Next Article in Journal
Genome Evolution from Random Ligation of RNAs of Autocatalytic Sets
Next Article in Special Issue
Role of Terpenophenolics in Modulating Inflammation and Apoptosis in Cardiovascular Diseases: A Review
Previous Article in Journal
The Role of Filippi’s Glands in the Silk Moths Cocoon Construction
Previous Article in Special Issue
Diabetes Mellitus and Cardiovascular Diseases: Nutraceutical Interventions Related to Caloric Restriction
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

The Role of Anti-Inflammatory Adipokines in Cardiometabolic Disorders: Moving beyond Adiponectin

1
Asan Medical Center, Department of Internal Medicine, University of Ulsan College of Medicine, Seoul 05505, Korea
2
Asan Diabetes Center, Asan Medical Center, Seoul 05505, Korea
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2021, 22(24), 13529; https://doi.org/10.3390/ijms222413529
Submission received: 5 November 2021 / Revised: 9 December 2021 / Accepted: 13 December 2021 / Published: 16 December 2021
(This article belongs to the Special Issue Nutraceuticals in Cardiovascular and Metabolic Diseases)

Abstract

:
The global burden of obesity has multiplied owing to its rapidly growing prevalence and obesity-related morbidity and mortality. In addition to the classic role of depositing extra energy, adipose tissue actively interferes with the metabolic balance by means of secreting bioactive compounds called adipokines. While most adipokines give rise to inflammatory conditions, the others with anti-inflammatory properties have been the novel focus of attention for the amelioration of cardiometabolic complications. This review compiles the current evidence on the roles of anti-inflammatory adipokines, namely, adiponectin, vaspin, the C1q/TNF-related protein (CTRP) family, secreted frizzled-related protein 5 (SFRP5), and omentin-1 on cardiometabolic health. Further investigations on the mechanism of action and prospective human trials may pave the way to their clinical application as innovative biomarkers and therapeutic targets for cardiovascular and metabolic disorders.

1. Introduction

Obesity has been a growing threat worldwide owing to its association with various cardiometabolic disorders, including insulin resistance, type 2 diabetes mellitus (T2DM), fatty liver disease, and cardiovascular disease [1]. Accumulated evidence suggests that obesity-induced inflammation plays a key role in the pathogenesis of these disorders [1,2,3]. Obesity initiates chronic low-level inflammation, possibly by means of increased expression of pro-inflammatory genes and oxidative stress [2].
Contrary to the traditional concept of adipose tissue as an inert storage site for redundant energy, it is now highlighted as an endocrine organ that is involved in metabolic homeostasis. Since the characterization of leptin in 1994 [4], a number of adipocyte-derived compounds have been identified. These substances known as adipokines modulate appetite control, energy expenditure, insulin secretion, cardiovascular function, inflammation, immunity, adipogenesis, reproduction, and bone metabolism [5]. The inflammation and dysfunction of adipose tissue induce an alteration in adipokine secretion toward a pro-inflammatory, diabetogenic, and atherogenic pattern. Adipokines act not only locally by mediating the crosstalk between adipocytes, endothelial cells, and macrophages but also at the systemic level [2].
Most adipokines, including leptin, tumor necrosis factor-alpha (TNF-α), interleukin 6 (IL-6), resistin, or retinol-binding protein 4 (RBP4), are augmented in the obese state and promote inflammatory reactions, leading to obesity-associated comorbidities [6]. Contrarily, a smaller number of adipokines are produced by metabolically healthy adipose tissue to decrease inflammation and have protective effects on metabolic dysfunction [1]. This review focuses on the beneficial role of several anti-inflammatory adipokines of our interest in cardiometabolic disorders.

2. Adiponectin

2.1. Biological Actions

Adiponectin is one of the most extensively investigated adipokines since its first description in 1995 [7]. It is primarily produced by white adipose tissue, although additional sources, such as skeletal muscles or cardiomyocytes, have been known [8,9]. Circulating adiponectin exists in three different oligomers: low-molecular-weight trimer, middle-molecular-weight hexamer, and high-molecular-weight (HMW) multimer. HMW adiponectin is the principal bioactive form in metabolic tissues [10]. Activation of the two recognized receptors for adiponectin, AdipoR1 and R2, is linked to the stimulation of AMP-activated protein kinase (AMPK), p38 mitogen-activated protein kinase, and peroxisome proliferator-activated receptor alpha (PPARα) [11].
Adiponectin suppresses M1, which induces inflammation and insulin resistance while activating M2, which intensifies the anti-inflammatory response and oxidative metabolism [12,13,14]. Anti-inflammatory cytokines such as IL-10 are stimulated, and pro-inflammatory factors including TNF-α, interferon-gamma (IFN-γ), and vascular cell adhesion molecule-1 (VCAM-1) are inhibited by adiponectin [15,16]. Meanwhile, adiponectin inhibits hepatic glucose production, as well as promotes glucose utilization and fatty acid oxidation of the skeletal muscles via the stimulation of the AMPK and PPARα pathway [17,18,19,20] (Figure 1). Cytokine- or ceramide-induced β cell apoptosis is blocked by adiponectin, further contributing to insulin sensitivity [21,22]. Moreover, augmented nitric oxide (NO) synthesis by AMPK following the adiponectin-mediated phosphorylation of endothelial nitric oxide synthase (eNOS) attenuates vasoconstriction and progression of atherosclerosis [23,24]. Adiponectin also interferes with vascular smooth muscle cell (VSMC) hypertrophy [25]. Collectively, adiponectin has anti-inflammatory, insulin-sensitizing, and anti-atherogenic effects.
Several anti-diabetic medications are also recognized to intensify adiponectin expression (Figure 1). For instance, pioglitazone not only upregulates the adiponectin levels in adipose tissue but also affects the multimerization of adiponectin into the potent, HMW form [26]. Likewise, glucagon-like peptide-1 (GLP-1) analogs directly enhance the secretion of adiponectin via protein kinase A (PKA) signaling in vitro [27]. Lastly, T2DM mice administered with sodium-glucose cotransporter-2 (SGLT2) inhibitors exhibit elevation of plasma adiponectin levels [28]. Although further studies are needed, adiponectin may be the mediator of the cardiovascular benefits of pioglitazone, GLP-1 analogs, and SGLT2 inhibitors, which have been established by randomized controlled trials [29,30,31].
Recently, adiponectin is suggested to be a downstream mediator of fibroblast growth factor (FGF) 21, which is another potent modulator of glucose and lipid metabolism produced in adipose tissue [32]. The expression of adiponectin is stimulated by the acute injection of FGF21 through activating peroxisome proliferator-activated receptor gamma (PPARγ) [33]. In addition, the resistance to FGF21 inhibits adiponectin secretion [34]. Laboratory studies have advocated that adiponectin mediates the metabolic effects of FGF21, although the direct mechanism needs to be specified.

2.2. Human Relevance

Plasma adiponectin levels are reduced in patients with obesity [35], T2DM [36,37], atherosclerosis [38], or hypertension [39]. Clinical studies have shown that the VLDL and HDL levels are inversely and positively correlated with the adiponectin levels, respectively [40,41]. A recent cohort study proposed that low adiponectin levels may predict the diminished capability of cholesterol efflux regardless of body mass index (BMI) [42]. Along with an inverse relationship to total fat mass [35], adiponectin secretion is also regulated by the quality of adipose tissue [43]. Metabolically healthy but obese individuals tend to have higher levels of adiponectin compared with their unhealthy counterparts with a similar amount of adipose tissue [44]. In addition, the disorganized formation of adiponectin isoforms may be associated with cardiometabolic disorders. Patients with coronary artery disease (CAD) show a lower proportion of HMW multimer in contrast to a higher trimeric form. Likewise, only HMW adiponectin is increased following weight loss in obese patients [45]. The augmentation of adiponectin action through increasing its level or enhancing the signaling pathways may be a therapeutic approach for cardiometabolic diseases. Human levels of circulating adiponectin are positively affected by weight loss with a low-calorie diet [46]; medications, such as sibutramine or phentermine/topiramate [47,48]; and bariatric surgery [49].
Recently, some studies emphasized the contradictory results indicating that high adiponectin levels are associated with unfavorable cardiovascular and other metabolic outcomes [50,51,52]. This contradiction, referred to as the adiponectin paradox, might be attributed to a compensatory response in cardiovascular disorders, instead of precipitating them. Reduced adiponectin clearance is another possible explanation considering that hyperadiponectinemia is frequently observed in patients who have both cardiovascular diseases and hepatic or renal damage [43]. Finally, adiponectin isoforms rather than concentration may have a detrimental role in cardiovascular homeostasis. Even though the adiponectin paradox makes it cumbersome to apply adiponectin as a biomarker, it is still a promising therapeutic target for cardiometabolic disorders.

3. Vaspin

3.1. Biological Actions

Visceral adipose tissue-derived serpin protease inhibitor (vaspin) belongs to serpin family A member 12 (serpin A12) in accordance with the serpin nomenclature [53]. It was first isolated from the Otsuka Long-Evans Tokushima fatty (OLETF) rat, a T2DM animal model with obesity [54]. Vaspin is expressed in not only visceral and subcutaneous adipose tissues but also other organs, including the pancreas, liver, stomach, and skin [55]. Vaspin binds to a 78 kDa glucose-regulated protein and initiates multiple kinase signaling pathways such as AMPK and Akt [56]. Two protease targets of vaspin have been discovered so far, that is, kallikrein 7 and kallikrein 14, which are inhibited by vaspin through the classical serpin mechanism [57,58].
Vaspin has beneficial effects on glucose metabolism, vascular health, appetite control, and lipid profile (Figure 2). Vaspin expression in adipose tissue and plasma vaspin concentration increases in the highest point of insulin resistance and obesity but decreases with the aggravation of T2DM and body weight loss in the OLETF rat [54]. This conflicting tendency of vaspin contrary to adiponectin may imply the compensatory role of vaspin in metabolic dysfunction. Obviously, in vivo, the infusion of recombinant vaspin or transgenic vaspin overexpression enhances glucose tolerance and insulin sensitivity [54,59,60]. Vaspin promotes pancreatic islet cell secretion, protects β cells from nuclear factor-kappa B (NF-κB)-mediated inflammatory damage, and reduces glucose production in the liver [61,62].
Counteracting vascular inflammation is one of the major capacities of vaspin for preserving endothelial function. Vaspin attenuates NF-κB activity in response to the reduced secretion of inflammatory cytokines in the AMPK-dependent pathway [63,64,65]. The macrophage phenotype is shifted by vaspin toward anti-inflammatory M2 rather than M1 with the downregulation of NF-κB and upregulation of PPARγ [59]. Moreover, vaspin dampens the generation of reactive oxygen species (ROS) and oxidative stress-induced apoptosis of mesenchymal stem cells (MSCs) [66,67,68,69]. The progression of aortic atherosclerosis is mitigated by vaspin through impeding intimal proliferation and plaque instability [59,70]. We also demonstrated that vaspin was involved in the aortic vasorelaxation following the augmentation of eNOS activity via favorable effects on the signal transducer and activator of transcription 3 and asymmetric dimethylarginine system [71]. Furthermore, vaspin modulates feeding regulation and lipid metabolism. Both the peripheral and central application of vaspin reduce food intake [72], partly due to the downregulated expression of the hypothalamic orexigenic neuropeptides [73]. High-dose vaspin infusion decreases the free fatty acid and triglyceride levels as well as promotes cholesterol efflux via the upregulation of ATP-binding cassette transporter A1 (ABCA1) in macrophages [59,74]. These biological actions of vaspin support its role as an anti-atherogenic adipokine.

3.2. Human Relevance

Compared with non-obese controls, serum levels of vaspin are higher in obese subjects with even normal body weight [75]. A meta-analysis verified significantly higher vaspin concentrations in subjects with T2DM compared with non-diabetic subjects [76]. Increased vaspin concentrations in T2DM patients are also associated with the elevated risk of diabetic complications, such as CAD or diabetic retinopathy [77,78]. The vaspin levels decrease as insulin sensitivity improves through exercise, anti-diabetic medication such as metformin and rosiglitazone, and bariatric surgery [79,80,81,82]. Meanwhile, variants of the vaspin rs2236242 gene have been found to be correlated with the development of T2DM independently of obesity [83]. Contrarily, treatment with atorvastatin or rosuvastatin has been shown to increase the vaspin levels in patients with non-significant carotid atherosclerosis and acute coronary syndrome, respectively [84,85]. Low plasma vaspin levels are significantly associated with a higher risk of preclinical carotid atherosclerosis [84], acute coronary syndrome, coronary in-stent restenosis after percutaneous coronary intervention (PCI) [86], and poorer prognosis after myocardial infarction (MI) [87]. The contrasting results of the association between the vaspin levels and cardiometabolic health might have originated from the different study designs.
Vaspin has been investigated as a diagnostic tool for cardiometabolic disorders. A retrospective observational study on individuals with chest pain implied the availability of vaspin as a predictive biomarker of major adverse cardiovascular events (MACE) [88]. The vaspin expression levels significantly indicated the risk of ischemic stroke in a prospective study of T2DM patients [89]. In women with polycystic ovary syndrome (PCOS), serum vaspin concentrations can make a distinction between individuals with a higher diabetogenic risk [90]. The vaspin levels also predict metabolic syndrome as a single entity, as well as some of its components [91,92]. Taken together, beginning with the identification from the diabetic animals, vaspin is expected to be applicable in cardiometabolic diseases.

4. C1q/TNF-Related Protein (CTRP) Family

The CTRP family is a conserved group of adiponectin paralogs, containing a structural similarity to adiponectin. Along with the epicardial adipose tissue, which is the main secretory organ, various sources, such as the heart, liver, kidney, and muscles, have been recognized [93]. Each member of the 15 identified CTRP isoforms has a distinct function. To sum up the latest research, CTRP1, CTRP3, CTRP5, CTRP6, CTRP9, CTRP12, CTRP13, and CTRP15 engage in the development of cardiometabolic diseases, among which CTRP1 and CTRP5 promote the pro-inflammatory response, whereas the others favor the opposite [94] (Figure 3).

4.1. CTRP9

4.1.1. Biological Actions

Of all its family members, CTRP9 has attracted the most attention following its discovery in 2009 [95]. CTRP9 shares the greatest homology with adiponectin involving 51% amino acid overlap [95]. In contrast to adiponectin that maintains the full length in the plasma, the major circulatory and biologically active structure of CTRP9 is the globular form produced by the proteolytic cleavage of trimeric complexes [96]. The heart is the third richest organ for CTRP9 distribution, and cardiac function is significantly influenced by CTRP9, making CTRP9 not only an adipokine but also a cardiokine [97]. Currently, two receptors have been introduced: AdipoR1 and N-cadherin, which is a cell surface marker of MSCs [98,99].
The administration of CTRP9 exerts anti-inflammatory effects by downregulating TNF-α with the downstream mediators through the activation of the AdipoR1/AMPK/NF-kB signaling cascade [100,101]. In addition, it interferes with macrophage polarization, stimulating the M1 to M2 transition [102]. The targeted deletion of murine CTRP9 leads to obesity, insulin resistance, and hepatic steatosis through both the central and peripheral mechanisms with the overexpression of orexigenic peptides in the hypothalamus, blunting hepatic insulin signaling, and upregulating the lipogenic genes [103]. Recombinant globular CTRP9 elevates the HDL levels while reducing the LDL and triglyceride levels, via the enhancement of cholesterol efflux by accelerating AdipoR1/AMPK signaling and inhibition of lipoprotein uptake by suppressing lectin-like oxidized low-density lipoprotein receptor 1 [100,104,105,106]. In vivo and in vitro studies have shown that the inhibitory effect of CTRP9 on VSMC proliferation, neointimal hyperplasia, and platelet activation prevents the progression of atherosclerosis [107,108,109]. The trans-differentiation of VSMCs into macrophage-like cells aggravates the inflammatory and proliferative response as well as debilitates the vascular contractile function, which is antagonized by CTRP9 [100]. Furthermore, CTRP9 serves as a defender of endothelial function by facilitating vascular relaxation and ischemia-induced revascularization through the AMPK/Akt/eNOS pathway [110,111,112]. It downregulates ROS production and apoptosis of endothelial cells [113,114,115], along with promoting the phagocytic removal of apoptotic cells to deter subsequent necrosis and inflammation [116]. Likewise, we have shown that palmitic acid-dependent endothelial senescence, which is a significant risk factor of vascular atherosclerosis, was abrogated by CTRP9-mediated restoration of AdipoR1-/AMPK-mediated autophagy [117]. In addition, the anti-fibrotic action of CTRP9 alleviates the post-MI pathologic cardiac remodeling and ventricular dysfunction [118]. Altogether, these actions of CTRP9 contribute to anti-inflammation and anti-atherogenesis.

4.1.2. Human Relevance

Two retrospective clinical studies on the association of the CAD and CTRP9 levels presented conflicting results [119,120]. Nonetheless, numerous human data favor the protective effects of CTRP9 on the cardiovascular system. Serum levels of CTRP9 along with CTRP3 are reduced in proportion to the severity of heart failure with reduced ejection fraction [121]. In patients who underwent kidney transplantation, plasma CTRP9 concentration is an improving factor of the aortic calcification [122]. In addition, elevated CTRP9 levels are correlated with less restenosis following the implantation of cerebrovascular stent [123].
Similar to coronary atherosclerosis, antecedent reports on plasma CTRP9 concentrations in patients with metabolic syndrome showed inconsistent trends [124,125]. Obese patients are associated with higher CTRP9 levels, which are downregulated following bariatric surgery [126]. Conversely, serum CTRP9 levels are significantly lower in patients with gestational diabetes mellitus (GDM) [127]. It is cautiously supposed that the CTRP9 levels fluctuate to compensate in the early stage of metabolic dysfunction, taking into account that the CTRP9 expression is increased at 8 weeks of age but decreased 4 weeks later in leptin-deficient obese mice [95]. Several clinical studies of diabetic patients suggested plasma CTRP9 levels as reliable biomarkers for cardiac autonomic neuropathy, carotid intima-media thickness (IMT), and arterial stiffness represented by brachial-ankle pulse wave velocity (baPWV) [128,129,130]. Likewise, CTRP9 levels are independent risk factors for the aggravation of GDM [131].

4.2. Other Members of the CTRP Family with Anti-Inflammatory Effects

4.2.1. Biological Actions

In conjunction with the most renowned CTRP9, other anti-inflammatory isoforms, such as CTRP3, CTRP12, and CTRP13, exert insulin-sensitizing, anti-atherosclerotic, and cardioprotective properties. CTRP3 remarkably decreases the inflammatory cytokines, suppresses cellular apoptosis, and equilibrates the opposing effects of endothelin-1 and NO by prompting the phosphatidylinositol 3-kinase (PI3K), Akt, and eNOS expressions [132]. CTRP12, also referred to as adipolin, is inversely correlated with IL-6 and TNF-α, and encourages the M2 phenotype of macrophage polarization [133,134]. CTRP3 inhibits hepatic glucose output [135], and both CTRP12 and CTRP13 repress gluconeogenesis, intensify the glucose uptake of various organs, and relieve hepatic insulin resistance [136,137]. Additionally, CTRP12 disturbs the neointimal thickening by alleviating vascular cell proliferation and lipid accumulation following the promotion of ABCA1-mediated cholesterol efflux [134,138]. CTRP13-infused mice are protected from atherosclerotic plaque formation with accelerated autophagy, downregulated lipid uptake, and inhibited foam cell migration [139].
Post-MI cardiac impairment is mitigated with the overexpression of CTRP3 by preserving the survival, migration, and antioxidant capacity of MSCs [140]. The anti-fibrotic, anti-apoptotic, and angiogenic functions of CTRP3 further ameliorate adverse cardiac remodeling after MI [141,142]. Conversely, CTRP3 is activated in dysfunctional human hearts, and it rather develops cardiac hypertrophy in response to overpressure [143]. This discrepancy may have derived from different trial models, which needs to be further elucidated. For CTRP12, the beneficial effects on myocardial survival and ventricular contractile function have been confirmed recently [144].

4.2.2. Human Relevance

A longitudinal study demonstrated that the circulating CTRP3 levels are negatively related to obesity, non-alcoholic fatty liver disease, and the indicators of pancreatic β-cell function [145]. Reflecting the compensative role, serum CTRP3 levels are increased in the pre-diabetic state but decreased in T2DM patients [146,147]. Metformin treatment upregulates the concentrations of CTRP3 and CTRP12 in humans [148,149]. Likewise, the CTRP13 levels present an inverse relationship with the adverse metabolic factors including BMI, insulin, lipidemia, and carotid IMT [150].
The CTRP3 levels are reduced in patients with CAD, acute aortic dissection, and heart failure [151,152,153]. Similarly, the CTRP12 levels are negatively correlated with the severity of CAD [154]. In the meantime, complement 1q (C1q), a complement subcomponent that provokes the classical immune pathway, can bind to adiponectin [155]. Plasma C1q-adiponectin/total adiponectin ratio is positively associated with atherosclerosis measured by vascular ultrasonography and can serve as an indicator of CAD [156,157,158,159].
To date, the anti-inflammatory members of the CTRP family show a generally compatible contribution to cardiovascular and metabolic homeostasis, despite the inconsistent findings in some studies.

5. Secreted Frizzled-Related Protein 5 (SFRP5)

5.1. Biological Actions

Wingless-type family member 5a (WNT5A), a glycopeptide secreted by adipose tissue macrophages, belongs to a non-canonical member of the Wnt family [160]. WNT5A has been implicated in cellular inflammation, proliferation, and migration to notably develop atherosclerosis and insulin resistance [6,161]. In persistent low-grade inflammation, the actions of WNT5A are offset by SFRP5 which is produced by healthy adipose tissues [162] (Figure 4). SFRP5 is one of the five identified SFRP families, which is the largest group of Wnt inhibitors. According to Ouchi et al., SFRP5 is substantially expressed in white adipose tissue in addition to other insulin targets, including the liver, muscle, and beta cells [163]. Although both the canonical and non-canonical Wnt pathways are impeded by SFRP5, the blockage of WNT5A signaling has been established to play a dominant role in the modulation of metabolic homeostasis [164,165].
The anti-inflammatory function of SFRP5 is exerted by the neutralization of c-Jun N-terminal kinases (JNK) in the downstream of the Wnt signaling pathway, to repress the production of inflammatory cytokines [162,166] (Figure 4). SFRP5 facilitates adipogenic differentiation and adipocyte expansion [167,168]. Despite the augmented expression of SFRP5 in adipose tissues, it seems to decline following the aggravation of metabolic dysfunction, considering the transient upregulation of SFRP5 with milder inflammation in adipocytes and ensuing drop after the development of obesity. In the same way, SFRP5 knockout mice acquire severe insulin resistance and hepatic steatosis with a greater macrophage accumulation in their adipocytes [162]. SFRP5 suppresses beta cell proliferation but enhances glucose-dependent insulin production, indicating the protective role on beta cell function [169]. Moreover, the central infusion of SFRP5 in vivo ameliorates glucose metabolism through brain-hepatic circuits, with less food intake, more energy expenditure, and potentiated hypothalamic neurons engaged in insulin activity, while inhibiting hepatic gluconeogenesis and lipogenesis via the hepatic vagus nerve [170].
The upregulation of SFRP5 significantly attenuates apoptosis of human endothelial cells, whereas the overexpression of WNT5A or knockdown of SFRP5 exhibits the opposite outcomes [171,172]. We have previously substantiated that SFRP5 dose dependently reverses impaired vasorelaxation by increasing the NO production via blockage of the WNT5A/JNK pathway [173]. SFRP5 also restricts VSMC proliferation and migration as well as decreases ROS generation in the murine aorta [174]. Meanwhile, SFRP5 exerts the restoring function in ischemic heart disease, as evidenced by SFRP5-deficient mice presenting more extensive inflammation, apoptosis, infarct size, and cardiac dysfunction consequent to ischemia/reperfusion injury [175]. SFRP5 also interferes with the survival, proliferation, and migration of cardiac fibroblasts [176].

5.2. Human Relevance

Obesity predisposes to decreased SFRP5 and increased WNT5A expression, which has been confirmed in human cohorts with atherosclerosis [177]. Cross-sectional studies have demonstrated that higher SFRP5 levels are negatively associated with odds of prediabetes and T2DM, along with multiple risk factors, such as systolic blood pressure, BMI, and C-reactive protein levels [178,179]. Likewise, lifestyle intervention with significant weight loss and anti-diabetic medications, including metformin and liraglutide, increase the SFRP5 levels in humans [180,181,182]. Numerous clinical trials showed the association between higher SFRP5 levels and a favorable lipid profile [178,179,183,184]. Contrarily, another study suggested that elevated SFRP5 concentrations are rather correlated with a higher risk of T2DM [185]. Further investigation is needed to determine whether the SFRP5 levels vary according to the progression of metabolic dysfunction.
Corresponding to in vitro and in vivo studies, clinical trials have indicated the salutary effects of SFRP5 on vascular health. In the adipocytes of patients with the peripheral arterial disease (PAD), the plasma concentrations of WNT5A are significantly increased but those of SFRP5 are significantly decreased [186]. Similarly, the serum SFRP5 levels are lower in chronic kidney disease subjects with vascular calcification compared with those without calcification [187]. On the other hand, we reported the positive correlation between SFRP5 concentrations and baPWV based on the multivariate linear regression analysis of T2DM patients. This discrepancy in the SFRP5 levels may be interpreted as a compensatory rise in incipient atherosclerosis, taking into account the restoration of endothelial NO synthesis by SFRP5 [173].
Human data concerning the role of SFRP5 on CAD have been discordant. CAD patients have higher plasma WNT5A levels and lower SFRP5 levels compared with those without CAD, independent of the conventional risk factors [177]. An experiment using the mRNA levels of SFRP5 and WNT5A from epicardial adipose tissue biopsies of CAD patients reconfirmed the results [188]. Additionally, the WNT5A concentration is independently correlated with the occurrence and advancement of coronary calcification [177]. In the acute phase of ST-segment elevation MI, high SFRP5 concentrations are significantly related to early myocardial recovery after primary PCI [189]. Furthermore, a prospective cohort study proposed SFRP5 as a novel prognostic marker for heart failure on the basis of enhanced discrimination by conjoining SFRP5 to the traditional risk factors [190]. On the contrary, elevated SFRP5 levels predicted the incidence of MACE in a 4-year prospective study [191]. In spite of the conflicting outcomes regarding the relationship of the SFRP5 levels with insulin resistance and CAD, the majority of up-to-date research supports the beneficial role of SFRP5 in cardiometabolic health. Future research on the temporal variation of SFRP concentration during the disease course as well as the mechanism of action is required.

6. Omentin-1

6.1. Biological Actions

Omentin is a novel adipokine comprising 313 amino acids [192]. It is preferentially produced by visceral adipose tissue rather than subcutaneous adipose tissue. Omentin-1 and omentin-2 are two identified isoforms showing analogous gene expression, among which omentin-1 is a dominant form in the human circulation [193]. Cumulative experimental studies have demonstrated the direct intervention of omentin-1 in anti-inflammation, insulin sensitization, and cardiovascular health mainly through the cascade of AMPK/Akt/NF-kB/extracellular signal-regulated kinase (ERK), JNK, and p38 [194]. The antagonistic effects of omentin-1 on inflammation and atherosclerosis have been confirmed by various in vivo and in vitro studies. Transgenic mice with the human omentin gene show decreased expression of pro-inflammatory cytokines via the stimulation of Akt [195]. Omentin-1 also suppresses macrophage aggregation, apoptosis, foam cell formation, and M1 phenotype [195,196,197]. Previously built atherosclerotic lesions are diminished by omentin-1 through reducing lipid content and necrotic core [195,196]. Neointimal hyperplasia and VSMC proliferation are attenuated by omentin via the AMPK/ERK pathway [198]. Likewise, omentin-1 overexpression mitigates arterial calcification through PI3K/Akt-dependent mechanisms [199].
Cardiovascular dysfunction is also ameliorated by omentin-1. Bioavailability of NO is improved by omentin-mediated phosphorylation of eNOS to promote vasorelaxation and revascularization in contracted artery [200,201,202]. Omentin-derived activation of the canonical pathway of AMPK/Akt/ERK attenuates myocyte apoptosis, hypertrophy, and fibrosis following myocardial ischemia [203,204]. Similarly, overexpression of omentin-1 enhances parameters of cardiac function and cardiac mitochondrial activities possibly through upregulating glycogen synthase kinase-3 beta pathway in the rat model [205].

6.2. Human Relevance

Investigations on the correlation of human plasma levels of omentin-1 and cardiometabolic disorders display remarkably consistent results. Cross-sectional studies determined significantly lower omentin-1 levels in obese individuals compared with lean controls [193,206]. Even after adjusting for BMI, both serum and adipocyte levels of omentin-1 are inversely associated with the presence of metabolic syndrome [207]. A negative correlation of the serum omentin-1 levels with T2DM and GDM has also been reported [206,208]. Moreover, plasma omentin-1 is further reduced in T2DM patients with diabetic complications including retinopathy, peripheral neuropathy, nephropathy, and PAD than in those without complications [209,210]. In addition to decreased omentin-1 levels, patients with diabetic nephropathy exhibit significantly increased IL-6 which plays a critical role in immune response [211]. Serum IL-6 levels are independent factors determining omentin-1 levels, indicating the crosstalk between the adipose tissue and immune system [212].
Decreased plasma omentin-1 levels are not only significantly associated with CAD but also reflective of the disease severity, with lower values in acute coronary syndrome than in stable angina pectoris [213,214]. Biscetti et al. performed a prospective cohort study with T2DM subjects who have PAD and chronic limb-threatening ischemia to reveal that declined omentin-1 levels at baseline are related to higher risks of MACE and major adverse limb events [215]. On the other hand, higher plasma omentin levels are associated with enhanced cardiac function in patients with MI and heart failure [203,216]. Omentin-1 is also thought to influence blood pressure and lipid profile regulation. Individuals with stage 1 and 2 hypertension have significantly lower omentin-1 in circulation compared with their normotensive counterparts [217]. HDL and VLDL concentrations are correlated with omentin-1 levels positively and negatively, respectively [193,207].
The possibility of using circulating omentin-1 levels as biomarkers for cardiovascular diseases has been raised. Reduced baseline levels of omentin-1 predict worse functional prognosis, carotid plaque instability, and 1-year mortality in patients with acute ischemic stroke [218,219,220,221], as well as poorer clinical outcomes for acute intracerebral hemorrhage [222]. Lower omentin-1 levels are also prognostic markers of frequent MACE, severe angiographic CAD, and incomplete coronary collateral circulation [215,223,224]. In addition, plasma omentin-1 levels are independent determinants of several vascular complications of diabetic patients including carotid plaque, arterial stiffness measured by baPWV, PAD, and retinopathy [210,225,226,227].
Upregulation of circulating omentin-1 levels may be a novel therapeutic approach to delay the progression of cardiometabolic diseases. A recent prospective cohort study of obese or overweight children discovered that both short-term and long-term lifestyle interventions correlated positively with omentin-1 levels [228]. Similarly, a high-intensity exercise program of 12-week duration and weight loss after biliopancreatic diversion surgery augments plasma omentin-1 levels [229,230]. The widely prescribed medications such as metformin and atorvastatin are also expected to increase serum levels of omentin-1 [84,231]. In conclusion, coherent results of animal and human data on the favoring effect of omentin-1 on the cardiometabolic system may allow omentin-1 to be developed as a predictive marker or therapeutic medium.

7. Conclusions

Obesity, beyond its definition indicating the static state of excessive fat accumulation, accompanies chronic low-grade inflammation and is associated with a broad spectrum of cardiometabolic diseases, including T2DM, dyslipidemia, CAD, and fatty liver disease. Likewise, the pleiotropic effects of adipokines on metabolic homeostasis have also been investigated. Together with the autocrine and paracrine actions on local adipose tissue and macrophages, adipokines reach the systemic vasculature to actively interact with the central nervous system, pancreatic islets, or liver. This review specifically summarizes the cardiometabolic function of anti-inflammatory adipokines, covering from prototypical adiponectin to the more recently discovered omentin-1. The present review is distinguished by its explicit partition of experimental and clinical data as well as the introduction of diverse, up-to-date research on humans, especially regarding the applicability as biomarkers and therapeutic targets. These advantages allow for a better understanding of current knowledge and constraints, along with an outline of future study.
A number of retrospective clinical studies have implied the predictive value of adipokines for the risk of cardiovascular disorders and the capability as biomarkers replacing more complicated diagnostic tests. Contrary to the consistent tendency presented in the animal data, human trials showed incompatible results of the associations between the serum adipokine levels and diseases (Table 1). More mechanistic explanations and prospective human trials are needed to overcome the limitations of animal studies and contradictory outcomes. The exploration of the dynamic expression of adipokines, rather than the single serum concentration, will confirm their role as prognostic markers for cardiovascular diseases. Additionally, further clinical investigation concerning the relationship of demographic factors such as age, sex, and ethnicity with the cardiometabolic effects of adipokines may be valuable. Furthermore, expanding from the gathered animal data indicating that the upregulation or exogenous administration of each substance exhibits therapeutic effects on vascular and metabolic function, the manipulation of the axis connecting the anti-inflammatory adipokines and target organs may be the blueprint for the future approaches in managing cardiometabolic disorders. Reasonable strategies for applying adipokines as biotargets are manufacturing the analogs which are stable in the circulation to sustain enough therapeutic concentration, in addition to influencing directly the action target and downstream signaling.

Author Contributions

Conceptualization, C.H.J.; investigation, H.N.J. and C.H.J.; writing—original draft preparation, H.N.J.; writing—review and editing, C.H.J. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Acknowledgments

This work was supported by the Basic Science Research Program through the National Research Foundation of Korea (NRF) funded by the Ministry of Education (NRF-2020R1A2C1101977: C.H.J.). These funding sources had no roles in the design of the study; the collection, analysis, and interpretation of data; writing of the article; or the decision to submit the article for publication.

Conflicts of Interest

The authors declare no conflict of interest.

Abbreviations

STEMI, ST-segment elevation MI; T2DM, type 2 diabetes mellitus; N/R, not reported; MetS, metabolic syndrome; CVD, cardiovascular disease; HF, heart failure; CAD, coronary artery disease; NAFLD, non-alcoholic fatty liver disease; PCOS, polycystic ovary syndrome; ACS, acute coronary syndrome; MI, myocardial infarction; MACE, major adverse cardiac events; IS, ischemic stroke; HFrEF, heart failure with reduced ejection fraction; GDM, gestational diabetes mellitus; CKD, chronic kidney disease; IMT, intima-media thickness; baPWV, brachial ankle pulse wave velocity; SAP, stable angina pectoris; VT, ventricular tachycardia; BMI, body mass index; FPG, fasting plasma glucose; TC, total cholesterol; PAD, peripheral arterial disease; LVEF, left ventricular ejection fraction; CTLI, chronic limb-threatening ischemia; MALE, major adverse limb events; EF, ejection fraction; CI, cerebral infarction.

References

  1. Ouchi, N.; Parker, J.L.; Lugus, J.J.; Walsh, K. Adipokines in inflammation and metabolic disease. Nat. Rev. Immunol. 2011, 11, 85–97. [Google Scholar] [CrossRef] [PubMed]
  2. Shoelson, S.E.; Herrero, L.; Naaz, A. Obesity, inflammation, and insulin resistance. Gastroenterology 2007, 132, 2169–2180. [Google Scholar] [CrossRef] [PubMed]
  3. Wellen, K.E.; Hotamisligil, G.S. Inflammation, stress, and diabetes. J. Clin. Investig. 2005, 115, 1111–1119. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  4. Zhang, Y.; Proenca, R.; Maffei, M.; Barone, M.; Leopold, L.; Friedman, J.M. Positional cloning of the mouse obese gene and its human homologue. Nature 1994, 372, 425–432. [Google Scholar] [CrossRef]
  5. Blüher, M. Adipokines—Removing road blocks to obesity and diabetes therapy. Mol. Metab. 2014, 3, 230–240. [Google Scholar] [CrossRef] [PubMed]
  6. Recinella, L.; Orlando, G.; Ferrante, C.; Chiavaroli, A.; Brunetti, L.; Leone, S. Adipokines: New Potential Therapeutic Target for Obesity and Metabolic, Rheumatic, and Cardiovascular Diseases. Front. Physiol. 2020, 11, 578966. [Google Scholar] [CrossRef] [PubMed]
  7. Scherer, P.E.; Williams, S.; Fogliano, M.; Baldini, G.; Lodish, H.F. A Novel Serum Protein Similar to C1q, Produced Exclusively in Adipocytes. J. Biol. Chem. 1995, 270, 26746–26749. [Google Scholar] [CrossRef] [Green Version]
  8. Martinez-Huenchullan, S.F.; Tam, C.S.; Ban, L.A.; Ehrenfeld-Slater, P.; Mclennan, S.V.; Twigg, S.M. Skeletal muscle adiponectin induction in obesity and exercise. Metabolism 2020, 102, 154008. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  9. Natarajan, R.; Salloum, F.N.; Fisher, B.J.; Kukreja, R.C.; Fowler, A.A. Hypoxia Inducible Factor-1 Upregulates Adiponectin in Diabetic Mouse Hearts and Attenuates Post-Ischemic Injury. J. Cardiovasc. Pharmacol. 2008, 51, 178–187. [Google Scholar] [CrossRef]
  10. Achari, A.E.; Jain, S.K. Adiponectin, a Therapeutic Target for Obesity, Diabetes, and Endothelial Dysfunction. Int. J. Mol. Sci. 2017, 18, 1321. [Google Scholar] [CrossRef] [Green Version]
  11. Fang, H.; Judd, R.L. Adiponectin Regulation and Function. Compr. Physiol. 2011, 8, 1031–1063. [Google Scholar]
  12. Xuan, D.; Han, Q.; Tu, Q.; Zhang, L.; Yu, L.; Murry, D.; Tu, T.; Tang, Y.; Lian, J.B.; Stein, G.S.; et al. Epigenetic Modulation in Periodontitis: Interaction of Adiponectin and JMJD3-IRF4 Axis in Macrophages. J. Cell. Physiol. 2015, 231, 1090–1096. [Google Scholar] [CrossRef] [Green Version]
  13. Luo, Y.; Liu, M. Adiponectin: A versatile player of innate immunity. J. Mol. Cell Biol. 2016, 8, 120–128. [Google Scholar] [CrossRef]
  14. Bourlier, V.; Bouloumie, A. Role of macrophage tissue infiltration in obesity and insulin resistance. Diabetes Metab. 2009, 35, 251–260. [Google Scholar] [CrossRef] [PubMed]
  15. Wolf, A.M.; Wolf, D.; Rumpold, H.; Enrich, B.; Tilg, H. Adiponectin induces the anti-inflammatory cytokines IL-10 and IL-1RA in human leukocytes. Biochem. Biophys. Res. Commun. 2004, 323, 630–635. [Google Scholar] [CrossRef] [PubMed]
  16. Okamoto, Y.; Kihara, S.; Ouchi, N.; Nishida, M.; Arita, Y.; Kumada, M.; Ohashi, K.; Sakai, N.; Shimomura, I.; Kobayashi, H.; et al. Adiponectin Reduces Atherosclerosis in Apolipoprotein E-Deficient Mice. Circulation 2002, 106, 2767–2770. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  17. Combs, T.P.; Berg, A.H.; Obici, S.; Scherer, P.E.; Rossetti, L. Endogenous glucose production is inhibited by the adipose-derived protein Acrp30. J. Clin. Investig. 2001, 108, 1875–1881. [Google Scholar] [CrossRef]
  18. Combs, T.P.; Marliss, E.B. Adiponectin signaling in the liver. Rev. Endocr. Metab. Disord. 2014, 15, 137–147. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  19. Schindler, M.; Pendzialek, M.; Grybel, K.J.; Seeling, T.; Gürke, J.; Fischer, B.; Santos, A.N. Adiponectin stimulates lipid metabolism via AMPK in rabbit blastocysts. Hum. Reprod. 2017, 32, 1382–1392. [Google Scholar] [CrossRef] [Green Version]
  20. Yamauchi, T.; Kamon, J.; Minokoshi, Y.; Ito, Y.; Waki, H.; Uchida, S.; Yamashita, S.; Noda, M.; Kita, S.; Ueki, K.; et al. Adiponectin stimulates glucose utilization and fatty-acid oxidation by activating AMP-activated protein kinase. Nat. Med. 2002, 8, 1288–1295. [Google Scholar] [CrossRef] [PubMed]
  21. Rakatzi, I.; Mueller, H.; Ritzeler, O.; Tennagels, N.; Eckel, J. Adiponectin counteracts cytokine- and fatty acid-induced apoptosis in the pancreatic beta-cell line INS-1. Diabetologia 2004, 47, 249–258. [Google Scholar] [CrossRef] [Green Version]
  22. Holland, W.L.; Miller, R.A.; Wang, Z.V.; Sun, K.; Barth, B.M.; Bui, H.H.; Davis, K.E.; Bikman, B.T.; Halberg, N.; Rutkowski, J.M.; et al. Receptor-mediated activation of ceramidase activity initiates the pleiotropic actions of adiponectin. Nat. Med. 2011, 17, 55–63. [Google Scholar] [CrossRef] [Green Version]
  23. Deng, G.; Long, Y.; Yu, Y.R.; Li, M.R. Adiponectin directly improves endothelial dysfunction in obese rats through the AMPK-eNOS Pathway. Int. J. Obes. 2010, 34, 165–171. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  24. Cai, X.; Li, X.; Li, L.; Huang, X.Z.; Liu, Y.S.; Chen, L.; Zhang, K.; Wang, L.; Li, X.; Song, J.; et al. Adiponectin reduces carotid atherosclerotic plaque formation in ApoE-/- mice: Roles of oxidative and nitrosative stress and inducible nitric oxide synthase. Mol. Med. Rep. 2015, 11, 1715–1721. [Google Scholar] [CrossRef] [Green Version]
  25. Nour-Eldine, W.; Ghantous, C.M.; Zibara, K.; Dib, L.; Issaa, H.; Itani, H.A.; El-Zein, N.; Zeidan, A. Adiponectin Attenuates Angiotensin II-Induced Vascular Smooth Muscle Cell Remodeling through Nitric Oxide and the RhoA/ROCK Pathway. Front. Pharmacol. 2016, 7, 86. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  26. Bodles, A.M.; Banga, A.; Rasouli, N.; Ono, F.; Kern, P.A.; Owens, R.J. Pioglitazone increases secretion of high-molecular-weight adiponectin from adipocytes. Am. J. Physiol. Metab. 2006, 291, E1100–E1105. [Google Scholar] [CrossRef] [PubMed]
  27. Chung, L.T.K.; Hosaka, T.; Yoshida, M.; Harada, N.; Sakaue, H.; Sakai, T.; Nakaya, Y. Exendin-4, a GLP-1 receptor agonist, directly induces adiponectin expression through protein kinase A pathway and prevents inflammatory adipokine expression. Biochem. Biophys. Res. Commun 2009, 390, 613–618. [Google Scholar] [CrossRef] [PubMed]
  28. Tahara, A.; Takasu, T.; Yokono, M.; Imamura, M.; Kurosaki, E. Characterization and comparison of sodium-glucose cotransporter 2 inhibitors: Part 2. Antidiabetic effects in type 2 diabetic mice. J. Pharmacol. Sci. 2016, 131, 198–208. [Google Scholar] [CrossRef] [Green Version]
  29. Dormandy, J.A.; Charbonnel, B.; Eckland, D.J.A.; Erdmann, E.; Massi-Benedetti, M.; Moules, I.K.; Skene, A.M.; Tan, M.H.; Lefèbvre, P.J.; Murray, G.D.; et al. Secondary prevention of macrovascular events in patients with type 2 diabetes in the PROactive Study (PROspective pioglitAzone Clinical Trial In macroVascular Events): A randomised controlled trial. Lancet 2005, 366, 1279–1289. [Google Scholar] [CrossRef]
  30. Bethel, M.A.; Patel, R.A.; Merrill, P.; Lokhnygina, Y.; Buse, J.B.; Mentz, R.J.; Pagidipati, N.J.; Chan, J.C.; Gustavson, S.M.; Iqbal, N.; et al. Cardiovascular outcomes with glucagon-like peptide-1 receptor agonists in patients with type 2 diabetes: A meta-analysis. Lancet Diabetes Endocrinol. 2017, 6, 105–113. [Google Scholar] [CrossRef]
  31. Zelniker, T.A.; Wiviott, S.D.; Raz, I.; Im, K.; Goodrich, E.L.; Bonaca, M.P.; Mosenzon, O.; Kato, E.T.; Cahn, A.; Furtado, R.H.; et al. SGLT2 inhibitors for primary and secondary prevention of cardiovascular and renal outcomes in type 2 diabetes: A systematic review and meta-analysis of cardiovascular outcome trials. Lancet 2019, 393, 31–39. [Google Scholar] [CrossRef]
  32. Kliewer, S.A.; Mangelsdorf, D.J. Fibroblast growth factor 21: From pharmacology to physiology. Am. J. Clin. Nutr. 2010, 91, S254–S257. [Google Scholar] [CrossRef] [Green Version]
  33. Holland, W.L.; Adams, A.C.; Brozinick, J.T.; Bui, H.H.; Miyauchi, Y.; Kusminski, C.M.; Bauer, S.M.; Wade, M.; Singhal, E.; Cheng, C.C.; et al. An FGF21-Adiponectin-Ceramide Axis Controls Energy Expenditure and Insulin Action in Mice. Cell Metab. 2013, 17, 790–797. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  34. Lin, Z.; Tian, H.; Lam, K.S.; Lin, S.; Hoo, R.C.; Konishi, M.; Itoh, N.; Wang, Y.; Bornstein, S.R.; Xu, A.; et al. Adiponectin Mediates the Metabolic Effects of FGF21 on Glucose Homeostasis and Insulin Sensitivity in Mice. Cell Metab. 2013, 17, 779–789. [Google Scholar] [CrossRef] [Green Version]
  35. Gariballa, S.; Alkaabi, J.; Yasin, J.; Al Essa, A. Total adiponectin in overweight and obese subjects and its response to visceral fat loss. BMC Endocr. Disord. 2019, 19, 55. [Google Scholar] [CrossRef] [PubMed]
  36. Liu, C.; Feng, X.; Li, Q.; Wang, Y.; Li, Q.; Hua, M. Adiponectin, TNF-α and inflammatory cytokines and risk of type 2 diabetes: A systematic review and meta-analysis. Cytokine 2016, 86, 100–109. [Google Scholar] [CrossRef]
  37. Lindberg, S.; Jensen, J.S.; Pedersen, S.H.; Galatius, S.; Frystyk, J.; Flyvbjerg, A.; Bjerre, M.; Mogelvang, R. Low Adiponectin Levels and Increased Risk of Type 2 Diabetes in Patients with Myocardial Infarction. Diabetes Care 2014, 37, 3003–3008. [Google Scholar] [CrossRef] [Green Version]
  38. Kou, H.; Deng, J.; Gao, D.; Song, A.; Han, Z.; Wei, J.; Jin, X.; Ma, R.; Zheng, Q. Relationship among adiponectin, insulin resistance and atherosclerosis in non-diabetic hypertensive patients and healthy adults. Clin. Exp. Hypertens. 2018, 40, 656–663. [Google Scholar] [CrossRef]
  39. Kim, D.H.; Kim, C.; Ding, E.L.; Townsend, M.K.; Lipsitz, L.A. Adiponectin levels and the risk of hypertension: A systematic review and meta-analysis. Hypertension 2013, 62, 27–32. [Google Scholar] [CrossRef] [Green Version]
  40. Yoshida, H.; Hirowatari, Y.; Kurosawa, H.; Tada, N. Implications of decreased serum adiponectin for type IIb hyperlipidaemia and increased cholesterol levels of very-low-density lipoprotein in type II diabetic patients. Clin. Sci. 2005, 109, 297–302. [Google Scholar] [CrossRef] [Green Version]
  41. Tomono, Y.; Hiraishi, C.; Yoshida, H. Age and sex differences in serum adiponectin and its association with lipoprotein fractions. Ann. Clin. Biochem. Int. J. Lab. Med. 2018, 55, 165–171. [Google Scholar] [CrossRef]
  42. Marsche, G.; Zelzer, S.; Meinitzer, A.; Kern, S.; Meissl, S.; Pregartner, G.; Weghuber, D.; Almer, G.; Mangge, H. Adiponectin Predicts High-Density Lipoprotein Cholesterol Efflux Capacity in Adults Irrespective of Body Mass Index and Fat Distribution. J. Clin. Endocrinol. Metab. 2017, 102, 4117–4123. [Google Scholar] [CrossRef] [PubMed]
  43. Zhao, S.; Kusminski, C.M.; Scherer, P.E. Adiponectin, Leptin and Cardiovascular Disorders. Circ. Res. 2021, 128, 136–149. [Google Scholar] [CrossRef] [PubMed]
  44. Doumatey, A.P.; Bentley, A.R.; Zhou, J.; Huang, H.; Adeyemo, A.; Rotimi, C.N. Paradoxical Hyperadiponectinemia is Associated with the Metabolically Healthy Obese (MHO) Phenotype in African Americans. J. Endocrinol. Metab. 2012, 2, 51–65. [Google Scholar] [CrossRef] [Green Version]
  45. Kobayashi, H.; Ouchi, N.; Kihara, S.; Walsh, K.; Kumada, M.; Abe, Y.; Funahashi, T.; Matsuzawa, Y. Selective Suppression of Endothelial Cell Apoptosis by the High Molecular Weight Form of Adiponectin. Circ. Res. 2004, 94, e27–e31. [Google Scholar] [CrossRef] [PubMed]
  46. Salehi-Abargouei, A.; Izadi, V.; Azadbakht, L. The Effect of Low Calorie Diet on Adiponectin Concentration: A Systematic Review and Meta-Analysis. Horm. Metab. Res. 2015, 47, 549–555. [Google Scholar] [CrossRef]
  47. De Vincentis, A.; Pedone, C.; Gentilucci, U.V.; Picardi, A.; Derosa, G.; Maffioli, P.; Sahebkar, A. Effect of Sibutramine on Plasma C-Reactive Protein, Leptin and Adipon ectin Concentrations: A Systematic Review and Meta-Analysis of Randomized Contr olled Trials. Curr. Pharm. Des. 2017, 23, 870–878. [Google Scholar] [CrossRef]
  48. Garvey, W.T.; Ryan, D.H.; Henry, R.; Bohannon, N.J.; Toplak, H.; Schwiers, M.; Troupin, B.; Day, W.W. Prevention of Type 2 Diabetes in Subjects with Prediabetes and Metabolic Syndrome Treated with Phentermine and Topiramate Extended Release. Diabetes Care 2013, 37, 912–921. [Google Scholar] [CrossRef] [Green Version]
  49. Khosravi-Largani, M.; Nojomi, M.; Aghili, R.; Otaghvar, H.A.; Tanha, K.; Seyedi, S.H.S.; Mottaghi, A. Evaluation of all Types of Metabolic Bariatric Surgery and its Consequences: A Systematic Review and Meta-Analysis. Obes. Surg. 2019, 29, 651–690. [Google Scholar] [CrossRef]
  50. Woodward, L.; Akoumianakis, I.; Antoniades, C. Unravelling the adiponectin paradox: Novel roles of adiponectin in the reg-ulation of cardiovascular disease. Br. J. Pharmacol. 2017, 174, 4007–4020. [Google Scholar] [CrossRef]
  51. Wannamethee, S.G.; Whincup, P.; Lennon, L.; Sattar, N. Circulating Adiponectin Levels and Mortality in Elderly Men with and Without Cardiovascular Disease and Heart Failure. Arch. Intern. Med. 2007, 167, 1510–1517. [Google Scholar] [CrossRef]
  52. McEntegart, M.B.; Awede, B.; Petrie, M.C.; Sattar, N.; Dunn, F.G.; Macfarlane, N.G.; McMurray, J.J. Increase in serum adiponectin concentration in patients with heart failure and cachexia: Relationship with leptin, other cytokines, and B-type natriuretic peptide. Eur. Heart J. 2007, 28, 829–835. [Google Scholar] [CrossRef]
  53. Tindall, C.A.; Dommel, S.; Riedl, V.; Ulbricht, D.; Hanke, S.; Sträter, N.; Heiker, J.T. Membrane Phospholipids and Polyphosphates as Cofactors and Binding Molecules of SERPINA12 (vaspin). Molecules 2020, 25, 1992. [Google Scholar] [CrossRef] [PubMed]
  54. Hida, K.; Wada, J.; Eguchi, J.; Zhang, H.; Baba, M.; Seida, A.; Hashimoto, I.; Okada, T.; Yasuhara, A.; Nakatsuka, A.; et al. Visceral adipose tissue-derived serine protease inhibitor: A unique insulin-sensitizing adipocytokine in obesity. Proc. Natl. Acad. Sci. USA 2005, 102, 10610–10615. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  55. Weiner, J.; Zieger, K.; Pippel, J.; Heiker, J.T. Molecular Mechanisms of Vaspin Action—From Adipose Tissue to Skin and Bone, from Blood Vessels to the Brain. Adv. Exp. Med. Biol. 2019, 1111, 159–188. [Google Scholar] [PubMed] [Green Version]
  56. Kurowska, P.; Mlyczyńska, E.; Dawid, M.; Jurek, M.; Klimczyk, D.; Dupont, J.; Rak, A. Review: Vaspin (SERPINA12) Expression and Function in Endocrine Cells. Cells 2021, 10, 1710. [Google Scholar] [CrossRef]
  57. Heiker, J.T.; Klöting, N.; Kovacs, P.; Kuettner, E.B.; Sträter, N.; Schultz, S.; Kern, M.; Stumvoll, M.; Blüher, M.; Beck-Sickinger, A.G. Vaspin inhibits kallikrein 7 by serpin mechanism. Experientia 2013, 70, 2569–2583. [Google Scholar] [CrossRef] [Green Version]
  58. Ulbricht, D.; Tindall, C.A.; Oertwig, K.; Hanke, S.; Sträter, N.; Heiker, J.T. Kallikrein-related peptidase 14 is the second KLK protease targeted by the serpin vaspin. Biol. Chem. 2018, 399, 1079–1084. [Google Scholar] [CrossRef] [PubMed]
  59. Sato, K.; Shirai, R.; Yamaguchi, M.; Yamashita, T.; Shibata, K.; Okano, T.; Mori, Y.; Matsuyama, T.A.; Ishibashi-Ueda, H.; Hirano, T.; et al. Anti-Atherogenic Effects of Vaspin on Human Aortic Smooth Muscle Cell/Macrophage Responses and Hyper-lipidemic Mouse Plaque Phenotype. Int. J. Mol. Sci. 2018, 19, 1732. [Google Scholar] [CrossRef] [Green Version]
  60. Nakatsuka, A.; Wada, J.; Iseda, I.; Teshigawara, S.; Higashio, K.; Murakami, K.; Kanzaki, M.; Inoue, K.; Terami, T.; Katayama, A.; et al. Vaspin is an adipokine ameliorating ER stress in obesity as a ligand for cell-surface GRP78/MTJ-1 complex. Diabetes 2012, 61, 2823–2832. [Google Scholar] [CrossRef] [Green Version]
  61. Liu, S.; Li, X.; Wu, Y.; Duan, R.; Zhang, J.; Du, F.; Zhang, Q.; Li, Y.; Li, N. Effects of vaspin on pancreatic β cell secretion via PI3K/Akt and NF-κB signaling pathways. PLoS ONE 2017, 12, e0189722. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  62. Luo, X.; Li, K.; Zhang, C.; Yang, G.; Yang, M.; Jia, Y.; Zhang, L.; Ma, Z.A.; Boden, G.; Li, L. Central administration of vaspin inhibits glucose production and augments hepatic insulin signaling in high-fat-diet-fed rat. Int. J. Obes. 2016, 40, 947–954. [Google Scholar] [CrossRef] [PubMed]
  63. Liu, S.; Dong, Y.; Wang, T.; Zhao, S.; Yang, K.; Chen, X.; Zheng, C. Vaspin inhibited proinflammatory cytokine induced activation of nuclear factor-kappa B and its downstream mol-ecules in human endothelial EA.hy926 cells. Diabetes Res. Clin. Pract. 2014, 103, 482–488. [Google Scholar] [CrossRef]
  64. Jung, C.H.; Lee, M.J.; Kang, Y.M.; La Lee, Y.; Yoon, H.K.; Kang, S.-W.; Lee, W.J.; Park, J.-Y. Vaspin inhibits cytokine-induced nuclear factor-kappa B activation and adhesion molecule expression via AMP-activated protein kinase activation in vascular endothelial cells. Cardiovasc. Diabetol. 2014, 13, 41. [Google Scholar] [CrossRef] [Green Version]
  65. Zieger, K.; Weiner, J.; Krause, K.; Schwarz, M.; Kohn, M.; Stumvoll, M.; Blüher, M.; Heiker, J.T. Vaspin suppresses cytokine-induced inflammation in 3T3-L1 adipocytes via inhibition of NFκB pathway. Mol. Cell. Endocrinol. 2018, 460, 181–188. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  66. Sakamoto, Y.; Kameshima, S.; Kakuda, C.; Okamura, Y.; Kodama, T.; Okada, M.; Yamawaki, H. Visceral adipose tissue-derived serine protease inhibitor prevents the development of monocrotaline-induced pulmonary arterial hypertension in rats. Pflugers Arch. 2017, 469, 1425–1432. [Google Scholar] [CrossRef] [PubMed]
  67. Qi, D.; Wang, D.; Zhang, C.; Tang, X.; He, J.; Zhao, Y.; Deng, W.; Deng, X. Vaspin protects against LPS-induced ARDS by inhibiting inflammation, apoptosis and reactive oxygen species gen-eration in pulmonary endothelial cells via the Akt/GSK-3β pathway. Int. J. Mol. Med. 2017, 40, 1803–1817. [Google Scholar]
  68. Jung, C.H.; Lee, W.J.; Hwang, J.Y.; Seol, S.M.; Kim, Y.M.; La Lee, Y.; Park, J.-Y. Vaspin protects vascular endothelial cells against free fatty acid-induced apoptosis through a phosphatidylinositol 3-kinase/Akt pathway. Biochem. Biophys. Res. Commun. 2011, 413, 264–269. [Google Scholar] [CrossRef]
  69. Zhu, X.; Zhang, L.; Chen, Y.; Chen, B.; Huang, H.; Lv, J.; Hu, S.; Shen, J. Vaspin protects mouse mesenchymal stem cells from oxidative stress-induced apoptosis through the MAPK/p38 pathway. Mol. Cell. Biochem. 2019, 462, 107–114. [Google Scholar] [CrossRef]
  70. Nakatsuka, A.; Wada, J.; Iseda, I.; Teshigawara, S.; Higashio, K.; Murakami, K.; Kanzaki, M.; Inoue, K.; Terami, T.; Katayama, A.; et al. Visceral adipose tissue-derived serine proteinase inhibitor inhibits apoptosis of endothelial cells as a ligand for the cell-surface GRP78/voltage-dependent anion channel complex. Circ. Res. 2013, 112, 771–780. [Google Scholar] [CrossRef] [Green Version]
  71. Jung, C.H.; Lee, W.J.; Hwang, J.Y.; Lee, M.J.; Seol, S.M.; Kim, Y.M.; La Lee, Y.; Kim, H.S.; Kim, M.-S.; Park, J.-Y. Vaspin Increases Nitric Oxide Bioavailability through the Reduction of Asymmetric Dimethylarginine in Vascular Endothelial Cells. PLoS ONE 2012, 7, e52346. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  72. Klöting, N.; Kovacs, P.; Kern, M.; Heiker, J.T.; Fasshauer, M.; Schön, M.R.; Stumvoll, M.; Beck-Sickinger, A.G.; Blüher, M. Central vaspin administration acutely reduces food intake and has sustained blood glucose-lowering effects. Diabetologia 2011, 54, 1819–1823. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  73. Brunetti, L.; Di Nisio, C.; Recinella, L.; Chiavaroli, A.; Leone, S.; Ferrante, C.; Orlando, G.; Vacca, M. Effects of vaspin, chemerin and omentin-1 on feeding behavior and hypothalamic peptide gene expression in the rat. Peptides 2011, 32, 1866–1871. [Google Scholar] [CrossRef] [PubMed]
  74. Gao, J.H.; Zeng, M.Y.; Yu, X.H.; Zeng, G.F.; He, L.H.; Zheng, X.L.; Zhang, D.W.; Ouyang, X.P.; Tang, C.K. Visceral adipose tissue-derived serine protease inhibitor accelerates cholesterol efflux by up-regulating ABCA1 expression via the NF-κB/miR-33a pathway in THP-1 macropahge-derived foam cells. Biochem. Biophys. Res. Commun. 2018, 500, 318–324. [Google Scholar] [CrossRef]
  75. Taheri, E.; Hosseini, S.; Qorbani, M.; Mirmiran, P. Association of adipocytokines with lipid and glycemic profiles in women with normal weight obesity. BMC Endocr. Disord. 2020, 20, 171. [Google Scholar] [CrossRef] [PubMed]
  76. Feng, R.; Li, Y.; Wang, C.; Luo, C.; Liu, L.; Chuo, F.; Li, Q.; Sun, C. Higher vaspin levels in subjects with obesity and type 2 diabetes mellitus: A meta-analysis. Diabetes Res. Clin. Pract. 2014, 106, 88–94. [Google Scholar] [CrossRef] [PubMed]
  77. Hao, F.; Zhang, H.; Zhu, J.; Kuang, H.; Yu, Q.; Bai, M.; Mu, J. Association between vaspin level and coronary artery disease in patients with type 2 diabetes. Diabetes Res. Clin. Pract. 2016, 113, 26–32. [Google Scholar] [CrossRef]
  78. Yang, H.; Huang, Y.; Gai, C.; Chai, G.; Lee, S. Serum vaspin levels are positively associated with diabetic retinopathy in patients with type 2 diabetes mellitus. J. Diabetes Investig. 2020, 12, 566–573. [Google Scholar] [CrossRef] [PubMed]
  79. Jia, G.Y.; Han, T.; Gao, L.; Wang, L.; Wang, S.C.; Yang, L.; Zhang, J.; Guan, Y.Y.; Yan, N.N.; Yu, H.Y.; et al. Effect of aerobic exercise and resistance exercise in improving non-alcoholic fatty liver disease: A randomized controlled trial. Zhonghua Gan Zang Bing Za Zhi 2018, 26, 34–41. [Google Scholar]
  80. Tan, B.K.; Heutling, D.; Chen, J.; Farhatullah, S.; Adya, R.; Keay, S.D.; Kennedy, C.R.; Lehnert, H.; Randeva, H.S. Metformin Decreases the Adipokine Vaspin in Overweight Women with Polycystic Ovary Syndrome Concomitant with Improvement in Insulin Sensitivity and a Decrease in Insulin Resistance. Diabetes 2008, 57, 1501–1507. [Google Scholar] [CrossRef] [Green Version]
  81. Zhang, L.; Li, L.; Yang, M.; Liu, H.; Yang, G. Elevated circulating vaspin levels were decreased by rosiglitazone therapy in T2DM patients with poor glycemic control on metformin alone. Cytokine 2011, 56, 399–402. [Google Scholar] [CrossRef]
  82. Golpaie, A.; Tajik, N.; Masoudkabir, F.; Karbaschian, Z.; Talebpour, M.; Hosseini, M.; Hosseinzadeh-Attar, M.J. Short-term effect of weight loss through restrictive bariatric surgery on serum levels of vaspin in morbidly obese subjects. Eur. Cytokine Netw. 2011, 22, 181–186. [Google Scholar] [CrossRef] [Green Version]
  83. Li, J.; Li, Q.; Zhu, Y.; Wang, Y.; Gao, C.; Li, X.; Ji, T.; Bai, S. Association of vaspin rs2236242 gene variants with type 2 diabetes and obesity in a Chinese population: A prospective, single-center study. J. Cell. Physiol. 2019, 234, 16097–16101. [Google Scholar] [CrossRef] [PubMed]
  84. Kadoglou, N.P.; Kassimis, G.; Patsourakos, N.; Kanonidis, I.; Valsami, G. Omentin-1 and vaspin serum levels in patients with pre-clinical carotid atherosclerosis and the effect of statin therapy on them. Cytokine 2021, 138, 155364. [Google Scholar] [CrossRef]
  85. Al-Kuraishy, H.M.; Al-Gareeb, A.I.; Al-Buhadilly, A.K. Rosuvastatin Improves Vaspin Serum Levels in Obese Patients with Acute Coronary Syndrome. Diseases 2018, 6, 9. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  86. Kastl, S.P.; Katsaros, K.M.; Krychtiuk, K.A.; Jägersberger, G.; Kaun, C.; Huber, K.; Wojta, J.; Speidl, W.S. The adipokine vaspin is associated with decreased coronary in-stent restenosis in vivo and inhibits migration of human coronary smooth muscle cells in vitro. PLoS ONE 2020, 15, e0232483. [Google Scholar] [CrossRef]
  87. Zhang, B.; Peng, W.; Wang, K.; Li, H.; Xu, Y. Vaspin as a Prognostic Marker in Patients with Acute Myocardial Infarction. Heart Lung Circ. 2016, 25, 257–264. [Google Scholar] [CrossRef]
  88. Ji, S.; Kou, W.; Luan, P.; Jian, W.; Zhuang, J.; Xu, X.; Zhao, Y.; Li, H.; Peng, W. Plasma vaspin is an effective biomarker for evaluation of future cardiovascular events in patients with chest pain: A 5-year retrospective observational study. Ann. Transl. Med. 2020, 8, 479. [Google Scholar] [CrossRef]
  89. Rashad, N.M.; Ahmed, H.S.; Ashour, W.M.R.; Yousef, M.S. Association of vaspin gene expression and its serum level on the risk of ischemic stroke in type 2 diabetic Egyptian patients: Prospective case-control study. Biotechnol. Appl. Biochem. 2020, 67, 912–919. [Google Scholar] [CrossRef] [PubMed]
  90. Cakal, E.; Ustun, Y.; Engin-Ustun, Y.; Ozkaya, M.; Kilinç, M. Serum vaspin and C-reactive protein levels in women with polycystic ovaries and polycystic ovary syndrome. Gynecol. Endocrinol. 2011, 27, 491–495. [Google Scholar] [CrossRef]
  91. Esteghamati, A.; Noshad, S.; Mousavizadeh, M.; Zandieh, A.; Nakhjavani, M. Association of vaspin with metabolic syndrome: The pivotal role of insulin resistance. Diabetes Metab. J. 2014, 38, 143–149. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  92. Buyukinan, M.; Atar, M.; Can, U.; Pirgon, O.; Guzelant, A.; Deniz, I. The Association Between Serum Vaspin and Omentin-1 Levels in Obese Children with Metabolic Syndrome. Metab. Syndr. Relat. Disord. 2018, 16, 76–81. [Google Scholar] [CrossRef]
  93. Si, Y.; Fan, W.; Sun, L. A Review of the Relationship Between CTRP Family and Coronary Artery Disease. Curr. Atheroscler. Rep. 2020, 22, 1–7. [Google Scholar] [CrossRef]
  94. Xie, Y.; Meng, Z.; Gao, J.; Liu, C.; Wang, J.; Guo, R.; Zhao, J.; Lopez, B.; Christopher, T.; Lee, D.; et al. C1q Complement/Tumor Necrosis Factor-Associated Proteins in Cardiovascular Disease and COVID-19. Proteomes 2021, 9, 12. [Google Scholar] [CrossRef]
  95. Wong, G.W.; Krawczyk, S.A.; Kitidis-Mitrokostas, C.; Ge, G.; Spooner, E.; Hug, C.; Gimeno, R.; Lodish, H.F. Identification and characterization of CTRP9, a novel secreted glycoprotein, from adipose tissue that reduces serum glucose in mice and forms heterotrimers with adiponectin. FASEB J. 2009, 23, 241–258. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  96. Yuan, Y.; Lau, W.B.; Su, H.; Sun, Y.; Yi, W.; Du, Y.; Christopher, T.; Lopez, B.; Wang, Y.; Ma, X.L. C1q-TNF-related protein-9, a novel cardioprotetcive cardiokine, requires proteolytic cleavage to generate a bio-logically active globular domain isoform. Am. J. Physiol. Endocrinol. Metab. 2015, 308, E891–E898. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  97. Su, H.; Yuan, Y.; Wang, X.M.; Lau, W.B.; Wang, Y.; Wang, X.; Gao, E.; Koch, W.J.; Ma, X.L. Inhibition of CTRP9, a novel and cardiac-abundantly expressed cell survival molecule, by TNFα-initiated oxidative signaling contributes to exacerbated cardiac injury in diabetic mice. Basic Res. Cardiol. 2013, 108, 315. [Google Scholar] [CrossRef]
  98. Yu, X.-H.; Zhang, D.; Zheng, X.-L.; Tang, C.-K. C1q tumor necrosis factor-related protein 9 in atherosclerosis: Mechanistic insights and therapeutic potential. Atherosclerosis 2018, 276, 109–116. [Google Scholar] [CrossRef]
  99. Yan, W.; Guo, Y.; Tao, L.; Lau, W.B.; Gan, L.; Yan, Z.; Guo, R.; Gao, E.; Wong, G.W.; Koch, W.L.; et al. C1q/Tumor Necrosis Factor-Related Protein-9 Regulates the Fate of Implanted Mesenchymal Stem Cells and Mobilizes Their Protective Effects Against Ischemic Heart Injury via Multiple Novel Signaling Pathways. Circulation 2017, 136, 2162–2177. [Google Scholar] [CrossRef]
  100. Liu, Q.; Zhang, H.; Lin, J.; Zhang, R.; Chen, S.; Liu, W.; Sun, M.; Du, W.; Hou, J.; Yu, B. C1q/TNF-related protein 9 inhibits the cholesterol-induced Vascular smooth muscle cell phenotype switch and cell dysfunction by activating AMP-dependent kinase. J. Cell. Mol. Med. 2017, 21, 2823–2836. [Google Scholar] [CrossRef]
  101. Jung, C.H.; Lee, M.J.; Kang, Y.M.; La Lee, Y.; Seol, S.M.; Yoon, H.K.; Kang, S.-W.; Lee, W.J.; Park, J.-Y. C1q/TNF-related protein-9 inhibits cytokine-induced vascular inflammation and leukocyte adhesiveness via AMP-activated protein kinase activation in endothelial cells. Mol. Cell. Endocrinol. 2016, 419, 235–243. [Google Scholar] [CrossRef]
  102. Liu, M.; Yin, L.; Li, W.; Hu, J.; Wang, H.; Ye, B.; Tang, Y.; Huang, C. C1q/TNF-related protein-9 promotes macrophage polarization and improves cardiac dysfunction after myocardial infarction. J. Cell. Physiol. 2019, 234, 18731–18747. [Google Scholar] [CrossRef] [Green Version]
  103. Wei, Z.; Lei, X.; Petersen, P.S.; Aja, S.; Wong, G.W. Targeted deletion of C1q/TNF-related protein 9 increases food intake, decreases insulin sensitivity, and promotes hepatic steatosis in mice. Am. J. Physiol. Endocrinol. Metab. 2014, 306, E779–E790. [Google Scholar] [CrossRef] [Green Version]
  104. Yang, G.; Qin, C.; Wang, B.; Jia, J.; Yuan, X.; Sun, C.; Li, W. Molecular identification and functional analysis of Ctrp9 in Epinephelus coioides. J. Mol. Endocrinol. 2017, 58, 179–191. [Google Scholar] [CrossRef]
  105. Zhang, L.; Liu, Q.; Zhang, H.; Wang, X.-D.; Chen, S.-Y.; Yang, Y.; Lv, H.; Hou, J.-B.; Yu, B. C1q/TNF-Related Protein 9 Inhibits THP-1 Macrophage Foam Cell Formation by Enhancing Autophagy. J. Cardiovasc. Pharmacol. 2018, 72, 167–175. [Google Scholar] [CrossRef] [PubMed]
  106. Lei, S.; Chen, J.; Song, C.; Li, J.; Zuo, A.; Xu, D.; Li, T.; Guo, Y. CTRP9 alleviates foam cells apoptosis by enhancing cholesterol efflux. Mol. Cell. Endocrinol. 2021, 522, 111138. [Google Scholar] [CrossRef] [PubMed]
  107. Uemura, Y.; Shibata, R.; Ohashi, K.; Enomoto, T.; Kambara, T.; Yamamoto, T.; Ogura, Y.; Yuasa, D.; Joki, Y.; Matsuo, K.; et al. Adipose-derived factor CTRP9 attenuates vascular smooth muscle cell proliferation and neointimal formation. FASEB J. 2013, 27, 25–33. [Google Scholar] [CrossRef]
  108. Li, Y.X.; Run, L.; Shi, T.; Zhang, Y.J. CTRP9 regulates hypoxia-mediated human pulmonary artery smooth muscle cell proliferation, apoptosis and migration via TGF-β1/ERK1/2 signaling pathway. Biochem. Biophys. Res. Commun. 2017, 490, 1319–1325. [Google Scholar] [CrossRef]
  109. Wang, W.; Lau, W.B.; Wang, Y.; Ma, X.; Li, R. Reduction of CTRP9, a novel anti-platelet adipokine, contributes to abnormal platelet activity in diabetic animals. Cardiovasc. Diabetol. 2016, 15, 6. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  110. Zheng, Q.; Yuan, Y.; Yi, W.; Lau, W.B.; Wang, Y.; Wang, X.; Sun, Y.; Lopez, B.L.; Christopher, T.A.; Peterson, J.M.; et al. C1q/TNF-related proteins, a family of novel adipokines, induce vascular relaxation through the adiponectin receptor-1/AMPK/eNOS/nitric oxide signaling pathway. Arterioscler. Thromb. Vasc. Biol. 2011, 31, 2616–2623. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  111. Yamaguchi, S.; Shibata, R.; Ohashi, K.; Enomoto, T.; Ogawa, H.; Otaka, N.; Hiramatsu-Ito, M.; Masutomi, T.; Kawanishi, H.; Murohara, T.; et al. C1q/TNF-Related Protein 9 Promotes Revascularization in Response to Ischemia via an eNOS-Dependent Manner. Front. Pharmacol. 2020, 11, 1313. [Google Scholar] [CrossRef]
  112. Li, Y.; Geng, X.; Wang, H.; Cheng, G.; Xu, S. CTRP9 Ameliorates Pulmonary Arterial Hypertension Through Attenuating Inflammation and Improving Endothelial Cell Survival and Function. J. Cardiovasc. Pharmacol. 2016, 67, 394–401. [Google Scholar] [CrossRef] [PubMed]
  113. Cheng, L.; Li, B.; Chen, X.; Su, J.; Wang, H.; Yu, S.; Zheng, Q. CTRP9 induces mitochondrial biogenesis and protects high glucose-induced endothelial oxidative damage via AdipoR1 -SIRT1- PGC-1α activation. Biochem. Biophys. Res. Commun. 2016, 477, 685–691. [Google Scholar] [CrossRef] [PubMed]
  114. Sun, H.; Zhu, X.; Zhou, Y.; Cai, W.; Qiu, L. C1q/TNF-Related Protein-9 Ameliorates Ox-LDL-Induced Endothelial Dysfunction via PGC-1α/AMPK-Mediated Antioxidant Enzyme Induction. Int. J. Mol. Sci. 2017, 18, 1097. [Google Scholar] [CrossRef] [Green Version]
  115. Niemann, B.; Li, L.; Siegler, D.; Siegler, B.H.; Knapp, F.; Hanna, J.; Aslam, M.; Kracht, M.; Schulz, R.; Rohrbach, S. CTRP9 Mediates Protective Effects in Cardiomyocytes via AMPK- and Adiponectin Receptor-Mediated Induction of Anti-Oxidant Response. Cells 2020, 9, 1229. [Google Scholar] [CrossRef]
  116. Song, C.X.; Chen, J.Y.; Li, N.; Guo, Y. CTRP9 Enhances Efferocytosis in Macrophages via MAPK/Drp1-Mediated Mitochondrial Fission and Adi-poR1-Induced Immunometabolism. J. Inflamm. Res. 2021, 14, 1007–1017. [Google Scholar] [CrossRef]
  117. Lee, J.; Yoo, J.H.; Kim, H.S.; Cho, Y.K.; La Lee, Y.; Lee, W.J.; Park, J.-Y.; Jung, C.H. C1q/TNF-related protein-9 attenuates palmitic acid-induced endothelial cell senescence via increasing autophagy. Mol. Cell. Endocrinol. 2021, 521, 111114. [Google Scholar] [CrossRef]
  118. Sun, Y.; Yi, W.; Yuan, Y.; Lau, W.B.; Yi, D.; Wang, X.; Wang, Y.; Su, H.; Wang, X.; Gao, E.; et al. C1q/Tumor Necrosis Factor–Related Protein-9, a Novel Adipocyte-Derived Cytokine, Attenuates Adverse Remodeling in the Ischemic Mouse Heart via Protein Kinase a Activation. Circulation 2013, 128 (Suppl. 1), S113–S120. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  119. Wang, J.; Hang, T.; Cheng, X.M.; Li, D.M.; Zhang, Q.G.; Wang, L.J.; Peng, Y.P.; Gong, J.B. Associations of C1q/TNF-Related Protein-9 Levels in Serum and Epicardial Adipose Tissue with Coronary Atherosclerosis in Humans. Biomed. Res. Int. 2015, 2015, 971683. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  120. Moradi, N.; Fadaei, R.; Emamgholipour, S.; Kazemian, E.; Panahi, G.; Vahedi, S.; Saed, L.; Fallah, S. Association of circulating CTRP9 with soluble adhesion molecules and inflammatory markers in patients with type 2 diabetes mellitus and coronary artery disease. PLoS ONE 2018, 13, e0192159. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  121. Gao, C.; Zhao, S.; Lian, K.; Mi, B.; Si, R.; Tan, Z.; Fu, F.; Wang, S.; Wang, R.; Ma, X.; et al. C1q/TNF-related protein 3 (CTRP3) and 9 (CTRP9) concentrations are decreased in patients with heart failure and are associated with increased morbidity and mortality. BMC Cardiovasc. Disord. 2019, 19, 139. [Google Scholar] [CrossRef] [PubMed]
  122. Miyatake, N.; Adachi, H.; Nomura-Nakayama, K.; Okada, K.; Okino, K.; Hayashi, N.; Fujimoto, K.; Furuichi, K.; Yokoyama, H. Circulating CTRP9 correlates with the prevention of aortic calcification in renal allograft recipients. PLoS ONE 2020, 15, e0226526. [Google Scholar] [CrossRef] [Green Version]
  123. Pan, J.; Cui, X.; Wang, G.; Xue, K.; Hu, J.; Zhou, L. Predictive value of serum CTRP9 and STIM1 for restenosis after cerebrovascular stent implantation and its relationship with vasoactive substances and inflammatory cytokines. Exp. Ther. Med. 2020, 20, 2617–2622. [Google Scholar] [CrossRef] [PubMed]
  124. Jia, Y.; Luo, X.; Ji, Y.; Xie, J.; Jiang, H.; Fu, M.; Li, X. Circulating CTRP9 levels are increased in patients with newly diagnosed type 2 diabetes and correlated with insulin resistance. Diabetes Res. Clin. Pract. 2017, 131, 116–123. [Google Scholar] [CrossRef]
  125. Hwang, Y.-C.; Oh, S.W.; Park, S.-W.; Park, C.-Y. Association of serum C1q/TNF-Related Protein-9 (CTRP9) concentration with visceral adiposity and metabolic syndrome in humans. Int. J. Obes. 2014, 38, 1207–1212. [Google Scholar] [CrossRef] [PubMed]
  126. Wolf, R.M.; Steele, K.E.; Peterson, L.A.; Zeng, X.; Jaffe, A.; Schweitzer, M.A.; Magnuson, T.H.; Wong, G.W. C1q/TNF-Related Protein-9 (CTRP9) Levels Are Associated with Obesity and Decrease Following Weight Loss Surgery. J. Clin. Endocrinol. Metab. 2016, 101, 2211–2217. [Google Scholar] [CrossRef]
  127. Xia, L.; Zhang, H.; Shi, Q.; Zhang, X.; Wang, C.; Lin, G. Protective Role of CTRP3 and CTRP9 in the Development of Gestational Diabetes Mellitus. Clin. Lab. 2020, 66. [Google Scholar] [CrossRef] [PubMed]
  128. Yang, J.; Zhao, D.; Chen, Y.; Ma, Y.; Shi, X.; Wang, X.; Lv, Y.; Yuan, H. Association of serum CTRP9 levels with cardiac autonomic neuropathy in patients with type 2 diabetes mellitus. J. Diabetes Investig. 2021, 12, 1442–1451. [Google Scholar] [CrossRef]
  129. Asada, M.; Morioka, T.; Yamazaki, Y.; Kakutani, Y.; Kawarabayashi, R.; Motoyama, K.; Mori, K.; Fukumoto, S.; Shioi, A.; Shoji, T.; et al. Plasma C1q/TNF-Related Protein-9 Levels are Associated with Atherosclerosis in Patients with Type 2 Diabetes without Renal Dysfunction. J. Diabetes Res. 2016, 2016, 8624313. [Google Scholar] [CrossRef] [Green Version]
  130. Jung, C.H.; Lee, M.J.; Kang, Y.M.; Jang, J.E.; Leem, J.; La Lee, Y.; Seol, S.M.; Yoon, H.K.; Lee, W.J.; Park, J.-Y. Association of Serum C1q/TNF-Related Protein-9 Concentration with Arterial Stiffness in Subjects with Type 2 Diabetes. J. Clin. Endocrinol. Metab. 2014, 99, E2477–E2484. [Google Scholar] [CrossRef] [Green Version]
  131. Na, N.; Ji, M. Role of First-Trimester Serum C1q/TNF-Related Protein 9 in Gestational Diabetes Mellitus. Clin. Lab. 2020, 66. [Google Scholar] [CrossRef] [PubMed]
  132. Chen, L.; Qin, L.; Liu, X.; Meng, X. CTRP3 Alleviates Ox-LDL-Induced Inflammatory Response and Endothelial Dysfunction in Mouse Aortic Endo-thelial Cells by Activating the PI3K/Akt/eNOS Pathway. Inflammation 2019, 42, 1350–1359. [Google Scholar] [CrossRef]
  133. Fadaei, R.; Moradi, N.; Kazemi, T.; Chamani, E.; Azdaki, N.; Moezibady, S.A.; Shahmohamadnejad, S.; Fallah, S. Decreased serum levels of CTRP12/adipolin in patients with coronary artery disease in relation to inflammatory cytokines and insulin resistance. Cytokine 2019, 113, 326–331. [Google Scholar] [CrossRef] [PubMed]
  134. Wang, G.; Chen, J.-J.; Deng, W.-Y.; Ren, K.; Yin, S.-H.; Yu, X.-H. CTRP12 ameliorates atherosclerosis by promoting cholesterol efflux and inhibiting inflammatory response via the miR-155-5p/LXRα pathway. Cell Death Dis. 2021, 12, 254. [Google Scholar] [CrossRef]
  135. Peterson, J.; Wei, Z.; Wong, G.W. C1q/TNF-related Protein-3 (CTRP3), a Novel Adipokine That Regulates Hepatic Glucose Output. J. Biol. Chem. 2010, 285, 39691–39701. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  136. Wei, Z.; Peterson, J.M.; Wong, G.W. Metabolic regulation by C1q/TNF-related protein-13 (CTRP13): Activation of AMP-activated protein kinase and suppression of fatty acid-induced JNK signaling. J. Biol. Chem. 2011, 286, 15652–15665. [Google Scholar] [CrossRef] [Green Version]
  137. Wei, Z.; Peterson, J.; Lei, X.; Cebotaru, L.; Wolfgang, M.J.; Baldeviano, G.C.; Wong, G.W. C1q/TNF-related Protein-12 (CTRP12), a Novel Adipokine That Improves Insulin Sensitivity and Glycemic Control in Mouse Models of Obesity and Diabetes*. J. Biol. Chem. 2012, 287, 10301–10315. [Google Scholar] [CrossRef] [Green Version]
  138. Ogawa, H.; Ohashi, K.; Ito, M.; Shibata, R.; Kanemura, N.; Yuasa, D.; Kambara, T.; Matsuo, K.; Hayakawa, S.; Hiramatsu-Ito, M.; et al. Adipolin/CTRP12 protects against pathological vascular remodelling through suppression of smooth muscle cell growth and macrophage inflammatory response. Cardiovasc. Res. 2020, 116, 237–249. [Google Scholar] [CrossRef]
  139. Wang, C.; Xu, W.; Liang, M.; Huang, D.; Huang, K. CTRP13 inhibits atherosclerosis via autophagy-lysosome-dependent degradation of CD36. FASEB J. 2019, 33, 2290–2300. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  140. Zhang, Z.; Zhu, L.; Feng, P.; Tan, Y.; Zhang, B.; Gao, E.; Wang, X.; Fan, C.; Wang, X.; Yi, W.; et al. C1q/tumor necrosis factor-related protein-3-engineered mesenchymal stromal cells attenuate cardiac impairment in mice with myocardial infarction. Cell Death Dis. 2019, 10, 530. [Google Scholar] [CrossRef] [PubMed]
  141. Wu, D.; Lei, H.; Wang, J.Y.; Zhang, C.L.; Feng, H.; Fu, F.Y.; Li, L.; Wu, L.L. CTRP3 attenuates post-infarct cardiac fibrosis by targeting Smad3 activation and inhibiting myofibroblast differentiation. J. Mol. Med. 2015, 93, 1311–1325. [Google Scholar] [CrossRef] [PubMed]
  142. Yi, W.; Sun, Y.; Yuan, Y.; Lau, W.B.; Zheng, Q.; Wang, X.; Wang, Y.; Shang, X.; Gao, E.; Koch, W.J.; et al. C1q/Tumor Necrosis Factor-Related Protein-3, a Newly Identified Adipokine, Is a Novel Antiapoptotic, Proangiogenic, and Cardioprotective Molecule in the Ischemic Mouse Heart. Circulation 2012, 125, 3159–3169. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  143. Ma, Z.-G.; Yuan, Y.-P.; Zhang, X.; Xu, S.-C.; Kong, C.-Y.; Song, P.; Li, N.; Tang, Q.-Z. C1q-tumour necrosis factor-related protein-3 exacerbates cardiac hypertrophy in mice. Cardiovasc. Res. 2019, 115, 1067–1077. [Google Scholar] [CrossRef]
  144. Takikawa, T.; Ohashi, K.; Ogawa, H.; Otaka, N.; Kawanishi, H.; Fang, L.; Ozaki, Y.; Eguchi, S.; Tatsumi, M.; Takefuji, M.; et al. Adipolin/C1q/Tnf-related protein 12 prevents adverse cardiac remodeling after myocardial infarction. PLoS ONE 2020, 15, e0243483. [Google Scholar] [CrossRef]
  145. Zhou, W.; Wang, Y.; Wu, Y.; Yang, J.; Xu, L.; Yang, Y. Serum CTRP3 level is inversely associated with nonalcoholic fatty liver disease: A 3-y longitudinal study. Clin. Chim. Acta 2018, 479, 79–83. [Google Scholar] [CrossRef]
  146. Choi, K.M.; Hwang, S.Y.; Hong, H.C.; Yang, S.J.; Choi, H.Y.; Yoo, H.J.; Lee, K.W.; Nam, M.S.; Park, Y.S.; Woo, J.T.; et al. C1q/TNF-Related Protein-3 (CTRP-3) and Pigment Epithelium-Derived Factor (PEDF) Concentrations in Patients with Type 2 Diabetes and Metabolic Syndrome. Diabetes 2012, 61, 2932–2936. [Google Scholar] [CrossRef] [Green Version]
  147. Moradi, N.; Fadaei, R.; Khamseh, M.E.; Nobakht, A.; Rezaei, M.J.; Aliakbari, F.; Vatannejad, A.; Hosseini, J. Serum levels of CTRP3 in diabetic nephropathy and its relationship with insulin resistance and kidney function. PLoS ONE 2019, 14, e0215617. [Google Scholar] [CrossRef] [Green Version]
  148. Flehmig, G.; Scholz, M.; Klöting, N.; Fasshauer, M.; Tönjes, A.; Stumvoll, M.; Youn, B.-S.; Blüher, M. Identification of Adipokine Clusters Related to Parameters of Fat Mass, Insulin Sensitivity and Inflammation. PLoS ONE 2014, 9, e99785. [Google Scholar] [CrossRef]
  149. Tan, B.K.; Chen, J.; Hu, J.; Amar, O.; Mattu, H.S.; Ramanjaneya, M.; Patel, V.; Lehnert, H.; Randeva, H.S. Circulatory changes of the novel adipokine adipolin/CTRP12 in response to metformin treatment and an oral glucose challenge in humans. Clin. Endocrinol. 2014, 81, 841–846. [Google Scholar] [CrossRef] [PubMed]
  150. Shanaki, M.; Fadaei, R.; Moradi, N.; Emamgholipour, S.; Poustchi, H. The Circulating CTRP13 in Type 2 Diabetes and Non-Alcoholic Fatty Liver Patients. PLoS ONE 2016, 11, e0168082. [Google Scholar] [CrossRef]
  151. Choi, K.M.; Hwang, S.Y.; Hong, H.C.; Choi, H.Y.; Yoo, H.J.; Youn, B.-S.; Baik, S.H.; Seo, H.S. Implications of C1q/TNF-related protein-3 (CTRP-3) and progranulin in patients with acute coronary syndrome and stable angina pectoris. Cardiovasc. Diabetol. 2014, 13, 14. [Google Scholar] [CrossRef] [Green Version]
  152. Jiang, H.; Wang, M.; Ye, J.; Liu, J.; Wang, Z.; Xu, Y.; Ye, D.; Wan, J. Serum Levels of Complement-C1q/Tumor Necrosis Factor-Related Protein-3 Decreased in Patients with Acute Aortic Dissection. Am. J. Cardiol. 2018, 122, 1244–1248. [Google Scholar] [CrossRef] [PubMed]
  153. Yildirim, A.; Sumbul, H.E.; Koca, H.; Kucukosmanoglu, M.; Icen, Y.K.; Koc, M. Complement C1q/Tumor Necrosis Factor-Related Protein-3 (CTRP3) is Significantly Decreased in Patients with Heart Failure and Closely Related with Ventricular Tachycardia. Acta. Cardiol. Sin. 2021, 37, 278–285. [Google Scholar] [PubMed]
  154. Shahraki, Z.N.; Azimi, H.; Ilchi, N.; Borj, M.R.; Pourghadamyari, H.; Mosallanejad, S.; Abbaszadeh-Goudarzi, K.; Sahebkar, A. Circulating C1q/TNF-related protein-12 levels are associated with the severity of coronary artery disease. Cytokine 2021, 144, 155545. [Google Scholar] [CrossRef]
  155. Schumaker, V.N.; Zavodszky, P.; Poon, P.H. Activation of the first component of complement. Annu. Rev. Immunol. 1987, 5, 21–42. [Google Scholar] [CrossRef]
  156. Hirata, A.; Kishida, K.; Kobayashi, H.; Nakatsuji, H.; Funahashi, T.; Shimomura, I. Correlation between serum C1q-adiponectin/total adiponectin ratio and polyvascular lesions detected by vascular ultrasonography in Japanese type 2 diabetics. Metabolism 2013, 62, 376–385. [Google Scholar] [CrossRef] [PubMed]
  157. Hirata, A.; Kishida, K.; Nakatsuji, H.; Kobayashi, H.; Funahashi, T.; Shimomura, I. High serum C1q-adiponectin/total adiponectin ratio correlates with coronary artery disease in Japanese type 2 diabetics. Metabolism 2013, 62, 578–585. [Google Scholar] [CrossRef]
  158. Kishida, K.; Nakagawa, Y.; Kobayashi, H.; Yanagi, K.; Funahashi, T.; Shimomura, I. Increased serum C1q-binding adiponectin complex to total-adiponectin ratio in men with multi-vessel coronary disease. Diabetol. Metab. Syndr. 2014, 6, 64. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  159. Hong, E.S.; Lim, C.; Choi, H.Y.; Ku, E.J.; Kim, K.M.; Moon, J.H.; Lim, S.; Park, K.S.; Jang, H.C.; Choi, S.H. The amount of C1q–adiponectin complex is higher in the serum and the complex localizes to perivascular areas of fat tissues and the intimal–medial layer of blood vessels of coronary artery disease patients. Cardiovasc. Diabetol. 2015, 14, 50. [Google Scholar] [CrossRef] [Green Version]
  160. Bilkovski, R.; Schulte, D.M.; Oberhauser, F.; Mauer, J.; Hampel, B.; Gutschow, C.; Krone, W.; Laudes, M. Adipose tissue macrophages inhibit adipogenesis of mesenchymal precursor cells via wnt-5a in humans. Int. J. Obes. 2011, 35, 1450–1454. [Google Scholar] [CrossRef] [Green Version]
  161. Pashirzad, M.; Shafiee, M.; Rahmani, F.; Behnam-Rassouli, R.; Hoseinkhani, F.; Ryzhikov, M.; Binabaj, M.M.; Parizadeh, M.R.; Avan, A.; Hassanian, S.M. Role of Wnt5a in the Pathogenesis of Inflammatory Diseases. J. Cell. Physiol. 2017, 232, 1611–1616. [Google Scholar] [CrossRef] [PubMed]
  162. Ouchi, N.; Higuchi, A.; Ohashi, K.; Oshima, Y.; Gokce, N.; Shibata, R.; Akasaki, Y.; Shimono, A.; Walsh, K. Sfrp5 Is an Anti-Inflammatory Adipokine That Modulates Metabolic Dysfunction in Obesity. Science 2010, 329, 454–457. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  163. Rydzewska, M.; Nikołajuk, A.; Matulewicz, N.; Stefanowicz, M.; Karczewska-Kupczewska, M. Serum secreted frizzled-related protein 5 in relation to insulin sensitivity and its regulation by insulin and free fatty acids. Endocrine 2021, 74, 300–307. [Google Scholar] [CrossRef]
  164. Tong, S.; Ji, Q.; Du, Y.; Zhu, X.; Zhu, C.; Zhou, Y. Sfrp5/Wnt Pathway: A Protective Regulatory System in Atherosclerotic Cardiovascular Disease. J. Interferon Cytokine Res. 2019, 39, 472–482. [Google Scholar] [CrossRef] [Green Version]
  165. Wang, D.; Zhang, Y.; Shen, C. Research update on the association between SFRP5, an anti-inflammatory adipokine, with obesity, type 2 diabetes mellitus and coronary heart disease. J. Cell. Mol. Med. 2020, 24, 2730–2735. [Google Scholar] [CrossRef]
  166. Sun, M.; Wang, W.; Min, L.; Chen, C.; Li, Q.; Weng, W. Secreted frizzled-related protein 5 (SFRP5) protects ATDC5 cells against LPS-induced inflammation and apoptosis via inhibiting Wnt5a/JNK pathway. J. Orthop. Surg. Res. 2021, 16, 129. [Google Scholar] [CrossRef] [PubMed]
  167. Christodoulides, C.; Lagathu, C.; Sethi, J.K.; Vidal-Puig, A. Adipogenesis and WNT signalling. Trends Endocrinol. Metab. 2009, 20, 16–24. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  168. Mori, H.; Prestwich, T.C.; Reid, M.; Longo, K.A.; Gerin, I.; Cawthorn, W.; Susulic, V.S.; Krishnan, V.; Greenfield, A.; MacDougald, O. Secreted frizzled-related protein 5 suppresses adipocyte mitochondrial metabolism through WNT inhibition. J. Clin. Investig. 2012, 122, 2405–2416. [Google Scholar] [CrossRef] [Green Version]
  169. Carstensen-Kirberg, M.; Röhrig, K.; Niersmann, C.; Ouwens, D.M.; Belgardt, B.F.; Roden, M.; Herder, C. Sfrp5 increases glucose-stimulated insulin secretion in the rat pancreatic beta cell line INS-1E. PLoS ONE 2019, 14, e0213650. [Google Scholar] [CrossRef]
  170. Li, Y.; Tian, M.; Yang, M.; Yang, G.; Chen, J.; Wang, H.; Liu, D.; Wang, H.; Deng, W.; Zhu, Z.; et al. Central Sfrp5 regulates hepatic glucose flux and VLDL-triglyceride secretion. Metabolism 2020, 103, 154029. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  171. Wang, X.; Peng, Q.; Jiang, F.; Xue, L.; Li, J.; Fan, Z.; Chen, P.; Chen, G.; Cai, Y. Secreted frizzled-related protein 5 protects against oxidative stress-induced apoptosis in human aortic endothelial cells via downregulation of Bax. J. Biochem. Mol. Toxicol. 2017, 31, e21978. [Google Scholar] [CrossRef] [PubMed]
  172. Liu, W.; Ji, Y.; Chu, H.; Wang, M.; Yang, B.; Yin, C. SFRP5 mediates downregulation of the wnt5a/caveolin-1/JNK signaling pathway. J. Endocrinol. 2020, 247, 263–272. [Google Scholar] [CrossRef] [PubMed]
  173. Cho, Y.K.; Kang, Y.M.; Lee, S.E.; La Lee, Y.; Seol, S.M.; Lee, W.J.; Park, J.-Y.; Jung, C.H. Effect of SFRP5 (Secreted Frizzled–Related Protein 5) on the WNT5A (Wingless-Type Family Member 5A)-Induced Endothelial Dysfunction and Its Relevance with Arterial Stiffness in Human Subjects. Arterioscler. Thromb. Vasc. Biol. 2018, 38, 1358–1367. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  174. Teliewubai, J.; Ji, H.; Lu, Y.; Bai, B.; Yu, S.; Chi, C.; Xu, Y.; Zhang, Y. SFRP5 serves a beneficial role in arterial aging by inhibiting the proliferation, migration and inflammation of smooth muscle cells. Mol. Med. Rep. 2018, 18, 4682–4690. [Google Scholar] [CrossRef] [Green Version]
  175. Nakamura, K.; Sano, S.; Fuster, J.J.; Kikuchi, R.; Shimizu, I.; Ohshima, K.; Katanasaka, Y.; Ouchi, N.; Walsh, K. Secreted Frizzled-related Protein 5 Diminishes Cardiac Inflammation and Protects the Heart from Ischemia/Reperfusion Injury. J. Biol. Chem. 2016, 291, 2566–2575. [Google Scholar] [CrossRef] [Green Version]
  176. Bie, Z.-D.; Sun, L.-Y.; Geng, C.-L.; Meng, Q.-G.; Lin, X.-J.; Wang, Y.-F.; Wang, X.-B.; Yang, J. MiR-125b regulates SFRP5 expression to promote growth and activation of cardiac fibroblasts. Cell Biol. Int. 2016, 40, 1224–1234. [Google Scholar] [CrossRef]
  177. Akoumianakis, I.; Sanna, F.; Margaritis, M.; Badi, I.; Akawi, N.; Herdman, L.; Coutinho, P.; Fagan, H.; Antonopoulos, A.S.; Oikonomou, E.K.; et al. Adipose tissue-derived WNT5A regulates vascular redox signaling in obesity via USP17/RAC1-mediated activation of NADPH oxidases. Sci. Transl. Med. 2019, 11, eaav5055. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  178. Carstensen-Kirberg, M.; Kannenberg, J.M.; Huth, C.; Meisinger, C.; Koenig, W.; Heier, M.; Peters, A.; Rathmann, W.; Roden, M.; Herder, C.; et al. Inverse associations between serum levels of secreted frizzled-related protein-5 (SFRP5) and multiple cardiometabolic risk factors: KORA F4 study. Cardiovasc. Diabetol. 2017, 16, 109. [Google Scholar] [CrossRef]
  179. Bai, Y.; Du, Q.; Jiang, R.; Zhang, L.; Du, R.; Wu, N.; Li, P.; Li, L. Secreted Frizzled-Related Protein 5 is Associated with Glucose and Lipid Metabolism Related Metabolic Syndrome Components Among Adolescents in Northeastern China. Diabetes Metab. Syndr. Obes. 2021, 14, 2735–2742. [Google Scholar] [CrossRef]
  180. Tan, X.; Wang, X.; Chu, H.; Liu, H.; Yi, X.; Xiao, Y. SFRP5 correlates with obesity and metabolic syndrome and increases after weight loss in children. Clin. Endocrinol. 2014, 81, 363–369. [Google Scholar] [CrossRef] [PubMed]
  181. He, X.; Ma, H. Correlation Between Circulating Levels of Secreted Frizzled-Related Protein 5 and Type 2 Diabetic Patients and Subjects with Impaired-Glucose Regulation. Diabetes Metab. Syndr. Obes. 2020, 13, 1243–1250. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  182. Hu, W.; Li, L.; Yang, M.; Luo, X.; Ran, W.; Liu, D.; Xiong, Z.; Liu, H.; Yang, G. Circulating Sfrp5 Is a Signature of Obesity-Related Metabolic Disorders and Is Regulated by Glucose and Liraglutide in Humans. J. Clin. Endocrinol. Metab. 2013, 98, 290–298. [Google Scholar] [CrossRef]
  183. Almario, R.U.; Karakas, S.E. Roles of Circulating WNT-Signaling Proteins and WNT-Inhibitors in Human Adiposity, Insulin Resistance, Insulin Secretion, and Inflammation. Horm. Metab. Res. 2015, 47, 152–157. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  184. Xu, Q.; Wang, H.; Li, Y.; Wang, J.; Lai, Y.; Gao, L.; Lei, L.; Yang, G.; Liao, X.; Fang, X.; et al. Plasma Sfrp5 levels correlate with determinants of the metabolic syndrome in Chinese adults. Diabetes Metab. Res. Rev. 2017, 33, e2896. [Google Scholar] [CrossRef] [PubMed]
  185. Lu, Y.C.; Wang, C.P.; Hsu, C.C.; Chiu, C.A.; Yu, T.H.; Hung, W.C.; Lu, L.F.; Chung, F.M.; Tsai, I.T.; Lin, H.C.; et al. Circulating secreted frizzled-related protein 5 (Sfrp5) and wingless-type MMTV integration site family member 5a (Wnt5a) levels in patients with type 2 diabetes mellitus. Diabetes Metab. Res. Rev. 2013, 29, 551–556. [Google Scholar]
  186. Wang, B.; Pan, Y.; Yang, G.; Yu, W.; Liu, H.; Bai, B. Sfrp5/Wnt5a and leptin/adiponectin levels in the serum and the periarterial adipose tissue of patients with peripheral arterial occlusive disease. Clin. Biochem. 2021, 87, 46–51. [Google Scholar] [CrossRef]
  187. Oh, Y.J.; Kim, H.; Kim, A.J.; Ro, H.; Chang, J.H.; Lee, H.H.; Chung, W.; Jun, H.-S.; Jung, J.Y. Reduction of Secreted Frizzled-Related Protein 5 Drives Vascular Calcification through Wnt3a-Mediated Rho/ROCK/JNK Signaling in Chronic Kidney Disease. Int. J. Mol. Sci. 2020, 21, 3539. [Google Scholar] [CrossRef]
  188. Tong, S.; Du, Y.; Ji, Q.; Dong, R.; Cao, J.; Wang, Z.; Li, W.; Zeng, M.; Chen, H.; Zhu, X.; et al. Expression of Sfrp5/Wnt5a in human epicardial adipose tissue and their relationship with coronary artery disease. Life Sci. 2020, 245, 117338. [Google Scholar] [CrossRef]
  189. Du, Y.; Zhao, Y.; Zhu, Y.; Hu, C.; Zhang, J.; Ji, Q.; Liu, W.; Han, H.; Yang, L.; Zhang, D.; et al. High Serum Secreted Frizzled-Related Protein 5 Levels Associates with Early Improvement of Cardiac Function Following ST-Segment Elevation Myocardial Infarction Treated by Primary Percutaneous Coronary Intervention. J. Atheroscler. Thromb. 2019, 26, 868–878. [Google Scholar] [CrossRef] [Green Version]
  190. Wu, J.; Zheng, H.; Liu, X.; Chen, P.; Zhang, Y.; Luo, J.; Kuang, J.; Li, J.; Yang, Y.; Ma, T.; et al. Prognostic Value of Secreted Frizzled-Related Protein 5 in Heart Failure Patients with and Without Type 2 Diabetes Mellitus. Circ. Heart Fail. 2020, 13, e007054. [Google Scholar] [CrossRef] [PubMed]
  191. Ji, H.; Li, H.; Zhuang, J.; Su, Y.; Wen, J.; Zhang, J.; Zhao, D.; Zhang, Y.; Xu, Y. High serum level of secreted frizzled-related protein 5 (sfrp5) is associated with future cardiovascular events. Cardiovasc. Ther. 2017, 2, e115. [Google Scholar]
  192. Yang, R.-Z.; Lee, M.-J.; Hu, H.; Pray, J.; Wu, H.-B.; Hansen, B.C.; Shuldiner, A.R.; Fried, S.K.; McLenithan, J.C.; Gong, D.-W. Identification of omentin as a novel depot-specific adipokine in human adipose tissue: Possible role in modulating insulin action. Am. J. Physiol. Endocrinol. Metab. 2006, 290, E1253–E1261. [Google Scholar] [CrossRef] [PubMed]
  193. De Souza Batista, C.M.; Yang, R.-Z.; Lee, M.-J.; Glynn, N.M.; Yu, D.-Z.; Pray, J.; Ndubuizu, K.; Patil, S.; Schwartz, A.; Kligman, M.; et al. Omentin Plasma Levels and Gene Expression Are Decreased in Obesity. Diabetes 2007, 56, 1655–1661. [Google Scholar] [CrossRef] [Green Version]
  194. Watanabe, T.; Watanabe-Kominato, K.; Takahashi, Y.; Kojima, M.; Watanabe, R. Adipose Tissue-Derived Omentin-1 Function and Regulation. Compr. Physiol. 2017, 7, 765–781. [Google Scholar] [PubMed]
  195. Hiramatsu-Ito, M.; Shibata, R.; Ohashi, K.; Uemura, Y.; Kanemura, N.; Kambara, T.; Enomoto, T.; Yuasa, D.; Matsuo, K.; Ito, M.; et al. Omentin attenuates atherosclerotic lesion formation in apolipoprotein E-deficient mice. Cardiovasc. Res. 2016, 110, 107–117. [Google Scholar] [CrossRef] [Green Version]
  196. Lin, X.; Sun, Y.; Yang, S.; Yu, M.; Pan, L.; Yang, J.; Yang, J.; Shao, Q.; Liu, J.; Liu, Y.; et al. Omentin-1 Modulates Macrophage Function via Integrin Receptors αvβ3 and αvβ5 and Reverses Plaque Vulnerability in Animal Models of Atherosclerosis. Front. Cardiovasc. Med. 2021, 8, 757926. [Google Scholar] [CrossRef] [PubMed]
  197. Watanabe, K.; Watanabe, R.; Konii, H.; Shirai, R.; Sato, K.; Matsuyama, T.-A.; Ishibashi-Ueda, H.; Koba, S.; Kobayashi, Y.; Hirano, T.; et al. Counteractive effects of omentin-1 against atherogenesis. Cardiovasc. Res. 2016, 110, 118–128. [Google Scholar] [CrossRef]
  198. Uemura, Y.; Shibata, R.; Kanemura, N.; Ohashi, K.; Kambara, T.; Hiramatsu-Ito, M.; Enomoto, T.; Yuasa, D.; Joki, Y.; Matsuo, K.; et al. Adipose-derived protein omentin prevents neointimal formation after arterial injury. FASEB J. 2015, 29, 141–151. [Google Scholar] [CrossRef]
  199. Xie, H.; Xie, P.-L.; Wu, X.-P.; Chen, S.-M.; Zhou, H.-D.; Yuan, L.-Q.; Sheng, Z.-F.; Tang, S.-Y.; Luo, X.-H.; Liao, E.-Y. Omentin-1 attenuates arterial calcification and bone loss in osteoprotegerin-deficient mice by inhibition of RANKL expression. Cardiovasc. Res. 2011, 92, 296–306. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  200. Leandro, A.; Queiroz, M.; Azul, L.; Seiça, R.; Sena, C.M. Omentin: A novel therapeutic approach for the treatment of endothelial dysfunction in type 2 diabetes. Free. Radic. Biol. Med. 2021, 162, 233–242. [Google Scholar] [CrossRef]
  201. Yamawaki, H.; Tsubaki, N.; Mukohda, M.; Okada, M.; Hara, Y. Omentin, a novel adipokine, induces vasodilation in rat isolated blood vessels. Biochem. Biophys. Res. Commun. 2010, 393, 668–672. [Google Scholar] [CrossRef]
  202. Maruyama, S.; Shibata, R.; Kikuchi, R.; Izumiya, Y.; Rokutanda, T.; Araki, S.; Kataoka, Y.; Ohashi, K.; Daida, H.; Kihara, S.; et al. Fat-derived Factor Omentin Stimulates Endothelial Cell Function and Ischemia-induced Revascularization via Endothelial Nitric Oxide Synthase-dependent Mechanism. J. Biol. Chem. 2012, 287, 408–417. [Google Scholar] [CrossRef] [Green Version]
  203. Kataoka, Y.; Shibata, R.; Ohashi, K.; Kambara, T.; Enomoto, T.; Uemura, Y.; Ogura, Y.; Yuasa, D.; Matsuo, K.; Nagata, T.; et al. Omentin Prevents Myocardial Ischemic Injury Through AMP-Activated Protein Kinase- and Akt-Dependent Mechanisms. J. Am. Coll. Cardiol. 2014, 63, 2722–2733. [Google Scholar] [CrossRef] [Green Version]
  204. Matsuo, K.; Shibata, R.; Ohashi, K.; Kambara, T.; Uemura, Y.; Hiramatsu-Ito, M.; Enomoto, T.; Yuasa, D.; Joki, Y.; Ito, M.; et al. Omentin functions to attenuate cardiac hypertrophic response. J. Mol. Cell. Cardiol. 2015, 79, 195–202. [Google Scholar] [CrossRef] [PubMed]
  205. Jin, Z.; Xia, F.; Dong, J.; Lin, T.; Cai, Y.; Chen, J.; Chen, X.; Huang, Z.; Wang, Q.; Chen, H.; et al. Omentin-1 attenuates glucocorticoid-induced cardiac injury by phosphorylating GSK3β. J. Mol. Endocrinol. 2021, 66, 273–283. [Google Scholar] [CrossRef]
  206. Zhang, Q.; Zhu, L.; Zheng, M.; Fan, C.; Li, Y.; Zhang, D.; He, Y.; Yang, H. Changes of serum omentin-1 levels in normal subjects, type 2 diabetes and type 2 diabetes with overweight and obesity in Chinese adults. Ann. Endocrionol 2014, 75, 171–175. [Google Scholar] [CrossRef]
  207. Jialal, I.; Devaraj, S.; Kaur, H.; Adams-Huet, B.; Bremer, A.A. Increased Chemerin and Decreased Omentin-1 in Both Adipose Tissue and Plasma in Nascent Metabolic Syndrome. J. Clin. Endocrinol. Metab. 2013, 98, E514–E517. [Google Scholar] [CrossRef] [PubMed]
  208. Peña-Cano, M.I.; Valencia-Ortega, J.; Morales-Ávila, E.; Díaz-Velázquez, M.F.; Gómez-Díaz, R.; Saucedo, R. Omentin-1 and its relationship with inflammatory factors in maternal plasma and visceral adipose tissue of women with gestational diabetes mellitus. J. Endocrinol. Investig. 2021, 1–10. [Google Scholar] [CrossRef] [PubMed]
  209. Latif, A.H.E.; Anwar, S.; Gautham, K.S.; Kadurei, F.; Ojo, R.O.; Hafizyar, F.; Haroon, D.M.; Rakesh, F.; Talpur, A.S. Association of Plasma Omentin-1 Levels with Diabetes and Its Complications. Cureus 2021, 13, e18203. [Google Scholar] [CrossRef]
  210. Biscetti, F.; Nardella, E.; Bonadia, N.; Angelini, F.; Pitocco, D.; Santoliquido, A.; Filipponi, M.; Landolfi, R.; Flex, A. Association between plasma omentin-1 levels in type 2 diabetic patients and peripheral artery disease. Cardiovasc. Diabetol. 2019, 18, 74–77. [Google Scholar] [CrossRef]
  211. Senthilkumar, G.P.; Anithalekshmi, M.S.; Yasir, M.; Parameswaran, S.; Packirisamy, R.M.; Bobby, Z. Role of omentin 1 and IL-6 in type 2 diabetes mellitus patients with diabetic nephropathy. Diabetes Metab. Syndr. Clin. Res. Rev. 2018, 12, 23–26. [Google Scholar] [CrossRef] [PubMed]
  212. El-Mesallamy, H.O.; El-Derany, M.O.; Hamdy, N.M. Serum omentin-1 and chemerin levels are interrelated in patients with Type 2 diabetes mellitus with or without ischaemic heart disease. Diabet. Med. 2011, 28, 1194–1200. [Google Scholar] [CrossRef]
  213. Bai, P.; Abdullah, F.; Lodi, M.; Sarhadi, M.; Dilip, A.; Shahab, S.; Yasir, F.; Jahangir, M. Association between Coronary Artery Disease and Plasma Omentin-1 Levels. Cureus 2021, 13, e17347. [Google Scholar] [CrossRef]
  214. Zhong, X.; Zhang, H.-Y.; Tan, H.; Zhou, Y.; Liu, F.-L.; Chen, F.-Q.; Shang, D.-Y. Association of serum omentin-1 levels with coronary artery disease. Acta Pharmacol. Sin. 2011, 32, 873–878. [Google Scholar] [CrossRef] [PubMed]
  215. Biscetti, F.; Nardella, E.; Rando, M.M.; Cecchini, A.L.; Angelini, F.; Cina, A.; Iezzi, R.; Filipponi, M.; Santoliquido, A.; Pitocco, D.; et al. Association between omentin-1 and major cardiovascular events after lower extremity endovascular revascu-larization in diabetic patients: A prospective cohort study. Cardiovasc. Diabetol. 2020, 19, 170. [Google Scholar] [CrossRef]
  216. Narumi, T.; Watanabe, T.; Kadowaki, S.; Kinoshita, D.; Yokoyama, M.; Honda, Y.; Otaki, Y.; Nishiyama, S.; Takahashi, H.; Arimoto, T.; et al. Impact of serum omentin-1 levels on cardiac prognosis in patients with heart failure. Cardiovasc. Diabetol. 2014, 13, 84. [Google Scholar] [CrossRef] [Green Version]
  217. Çelik, M.; Nar, R.; Nar, G.; Sökmen, E.; Günver, G. Serum omentin-1 levels in hypertensive patients. J. Hum. Hypertens 2021, 35, 290–295. [Google Scholar] [CrossRef]
  218. Xu, T.; Zuo, P.; Wang, Y.; Gao, Z.; Ke, K. Serum omentin-1 is a novel biomarker for predicting the functional outcome of acute ischemic stroke patients. Clin. Chem. Lab. Med. 2018, 56, 350–355. [Google Scholar] [CrossRef] [PubMed]
  219. Yang, J.; Gao, Y. Clinical relevance of serum omentin-1 levels as a biomarker of prognosis in patients with acute cerebral infarction. Brain Behav. 2020, 10, e01678. [Google Scholar] [CrossRef] [PubMed]
  220. Xu, T.; Zuo, P.; Cao, L.; Gao, Z.; Ke, K. Omentin-1 is Associated with Carotid Plaque Instability among Ischemic Stroke Patients. J. Atheroscler. Thromb. 2018, 25, 505–511. [Google Scholar] [CrossRef] [Green Version]
  221. Xu, T.; Li, Y.; Su, Y.; Zuo, P.; Gao, Z.; Ke, K. Serum omentin-1 and risk of one-year mortality in patients with ischemic stroke. Clin. Chim. Acta 2020, 505, 167–171. [Google Scholar] [CrossRef] [PubMed]
  222. Zhang, G.-H.; Ye, Z.-H.; Guan, H.-J.; Guo, M.; Zhou, X.-X.; Xu, Y.-Y. Impact of serum omentin-1 concentrations on functional outcome among acute intracerebral hemorrhage patients. Clin. Chim. Acta 2020, 503, 169–174. [Google Scholar] [CrossRef]
  223. Onur, I.; Oz, F.; Yildiz, S.; Oflaz, H.; Sigirci, S.; Elitok, A.; Pilten, S.; Karaayvaz, E.B.; Cizgici, A.Y.; Kaya, M.G.; et al. Serum Omentin 1 Level Is Associated with Coronary Artery Disease and Its Severity in Postmenopausal Women. Angiology 2014, 65, 896–900. [Google Scholar] [CrossRef] [PubMed]
  224. Zhou, J.-P.; Tong, X.-Y.; Zhu, L.-P.; Luo, J.-M.; Luo, Y.; Bai, Y.-P.; Li, C.-C.; Zhang, G.-G. Plasma Omentin-1 Level as a Predictor of Good Coronary Collateral Circulation. J. Atheroscler. Thromb. 2017, 24, 940–948. [Google Scholar] [CrossRef] [Green Version]
  225. Yoo, H.J.; Hwang, S.Y.; Hong, H.C.; Choi, H.Y.; Yang, S.J.; Seo, J.A.; Kim, S.G.; Kim, N.H.; Choi, K.M.; Choi, D.S.; et al. Association of circulating omentin-1 level with arterial stiffness and carotid plaque in type 2 diabetes. Cardiovasc. Diabetol. 2011, 10, 103. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  226. Onur, I.; Oz, F.; Yildiz, S.; Kuplay, H.; Yucel, C.; Sigirci, S.; Elitok, A.; Pilten, S.; Kasali, K.; Cizgici, A.Y.; et al. A decreased serum omentin-1 level may be an independent risk factor for peripheral arterial disease. Int Angiol. 2014, 33, 455–460. [Google Scholar]
  227. Yasir, M.; Senthilkumar, G.P.; Jayashree, K.; Babu, K.R.; Vadivelan, M.; Palanivel, C. Association of serum omentin-1, apelin and chemerin concentrations with the presence and severity of diabetic retinopathy in type 2 diabetes mellitus patients. Arch. Physiol. Biochem. 2019, 1–8. [Google Scholar] [CrossRef]
  228. Siegrist, M.; Heitkamp, M.; Braun, I.; Vogg, N.; Haller, B.; Langhof, H.; Koenig, W.; Halle, M. Changes of omentin-1 and chemerin during 4 weeks of lifestyle intervention and 1 year follow-up in children with obesity. Clin. Nutr. 2021, 40, 5648–5654. [Google Scholar] [CrossRef]
  229. Atashak, S.; Stannard, S.R.; Daraei, A.; Soltani, M.; Saeidi, A.; Moradi, F.; Laher, I.; Hackney, A.C.; Zouhal, H. High-intensity Interval Training Improves Lipocalin-2 and Omentin-1 Levels in Men with Obesity. Int. J. Sports Med. 2021, 1–16. [Google Scholar] [CrossRef]
  230. De Luis, D.A.; Calvo, S.G.; Gomez, J.J.L.; Izaola, O.; Primo, D.; Pacheco, D.; Aller, R. Omentin-1 Changes following Biliopancreatic Diversion and Relationship with Cardiovascular Risk Factors. Ann. Nutr. Metab. 2018, 73, 106–112. [Google Scholar] [CrossRef]
  231. Alkuraishy, H.M.; Al-Gareeb, A.I. New Insights into the Role of Metformin Effects on Serum Omentin-1 Levels in Acute My-ocardial Infarction: Cross-Sectional Study. Emerg. Med. Int. 2015, 2015, 283021. [Google Scholar] [CrossRef] [PubMed] [Green Version]
Figure 1. Anti-diabetic potential of adiponectin. Adiponectin enhances insulin sensitivity through multiorgan involvement to alleviate the progression of diabetes mellitus. Cumulative evidence has supported the favorable actions of anti-diabetic medications on adiponectin. The up arrows denote the increase, while the down arrows denote the opposite.
Figure 1. Anti-diabetic potential of adiponectin. Adiponectin enhances insulin sensitivity through multiorgan involvement to alleviate the progression of diabetes mellitus. Cumulative evidence has supported the favorable actions of anti-diabetic medications on adiponectin. The up arrows denote the increase, while the down arrows denote the opposite.
Ijms 22 13529 g001
Figure 2. Beneficial effects of vaspin on metabolic health. Vaspin affects a broad range of organs simultaneously to enhance metabolic health. Aside from its anti-inflammatory function, vaspin intervenes in energy regulation, glycemic control, lipid profile, and vascular function. The up arrows denote the increase, while the down arrows denote the opposite.
Figure 2. Beneficial effects of vaspin on metabolic health. Vaspin affects a broad range of organs simultaneously to enhance metabolic health. Aside from its anti-inflammatory function, vaspin intervenes in energy regulation, glycemic control, lipid profile, and vascular function. The up arrows denote the increase, while the down arrows denote the opposite.
Ijms 22 13529 g002
Figure 3. Major C1q/TNF-related protein (CTRP) isoforms participating in cardiometabolic homeostasis. The respective members of the CTRP family exert different actions on each organ, among which CTRP3, CTRP6, CTRP9, CTRP12, and CTRP13 promote an anti-inflammatory response. CTRP1 and CTRP5 are recognized to trigger inflammation and atherogenesis, whereas CTRP1 plays a protective function in insulin sensitivity and ischemic heart disease. However, the role of CTRP5 in diabetes and cardiac disease is still unclear.
Figure 3. Major C1q/TNF-related protein (CTRP) isoforms participating in cardiometabolic homeostasis. The respective members of the CTRP family exert different actions on each organ, among which CTRP3, CTRP6, CTRP9, CTRP12, and CTRP13 promote an anti-inflammatory response. CTRP1 and CTRP5 are recognized to trigger inflammation and atherogenesis, whereas CTRP1 plays a protective function in insulin sensitivity and ischemic heart disease. However, the role of CTRP5 in diabetes and cardiac disease is still unclear.
Ijms 22 13529 g003
Figure 4. Schematic view on the association of secreted frizzled-related protein 5 (SFRP5) and wingless-type family member 5a (WNT5A)/c-Jun N-terminal kinases (JNK). WNT5A and the downstream JNK signaling facilitate inflammation and cardiometabolic impairment, which is antagonized by SFRP5.
Figure 4. Schematic view on the association of secreted frizzled-related protein 5 (SFRP5) and wingless-type family member 5a (WNT5A)/c-Jun N-terminal kinases (JNK). WNT5A and the downstream JNK signaling facilitate inflammation and cardiometabolic impairment, which is antagonized by SFRP5.
Ijms 22 13529 g004
Table 1. Summary of the human studies for the association of adipokine levels with cardiometabolic parameters.
Table 1. Summary of the human studies for the association of adipokine levels with cardiometabolic parameters.
Author, Year [ref] *Study Design ParticipantsMean Age (years)Men (%)Ethnicity Outcomes or ParametersCardiometabolic Health Association §
AdiponectinGariballa et al., 2019 [35]PC193 overweight and obese subjects36.07.0AsianVisceral fatPositive
Lindberg et al., 2014 [37]PC666 patients with STEMI, without diabetes63.574.3WhiteIncident T2DMPositive
Kou et al., 2018 [38]CC309 subjectsN/RN/RAsianAtherogenic index of plasmaPositive
Yoshida et al., 2005 [40]CS56 patients with T2DMN/RN/RAsianVLDL levelsPositive
Tomono et al., 2018 [41]CS174 subjects without diabetes67.945.4AsianHDL levelsPositive
Doumatey et al., 2012 [44]CS822 subjects43.344.3BlackObesity and MetSPositive
Garvey et al., 2014 [48]RC475 subjects with prediabetes and/or MetS52.035.2MultiracialPhentermine/topiramatePositive
Wannamethee et al., 2007 [51]PC4046 men68.7100WhiteAll-cause and CVD mortalityNegative
McEntegart et al., 2007 [52]CS47 patients with CAD67.383.0WhiteHF and cachexiaNegative
VaspinTaheri et al., 2020 [75]CS70 women29.00AsianObesityNegative
Hao et al., 2016 [77]CS348 subjects52.858.0AsianT2DM and CADNegative
Yang et al., 2021 [78]CS372 patients with T2DM53.055.6AsianDiabetic retinopathyNegative
Jia et al., 2018 [79]RC474 patients with NAFLDN/RN/RAsianExerciseNegative
Tan et al., 2008 [80]NRI21 women with PCOS28.00WhiteMetforminNegative
Zhang et al., 2011 [81]NRI31 patients with T2DM55.335.5AsianRosiglitazoneNegative
Golpaie et al., 2011 [82]NRI30 obese subjects32.530.0AsianRestrictive bariatric surgeryNegative
Kadoglou et al., 2021 [84]NRI84 subjects with preclinical carotid atherosclerosis62.046.4WhiteAtorvastatinPositive
Al-Kuraishy et al., 2018 [85]RC110 patients with ACS48.665.5AsianRosuvastatinPositive
Kastl et al., 2020 [86]PC85 patients with CAD64.077.6WhiteIn-stent restenosisPositive
Zhang et al., 2016 [87]PC80 patients with MI68.081.2AsianMACEPositive
Ji et al., 2020 [88]RC197 subjects with chest pain65.056.9AsianMACEPositive
Rashad et al., 2020 [89]PCC90 patients with T2DM58.755.6AsianISNegative
Cakal et al., 2011 [90]CS71 womenN/R0AsianDiabetogenic riskNegative
Esteghamati et al., 2014 [91]CS145 subjects49.442.8AsianMetSNegative
Buyukinan et al., 2018 [92]CS121 obese childrenN/R34.7AsianMetSNegative
CTRP9Wang et al., 2015 [119]CS362 subjects62.172.1AsianCADPositive
Moradi et al., 2018 [120]CS337 subjects58.070.0AsianCAD and T2DMNegative
Gao et al., 2019 [121]PC344 subjects56.269.2AsianHFrEFPositive
Miyatake et al., 2020 [122]RC50 recipients of kidney transplantation 31.566.0AsianAortic calcificationPositive
Pan et al., 2020 [123]CS128 CI patients with cerebrovascular stent54.058.6AsianRestenosis after cerebrovascular stentPositive
Jia et al., 2017 [124]CS306 subjects52.048.0AsianIncident T2DM and obesityNegative
Hwang et al., 2014 [125]CS221 subjects46.063.3AsianMetSPositive
Wolf et al., 2016 [126]NRI21 obese subjectsN/R14.0MultiracialBariatric surgeryNegative
Xia et al., 2020 [127]CS259 pregnant womenN/R0AsianGDMPositive
Yang et al., 2021 [128]CS262 patients with T2DM55.068.3AsianCardiac autonomic neuropathyPositive
Asada et al., 2016 [129]CS258 patients with T2DM without CKD62.054.3AsianCarotid IMTNegative
Jung et al., 2014 [130]CS278 patients with T2DM58.360.8AsianbaPWVNegative
Na et al., 2020 [131]CS133 pregnant womenN/R0AsianGDMPositive
CTRP3Zhou et al., 2018 [145]PC313 subjectsN/RN/RAsianIncident NAFLDPositive
Choi et al., 2012 [146]CS345 subjects51.838.2AsianPrediabetes, T2DM, and MetSNegative
Moradi et al., 2019 [147]CS164 subjects58.062.8AsianT2DM and diabetic nephropathyPositive
Flehmig et al., 2014 [148]CS141 obese subjects48.047.5WhiteMetforminPositive
Choi et al., 2014 [151]CS362 subjects60.467.4AsianACS and SAPPositive
Jiang et al., 2018 [152]CS108 subjects56.371.3AsianAcute aortic dissectionPositive
Yildirim et al., 2021 [153]CS118 subjects64.466.1AsianHFrEF and VTPositive
CTRP12Tan et al., 2014 [149]NRI21 women with PCOS28.00WhiteMetforminPositive
Nadimi et al., 2021 [154]CS250 subjects58.554.8AsianCAD severityPositive
CTRP13Shanaki et al., 2016 [150]CS86 men54.0100AsianBMI, visceral fat, and IMTPositive
SFRP5Akoumianakis et al., 2019 [177]CC140 subjects64.045.0WhiteCADPositive
Carstensen-Kirberg et al., 2017 [178]CS1096 subjects70.251.5WhitePrediabetes, T2DM, BMI, and HDL-CPositive
Bai et al., 2021 [179]CS684 adolescents13.754.2AsianFPG and TCPositive
Tan et al., 2014 [180]NRI31 obese children11.071.0AsianLifestyle interventionPositive
He et al., 2020 [181]NRI111 patients with T2DM57.046.8AsianMetforminPositive
Hu et al., 2013 [182]NRI30 patients with T2DMN/RN/RAsianLiraglutidePositive
Almario et al., 2015 [183]CS84 women36.10WhiteWeight and cholesterolPositive
Xu et al., 2017 [184]CS284 subjects53.453.9AsianMetSPositive
Lu et al., 2013 [185]CS124 subjects59.556.5AsianT2DMNegative
Wang et al., 2021 [186]CS114 subjects67.351.8AsianPADPositive
Oh et al., 2020 [187]CS120 subjects51.719.3AsianVascular calcificationPositive
Cho et al., 2018 [173]CS282 patients with T2DM58.063.6AsianbaPWVNegative
Tong et al., 2020 [188]CS87 subjects61.556.3AsianCADPositive
Du et al., 2019 [189]PC85 patients with STEMI55.776.5AsianEarly improvement of LVEFPositive
Wu et al., 2020 [190]PC833 patients with HF65.957.4AsianComposite of all-cause mortality or HF rehospitalizationPositive
Ji et al., 2017 [191]PC168 subjects65.055.4AsianMACENegative
Omentin-1Batista et al., 2007 [193]CS91 subjects43.742.9WhiteObesityPositive
Zhang et al., 2014 [206]CS120 subjects66.351.7AsianT2DM and obesityPositive
Jialal et al., 2013 [207]CS75 subjects51.678.7WhiteMetSPositive
Peña-Cano et al., 2021 [208]CS231 pregnant women29.50HispanicGDMPositive
Latif et al., 2021 [209]CS500 patients with T2DM53.052.0AsianDiabetic complicationsPositive
Biscetti et al., 2019 [210]S600 patients with T2DM74.768.2WhitePADPositive
Senthilkumar et al., 2018 [211]CS82 patients with T2DM48.5N/RAsianDiabetic nephropathyPositive
El-Mesallamy et al., 2011 [212]CS90 subjects57.374.4AsianT2DMPositive
Bai et al., 2021 [213]CS600 subjects52.561.2AsianCADPositive
Zhong et al., 2011 [214]CS207 subjects61.269.1AsianACS and SAPPositive
Biscetti et al., 2020 [215]PC207 patients with T2DM and CTLI75.069.6WhiteMACE and MALEPositive
Kataoka et al., 2014 [203]CS20 patients with acute MI62.565.0AsianMyocardial salvage index and EFPositive
Narumi et al., 2014 [216]PC136 patients with HF72.055.9AsianCardiac death or HF rehospitalizationPositive
Çelik et al., 2021 [217]CS121 subjects49.648.8AsianHypertensionPositive
Xu et al., 2018 [218]PC266 patients with ISN/RN/RAsianFunctional outcomePositive
Yang et al., 2020 [219]PC109 patients with CI62.862.4AsianFunctional prognosisPositive
Xu et al., 2018 [220]CS173 patients with ISN/RN/RAsianUnstable carotid plaquePositive
Xu et al., 2020 [221]PC303 patients with IS66.864.7Asian1 year mortalityPositive
Zhang et al., 2020 [222]PC104 patients with hemorrhagic stroke68.054.8AsianFunctional outcomePositive
Onur et al., 2014 [223]CS193 women67.30AsianAngiographic CADPositive
Zhou et al., 2017 [224]CS142 patients with CADN/RN/RAsianCoronary collateral circulationPositive
Yoo et al., 2011 [225]CS90 subjects54.541.1AsianT2DM and carotid plaquePositive
Onur et al., 2014 [226]CS173 subjectsN/RN/RAsianPADPositive
Yasir et al., 2019 [227]CS167 patients with T2DMN/RN/RAsianDiabetic retinopathyPositive
Siegrist et al., 2021 [228]NRI156 overweight and obese children14.044.9WhiteLifestyle interventionPositive
Atashak et al., 2021 [229]RC30 obese menN/R100MultiracialExercisePositive
Luis et al., 2018 [230]NRI24 obese subjectsN/RN/RWhiteBiliopancreatic diversion surgeryPositive
Kadoglou et al., 2021 [84]NRI84 subjects with preclinical carotid atherosclerosis62.046.4WhiteAtorvastatinPositive
Alkuraishy et al., 2015 [231]CS85 patients with T2DM and acute MI57.560.0WhiteMetforminPositive
* Articles were listed in the order of citation in the text. Meta-analyses were excluded. RC, randomized controlled; NRI, nonrandomized interventional; PC, prospective cohort; RC, retrospective cohort; CC, case-control; PCC, prospective case-control; CS, cross-sectional. One of the following ethnic groups: white, Asian, Hispanic, black, and multiracial. § Labeled as positive if the increasing adipokine levels are associated with the positive direction of cardiometabolic health, and as negative in the case of the inverse association.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Jung, H.N.; Jung, C.H. The Role of Anti-Inflammatory Adipokines in Cardiometabolic Disorders: Moving beyond Adiponectin. Int. J. Mol. Sci. 2021, 22, 13529. https://doi.org/10.3390/ijms222413529

AMA Style

Jung HN, Jung CH. The Role of Anti-Inflammatory Adipokines in Cardiometabolic Disorders: Moving beyond Adiponectin. International Journal of Molecular Sciences. 2021; 22(24):13529. https://doi.org/10.3390/ijms222413529

Chicago/Turabian Style

Jung, Han Na, and Chang Hee Jung. 2021. "The Role of Anti-Inflammatory Adipokines in Cardiometabolic Disorders: Moving beyond Adiponectin" International Journal of Molecular Sciences 22, no. 24: 13529. https://doi.org/10.3390/ijms222413529

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