Skip to main content
Erschienen in: Journal of Translational Medicine 1/2022

Open Access 01.12.2022 | Review

Role of Chemerin/ChemR23 axis as an emerging therapeutic perspective on obesity-related vascular dysfunction

verfasst von: Yingying Xie, Ling Liu

Erschienen in: Journal of Translational Medicine | Ausgabe 1/2022

insite
INHALT
download
DOWNLOAD
print
DRUCKEN
insite
SUCHEN

Abstract

Sufficient epidemiological investigations demonstrate that there is a close correlation between obesity and vascular dysfunction. Nevertheless, specific mechanisms underlying this link remain currently unclear. Given the crucial and decisive role of vascular dysfunction in multitudinous diseases, various hypotheses had been proposed and numerous experiments were being carried out. One recognized view is that increased adipokine secretion following the expanded mass of white adipose tissue due to obesity contributes to the regulation of vascular function. Chemerin, as a neo-adipokine, whose systemic level is elevated in obesity, is believed as a regulator of adipogenesis, inflammation, and vascular dysfunction via binding its cell surface receptor, chemR23. Hence, this review aims to focus on the up-to-date proof on chemerin/chemR23 axis-relevant signaling pathways, emphasize the multifarious impacts of chemerin/chemR23 axis on vascular function regulation, raise certain unsettled questions to inspire further investigations, and explore the therapeutic possibilities targeting chemerin/chemR23.
Hinweise

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Abkürzungen
WAT
White adipose tissue
PVAT
Perivascular adipose tissue
GTP
Guanosine 5'-triphosphate
GPCR
GTP-binding protein-coupled receptors
CMKLR1
Chemokine like receptor 1
CPN
Carboxypeptidases N
CPB
Carboxypeptidases B
GPR1
G protein-coupled receptor 1
CCRL2
C–C motif receptor-like 2
RvE1
Resolvin E1
EPA
Eicosapentaenoic acid
ω-3 PUFAs
Omega-3 polyunsaturated fatty acids
BRIGHT
British Genetics of Hypertension
CCL
C–C motif chemokine ligand
BMI
Body mass index
SAT
Subcutaneous adipose tissue
VAT
Visceral adipose tissue
WHR
Waist-to-hip ratio
TNFα
Tumor necrosis factor α
CRP
C-reactive protein
HFD
High-fat diet
DIO
Diet-induced-obesity
pDCs
Plasmacytoid dendritic cells
ECs
Endothelial cells
VSMCs
Vascular smooth muscle cells
ASO
Antisense oligonucleotides
ERK
Extracellular regulated protein kinases
MAPK
Mitogen-activated protein kinase
shRNA
Short hairpin RNA
IL
Interleukin
ROS
Reactive oxygen species
NO
Nitric oxide
eNOS
Endothelial nitric oxide synthase
Apo
Apolipoprotein
MCP-1
Monocyte chemoattractant protein-1
UAP
Unstable angina pectoris
AMI
Acute myocardial
PAH
Pulmonary hypertension
AV
Aortic valve
AAA
Abdominal aotic aneurysm

Introduction

Over the years, obesity has become a worldwide public health incident [1, 2], which is mainly caused by the accumulation of white adipose tissue (WAT) [3]. Besides, obesity is also an acknowledged predisposing and risk factor for types of disorders, including diabetes, dyslipidemia, vascular dysfunction and so on [4]. Vascular dysfunction then leads to multi-system diseases, such as cardiovascular system diseases (atherosclerosis, hypertension) [5, 6], respiratory system diseases (pulmonary hypertension, adult respiratory distress syndrome) [7, 8], digestive system diseases (Budd-Chiari syndrome, severe acute pancreatitis) [9, 10] and so on.
WAT is a well-acknowledged human endocrine organ with the secretion of various adipokines (i.e. adipocytokines), in addition to serving as energy storage [11]. Adipokines are the conditioning agents of adipogenesis, vascular function, glucose and insulin metabolisms [1215], whose regulating ability becomes more pronounced with the expanded mass of WAT due to obesity. Chemerin is also a protein whose systemic level will increase in obesity and plays an extremely important role in regulating vascular function through binding its receptor, chemR23. Notably, chemerin can be activated in obesity, transforming from inert prochemerin to activated chemerin [16].
Interestingly, chemerin is more highly expressed in perivascular adipose tissue (PVAT) than in subcutaneous or visceral adipose tissue [17], which provides a mechanistic explanation for the regulation of vascular function by chemerin, partly due to the natural location advantage and the absence of mechanical barrier between PVAT and blood vessels [18]. Evidence had shown that chemerin/chemR23 axis exerted significant effects on the regulation of vascular function, but the detailed mechanisms and pathways mediated by this axis remained controversial.
In this review, we introduce the formation of chemerin via COOH-terminal processing and the alteration of expressions of both chemerin and chemR23 in obesity. Furthermore, the role of chemerin/chemR23 axis in vascular dysfunction, especially when the body is in the state of obesity is described in detail.

Chemerin feature and processing

Chemerin is a 16 KDa protein and encoded by tazarotene induced gene 2 (i.e. retinoic acid receptor responder 2). It was initially reported as a synthetic retinoid gene in psoriatic skin lesions in 1997 [19, 20]. In previous studies, chemerin was identified as a natural ligand of orphan guanosine 5'-triphosphate (GTP)-binding protein-coupled receptors (GPCR), also known as chemokine like receptor 1 (CMKLR1) or chemR23 [18]. There exist three phases before chemerin evolves into active modalities: pre-prochemerin, prochemerin, and chemerin stages [22]. So-called pre-prochemerin (1–163) is a 163-amino-acid long protein without biological activity and directly encoded by tazarotene induced gene 2. It transforms into prochemerin (21–163), a 143-amino-acid long protein with low biological activity, by getting rid of 20 NH2-terminal amino acids in a proteolytic cleavage way [20, 23]. And prochemerin undergoes extracellular COOH-terminal processing by certain proteases [24], ending up as chemerin with higher biological activity and chemR23 receptor binding ability [20].
It is worth mentioning that different types of proteases simultaneously cut prochemerin into diverse length products that differ in COOH-terminal amino acids and potency for chemR23 activation. For instance, the first enzymes shown to activate prochemerin are called elastase and cathepsin G [24], both belonging to neutrophil proteases species. The former removes the 6, 8, or 11 COOH-terminal amino acids to produce three forms, chemerin (21–157), (21–155), or (21–152), respectively [21]. Among them, chemerin (21–157) holds the highest activity. While the latter clears the 7 COOH-terminal amino acids to produce one fragment, chemerin (21–156) [25], which is the second most active form next to chemerin (21–157), and both are the two active forms in the human body.
Prochemerin is also cut into chemerin (21–158) with low activity mediated by tryptase and plasmin through sweeping the 5 COOH-terminal amino acids away. Chemerin (21–158) undergoes the second processing to complement this activity by carboxypeptidases N or B (CPN or CPB), ultimately forming chemerin (21–157) [26, 27]. This illustrates that the segments formed by the COOH-terminal processing might be also the substrates in the subsequent processing.
In addition to the above typical proteases leading to the formation of various fragments, there are some enzymes (e.g., mast cell chymase and angiotensin-converting enzyme) having been reported to cleave certain fragments to generate relatively inactive modalities, including chemerin (21–154) and chemerin (21–155) [28, 29].
Taken together, chemerin is conscripted to undergo proteolytic cleavages to remove amino acids located in NH2-terminal and COOH-terminal by multitudinous proteases before the formation of diverse isoforms (Table 1), which differ in their activities. Most of them hold a low activity or no activity, even antagonizing the active isoforms, generally referring to chemerin (21–157). This manifests that a complex regulatory network controls chemerin bioactivity.
Noticeably, excessive chemerin tends to be activated when the body is in the state of obesity with the accelerated COOH-terminal processing. To some extent, it is a hint that chemerin seems to have inextricable relation to obesity.

Chemerin receptor types and characteristics

The chemerin receptors that have been recognized and understood mainly include chemR23(i.e. GPCR or CMKLR1), G protein-coupled receptor (GPR)1 and chemokine (C–C motif) receptor-like (CCRL)2. They are all located at the surface of cells but distinct from each other in the affinity of binding to chemerin, signaling, and internalization of the chemerin-receptor complex (Fig. 1).

ChemR23

Chemerin is a natural ligand of chemR23. As the well-deserved nature receptor with the highest affinity binding, efficient signaling and internalization, chemR23 is presently the only one reflecting the veritable activity of chemerin and accurately representing chemerin targeting sites among the three receptors. Chemerin active forms were observed only through measuring the activity on chemR23-expressing cells [25]. In other words, where there is chemR23, there is chemerin combining target. Besides, chemR23 is structurally relevant to a suite of chemokines, such as complement fragments (e.g., C5a, C3a) and prostaglandin D2 [30] which also cover GPR1 and the orphan receptors GPR32 and GPR33 [25].
Furthermore, chemR23 has a second ligand named resolvin E1 (RvE1), a new bioactive oxygenated product of the essential fatty eicosapentaenoic acid (EPA), which is one of the main types of omega-3 polyunsaturated fatty acids (ω-3 PUFAs), existing in fish oils [31, 32]. Based on the powerfully anti-inflammatory role of ω-3 PUFAs in cardiovascular diseases, the combination of chemR23 and RvE1 is increasingly taken for a salutary one, differing from chemerin/chemR23 axis, even quite the opposite [33, 34].

GPR1 and CCRL2

In addition to chemR23, GPR1 and CCRL2 are universally acknowledged as two other receptor types of chemerin [35]. GPR1 structurally resembles ChemR23, which is concretely embodied in a 37% similar sequence identity between them [36]. That is why GPR1 is taken as a potential candidate in binding and activating chemerin in addition to chemR23. Meanwhile, GPR1 was mapped genome-wide of human loci for essential hypertension, involving the British Genetics of Hypertension (BRIGHT) study in 2003 [37]. Regrettably, despite high-affinity binding, GPR1 shows weak signaling. It has been found that chemerin elicits potent constrictor actions via chemR23, not GPR1 [38].
CCRL2 is also referred to as Eo1 in mice, and chemokine receptor (HCR) in humans. Though previous thought of as leukocyte chemoattractant receptor binding the chemokines C–C motif chemokine ligand (CCL)2, CCL5, CCL7, and CCL8 [39], CCRL2 is latterly depicted as a third receptor for chemerin with high-affinity binding [40]. Unlike the first two receptors, CCRL2 shows no signaling or internalization. Whereas, the conclusion cannot be drawn that CCRL2 is disqualified as the receptor to bind chemerin and activate chemerin/chemR23 axis. Instead, CCRL2 is proposed for elevating the local concentration of chemerin and presenting chemerin to GPR1 and chemR23 nearby, implying its role as a regulator of chemerin concentration and a mediator of chemerin transfer [40].
It appears that the other two receptors, GPR1 and CCRL2 are likely to participate in the peculiarity manifestations of chemerin, but both take effect with mechanisms relying on chemR23 more or less.

The changes of chemerin/chemR23 axis in obesity

Increased expression of chemerin in obesity

Evidence from clinical observations

Chemerin concentrations in obese patients
Chemerin is a secreted protein that is detected in the plasma or serum. Generally speaking, it is a trend that women and the elderly have higher circulating chemerin concentrations than men and the young [41, 42]. Though chemerin concentration partly varies with age and gender, it always fluctuates from 90 to 200 ng/mL in non-obese populations. Owing to the varying degree of obesity, the range of chemerin concentration fluctuation is too large to be represented simply by a numerical value in obese populations.
Circulating chemerin concentration is certainly higher in obese patients than lean ones, which has been confirmed by numerous studies [43, 44]. Obesity is often classified into different grades by body mass index (BMI), so, previous studies elucidated that plasma chemerin concentration positively correlates with BMI [43]. A rising view is that BMI puts more emphasis on the amount of subcutaneous adipose tissue (SAT), not visceral adipose tissue (VAT), which was confirmed by stepwise multiple regression analysis [45]. And VAT is better represented by waistline or waist-to-hip ratio (WHR). Given that chemerin was reported to be expressed more in VAT, particularly in PVAT, than in SAT [4648], relevant evidence supported serum chemerin concentration was also positively correlated with waistline or WHR, both of which were more representative of circulating chemerin than BMI [42, 49]. Besides, studies about aerobic exercise [50] and losing weight [51] as lateral proofs verified that chemerin can be reduced through the cut-down of WAT mass.
Chemerin mRNA and protein expressions in obese patients
Similar to the up-regulation of circulating chemerin concentration due to obesity, chemerin protein expression is increased in tissues from obese patients. A study described that compared with the WAT of non-obese individuals, the WAT from obese donors produced higher chemerin protein mass [52]. In addition to WAT, other tissues like skeletal muscle also show a similar trend [52].
The mRNA expression of chemerin is also elevated in obese individuals. It was observed that chemerin mRNA expression was dramatically higher expressed in omental adipose tissue and SAT of obese patients, and weight loss induced by bariatric surgery led to a lower mRNA expression of chemerin than before [43]. Additionally, chemerin mRNA expression was elevated significantly in obese patients with non-alcoholic fatty liver disease [53]. In 56 morbidly obese women (BMI > 40 kg/m2), hepatic chemerin mRNA expression was induced [53, 54]. Increased expressions of other inflammatory factors, such as tumor necrosis factor α (TNFα) and lipopolysaccharide, were also detected in WAT of obese patients when chemerin expression was elevated. It indicates that inflammation could enhance chemerin production in WAT and other tissues [49, 55].
The conjecture that circulating chemerin protein depends on chemerin mRNA expression in WAT is reasonable in many cases. However, chemerin mRNA and protein expressions are not always synchronously modulated. For instance, chemerin mRNA expressions in SAT and VAT seemed not to be associated with serum chemerin concentration [56]. A study even revealed the negative correlation of chemerin mRNA expression in SAT with the systemic level of chemerin [57]. These seemingly contradictory experimental results manifest, to some extent, that chemerin is partly regulated by unknown post-transcriptional mechanisms.

Evidence from animal experiments

Chemerin concentration in obese animals
There are usually two types of experimental models associated with obesity. The first is mice with genetic deficiency, such as db/db mice (leptin receptor-deficient) and ob/ob mice (leptin-deficient). The second is obese mice induced by a high-fat diet (HFD), called diet-induced-obesity (DIO) mice. Circulating chemerin concentration in db/db mice was approximately two-fold higher than that in wild-type (WT) mice [58]. Female DIO mice had higher systemic chemerin concentrations compared to mice fed the control chow diet [59]. Moreover, compared with db/db mice treated with vehicle, CCX832 (chemR23 antagonist)treatment decreased body weight of db/db mice, accompanied by reduction of chemR23 protein expression. It is suggested that chemerin/chemR23 axis could be involved in the development of obesity.
Chemerin protein and mRNA expressions in obese animals
In terms of tissues, protein expression of chemerin varies with different parts, the highest in WAT and liver [61], followed by brown adipose tissue, lung, heart, ovary, and kidney [61]. Female DIO mice had markedly elevated chemerin protein expression in SAT and VAT [62]. Chemerin protein expression was up-regulated prominently in the gonadal adipose tissue of ob/ob mice [63]. Of interest, the level of chemerin protein expression in WAT of obese animal models was observed to go down after injection of 0.5 µg/g leptin, demonstrating that leptin resistance may be one of the reasons for elevated chemerin protein expression in obesity.
Hepatic chemerin mRNA expression of db/db mice was twofold higher than that of WT mice [58]. Hepatic chemerin mRNA expression was also induced in DIO mice [64]. In ob/ob mice, chemerin mRNA was not significantly changed in the liver, but elevated in the skeletal muscle [58], indicating the participation of chemerin in skeletal muscle insulin resistance via underlying mechanisms.

Evidence from cell researches

In physiological conditions, besides mature adipocytes and preadipocytes, chemerin was expressed in types of cell populations, like epithelial cells [65], endothelial cells [66, 67], fibroblasts [68], chondrocytes, and platelets [27], because those cells contain prochemerin transcripts. Sherd of evidence manifested that platelets also stockpiled a small amount of chemerin and released it under the stimulation of platelet-activating factors as platelet activity fluctuated [27].
At the cellular level, obesity is characterized by an increase in adipocyte cell size (hypertrophy), adipocyte cell number (hyperplasia), or both [27]. Studies showed that chemerin was detected on the 6th day (the early stage of differentiation) and increased dramatically on the 9th day (the end of differentiation) in 3T3-L1 preadipocytes, revealing the up-regulated expression of chemerin during adipogenic differentiation.

Increased expression of chemR23 in obesity

Evidence from obese patients

Interestingly, both chemR23 protein and mRNA expression show similar trends to chemerin in obese patients. ChemR23 protein expression was increased prominently in adipocytes and skeletal muscle cells of obese subjects when compared to lean, healthy ones [52]. Hepatic chemR23 mRNA expression was likewise induced markedly in patients suffering from non-alcoholic fatty liver disease [53]. ChemR23 mRNA expression in VATvwas dramatically higher in obese patients than that in lean volunteers. What’s more, there was a significant correlation and synchronism between chemerin and chemR23 mRNA expressions [53]. Bariatric surgery-induced weight loss reduced chemR23 protein and mRNA expressions in obese patients [43]. These findings were obtained from tissue samples of obese individuals. In order to further explore its possible mechanism, related investigation needs to be carried out in obese animal models.

Evidence from animal experiments

Through experimental animals, scientists can know more about the tissue distribution of chemR23. Under physiological conditions, chemR23 is expressed in WAT and hematopoietic tissues such as the thymus, bone marrow, spleen, fetal liver, and lymphoid organs [4, 69, 70]. While in obese animals, chemR23 protein expression in WAT was affected a lot. For example, chemR23 protein expression was increased markedly in SAT and VAT of obese rodents, including female DIO mice [59], Psammomys obesus [59], db/db mice and ob/ob mice [63]. What’s more, chemR23 protein expression was even up to five times higher than that of WT mice in epididymal adipose tissue of ob/ob mice. Interestingly, the injection of 0.5 µg/g leptin reduced chemR23 protein expression in WAT of ob/ob and db/db mice [63], indicating that leptin deficiency or resistance partly contributes to the elevated chemR23 protein expression in WAT of obese animals.

Evidence from cell researches

ChemR23 is expressed in various types of cells, covering blood monocytes, monocyte-derived human macrophages [65], immature dendritic cells [23, 71], plasmacytoid dendritic cells (pDCs) [72], microglial cells, and natural killer cells [73], and low levels in non-irritant CD4+ T lymphocytes, polymorphonuclear cells [73], even leukocyte population, where chemerin is not expressed.
Although no increase in the protein and mRNA expressions of chemr23 was observed in 3T3-L1 preadipocytes [59]. The knock-down even loss of chemR23 in 3T3-L1 preadipocytes indeed obstructed adipogenesis through lowering gene expression of adipogenesis like peroxisome proliferator-activated receptor-γ and sterol regulatory element-binding protein 2 [50, 74, 75]. It demonstrates that chemR23 promoted adipogenesis and further led to fat accumulation through enhancing capacity rather than expressions.

Increased activity of chemerin in obesity

The biological activity of chemerin is influenced as well as its expression by obesity. As mentioned above, chemerin becomes the most active form, chemerin(21–157), through COOH-terminal processing. By using specific enzyme-linked immunosorbent assays for different chemerin forms, chemerin (21–157) concentration in obese subjects was 1000-fold higher than that in non-obese subjects, accompanied by the increase of plasma C-reactive protein (CRP) level. This indicates that chemerin activity increased greatly in obesity perhaps by inflammation-mediated COOH-terminal processing [16].

Vascular dysfunction mediated by chemerin/chemR23 axis in obesity

Blood vessels are the largest network structure of the human body, mainly composed of the intima, media, adventitia, and PVAT surrounding the adventitia [52]. Endothelial cells (ECs) from intima and vascular smooth muscle cells (VSMCs) from media are recognized as the most pivotal cell populations for normal vascular function. In addition, PVAT attracts much attention recently because diverse adipokines from PVAT regulate vascular function more quickly than those from adipose tissue in other areas [76]. Thus, ECs, VSMCs, and PVAT maintain vascular homeostasis together. In other words, alters of structures or features of any one of them could induce vascular dysfunction.

Dysfunction of PVAT

PVAT is a double-edged sword in regulating vascular function. Under physiological conditions, PVAT often serves as mechanical support for blood vessels, with stable adipokines secretions. Immunohistochemistry, quantitative real-time polymerase chain reaction and western blot had validated a robust expression of chemerin in PVAT. But in vascular pathologies caused by detrimental states like obesity, PVAT expands in volume, along with increased secretions of adipokines [77, 78]. For example, chemerin and chemR23 protein expressions were higher in PVAT of DIO rats after a four-week HFD [48]. Chemerin antisense oligonucleotides (ASO) with whole-body activity reduced chemerin in PVAT and partially reversed the chemerin/chemR23 axis-induced vascular dysfunction [79]. Broadly speaking, the currently ample evidence supports that chemerin is such a typical adipokine that elevates in obese PVAT and then contributes to vascular dysfunction by further acting on the constituent cells of blood vessels like ECs and VSMCs via various mechanisms and pathways.

Endothelial dysfunction

Enhanced ECs proliferation and migration capacity

Proliferation and migration of ECs driven by proangiogenic molecules lead to angiogenesis, which is the pathophysiological basis of some diseases, such as metastasis of the tumor, atherosclerosis [80], and so on. A mass of in-vitro experiments had manifested that chemerin/chemR23 axis stimulated capillary-like structure formation via promoting the proliferation of ECs, and chemerin functioned as a chemoattractant for ECs to hasten migration [80, 81]. Chemerin/chemR23 axis-induced angiogenesis depends on p38 mitogen-activated protein kinase (MAPK) and extracellular regulated protein kinases (ERK) 1/2 pathway in human umbilical vein endothelial cells [66, 82, 83]. Matrix metalloproteinases-2/9 were also found to get involved in the proliferation and migration of ECs through degrading extracellular matrix in a chemerin dose-dependent way [66]. Besides, new evidence suggested that chemerin/chemR23 axis promoted angiogenesis through enhanced autophagy. It illustrates that chemerin/chemR23 axis enhances ECs proliferation and migration capacity through diversified pathways and mechanisms.
Moreover, CCX832 and the knockdown of chemR23 largely reversed chemerin/chemR23 axis-induced angiogenesis. For example, CCX832 reversed angiogenesis through decreased expression of P38 MAPK, ERK1/2 and matrix metalloproteinases-2/9. Additionally, angiogenesis was also reversed through knockdown of chemR23 by short hairpin RNA (shRNA), along with the down-regulated expression of autophagy-related genes [8486]. It not only verifies the effect of chemerin on ECs capacity, but also indicates that ChemR23 can be taken as the target to block obesity-related angiogenesis.

Elevated expressions and levels of endothelial inflammatory factors

Activated ECs can release a series of inflammatory factors, such as interleukin (IL-6), TNFα and CRP, which leads to abnormal endothelial secretion and inflammation in the blood vessel wall [87]. Besides, elevated expressions of intercellular adhesion molecule 1 and E-selectin have been regarded as the symbols of vascular endothelial activation. It was shown that increased circulating chemerin concentration was accompanied by the elevated level of CRP in obese children and expressions of intercellular adhesion molecule 1 and E-selectin in human coronary artery endothelial cells [88]. Simultaneously, certain inflammatory cytokines such as TNF-α, IL-1β, and IL-6 augmented chemR23 expression in ECs in turn [66]. Moreover, the elevated levels of inflammatory cytokines induced by chemerin/chemR23 axis increased monocyte attachment to ECs [67, 89]. It suggests that the activation of chemerin/chemR23 axis promotes endothelial dysfunction partly through inflammatory mechanisms.

Excessive production of reactive oxygen species in ECs

Oxidative stress is associated with multiple pathological processes [90]. As a momentous product of oxidative stress, reactive oxygen species (ROS) is another key factor that contributes to endothelial dysfunction [87, 91]. On one hand, the production of ROS was increased in human aorta endothelial cells with chemerin stimulation, and the chemerin-induced ROS generation was inhibited by N-acetylcysteine, a ROS scavenger [85]. On the other hand, the knock-down of chemR23 decreased the ROS generation [85]. It supported that chemerin/chemR23 axis induced ROS generation. Moreover, as the main source of intracellular ROS production, mitochondria are closely related to chemerin/chemR23 axis [92]. It’s reported that chemerin-treated ECs showed enhanced mitochondrial ROS generation and the mitochondria-targeted antioxidant, Mito-TEMPO significantly suppressed the chemerin-mediated ROS production. Furthermore, oxidative stress induced by chemerin/chemR23 axis subsequently triggers autophagy and apoptosis of ECs, which further impairs the vascular integrity and function [85].

Reduced production of nitric oxide in ECs

Nitric oxide (NO) is recognized as an effective vasodilator released by ECs and maintains vascular tone and homeostasis [93]. NO within ECs is mainly produced via endothelial nitric oxide synthase (eNOS) [94]. Chemerin decreased eNOS generation and enhanced NO breakdown, which ultimately led to NO reduction in ECs. Additionally, other potential mechanisms, including eNOS uncoupling, increased O2-generation, and reduced NO-dependent cGMP signaling could participate in chemerin/chemR23 axis-related endothelial dysfunction [95].

Dysfunction of VSMCs

Enhanced proliferation and migration capacity of VSMCs

It has been approved that VSMCs proliferation and migration are involved in the pathophysiological process of vascular remodeling [96, 97]. The abnormal vascular structure is accompanied by vascular dysfunction to a great extent. After stimulating VSMCs by chemerin (100 ng/mL) for 20 min, enhanced proliferation and migration capacity of VSMCs could be found. And CCX832 ameliorated VSMCs proliferation and migration. Furthermore, new evidence demonstrated that chemerin/chemR23 axis promoted proliferation and migration of VSMCs through MAPK signaling [98], Akt/ERK signaling [99], endothelin-1 dependent pathway [100], and enhanced autophagy [101]. To some extent, chemerin/chemR23 axis could affect VSMCs capacity through multiple regulatory networks.

Excessive apoptosis of VSMCs

Excessive apoptosis is another adverse event for dysfunction of VSMCs in addition to increased proliferation and migration capacity [102]. After stimulating VSMCs by chemerin (100 ng/mL) for 6 h, obvious apoptosis of VSMCs was observed. Similarly, CCX832 improved such apoptosis-associated VSMCs dysfunction [89]. Interestingly, the intervention time of chemerin-induced proliferation and migration was obviously longer than that of chemerin-induced apoptosis. It may indicate that the early effect of chemerin is to promote proliferation and migration of VSMCs, resulting in thickening of the media while the late effect enhanced apoptosis ultimately leading to vascular weakness. This further suggests that chemerin could play different roles in different stages of vascular dysfunction.

Augmentation of oxidative stress of VSMCs

Oxidative stress has been identified to exert important effects on deteriorating vascular function through enhanced inflammation, proliferation and migration, apoptosis and so on [103106]. The generation of ROS was increased in chemerin-incubated VSMCs. However, CCX832 or knockdown of chemR23 by shRNA decreased the production of ROS. It indicates that chemerin/chemR23 axis could induce VSMCs dysfunction partly through an oxidative mechanism [103106].

Up-regulation of Inflammatory factors of VSMCs

There is no doubt that inflammation is involved in the dysfunction of VSMCs and inflammatory factors could impair VSMCs capacity through multiple pathways, such as promoted proliferation and migration, aging and apoptosis [97]. After stimulation with chemerin, the expressions of certain inflammatory cytokines such as IL-1β, IL-6 and monocyte chemoattractant protein-1 (MCP-1) were found to up-regulate in VSMCs. Conversely, CCX832 ameliorated the increase of these inflammatory cytokines [89].

Cardiovascular diseases mediated by chemerin/chemR23 axis

Atherosclerosis and acute coronary syndrome (ACS)

The scientists assessed the severity of aortic and coronary arteries by the AHA classification through the angiography completed before the patients died, and then detected chemerin and chemR23 proteins in those arterial specimens by immunohistochemistry. Strong chemerin immunopositivity was observed in PVAT, VSMCs, and foam cells in atherosclerotic lesions, and accompanied by a positive correlation with the severity of atherosclerosis [107]. Nevertheless, chemerin is not recommended as a predictor of human atherosclerosis [108, 109]. Or rather, the predictive value of chemerin immeasurably could hinge on the region of affected arteries and stage of disease [110].
The analogous conclusion in experimental animals was drawn that chemerin accelerated the progression of atherosclerosis in apolipoprotein (Apo) E−/− mice with HFD by adenovirus transfection for knockdown or overexpression of chemerin gene into the aorta or pDCs [111]. In addition, the knockout of chemR23 in pDCs of ApoE−/− mice restricted the formation and progression of atherosclerotic plaque [112]. This pro-atherosclerotic effect is induced by chemerin/chemR23 axis partly through promoting adhesion and migration of ECs [113], inflammation [114], and proliferation of VSMCs.
ACS, including unstable angina pectoris (UAP) and acute myocardial infarction (AMI), is the most feared consequence of partial or complete thrombotic vessel occlusion caused by disruption of a certain coronary atherosclerotic plaque. The plasma chemerin concentration was notably higher in patients with ACS than in those with stable angina pectoris and controls, and the increase of chemerin concentration was synchronized with the elevation of CRP concentration [115118]. Hence, chemerin could be considered as a novel predictor of ACS. What's more, the average chemerin concentration in AMI is higher than that in UAP [117].

Hypertension

Compared with healthy controls, adult patients with primary hypertension had significantly higher serum chemerin concentrations [117]. Circulating chemerin concentration also increased even in obese children with elevated systolic blood pressure and not diagnosed with hypertension [119, 120]. Although ample evidence appealed that chemerin concentration was highly positively correlated with blood pressure, more clinical trials are needed to support the view prop up the perspective that chemerin is a predictor of human obesity-induced hypertension.
Obese rats with elevated serum chemerin concentrations showed a tendency of susceptibility to hypertension [48]. There was a certain drop in mean and systolic blood pressures in the chemerin knockout rats [121]. Besides dysfunctions of ECs, VSMCs and PVAT, chemerin/chemR23 axis induced hypertension also through enhanced arterial contraction [26, 38, 47] and sensitivity of the sympathetic nervous system [122].

Pulmonary arterial hypertension (PAH)

The protein expressions of chemerin and chemR23 were elevated in PAH model rats. And chemerin-9 (chemR23 agonist) induced contraction of the isolated intrapulmonary artery through increasing chemR23 protein expression in VSMCs [123]. Although animal experiments indicated that chemerin/chemR23 axis was involved in the development of PAH, there was still a lack of abundant clinical observations to prove this correlation in patients with PAH.
Aortic valve (AV) stenosis
Degenerative AV stenosis is one of the most common cardiovascular diseases currently in the elderly, which is classified into mild, moderate, severe according to the stricture degree of AV. In comparison with the controls, serum chemerin concentration was observably higher in patients with mild and moderate AV stenosis. Interestingly enough, the patients with the severe stenosis had lower circulating chemerin concentration than those in the mild and moderate stenosis, for which inactivation of inflammation might account [123, 124]. So, chemerin was proposed as a good predictor for mild AV stenosis and chemerin > 38.60 ng/mL was selected as the cut-off point for the diagnosis of mild AV stenosis. [125].

Abdominal aortic aneurysm (AAA)

AAA is a progressive segmental abdominal aortic dilation. The circulating chemerin concentration was higher in patients suffering from AAA than healthy controls. Besides, compared with the normal abdominal aortic samples, higher mRNA expressions of chemerin and chemR23 were detected in the focus tissues from patients with AAA [123]. This demonstrated that chemerin/chemR23 axis was involved in AAA progression.
In previous animal experiments, scientists found that DIO mice and ob/ob mice were more prone to AAA than WT mice [126, 127], but the potential mechanisms have not been explored yet. Therefore, we reasonably speculate that chemerin/chemR23 axis could partly promote the formation of AAA in obese mice. (Fig. 2).

Arterial calcification and arterial stiffness

Arterial calcification and arterial stiffness are independent predictors of cardiovascular risk and mortality. Both processes reinforce one another, creating a vicious cycle. However, chemerin/chemR23 axis seemingly induced contradictory results in the occurrence and progression of arterial calcification and arterial stiffness. On one hand, there was a significantly positive correlation between the circulating chemerin concentration and arterial stiffness in obese patients [120, 128, 129]. On the other hand, chemerin-9 (chemR23 agonist) increased the expression of a calcification inhibitor, matrix gla protein, and reduced phosphate-induced calcification in VSMCs. And the aforementioned effect on arterial calcification mediated by chemerin-9 was not observed VSMCs lacking chemR23. It indicates that chemerin/chemR23 axis may restrain the occurrence and development of arterial calcification [130].

Future therapeutics for chemrin/chemR23 axis

Although we have a general idea of the significant role of chemerin/chemR23 axis in obesity-related vascular dysfunction, it remains a pity that there is almost no drug circulating in the market targeting chemerin/chemR23 axis. Through a variety of related studies, several drugs targeting chemerin/chemR23 axis are still considered to have potential and feasibility for use in humans. They include CCX832, RvE1, chemerin-9, chemerin ASO, and nano-antioxidant.

CCX832

Numerous in vivo and in vitro studies have revealed that chemR23 inhibitor, CCX832, greatly reversed chemerin/chemR23 axis-induced vascular dysfunction.
The effect of CCX832 on improving chemerin/chemR23 axis-induced vascular dysfunction has been verified in various cells and in vitro vessels from multiple sources. For example, CCX832 improved chemerin-induced vascular inflammation in human microvascular ECs and human aorta ECs [60, 66]. CCX832 reversed oxidative stress in human aortic smooth muscle cells and VSMCs from mesenteric arteries of C57BL/6 J mice [38, 47]. Besides, CCX832 inhibited abnormal contraction in the isolated rat mesenteric artery, rat thoracic aorta, human pulmonary artery and human coronary artery [38, 131, 132].
Moreover, oral and intravenous administration of CCX832 ameliorated vascular insulin sensitivity in diabetic obese db/db mice and DIO mice [60]. Although CCX achieved phased success in phase 1 clinical trial of patients with psoriasis in 2012, its development was discontinued in 2013 for unknown reasons. Even so, CCX832 was still used in animal and cell experiments and obtained good experimental results. This manifests that CCX832 does play an essential role in blocking chemerin/chemR23 axis signaling and improving the chemerin/chemR23 axis-induced vascular dysfunction. Hence, we should strengthen the development of chemR23 antagonists to find other chemR23 antagonists that can not only replace CCX832 in experiments but also have safety and effectiveness in humans.

RvE1

RvE1, a new bioactive oxygenated product of EPA, is proved to be another ligand of chemR23. Meanwhile, RvE1 is a specialized pro-resolving mediator (SPMs), which are involved in promoting the resolution of inflammation [133, 134]. RvE1 plays a crucial role in improving vascular function and reducing cardiovascular risk through its downstream specific receptor axis, namely ERV1/chemR23 [135, 136].
A mass of in vivo and in vitro studies have demonstrated that RVE1 exerts powerful cardiovascular benefits, including inhibiting the progression of atherosclerotic plaques [137, 138], reducing the formation of new intima [139], preventing vascular endothelial injury, regulating vasoconstriction [132], restraining vascular calcification [140, 141] and so on. What’s more, multiple clinical trials have also supplemented the cardiovascular benefits of EPA [142145]. Based on this, RvE1 is speculated rationally to redress vascular dysfunction partly through competing with chemerin for the binding site of chemR23.

Chemerin-9 and other chemerin isoforms

Chemerin-9, also known as C9, is a synthetic small molecule fragment of chemerin. Although it is biologically active as an analog of chemerin and an agonist of chemR23. Chemerin-9 seemingly plays an anti-inflammatory and positive role in regulating vascular function in most cases, which is the exact opposite of chemerin.
A study described that chemerin-9 attenuated the formation of AAA in obese mice [146]. And the infusion of chemerin-9 significantly decreased the areas of aortic atherosclerotic lesions, accompanied by the reduction of TNF-α [147]. It is well known that chemerin-9 improved vascular function. But interestingly, some studies revealed that chemerin-9 might be involved in the evolvement of hypertension and PAH by inducing arterial contraction [147]. The detailed mechanism is worth exploring.
In addition to chemerin-9, there are other active fragments, such as chemerin-13 (C13). Chemerin-13, similar to chemerin-9, is also an agonist of chemR23, but few studies have been conducted on the treatment of chemerin/chemR23 axis-induced vascular dysfunction with it. More studies are needed to explore the difference between chemerin-13 and chemerin-9 in regulating vascular function.
It will necessitate investigations of the differences between chemerin-13 and chemerin-9 in regulating vascular function and further explorations of the therapeutic possibilities of more fragments or isoforms of chemerin.

Chemerin antisense oligonucleotide

ASO is a class of molecular drugs that inhibit the expression of target gene DNA or mRNA by sequence-specific binding and regulation at the gene level. It has been found that chemerin ASO with whole-body activity dramatically decreases blood pressure in rats [57, 148]. This opens up the possibility of chemerin ASO in the treatment of chemerin/chemR23 axis-induced vascular dysfunction in humans. Hence, the experimental and clinical studies of chemerin ASO are strongly advocated to provide more diversified options for the treatment of chemerin/chemR23 axis-induced dysfunction.

Nano-antioxidant

Considering the critical role of oxidative stress on the vascular dysfunction induced by chemerin/chemR23 axis, inhibition of ROS production can reduce chemerin expression to a certain extent, further improving vascular function. Therefore, the application of antioxidants provides a novel direction for the treatment of chemerin/chemR23 axis-induced vascular dysfunction. To date, the effectiveness of traditional antioxidants has been limited for many reasons, such as gastrointestinal degradation, first-pass effect, and/or instability during storage [149]. The combination of nanotechnology and antioxidants can effectively avoid these problems. Nano-antioxidants not only have higher physicochemical stability as well as biological activity, but also have improved ROS scavenging ability [92]. Recently, several types of these nano-antioxidants have demonstrated potential utility in nanomedicine [150, 151]. Thus, The application of nano antioxidant technology is expected to ameliorate obesity-related vascular dysfunction.

Conclusion

The significance of chemerin has been illustrated in many researches. Circulating chemerin concentrations are related to the development and progression of multiple system diseases and even are used as a biomarker for the diagnosis of cardiovascular diseases like ACS, AV stenosis, AAA, and so on. The chemerin/chemR23 axis is a complex network strictly involved in the occurrence and development of obesity and regulation of vascular function, which is supported by abundant rationale from clinical and experimental observations. However, the concrete mechanisms and pathways of chemerin/chemR23 axis displaying in these disorders remain a mystery and worth exploring. Thus, uncovering the answer to key questions (Fig. 3) probably conduces to deeply understand and better define the relationship among this axis, obesity, and vascular dysfunction. Moreover, it is helpful to seek the possibility of other treatments in addition to CCX832, RvE1, chemerin ASO, chemerin-9, nano-antioxidants even other alternative and salutary isoforms of chemerin.
Table 1
Characteristics of different chemerin isoforms
https://static-content.springer.com/image/art%3A10.1186%2Fs12967-021-03220-7/MediaObjects/12967_2021_3220_Tab1_HTML.png

Acknowledgements

Not applicable.

Declarations

Not applicable.
Not applicable.

Competing interests

There are no conflicts of interest.
Open AccessThis article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://​creativecommons.​org/​licenses/​by/​4.​0/​. The Creative Commons Public Domain Dedication waiver (http://​creativecommons.​org/​publicdomain/​zero/​1.​0/​) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
insite
INHALT
download
DOWNLOAD
print
DRUCKEN
Literatur
1.
Zurück zum Zitat Stefan N, et al. Metabolically healthy obesity: epidemiology, mechanisms, and clinical implications. Lancet Diabetes Endocrinol. 2013;1(2):152–62. PubMed Stefan N, et al. Metabolically healthy obesity: epidemiology, mechanisms, and clinical implications. Lancet Diabetes Endocrinol. 2013;1(2):152–62. PubMed
2.
Zurück zum Zitat Saxton SN, et al. Mechanistic links between obesity, diabetes, and blood pressure: role of perivascular adipose tissue. Physiol Rev. 2019;99(4):1701–63.PubMed Saxton SN, et al. Mechanistic links between obesity, diabetes, and blood pressure: role of perivascular adipose tissue. Physiol Rev. 2019;99(4):1701–63.PubMed
3.
Zurück zum Zitat Reilly SM, Saltiel AR. Adapting to obesity with adipose tissue inflammation. Nat Rev Endocrinol. 2017;13(11):633–43. Reilly SM, Saltiel AR. Adapting to obesity with adipose tissue inflammation. Nat Rev Endocrinol. 2017;13(11):633–43.
4.
Zurück zum Zitat Meakin PJ, et al. Elevated circulating amyloid concentrations in obesity and diabetes promote vascular dysfunction. J Clin Invest. 2020;130(8):4104–17.PubMedPubMedCentral Meakin PJ, et al. Elevated circulating amyloid concentrations in obesity and diabetes promote vascular dysfunction. J Clin Invest. 2020;130(8):4104–17.PubMedPubMedCentral
5.
Zurück zum Zitat Kim HW, et al. Perivascular adipose tissue and vascular perturbation/atherosclerosis. Arterioscler Thromb Vasc Biol. 2020;40(11):2569–76.PubMedPubMedCentral Kim HW, et al. Perivascular adipose tissue and vascular perturbation/atherosclerosis. Arterioscler Thromb Vasc Biol. 2020;40(11):2569–76.PubMedPubMedCentral
6.
Zurück zum Zitat Rubio-Ruiz ME, et al. Aging in blood vessels. Medicinal agents FOR systemic arterial hypertension in the elderly. Ageing Res Rev. 2014;18:132–47.PubMed Rubio-Ruiz ME, et al. Aging in blood vessels. Medicinal agents FOR systemic arterial hypertension in the elderly. Ageing Res Rev. 2014;18:132–47.PubMed
7.
Zurück zum Zitat Barratt S, Millar A. Vascular remodelling in the pathogenesis of idiopathic pulmonary fibrosis. QJM. 2014;107(7):515–9.PubMedPubMedCentral Barratt S, Millar A. Vascular remodelling in the pathogenesis of idiopathic pulmonary fibrosis. QJM. 2014;107(7):515–9.PubMedPubMedCentral
9.
Zurück zum Zitat DeLeve LD. Vascular liver diseases. Curr Gastroenterol Rep. 2003;5(1):63–70.PubMed DeLeve LD. Vascular liver diseases. Curr Gastroenterol Rep. 2003;5(1):63–70.PubMed
11.
Zurück zum Zitat Heinonen S, et al. White adipose tissue mitochondrial metabolism in health and in obesity. Obes Rev. 2020;21(2):e12958.PubMed Heinonen S, et al. White adipose tissue mitochondrial metabolism in health and in obesity. Obes Rev. 2020;21(2):e12958.PubMed
12.
Zurück zum Zitat Catalina MO, et al. New Insights into Adipokines as Potential Biomarkers for Type-2 Diabetes Mellitus. Curr Med Chem. 2019;26(22):4119–44.PubMed Catalina MO, et al. New Insights into Adipokines as Potential Biomarkers for Type-2 Diabetes Mellitus. Curr Med Chem. 2019;26(22):4119–44.PubMed
13.
Zurück zum Zitat Dyck DJ. Adipokines as regulators of muscle metabolism and insulin sensitivity. Appl Physiol Nutr Metab. 2009;34(3):396–402.PubMed Dyck DJ. Adipokines as regulators of muscle metabolism and insulin sensitivity. Appl Physiol Nutr Metab. 2009;34(3):396–402.PubMed
14.
Zurück zum Zitat Gu P, Xu A. Interplay between adipose tissue and blood vessels in obesity and vascular dysfunction. Rev Endocr Metab Disord. 2013;14(1):49–58.PubMed Gu P, Xu A. Interplay between adipose tissue and blood vessels in obesity and vascular dysfunction. Rev Endocr Metab Disord. 2013;14(1):49–58.PubMed
15.
Zurück zum Zitat Maresca F, et al. Adipokines, vascular wall, and cardiovascular disease: a focused overview of the role of adipokines in the pathophysiology of cardiovascular disease. Angiology. 2015;66(1):8–24.PubMed Maresca F, et al. Adipokines, vascular wall, and cardiovascular disease: a focused overview of the role of adipokines in the pathophysiology of cardiovascular disease. Angiology. 2015;66(1):8–24.PubMed
16.
Zurück zum Zitat Chang SS, et al. Chemerin activation in human obesity. Obesity (Silver Spring). 2016;24(7):1522–9. Chang SS, et al. Chemerin activation in human obesity. Obesity (Silver Spring). 2016;24(7):1522–9.
17.
Zurück zum Zitat Han J, et al. Serum chemerin levels are associated with abdominal visceral fat in type 2 diabetes. J Korean Med Sci. 2016;31(6):924–31.PubMedPubMedCentral Han J, et al. Serum chemerin levels are associated with abdominal visceral fat in type 2 diabetes. J Korean Med Sci. 2016;31(6):924–31.PubMedPubMedCentral
18.
Zurück zum Zitat Boa B, et al. Exercise effects on perivascular adipose tissue: endocrine and paracrine determinants of vascular function. Br J Pharmacol. 2017;174(20):3466–81.PubMedPubMedCentral Boa B, et al. Exercise effects on perivascular adipose tissue: endocrine and paracrine determinants of vascular function. Br J Pharmacol. 2017;174(20):3466–81.PubMedPubMedCentral
19.
Zurück zum Zitat Roh SG, et al. Chemerin–a new adipokine that modulates adipogenesis via its own receptor. Biochem Biophys Res Commun. 2007;362(4):1013–8.PubMed Roh SG, et al. Chemerin–a new adipokine that modulates adipogenesis via its own receptor. Biochem Biophys Res Commun. 2007;362(4):1013–8.PubMed
20.
Zurück zum Zitat Meder W, et al. Characterization of human circulating TIG2 as a ligand for the orphan receptor ChemR23. FEBS Lett. 2003;555(3):495–9. PubMed Meder W, et al. Characterization of human circulating TIG2 as a ligand for the orphan receptor ChemR23. FEBS Lett. 2003;555(3):495–9. PubMed
21.
Zurück zum Zitat Kaur J, et al. Chemerin in human cardiovascular disease. Vascul Pharmacol. 2018;110:1–6.PubMed Kaur J, et al. Chemerin in human cardiovascular disease. Vascul Pharmacol. 2018;110:1–6.PubMed
22.
Zurück zum Zitat Kennedy AJ, Davenport AP. International union of basic and clinical pharmacology ciii: chemerin receptors CMKLR1 (Chemerin(1)) and GPR1 (Chemerin(2)) nomenclature, pharmacology, and function. Pharmacol Rev. 2018;70(1):174–96.PubMedPubMedCentral Kennedy AJ, Davenport AP. International union of basic and clinical pharmacology ciii: chemerin receptors CMKLR1 (Chemerin(1)) and GPR1 (Chemerin(2)) nomenclature, pharmacology, and function. Pharmacol Rev. 2018;70(1):174–96.PubMedPubMedCentral
23.
Zurück zum Zitat Wittamer V, et al. Specific recruitment of antigen-presenting cells by chemerin, a novel processed ligand from human inflammatory fluids. J Exp Med. 2003;198(7):977–85.PubMedPubMedCentral Wittamer V, et al. Specific recruitment of antigen-presenting cells by chemerin, a novel processed ligand from human inflammatory fluids. J Exp Med. 2003;198(7):977–85.PubMedPubMedCentral
24.
Zurück zum Zitat Wittamer V, et al. Neutrophil-mediated maturation of chemerin: a link between innate and adaptive immunity. J Immunol. 2005;175(1):487–93.PubMed Wittamer V, et al. Neutrophil-mediated maturation of chemerin: a link between innate and adaptive immunity. J Immunol. 2005;175(1):487–93.PubMed
25.
Zurück zum Zitat Bondue B, Wittamer V, Parmentier M. Chemerin and its receptors in leukocyte trafficking, inflammation and metabolism. Cytokine Growth Factor Rev. 2011;22(5–6):331–8.PubMed Bondue B, Wittamer V, Parmentier M. Chemerin and its receptors in leukocyte trafficking, inflammation and metabolism. Cytokine Growth Factor Rev. 2011;22(5–6):331–8.PubMed
26.
Zurück zum Zitat Zabel BA, et al. Chemoattractants, extracellular proteases, and the integrated host defense response. Exp Hematol. 2006;34(8):1021–32.PubMed Zabel BA, et al. Chemoattractants, extracellular proteases, and the integrated host defense response. Exp Hematol. 2006;34(8):1021–32.PubMed
27.
Zurück zum Zitat Du XY, et al. Regulation of chemerin bioactivity by plasma carboxypeptidase N, carboxypeptidase B (activated thrombin-activable fibrinolysis inhibitor), and platelets. J Biol Chem. 2009;284(2):751–8.PubMedPubMedCentral Du XY, et al. Regulation of chemerin bioactivity by plasma carboxypeptidase N, carboxypeptidase B (activated thrombin-activable fibrinolysis inhibitor), and platelets. J Biol Chem. 2009;284(2):751–8.PubMedPubMedCentral
28.
Zurück zum Zitat John H, et al. Quantification of angiotensin-converting-enzyme-mediated degradation of human chemerin 145–154 in plasma by matrix-assisted laser desorption/ionization-time-of-flight mass spectrometry. Anal Biochem. 2007;362(1):117–25.PubMed John H, et al. Quantification of angiotensin-converting-enzyme-mediated degradation of human chemerin 145–154 in plasma by matrix-assisted laser desorption/ionization-time-of-flight mass spectrometry. Anal Biochem. 2007;362(1):117–25.PubMed
29.
Zurück zum Zitat Guillabert A, et al. Role of neutrophil proteinase 3 and mast cell chymase in chemerin proteolytic regulation. J Leukoc Biol. 2008;84(6):1530–8.PubMed Guillabert A, et al. Role of neutrophil proteinase 3 and mast cell chymase in chemerin proteolytic regulation. J Leukoc Biol. 2008;84(6):1530–8.PubMed
30.
Zurück zum Zitat Gantz I, et al. Molecular cloning of a novel receptor (CMKLR1) with homology to the chemotactic factor receptors. Cytogenet Cell Genet. 1996;74(4):286–90.PubMed Gantz I, et al. Molecular cloning of a novel receptor (CMKLR1) with homology to the chemotactic factor receptors. Cytogenet Cell Genet. 1996;74(4):286–90.PubMed
31.
Zurück zum Zitat Zhang TT, et al. Health benefits of dietary marine DHA/EPA-enriched glycerophospholipids. Prog Lipid Res. 2019;75:100997.PubMed Zhang TT, et al. Health benefits of dietary marine DHA/EPA-enriched glycerophospholipids. Prog Lipid Res. 2019;75:100997.PubMed
32.
Zurück zum Zitat Arita M, et al. Stereochemical assignment, antiinflammatory properties, and receptor for the omega-3 lipid mediator resolvin E1. J Exp Med. 2005;201(5):713–22.PubMedPubMedCentral Arita M, et al. Stereochemical assignment, antiinflammatory properties, and receptor for the omega-3 lipid mediator resolvin E1. J Exp Med. 2005;201(5):713–22.PubMedPubMedCentral
33.
Zurück zum Zitat Weinberg RL, et al. Cardiovascular Impact of Nutritional Supplementation With Omega-3 Fatty Acids: JACC Focus Seminar. J Am Coll Cardiol. 2021;77(5):593–608.PubMed Weinberg RL, et al. Cardiovascular Impact of Nutritional Supplementation With Omega-3 Fatty Acids: JACC Focus Seminar. J Am Coll Cardiol. 2021;77(5):593–608.PubMed
34.
Zurück zum Zitat Schulze MB, et al. Intake and metabolism of omega-3 and omega-6 polyunsaturated fatty acids: nutritional implications for cardiometabolic diseases. Lancet Diabetes Endocrinol. 2020;8(11):915–30.PubMed Schulze MB, et al. Intake and metabolism of omega-3 and omega-6 polyunsaturated fatty acids: nutritional implications for cardiometabolic diseases. Lancet Diabetes Endocrinol. 2020;8(11):915–30.PubMed
35.
Zurück zum Zitat Ernst MC, Sinal CJ. Chemerin: at the crossroads of inflammation and obesity. Trends Endocrinol Metab. 2010;21(11):660–7.PubMed Ernst MC, Sinal CJ. Chemerin: at the crossroads of inflammation and obesity. Trends Endocrinol Metab. 2010;21(11):660–7.PubMed
36.
Zurück zum Zitat Barnea G, et al. The genetic design of signaling cascades to record receptor activation. Proc Natl Acad Sci U S A. 2008;105(1):64–9.PubMed Barnea G, et al. The genetic design of signaling cascades to record receptor activation. Proc Natl Acad Sci U S A. 2008;105(1):64–9.PubMed
37.
Zurück zum Zitat Caulfield M, et al. Genome-wide mapping of human loci for essential hypertension. Lancet. 2003;361(9375):2118–23.PubMed Caulfield M, et al. Genome-wide mapping of human loci for essential hypertension. Lancet. 2003;361(9375):2118–23.PubMed
39.
Zurück zum Zitat Biber K, et al. Expression of L-CCR in HEK 293 cells reveals functional responses to CCL2, CCL5, CCL7, and CCL8. J Leukoc Biol. 2003;74(2):243–51.PubMed Biber K, et al. Expression of L-CCR in HEK 293 cells reveals functional responses to CCL2, CCL5, CCL7, and CCL8. J Leukoc Biol. 2003;74(2):243–51.PubMed
40.
Zurück zum Zitat Zabel BA, et al. Mast cell-expressed orphan receptor CCRL2 binds chemerin and is required for optimal induction of IgE-mediated passive cutaneous anaphylaxis. J Exp Med. 2008;205(10):2207–20.PubMedPubMedCentral Zabel BA, et al. Mast cell-expressed orphan receptor CCRL2 binds chemerin and is required for optimal induction of IgE-mediated passive cutaneous anaphylaxis. J Exp Med. 2008;205(10):2207–20.PubMedPubMedCentral
41.
Zurück zum Zitat Bozaoglu K, et al. Chemerin is a novel adipokine associated with obesity and metabolic syndrome. Endocrinology. 2007;148(10):4687–94.PubMed Bozaoglu K, et al. Chemerin is a novel adipokine associated with obesity and metabolic syndrome. Endocrinology. 2007;148(10):4687–94.PubMed
42.
Zurück zum Zitat Bozaoglu K, et al. Chemerin is associated with metabolic syndrome phenotypes in a Mexican-American population. J Clin Endocrinol Metab. 2009;94(8):3085–8.PubMedPubMedCentral Bozaoglu K, et al. Chemerin is associated with metabolic syndrome phenotypes in a Mexican-American population. J Clin Endocrinol Metab. 2009;94(8):3085–8.PubMedPubMedCentral
43.
Zurück zum Zitat Chakaroun R, et al. Effects of weight loss and exercise on chemerin serum concentrations and adipose tissue expression in human obesity. Metabolism. 2012;61(5):706–14.PubMed Chakaroun R, et al. Effects of weight loss and exercise on chemerin serum concentrations and adipose tissue expression in human obesity. Metabolism. 2012;61(5):706–14.PubMed
44.
Zurück zum Zitat Ba HJ, et al. Serum Chemerin Levels Correlate With Determinants of Metabolic Syndrome in Obese Children and Adolescents. Clin Med Insights Pediatr. 2019;13:1179556519853780.PubMedPubMedCentral Ba HJ, et al. Serum Chemerin Levels Correlate With Determinants of Metabolic Syndrome in Obese Children and Adolescents. Clin Med Insights Pediatr. 2019;13:1179556519853780.PubMedPubMedCentral
45.
Zurück zum Zitat Ferland DJ, et al. Chemerin-induced arterial contraction is G(i)- and calcium-dependent. Vascul Pharmacol. 2017;88:30–41.PubMed Ferland DJ, et al. Chemerin-induced arterial contraction is G(i)- and calcium-dependent. Vascul Pharmacol. 2017;88:30–41.PubMed
46.
Zurück zum Zitat Cheon DY, et al. Serum Chemerin Levels are Associated with Visceral Adiposity, Independent of Waist Circumference, in Newly Diagnosed Type 2 Diabetic Subjects. Yonsei Med J. 2017;58(2):319–25.PubMedPubMedCentral Cheon DY, et al. Serum Chemerin Levels are Associated with Visceral Adiposity, Independent of Waist Circumference, in Newly Diagnosed Type 2 Diabetic Subjects. Yonsei Med J. 2017;58(2):319–25.PubMedPubMedCentral
47.
Zurück zum Zitat Watts SW, et al. Chemerin connects fat to arterial contraction. Arterioscler Thromb Vasc Biol. 2013;33(6):1320–8.PubMedPubMedCentral Watts SW, et al. Chemerin connects fat to arterial contraction. Arterioscler Thromb Vasc Biol. 2013;33(6):1320–8.PubMedPubMedCentral
48.
Zurück zum Zitat Weng C, et al. Effects of chemerin/CMKLR1 in obesity-induced hypertension and potential mechanism. Am J Transl Res. 2017;9(6):3096–104.PubMedPubMedCentral Weng C, et al. Effects of chemerin/CMKLR1 in obesity-induced hypertension and potential mechanism. Am J Transl Res. 2017;9(6):3096–104.PubMedPubMedCentral
49.
Zurück zum Zitat Feng X, et al. Elevated levels of serum chemerin in patients with obstructive sleep apnea syndrome. Biomarkers. 2012;17(3):248–53.PubMed Feng X, et al. Elevated levels of serum chemerin in patients with obstructive sleep apnea syndrome. Biomarkers. 2012;17(3):248–53.PubMed
50.
Zurück zum Zitat Lloyd JW, et al. Effect of an acute bout of aerobic exercise on chemerin levels in obese adults. Diabetes Metab Syndr. 2016;10(1):37–42.PubMed Lloyd JW, et al. Effect of an acute bout of aerobic exercise on chemerin levels in obese adults. Diabetes Metab Syndr. 2016;10(1):37–42.PubMed
51.
Zurück zum Zitat Shin HY, Park S, Lee JW. Positive association between the changes in chemerin and adiponectin levels after weight reduction. Endocr Res. 2017;42(4):287–95.PubMed Shin HY, Park S, Lee JW. Positive association between the changes in chemerin and adiponectin levels after weight reduction. Endocr Res. 2017;42(4):287–95.PubMed
52.
Zurück zum Zitat Sell H, et al. Chemerin is a novel adipocyte-derived factor inducing insulin resistance in primary human skeletal muscle cells. Diabetes. 2009;58(12):2731–40.PubMedPubMedCentral Sell H, et al. Chemerin is a novel adipocyte-derived factor inducing insulin resistance in primary human skeletal muscle cells. Diabetes. 2009;58(12):2731–40.PubMedPubMedCentral
53.
Zurück zum Zitat Döcke S, et al. Elevated hepatic chemerin mRNA expression in human non-alcoholic fatty liver disease. Eur J Endocrinol. 2013;169(5):547–57.PubMed Döcke S, et al. Elevated hepatic chemerin mRNA expression in human non-alcoholic fatty liver disease. Eur J Endocrinol. 2013;169(5):547–57.PubMed
54.
Zurück zum Zitat Ernst MC, et al. Disruption of the chemokine-like receptor-1 (CMKLR1) gene is associated with reduced adiposity and glucose intolerance. Endocrinology. 2012;153(2):672–82.PubMed Ernst MC, et al. Disruption of the chemokine-like receptor-1 (CMKLR1) gene is associated with reduced adiposity and glucose intolerance. Endocrinology. 2012;153(2):672–82.PubMed
55.
Zurück zum Zitat Catalán V, et al. Increased levels of chemerin and its receptor, chemokine-like receptor-1, in obesity are related to inflammation: tumor necrosis factor-α stimulates mRNA levels of chemerin in visceral adipocytes from obese patients. Surg Obes Relat Dis. 2013;9(2):306–14.PubMed Catalán V, et al. Increased levels of chemerin and its receptor, chemokine-like receptor-1, in obesity are related to inflammation: tumor necrosis factor-α stimulates mRNA levels of chemerin in visceral adipocytes from obese patients. Surg Obes Relat Dis. 2013;9(2):306–14.PubMed
56.
Zurück zum Zitat Wolfs MG, et al. Determining the association between adipokine expression in multiple tissues and phenotypic features of non-alcoholic fatty liver disease in obesity. Nutr Diabetes. 2015;5(2):e146.PubMedPubMedCentral Wolfs MG, et al. Determining the association between adipokine expression in multiple tissues and phenotypic features of non-alcoholic fatty liver disease in obesity. Nutr Diabetes. 2015;5(2):e146.PubMedPubMedCentral
57.
Zurück zum Zitat Alfadda AA, et al. Differential patterns of serum concentration and adipose tissue expression of chemerin in obesity: adipose depot specificity and gender dimorphism. Mol Cells. 2012;33(6):591–6.PubMedPubMedCentral Alfadda AA, et al. Differential patterns of serum concentration and adipose tissue expression of chemerin in obesity: adipose depot specificity and gender dimorphism. Mol Cells. 2012;33(6):591–6.PubMedPubMedCentral
58.
Zurück zum Zitat Ernst MC, et al. Chemerin exacerbates glucose intolerance in mouse models of obesity and diabetes. Endocrinology. 2010;151(5):1998–2007.PubMed Ernst MC, et al. Chemerin exacerbates glucose intolerance in mouse models of obesity and diabetes. Endocrinology. 2010;151(5):1998–2007.PubMed
59.
Zurück zum Zitat Bauer S, et al. Sterol regulatory element-binding protein 2 (SREBP2) activation after excess triglyceride storage induces chemerin in hypertrophic adipocytes. Endocrinology. 2011;152(1):26–35.PubMed Bauer S, et al. Sterol regulatory element-binding protein 2 (SREBP2) activation after excess triglyceride storage induces chemerin in hypertrophic adipocytes. Endocrinology. 2011;152(1):26–35.PubMed
60.
Zurück zum Zitat Neves KB, et al. Chemerin receptor blockade improves vascular function in diabetic obese mice via redox-sensitive and Akt-dependent pathways. Am J Physiol Heart Circ Physiol. 2018;315(6):H1851–60.PubMedPubMedCentral Neves KB, et al. Chemerin receptor blockade improves vascular function in diabetic obese mice via redox-sensitive and Akt-dependent pathways. Am J Physiol Heart Circ Physiol. 2018;315(6):H1851–60.PubMedPubMedCentral
61.
Zurück zum Zitat Rourke JL, Dranse HJ, Sinal CJ. Towards an integrative approach to understanding the role of chemerin in human health and disease. Obes Rev. 2013;14(3):245–62.PubMed Rourke JL, Dranse HJ, Sinal CJ. Towards an integrative approach to understanding the role of chemerin in human health and disease. Obes Rev. 2013;14(3):245–62.PubMed
64.
Zurück zum Zitat Krautbauer S, et al. Chemerin is highly expressed in hepatocytes and is induced in non-alcoholic steatohepatitis liver. Exp Mol Pathol. 2013;95(2):199–205.PubMed Krautbauer S, et al. Chemerin is highly expressed in hepatocytes and is induced in non-alcoholic steatohepatitis liver. Exp Mol Pathol. 2013;95(2):199–205.PubMed
65.
Zurück zum Zitat Luangsay S, et al. Mouse ChemR23 is expressed in dendritic cell subsets and macrophages, and mediates an anti-inflammatory activity of chemerin in a lung disease model. J Immunol. 2009;183(10):6489–99.PubMed Luangsay S, et al. Mouse ChemR23 is expressed in dendritic cell subsets and macrophages, and mediates an anti-inflammatory activity of chemerin in a lung disease model. J Immunol. 2009;183(10):6489–99.PubMed
66.
Zurück zum Zitat Kaur J, et al. Identification of chemerin receptor (ChemR23) in human endothelial cells: chemerin-induced endothelial angiogenesis. Biochem Biophys Res Commun. 2010;391(4):1762–8.PubMed Kaur J, et al. Identification of chemerin receptor (ChemR23) in human endothelial cells: chemerin-induced endothelial angiogenesis. Biochem Biophys Res Commun. 2010;391(4):1762–8.PubMed
67.
Zurück zum Zitat Dimitriadis GK, et al. Chemerin induces endothelial cell inflammation: activation of nuclear factor-kappa beta and monocyte-endothelial adhesion. Oncotarget. 2018;9(24):16678–90.PubMedPubMedCentral Dimitriadis GK, et al. Chemerin induces endothelial cell inflammation: activation of nuclear factor-kappa beta and monocyte-endothelial adhesion. Oncotarget. 2018;9(24):16678–90.PubMedPubMedCentral
68.
Zurück zum Zitat Vermi W, et al. Role of ChemR23 in directing the migration of myeloid and plasmacytoid dendritic cells to lymphoid organs and inflamed skin. J Exp Med. 2005;201(4):509–15.PubMedPubMedCentral Vermi W, et al. Role of ChemR23 in directing the migration of myeloid and plasmacytoid dendritic cells to lymphoid organs and inflamed skin. J Exp Med. 2005;201(4):509–15.PubMedPubMedCentral
70.
Zurück zum Zitat Arita M, et al. Resolvin E1 selectively interacts with leukotriene B4 receptor BLT1 and ChemR23 to regulate inflammation. J Immunol. 2007;178(6):3912–7.PubMed Arita M, et al. Resolvin E1 selectively interacts with leukotriene B4 receptor BLT1 and ChemR23 to regulate inflammation. J Immunol. 2007;178(6):3912–7.PubMed
71.
Zurück zum Zitat Kulig P, et al. Regulation of chemerin chemoattractant and antibacterial activity by human cysteine cathepsins. J Immunol. 2011;187(3):1403–10.PubMed Kulig P, et al. Regulation of chemerin chemoattractant and antibacterial activity by human cysteine cathepsins. J Immunol. 2011;187(3):1403–10.PubMed
72.
Zurück zum Zitat Graham KL, et al. Chemokine-like receptor-1 expression by central nervous system-infiltrating leukocytes and involvement in a model of autoimmune demyelinating disease. J Immunol. 2009;183(10):6717–23.PubMed Graham KL, et al. Chemokine-like receptor-1 expression by central nervous system-infiltrating leukocytes and involvement in a model of autoimmune demyelinating disease. J Immunol. 2009;183(10):6717–23.PubMed
73.
Zurück zum Zitat Parolini S, et al. The role of chemerin in the colocalization of NK and dendritic cell subsets into inflamed tissues. Blood. 2007;109(9):3625–32.PubMed Parolini S, et al. The role of chemerin in the colocalization of NK and dendritic cell subsets into inflamed tissues. Blood. 2007;109(9):3625–32.PubMed
74.
Zurück zum Zitat Lin X, et al. Aerobic exercise decreases chemerin/CMKLR1 in the serum and peripheral metabolic organs of obesity and diabetes rats by increasing PPARγ. Nutr Metab (Lond). 2019;16:17. Lin X, et al. Aerobic exercise decreases chemerin/CMKLR1 in the serum and peripheral metabolic organs of obesity and diabetes rats by increasing PPARγ. Nutr Metab (Lond). 2019;16:17.
75.
Zurück zum Zitat Kukla M, et al. Potential role of leptin, adiponectin and three novel adipokines–visfatin, chemerin and vaspin–in chronic hepatitis. Mol Med. 2011;17(11–12):1397–410.PubMedPubMedCentral Kukla M, et al. Potential role of leptin, adiponectin and three novel adipokines–visfatin, chemerin and vaspin–in chronic hepatitis. Mol Med. 2011;17(11–12):1397–410.PubMedPubMedCentral
76.
Zurück zum Zitat Zhang W, et al. PI3K inhibition protects mice from NAFLD by down-regulating CMKLR1 and NLRP3 in Kupffer cells. J Physiol Biochem. 2017;73(4):583–94.PubMed Zhang W, et al. PI3K inhibition protects mice from NAFLD by down-regulating CMKLR1 and NLRP3 in Kupffer cells. J Physiol Biochem. 2017;73(4):583–94.PubMed
77.
Zurück zum Zitat Nosalski R, Guzik TJ. Perivascular adipose tissue inflammation in vascular disease. Br J Pharmacol. 2017;174(20):3496–513.PubMedPubMedCentral Nosalski R, Guzik TJ. Perivascular adipose tissue inflammation in vascular disease. Br J Pharmacol. 2017;174(20):3496–513.PubMedPubMedCentral
78.
Zurück zum Zitat Qi XY, et al. Perivascular adipose tissue (PVAT) in atherosclerosis: a double-edged sword. Cardiovasc Diabetol. 2018;17(1):134.PubMedPubMedCentral Qi XY, et al. Perivascular adipose tissue (PVAT) in atherosclerosis: a double-edged sword. Cardiovasc Diabetol. 2018;17(1):134.PubMedPubMedCentral
79.
Zurück zum Zitat Ferland DJ, et al. Whole-body but not hepatic knockdown of chemerin by antisense oligonucleotide decreases blood pressure in rats. J Pharmacol Exp Ther. 2018;365(2):212–8.PubMedPubMedCentral Ferland DJ, et al. Whole-body but not hepatic knockdown of chemerin by antisense oligonucleotide decreases blood pressure in rats. J Pharmacol Exp Ther. 2018;365(2):212–8.PubMedPubMedCentral
80.
Zurück zum Zitat Sluimer JC, Daemen MJ. Novel concepts in atherogenesis: angiogenesis and hypoxia in atherosclerosis. J Pathol. 2009;218(1):7–29.PubMed Sluimer JC, Daemen MJ. Novel concepts in atherogenesis: angiogenesis and hypoxia in atherosclerosis. J Pathol. 2009;218(1):7–29.PubMed
81.
Zurück zum Zitat Rupnick MA, et al. Adipose tissue mass can be regulated through the vasculature. Proc Natl Acad Sci U S A. 2002;99(16):10730–5.PubMedPubMedCentral Rupnick MA, et al. Adipose tissue mass can be regulated through the vasculature. Proc Natl Acad Sci U S A. 2002;99(16):10730–5.PubMedPubMedCentral
82.
Zurück zum Zitat Hobson SR, et al. Melatonin improves endothelial function in vitro and prolongs pregnancy in women with early-onset preeclampsia. J Pineal Res. 2018;65(3):e12508.PubMed Hobson SR, et al. Melatonin improves endothelial function in vitro and prolongs pregnancy in women with early-onset preeclampsia. J Pineal Res. 2018;65(3):e12508.PubMed
83.
Zurück zum Zitat Corre I, Paris F, Huot J. The p38 pathway, a major pleiotropic cascade that transduces stress and metastatic signals in endothelial cells. Oncotarget. 2017;8(33):55684–714.PubMedPubMedCentral Corre I, Paris F, Huot J. The p38 pathway, a major pleiotropic cascade that transduces stress and metastatic signals in endothelial cells. Oncotarget. 2017;8(33):55684–714.PubMedPubMedCentral
84.
Zurück zum Zitat Du J, et al. Role of autophagy in angiogenesis in aortic endothelial cells. Am J Physiol Cell Physiol. 2012;302(2):C383–91.PubMed Du J, et al. Role of autophagy in angiogenesis in aortic endothelial cells. Am J Physiol Cell Physiol. 2012;302(2):C383–91.PubMed
85.
Zurück zum Zitat Shen W, et al. Oxidative stress mediates chemerin-induced autophagy in endothelial cells. Free Radic Biol Med. 2013;55:73–82.PubMed Shen W, et al. Oxidative stress mediates chemerin-induced autophagy in endothelial cells. Free Radic Biol Med. 2013;55:73–82.PubMed
87.
Zurück zum Zitat Haybar H, et al. Endothelial Cells: From Dysfunction Mechanism to Pharmacological Effect in Cardiovascular Disease. Cardiovasc Toxicol. 2019;19(1):13–22.PubMed Haybar H, et al. Endothelial Cells: From Dysfunction Mechanism to Pharmacological Effect in Cardiovascular Disease. Cardiovasc Toxicol. 2019;19(1):13–22.PubMed
88.
Zurück zum Zitat Landgraf K, et al. Chemerin as a mediator between obesity and vascular inflammation in children. J Clin Endocrinol Metab. 2012;97(4):E556–64.PubMed Landgraf K, et al. Chemerin as a mediator between obesity and vascular inflammation in children. J Clin Endocrinol Metab. 2012;97(4):E556–64.PubMed
89.
Zurück zum Zitat Neves KB, et al. Chemerin regulates crosstalk between adipocytes and vascular cells through nox. Hypertension. 2015;66(3):657–66.PubMed Neves KB, et al. Chemerin regulates crosstalk between adipocytes and vascular cells through nox. Hypertension. 2015;66(3):657–66.PubMed
90.
Zurück zum Zitat Khatri JJ, et al. Vascular oxidant stress enhances progression and angiogenesis of experimental atheroma. Circulation. 2004;109(4):520–5.PubMed Khatri JJ, et al. Vascular oxidant stress enhances progression and angiogenesis of experimental atheroma. Circulation. 2004;109(4):520–5.PubMed
91.
Zurück zum Zitat Zhang W, et al. Sirt1 inhibits oxidative stress in vascular endothelial cells. Oxid Med Cell Longev. 2017;2017:7543973.PubMedPubMedCentral Zhang W, et al. Sirt1 inhibits oxidative stress in vascular endothelial cells. Oxid Med Cell Longev. 2017;2017:7543973.PubMedPubMedCentral
92.
Zurück zum Zitat Eftekhari A, et al. The promising future of nano-antioxidant therapy against environmental pollutants induced-toxicities. Biomed Pharmacother. 2018;103:1018–27.PubMed Eftekhari A, et al. The promising future of nano-antioxidant therapy against environmental pollutants induced-toxicities. Biomed Pharmacother. 2018;103:1018–27.PubMed
93.
Zurück zum Zitat Engin A. Endothelial dysfunction in obesity. Adv Exp Med Biol. 2017;960:345–79.PubMed Engin A. Endothelial dysfunction in obesity. Adv Exp Med Biol. 2017;960:345–79.PubMed
94.
Zurück zum Zitat Siragusa M, Fleming I. The eNOS signalosome and its link to endothelial dysfunction. Pflugers Arch. 2016;468(7):1125–37.PubMed Siragusa M, Fleming I. The eNOS signalosome and its link to endothelial dysfunction. Pflugers Arch. 2016;468(7):1125–37.PubMed
95.
Zurück zum Zitat Neves KB, et al. Chemerin reduces vascular nitric oxide/cGMP signalling in rat aorta: a link to vascular dysfunction in obesity? Clin Sci (Lond). 2014;127(2):111–22. Neves KB, et al. Chemerin reduces vascular nitric oxide/cGMP signalling in rat aorta: a link to vascular dysfunction in obesity? Clin Sci (Lond). 2014;127(2):111–22.
96.
Zurück zum Zitat Frismantiene A, et al. Cadherins in vascular smooth muscle cell (patho)biology: Quid nos scimus? Cell Signal. 2018;45:23–42.PubMed Frismantiene A, et al. Cadherins in vascular smooth muscle cell (patho)biology: Quid nos scimus? Cell Signal. 2018;45:23–42.PubMed
97.
Zurück zum Zitat Chistiakov DA, Orekhov AN, Bobryshev YV. Vascular smooth muscle cell in atherosclerosis. Acta Physiol (Oxf). 2015;214(1):33–50. Chistiakov DA, Orekhov AN, Bobryshev YV. Vascular smooth muscle cell in atherosclerosis. Acta Physiol (Oxf). 2015;214(1):33–50.
98.
Zurück zum Zitat Wen J, et al. Chemerin stimulates aortic smooth muscle cell proliferation and migration via activation of autophagy in VSMCs of metabolic hypertension rats. Am J Transl Res. 2019;11(3):1327–42.PubMedPubMedCentral Wen J, et al. Chemerin stimulates aortic smooth muscle cell proliferation and migration via activation of autophagy in VSMCs of metabolic hypertension rats. Am J Transl Res. 2019;11(3):1327–42.PubMedPubMedCentral
99.
Zurück zum Zitat Kunimoto H, et al. Chemerin promotes the proliferation and migration of vascular smooth muscle and increases mouse blood pressure. Am J Physiol Heart Circ Physiol. 2015;309(5):H1017–28.PubMed Kunimoto H, et al. Chemerin promotes the proliferation and migration of vascular smooth muscle and increases mouse blood pressure. Am J Physiol Heart Circ Physiol. 2015;309(5):H1017–28.PubMed
100.
Zurück zum Zitat Hanthazi A, et al. Chemerin added to endothelin-1 promotes rat pulmonary artery smooth muscle cell proliferation and migration. Front Physiol. 2020;11:926.PubMedPubMedCentral Hanthazi A, et al. Chemerin added to endothelin-1 promotes rat pulmonary artery smooth muscle cell proliferation and migration. Front Physiol. 2020;11:926.PubMedPubMedCentral
101.
Zurück zum Zitat Han JH, et al. Regulation of autophagy by controlling Erk1/2 and mTOR for platelet-derived growth factor-BB-mediated vascular smooth muscle cell phenotype shift. Life Sci. 2021;267:118978.PubMed Han JH, et al. Regulation of autophagy by controlling Erk1/2 and mTOR for platelet-derived growth factor-BB-mediated vascular smooth muscle cell phenotype shift. Life Sci. 2021;267:118978.PubMed
102.
Zurück zum Zitat Rudijanto A. The role of vascular smooth muscle cells on the pathogenesis of atherosclerosis. Acta Med Indones. 2007;39(2):86–93.PubMed Rudijanto A. The role of vascular smooth muscle cells on the pathogenesis of atherosclerosis. Acta Med Indones. 2007;39(2):86–93.PubMed
103.
Zurück zum Zitat Satoh K, Nigro P, Berk BC. Oxidative stress and vascular smooth muscle cell growth: a mechanistic linkage by cyclophilin A. Antioxid Redox Signal. 2010;12(5):675–82.PubMedPubMedCentral Satoh K, Nigro P, Berk BC. Oxidative stress and vascular smooth muscle cell growth: a mechanistic linkage by cyclophilin A. Antioxid Redox Signal. 2010;12(5):675–82.PubMedPubMedCentral
104.
Zurück zum Zitat Das S, et al. A novel angiotensin II-induced long noncoding RNA giver regulates oxidative stress, inflammation, and proliferation in vascular smooth muscle cells. Circ Res. 2018;123(12):1298–312.PubMedPubMedCentral Das S, et al. A novel angiotensin II-induced long noncoding RNA giver regulates oxidative stress, inflammation, and proliferation in vascular smooth muscle cells. Circ Res. 2018;123(12):1298–312.PubMedPubMedCentral
105.
Zurück zum Zitat Cui L, et al. Mitoquinone attenuates vascular calcification by suppressing oxidative stress and reducing apoptosis of vascular smooth muscle cells via the Keap1/Nrf2 pathway. Free Radic Biol Med. 2020;161:23–31.PubMed Cui L, et al. Mitoquinone attenuates vascular calcification by suppressing oxidative stress and reducing apoptosis of vascular smooth muscle cells via the Keap1/Nrf2 pathway. Free Radic Biol Med. 2020;161:23–31.PubMed
107.
Zurück zum Zitat Kostopoulos CG, et al. Chemerin and CMKLR1 expression in human arteries and periadventitial fat: a possible role for local chemerin in atherosclerosis? BMC Cardiovasc Disord. 2014;14:56.PubMedPubMedCentral Kostopoulos CG, et al. Chemerin and CMKLR1 expression in human arteries and periadventitial fat: a possible role for local chemerin in atherosclerosis? BMC Cardiovasc Disord. 2014;14:56.PubMedPubMedCentral
108.
Zurück zum Zitat Aydın K, et al. Chemerin is not associated with subclinical atherosclerosis markers in prediabetes and diabetes. Anatol J Cardiol. 2016;16(10):749–55.PubMed Aydın K, et al. Chemerin is not associated with subclinical atherosclerosis markers in prediabetes and diabetes. Anatol J Cardiol. 2016;16(10):749–55.PubMed
109.
Zurück zum Zitat Lehrke M, et al. Chemerin is associated with markers of inflammation and components of the metabolic syndrome but does not predict coronary atherosclerosis. Eur J Endocrinol. 2009;161(2):339–44.PubMed Lehrke M, et al. Chemerin is associated with markers of inflammation and components of the metabolic syndrome but does not predict coronary atherosclerosis. Eur J Endocrinol. 2009;161(2):339–44.PubMed
110.
Zurück zum Zitat Zylla S, et al. Association of circulating chemerin with subclinical parameters of atherosclerosis: results of a population-based study. Arterioscler Thromb Vasc Biol. 2018;38(7):1656–64.PubMedPubMedCentral Zylla S, et al. Association of circulating chemerin with subclinical parameters of atherosclerosis: results of a population-based study. Arterioscler Thromb Vasc Biol. 2018;38(7):1656–64.PubMedPubMedCentral
111.
Zurück zum Zitat Liu H, et al. Adipokine chemerin stimulates progression of atherosclerosis in ApoE(−/−) Mice. Biomed Res Int. 2019;2019:7157865.PubMedPubMedCentral Liu H, et al. Adipokine chemerin stimulates progression of atherosclerosis in ApoE(−/−) Mice. Biomed Res Int. 2019;2019:7157865.PubMedPubMedCentral
112.
Zurück zum Zitat van der Vorst E, et al. Hematopoietic chemR23 (Chemerin Receptor 23) fuels atherosclerosis by sustaining an M1 macrophage-phenotype and guidance of plasmacytoid dendritic cells to murine lesions-brief report. Arterioscler Thromb Vasc Biol. 2019;39(4):685–93.PubMed van der Vorst E, et al. Hematopoietic chemR23 (Chemerin Receptor 23) fuels atherosclerosis by sustaining an M1 macrophage-phenotype and guidance of plasmacytoid dendritic cells to murine lesions-brief report. Arterioscler Thromb Vasc Biol. 2019;39(4):685–93.PubMed
113.
Zurück zum Zitat Jia J, et al. Chemerin enhances the adhesion and migration of human endothelial progenitor cells and increases lipid accumulation in mice with atherosclerosis. Lipids Health Dis. 2020;19(1):207.PubMedPubMedCentral Jia J, et al. Chemerin enhances the adhesion and migration of human endothelial progenitor cells and increases lipid accumulation in mice with atherosclerosis. Lipids Health Dis. 2020;19(1):207.PubMedPubMedCentral
114.
Zurück zum Zitat Lachine NA, et al. Serum chemerin and high-sensitivity C reactive protein as markers of subclinical atherosclerosis in Egyptian patients with type 2 diabetes. Ther Adv Endocrinol Metab. 2016;7(2):47–56.PubMedPubMedCentral Lachine NA, et al. Serum chemerin and high-sensitivity C reactive protein as markers of subclinical atherosclerosis in Egyptian patients with type 2 diabetes. Ther Adv Endocrinol Metab. 2016;7(2):47–56.PubMedPubMedCentral
115.
Zurück zum Zitat Xiang H, et al. Changes in serum levels of adipokine after treatment in children with Kawasaki disease. Zhongguo Dang Dai Er Ke Za Zhi. 2020;22(1):53–7.PubMed Xiang H, et al. Changes in serum levels of adipokine after treatment in children with Kawasaki disease. Zhongguo Dang Dai Er Ke Za Zhi. 2020;22(1):53–7.PubMed
116.
Zurück zum Zitat Liang Z, et al. The elevated levels of plasma chemerin and C-reactive protein in patients with acute coronary syndrome. Xi Bao Yu Fen Zi Mian Yi Xue Za Zhi. 2015;31(7):953–6.PubMed Liang Z, et al. The elevated levels of plasma chemerin and C-reactive protein in patients with acute coronary syndrome. Xi Bao Yu Fen Zi Mian Yi Xue Za Zhi. 2015;31(7):953–6.PubMed
117.
Zurück zum Zitat Ji Q, et al. Chemerin is a novel biomarker of acute coronary syndrome but not of stable angina pectoris. Cardiovasc Diabetol. 2014;13:145.PubMedPubMedCentral Ji Q, et al. Chemerin is a novel biomarker of acute coronary syndrome but not of stable angina pectoris. Cardiovasc Diabetol. 2014;13:145.PubMedPubMedCentral
118.
Zurück zum Zitat Aronis KN, et al. Chemerin levels as predictor of acute coronary events: a case-control study nested within the veterans affairs normative aging study. Metabolism. 2014;63(6):760–6.PubMed Aronis KN, et al. Chemerin levels as predictor of acute coronary events: a case-control study nested within the veterans affairs normative aging study. Metabolism. 2014;63(6):760–6.PubMed
119.
Zurück zum Zitat Gu P, et al. Elevating circulation chemerin level is associated with endothelial dysfunction and early atherosclerotic changes in essential hypertensive patients. J Hypertens. 2015;33(8):1624–32.PubMed Gu P, et al. Elevating circulation chemerin level is associated with endothelial dysfunction and early atherosclerotic changes in essential hypertensive patients. J Hypertens. 2015;33(8):1624–32.PubMed
120.
Zurück zum Zitat Wójcik M, et al. Circulating chemerin level may be associated with early vascular pathology in obese children without overt arterial hypertension - preliminary results. J Pediatr Endocrinol Metab. 2020;33(6):729–34.PubMed Wójcik M, et al. Circulating chemerin level may be associated with early vascular pathology in obese children without overt arterial hypertension - preliminary results. J Pediatr Endocrinol Metab. 2020;33(6):729–34.PubMed
122.
Zurück zum Zitat Ferland DJ, Mullick AE, Watts SW. Chemerin as a driver of hypertension: a consideration. Am J Hypertens. 2020;33(11):975–86.PubMedPubMedCentral Ferland DJ, Mullick AE, Watts SW. Chemerin as a driver of hypertension: a consideration. Am J Hypertens. 2020;33(11):975–86.PubMedPubMedCentral
123.
Zurück zum Zitat Omori A, et al. Chemerin-9-induced contraction was enhanced through the upregulation of smooth muscle chemokine-like receptor 1 in isolated pulmonary artery of pulmonary arterial hypertensive rats. Pflugers Arch. 2020;472(3):335–42.PubMed Omori A, et al. Chemerin-9-induced contraction was enhanced through the upregulation of smooth muscle chemokine-like receptor 1 in isolated pulmonary artery of pulmonary arterial hypertensive rats. Pflugers Arch. 2020;472(3):335–42.PubMed
124.
Zurück zum Zitat Lurins J, et al. Impact of several proinflammatory and cell degradation factors in patients with aortic valve stenosis. Exp Ther Med. 2019;17(4):2433–42.PubMedPubMedCentral Lurins J, et al. Impact of several proinflammatory and cell degradation factors in patients with aortic valve stenosis. Exp Ther Med. 2019;17(4):2433–42.PubMedPubMedCentral
125.
Zurück zum Zitat Lurins J, et al. Increased serum chemerin level to predict early onset of aortic valve stenosis. Biomed Rep. 2018;8(1):31–6.PubMed Lurins J, et al. Increased serum chemerin level to predict early onset of aortic valve stenosis. Biomed Rep. 2018;8(1):31–6.PubMed
126.
Zurück zum Zitat Liu CL, et al. Adipocytes promote interleukin-18 binding to its receptors during abdominal aortic aneurysm formation in mice. Eur Heart J. 2020;41(26):2456–68.PubMed Liu CL, et al. Adipocytes promote interleukin-18 binding to its receptors during abdominal aortic aneurysm formation in mice. Eur Heart J. 2020;41(26):2456–68.PubMed
127.
Zurück zum Zitat Police SB, et al. Obesity promotes inflammation in periaortic adipose tissue and angiotensin II-induced abdominal aortic aneurysm formation. Arterioscler Thromb Vasc Biol. 2009;29(10):1458–64.PubMedPubMedCentral Police SB, et al. Obesity promotes inflammation in periaortic adipose tissue and angiotensin II-induced abdominal aortic aneurysm formation. Arterioscler Thromb Vasc Biol. 2009;29(10):1458–64.PubMedPubMedCentral
128.
Zurück zum Zitat Yoo HJ, et al. Circulating chemerin level is independently correlated with arterial stiffness. J Atheroscler Thromb. 2012;19(1):59–66.PubMed Yoo HJ, et al. Circulating chemerin level is independently correlated with arterial stiffness. J Atheroscler Thromb. 2012;19(1):59–66.PubMed
129.
Zurück zum Zitat Manco M, et al. Arterial stiffness, thickness and association to suitable novel markers of risk at the origin of cardiovascular disease in obese children. Int J Med Sci. 2017;14(8):711–20.PubMedPubMedCentral Manco M, et al. Arterial stiffness, thickness and association to suitable novel markers of risk at the origin of cardiovascular disease in obese children. Int J Med Sci. 2017;14(8):711–20.PubMedPubMedCentral
130.
Zurück zum Zitat Carracedo M, et al. Chemerin inhibits vascular calcification through ChemR23 and is associated with lower coronary calcium in chronic kidney disease. J Intern Med. 2019;286(4):449–57.PubMedPubMedCentral Carracedo M, et al. Chemerin inhibits vascular calcification through ChemR23 and is associated with lower coronary calcium in chronic kidney disease. J Intern Med. 2019;286(4):449–57.PubMedPubMedCentral
131.
Zurück zum Zitat Darios ES, et al. The adipokine chemerin amplifies electrical field-stimulated contraction in the isolated rat superior mesenteric artery. Am J Physiol Heart Circ Physiol. 2016;311(2):H498-507.PubMedPubMedCentral Darios ES, et al. The adipokine chemerin amplifies electrical field-stimulated contraction in the isolated rat superior mesenteric artery. Am J Physiol Heart Circ Physiol. 2016;311(2):H498-507.PubMedPubMedCentral
132.
Zurück zum Zitat Jannaway M, et al. Resolvin E1, resolvin D1 and resolvin D2 inhibit constriction of rat thoracic aorta and human pulmonary artery induced by the thromboxane mimetic U46619. Br J Pharmacol. 2018;175(7):1100–8.PubMedPubMedCentral Jannaway M, et al. Resolvin E1, resolvin D1 and resolvin D2 inhibit constriction of rat thoracic aorta and human pulmonary artery induced by the thromboxane mimetic U46619. Br J Pharmacol. 2018;175(7):1100–8.PubMedPubMedCentral
133.
Zurück zum Zitat Carracedo M, et al. The resolution of inflammation through omega-3 fatty acids in atherosclerosis, intimal hyperplasia, and vascular calcification. Semin Immunopathol. 2019;41(6):757–66.PubMedPubMedCentral Carracedo M, et al. The resolution of inflammation through omega-3 fatty acids in atherosclerosis, intimal hyperplasia, and vascular calcification. Semin Immunopathol. 2019;41(6):757–66.PubMedPubMedCentral
134.
Zurück zum Zitat Chen J, et al. RvE1 attenuates polymicrobial sepsis-induced cardiac dysfunction and enhances bacterial clearance. Front Immunol. 2020;11:2080.PubMedPubMedCentral Chen J, et al. RvE1 attenuates polymicrobial sepsis-induced cardiac dysfunction and enhances bacterial clearance. Front Immunol. 2020;11:2080.PubMedPubMedCentral
135.
Zurück zum Zitat Bäck M, Hansson GK. Omega-3 fatty acids, cardiovascular risk, and the resolution of inflammation. FASEB J. 2019;33(2):1536–9.PubMed Bäck M, Hansson GK. Omega-3 fatty acids, cardiovascular risk, and the resolution of inflammation. FASEB J. 2019;33(2):1536–9.PubMed
136.
Zurück zum Zitat Laguna-Fernandez A, et al. ERV1/ChemR23 signaling protects against atherosclerosis by modifying oxidized low-density lipoprotein uptake and phagocytosis in macrophages. Circulation. 2018;138(16):1693–705.PubMedPubMedCentral Laguna-Fernandez A, et al. ERV1/ChemR23 signaling protects against atherosclerosis by modifying oxidized low-density lipoprotein uptake and phagocytosis in macrophages. Circulation. 2018;138(16):1693–705.PubMedPubMedCentral
137.
Zurück zum Zitat Hasturk H, et al. Resolvin E1 (RvE1) Attenuates Atherosclerotic Plaque Formation in Diet and Inflammation-Induced Atherogenesis. Arterioscler Thromb Vasc Biol. 2015;35(5):1123–33.PubMedPubMedCentral Hasturk H, et al. Resolvin E1 (RvE1) Attenuates Atherosclerotic Plaque Formation in Diet and Inflammation-Induced Atherogenesis. Arterioscler Thromb Vasc Biol. 2015;35(5):1123–33.PubMedPubMedCentral
138.
Zurück zum Zitat Salic K, et al. Resolvin E1 attenuates atherosclerosis in absence of cholesterol-lowering effects and on top of atorvastatin. Atherosclerosis. 2016;250:158–65.PubMed Salic K, et al. Resolvin E1 attenuates atherosclerosis in absence of cholesterol-lowering effects and on top of atorvastatin. Atherosclerosis. 2016;250:158–65.PubMed
139.
Zurück zum Zitat Liu G, et al. Resolvin E1 attenuates injury-induced vascular neointimal formation by inhibition of inflammatory responses and vascular smooth muscle cell migration. FASEB J. 2018;32(10):5413–25.PubMed Liu G, et al. Resolvin E1 attenuates injury-induced vascular neointimal formation by inhibition of inflammatory responses and vascular smooth muscle cell migration. FASEB J. 2018;32(10):5413–25.PubMed
140.
141.
Zurück zum Zitat Carracedo M, et al. The G-protein coupled receptor ChemR23 determines smooth muscle cell phenotypic switching to enhance high phosphate-induced vascular calcification. Cardiovasc Res. 2019;115(10):1557–66.PubMed Carracedo M, et al. The G-protein coupled receptor ChemR23 determines smooth muscle cell phenotypic switching to enhance high phosphate-induced vascular calcification. Cardiovasc Res. 2019;115(10):1557–66.PubMed
142.
Zurück zum Zitat Fisk HL, et al. A novel n-3 glyceride mixture enhances enrichment of EPA and DHA after single dosing in healthy older adults: results from a double-blind crossover trial. Br J Nutr. 2021;126(2):244–52.PubMed Fisk HL, et al. A novel n-3 glyceride mixture enhances enrichment of EPA and DHA after single dosing in healthy older adults: results from a double-blind crossover trial. Br J Nutr. 2021;126(2):244–52.PubMed
143.
Zurück zum Zitat Nakao K, et al. Effect of eicosapentaenoic acid/docosahexaenoic acid on coronary high-intensity plaques detected with non-contrast T1-weighted imaging (the AQUAMARINE EPA/DHA study): study protocol for a randomized controlled trial. Trials. 2018;19(1):12.PubMedPubMedCentral Nakao K, et al. Effect of eicosapentaenoic acid/docosahexaenoic acid on coronary high-intensity plaques detected with non-contrast T1-weighted imaging (the AQUAMARINE EPA/DHA study): study protocol for a randomized controlled trial. Trials. 2018;19(1):12.PubMedPubMedCentral
144.
Zurück zum Zitat Kris-Etherton PM, et al. Recent clinical trials shed new light on the cardiovascular benefits of omega-3 fatty acids. Methodist Debakey Cardiovasc J. 2019;15(3):171–8.PubMedPubMedCentral Kris-Etherton PM, et al. Recent clinical trials shed new light on the cardiovascular benefits of omega-3 fatty acids. Methodist Debakey Cardiovasc J. 2019;15(3):171–8.PubMedPubMedCentral
145.
Zurück zum Zitat Golzari MH, et al. Effect of Eicosapentaenoic acid (EPA) supplementation on cardiovascular markers in patients with type 2 diabetes mellitus: a randomized, double-blind, placebo-controlled trial. Diabetes Metab Syndr. 2018;12(3):411–5.PubMed Golzari MH, et al. Effect of Eicosapentaenoic acid (EPA) supplementation on cardiovascular markers in patients with type 2 diabetes mellitus: a randomized, double-blind, placebo-controlled trial. Diabetes Metab Syndr. 2018;12(3):411–5.PubMed
146.
Zurück zum Zitat Chen S, et al. Chemerin-9 attenuates experimental abdominal aortic aneurysm formation in ApoE(−/−) Mice. J Oncol. 2021;2021:6629204.PubMedPubMedCentral Chen S, et al. Chemerin-9 attenuates experimental abdominal aortic aneurysm formation in ApoE(−/−) Mice. J Oncol. 2021;2021:6629204.PubMedPubMedCentral
147.
Zurück zum Zitat Sato K, et al. Chemerin-9, a potent agonist of chemerin receptor (ChemR23), prevents atherogenesis. Clin Sci (Lond). 2019;133(16):1779–96. Sato K, et al. Chemerin-9, a potent agonist of chemerin receptor (ChemR23), prevents atherogenesis. Clin Sci (Lond). 2019;133(16):1779–96.
148.
Zurück zum Zitat Ferland DJ, et al. Different blood pressure responses in hypertensive rats following chemerin mRNA inhibition in dietary high fat compared to dietary high-salt conditions. Physiol Genomics. 2019;51(11):553–61.PubMedPubMedCentral Ferland DJ, et al. Different blood pressure responses in hypertensive rats following chemerin mRNA inhibition in dietary high fat compared to dietary high-salt conditions. Physiol Genomics. 2019;51(11):553–61.PubMedPubMedCentral
149.
Zurück zum Zitat Hu B, et al. Food macromolecule based nanodelivery systems for enhancing the bioavailability of polyphenols. J Food Drug Anal. 2017;25(1):3–15.PubMed Hu B, et al. Food macromolecule based nanodelivery systems for enhancing the bioavailability of polyphenols. J Food Drug Anal. 2017;25(1):3–15.PubMed
150.
Zurück zum Zitat Hasanzadeh M, et al. Poly arginine-graphene quantum dots as a biocompatible and non-toxic nanocomposite: layer-by-layer electrochemical preparation, characterization and non-invasive malondialdehyde sensory application in exhaled breath condensate. Mater Sci Eng C Mater Biol Appl. 2017;75:247–58.PubMed Hasanzadeh M, et al. Poly arginine-graphene quantum dots as a biocompatible and non-toxic nanocomposite: layer-by-layer electrochemical preparation, characterization and non-invasive malondialdehyde sensory application in exhaled breath condensate. Mater Sci Eng C Mater Biol Appl. 2017;75:247–58.PubMed
151.
Zurück zum Zitat Hasanzadeh M, et al. An innovative immunosensor for ultrasensitive detection of breast cancer specific carbohydrate (CA 15–3) in unprocessed human plasma and MCF-7 breast cancer cell lysates using gold nanospear electrochemically assembled onto thiolated graphene quantum dots. Int J Biol Macromol. 2018;114:1008–17.PubMed Hasanzadeh M, et al. An innovative immunosensor for ultrasensitive detection of breast cancer specific carbohydrate (CA 15–3) in unprocessed human plasma and MCF-7 breast cancer cell lysates using gold nanospear electrochemically assembled onto thiolated graphene quantum dots. Int J Biol Macromol. 2018;114:1008–17.PubMed
Metadaten
Titel
Role of Chemerin/ChemR23 axis as an emerging therapeutic perspective on obesity-related vascular dysfunction
verfasst von
Yingying Xie
Ling Liu
Publikationsdatum
01.12.2022
Verlag
BioMed Central
Erschienen in
Journal of Translational Medicine / Ausgabe 1/2022
Elektronische ISSN: 1479-5876
DOI
https://doi.org/10.1186/s12967-021-03220-7

Weitere Artikel der Ausgabe 1/2022

Journal of Translational Medicine 1/2022 Zur Ausgabe

Leitlinien kompakt für die Innere Medizin

Mit medbee Pocketcards sicher entscheiden.

Seit 2022 gehört die medbee GmbH zum Springer Medizin Verlag

Die „Zehn Gebote“ des Endokarditis-Managements

30.04.2024 Endokarditis Leitlinie kompakt

Worauf kommt es beim Management von Personen mit infektiöser Endokarditis an? Eine Kardiologin und ein Kardiologe fassen die zehn wichtigsten Punkte der neuen ESC-Leitlinie zusammen.

Strenge Blutdruckeinstellung lohnt auch im Alter noch

30.04.2024 Arterielle Hypertonie Nachrichten

Ältere Frauen, die von chronischen Erkrankungen weitgehend verschont sind, haben offenbar die besten Chancen, ihren 90. Geburtstag zu erleben, wenn ihr systolischer Blutdruck < 130 mmHg liegt. Das scheint selbst für 80-Jährige noch zu gelten.

Frauen bekommen seltener eine intensive Statintherapie

30.04.2024 Statine Nachrichten

Frauen in den Niederlanden erhalten bei vergleichbarem kardiovaskulärem Risiko seltener eine intensive Statintherapie als Männer. Ihre LDL-Zielwerte erreichen sie aber fast ähnlich oft.

Reizdarmsyndrom: Diäten wirksamer als Medikamente

29.04.2024 Reizdarmsyndrom Nachrichten

Bei Reizdarmsyndrom scheinen Diäten, wie etwa die FODMAP-arme oder die kohlenhydratreduzierte Ernährung, effektiver als eine medikamentöse Therapie zu sein. Das hat eine Studie aus Schweden ergeben, die die drei Therapieoptionen im direkten Vergleich analysierte.

Update Innere Medizin

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.