Next Article in Journal
Correction: Jellinger, K.A. Pathomechanisms of Vascular Depression in Older Adults. Int. J. Mol. Sci. 2022, 23, 308
Next Article in Special Issue
Circadian Disruption and Consequences on Innate Immunity and Inflammatory Response
Previous Article in Journal
The Arabidopsis thaliana–Streptomyces Interaction Is Controlled by the Metabolic Status of the Holobiont
Previous Article in Special Issue
Transcriptional Response of Circadian Clock Genes to an ‘Artificial Light at Night’ Pulse in the Cricket Gryllus bimaculatus
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Role of Circadian Transcription Factor Rev-Erb in Metabolism and Tissue Fibrosis

1
Research Unit of Biomedicine, Medical Research Center, Faculty of Medicine, University of Oulu, 90220 Oulu, Finland
2
Department of Nutrition and Dietetics, Faculty of Health Sciences, Marmara University, 34854 Istanbul, Turkey
3
Oulu University Hospital, University of Oulu, 90220 Oulu, Finland
4
Pediatric Gastroenterology and Metabolic Diseases, Pediatric Institute, Poznan University of Medical Sciences, 60-572 Poznań, Poland
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2022, 23(21), 12954; https://doi.org/10.3390/ijms232112954
Submission received: 5 September 2022 / Revised: 21 October 2022 / Accepted: 22 October 2022 / Published: 26 October 2022

Abstract

:
Circadian rhythms significantly affect metabolism, and their disruption leads to cardiometabolic diseases and fibrosis. The clock repressor Rev-Erb is mainly expressed in the liver, heart, lung, adipose tissue, skeletal muscles, and brain, recognized as a master regulator of metabolism, mitochondrial biogenesis, inflammatory response, and fibrosis. Fibrosis is the response of the body to injuries and chronic inflammation with the accumulation of extracellular matrix in tissues. Activation of myofibroblasts is a key factor in the development of organ fibrosis, initiated by hormones, growth factors, inflammatory cytokines, and mechanical stress. This review summarizes the importance of Rev-Erb in ECM remodeling and tissue fibrosis. In the heart, Rev-Erb activation has been shown to alleviate hypertrophy and increase exercise capacity. In the lung, Rev-Erb agonist reduced pulmonary fibrosis by suppressing fibroblast differentiation. In the liver, Rev-Erb inhibited inflammation and fibrosis by diminishing NF-κB activity. In adipose tissue, Rev- Erb agonists reduced fat mass. In summary, the results of multiple studies in preclinical models demonstrate that Rev-Erb is an attractive target for positively influencing dysregulated metabolism, inflammation, and fibrosis, but more specific tools and studies would be needed to increase the information base for the therapeutic potential of these substances interfering with the molecular clock.

1. Introduction

Fibrosis is the body’s response to injuries, and chronic inflammation is increasingly recognized as an important cause of morbidity and mortality [1]. It has been estimated that fibrosis in different organs is associated with 45% of all deaths in the industrialized world [2]. Fibrosis is defined as “an excessive accumulation of extracellular matrix (ECM) proteins in tissues, preventing functions and dynamically remodeling” [3,4]. After an injury, molecules from damaged cells induce an inflammatory reaction with the attraction of cells e.g., monocytes, neutrophils, and other immune cells and increased fibroblasts differentiation into ECM-producing myofibroblasts [5]. ECM is a noncellular component essential for tissue differentiation, morphogenesis, and homeostasis and includes fibrous proteins collagens, fibronectins, elastin, laminins, and hydrophilic proteoglycans [6]. During fibrosis, the functional cells such as cardiomyocytes, hepatocytes, acinar cells, podocytes, alveolar cells, and neurons are being replaced by ECM proteins such as collagen and fibronectin in the tissues [3,7]. Various factors such as hereditary disorders, age, obesity, diabetes and hypertension, hypercholesterolemia, persistent infections, and recurrent exposure to toxins, irritants/smoke affect wound healing after injury, resulting either in progressive fibrosis or healthy tissue repair [8]. Collagen is the most abundant fibrous protein of ECM, which provides tensile strength, regulates cell adhesions, and supports chemotaxis and tissue development [9]. Myofibroblasts synthesize ECM proteins and are well-known fibrosis effectors in various tissues (Figure 1) [10].
Myofibroblasts are spindle or stellate shape fusiform cells with a similar ultrastructure as smooth muscle cells and are typically found in scar and granulation tissues and stroma of tumors [11]. These cells possess both secretory characteristics of fibroblasts and contractile characteristics of smooth muscles and play vital roles in swiftly repairing injured tissues. Myofibroblasts lack smooth muscle markers desmin and myosin and display elevated levels of stress fibers, exosomes, and microvesicles [12]. They typically contain elongated and serrated nuclei, multiple dendritic processes, extensive rough ER, fibrillar collagens, and microfilament proteins [13,14]. Myofibroblasts synthesize and secrete ECM proteins such as collagens (e.g., I and III), a specialized isoform of fibronectin, periostin, and cadherin-2, and -11 [13]. In addition, they secrete numbers of cytokines, inflammatory mediators (CO, H2O2, NO, O-radical, HETE, PAF, and prostacyclin), and growth factors (IGF, PDGF TGF-β) maintaining the inflammatory response to an injury [15]. Cell-adhesion molecules (CAMs) such as ICAM1 and VCAM1 are expressed in subtypes of myofibroblasts, and induce communications with immune cells such as T-cells and neutrophils [16,17].
Transdifferentiation of quiescent mesenchymal cells to activated myofibroblasts is a key event in the pathogenesis of fibrosis [18]. The potential progenitor cells for myofibroblasts consist of fibroblasts, fibrocytes, epithelial and endothelial cells, and other mesenchymal cells such as pericytes, depending on the insult and organ [19,20]. The early changes occur during myofibroblast formation with mitochondrial changes and ROS production [21,22]. Recently, it has been demonstrated that mitochondrial Ca2+ signaling is a key regulator of myofibroblast differentiation and fibrosis [21]. The differentiation of myofibroblasts is initiated by various factors such as hormones (endothelin-1 and angiotensin II), growth factors (TGF-β and VEGF), and inflammatory cytokines (TNF-α, IL-1, and IL-6), Ca2+, and mechanical stress [23,24], which are linked to the circadian rhythm [25].

2. Circadian Rhythm

Circadian rhythm is an autonomous self-sustained oscillation of 24 h, which regulates the metabolism of organisms [26,27]. The circadian clock drives the daily rhythm and controls coordinated oscillations of many neuroendocrine, signaling, and metabolic pathways [28]. Disturbances of these interactions, as in shift work, transcontinental fights, and irregular eating patterns, are increasingly recognized as risk factors for CNS, metabolic, cardiovascular diseases, and cancer (Figure 2) [29,30,31].
At the cellular level, circadian clock genes consist of transcription-translation feedback loops, in which Clock and brain and muscle Arnt-like protein-1 (Bmal1) drive expression of 2 negative-feedback arms controlled by Period/Cryptochrome (Per/Cry) and the two paralogs, Rev-Erbα and Rev-Erbβ [32]. In turn, these negative-feedback arms repress Bmal1/Clock transactivation function (Per/Cry) or Bmal1 expression (Rev-Erbα/β) and Bmal1 activation (ROR) (Figure 3).
Circadian rhythms are influenced by light, physical activity, diet, aging, and disease states [33,34]. Genetic polymorphisms within the core clock genes Bmal1 and Clock are associated with metabolic diseases such as obesity, type 2 diabetes, and hypertension [35,36]. More than half of human genes display circadian oscillations in at least one body tissue or organ [37]. Changes in circadian transcriptions affect major homeostatic mechanisms, including stem cell regulation, mitochondrial function, and immune responses. Mitochondria exhibit a bidirectional relationship with the circadian clock via a reciprocal interaction between peroxisome proliferator-activated receptor gamma co-activator 1-alpha (PGC-1α) and Bmal1 [38]. In addition, fatty acid oxidation enzymes and electron transfer flavoproteins also follow circadian oscillation [39]. Furthermore, clock-controlled genes (CCGs) such as O-linked beta-D-N-acetylglucosamine (O-GlcNAc) signaling, nicotinamide adenine dinucleotide (NAD+)-dependent sensors, nicotinamide phosphoribosyl transferase (NAMPT), silent mating type information regulation 2 homolog 1 (SIRT1), and 5′-adenosine monophosphate-activated protein kinase (AMPK) link the clocks with metabolic regulators [40,41,42]. Nuclear receptors (NRs) exhibit circadian oscillations in metabolic tissues, in addition to direct transcriptional regulation of Bmal1 via ROR and Rev-Erb. Circadian clocks regulate various immune markers such as interleukin IL-2, IL-10, IL-6, IL-1β, and tumor necrosis factor-α (TNF-α) [43,44]. Disruption of the circadian rhythm increases inflammatory markers such as IL-6 and TNF-α, affecting circulating lymphocytes and natural killer (NK) cells [45]. Inflammation-related genes such as nuclear factor erythroid2-related factor 2 (NRF2), nuclear factor kappa B (NF-κB), chemokine ligands, and PPARs are strongly linked with the molecular clocks [46,47,48].

Circadian Repressor Rev-Erb

Rev-Erbα (Nr1d1) and Rev-Erbβ (Nr1d2) are members of the nuclear receptor (NRs) superfamily and have significant roles in the maintenance of circadian rhythm, metabolic process, and immune functions [49,50]. They have distinct features among NRs because their ligand binding domain lack activation function-2 (AF2) at C-terminus, which is essential for interacting with co-activators and gene transcription [51]. Rev-Erb acts as constitutive repressors of circadian transcription either passively by competing with the transcriptional activator retinoic acid-related orphan receptor (ROR) at binding elements (RORE) [52,53], or actively by recruiting the NCoR-HDAC3 corepressor complex [54,55]. Rev-Erbs regulate glucose, lipids, and energy metabolism as well as adipogenesis and inflammation (Figure 4) [56,57].
Synthetic Rev-Erb agonists modulate metabolic states by altering central and peripheral clocks [49,58]. The Rev-Erb agonist (SR9009) reduced obesity and improved plasma lipid profiles in DIO mice by increasing energy expenditure [58,59]. In addition, the Rev-Erb agonist had beneficial effects on jet lag, sleep disturbances, metabolic diseases, inflammation, and cancer [60,61,62]. Daily administration of the Rev-Erb agonist (SR9011) reduced weight gain, fat mass, and expression of lipogenic genes in normal mice [58,63]. Gibbs et al. showed that genetic knockdown of Rev-Erbα increased IL-6 expression in response to lipopolysaccharides (LPS) and the Rev-Erbα ligand (GSK4112) inhibited the production and release of IL-6 in human macrophages [62]. Furthermore, the author reported that in the absence of Rev-Erbα, the inhibitory effect of GSK4112 on IL-6 response was abolished, indicating that Rev-Erbα regulates the production and secretions of inflammatory cytokines [62].

3. Fibrotic Mediators

Two major mediators of tissue fibrosis are transforming growth factor-β (TGF-β) [64] and hypoxia-inducible factor 1 alpha (HIF-1α) [65]. Proteases such as plasmin, cathepsin, thrombospondin, calpain, integrin-αvβ6, and MMPs activate TGF-β, which is a potential target for antifibrotic drugs [66,67]. TGF-β affects the circadian clock gene machinery by inducing expression of Bmal1 and Clock and inhibiting expressions of Per1/2, Rev-Erbα, RORα, and cold-inducible RNA binding protein (Cirbp) (Figure 5) [68,69]. TGF-β2 inhibits the expression of circadian clock genes; Per, Rev-Erbα, and CCGs; D-site albumin promoter binding protein (DBP) and thyrotroph embryonic factor (TEF), without altering Bmal1 expression in NIH3T3 fibroblasts and HT22 neurons [70]. TGF-β signaling is influenced by Rev-Erb and Bmal1, and TGF-β itself alters Rev-Erb and Bmal1 expressions [71,72]. Hypoxia increases the expression of proteases such as MMPs and thrombospondin 1 (TSP-1) activating TGF-β [73].
HIF-1α activation increases the production of proinflammatory cytokine TNF-α, IL-1β, IL-6, and IL-8, while HIF-2α reduces cytokine productions [74,75]. Clock genes respond to acute hypoxia in a tissue-specific manner with an inter-tissue clock misalignment [76]. HIF-1α activation disrupted the circadian rhythm by dampening the oscillation amplitudes of Bmal1 and Per2 in a dose-dependent manner in myoblasts and osteosarcoma (U2OS) cells [39,77]. PER2 enhances HIF-1α activity by facilitating the recruitment of HIF-1α to the hypoxia-response element (HRE) on the VEGF promoter [78]. Cry1 inhibits HIF-1α transactivation and reduces the half-life of HIF-1α [79].
Other important molecular pathways for tissue fibrosis include platelet-derived growth factor (PDGF), connective-tissue growth factor (CTGF), vasoactive peptides (especially angiotensin II and endothelin-1), and integrins [64,80]. TNF-α, CTGF, PDGF, and FGF induce fibrosis by increasing ECM accumulation via myofibroblast differentiation [81]. In addition, various cytokines such as IL-4, IL-13, IL-17, IL-25, and IL-33 induce fibrosis in different organs [82,83,84,85].
The role of Rev-Erb in different organs will be discussed in the following chapters.

4. Effect of Rev-Erb in Tissue Fibrosis

4.1. Rev-Erb in Heart Fibrosis

The prevalence of heart diseases are continuously rising, and globally, approximately 64.3 million people are affected by heart failure (HF) [86]. Recently, a meta-analysis showed that the prevalence of ‘all types’ heart failure in developed countries is around 11.8% in those aged 65 years and older [87]. Ischemic injury leads to cardiac remodeling and fibrosis, resulting in HF. Cardiac injuries lead to the development of cardiac fibrosis, which is associated with increased ECM proteins in the myocardium. The ECM network of the heart mainly consists of collagen 1 (Col1; 85% of the total myocardial collagen), providing tensile strength with minor amounts of collagen III (Col3), which provides elasticity [88]. The fibrotic response is triggered by growth factors and proteases in the cardiac ECM after injury [89].
Rodents and human hearts displayed robust circadian rhythms [90,91]. Studies in murine models demonstrated that genetic disruption of the circadian clock gene Bmal1 resulted in cardiomyopathy and reduced lifespan [92,93]. Clock-deficient mice developed age-dependent cardiac hypertrophy and interstitial fibrosis at 21 months of age [94]. Furthermore, shift workers are more prone to cardiovascular diseases such as atrial fibrillation and coronary heart disease (CHD) [95].
Rev-Erb agonist (SR9009) prevented cardiomyocyte hypertrophy, reduced fibrosis, and halted the progression of HF in mice [96]. In addition, the Rev-Erb agonist (SR9009) improved left ventricles (LV) function and survival after myocardial infarction (MI) (Table 1) [97]. SR9009 decreased the expression of cytokines IL-6, monocyte chemoattractant protein-1 (MCP-1), MMP9, and immune cells (neutrophil and proinflammatory macrophages) infiltration in the infarcted heart [97]. In pressure-induced cardiac hypertrophy by transverse aortic constriction (TAC) in mice the Rev-Erb agonist (SR9009) reduced protein kinase B (AKT) expression and cardiac hypertrophy [94]. In LDL-receptor-deficient mice fed with a western diet, Rev-Erb agonist (SR9009) reduced atherosclerosis [98]. The SR9009-treated mice had a reduced ratio of proinflammatory M1 macrophages to the anti-inflammatory M2 macrophages, indicating that the reduction in atherosclerotic plaque is due to the anti-inflammatory activity of the Rev-Erb agonist [98].
Overall, the effects of Rev-Erbs on cardiac functions are only sparsely studied and would need further investigation. In addition, human studies regarding the role of the circadian clock in cardiac fibrosis are lacking.

4.2. Rev-Erb in Lung Fibrosis

Inflammatory lung diseases such as COPD and asthma frequently show daily variations in symptoms, and genes involved in lung functions display circadian rhythms [124,125]. Circadian clock genes play an important role in pulmonary fibrosis, COPD, and lung cancer [99,126]. Bmal1 deletion in lung epithelium enhanced the inflammatory reaction in response to cigarette smoke and lipopolysaccharide (LPS) [102,127]. Dong et al. (2016) reported that TGF-β induction increased Bmal1 and decreased Rev-Erbα and RORα expression in the lung while silencing Bmal1 reduced TGF-β–induced endothelium mesenchymal transition (EMT) and MMP9 production in lung epithelium [128]. Recently, Cunningham et al. demonstrated that circadian repressor Rev-Erbα inhibited myofibroblast differentiation and collagen secretion in cultured fibroblasts and lung tissues of pulmonary fibrosis patients (Table 1) [99]. Mice with emphysema had lower expression of Rev-Erbα genes and proteins in the lung tissues [102]. Rev-Erb−/− mice displayed increased pulmonary myofibroblast activating markers such as collagen-1 and α-SMA [99]. Sundar et al. demonstrated that Rev-Erbα knockout mice had an increased neutrophils influx in the lungs and proinflammatory cytokines (IL-6, MCP-1, keratinocyte chemoattractant (KC)) release after exposure to cigarette smoke or LPS [100]. Smokers and COPD patients showed lower Rev-Erbα mRNA and protein expression compared to nonsmokers in PBMCs, sputum, and lung tissues [101]. The Rev-Erb agonist (GSK 4112) suppressed TGF-β–induced fibroblast differentiation by inhibiting activation of mesenchymal markers in human fetal lung fibroblast 1 cells [115] and attenuated both LPS and cigarette smoking-induced inflammatory responses and pulmonary fibrosis [114,127] in human small airway epithelial cells and mouse lung fibroblasts [100].

4.3. Rev-Erb in Liver Fibrosis

Liver fibrosis results from a chronic liver injury, e.g., alcohol or overnutrition, which may lead to liver cirrhosis and hepatocellular carcinoma (HCC). The fibrous tissues disrupt the normal architecture and functions of the liver and increase portal vein pressure [129]. Apoptotic hepatocytes activate quiescent hepatic stellate cells (HSCs) to transdifferentiate into myofibroblasts via TGF-β [130,131]. The activated HSCs induce ECM deposition in liver tissues via tissue TIMPs, which upregulate TNFα and IL-1β expression [132]. During liver injury, expression of inflammatory cytokines such as TNF-α, TGF-β, and platelet-derived growth factor (PDGF) increase, which exacerbate the progression of liver fibrosis by activation of the Ras-MAPK, PI3K-AKT/PKB, and PKC pathways [133].
The liver is the main organ for the control of lipid homeostasis, which is disrupted in clock mutant mice [134,135]. In mice liver, app. 17% of lipids display robust circadian rhythmicity [136], with similar results obtained in a human lipidomic study [137]. In the liver, Rev-Erbα regulates multiple genes involved in metabolic pathways [54]. Rev-Erbα knockout mice exhibit increased plasma VLDL concentrations and APOC-III expression [103]. Hepatic triglyceride levels were increased in the liver-specific knockout of Rev-Erbα/β mice fed with HFD [105]. These mice had an enhanced rhythmic expression of sterol regulatory element binding transcription factor 1 (Srebf1), which regulates de novo lipogenesis by its target genes [105]. Similar results on Srebf1, cholesterol, and bile acid metabolism were reported in whole-body Rev-Erbα knockout mice [138]. Rev-Erbα null mice had hepatic steatosis and reduced bile acid synthesis, which were aggravated by the additional knockdown of Rev-Erbβ [55,104]. The disrupted circadian behavior was more distinct in the Rev-Erbα/β double-knockout model, suggesting additive functions of Rev-Erbα and Rev-Erbβ [109]. Rev-Erbα expression is upregulated in activated HSCs, and an increased hepatic mRNA and protein expression were found in CCL4-treated mice [120,139]. Primary hepatocytes from Rev-Erbα−/− mice had markedly increased lipid accumulation and Cyp4a10 and Cyp4a14 expression [106]. Cyp4a10 and Cyp4a14 are highly expressed in the liver converting arachidonic acid to 20-hydroxyeicosatetraenoic acid, which induce inflammation via ROS generation [140]. Cyp4a14 deficient mice had reduced lipid accumulation, hepatic inflammation, and fibrosis in methionine and choline-deficient diet-induced nonalcoholic steatohepatitis (NASH) [113], indicating that Rev-Erbα acts as a transcriptional repressor of Cyp4a10 and Cyp4a14.
In addition to genetically modified animals, synthetic Rev-Erb ligands influenced lipid metabolism. The Rev-Erb agonist (SR9009) displayed beneficial effects such as weight loss and reduced plasma triglycerides and cholesterol [63]. The Rev-Erb agonist (SR9009) inhibited cholesterol biosynthesis by suppressing liver Hmgcr and Srebf2 [116]. In vitro and in vivo studies demonstrated that Rev-Erbα agonist reduced fibrosis in rat HSCs and CCL4-induced liver fibrosis [120,121]. The Rev-Erb agonist (SR9009) inhibited HSCs proliferation by inhibition of the AKT/mTOR/P70S6K pathway [117]. Furthermore, Rev-Erb agonist (SR9009) prevented alcohol-induced liver injury in mice by downregulating Cyp4a1 expression [106]. Rev-Erb agonist (SR9009) reduced hepatic inflammation (IL-1α, IL-1β, Ifnγ, and TNFα) and fibrosis [118] via decreased expression of profibrotic genes (TGFβ, STAT1 Col3A1, Acta2, MMP13 and TIMP1 in a NASH mouse model (ob/ob mice fed a high-fat, high-fructose and high cholesterol diet)) [141]. Rev-Erb agonist (SR9009) protected against CCL4-induced liver fibrosis in mice by upregulation of the expression of the clock genes Bmal1, Clock, Per2, Cry1, and RORα [120]. In NAFLD patients, studies of Rev-Erbα are sparse; however, a significant down-regulation of Rev-Erbα was reported in pediatric patients [142], suggesting that Rev-Erb agonist could be a novel target for the prevention of liver fibrosis.

4.4. Rev-Erb in Adipose Tissue Fibrosis

Adipocyte hypertrophy and hyperplasia are characteristic features of obesity, which trigger inflammation and, eventually, adipose tissue fibrosis [143]. White adipose tissue (WAT) is a complex organ that includes a number of cell types in addition to adipocytes, such as preadipocytes, endothelial cells, fibroblasts, and immune cells (macrophages, mast cells, B and T cells) [144]. An increase in adipocyte size during obesity results in hypoxia due to inadequate blood supply and increased oxygen consumption [145,146]. Hypoxia increases the expression of genes such as vascular endothelial growth factor (VEGF) and MMP2 and MMP9 in AT affecting vascularization, ECM remodeling, and fibrosis [147]. Enlarged adipocytes are more dysfunctional and produce a higher amount of fatty acid, TNFα, IL-6, IL-8, and MCP-1 and display reduced mitochondrial size [148,149]. Inflammation in AT leads to obesity-associated fibrosis, and macrophages play a major role in WAT fibrosis by Toll-Like Receptor 4 (TLR4) activation [5,150]. TLR4 activation stimulates TGFβ-1 and ECM production as well as fibroblast differentiation by macrophage-inducible C-type lectin [150,151]. The enlarged adipocytes trigger the recruitment of immune cells driving AT inflammation and fibrosis [152,153,154].
Several studies demonstrated that functional peripheral clocks are present in various adipose depots of animals and humans [155,156]. Disruption of the circadian rhythm by constant light exposure increased body weight gain, despite similar caloric intakes and total activity in mice [157]. Visceral AT in diet-induced or genetically obese mice had a dampened circadian rhythm with metabolic dysfunctions [158,159]. In addition, obese and diabetic mice (db/db) displayed disrupted rhythm and higher expressions of collagen I, III, and VI in AT compared to control mice [160]. Collagen VI (Col6) is a major ECM protein in adipose tissue of humans and mice and accumulates at higher rates under obese and diabetic conditions [161,162,163]. Col6α3 elevates systemic inflammations and causes insulin resistance and fibrosis in adipose tissue by inducing TGFβ-1 expression [162,164]. Clock knockout mice displayed visceral adiposity with AT hypertrophy [135,165]. AT-specific deletion of Bmal1 or Cry in mice resulted in higher adiposity, hypertrophic adipocytes, and crown-like structures under HFD [166].
Rev-Erbα expression was upregulated during adipogenesis in 3T3L1 cells and the synthetic Rev-Erb agonist induced adipocyte differentiation [122]. Rev-Erbα knockout mice displayed adiposity and hypertrophied adipocytes, which was aggravated by HFD feeding [107,108]. These mice did not show hyperglycemia due to increased uptake of fatty acid in the muscle by increased lipoprotein lipase (Lpl) gene expression in adipose tissue and muscles [108]. Rev-Erbα downregulated the triglyceride synthesis enzymes Lpl and fibroblast growth factor-21 (FGF21) [108,167]. In Rev-Erb−/− mice, increased plasma adiponectin and its expression in WAT were found [107]. These results suggest that Rev-Erbs improve insulin sensitivity. Mice with adipocyte-specific knockout of Rev-Erbα developed obesity only under a high-fat diet, indicating that Rev-Erbα regulates WAT metabolism in a state-dependent manner [168]. These results suggest that Rev-Erbα does not enforce rhythmic repression of metabolic circuits under basal conditions but rather affects tissue responses to the altered metabolic states. Rev-Erbα expression was decreased in both omental adipocytes and adipocytes of obese subjects due to reduced Bmal1 binding [159]. Rev-Erbα expression was increased in subcutaneous fat after weight loss in overweight subjects [169]. The Rev-Erb agonist (SR9011) reduced fat mass and lipogenic gene expression in mice liver [58]. In addition, Solt et al. confirmed that the Rev-Erb agonist (SR9009) reduced adiposity and inflammation in diet-induced obese mice [58].
These studies demonstrate that Rev-Erbα alleviates metabolic alterations in adipose tissue by reducing inflammation.

4.5. Rev-Erb in Skeletal Muscle Fibrosis

Skeletal muscle (SM) accounts for 30–40% of total body mass and regulates whole-body metabolism and energy homeostasis. ECM contains myofibers, nerves, and blood vessels in SM and constitutes up to 10% of the SM weight [170,171]. The major roles of ECM in SM include force transmission, repair, and maintenance of muscle fibers after injury [172]. SM has a high regeneration ability, and SM clocks affect proteostasis, lipid metabolism, and muscle functions [173]. Hodge et al. [174] reported that of 1628 circadian genes in SM, 62% had metabolic roles, mainly in carbohydrate metabolism. Clock disruptions in SM aggravated metabolic dysfunction and muscle atrophy [175,176]. Muscle-specific Bmal1 knockout mice showed impaired glucose uptake and reduced glucose oxidation in SM without change in glucose tolerance [177], suggesting that reduced muscle insulin sensitivity was most likely compensated by other insulin-sensitive tissues [178].
Rev-Erb is highly expressed in SM with similar oscillations as in the liver and AT [56,179]. Rev-Erbα regulates muscle mass and muscle fiber type distribution and is selectively expressed in glycolytic type IIB and intermediate type IIA muscle fibers [180]. Rev-Erbα−/− mice displayed substantially reduced muscle mass with increased expression of atrophy-related genes (atrogenes) [110]. In addition, Rev-Erbα/β double-knockout mice exhibited altered circadian wheel-running behavior [109]. Rev-Erb−/− mice showed reduced running capacity due to reduced mitochondrial biogenesis and functions by deactivating the serine/threonine kinase 11 (STK11)-AMPK-SIRT-1-PGC-1α signaling pathway [59]. The regenerative capacity of muscle was reduced in Rev-Erb−/− mice; however, partial loss of Rev-Erbα increased regenerative capacity in response to injury [111]. Rev-Erbα overexpression increased exercise capacity, mitochondrial content, and activity in C2C12 myocytes via AMPK [59] and reduced dexamethasone-induced atrophy-related genes in myoblast cells and loss of muscle mass in mice [110]. Rev-Erbβ overexpression affected lipid metabolism and energy expenditure in SM by downregulating Cd36, Fabp-3, and -4 mRNA and upregulating IL-6 expression in mouse myogenic C2C12 cells [181]. Rev-Erbα overexpression alleviated autophagy and reduced oxidative stress in skeletal muscles by directly inhibiting genes such as Ulk1 and perkin (Park2) involved in mitochondrial clearance and mitophagy [59,182]. Welch Billion et al. [123] reported that the Rev-Erb (antagonist SR8278) improved mitochondrial biogenesis and muscle functions, and fibrosis in dystrophic mice by activating Wnt signaling. The group suggested differences in mitochondrial biogenesis in fully differentiated versus proliferating muscles using a dystrophic mouse model [123].

4.6. Rev-Erb in Kidney Fibrosis

Fibrosis is present in chronic kidney disease (CKD) and is characterized by excessive deposition of ECM in the tubular interstitium and glomeruli. CKD is one of the most prominent causes of death and suffering globally [183,184], affecting about 850 million individuals worldwide [185]. Type 2 diabetes and ischemic/hypertensive nephropathy are the two most common causes of CKD in developed nations. Factors that contribute to CKD development include, e.g. xenobiotics, toxins, infections, mechanical obstruction, and autoimmune diseases [186]. The renal medulla is especially prone to hypoxia with increased expression of fibrillar Col I and Col III but also contains Col IV, V, heparan, fibronectin, and laminin [187,188]. The cellular mechanism of renal fibrosis is not yet clear, and there is no specific therapy to halt its progression. In addition, the origin of myofibroblasts in the kidney is controversial and considered to arise from different cell types such as fibroblasts, endothelium, epithelium, and podocytes [189,190].
The kidney is under the control of an intrinsic clock, affecting blood flow, glomerular filtration rate (GFR), water, and electrolyte transport [191,192,193]. Partial nephrectomy (5/6Nx) caused glomerulosclerosis, interstitial fibrosis and induced Bmal1, Clock, and Rev-Erbα expression [194]. Rev-Erbα and Rev-Erbβ were downregulated, and CCGs related to vascular integrity, endothelial function, inflammation, and thrombogenesis were severely deregulated in calcified aortas of rats in CKD [195]. Cisplatin-induced increases in blood urea nitrogen (BUN), serum creatinine, kidney injury molecule (KIM-1), and neutrophil gelatinase-associated lipocalin (NGAL1) were reduced in Bmal1 knockout mice [196]. Kidney fibrosis induced by ureteral obstruction was ameliorated in Bmal1 deficient mice [197,198]. Clock-deficient mice showed severe kidney fibrosis via activation of cyclooxygenase 2 in the unilateral ureteral obstruction model of kidney injury [199]. In Clock mutant mice, adenine-induced CKD was exacerbated by increased MMPs and 2,8-dehydroxyadenine, and fibrosis markers deposition [200]. In contrast, clock mutant mice (Clk/Clk deletion of exon 19 in the Clock locus) ameliorated kidney injury by increased GFR and reduced creatinine, BUN, and fibrosis markers in partially nephrectomized (5/6Nx) mice [201]. The different results on the influence of clocks on kidney functions could be due to various methodologies of acute kidney injury (AKI) and CKD.
Rev-Erbα−/− and Rev-Erbβ−/− mice showed a decreased sensitivity to folic acid-induced AKI and diminished disease severity [112]. The authors reported that Rev-Erbα/β inhibition reduced folic-acid-induced inflammatory cytokine (TNFα, IL-1β) secretion, renal injury (KIM-1 and NGAL), and profibrotic (TGF-β, αMSA) gene expression [112]. Rev-Erbα expression was increased in the renal cortex and medulla of spontaneously hypertensive rats (SHRs) and stroke-prone SHR rats (SHRSP), compared to normal Wistar rats [202]. Dexamethasone administration to tubular epithelial cells of mice increased Rev-Erbα, Fragile X mental retardation autosomal homolog 1 (Fxr1), and phosphoribosyl pyrophosphate amidotransferase (Ppat) expression [202,203]. Fxr1 and Ppat are involved in the formation of circadian rhythm in kidney tubules [202]. Rev-Erb antagonist (SR8278) reduced kidney damage in wild-type mice by increasing gene and protein expressions of solute carrier family 7 member 11 (Slc7a11) and heme oxygenase (HO1), which were elevated in Rev-Erbβ−/− mice [112]. These studies demonstrate the significantly emerging role of Rev-Erb in the kidney, but it is not yet well understood and needs further investigations.

5. Summary and Conclusions

Circadian rhythms are self-controlled oscillations in all organs regulating body functions by directly driving the cyclic expression of nuclear receptors (NRs) and their ligands. As members of this superfamily, the circadian repressor Rev-Erbs regulate energy metabolism, inflammation, and fibrosis. Rev-Erb activation is therapeutically beneficial for alleviating tissue fibrosis in various organs such as the liver, heart, and lungs. The role of Rev-Erb in adipose tissue fibrosis is not fully understood and needs further investigation. Preclinical studies demonstrated the anti-inflammatory and antifibrotic effects of Rev-Erbα; however, limited results on inflammation and tissue fibrosis in clinical settings have been reported.
Rev-Erbs are promising therapeutic targets for the treatment of cardiometabolic diseases and tissue fibrosis, but this is challenging for drug development due to offsite targets. Several Rev-Erb agonists and antagonists were developed, but currently, there is no specific agonist or antagonist for Rev-Erbα, which is the major player in circadian rhythm regulations compared to Rev-Erbβ. More mechanistic and clinical studies would be necessary to further investigate the roles of Rev-Erbs in fibrotic conditions. Target-specific compounds selective to Rev-Erbα are needed for optimal benefits. Rev-Erb synthetic ligands have poor pharmacokinetic parameters, which hinder the development of these agents. Human studies are required to determine whether modulation of Rev-Erbα/β by synthetic ligands in different organs is beneficial in terms of alleviation of cardiometabolic diseases, inflammation, and tissue fibrosis.

Author Contributions

G.S.R. conceptualized and wrote the first draft of the manuscript. N.S. and Y.K. contributed to the writing of the manuscript. K.-H.H. supervised and finalized the manuscript. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Conflicts of Interest

Authors declare no conflict of interest.

References

  1. Franceschi, C.; Campisi, J. Chronic Inflammation (Inflammaging) and Its Potential Contribution to Age-Associated Diseases. J. Gerontol. A. Biol. Sci. Med. Sci. 2014, 69 (Suppl. S1), S4–S9. [Google Scholar] [CrossRef] [PubMed]
  2. Henderson, N.C.; Rieder, F.; Wynn, T.A. Fibrosis: From Mechanisms to Medicines. Nature 2020, 587, 555–566. [Google Scholar] [CrossRef] [PubMed]
  3. Buechler, C.; Krautbauer, S.; Eisinger, K. Adipose Tissue Fibrosis. World J. Diabetes 2015, 6, 548. [Google Scholar] [CrossRef] [PubMed]
  4. Bonnans, C.; Chou, J.; Werb, Z. Remodelling the Extracellular Matrix in Development and Disease. Nat. Rev. Mol. Cell Biol. 2014, 15, 786–801. [Google Scholar] [CrossRef] [PubMed]
  5. Wynn, T.A.; Ramalingam, T.R. Mechanisms of Fibrosis: Therapeutic Translation for Fibrotic Disease. Nat. Med. 2012, 18, 1028–1040. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  6. Theocharis, A.D.; Manou, D.; Karamanos, N.K. The Extracellular Matrix as a Multitasking Player in Disease. FEBS J. 2019, 286, 2830–2869. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  7. Klingberg, F.; Hinz, B.; White, E.S. The Myofibroblast Matrix: Implications for Tissue Repair and Fibrosis. J. Pathol. 2013, 229, 298. [Google Scholar] [CrossRef] [Green Version]
  8. Wynn, T.A. Cellular and Molecular Mechanisms of Fibrosis. J. Pathol. 2008, 214, 199–210. [Google Scholar] [CrossRef] [Green Version]
  9. Frantz, C.; Stewart, K.M.; Weaver, V.M. The Extracellular Matrix at a Glance. J. Cell Sci. 2010, 123, 4195. [Google Scholar] [CrossRef] [Green Version]
  10. Hinz, B.; Phan, S.H.; Thannickal, V.J.; Galli, A.; Bochaton-Piallat, M.L.; Gabbiani, G. The Myofibroblast: One Function, Multiple Origins. Am. J. Pathol. 2007, 170, 1807–1816. [Google Scholar] [CrossRef]
  11. Darby, I.A.; Laverdet, B.; Bonté, F.; Desmoulière, A. Fibroblasts and Myofibroblasts in Wound Healing. Clin. Cosmet. Investig. Dermatol. 2014, 7, 301–311. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  12. Petrov, V.V.; Fagard, R.H.; Lijnen, P.J. Stimulation of Collagen Production by Transforming Growth Factor-Beta1 during Differentiation of Cardiac Fibroblasts to Myofibroblasts. Hypertension 2002, 39, 258–263. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  13. Gibb, A.A.; Lazaropoulos, M.P.; Elrod, J.W. Myofibroblasts and Fibrosis: Mitochondrial and Metabolic Control of Cellular Differentiation. Circ. Res. 2020, 127, 427–447. [Google Scholar] [CrossRef] [PubMed]
  14. Hinz, B. Formation and Function of the Myofibroblast during Tissue Repair. J. Investig. Dermatol. 2007, 127, 526–537. [Google Scholar] [CrossRef] [PubMed]
  15. Baum, J.; Duffy, H.S. Fibroblasts and Myofibroblasts: What Are We Talking About? J. Cardiovasc. Pharmacol. 2011, 57, 376. [Google Scholar] [CrossRef]
  16. Fontani, F.; Domazetovic, V.; Marcucci, T.; Vincenzini, M.T.; Iantomasi, T. Tumor Necrosis Factor-Alpha Up-Regulates ICAM-1 Expression and Release in Intestinal Myofibroblasts by Redox-Dependent and -Independent Mechanisms. J. Cell. Biochem. 2016, 117, 370–381. [Google Scholar] [CrossRef] [Green Version]
  17. Domazetovic, V.; Bonanomi, A.G.; Stio, M.; Vincenzini, M.T.; Iantomasi, T. Resveratrol Decreases TNFα-Induced ICAM-1 Expression and Release by Sirt-1-Independent Mechanism in Intestinal Myofibroblasts. Exp. Cell Res. 2019, 382, 111479. [Google Scholar] [CrossRef]
  18. Hinz, B.; Phan, S.H.; Thannickal, V.J.; Prunotto, M.; Desmoulire, A.; Varga, J.; De Wever, O.; Mareel, M.; Gabbiani, G. Recent Developments in Myofibroblast Biology: Paradigms for Connective Tissue Remodeling. Am. J. Pathol. 2012, 180, 1340–1355. [Google Scholar] [CrossRef]
  19. Kida, Y.; Duffield, J.S. Pivotal Role of Pericytes in Kidney Fibrosis. Clin. Exp. Pharmacol. Physiol. 2011, 38, 467–473. [Google Scholar] [CrossRef] [Green Version]
  20. Di Carlo, S.E.; Peduto, L. The Perivascular Origin of Pathological Fibroblasts. J. Clin. Investig. 2018, 128, 54–63. [Google Scholar] [CrossRef]
  21. Lombardi, A.A.; Gibb, A.A.; Arif, E.; Kolmetzky, D.W.; Tomar, D.; Luongo, T.S.; Jadiya, P.; Murray, E.K.; Lorkiewicz, P.K.; Hajnóczky, G.; et al. Mitochondrial Calcium Exchange Links Metabolism with the Epigenome to Control Cellular Differentiation. Nat. Commun. 2019, 10, 4509. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  22. Jain, M.; Rivera, S.; Monclus, E.A.; Synenki, L.; Zirk, A.; Eisenbart, J.; Feghali-Bostwick, C.; Mutlu, G.M.; Budinger, G.R.S.; Chandel, N.S. Mitochondrial Reactive Oxygen Species Regulate Transforming Growth Factor-β Signaling. J. Biol. Chem. 2013, 288, 770–777. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  23. Tomasek, J.J.; Gabbiani, G.; Hinz, B.; Chaponnier, C.; Brown, R.A. Myofibroblasts and Mechano-Regulation of Connective Tissue Remodelling. Nat. Rev. Mol. Cell Biol. 2002, 3, 349–363. [Google Scholar] [CrossRef] [PubMed]
  24. Stempien-Otero, A.; Kim, D.H.; Davis, J. Molecular Networks Underlying Myofibroblast Fate and Fibrosis. J. Mol. Cell. Cardiol. 2016, 97, 153–161. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  25. Xu, H.; Huang, L.; Zhao, J.; Chen, S.; Liu, J.; Li, G. The Circadian Clock and Inflammation: A New Insight. Clin. Chim. Acta. 2021, 512, 12–17. [Google Scholar] [CrossRef] [PubMed]
  26. Dunlap, J.C. Molecular Bases for Circadian Clocks. Cell 1999, 96, 271–290. [Google Scholar] [CrossRef] [Green Version]
  27. Mitsui, A.; Kumazawa, S.; Takahashi, A.; Ikemoto, H.; Cao, S.; Arai, T. Strategy by Which Nitrogen-Fixing Unicellular Cyanobacteria Grow Photoautotrophically. Nature 1986, 323, 720–722. [Google Scholar] [CrossRef]
  28. Vollmers, C.; Gill, S.; DiTacchio, L.; Pulivarthy, S.R.; Le, H.D.; Panda, S. Time of Feeding and the Intrinsic Circadian Clock Drive Rhythms in Hepatic Gene Expression. Proc. Natl. Acad. Sci. USA 2009, 106, 21453–21458. [Google Scholar] [CrossRef] [Green Version]
  29. Bass, J.; Takahashi, J.S. Circadian Integration of Metabolism and Energetics. Science 2010, 330, 1349–1354. [Google Scholar] [CrossRef] [Green Version]
  30. Vetter, C.; Dashti, H.S.; Lane, J.M.; Anderson, S.G.; Schernhammer, E.S.; Rutter, M.K.; Saxena, R.; Scheer, F.A.J.L. Night Shift Work, Genetic Risk, and Type 2 Diabetes in the UK Biobank. Diabetes Care 2018, 41, 762–769. [Google Scholar] [CrossRef]
  31. Kim, H.J.; Choi, S.; Kim, K.; Park, H.; Kim, K.H.; Park, S.M. Association between Misalignment of Circadian Rhythm and Obesity in Korean Men: Sixth Korea National Health and Nutrition Examination Survey. Chronobiol. Int. 2020, 37, 272–280. [Google Scholar] [CrossRef] [PubMed]
  32. Man, K.; Loudon, A.; Chawla, A. Immunity around the Clock. Science 2016, 354, 999–1003. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  33. Dibner, C.; Schibler, U. Circadian Timing of Metabolism in Animal Models and Humans. J. Intern. Med. 2015, 277, 513–527. [Google Scholar] [CrossRef] [PubMed]
  34. Tahara, Y.; Aoyama, S.; Shibata, S. The Mammalian Circadian Clock and Its Entrainment by Stress and Exercise. J. Physiol. Sci. 2017, 67, 1–10. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  35. Scott, E.M.; Carter, A.M.; Grant, P.J. Association between Polymorphisms in the Clock Gene, Obesity and the Metabolic Syndrome in Man. Int. J. Obes. 2008, 32, 658–662. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  36. Woon, P.Y.; Kaisaki, P.J.; Bragança, J.; Bihoreau, M.T.; Levy, J.C.; Farrall, M.; Gauguier, D. Aryl Hydrocarbon Receptor Nuclear Translocator-like (BMAL1) Is Associated with Susceptibility to Hypertension and Type 2 Diabetes. Proc. Natl. Acad. Sci. USA 2007, 104, 14412–14417. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  37. Ruben, M.D.; Wu, G.; Smith, D.F.; Schmidt, R.E.; Francey, L.J.; Lee, Y.Y.; Anafi, R.C.; Hogenesch, J.B. A Database of Tissue-Specific Rhythmically Expressed Human Genes Has Potential Applications in Circadian Medicine. Sci. Transl. Med. 2018, 10, eaat8806. [Google Scholar] [CrossRef]
  38. Chaix, A.; Zarrinpar, A.; Panda, S. The Circadian Coordination of Cell Biology. J. Cell Biol. 2016, 215, 15–25. [Google Scholar] [CrossRef] [Green Version]
  39. Peek, C.B.; Levine, D.C.; Cedernaes, J.; Taguchi, A.; Kobayashi, Y.; Tsai, S.J.; Bonar, N.A.; McNulty, M.R.; Ramsey, K.M.; Bass, J. Circadian Clock Interaction with HIF1α Mediates Oxygenic Metabolism and Anaerobic Glycolysis in Skeletal Muscle. Cell Metab. 2017, 25, 86–92. [Google Scholar] [CrossRef] [Green Version]
  40. Liu, C.; Li, S.; Liu, T.; Borjigin, J.; Lin, J.D. Transcriptional Coactivator PGC-1alpha Integrates the Mammalian Clock and Energy Metabolism. Nature 2007, 447, 477–481. [Google Scholar] [CrossRef]
  41. Jee, H.U.; Yang, S.; Yamazaki, S.; Kang, H.; Viollet, B.; Foretz, M.; Chung, J.H. Activation of 5’-AMP-Activated Kinase with Diabetes Drug Metformin Induces Casein Kinase Iepsilon (CKIepsilon)-Dependent Degradation of Clock Protein MPer2. J. Biol. Chem. 2007, 282, 20794–20798. [Google Scholar] [CrossRef] [Green Version]
  42. Nakahata, Y.; Sahar, S.; Astarita, G.; Kaluzova, M.; Sassone-Corsi, P. Circadian Control of the NAD+ Salvage Pathway by CLOCK-SIRT1. Science 2009, 324, 654–657. [Google Scholar] [CrossRef] [PubMed]
  43. Coogan, A.N.; Wyse, C.A. Neuroimmunology of the Circadian Clock. Brain Res. 2008, 1232, 104–112. [Google Scholar] [CrossRef] [PubMed]
  44. Hayashi, M.; Shimba, S.; Tezuka, M. Characterization of the Molecular Clock in Mouse Peritoneal Macrophages. Biol. Pharm. Bull. 2007, 30, 621–626. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  45. Hui, L.; Hua, F.; Diandong, H.; Hong, Y. Effects of Sleep and Sleep Deprivation on Immunoglobulins and Complement in Humans. Brain. Behav. Immun. 2007, 21, 308–310. [Google Scholar] [CrossRef]
  46. Pekovic-Vaughan, V.; Gibbs, J.; Yoshitane, H.; Yang, N.; Pathiranage, D.; Guo, B.; Sagami, A.; Taguchi, K.; Bechtold, D.; Loudon, A.; et al. The Circadian Clock Regulates Rhythmic Activation of the NRF2/Glutathione-Mediated Antioxidant Defense Pathway to Modulate Pulmonary Fibrosis. Genes Dev. 2014, 28, 548–560. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  47. Wang, N.; Yang, G.; Jia, Z.; Zhang, H.; Aoyagi, T.; Soodvilai, S.; Symons, J.D.; Schnermann, J.B.; Gonzalez, F.J.; Litwin, S.E.; et al. Vascular PPARgamma Controls Circadian Variation in Blood Pressure and Heart Rate through Bmal1. Cell Metab. 2008, 8, 482–491. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  48. Baker, R.G.; Hayden, M.S.; Ghosh, S. NF-ΚB, Inflammation, and Metabolic Disease. Cell Metab. 2011, 13, 11–22. [Google Scholar] [CrossRef] [Green Version]
  49. Kojetin, D.J.; Burris, T.P. REV-ERB and ROR Nuclear Receptors as Drug Targets. Nat. Rev. Drug Discov. 2014, 13, 197–216. [Google Scholar] [CrossRef] [Green Version]
  50. Amir, M.; Chaudhari, S.; Wang, R.; Campbell, S.; Mosure, S.A.; Chopp, L.B.; Lu, Q.; Shang, J.; Pelletier, O.B.; He, Y.; et al. REV-ERBα Regulates T H 17 Cell Development and Autoimmunity. Cell Rep. 2018, 25, 3733–3749.e8. [Google Scholar] [CrossRef]
  51. Burke, L.; Downes, M.; Carozzi, A.; Giguère, V.; Muscat, G.E.O. Transcriptional Repression by the Orphan Steroid Receptor RVR/Rev-Erb Beta Is Dependent on the Signature Motif and Helix 5 in the E Region: Functional Evidence for a Biological Role of RVR in Myogenesis. Nucleic Acids Res. 1996, 24, 3481–3489. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  52. Yin, L.; Lazar, M.A. The Orphan Nuclear Receptor Rev-Erbalpha Recruits the N-CoR/Histone Deacetylase 3 Corepressor to Regulate the Circadian Bmal1 Gene. Mol. Endocrinol. 2005, 19, 1452–1459. [Google Scholar] [CrossRef] [PubMed]
  53. Preitner, N.; Damiola, F.; Luis-Lopez-Molina; Zakany, J.; Duboule, D.; Albrecht, U.; Schibler, U. The Orphan Nuclear Receptor REV-ERBalpha Controls Circadian Transcription within the Positive Limb of the Mammalian Circadian Oscillator. Cell 2002, 110, 251–260. [Google Scholar] [CrossRef]
  54. Zhang, Y.; Fang, B.; Emmett, M.J.; Damle, M.; Sun, Z.; Feng, D.; Armour, S.M.; Remsberg, J.R.; Jager, J.; Soccio, R.E.; et al. GENE REGULATION. Discrete Functions of Nuclear Receptor Rev-Erbα Couple Metabolism to the Clock. Science 2015, 348, 1488–1492. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  55. Feng, D.; Liu, T.; Sun, Z.; Bugge, A.; Mullican, S.E.; Alenghat, T.; Liu, X.S.; Lazar, M.A. A Circadian Rhythm Orchestrated by Histone Deacetylase 3 Controls Hepatic Lipid Metabolism. Science 2011, 331, 1315–1319. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  56. Yin, L.; Wu, N.; Curtin, J.C.; Qatanani, M.; Szwergold, N.R.; Reid, R.A.; Waitt, G.M.; Parks, D.J.; Pearce, K.H.; Wisely, G.B.; et al. Rev-Erbalpha, a Heme Sensor That Coordinates Metabolic and Circadian Pathways. Science 2007, 318, 1786–1789. [Google Scholar] [CrossRef]
  57. Gerhart-Hines, Z.; Lazar, M.A. Rev-Erbα and the Circadian Transcriptional Regulation of Metabolism. Diabetes. Obes. Metab. 2015, 17 (Suppl. S1), 12–16. [Google Scholar] [CrossRef] [Green Version]
  58. Solt, L.A.; Wang, Y.; Banerjee, S.; Hughes, T.; Kojetin, D.J.; Lundasen, T.; Shin, Y.; Liu, J.; Cameron, M.D.; Noel, R.; et al. Regulation of Circadian Behaviour and Metabolism by Synthetic REV-ERB Agonists. Nature 2012, 485, 62–68. [Google Scholar] [CrossRef] [Green Version]
  59. Woldt, E.; Sebti, Y.; Solt, L.A.; Duhem, C.; Lancel, S.; Eeckhoute, J.; Hesselink, M.K.C.; Paquet, C.; Delhaye, S.; Shin, Y.; et al. Rev-Erb-α Modulates Skeletal Muscle Oxidative Capacity by Regulating Mitochondrial Biogenesis and Autophagy. Nat. Med. 2013, 19, 1039–1046. [Google Scholar] [CrossRef]
  60. Sulli, G.; Rommel, A.; Wang, X.; Kolar, M.J.; Puca, F.; Saghatelian, A.; Plikus, M.V.; Verma, I.M.; Panda, S. Pharmacological Activation of REV-ERBs Is Lethal in Cancer and Oncogene-Induced Senescence. Nature 2018, 553, 351–355. [Google Scholar] [CrossRef]
  61. Banerjee, S.; Wang, Y.; Solt, L.A.; Griffett, K.; Kazantzis, M.; Amador, A.; El-Gendy, B.M.; Huitron-Resendiz, S.; Roberts, A.J.; Shin, Y.; et al. Pharmacological Targeting of the Mammalian Clock Regulates Sleep Architecture and Emotional Behaviour. Nat. Commun. 2014, 5, 5759. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  62. Gibbs, J.E.; Blaikley, J.; Beesley, S.; Matthews, L.; Simpson, K.D.; Boyce, S.H.; Farrow, S.N.; Else, K.J.; Singh, D.; Ray, D.W.; 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, 582–587. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  63. Dierickx, P.; Emmett, M.J.; Jiang, C.; Uehara, K.; Liu, M.; Adlanmerini, M.; Lazar, M.A. SR9009 Has REV-ERB-Independent Effects on Cell Proliferation and Metabolism. Proc. Natl. Acad. Sci. USA 2019, 116, 12147–12152. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  64. Rockey, D.C.; Bell, P.D.; Hill, J.A. Fibrosis—A Common Pathway to Organ Injury and Failure. N. Engl. J. Med. 2015, 372, 1138–1149. [Google Scholar] [CrossRef] [PubMed]
  65. Hasegawa, Y.; Ikeda, K.; Chen, Y.; Alba, D.L.; Stifler, D.; Shinoda, K.; Hosono, T.; Maretich, P.; Yang, Y.; Ishigaki, Y.; et al. Repression of Adipose Tissue Fibrosis through a PRDM16-GTF2IRD1 Complex Improves Systemic Glucose Homeostasis. Cell Metab. 2018, 27, 180–194.e6. [Google Scholar] [CrossRef] [Green Version]
  66. Gorelik, L.; Flavell, R.A. Transforming Growth Factor-Beta in T-Cell Biology. Nat. Rev. Immunol. 2002, 2, 46–53. [Google Scholar] [CrossRef]
  67. Munger, J.S.; Huang, X.; Kawakatsu, H.; Griffiths, M.J.D.; Dalton, S.L.; Wu, J.; Pittet, J.F.; Kaminski, N.; Garat, C.; Matthay, M.A.; et al. The Integrin Alpha v Beta 6 Binds and Activates Latent TGF Beta 1: A Mechanism for Regulating Pulmonary Inflammation and Fibrosis. Cell 1999, 96, 319–328. [Google Scholar] [CrossRef] [Green Version]
  68. Lopez, M.; Meier, D.; Müller, A.; Franken, P.; Fujita, J.; Fontana, A. Tumor Necrosis Factor and Transforming Growth Factor β Regulate Clock Genes by Controlling the Expression of the Cold Inducible RNA-Binding Protein (CIRBP). J. Biol. Chem. 2014, 289, 2736–2744. [Google Scholar] [CrossRef] [Green Version]
  69. Ertosun, M.G.; Kocak, G.; Ozes, O.N. The Regulation of Circadian Clock by Tumor Necrosis Factor Alpha. Cytokine Growth Factor Rev. 2019, 46, 10–16. [Google Scholar] [CrossRef]
  70. Gast, H.; Gordic, S.; Petrzilka, S.; Lopez, M.; Müller, A.; Gietl, A.; Hock, C.; Birchler, T.; Fontana, A. Transforming Growth Factor-Beta Inhibits the Expression of Clock Genes. Ann. N. Y. Acad. Sci. 2012, 1261, 79–87. [Google Scholar] [CrossRef]
  71. Kon, N.; Hirota, T.; Kawamoto, T.; Kato, Y.; Tsubota, T.; Fukada, Y. Activation of TGF-Beta/Activin Signalling Resets the Circadian Clock through Rapid Induction of Dec1 Transcripts. Nat. Cell Biol. 2008, 10, 1463–1469. [Google Scholar] [CrossRef] [PubMed]
  72. Sato, F.; Sato, H.; Jin, D.; Bhawal, U.K.; Wu, Y.; Noshiro, M.; Kawamoto, T.; Fujimoto, K.; Seino, H.; Morohashi, S.; et al. Smad3 and Snail Show Circadian Expression in Human Gingival Fibroblasts, Human Mesenchymal Stem Cell, and in Mouse Liver. Biochem. Biophys. Res. Commun. 2012, 419, 441–446. [Google Scholar] [CrossRef] [PubMed]
  73. Roth, K.J.; Copple, B.L. Role of Hypoxia-Inducible Factors in the Development of Liver Fibrosis. Cell. Mol. Gastroenterol. Hepatol. 2015, 1, 589–597. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  74. Imtiyaz, H.Z.; Williams, E.P.; Hickey, M.M.; Patel, S.A.; Durham, A.C.; Yuan, L.J.; Hammond, R.; Gimotty, P.A.; Keith, B.; Simon, M.C. Hypoxia-Inducible Factor 2alpha Regulates Macrophage Function in Mouse Models of Acute and Tumor Inflammation. J. Clin. Investig. 2010, 120, 2699–2714. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  75. Hu, F.; Liu, H.; Xu, L.; Li, Y.; Liu, X.; Shi, L.; Su, Y.; Qiu, X.; Zhang, X.; Yang, Y.; et al. Hypoxia-Inducible Factor-1α Perpetuates Synovial Fibroblast Interactions with T Cells and B Cells in Rheumatoid Arthritis. Eur. J. Immunol. 2016, 46, 742–751. [Google Scholar] [CrossRef] [Green Version]
  76. Manella, G.; Aviram, R.; Bolshette, N.; Muvkadi, S.; Golik, M.; Smith, D.F.; Asher, G. Hypoxia Induces a Time- And Tissue-Specific Response That Elicits Intertissue Circadian Clock Misalignment. Proc. Natl. Acad. Sci. USA 2020, 117, 779–786. [Google Scholar] [CrossRef]
  77. Wu, Y.; Tang, D.; Liu, N.; Xiong, W.; Huang, H.; Li, Y.; Ma, Z.; Zhao, H.; Chen, P.; Qi, X.; et al. Reciprocal Regulation between the Circadian Clock and Hypoxia Signaling at the Genome Level in Mammals. Cell Metab. 2017, 25, 73–85. [Google Scholar] [CrossRef] [Green Version]
  78. Kobayashi, M.; Morinibu, A.; Koyasu, S.; Goto, Y.; Hiraoka, M.; Harada, H. A Circadian Clock Gene, PER2, Activates HIF-1 as an Effector Molecule for Recruitment of HIF-1α to Promoter Regions of Its Downstream Genes. FEBS J. 2017, 284, 3804–3816. [Google Scholar] [CrossRef] [Green Version]
  79. Dimova, E.Y.; Jakupovic, M.; Kubaichuk, K.; Mennerich, D.; Chi, T.F.; Tamanini, F.; Oklejewicz, M.; Hänig, J.; Byts, N.; Mäkelä, K.A.; et al. The Circadian Clock Protein CRY1 Is a Negative Regulator of HIF-1α. iScience 2019, 13, 284–304. [Google Scholar] [CrossRef] [Green Version]
  80. Henderson, N.C.; Arnold, T.D.; Katamura, Y.; Giacomini, M.M.; Rodriguez, J.D.; McCarty, J.H.; Pellicoro, A.; Raschperger, E.; Betsholtz, C.; Ruminski, P.G.; et al. Targeting of Av Integrin Identifies a Core Molecular Pathway That Regulates Fibrosis in Several Organs. Nat. Med. 2013, 19, 1617–1624. [Google Scholar] [CrossRef]
  81. Hewlett, J.C.; Kropski, J.A.; Blackwell, T.S. Idiopathic Pulmonary Fibrosis: Epithelial-Mesenchymal Interactions and Emerging Therapeutic Targets. Matrix Biol. 2018, 71–72, 112–127. [Google Scholar] [CrossRef] [PubMed]
  82. Vannella, K.M.; Ramalingam, T.R.; Borthwick, L.A.; Barron, L.; Hart, K.M.; Thompson, R.W.; Kindrachuk, K.N.; Cheever, A.W.; White, S.; Budelsky, A.L.; et al. Combinatorial Targeting of TSLP, IL-25, and IL-33 in Type 2 Cytokine-Driven Inflammation and Fibrosis. Sci. Transl. Med. 2016, 8, 337ra65. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  83. Gieseck, R.L.; Wilson, M.S.; Wynn, T.A. Type 2 Immunity in Tissue Repair and Fibrosis. Nat. Rev. Immunol. 2018, 18, 62–76. [Google Scholar] [CrossRef]
  84. Park, M.J.; Moon, S.J.; Lee, E.J.; Jung, K.A.; Kim, E.K.; Kim, D.S.; Lee, J.H.; Kwok, S.K.; Min, J.K.; Park, S.H.; et al. IL-1-IL-17 Signaling Axis Contributes to Fibrosis and Inflammation in Two Different Murine Models of Systemic Sclerosis. Front. Immunol. 2018, 9, 1611. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  85. Sun, B.; Wang, H.; Zhang, L.; Yang, X.; Zhang, M.; Zhu, X.; Ji, X.; Wang, H. Role of Interleukin 17 in TGF-β Signaling-Mediated Renal Interstitial Fibrosis. Cytokine 2018, 106, 80–88. [Google Scholar] [CrossRef] [PubMed]
  86. James, S.L.; Abate, D.; Abate, K.H.; Abay, S.M.; Abbafati, C.; Abbasi, N.; Abbastabar, H.; Abd-Allah, F.; Abdela, J.; Abdelalim, A.; et al. Global, Regional, and National Incidence, Prevalence, and Years Lived with Disability for 354 Diseases and Injuries for 195 Countries and Territories, 1990-2017: A Systematic Analysis for the Global Burden of Disease Study 2017. Lancet 2018, 392, 1789–1858. [Google Scholar] [CrossRef] [Green Version]
  87. Groenewegen, A.; Rutten, F.H.; Mosterd, A.; Hoes, A.W. Epidemiology of Heart Failure. Eur. J. Heart Fail. 2020, 22, 1342–1356. [Google Scholar] [CrossRef]
  88. Kwak, H.-B. Aging, Exercise, and Extracellular Matrix in the Heart. J. Exerc. Rehabil. 2013, 9, 338–347. [Google Scholar] [CrossRef]
  89. Kong, P.; Christia, P.; Frangogiannis, N.G. The Pathogenesis of Cardiac Fibrosis. Cell. Mol. Life Sci. 2014, 71, 549–574. [Google Scholar] [CrossRef] [Green Version]
  90. Leibetseder, V.; Humpeler, S.; Svoboda, M.; Schmid, D.; Thalhammer, T.; Zuckermann, A.; Marktl, W.; Ekmekcioglu, C. Clock Genes Display Rhythmic Expression in Human Hearts. Chronobiol. Int. 2009, 26, 621–636. [Google Scholar] [CrossRef]
  91. Young, M.E.; Razeghi, P.; Taegtmeyer, H. Clock Genes in the Heart: Characterization and Attenuation with Hypertrophy. Circ. Res. 2001, 88, 1142–1150. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  92. Lefta, M.; Campbell, K.S.; Feng, H.Z.; Jin, J.P.; Esser, K.A. Development of Dilated Cardiomyopathy in Bmal1-Deficient Mice. Am. J. Physiol. Heart Circ. Physiol. 2012, 303, H475–H485. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  93. Young, M.E.; Brewer, R.A.; Peliciari-Garcia, R.A.; Collins, H.E.; He, L.; Birky, T.L.; Peden, B.W.; Thompson, E.G.; Ammons, B.J.; Bray, M.S.; et al. Cardiomyocyte-Specific BMAL1 Plays Critical Roles in Metabolism, Signaling, and Maintenance of Contractile Function of the Heart. J. Biol. Rhythms 2014, 29, 257–276. [Google Scholar] [CrossRef] [PubMed]
  94. Alibhai, F.J.; LaMarre, J.; Reitz, C.J.; Tsimakouridze, E.V.; Kroetsch, J.T.; Bolz, S.S.; Shulman, A.; Steinberg, S.; Burris, T.P.; Oudit, G.Y.; et al. Disrupting the Key Circadian Regulator CLOCK Leads to Age-Dependent Cardiovascular Disease. J. Mol. Cell. Cardiol. 2017, 105, 24–37. [Google Scholar] [CrossRef] [Green Version]
  95. Wang, N.; Sun, Y.; Zhang, H.; Wang, B.; Chen, C.; Wang, Y.; Chen, J.; Tan, X.; Zhang, J.; Xia, F.; et al. Long-Term Night Shift Work Is Associated with the Risk of Atrial Fibrillation and Coronary Heart Disease. Eur. Heart J. 2021, 42, 4180–4188. [Google Scholar] [CrossRef]
  96. Zhang, L.; Zhang, R.; Tien, C.L.; Chan, R.E.; Sugi, K.; Fu, C.; Griffin, A.C.; Shen, Y.; Burris, T.P.; Liao, X.; et al. REV-ERBα Ameliorates Heart Failure through Transcription Repression. JCI Insight 2017, 2, e95177. [Google Scholar] [CrossRef] [Green Version]
  97. Stujanna, E.N.; Murakoshi, N.; Tajiri, K.; Xu, D.; Kimura, T.; Qin, R.; Feng, D.; Yonebayashi, S.; Ogura, Y.; Yamagami, F.; et al. Rev-Erb Agonist Improves Adverse Cardiac Remodeling and Survival in Myocardial Infarction through an Anti-Inflammatory Mechanism. PLoS ONE 2017, 12, e0189330. [Google Scholar] [CrossRef] [Green Version]
  98. Sitaula, S.; Billon, C.; Kamenecka, T.M.; Solt, L.A.; Burris, T.P. Suppression of Atherosclerosis by Synthetic REV-ERB Agonist. Biochem. Biophys. Res. Commun. 2015, 460, 566–571. [Google Scholar] [CrossRef] [Green Version]
  99. Cunningham, P.S.; Meijer, P.; Nazgiewicz, A.; Anderson, S.G.; Borthwick, L.A.; Bagnall, J.; Kitchen, G.B.; Lodyga, M.; Begley, N.; Venkateswaran, R.V.; et al. The Circadian Clock Protein REVERBα Inhibits Pulmonary Fibrosis Development. Proc. Natl. Acad. Sci. USA 2020, 117, 1139–1147. [Google Scholar] [CrossRef] [Green Version]
  100. Sundar, I.K.; Rashid, K.; Sellix, M.T.; Rahman, I. The Nuclear Receptor and Clock Gene REV-ERBα Regulates Cigarette Smoke-Induced Lung Inflammation. Biochem. Biophys. Res. Commun. 2017, 493, 1390–1395. [Google Scholar] [CrossRef]
  101. Yao, H.; Sundar, I.K.; Huang, Y.; Gerloff, J.; Sellix, M.T.; Sime, P.J.; Rahman, I. Disruption of Sirtuin 1-Mediated Control of Circadian Molecular Clock and Inflammation in Chronic Obstructive Pulmonary Disease. Am. J. Respir. Cell Mol. Biol. 2015, 53, 782–792. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  102. Hwang, J.W.; Sundar, I.K.; Yao, H.; Sellix, M.T.; Rahman, I. Circadian Clock Function Is Disrupted by Environmental Tobacco/Cigarette Smoke, Leading to Lung Inflammation and Injury via a SIRT1-BMAL1 Pathway. FASEB J. 2014, 28, 176–194. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  103. Raspé, E.; Duez, H.; Mansén, A.; Fontaine, C.; Fiévet, C.; Fruchart, J.C.; Vennström, B.; Staels, B. Identification of Rev-Erbα as a Physiological Repressor of ApoC-III Gene Transcription. J. Lipid Res. 2002, 43, 2172–2179. [Google Scholar] [CrossRef] [Green Version]
  104. Duez, H.; van der Veen, J.N.; Duhem, C.; Pourcet, B.; Touvier, T.; Fontaine, C.; Derudas, B.; Baugé, E.; Havinga, R.; Bloks, V.W.; et al. Regulation of Bile Acid Synthesis by the Nuclear Receptor Rev-Erbalpha. Gastroenterology 2008, 135, 689–698. [Google Scholar] [CrossRef] [PubMed]
  105. Guan, D.; Xiong, Y.; Trinh, T.M.; Xiao, Y.; Hu, W.; Jiang, C.; Dierickx, P.; Jang, C.; Rabinowitz, J.D.; Lazar, M.A. The Hepatocyte Clock and Feeding Control Chronophysiology of Multiple Liver Cell Types. Science 2020, 369, 1388–1395. [Google Scholar] [CrossRef] [PubMed]
  106. Yang, Z.; Smalling, R.V.; Huang, Y.; Jiang, Y.; Kusumanchi, P.; Bogaert, W.; Wang, L.; Delker, D.A.; Skill, N.J.; Han, S.; et al. The Role of SHP/REV-ERBα/CYP4A Axis in the Pathogenesis of Alcohol-Associated Liver Disease. JCI Insight 2021, 6, e140687. [Google Scholar] [CrossRef]
  107. Hand, L.E.; Usan, P.; Cooper, G.J.S.; Xu, L.Y.; Ammori, B.; Cunningham, P.S.; Aghamohammadzadeh, R.; Soran, H.; Greenstein, A.; Loudon, A.S.I.; et al. Adiponectin Induces A20 Expression in Adipose Tissue to Confer Metabolic Benefit. Diabetes 2015, 64, 128–136. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  108. Delezie, J.; Dumont, S.; Dardente, H.; Oudart, H.; Gréchez-Cassiau, A.; Klosen, P.; Teboul, M.; Delaunay, F.; Pévet, P.; Challet, E. The Nuclear Receptor REV-ERBα Is Required for the Daily Balance of Carbohydrate and Lipid Metabolism. FASEB J. 2012, 26, 3321–3335. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  109. Cho, H.; Zhao, X.; Hatori, M.; Yu, R.T.; Barish, G.D.; Lam, M.T.; Chong, L.W.; Ditacchio, L.; Atkins, A.R.; Glass, C.K.; et al. Regulation of Circadian Behaviour and Metabolism by REV-ERB-α and REV-ERB-β. Nature 2012, 485, 123–127. [Google Scholar] [CrossRef] [Green Version]
  110. Mayeuf-Louchart, A.; Thorel, Q.; Delhaye, S.; Beauchamp, J.; Duhem, C.; Danckaert, A.; Lancel, S.; Pourcet, B.; Woldt, E.; Boulinguiez, A.; et al. Rev-Erb-α Regulates Atrophy-Related Genes to Control Skeletal Muscle Mass. Sci. Rep. 2017, 7, 14383. [Google Scholar] [CrossRef]
  111. Welch, R.D.; Guo, C.; Sengupta, M.; Carpenter, K.J.; Stephens, N.A.; Arnett, S.A.; Meyers, M.J.; Sparks, L.M.; Smith, S.R.; Zhang, J.; et al. Rev-Erb Co-Regulates Muscle Regeneration via Tethered Interaction with the NF-Y Cistrome. Mol. Metab. 2017, 6, 703–714. [Google Scholar] [CrossRef] [PubMed]
  112. Guo, L.; Zhang, T.; Wang, F.; Chen, X.; Xu, H.; Zhou, C.; Chen, M.; Yu, F.; Wang, S.; Yang, D.; et al. Targeted Inhibition of Rev-Erb-α/β Limits Ferroptosis to Ameliorate Folic Acid-Induced Acute Kidney Injury. Br. J. Pharmacol. 2021, 178, 328–345. [Google Scholar] [CrossRef] [PubMed]
  113. Zhang, X.; Li, S.; Zhou, Y.; Su, W.; Ruan, X.; Wang, B.; Zheng, F.; Warner, M.; Gustafsson, J.Å.; Guan, Y. Ablation of Cytochrome P450 Omega-Hydroxylase 4A14 Gene Attenuates Hepatic Steatosis and Fibrosis. Proc. Natl. Acad. Sci. USA 2017, 114, 3181–3185. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  114. Pariollaud, M.; Gibbs, J.E.; Hopwood, T.W.; Brown, S.; Begley, N.; Vonslow, R.; Poolman, T.; Guo, B.; Saer, B.; Jones, D.H.; et al. Circadian Clock Component REV-ERBα Controls Homeostatic Regulation of Pulmonary Inflammation. J. Clin. Investig. 2018, 128, 2281–2296. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  115. Wang, Q.; Sundar, I.K.; Lucas, J.H.; Muthumalage, T.; Rahman, I. Molecular Clock REV-ERBα Regulates Cigarette Smoke-Induced Pulmonary Inflammation and Epithelial-Mesenchymal Transition. JCI Insight 2021, 6, e145200. [Google Scholar] [CrossRef]
  116. Sitaula, S.; Zhang, J.; Ruiz, F.; Burris, T.P. Rev-Erb Regulation of Cholesterologenesis. Biochem. Pharmacol. 2017, 131, 68–77. [Google Scholar] [CrossRef]
  117. Thomes, P.G.; Brandon-Warner, E.; Li, T.; Donohue, T.M.; Schrum, L.W. Rev-Erb Agonist and TGF-β Similarly Affect Autophagy but Differentially Regulate Hepatic Stellate Cell Fibrogenic Phenotype. Int. J. Biochem. Cell Biol. 2016, 81, 137–147. [Google Scholar] [CrossRef]
  118. Griffett, K.; Bedia-Diaz, G.; Elgendy, B.; Burris, T.P. REV-ERB Agonism Improves Liver Pathology in a Mouse Model of NASH. PLoS ONE 2020, 15, e0236000. [Google Scholar] [CrossRef]
  119. Feng, G.; Li, X.P.; Niu, C.Y.; Liu, M.L.; Yan, Q.Q.; Fan, L.P.; Li, Y.; Zhang, K.L.; Gao, J.; Qian, M.R.; et al. Bioinformatics Analysis Reveals Novel Core Genes Associated with Nonalcoholic Fatty Liver Disease and Nonalcoholic Steatohepatitis. Gene 2020, 742, 144549. [Google Scholar] [CrossRef]
  120. González-Fernández, B.; Sánchez, D.I.; Crespo, I.; San-Miguel, B.; de Urbina, J.O.; González-Gallego, J.; Tuñón, M.J. Melatonin Attenuates Dysregulation of the Circadian Clock Pathway in Mice With CCl4-Induced Fibrosis and Human Hepatic Stellate Cells. Front. Pharmacol. 2018, 9, 556. [Google Scholar] [CrossRef]
  121. Li, T.; Eheim, A.L.; Klein, S.; Uschner, F.E.; Smith, A.C.; Brandon-Warner, E.; Ghosh, S.; Bonkovsky, H.L.; Trebicka, J.; Schrum, L.W. Novel Role of Nuclear Receptor Rev-Erbα in Hepatic Stellate Cell Activation: Potential Therapeutic Target for Liver Injury. Hepatology 2014, 59, 2383–2396. [Google Scholar] [CrossRef] [PubMed]
  122. Kumar, N.; Solt, L.A.; Wang, Y.; Rogers, P.M.; Bhattacharyya, G.; Kamenecka, T.M.; Stayrook, K.R.; Crumbley, C.; Floyd, Z.E.; Gimble, J.M.; et al. Regulation of Adipogenesis by Natural and Synthetic REV-ERB Ligands. Endocrinology 2010, 151, 3015–3025. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  123. Welch, R.D.; Billon, C.; Valfort, A.C.; Burris, T.P.; Flaveny, C.A. Pharmacological Inhibition of REV-ERB Stimulates Differentiation, Inhibits Turnover and Reduces Fibrosis in Dystrophic Muscle. Sci. Rep. 2017, 7, 1–13. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  124. Hadden, H.; Soldin, S.J.; Massaro, D. Circadian Disruption Alters Mouse Lung Clock Gene Expression and Lung Mechanics. J. Appl. Physiol. 2012, 113, 385–392. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  125. Tsai, C.L.; Brenner, B.E.; Camargo, C.A. Circadian-Rhythm Differences among Emergency Department Patients with Chronic Obstructive Pulmonary Disease Exacerbation. Chronobiol. Int. 2007, 24, 699–713. [Google Scholar] [CrossRef] [PubMed]
  126. Papagiannakopoulos, T.; Bauer, M.R.; Davidson, S.M.; Heimann, M.; Subbaraj, L.; Bhutkar, A.; Bartlebaugh, J.; Vander Heiden, M.G.; Jacks, T. Circadian Rhythm Disruption Promotes Lung Tumorigenesis. Cell Metab. 2016, 24, 324. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  127. Gibbs, J.; Ince, L.; Matthews, L.; Mei, J.; Bell, T.; Yang, N.; Saer, B.; Begley, N.; Poolman, T.; Pariollaud, M.; et al. An Epithelial Circadian Clock Controls Pulmonary Inflammation and Glucocorticoid Action. Nat. Med. 2014, 20, 919–926. [Google Scholar] [CrossRef] [Green Version]
  128. Dong, C.; Gongora, R.; Sosulski, M.L.; Luo, F.; Sanchez, C.G. Regulation of Transforming Growth Factor-Beta1 (TGF-Β1)-Induced pro-Fibrotic Activities by Circadian Clock Gene BMAL1. Respir. Res. 2016, 17, 4. [Google Scholar] [CrossRef] [Green Version]
  129. Kisseleva, T.; Brenner, D.A. Mechanisms of Fibrogenesis. Exp. Biol. Med. 2008, 233, 109–122. [Google Scholar] [CrossRef]
  130. Wallace, M.C.; Friedman, S.L.; Mann, D.A. Emerging and Disease-Specific Mechanisms of Hepatic Stellate Cell Activation. Semin. Liver Dis. 2015, 35, 107–118. [Google Scholar] [CrossRef]
  131. Mederacke, I.; Hsu, C.C.; Troeger, J.S.; Huebener, P.; Mu, X.; Dapito, D.H.; Pradere, J.P.; Schwabe, R.F. Fate Tracing Reveals Hepatic Stellate Cells as Dominant Contributors to Liver Fibrosis Independent of Its Aetiology. Nat. Commun. 2013, 4, 2823. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  132. Böker, K.H.W.; Pehle, B.; Steinmetz, C.; Breitenstein, K.; Bahr, M.; Lichtinghagen, R. Tissue Inhibitors of Metalloproteinases in Liver and Serum/Plasma in Chronic Active Hepatitis C and HCV-Induced Cirrhosis. Hepatogastroenterology 2000, 47, 812–819. [Google Scholar] [PubMed]
  133. Berardis, S.; Sattwika, P.D.; Najimi, M.; Sokal, E.M. Use of Mesenchymal Stem Cells to Treat Liver Fibrosis: Current Situation and Future Prospects. World J. Gastroenterol. 2015, 21, 742–758. [Google Scholar] [CrossRef] [PubMed]
  134. Sahar, S.; Sassone-Corsi, P. Regulation of Metabolism: The Circadian Clock Dictates the Time. Trends Endocrinol. Metab. 2012, 23, 1–8. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  135. Turek, F.W.; Joshu, C.; Kohsaka, A.; Lin, E.; Ivanova, G.; McDearmon, E.; Laposky, A.; Losee-Olson, S.; Easton, A.; Jensen, D.R.; et al. Obesity and Metabolic Syndrome in Circadian Clock Mutant Mice. Science 2005, 308, 1043–1045. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  136. Adamovich, Y.; Rousso-Noori, L.; Zwighaft, Z.; Neufeld-Cohen, A.; Golik, M.; Kraut-Cohen, J.; Wang, M.; Han, X.; Asher, G. Circadian Clocks and Feeding Time Regulate the Oscillations and Levels of Hepatic Triglycerides. Cell Metab. 2014, 19, 319–330. [Google Scholar] [CrossRef] [Green Version]
  137. Dallmann, R.; Viola, A.U.; Tarokh, L.; Cajochen, C.; Brown, S.A. The Human Circadian Metabolome. Proc. Natl. Acad. Sci. USA 2012, 109, 2625–2629. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  138. Le Martelot, G.; Claudel, T.; Gatfield, D.; Schaad, O.; Kornmann, B.; Lo Sasso, G.; Moschetta, A.; Schibler, U. REV-ERBalpha Participates in Circadian SREBP Signaling and Bile Acid Homeostasis. PLoS Biol. 2009, 7, e1000181. [Google Scholar] [CrossRef] [Green Version]
  139. Zhou, D.; Wang, Y.; Chen, L.; Jia, L.; Yuan, J.; Sun, M.; Zhang, W.; Wang, P.; Zuo, J.; Xu, Z.; et al. Evolving Roles of Circadian Rhythms in Liver Homeostasis and Pathology. Oncotarget 2016, 7, 8625–8639. [Google Scholar] [CrossRef] [Green Version]
  140. Hardwick, J.P. Cytochrome P450 Omega Hydroxylase (CYP4) Function in Fatty Acid Metabolism and Metabolic Diseases. Biochem. Pharmacol. 2008, 75, 2263–2275. [Google Scholar] [CrossRef]
  141. Trevaskis, J.L.; Griffin, P.S.; Wittmer, C.; Neuschwander-Tetri, B.A.; Brunt, E.M.; Dolman, C.S.; Erickson, M.R.; Napora, J.; Parkes, D.G.; Roth, J.D. Glucagon-like Peptide-1 Receptor Agonism Improves Metabolic, Biochemical, and Histopathological Indices of Nonalcoholic Steatohepatitis in Mice. Am. J. Physiol. -Gastrointest. Liver Physiol. 2012, 302, 762–772. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  142. Elbel, E.E.; Lavine, J.E.; Downes, M.; Van Natta, M.; Yu, R.; Schwimmer, J.B.; Behling, C.; Brunt, E.M.; Tonascia, J.; Evans, R. Hepatic Nuclear Receptor Expression Associates with Features of Histology in Pediatric Nonalcoholic Fatty Liver Disease. Hepatol. Commun. 2018, 2, 1213–1226. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  143. Gustafson, B.; Gogg, S.; Hedjazifar, S.; Jenndahl, L.; Hammarstedt, A.; Smith, U. Inflammation and Impaired Adipogenesis in Hypertrophic Obesity in Man. Am. J. Physiol. Endocrinol. Metab. 2009, 297, E999–E1003. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  144. Exley, M.A.; Hand, L.; O’Shea, D.; Lynch, L. Interplay between the Immune System and Adipose Tissue in Obesity. J. Endocrinol. 2014, 223, R41–R48. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  145. Hosogai, N.; Fukuhara, A.; Oshima, K.; Miyata, Y.; Tanaka, S.; Segawa, K.; Furukawa, S.; Tochino, Y.; Komuro, R.; Matsuda, M.; et al. Adipose Tissue Hypoxia in Obesity and Its Impact on Adipocytokine Dysregulation. Diabetes 2007, 56, 901–911. [Google Scholar] [CrossRef] [Green Version]
  146. Lee, Y.S.; Kim, J.W.; Osborne, O.; Oh, D.Y.; Sasik, R.; Schenk, S.; Chen, A.; Chung, H.; Murphy, A.; Watkins, S.M.; et al. Increased Adipocyte O2 Consumption Triggers HIF-1α, Causing Inflammation and Insulin Resistance in Obesity. Cell 2014, 157, 1339–1352. [Google Scholar] [CrossRef] [Green Version]
  147. Lolmède, K.; De Durand Saint Front, V.; Galitzky, J.; Lafontan, M.; Bouloumié, A. Effects of Hypoxia on the Expression of Proangiogenic Factors in Differentiated 3T3-F442A Adipocytes. Int. J. Obes. Relat. Metab. Disord. 2003, 27, 1187–1195. [Google Scholar] [CrossRef] [Green Version]
  148. Wueest, S.; Rapold, R.A.; Rytka, J.M.; Schoenle, E.J.; Konrad, D. Basal Lipolysis, Not the Degree of Insulin Resistance, Differentiates Large from Small Isolated Adipocytes in High-Fat Fed Mice. Diabetologia 2009, 52, 541–546. [Google Scholar] [CrossRef] [Green Version]
  149. Giordano, A.; Murano, I.; Mondini, E.; Perugini, J.; Smorlesi, A.; Severi, I.; Barazzoni, R.; Scherer, P.E.; Cinti, S. Obese Adipocytes Show Ultrastructural Features of Stressed Cells and Die of Pyroptosis. J. Lipid Res. 2013, 54, 2423–2436. [Google Scholar] [CrossRef] [Green Version]
  150. Vila, I.K.; Badin, P.M.; Marques, M.A.; Monbrun, L.; Lefort, C.; Mir, L.; Louche, K.; Bourlier, V.; Roussel, B.; Gui, P.; et al. Immune Cell Toll-like Receptor 4 Mediates the Development of Obesity- and Endotoxemia-Associated Adipose Tissue Fibrosis. Cell Rep. 2014, 7, 1116–1129. [Google Scholar] [CrossRef]
  151. Tanaka, M.; Ikeda, K.; Suganami, T.; Komiya, C.; Ochi, K.; Shirakawa, I.; Hamaguchi, M.; Nishimura, S.; Manabe, I.; Matsuda, T.; et al. Macrophage-Inducible C-Type Lectin Underlies Obesity-Induced Adipose Tissue Fibrosis. Nat. Commun. 2014, 5, 4982. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  152. Deng, T.; Lyon, C.J.; Minze, L.J.; Lin, J.; Zou, J.; Liu, J.Z.; Ren, Y.; Yin, Z.; Hamilton, D.J.; Reardon, P.R.; et al. Class II Major Histocompatibility Complex Plays an Essential Role in Obesity-Induced Adipose Inflammation. Cell Metab. 2013, 17, 411–422. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  153. Marcelin, G.; Silveira, A.L.M.; Martins, L.B.; Ferreira, A.V.M.; Clément, K. Deciphering the Cellular Interplays Underlying Obesity-Induced Adipose Tissue Fibrosis. J. Clin. Investig. 2019, 129, 4032–4040. [Google Scholar] [CrossRef] [PubMed]
  154. Marcelin, G.; Ferreira, A.; Liu, Y.; Atlan, M.; Aron-Wisnewsky, J.; Pelloux, V.; Botbol, Y.; Ambrosini, M.; Fradet, M.; Rouault, C.; et al. A PDGFRα-Mediated Switch toward CD9 High Adipocyte Progenitors Controls Obesity-Induced Adipose Tissue Fibrosis. Cell Metab. 2017, 25, 673–685. [Google Scholar] [CrossRef] [Green Version]
  155. Zvonic, S.; Ptitsyn, A.A.; Conrad, S.A.; Scott, L.K.; Floyd, Z.E.; Kilroy, G.; Wu, X.; Goh, B.C.; Mynatt, R.L.; Gimble, J.M. Characterization of Peripheral Circadian Clocks in Adipose Tissues. Diabetes 2006, 55, 962–970. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  156. Wu, X.; Zvonic, S.; Floyd, Z.E.; Kilroy, G.; Goh, B.C.; Hernandez, T.L.; Eckel, R.H.; Mynatt, R.L.; Gimble, J.M. Induction of Circadian Gene Expression in Human Subcutaneous Adipose-Derived Stem Cells. Obesity 2007, 15, 2560–2570. [Google Scholar] [CrossRef] [PubMed]
  157. Fonken, L.K.; Workman, J.L.; Walton, J.C.; Weil, Z.M.; Morris, J.S.; Haim, A.; Nelson, R.J. Light at Night Increases Body Mass by Shifting the Time of Food Intake. Proc. Natl. Acad. Sci. USA 2010, 107, 18664–18669. [Google Scholar] [CrossRef] [Green Version]
  158. Ando, H.; Kumazaki, M.; Motosugi, Y.; Ushijima, K.; Maekawa, T.; Ishikawa, E.; Fujimura, A. Impairment of Peripheral Circadian Clocks Precedes Metabolic Abnormalities in Ob/Ob Mice. Endocrinology 2011, 152, 1347–1354. [Google Scholar] [CrossRef] [Green Version]
  159. Maury, E.; Navez, B.; Brichard, S.M. Circadian Clock Dysfunction in Human Omental Fat Links Obesity to Metabolic Inflammation. Nat. Commun. 2021, 12, 2388. [Google Scholar] [CrossRef]
  160. Huber, J.; Löffler, M.; Bilban, M.; Reimers, M.; Kadl, A.; Todoric, J.; Zeyda, M.; Geyeregger, R.; Schreiner, M.; Weichhart, T.; et al. Prevention of High-Fat Diet-Induced Adipose Tissue Remodeling in Obese Diabetic Mice by n-3 Polyunsaturated Fatty Acids. Int. J. Obes. 2007, 31, 1004–1013. [Google Scholar] [CrossRef]
  161. Divoux, A.; Tordjman, J.; Lacasa, D.; Veyrie, N.; Hugol, D.; Aissat, A.; Basdevant, A.; Guerre-Millo, M.; Poitou, C.; Zucker, J.D.; et al. Fibrosis in Human Adipose Tissue: Composition, Distribution, and Link with Lipid Metabolism and Fat Mass Loss. Diabetes 2010, 59, 2817–2825. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  162. Khan, T.; Muise, E.S.; Iyengar, P.; Wang, Z.V.; Chandalia, M.; Abate, N.; Zhang, B.B.; Bonaldo, P.; Chua, S.; Scherer, P.E. Metabolic Dysregulation and Adipose Tissue Fibrosis: Role of Collagen VI. Mol. Cell. Biol. 2009, 29, 1575–1591. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  163. Spencer, M.; Yao-Borengasser, A.; Unal, R.; Rasouli, N.; Gurley, C.M.; Zhu, B.; Peterson, C.A.; Kern, P.A. Adipose Tissue Macrophages in Insulin-Resistant Subjects Are Associated with Collagen VI and Fibrosis and Demonstrate Alternative Activation. Am. J. Physiol. Endocrinol. Metab. 2010, 299. [Google Scholar] [CrossRef] [PubMed]
  164. Sun, K.; Park, J.; Gupta, O.T.; Holland, W.L.; Auerbach, P.; Zhang, N.; Goncalves Marangoni, R.; Nicoloro, S.M.; Czech, M.P.; Varga, J.; et al. Endotrophin Triggers Adipose Tissue Fibrosis and Metabolic Dysfunction. Nat. Commun. 2014, 5, 3485. [Google Scholar] [CrossRef] [Green Version]
  165. Chaix, A.; Lin, T.; Le, H.D.; Chang, M.W.; Panda, S. Time-Restricted Feeding Prevents Obesity and Metabolic Syndrome in Mice Lacking a Circadian Clock. Cell Metab. 2019, 29, 303–319.e4. [Google Scholar] [CrossRef] [PubMed]
  166. Paschos, G.K.; Ibrahim, S.; Song, W.L.; Kunieda, T.; Grant, G.; Reyes, T.M.; Bradfield, C.A.; Vaughan, C.H.; Eiden, M.; Masoodi, M.; et al. Obesity in Mice with Adipocyte-Specific Deletion of Clock Component Arntl. Nat. Med. 2012, 18, 1768–1777. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  167. Jager, J.; Wang, F.; Fang, B.; Lim, H.W.; Peed, L.C.; Steger, D.J.; Won, K.J.; Kharitonenkov, A.; Adams, A.C.; Lazar, M.A. The Nuclear Receptor Rev-Erbα Regulates Adipose Tissue-Specific FGF21 Signaling. J. Biol. Chem. 2016, 291, 10867–10875. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  168. Hunter, A.L.; Pelekanou, C.E.; Barron, N.J.; Northeast, R.C.; Grudzien, M.; Adamson, A.D.; Downton, P.; Cornfield, T.; Cunningham, P.S.; Billaud, J.N.; et al. Adipocyte Nr1d1 Dictates Adipose Tissue Expansion during Obesity. Elife 2021, 10, e63324. [Google Scholar] [CrossRef]
  169. Pivovarova, O.; Gögebakan; Sucher, S.; Groth, J.; Murahovschi, V.; Kessler, K.; Osterhoff, M.; Rudovich, N.; Kramer, A.; Pfeiffer, A.F.H. Regulation of the Clock Gene Expression in Human Adipose Tissue by Weight Loss. Int. J. Obes. 2016, 40, 899–906. [Google Scholar] [CrossRef]
  170. Järvinen, T.A.H.; Järvinen, T.L.N.; Kääriäinen, M.; Kalimo, H.; Järvinen, M. Muscle Injuries: Biology and Treatment. Am. J. Sports Med. 2005, 33, 745–764. [Google Scholar] [CrossRef] [PubMed]
  171. Kjær, M. Role of Extracellular Matrix in Adaptation of Tendon and Skeletal Muscle to Mechanical Loading. Physiol. Rev. 2004, 84, 649–698. [Google Scholar] [CrossRef] [PubMed]
  172. Gillies, A.R.; Lieber, R.L. Structure and Function of the Skeletal Muscle Extracellular Matrix. Muscle Nerve 2011, 44, 318–331. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  173. Dyar, K.A.; Hubert, M.J.; Mir, A.A.; Ciciliot, S.; Lutter, D.; Greulich, F.; Quagliarini, F.; Kleinert, M.; Fischer, K.; Eichmann, T.O.; et al. Transcriptional Programming of Lipid and Amino Acid Metabolism by the Skeletal Muscle Circadian Clock. PLoS Biol. 2018, 16, e2005886. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  174. Hodge, B.A.; Wen, Y.; Riley, L.A.; Zhang, X.; England, J.H.; Harfmann, B.D.; Schroder, E.A.; Esser, K.A. The Endogenous Molecular Clock Orchestrates the Temporal Separation of Substrate Metabolism in Skeletal Muscle. Skelet. Muscle 2015, 5, 17. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  175. Zhang, H.; Liang, J.; Chen, N. Do Not Neglect the Role of Circadian Rhythm in Muscle Atrophy. Ageing Res. Rev. 2020, 63, 101155. [Google Scholar] [CrossRef] [PubMed]
  176. 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]
  177. Dyar, K.A.; Ciciliot, S.; Wright, L.E.; Biensø, R.S.; Tagliazucchi, G.M.; Patel, V.R.; Forcato, M.; Paz, M.I.P.; Gudiksen, A.; Solagna, F.; et al. Muscle Insulin Sensitivity and Glucose Metabolism Are Controlled by the Intrinsic Muscle Clock. Mol. Metab. 2013, 3, 29–41. [Google Scholar] [CrossRef] [PubMed]
  178. Kim, J.K.; Michael, M.D.; Previs, S.F.; Peroni, O.D.; Mauvais-Jarvis, F.; Neschen, S.; Kahn, B.B.; Kahn, C.R.; Shulman, G.I. Redistribution of Substrates to Adipose Tissue Promotes Obesity in Mice with Selective Insulin Resistance in Muscle. J. Clin. Investig. 2000, 105, 1791–1797. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  179. Yang, X.; Downes, M.; Yu, R.T.; Bookout, A.L.; He, W.; Straume, M.; Mangelsdorf, D.J.; Evans, R.M. Nuclear Receptor Expression Links the Circadian Clock to Metabolism. Cell 2006, 126, 801–810. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  180. Pircher, P.; Chomez, P.; Yu, F.; Vennström, B.; Larsson, L. Aberrant Expression of Myosin Isoforms in Skeletal Muscles from Mice Lacking the Rev-ErbAalpha Orphan Receptor Gene. Am. J. Physiol. Regul. Integr. Comp. Physiol. 2005, 288, R482–R490. [Google Scholar] [CrossRef]
  181. Ramakrishnan, S.N.; Lau, P.; Burke, L.J.; Muscat, G.E.O. Rev-Erbβ Regulates the Expression of Genes Involved in Lipid Absorption in Skeletal Muscle Cells: Evidence for Cross-Talk between Orphan Nuclear Receptors and Myokines. J. Biol. Chem. 2005, 280, 8651–8659. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  182. Narendra, D.; Tanaka, A.; Suen, D.F.; Youle, R.J. Parkin Is Recruited Selectively to Impaired Mitochondria and Promotes Their Autophagy. J. Cell Biol. 2008, 183, 795–803. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  183. Naghavi, M.; Wang, H.; Lozano, R.; Davis, A.; Liang, X.; Zhou, M.; Vollset, S.E.; Abbasoglu Ozgoren, A.; Abdalla, S.; Abd-Allah, F.; et al. Global, Regional, and National Age-Sex Specific All-Cause and Cause-Specific Mortality for 240 Causes of Death, 1990–2013: A Systematic Analysis for the Global Burden of Disease Study 2013. Lancet 2015, 385, 117–171. [Google Scholar] [CrossRef] [Green Version]
  184. Rhee, C.M.; Kovesdy, C.P. Epidemiology: Spotlight on CKD Deaths—Increasing Mortality Worldwide. Nat. Rev. Nephrol. 2015, 11, 199–200. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  185. Jager, K.J.; Kovesdy, C.; Langham, R.; Rosenberg, M.; Jha, V.; Zoccali, C. A Single Number for Advocacy and Communication-Worldwide More than 850 Million Individuals Have Kidney Diseases. Kidney Int. 2019, 96, 1048–1050. [Google Scholar] [CrossRef] [PubMed]
  186. Duffield, J.S. Cellular and Molecular Mechanisms in Kidney Fibrosis. J. Clin. Investig. 2014, 124, 2299–2306. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  187. Yoshioka, K.; Takemura, T.; Tohda, M.; Akano, N.; Miyamoto, H.; Ooshima, A.; Maki, S. Glomerular Localization of Type III Collagen in Human Kidney Disease. Kidney Int. 1989, 35, 1203–1211. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  188. Nerlich, A.G.; Schleicher, E. Identification of Lymph and Blood Capillaries by Immunohistochemical Staining for Various Basement Membrane Components. Histochemistry 1991, 96, 449–453. [Google Scholar] [CrossRef] [PubMed]
  189. Lebleu, V.S.; Taduri, G.; O’Connell, J.; Teng, Y.; Cooke, V.G.; Woda, C.; Sugimoto, H.; Kalluri, R. Origin and Function of Myofibroblasts in Kidney Fibrosis. Nat. Med. 2013, 19, 1047–1053. [Google Scholar] [CrossRef]
  190. Carew, R.M.; Wang, B.; Kantharidis, P. The Role of EMT in Renal Fibrosis. Cell Tissue Res. 2012, 347, 103–116. [Google Scholar] [CrossRef]
  191. Solocinski, K.; Gumz, M.L. The Circadian Clock in the Regulation of Renal Rhythms. J. Biol. Rhythms 2015, 30, 470–486. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  192. Voogel, A.J.; Koopman, M.G.; Hart, A.A.M.; Van Montfrans, G.A.; Arisz, L. Circadian Rhythms in Systemic Hemodynamics and Renal Function in Healthy Subjects and Patients with Nephrotic Syndrome. Kidney Int. 2001, 59, 1873–1880. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  193. Ansermet, C.; Centeno, G.; Nikolaeva, S.; Maillard, M.P.; Pradervand, S.; Firsov, D. The Intrinsic Circadian Clock in Podocytes Controls Glomerular Filtration Rate. Sci. Rep. 2019, 9, 16089. [Google Scholar] [CrossRef] [Green Version]
  194. Yoshida, Y.; Matsunaga, N.; Nakao, T.; Hamamura, K.; Kondo, H.; Ide, T.; Tsutsui, H.; Tsuruta, A.; Kurogi, M.; Nakaya, M.; et al. Alteration of Circadian Machinery in Monocytes Underlies Chronic Kidney Disease-Associated Cardiac Inflammation and Fibrosis. Nat. Commun. 2021, 12, 2783. [Google Scholar] [CrossRef] [PubMed]
  195. Egstrand, S.; Mace, M.L.; Olgaard, K.; Lewin, E. The Vascular Circadian Clock in Chronic Kidney Disease. Cells 2021, 10, 1769. [Google Scholar] [CrossRef] [PubMed]
  196. Zha, M.; Tian, T.; Xu, W.; Liu, S.; Jia, J.; Wang, L.; Yan, Q.; Li, N.; Yu, J.; Huang, L. The Circadian Clock Gene Bmal1 Facilitates Cisplatin-Induced Renal Injury and Hepatization. Cell Death Dis. 2020, 11, 446. [Google Scholar] [CrossRef] [PubMed]
  197. Zhang, J.; Liu, C.; Liang, Q.; Zheng, F.; Guan, Y.; Yang, G.; Chen, L. Postnatal Deletion of Bmal1 in Mice Protects against Obstructive Renal Fibrosis via Suppressing Gli2 Transcription. FASEB J. 2021, 35, e21530. [Google Scholar] [CrossRef] [PubMed]
  198. Kramann, R. Hedgehog Gli Signalling in Kidney Fibrosis. Nephrol. Dial. Transplant 2016, 31, 1989–1995. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  199. Chen, W.D.; Yeh, J.K.; Peng, M.T.; Shie, S.S.; Lin, S.L.; Yang, C.H.; Chen, T.H.; Hung, K.C.; Wang, C.C.; Hsieh, I.C.; et al. Circadian CLOCK Mediates Activation of Transforming Growth Factor-β Signaling and Renal Fibrosis through Cyclooxygenase 2. Am. J. Pathol. 2015, 185, 3152–3163. [Google Scholar] [CrossRef] [PubMed]
  200. Motohashi, H.; Tahara, Y.; Whittaker, D.S.; Wang, H.B.; Yamaji, T.; Wakui, H.; Haraguchi, A.; Yamazaki, M.; Miyakawa, H.; Hama, K.; et al. The Circadian Clock Is Disrupted in Mice with Adenine-Induced Tubulointerstitial Nephropathy. Kidney Int. 2020, 97, 728–740. [Google Scholar] [CrossRef] [PubMed]
  201. Matsunaga, N.; Ikeda, E.; Kakimoto, K.; Watanabe, M.; Shindo, N.; Tsuruta, A.; Ikeyama, H.; Hamamura, K.; Higashi, K.; Yamashita, T.; et al. Inhibition of G0/G1 Switch 2 Ameliorates Renal Inflammation in Chronic Kidney Disease. EBioMedicine 2016, 13, 262. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  202. Murata, Y.; Ueno, T.; Tanaka, S.; Kobayashi, H.; Okamura, M.; Hemmi, S.; Fuke, Y.; Matsumoto, Y.; Abe, M.; Fukuda, N. Identification of Clock Genes Related to Hypertension in Kidney From Spontaneously Hypertensive Rats. Am. J. Hypertens. 2020, 33, 1136. [Google Scholar] [CrossRef] [PubMed]
  203. Balsalobre, A.; Brown, S.A.; Marcacci, L.; Tronche, F.; Kellendonk, C.; Reichardt, H.M.; Schutz, G.; Schibler, U. Resetting of Circadian Time in Peripheral Tissues by Glucocorticoid Signaling. Science 2000, 289, 2344–2347. [Google Scholar] [CrossRef] [PubMed]
Figure 1. After injury, damaged cells recruit inflammatory cells (e.g., monocytes, neutrophils, and other immune cells), which promote fibroblasts differentiation into ECM-producing myofibroblasts, leading to fibrosis in various tissues.
Figure 1. After injury, damaged cells recruit inflammatory cells (e.g., monocytes, neutrophils, and other immune cells), which promote fibroblasts differentiation into ECM-producing myofibroblasts, leading to fibrosis in various tissues.
Ijms 23 12954 g001
Figure 2. Genetic factors (age and mutations in clock genes) and environmental factors (sleep disturbances, irregular diet, and shift work) disrupt circadian rhythm. Disturbances in the oscillations of the circadian clock lead to cardiometabolic diseases, CNS disorders, and cancer.
Figure 2. Genetic factors (age and mutations in clock genes) and environmental factors (sleep disturbances, irregular diet, and shift work) disrupt circadian rhythm. Disturbances in the oscillations of the circadian clock lead to cardiometabolic diseases, CNS disorders, and cancer.
Ijms 23 12954 g002
Figure 3. Clock and Bmal1 drive expression of two negative-feedback arms controlled by Period/Cryptochrome (Per/Cry) and the two paralogs, Rev-Erbα/β and ROR. In turn, these negative-feedback arms repress Bmal1/Clock transactivation function (Per/Cry) or Bmal1 expression (Rev-Erbα/β) and Bmal1 activation (ROR).
Figure 3. Clock and Bmal1 drive expression of two negative-feedback arms controlled by Period/Cryptochrome (Per/Cry) and the two paralogs, Rev-Erbα/β and ROR. In turn, these negative-feedback arms repress Bmal1/Clock transactivation function (Per/Cry) or Bmal1 expression (Rev-Erbα/β) and Bmal1 activation (ROR).
Ijms 23 12954 g003
Figure 4. Rev-Erbα/βs display robust 24 h oscillations and regulate the physiological functions of the organs. Rev-Erbs reduce adipogenesis and lipogenesis and increase bile acid synthesis in adipose tissue and liver, respectively. Rev-Erbs reduce cardiac hypertrophy and inflammatory cytokines in the heart, myofibroblast differentiation, and collagen production in the lungs. Rev-Erbs decrease myogenesis and increase mitochondrial function in skeletal muscles. Rev-Erbs increase injury and fibrosis in the kidney. (Ijms 23 12954 i001 = decrease, Ijms 23 12954 i002 = increase).
Figure 4. Rev-Erbα/βs display robust 24 h oscillations and regulate the physiological functions of the organs. Rev-Erbs reduce adipogenesis and lipogenesis and increase bile acid synthesis in adipose tissue and liver, respectively. Rev-Erbs reduce cardiac hypertrophy and inflammatory cytokines in the heart, myofibroblast differentiation, and collagen production in the lungs. Rev-Erbs decrease myogenesis and increase mitochondrial function in skeletal muscles. Rev-Erbs increase injury and fibrosis in the kidney. (Ijms 23 12954 i001 = decrease, Ijms 23 12954 i002 = increase).
Ijms 23 12954 g004
Figure 5. TGF-β inhibits the expression of the negative regulators of circadian clocks (Rev-Erbα/β, Per1/2, Cirbp) and prolongs the wake period. In addition, TGF-β reverses the wake period by stimulation of Clock and Bmal1 expression.
Figure 5. TGF-β inhibits the expression of the negative regulators of circadian clocks (Rev-Erbα/β, Per1/2, Cirbp) and prolongs the wake period. In addition, TGF-β reverses the wake period by stimulation of Clock and Bmal1 expression.
Ijms 23 12954 g005
Table 1. Role of Rev-Erb and their synthetic ligands in tissue fibrosis.
Table 1. Role of Rev-Erb and their synthetic ligands in tissue fibrosis.
A
Effector OrganRev-ErbFunctionsReference
Lung Knockout mice
  • Increased pulmonary myofibroblast activating markers such as collagen-1 and αSMA
  • Increased neutrophil lung influx and proinflammatory cytokines (IL-6, MCP-1)
[99,100]
Lower mRNA and protein expression
  • Smokers and COPD patients
[101]
Reduced Rev-Erbα mRNA and protein expression
  • Emphysema mice
[102]
LiverKnockout mice
  • Plasma VLDL concentrations and APOC-III expression
  • Hepatic steatosis and reduced bile acid synthesis
[55,103,104]
Knockout of Rev-Erbα/β mice fed with HFD
  • Hepatic triglyceride levels
[105]
Knockout in primary hepatocytes
  • CYP4A expression, lipid accumulation, and oxidative stress
[106]
Adipose tissueKnockout mice
  • Increased plasma adiponectin and its expression in WAT
[107,108]
  • Adiposity and hypertrophied adipocytes
[107,108]
Skeletal muscleRev-Erbα/β double-knockout mice
  • Altered lipid metabolism and circadian wheel-running behavior
[109]
Knockout mice
  • Reduced running capacity
  • Reduced skeletal muscle mass and muscle fiber cross-section area
  • Reduction of the regenerative capacity of muscle
[59,110,111]
Upregulation in Mice
  • Loss of muscle mass
[110]
Upregulation in C2C12 myocytes
  • Increases exercise capacity, mitochondrial content, and activity
[59]
Upregulation in myoblasts
  • ∙ Reduces dexamethasone-induced atrophy-related genes
[110]
KidneyRev-Erbα/β knockout mice
  • Decreased sensitivity to folic acid-induced acute kidney injury and diminished rhythm in disease severity
[112]
Rev-Erbβ knockout mice
  • Increasing gene and protein expressions of Slc7a11 and HO1
[112]
B
Rev-Erb ligandsEffector
organ
Animal ModelEffectsReference
SR9009 (Agonist)HeartTAC mice
  • Prevented cardiomyocyte hypertrophy, reduced fibrosis
[113]
Mice
  • Improved left ventricles (LV) function and survival after myocardial infarction
  • Decreased expression of cytokines IL-6, MCP-1, MMP9, and immune cells (neutrophil and proinflammatory macrophages) infiltration into the infarcted heart
  • Reduced ratio of proinflammatory M1 macrophages to anti-inflammatory M2 macrophages
[97]
LDL-receptor deficient mice fed with a western diet
  • Reduced atherosclerosis
[98]
GSK4112 (Agonist)Lungs Human small airway epithelial cells and mouse lung fibroblasts
  • Attenuated both LPS and cigarette smoking-induced inflammatory response and reduced pulmonary fibrosis
[100,114]
HFL-1 cells
  • Suppressed TGF-β–induced fibroblast differentiation
[115]
SR9009
(Agonist)
LiverMice
  • Induced weight loss and reduced plasma triglycerides and cholesterol
[63]
  • Inhibition of cholesterol biosynthesis
[116]
HSC
  • Inhibition of HSCs proliferation
[117]
Mice
  • Prevented alcohol-induced liver injury
[106]
NASH mice
  • Reduced hepatic inflammation (IL-1α, IL-1β, Ifnγ, and TNFα) and fibrosis
[118,119]
Ccl4-induced fibrosis in mice and Rat HSCs
  • Reduced fibrosis
[120,121]
SR6452
(Agonist)
Adipose
tissue
3T3L1 cells
  • Induction of adipocyte differentiation
[122]
SR9011 (Agonist) Mice
  • Reduced fat mass and lipogenic gene expression
[58]
SR9009 (Agonist) Mice
  • Induced weight loss and reduced plasma lipids
[58]
SR6452
(Agonist)
Skeletal
muscle
Mice
  • Regulation of atrophy-related genes
[110]
SR8278
(Antagonist)
Dystrophic mice
  • Improved mitochondrial biogenesis and muscle functions
[123]
GSK1362 (Inverse agonistKidney HEK293 cells
  • Inhibition of chemokines and cytokines
[114]
SR8278 (Antagonist) Mice
  • Reduced kidney damage
[112]
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Raza, G.S.; Sodum, N.; Kaya, Y.; Herzig, K.-H. Role of Circadian Transcription Factor Rev-Erb in Metabolism and Tissue Fibrosis. Int. J. Mol. Sci. 2022, 23, 12954. https://doi.org/10.3390/ijms232112954

AMA Style

Raza GS, Sodum N, Kaya Y, Herzig K-H. Role of Circadian Transcription Factor Rev-Erb in Metabolism and Tissue Fibrosis. International Journal of Molecular Sciences. 2022; 23(21):12954. https://doi.org/10.3390/ijms232112954

Chicago/Turabian Style

Raza, Ghulam Shere, Nalini Sodum, Yagmur Kaya, and Karl-Heinz Herzig. 2022. "Role of Circadian Transcription Factor Rev-Erb in Metabolism and Tissue Fibrosis" International Journal of Molecular Sciences 23, no. 21: 12954. https://doi.org/10.3390/ijms232112954

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