellular effectors of the innate immune system and involved in adipose tissue inflammation during cachexia [94]. In background of burn-induced cachexia, it was shown that polarized M2 macrophages are recruited to subcutaneous adipose tissue and secrete catecholamines to activate browning of white adipose [95]. Ly6C(hi) inflammatory monocytes exhibit diurnal variation, which controls their trafficking to sites of inflammation and polarization. Myeloid cell-specific deletion of Bmal1 induces expression of monocyte-attracting chemokines and disrupts rhythmic cycling of Ly6C(hi) monocytes, predisposing mice to development of pathologies associated with acute and chronic inflammation [96]. In background of atherosclerosis, Bmal1 deficiency in monocytes and macrophages promotes atherosclerosis by enhancing monocyte recruitment to atherosclerotic lesions and enhanced M2 to M1 macrophage transformation [97]. REV-ERBα also has a role in regulating inflammation in macrophages. REV-ERBα agonists SR9009 reduced the polarization of bone marrow-derived mouse macrophages to proinflammatory M1 macrophage while increasing the polarization of macrophages to anti-inflammatory M2 macrophages [98]. Moreover, REV-ERBα regulates the inflammatory infiltration of macrophages through the suppression of Ccl2 expression [90].

As discuss below, circadian clock system directly and indirectly regulates the expression and activation state of cytokines and chemokines, and the inflammatory cell migration and transformation. In cachexia, excessive adipose tissue expenditure is closely associated with production of inflammatory cytokines and recruitment of immune cells; however, whether circadian clock dysregulation involves in adipose tissue expenditure through affecting inflammatory cytokines and immune cell is still unknown. In this regard, future studies should determine the association between circadian clock system and inflammation in cachexia-induced adipose tissue wasting.

Conclusion and future directions

Collectively, a well-functioning circadian clock play important roles in most aspects of adipose biology, from adipogenesis to lipid storage, lipolysis and thermogenesis. Dysregulation of circadian rhythmicity is closely related to disordered lipid metabolism and exacerbated inflammation in adipose tissue, which are critical underlying mechanisms of metabolic disorders.

Nevertheless, our review only provides primary evidences for the link between metabolic disorders and circadian clock dysregulation, studies addressed causes and consequences of circadian clock disruption and adipose expenditure in cachexia are far less understood. In light of the clinical relevance of excessive adipose expenditure and the current lack of effective therapeutic options in cachexia patients, a certain refocusing of clinical and research efforts onto the intricate molecular mechanisms between the dysregulated circadian clock and adipose expenditure in cachexia seems to be necessary and valuable in the future. This field will be to identify circadian clock as a new target in adipose tissues for treating cachexia. Moreover, circadian rhythm-dependent treatment approaches such as “chronopharmacology” and “chrononutrition” should be considered in the treatment and prevention of cachexia.

Availability of data and materials

Not applicable.

Abbreviations

ClOCK:

Circadian locomotor output cycles kaput

BMAL1:

Brain and muscle ARNT-like 1 protein

PER:

Period

CRY:

Cryptochrome

REV-ERBα:

Nuclear receptor reverse ERBα

ROR:

Retinoic acid receptor-related orphan receptor

ROREs:

ROR response elements

UCP-1:

Uncoupling protein 1

FFAs:

Free fatty acids

CD36:

Fatty acid translocase CD36

FATP1:

Fatty acid transport protein 1

C/EBPα:

CCAAT/enhancer binding protein α

C/EBPβ:

CCAAT/enhancer-binding protein beta

PPARγ:

Peroxisome proliferator-activated receptor γ

SREBP-1c:

Sterol regulatory element-binding protein-1c

FAS:

Fatty acid synthase

ACC:

Acetyl-CoA carboxylase

Scd1:

Stearoyl CoA desaturase 1

G3P:

Glycerol-3-phosphate acyl transferase

DGAT2:

Diacylglycerol O-acyltransferase

PKA:

Activates protein kinase A

PKC:

Activates protein kinase C

ATGL:

Adipose triglyceride lipase

TAG:

Triacylglycerol

DAG:

Diacylglycerol

HSL:

Hormone‐sensitive lipase

MAG:

Monoacylglycerol

MGL:

Monoglyceride lipase

PDRM16:

PR domain-containing 16

PGC1α:

PPARγ coactivator 1α

FAS:

Fatty acid synthase

aP2:

Adipocyte protein 2

Fabp3:

Fatty acid-binding protein 3

Pbe:

Peroxisomal bifunctional enzyme

Cpt1α:

Mitochondrial carnitine palmitoyl transferase 1α

TNF-α:

Tumor necrosis factor α

IL-1:

Interleukin 1

IL-6:

Interleukin 6

Ccl2:

Chemokine (C–C motif) ligand 2

Ccl5:

Chemokine (C–C motif) ligand 5

NF-κB:

Nuclear factor kappa-B

References

  1. Gerhart-Hines Z, Lazar MA. Rev-erbα and the circadian transcriptional regulation of metabolism. Diabetes Obes Metab. 2015;17(Suppl 1):12–6.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  2. van der Spek R, Fliers E, la Fleur SE, Kalsbeek A. Daily gene expression rhythms in rat white adipose tissue do not differ between subcutaneous and intra-abdominal depots. Front Endocrinol (Lausanne). 2018;9:206.

    Article  Google Scholar 

  3. Sato TK, Panda S, Miraglia LJ, Reyes TM, Rudic RD, McNamara P, et al. A functional genomics strategy reveals Rora as a component of the mammalian circadian clock. Neuron. 2004;43(4):527–37.

    Article  CAS  PubMed  Google Scholar 

  4. Ueda HR, Hayashi S, Chen W, Sano M, Machida M, Shigeyoshi Y, et al. System-level identification of transcriptional circuits underlying mammalian circadian clocks. Nat Genet. 2005;37(2):187–92.

    Article  CAS  PubMed  Google Scholar 

  5. Preitner N, Damiola F, Lopez-Molina L, Zakany J, Duboule D, Albrecht U, et al. The orphan nuclear receptor REV-ERBalpha controls circadian transcription within the positive limb of the mammalian circadian oscillator. Cell. 2002;110(2):251–60.

    Article  CAS  PubMed  Google Scholar 

  6. Fonseca G, Farkas J, Dora E, von Haehling S, Lainscak M. Cancer cachexia and related metabolic dysfunction. Int J Mol Sci. 2020; 21(7).

  7. Janovska P, Melenovsky V, Svobodova M, Havlenova T, Kratochvilova H, Haluzik M, et al. Dysregulation of epicardial adipose tissue in cachexia due to heart failure: the role of natriuretic peptides and cardiolipin. J Cachexia Sarcopenia Muscle. 2020;11(6):1614–27.

    Article  PubMed  PubMed Central  Google Scholar 

  8. Barajas Galindo DE, Vidal-Casariego A, Calleja-Fernández A, Hernández-Moreno A, Pintor de la Maza B, Pedraza-Lorenzo M, et al. Appetite disorders in cancer patients: Impact on nutritional status and quality of life. Appetite. 2017; 114:23–27.

  9. Vest AR, Chan M, Deswal A, Givertz MM, Lekavich C, Lennie T, et al. Nutrition, obesity, and cachexia in patients with heart failure: a consensus statement from the Heart Failure Society of America Scientific Statements Committee. J Card Fail. 2019;25(5):380–400.

    Article  PubMed  Google Scholar 

  10. Dahlman I, Mejhert N, Linder K, Agustsson T, Mutch DM, Kulyte A, et al. Adipose tissue pathways involved in weight loss of cancer cachexia. Br J Cancer. 2010;102(10):1541–8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Byerley LO, Lee SH, Redmann S, Culberson C, Clemens M, Lively MO. Evidence for a novel serum factor distinct from zinc alpha-2 glycoprotein that promotes body fat loss early in the development of cachexia. Nutr Cancer. 2010;62(4):484–94.

    Article  CAS  PubMed  Google Scholar 

  12. Di Sebastiano KM, Yang L, Zbuk K, Wong RK, Chow T, Koff D, et al. Accelerated muscle and adipose tissue loss may predict survival in pancreatic cancer patients: the relationship with diabetes and anaemia. Br J Nutr. 2013;109(2):302–12.

    Article  PubMed  CAS  Google Scholar 

  13. Barreiro E. Models of disuse muscle atrophy: therapeutic implications in critically ill patients. Ann Transl Med. 2018;6(2):29.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  14. Gonzalez MC, Pastore CA, Orlandi SP, Heymsfield SB. Obesity paradox in cancer: new insights provided by body composition. Am J Clin Nutr. 2014;99(5):999–1005.

    Article  CAS  PubMed  Google Scholar 

  15. Martino JL, Stapleton RD, Wang M, Day AG, Cahill NE, Dixon AE, et al. Extreme obesity and outcomes in critically ill patients. Chest. 2011;140(5):1198–206.

    Article  PubMed  PubMed Central  Google Scholar 

  16. Diaz JJ Jr, Norris PR, Collier BR, Berkes MB, Ozdas A, May AK, et al. Morbid obesity is not a risk factor for mortality in critically ill trauma patients. J Trauma. 2009;66(1):226–31.

    PubMed  Google Scholar 

  17. Hurt RT, Frazier TH, McClave SA, Kaplan LM. Obesity epidemic: overview, pathophysiology, and the intensive care unit conundrum. JPEN J Parenter Enteral Nutr. 2011;35(5 Suppl):4s–13s.

    Article  CAS  PubMed  Google Scholar 

  18. Fearon KC, Glass DJ, Guttridge DC. Cancer cachexia: mediators, signaling, and metabolic pathways. Cell metab. 2012;16(2):153–66.

    Article  CAS  PubMed  Google Scholar 

  19. Das SK, Eder S, Schauer S, Diwoky C, Temmel H, Guertl B, et al. Adipose triglyceride lipase contributes to cancer-associated cachexia. Science (New York, NY). 2011;333(6039):233–8.

    Article  CAS  Google Scholar 

  20. Sun X, Feng X, Wu X, Lu Y, Chen K, Ye Y. Fat wasting is damaging: role of adipose tissue in cancer-associated cachexia. Front Cell Dev Biol. 2020;8:33.

    Article  PubMed  PubMed Central  Google Scholar 

  21. Galic S, Oakhill JS, Steinberg GR. Adipose tissue as an endocrine organ. Mol Cell Endocrinol. 2010;316(2):129–39.

    Article  CAS  PubMed  Google Scholar 

  22. Johnston JD, Ordovás JM, Scheer FA, Turek FW. Circadian rhythms, metabolism, and chrononutrition in rodents and humans. Adv Nutr. 2016;7(2):399–406.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Zechner R, Zimmermann R, Eichmann TO, Kohlwein SD, Haemmerle G, Lass A, et al. FAT SIGNALS–lipases and lipolysis in lipid metabolism and signaling. Cell Metab. 2012;15(3):279–91.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Trayhurn P, Beattie JH. Physiological role of adipose tissue: white adipose tissue as an endocrine and secretory organ. Proc Nutr Soc. 2001;60(3):329–39.

    Article  CAS  PubMed  Google Scholar 

  25. Concha F, Prado G, Quezada J, Ramirez A, Bravo N, Flores C, et al. Nutritional and non-nutritional agents that stimulate white adipose tissue browning. Rev Endocrinol Metab Disord. 2019;20(2):161–71.

    Article  CAS  Google Scholar 

  26. Wu J, Boström P, Sparks LM, Ye L, Choi JH, Giang AH, et al. Beige adipocytes are a distinct type of thermogenic fat cell in mouse and human. Cell. 2012;150(2):366–76.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Bartelt A, Heeren J. Adipose tissue browning and metabolic health. Nat Rev Endocrinol. 2014;10(1):24–36.

    Article  CAS  PubMed  Google Scholar 

  28. Rutkowski JM, Stern JH, Scherer PE. The cell biology of fat expansion. J Cell Biol. 2015;208(5):501–12.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Sun K, Kusminski CM, Scherer PE. Adipose tissue remodeling and obesity. J Clin Invest. 2011;121(6):2094–101.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Lanza-Jacoby S, Lansey SC, Miller EE, Cleary MP. Sequential changes in the activities of lipoprotein lipase and lipogenic enzymes during tumor growth in rats. Cancer Res. 1984;44(11):5062–7.

    CAS  PubMed  Google Scholar 

  31. Tsoli M, Schweiger M, Vanniasinghe AS, Painter A, Zechner R, Clarke S, et al.Depletion of white adipose tissue in cancer cachexia syndrome is associated with inflammatory signaling and disrupted circadian regulation. PLoS ONE. 2014, 9(3):e92966.

  32. Bing C, Russell S, Becket E, Pope M, Tisdale MJ, Trayhurn P, et al. Adipose atrophy in cancer cachexia: morphologic and molecular analysis of adipose tissue in tumour-bearing mice. Br J Cancer. 2006;95(8):1028–37.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Jeschke MG, Gauglitz GG, Finnerty CC, Kraft R, Mlcak RP, Herndon DN. Survivors versus nonsurvivors postburn: differences in inflammatory and hypermetabolic trajectories. Ann Surg. 2014;259(4):814–23.

    Article  PubMed  Google Scholar 

  34. Kulp GA, Herndon DN, Lee JO, Suman OE, Jeschke MG. Extent and magnitude of catecholamine surge in pediatric burned patients. Shock (Augusta, GA). 2010;33(4):369–74.

    Article  CAS  Google Scholar 

  35. Han J, Meng Q, Shen L, Wu G. Interleukin-6 induces fat loss in cancer cachexia by promoting white adipose tissue lipolysis and browning. Lipids Health Dis. 2018;17(1):14.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  36. Petruzzelli M, Schweiger M, Schreiber R, Campos-Olivas R, Tsoli M, Allen J, et al. A switch from white to brown fat increases energy expenditure in cancer-associated cachexia. Cell Metab. 2014;20(3):433–47.

    Article  CAS  PubMed  Google Scholar 

  37. Heier C, Haemmerle G. Fat in the heart: the enzymatic machinery regulating cardiac triacylglycerol metabolism. Biochim Biophys Acta. 2016;1861(10):1500–12.

    Article  CAS  PubMed  Google Scholar 

  38. Dalal S. Lipid metabolism in cancer cachexia. Ann Palliat Med. 2019;8(1):13–23.

    Article  PubMed  Google Scholar 

  39. Abdullahi A, Jeschke MG. White adipose tissue browning: a double-edged sword. Trends Endocrinol Metab. 2016;27(8):542–52.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. van Herpen NA, Schrauwen-Hinderling VB. Lipid accumulation in non-adipose tissue and lipotoxicity. Physiol Behav. 2008;94(2):231–41.

    Article  PubMed  CAS  Google Scholar 

  41. Silvério R, Lira FS, Oyama LM, Oller do Nascimento CM, Otoch JP, Alcântara PSM, et al. Lipases and lipid droplet-associated protein expression in subcutaneous white adipose tissue of cachectic patients with cancer. Lipids Health Dis. 2017, 16(1):159.

  42. Kliewer KL, Ke JY, Tian M, Cole RM, Andridge RR, Belury MA. Adipose tissue lipolysis and energy metabolism in early cancer cachexia in mice. Cancer Biol Ther. 2015;16(6):886–97.

    Article  CAS  PubMed  Google Scholar 

  43. Morrison SF, Madden CJ, Tupone D. Central neural regulation of brown adipose tissue thermogenesis and energy expenditure. Cell Metab. 2014;19(5):741–56.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Kir S, White JP, Kleiner S, Kazak L, Cohen P, Baracos VE, et al. Tumour-derived PTH-related protein triggers adipose tissue browning and cancer cachexia. Nature. 2014;513(7516):100–4.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Sidossis LS, Porter C, Saraf MK, Børsheim E, Radhakrishnan RS, Chao T, et al. Browning of subcutaneous white adipose tissue in humans after severe adrenergic stress. Cell Metab. 2015;22(2):219–27.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Patsouris D, Qi P, Abdullahi A, Stanojcic M, Chen P, Parousis A, et al. Burn induces browning of the subcutaneous white adipose tissue in mice and humans. Cell Rep. 2015;13(8):1538–44.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Vegiopoulos A, Rohm M, Herzig S. Adipose tissue: between the extremes. EMBO J. 2017;36(14):1999–2017.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Batista ML Jr, Peres SB, McDonald ME, Alcantara PS, Olivan M, Otoch JP, et al. Adipose tissue inflammation and cancer cachexia: possible role of nuclear transcription factors. Cytokine. 2012;57(1):9–16.

    Article  CAS  PubMed  Google Scholar 

  49. Reitz CJ, Alibhai FJ, de Lima-Seolin BG, Nemec-Bakk A, Khaper N, Martino TA. Circadian mutant mice with obesity and metabolic syndrome are resilient to cardiovascular disease. Am J Physiol Heart Circ Physiol. 2020;319(5):H1097-h1111.

    Article  CAS  PubMed  Google Scholar 

  50. Zhu Z, Hua B, Xu L, Yuan G, Li E, Li X, et al. CLOCK promotes 3T3-L1 cell proliferation via Wnt signaling. IUBMB Life. 2016;68(7):557–68.

    Article  CAS  PubMed  Google Scholar 

  51. Shimba S, Ishii N, Ohta Y, Ohno T, Watabe Y, Hayashi M, et al. Brain and muscle Arnt-like protein-1 (BMAL1), a component of the molecular clock, regulates adipogenesis. Proc Natl Acad Sci USA. 2005;102(34):12071–6.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. Zhang D, Tong X, Arthurs B, Guha A, Rui L, Kamath A, et al. Liver clock protein BMAL1 promotes de novo lipogenesis through insulin-mTORC2-AKT signaling. J Biol Chem. 2014;289(37):25925–35.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Fontaine C, Dubois G, Duguay Y, Helledie T, Vu-Dac N, Gervois P, et al. The orphan nuclear receptor Rev-Erbalpha is a peroxisome proliferator-activated receptor (PPAR) gamma target gene and promotes PPARgamma-induced adipocyte differentiation. J Biol Chem. 2003;278(39):37672–80.

    Article  CAS  PubMed  Google Scholar 

  54. Cho H, Zhao X, Hatori M, Yu RT, Barish GD, Lam MT, et al. Regulation of circadian behaviour and metabolism by REV-ERB-α and REV-ERB-β. Nature. 2012;485(7396):123–7.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. Solt LA, Wang Y, Banerjee S, Hughes T, Kojetin DJ, Lundasen T, et al. Regulation of circadian behaviour and metabolism by synthetic REV-ERB agonists. Nature. 2012;485(7396):62–8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  56. Griebel G, Ravinet-Trillou C, Beeské S, Avenet P, Pichat P. Mice deficient in cryptochrome 1 (cry1 (-/-)) exhibit resistance to obesity induced by a high-fat diet. Front Endocrinol (Lausanne). 2014;5:49.

    Article  Google Scholar 

  57. Sun S, Zhou L, Yu Y, Zhang T, Wang M. Knocking down clock control gene CRY1 decreases adipogenesis via canonical Wnt/β-catenin signaling pathway. Biochem Biophys Res Commun. 2018;506(3):746–53.

    Article  CAS  PubMed  Google Scholar 

  58. Aggarwal A, Costa MJ, Rivero-Gutiérrez B, Ji L, Morgan SL, Feldman BJ. The circadian clock regulates adipogenesis by a Per3 crosstalk pathway to Klf15. Cell Rep. 2017;21(9):2367–75.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  59. Suzuki M, Shimomura Y, Satoh Y. Diurnal changes in lipolytic activity of isolated fat cells and their increased responsiveness to epinephrine and theophylline with meal feeding in rats. J Nutr Sci Vitaminol (Tokyo). 1983;29(4):399–411.

    Article  CAS  Google Scholar 

  60. Dallmann R, Viola AU, Tarokh L, Cajochen C, Brown SA. The human circadian metabolome. Proc Natl Acad Sci USA. 2012;109(7):2625–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  61. Shostak A, Meyer-Kovac J, Oster H. Circadian regulation of lipid mobilization in white adipose tissues. Diabetes. 2013;62(7):2195–203.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  62. Shimba S, Ogawa T, Hitosugi S, Ichihashi Y, Nakadaira Y, Kobayashi M, et al. Deficient of a clock gene, brain and muscle Arnt-like protein-1 (BMAL1), induces dyslipidemia and ectopic fat formation. PLoS One. 2011, 6(9):e25231.

  63. Oelkrug R, Polymeropoulos ET, Jastroch M. Brown adipose tissue: physiological function and evolutionary significance. J Comp Physiol B. 2015;185(6):587–606.

    Article  CAS  PubMed  Google Scholar 

  64. Saito M, Okamatsu-Ogura Y, Matsushita M, Watanabe K, Yoneshiro T, Nio-Kobayashi J, et al. High incidence of metabolically active brown adipose tissue in healthy adult humans: effects of cold exposure and adiposity. Diabetes. 2009;58(7):1526–31.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  65. van Marken Lichtenbelt WD, Vanhommerig JW, Smulders NM, Drossaerts JM, Kemerink GJ, Bouvy ND, et al. Cold-activated brown adipose tissue in healthy men. N Engl J Med. 2009;360(15):1500–8.

    Article  PubMed  Google Scholar 

  66. van den Berg R, Kooijman S, Noordam R, Ramkisoensing A, Abreu-Vieira G, Tambyrajah LL, et al. A diurnal rhythm in brown adipose tissue causes rapid clearance and combustion of plasma lipids at wakening. Cell Rep. 2018;22(13):3521–33.

    Article  PubMed  CAS  Google Scholar 

  67. Zvonic S, Ptitsyn AA, Conrad SA, Scott LK, Floyd ZE, Kilroy G, et al. Characterization of peripheral circadian clocks in adipose tissues. Diabetes. 2006;55(4):962–70.

    Article  CAS  PubMed  Google Scholar 

  68. Froy O, Garaulet M. The circadian clock in white and brown adipose tissue: mechanistic, endocrine, and clinical aspects. Endocrinol Rev. 2018;39(3):261–73.

    Article  Google Scholar 

  69. Kooijman S, van den Berg R, Ramkisoensing A, Boon MR, Kuipers EN, Loef M, et al. Prolonged daily light exposure increases body fat mass through attenuation of brown adipose tissue activity. Proc Natl Acad Sci USA. 2015;112(21):6748–53.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  70. Turek FW, Joshu C, Kohsaka A, Lin E, Ivanova G, McDearmon E, et al. Obesity and metabolic syndrome in circadian Clock mutant mice. Science. 2005;308(5724):1043–5.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  71. Li S, Yu Q, Wang GX, Lin JD. The biological clock is regulated by adrenergic signaling in brown fat but is dispensable for cold-induced thermogenesis. PLoS ONE. 2013, 8(8): e70109.

  72. Hasan N, Nagata N, Morishige JI, Islam MT, Jing Z, Harada KI, et al. Brown adipocyte-specific knockout of Bmal1 causes mild but significant thermogenesis impairment in mice. Mol Metab. 2021;49:101202.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  73. Nam D, Guo B, Chatterjee S, Chen MH, Nelson D, Yechoor VK, et al. The adipocyte clock controls brown adipogenesis through the TGF-β and BMP signaling pathways. J Cell Sci. 2015;128(9):1835–47.

    CAS  PubMed  PubMed Central  Google Scholar 

  74. Nam D, Chatterjee S, Yin H, Liu R, Lee J, Yechoor VK, et al. Novel function of Rev-erbα in promoting brown adipogenesis. Sci Rep. 2015;5:11239.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  75. Gerhart-Hines Z, Feng D, Emmett MJ, Everett LJ, Loro E, Briggs ER, et al. The nuclear receptor Rev-erbα controls circadian thermogenic plasticity. Nature. 2013;503(7476):410–3.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  76. Lau P, Tuong ZK, Wang SC, Fitzsimmons RL, Goode JM, Thomas GP, et al. Rorα deficiency and decreased adiposity are associated with induction of thermogenic gene expression in subcutaneous white adipose and brown adipose tissue. Am J Physiol Endocrinol Metab. 2015;308(2):E159-171.

    Article  CAS  PubMed  Google Scholar 

  77. Chappuis S, Ripperger JA, Schnell A, Rando G, Jud C, Wahli W, et al. Role of the circadian clock gene Per2 in adaptation to cold temperature. Mol Metab. 2013;2:184–93.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  78. Tsoli M, Moore M, Burg D, Painter A, Taylor R, Lockie SH, et al. Activation of thermogenesis in brown adipose tissue and dysregulated lipid metabolism associated with cancer cachexia in mice. Can Res. 2012;72(17):4372–82.

    Article  CAS  Google Scholar 

  79. Loncar G, Springer J, Anker M, Doehner W, Lainscak M. Cardiac cachexia: hic et nunc. J Cachexia Sarcopenia Muscle. 2016;7(3):246–60.

    Article  PubMed  PubMed Central  Google Scholar 

  80. Rydén M, Agustsson T, Laurencikiene J, Britton T, Sjölin E, Isaksson B, et al. Lipolysis–not inflammation, cell death, or lipogenesis–is involved in adipose tissue loss in cancer cachexia. Cancer. 2008;113(7):1695–704.

    Article  PubMed  Google Scholar 

  81. Han J, Lu C, Meng Q, Halim A, Yean TJ, Wu G. Plasma concentration of interleukin-6 was upregulated in cancer cachexia patients and was positively correlated with plasma free fatty acid in female patients. Nutr Metab (Lond). 2019;16:80.

    Article  CAS  Google Scholar 

  82. Mauer J, Chaurasia B, Goldau J, Vogt MC, Ruud J, Nguyen KD, et al. Signaling by IL-6 promotes alternative activation of macrophages to limit endotoxemia and obesity-associated resistance to insulin. Nat Immunol. 2014;15(5):423–30.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  83. Jeanson Y, Carrière A, Casteilla L. A new role for browning as a redox and stress adaptive mechanism? Front Endocrinol (Lausanne). 2015;6:158.

    Article  Google Scholar 

  84. Niu S, Bian Z, Tremblay A, Luo Y, Kidder K, Mansour A, et al. Broad infiltration of macrophages leads to a proinflammatory state in streptozotocin-induced hyperglycemic mice. J Immunol. 2016;197(8):3293–301.

    Article  CAS  PubMed  Google Scholar 

  85. Maury E, Navez B, Brichard SM. Circadian clock dysfunction in human omental fat links obesity to metabolic inflammation. Nat Commun. 2021;12(1):2388.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  86. Bellet MM, Deriu E, Liu JZ, Grimaldi B, Blaschitz C, Zeller M, et al. Circadian clock regulates the host response to Salmonella. Proc Natl Acad Sci USA. 2013;110(24):9897–902.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  87. Curtis AM, Bellet MM, Sassone-Corsi P, O’Neill LA. Circadian clock proteins and immunity. Immunity. 2014;40(2):178–86.

    Article  CAS  PubMed  Google Scholar 

  88. Spengler ML, Kuropatwinski KK, Comas M, Gasparian AV, Fedtsova N, Gleiberman AS, et al. Core circadian protein CLOCK is a positive regulator of NF-κB-mediated transcription. Proc Natl Acad Sci USA. 2012;109(37):E2457-2465.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  89. Gibbs JE, Blaikley J, Beesley S, Matthews L, Simpson KD, Boyce SH, et al. The nuclear receptor REV-ERBα mediates circadian regulation of innate immunity through selective regulation of inflammatory cytokines. Proc Natl Acad Sci USA. 2012;109(2):582–7.

    Article  CAS  PubMed  Google Scholar 

  90. Sato S, Sakurai T, Ogasawara J, Takahashi M, Izawa T, Imaizumi K, et al. A circadian clock gene, Rev-erbα, modulates the inflammatory function of macrophages through the negative regulation of Ccl2 expression. J Immunol. 2014;192(1):407–17.

    Article  CAS  PubMed  Google Scholar 

  91. Pariollaud M, Gibbs JE, Hopwood TW, Brown S, Begley N, Vonslow R, et al. Circadian clock component REV-ERBα controls homeostatic regulation of pulmonary inflammation. J Clin Investig. 2018;128(6):2281–96.

    Article  PubMed  PubMed Central  Google Scholar 

  92. Xu H, Li H, Woo SL, Kim SM, Shende VR, Neuendorff N, et al. Myeloid cell-specific disruption of Period1 and Period2 exacerbates diet-induced inflammation and insulin resistance. J Biol Chem. 2014;289(23):16374–88.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  93. Narasimamurthy R, Hatori M, Nayak SK, Liu F, Panda S, Verma IM. Circadian clock protein cryptochrome regulates the expression of proinflammatory cytokines. Proc Natl Acad Sci USA. 2012;109(31):12662–7.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  94. Fabbiano S, Suárez-Zamorano N, Rigo D, Veyrat-Durebex C, Stevanovic Dokic A, Colin DJ, et al. Caloric restriction leads to browning of white adipose tissue through type 2 immune signaling. Cell Metab. 2016;24(3):434–46.

    Article  CAS  PubMed  Google Scholar 

  95. Abdullahi A, Auger C, Stanojcic M, Patsouris D, Parousis A, Epelman S, et al. Alternatively activated macrophages drive browning of white adipose tissue in burns. Ann Surg. 2019;269(3):554–63.

    Article  PubMed  Google Scholar 

  96. Nguyen KD, Fentress SJ, Qiu Y, Yun K, Cox JS, Chawla A. Circadian gene Bmal1 regulates diurnal oscillations of Ly6C(hi) inflammatory monocytes. Science. 2013;341(6153):1483–8.

    Article  CAS  PubMed  Google Scholar 

  97. Huo M, Huang Y, Qu D, Zhang H, Wong WT, Chawla A, et al. Myeloid Bmal1 deletion increases monocyte recruitment and worsens atherosclerosis. FASEB J. 2017;31(3):1097–106.

    Article  CAS  PubMed  Google Scholar 

  98. Sitaula S, Billon C, Kamenecka TM, Solt LA, Burris TP. Suppression of atherosclerosis by synthetic REV-ERB agonist. Biochem Biophys Res Commun. 2015;460(3):566–71.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

None.

Funding

This work was supported by the Chinese National Natural Science Foundation (Nos. 82004280 and 82074388).

Author information

Authors and Affiliations

Authors

Contributions

YW provided the overall concept and framework of the manuscript, DM search appropriate articles and draft the manuscript. XL, YW and LC revised the manuscript. All authors read and approved the manuscript.

Corresponding author

Correspondence to Yong Wang.

Ethics declarations

Ethics approval and consent to participate

Not applicable.

Consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Additional information

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Ma, D., Li, X., Wang, Y. et al. Excessive fat expenditure in cachexia is associated with dysregulated circadian rhythm: a review. Nutr Metab (Lond) 18, 89 (2021). https://doi.org/10.1186/s12986-021-00616-6

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s12986-021-00616-6

Keywords