Skip to main content

Emerging role of chemokine CC motif ligand 4 related mechanisms in diabetes mellitus and cardiovascular disease: friends or foes?

Abstract

Chemokines are critical components in pathology. The roles of chemokine CC motif ligand 4 (CCL4) and its receptor are associated with diabetes mellitus (DM) and atherosclerosis cardiovascular diseases. However, due to the complexity of these diseases, the specific effects of CCL4 remain unclear, although recent reports have suggested that multiple pathways are related to CCL4. In this review, we provide an overview of the role and potential mechanisms of CCL4 and one of its major receptors, fifth CC chemokine receptor (CCR5), in DM and cardiovascular diseases. CCL4-related mechanisms, including CCL4 and CCR5, might provide potential therapeutic targets in DM and/or atherosclerosis cardiovascular diseases.

Background

Chemokines and their seven-transmembrane, G-protein coupled receptors have been recognized as key mediators in the pathology of both diabetes mellitus (DM) and atherosclerosis cardiovascular diseases. These small molecular weight chemo-tactic cytokines recruit and physiologically direct cell migration. They can be broadly divided into four subfamilies, the CXC, CC, C, and CX3C families, according to their N-terminal cysteine -motifs [1]. Moreover, chemokines are also defined as “homeostatic” chemokines and “inflammatory” chemokines based on their functions. Homeostatic chemokines are constitutively secreted and involved in lymphocyte traffic, while inflammatory chemokines mediate pro-inflammatory signals and induce leukocyte recruitment to damaged tissue [2].

Diabetes patients have an increased risk of atherosclerosis cardiovascular diseases compared to people without diabetes. Diabetic vascular complications are the major cause of morbidity and early mortality in type 2 DM, suggesting the close link between type 2 DM and atherosclerosis cardiovascular disease [35]. Both type 2 DM and atherosclerosis cardiovascular diseases have been increasingly recognized as inflammatory related diseases. Recently, chemokine CC motif ligand 4 (CCL4), also known as macrophage inflammatory protein-1β (MIP-1β), a member of the CC chemokine family, was suggested to play a potential role in the development and/or progression of DM and atherosclerosis disease [6]. Besides, other chemokines such as chemerin may also related to inflammation and could act as novel biomarkers of acute coronary syndrome [7]. Therefore, inflammatory chemokines such as CCL4 might act as a possible node to link the presence of DM and atherosclerosis. However, the current CCL4 data are not consistent, especially for the complications of diabetes. This review focuses on the emerging evidence for the multiple roles of CCL4-related mechanisms, including those of CCL4 and one of its major receptors, fifth CC chemokine receptor (CCR5), in both experimental and clinical DM and atherosclerosis cardiovascular diseases. It may also provide the rationale for the potential role of CCL4-related mechanisms as therapeutic targets in clinical DM and atherosclerosis diseases.

Chemokine CC motif ligand 4 (CCL4, MIP-1β)

The molecular weight of CCL4 is 7.8 kDa. Its gene is located on chromosome 17 [6]. Murine and human CCL4 proteins are synthesized as 92 amino acid precursors. Mature secreted proteins of 69 amino acids are generated by peptidases that cleave hydrophobic signal peptides [8, 9]. The three-dimensional structure of human CCL4 has been identified by heteronuclear magnetic resonance. CCL4 exists as a symmetrical homodimer. The main secondary structure elements comprise a triple-stranded antiparallel β sheet and an NH2-terminus followed by a four-residue helical turn. In general, the CCL4 dimer is elongated and cylindrical [10].

CCL4 was first isolated from the culture medium of LPS-activated macrophages [11]. LPS can impair muscle glucose uptake by both the direct effects of inflammation on myocytes, as well as by indirect NO-driven cardiovascular dysfunction [12]. CCL4 may exhibit chemoattractive ability towards different cell types, including macrophages, natural killer cells, monocytes, immature dendritic cells, and coronary endothelial cells [1317]. In addition, CCL4 has been shown to induce calcium mobilization in natural killer cells, monocytes, leukocytes, vascular smooth muscle cells, and progenitor B cells [1416, 18, 19]. The upregulation of circulating CCL4 levels was also observed in patients with type 2 DM and/or clinical atherosclerosis cardiovascular diseases [2024].

The role of CCL4 in type 1 and type 2 diabetes mellitus

Serial chemokines have been shown to be associated with the progression of both type 1 and type 2 DM. While macrophages may be implicated in the destruction of islet cells with the increase in blood glucose and the progression of both type1 and type 2 DM, CCL4 is one of the major macrophage attractants [25, 26]. While resulting in β-cell death and early islet graft loss, inflammatory stimuli with a CD40-CD40L interaction could induce the secretion of CCL4 through the Raf/MEK/ERK and NF-κB pathways in pancreatic islets [27]. CCL4 may also be produced by human islet ductal cells, which could be suppressed by sirolimus, an inhibitor of cell proliferation [28]. Clinically, the circulating CCL4 concentrations may be increased not only in the multiple islet autoantibody-positive group type 1 diabetes patients but also in prediabetic patients [20]. High-risk individuals that later developed type 1 DM had the increased secretion of the pro-inflammatory chemokine CCL4 [22]. In addition, circulating concentrations of CCL4 may be similar between type 1 and type 2 DM patients, suggesting the universal involvement of CCL4 in different stages and types of DM [29].

However, one study reported that CCL4 was significantly lower in patients with type 1 DM than in controls [30]. It was also shown that release of CCL4 from monocytes was dose-dependently induced by adiponectin [31]. Interestingly, in type 2 diabetic patients, insulin infusion could significantly suppress the expression of CCL4 in mononuclear cells. These findings are relevant because insulin is anti-inflammatory [32, 33]. Insulin also suppresses RANTES, which is atherogenic, and eotaxin, which is allergogenic [34]. Both of them bind to CCR5 as their receptor. Besides, the deletion of insulin receptor in endothelial cells leads to atherosclerosis [35]. The administration of insulin to apo E-deleted animals could also inhibit atherogenesis, suggesting the potential anti-atherosclerosis effects of insulin [36].

Taken together, in most clinical diabetes patients, circulating CCL4 levels may be increased early with the dysfunction of β-cells, even before they are extensively damaged. Further, exogenous insulin supplementation may eventually reduce CCL4 expression, likely by unloading the β-cells, as circulating CCL4 levels were inversely associated with proinsulin levels [37]. Thus, CCL4 might be produced by, rather than induce, the initial inflammatory damage on islet β-cells.

Indeed, the in vivo pathogenesis role of CCL4 in experimental diabetes may be obscured. A protective rather than aggravated role was suggested in animal studies, mainly in nonobese diabetic (NOD) mouse models. NOD mice can spontaneously develop a form of type 1 diabetes that shares some features of the human disease [38]. It was previously shown in the NOD mouse model that Th1 MNCs could enter and/or accumulate in the pancreas more rapidly than Th2 MNCs and induce immune-associated diabetes. However, both subsets induce adhesion receptors on vascular endothelium coincident with their infiltration. In vitro, Th1 cells were also distinguished from Th2 cells by the capacity to synthesize several chemokines, including lymphotactin, monocyte chemoattractant protein-1, and CCL3 (MIP-1α), whereas both subsets produced CCL4 (MIP-1β) [39]. Although the Th1/Th2 paradigm provided a conceptual framework for diseases, the opposite results of Th2-skewing agents indicate that the Th1/Th2 paradigm is too simple to explain DM pathogenesis [4043], possibly because Th1/Th2 cytokines have pleiotropic functions beyond T helper cell phenotype modulation [44]. These studies indicate that Th1/Th2 modulation alone is insufficient for DM treatment. In addition, a recent study indicated the Treg control immune homeostasis via the CCL4/CCR5 pathway, suggesting a potential relationship between CCL4/CCR5 and Th17/Treg [45]. In another NOD mouse model, an elevated ratio of CCL3 to CCL4 in the pancreas was correlated with destructive insulitis and progression to diabetes [38]. Moreover, a decreased intra-pancreatic CCL3 to CCL4 ratio was also observed in nonobese diabetes-resistant mice. NOD.CCL3-/- mice but not NOD.CCL4-/- mice exhibited reduced destructive insulitis and were protected from type 1 DM. In NOD.Scid mice, which do not have functional T or B cells, CCL4 expression was undetectable in the pancreas, suggesting that the intra-pancreatic expression of CCL4 may depend on the presence of infiltrated MNCs. In NOD.Scid recipients, the neutralization of CCL3 with specific antibodies following transfer of diabetogenic T cells delayed the onset of diabetes, while the injection with anti-CCL4 mAb did not [46]. Furthermore, in Lewis rats, in vivo neutralization of the activity of CCL4 exacerbated the disease [47]. Blockade of IL-16 in vivo protected against type 1 DM in NOD mice by interfering with recruitment of T-cells to the pancreas, and this protection required the activity of CCL4 [48, 49]. Another study also showed that the protection from type 1 diabetes elicited by insulin-like growth factor (IGF)-I/IGF-binding protein-3 was mediated by the upregulation of CCL4 gene expression in pancreatic-draining lymph nodes, activation of the phosphatidylinositol 3-kinase and Akt/protein kinase B signaling pathway of β-cells, reduced β-cell apoptosis, and stimulation of β-cell replication [50]. It was further shown that exogenous CCL4 supplementation could suppress rather than accelerate inflammatory responses targeting islet β-cells [51]. Thus, the role of CCL4 is still undefined in DM.

Currently, almost all CCL4-related type 1 DM effects have been observed in the NOD mouse model. Ideally, more than one animal model should be investigated due to the complexity of DM [52]. Interventions in the NOD mouse studies have been reported [53]. First, the agent efficacy frequently varied when mice were treated at different ages. For example, early treatment with TNF-α exacerbated the disease but later treatment protected from disease [54]. These data suggested that the different modulations during disease progression might result in opposite effects. Second, different efficacies were observed in animal models. NOD mice and BB rats are both spontaneous autoimmune models of type 1 DM. However, nicotinamide and oral insulin showed protection in prediabetic NOD mice, but not in BB rats [5558]. Commonly, it is easy to prevent type 1 DM onset in NOD mice if treatment is initiated early, but more difficult later in disease. As many type 1 DM patients are identified at diabetes onset, agents for DM treatment rather than prevention are urgently needed. Interestingly, previous data showed that the intra-pancreatic CCL4 concentration was relatively lower than that of CCL3 in NOD mice [50]. However, clinical data revealed that CCL4 levels were higher than CCL3 levels [21]. On the other hand, concentrations of CCL4 did not differ between groups, but CCL3 was higher in patients with latent autoimmune diabetes and type 1 diabetes than in those with type 2 diabetes and control subjects [29]. Taken together, these data imply that CCL4 levels might be different because of the different stage of DM development or the complexity of DM. As a result, although anti-CCL4 showed protective effects when the CCL4 level was relatively lower in pre-diabetic NOD mice, the effects of anti-CCL4 should also be tested when the CCL4 level is higher, as in late DM and/or in other animal models of DM.

In summary, while CCL3 contributes to the development of type 1 DM, CCL4 might play a protective role in some experimental diabetes, especially the NOD type 1 DM model. However, the role of CCL4 is much less clear in type 2 DM and might be varied in different animal models of experimental diabetes. Future studies should be required to clarify the mechanistic insights and to evaluate the clinical impact of CCL4 in the development of either type 1 or type 2 DM in humans.

The role of CCL4 in atherosclerosis cardiovascular diseases

Previous studies showed that the inflammatory microenvironment influences cell recruitment and activation, opening new investigative fields for pathophysiological studies in cardiovascular diseases. In vitro, CCL4 was able to induce reactive oxygen species production and adhesion of THP-1 cells to human umbilical vein endothelial cells. CCL4 directly induced cell adhesion to endothelial cells through oxidative stress via PI3k–Rac1 cascades [23]. Also, macrophages under high glucose conditions released more CCL4. CCL4 secreted from macrophages under high glucose conditions is capable of inducing the endothelial expression of adhesion molecule such as E-selectin in an in vitro study [59]. Several studies have demonstrated that the E-selectin plasma level could be increased in patients with hypertension, type 2 DM, and atherosclerosis, and regard it as one of the risk factors for atherogenesis [6063]. Furthermore, LPS-induced CCL4 production from human monocytes was significantly and positively correlated with the total and LDL cholesterol concentration [64].

In the animal model of myocardial infarction, chemokine induction in the infarct heart mediates recruitment of leukocyte subsets with distinct properties. CCL4 and its receptor CCR5 were significantly induced in the infarct mouse myocardium [65, 66]. CCL4 was also upregulated in vulnerable atherosclerosis plaques and was expressed by T cells in advanced atherosclerotic lesions in stroke patients [67, 68]. Further, elevated serum CCL4 levels have been shown to be an independent predictor of stroke and cardiovascular events in an average follow-up period of 37.2 ± 19.9 months in a cohort of hypertensive patients [23]. Patients with the highest quartile of CCL4 level showed a higher risk of stroke and cardiovascular events [23]. These data support the potential role of CCL4 in atherosclerosis disease and plaque vulnerability. However, the in vivo evidence indicating the direct contribution of CCL4 to vascular and myocardial injury is still lacking.

While similar between the non-diabetic hypertensive patients and the normotensive controls, CCL4 levels were significantly higher in the hypertensive patients with type 2 DM than in the controls. Furthermore, in these patients, CCL4 concentrations could be decreased after treatment with nifedipine, a calcium-channel blocker with antihypertensive and vascular protection effects [69]. It seems that the modulation of CCL4 in some clinical cardiovascular diseases, such as hypertension, is mainly related to the presence of type 2 DM. Although increasing evidence supports the potential role of CCL4, the mechanistic insights and clinical impact of CCL4-related atherosclerosis cardiovascular disease should be further explored.

Chemokine receptor 5 (CCR5)

CCL4 mediates its biological effects by binding to cell surface CC chemokine receptors belonging to the G-protein-coupled receptor super family. The most well-known receptor of CCL4 is the fifth CC chemokine receptor (CCR5). CCR5 was cloned in 1996 [70] and shares 55 % amino acids identity with the first CC chemokine receptor [71]. Recent evidence has suggested that CCR5 is involved in human diseases such as infections and inflammatory diseases [7274]. In this part, we focus on the background of CCR5 and its interactions with CCL4 in DM and cardiovascular diseases.

The role of CCR5 in type 1 and type 2 diabetes mellitus

CCR5 may be linked with DM pathogenesis. In clinical studies, CCR5 gene polymorphisms were differentially related to the phenotypes of type 1 and type 2 DM [75, 76]. Patients with type 2 DM exhibit CCR5 over-expression on their peripheral mononuclear cells, and low-dose infusions of insulin can suppress the expression of CCR5 in mononuclear cells in obese type 2 diabetes patients [32].

In an animal model of experiment diabetes, CCR5 expression in the pancreas was associated with the development of insulitis and spontaneous type 1 DM [46]. CCR5 is also elevated in the superior cervical ganglion of type 2 diabetic rats [77]. Moreover, CCR5 is considered a novel link between obesity, adipose tissue inflammation, and insulin resistance [78, 79]. However, the data were not consistent in different animal models. In two previous studies, transient blockade of CCR5 with an anti-CCR5 mAb at 11–13 weeks of age or CCR5 deficiency significantly accelerated rather than prevented auto-immune type 1 diabetes in NOD mice [49, 80]. Furthermore, CCR5 expression at both the mRNA and protein levels was significantly increased when HUVECs were exposed to chronic high glucose [81].

Accordingly, these findings suggest the diverse roles of CCR5 in the progression of diabetes in experimental DM with different animal models. It should be further clarified if and how modulations of CCR5 could alter the development of human type 1 or type 2 DM.

The role of CCR5 in diabetic renal disease

In clinical studies, CCR5 gene polymorphisms are not only related to the phenotypes [75, 76] but also to the risk of nephropathy in type 1 and type 2 diabetes patients [8286]. During diabetic nephropathy, the production of chemokines, such as RANTES, CCL3, and CCL4, by glomerular and tubular cells has been reported in hyperglycemia [8789]. It has been reported that CCR5 mRNA was upregulated in microdissected glomeruli. In clinical diabetic nephropathy, CCR5 mRNA was overexpressed in the tubulointerstitial compartment but not expressed in microalbuminuria [90]. These chemokines are able to recruit kidney monocytes by interacting with the chemokine receptor CCR5 [91]. Interestingly, the presence of CCR5∆32 is associated with better survival in type 2 diabetic patients [76].

CCR5 in atherosclerosis cardiovascular disease

The upregulation of CCR5 may be associated with the development of atherosclerosis in type 2 diabetic patients [92]. It has been shown that the CCR5∆32 allele could be associated with decreased levels of C-reactive protein, intima-media thickness, and cardiovascular disease risk [9395]. Reduced early onset of coronary heart disease in women is linked with CCR5 [96]. Further, the variation at the CCR5 gene was suggested to modulate the age of onset of myocardial infarction [97]. The CCR5 expression in peripheral monocytes was also increased in obese women [98]. However, another series of clinical investigations found no effects of the CCR5∆32 polymorphism on coronary artery disease or myocardial infarction in other populations [99102]. Recently, it was shown that no chemokine receptor variant was associated with coronary artery disease, myocardial infarction or glucometabolic traits in large European ancestry cohorts [103]. Given the controversy in human studies, several investigations carried out on CCR5 in atherosclerosis mouse models, using receptor antagonism or genetic deletion, indicated that CCR5 may be important in plaque development [93, 104108]. Interestingly, CCR5 was specifically needed for CD4 T cell homing to the atherosclerotic plaques [109].

CCR5 was discovered in smooth muscles and atherosclerotic plaques. Increased CCR5 mRNA expression was discovered in unstable carotid atherosclerotic plaques compared with the stable ones [18, 110]. An animal study revealed the association between upregulated CCR5 and disease progression in a vascular injury model using targeted nanoparticles with PET/CT imaging [111]. CCR5 was also induced in murine myocardial infarct [66]. It may mediate vasoconstriction and stimulate intimal hyperplasia in human vessels in vitro [112]. In addition, 40 % of the mononuclear cells infiltrating the infarct myocardium expressed CCR5 [65]. Furthermore, CCR5 inhibition prevents cardiac dysfunction in the SIV/Macaque Model of HIV [113]. HIV-1 binds to CCR5 as a co-receptor to enter cell macrophages and glial cells [114]. While HIV-1 infection is atherogenic [115], higher monocyte CCR5 expression and plasma IL-6 may be associated with atherosclerosis in HIV-infected individuals [116]. CCR5 antagonists could retard early ritonavir-induced atherogenesis and advanced plaque progression [117]. These findings support the involvement of CCR5, although its function is still not completely known, in the mediation of atherosclerosis and cardiovascular diseases. In 2002, orally bioavailable, synthetic, small molecule antagonists of CCR5 were tested in clinical trials. The CCR5 antagonist was shown to inhibit the proinflammatory effects of CCR5 ligands related to the pathogenesis of inflammatory diseases [118]. It was also reported that the annexin A1 fragment Ac2-26 largely diminished arterial recruitment of myeloid cells in a FPR2-dependent fashion. However, this effect was not abolished in the presence of selective antagonists to CCR5 [119]. The potential role of CCR5 in clinical atherosclerosis disease awaits further clarification.

Conclusion

Recent findings demonstrated that both CCL4 and its receptor CCR5 play diverse roles in the inflammatory events underlying DM and cardiovascular diseases. Though it may attract macrophages to destroy islet cells, CCL4 could play a protective role in some experimental diabetes models, especially the NOD type 1 DM model. CCL4 might be the product rather than an inducer of the initial development of immune-related type 1 diabetes. However, given the specific pathological background in each individual DM disease model, the role of CCL4 should be fully investigated to ensure translation to clinical trial. On the other hand, CCL4 could also be upregulated in atherosclerosis and myocardial infarction to enhance adhesion molecule expression and accelerate the vascular inflammation response. While the findings on CCL4 neutralization may be contradictory, accumulating evidence showed the potential benefits of CCL4 blockade in experimental atherosclerosis disease but not in the development of DM. On the other hand, although the role of CCR5, a receptor of CCL4, may be diverse in different experimental DM models, experimental evidence favored the involvement of CCR5 in the progression of atherosclerosis cardiovascular disease. Accordingly, future experimental and clinical studies are worthwhile to clarify if anti-CCL4 mechanisms, including direct blocking of CCL4 and/or of CCR5, could be a promising therapeutic approach to retard the development of DM, atherosclerosis cardiovascular diseases or both.

Abbreviations

CCL3:

chemokine CC motif ligand 3

CCL4:

chemokine CC motif ligand 4

CCR5:

fifth CC chemokine receptor

DM:

diabetes mellitus

IGF-I:

insulin-like growth factor-I

MIP-1α:

macrophage inflammatory protein-1α

MIP-1β:

macrophage inflammatory protein-1β

NOD mice:

nonobese diabetic mice

References

  1. Rollins BJ. Chemokines. Blood. 1997;90(3):909–28.

    CAS  PubMed  Google Scholar 

  2. Moser B, Loetscher P. Lymphocyte traffic control by chemokines. Nat Immunol. 2001;2(2):123–8.

    Article  CAS  PubMed  Google Scholar 

  3. Fox CS, Coady S, Sorlie PD, D’Agostino RB Sr, Pencina MJ, Vasan RS, Meigs JB, Levy D, Savage PJ. Increasing cardiovascular disease burden due to diabetes mellitus: the Framingham Heart Study. Circulation. 2007;115(12):1544–50.

    Article  PubMed  Google Scholar 

  4. He Z, King GL. Microvascular complications of diabetes. Endocrinol Metab Clin North Am. 2004;33(1):215–38.

    Article  CAS  PubMed  Google Scholar 

  5. Vazzana N, Ranalli P, Cuccurullo C, Davi G. Diabetes mellitus and thrombosis. Thromb Res. 2012;129(3):371–7.

    Article  CAS  PubMed  Google Scholar 

  6. McColl SR, Hachicha M, Levasseur S, Neote K, Schall TJ. Uncoupling of early signal transduction events from effector function in human peripheral blood neutrophils in response to recombinant macrophage inflammatory proteins-1α and -1β. J Immunol. 1993;150(10):4550–60.

    CAS  PubMed  Google Scholar 

  7. Ji Q, Lin Y, Liang Z, Yu K, Liu Y, Fang Z, Liu L, Shi Y, Zeng Q, Chang C, et al. Chemerin is a novel biomarker of acute coronary syndrome but not of stable angina pectoris. Cardiovasc Diabetol. 2014;13:145.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  8. Proost P, Menten P, Struyf S, Schutyser E, De Meester I, Van Damme J. Cleavage by CD26/dipeptidyl peptidase IV converts the chemokine LD78β into a most efficient monocyte attractant and CCR1 agonist. Blood. 2000;96(5):1674–80.

    CAS  PubMed  Google Scholar 

  9. Menten P, Wuyts A, Van Damme J. Macrophage inflammatory protein-1. Cytokine Growth Factor Rev. 2002;13(6):455–81.

    Article  CAS  PubMed  Google Scholar 

  10. Lodi PJ, Garrett DS, Kuszewski J, Tsang ML, Weatherbee JA, Leonard WJ, Gronenborn AM, Clore GM. High-resolution solution structure of the beta chemokine hMIP-1β by multidimensional NMR. Science. 1994;263(5154):1762–7.

    Article  CAS  PubMed  Google Scholar 

  11. Schall TJ, Bacon KB. Chemokines, leukocyte trafficking, and inflammation. Curr Opin Immunol. 1994;6(6):865–73.

    Article  CAS  PubMed  Google Scholar 

  12. House LM 2nd, Morris RT, Barnes TM, Lantier L, Cyphert TJ, McGuinness OP, Otero YF. Tissue inflammation and nitric oxide-mediated alterations in cardiovascular function are major determinants of endotoxin-induced insulin resistance. Cardiovasc Diabetol. 2015;14:56.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  13. Maurer M, von Stebut E. Macrophage inflammatory protein-1. Int J Biochem Cell Biol. 2004;36(10):1882–6.

    Article  CAS  PubMed  Google Scholar 

  14. Loetscher P, Seitz M, Clark-Lewis I, Baggiolini M, Moser B. Activation of NK cells by CC chemokines. Chemotaxis, Ca2+ mobilization, and enzyme release. J Immunol. 1996;156(1):322–7.

    CAS  PubMed  Google Scholar 

  15. Taub DD, Sayers TJ, Carter CR, Ortaldo JR. Alpha and beta chemokines induce NK cell migration and enhance NK-mediated cytolysis. J Immunol. 1995;155(8):3877–88.

    CAS  PubMed  Google Scholar 

  16. Menten P, Struyf S, Schutyser E, Wuyts A, De Clercq E, Schols D, Proost P, Van Damme J. The LD78β isoform of MIP-1α is the most potent CCR5 agonist and HIV-1-inhibiting chemokine. J Clin Invest. 1999;104(4):R1–5.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Berger O, Gan X, Gujuluva C, Burns AR, Sulur G, Stins M, Way D, Witte M, Weinand M, Said J, et al. CXC and CC chemokine receptors on coronary and brain endothelia. Mol Med. 1999;5(12):795–805.

    CAS  PubMed  PubMed Central  Google Scholar 

  18. Schecter AD, Calderon TM, Berman AB, McManus CM, Fallon JT, Rossikhina M, Zhao W, Christ G, Berman JW, Taubman MB. Human vascular smooth muscle cells possess functional CCR5. J Biol Chem. 2000;275(8):5466–71.

    Article  CAS  PubMed  Google Scholar 

  19. Honczarenko M, Le Y, Glodek AM, Majka M, Campbell JJ, Ratajczak MZ, Silberstein LE. CCR5-binding chemokines modulate CXCL12 (SDF-1)-induced responses of progenitor B cells in human bone marrow through heterologous desensitization of the CXCR4 chemokine receptor. Blood. 2002;100(7):2321–9.

    Article  CAS  PubMed  Google Scholar 

  20. Hanifi-Moghaddam P, Kappler S, Seissler J, Muller-Scholze S, Martin S, Roep BO, Strassburger K, Kolb H, Schloot NC. Altered chemokine levels in individuals at risk of type 1 diabetes mellitus. Diab Med. 2006;23(2):156–63.

    Article  CAS  Google Scholar 

  21. Maier R, Weger M, Haller-Schober EM, El-Shabrawi Y, Wedrich A, Theisl A, Aigner R, Barth A, Haas A. Multiplex bead analysis of vitreous and serum concentrations of inflammatory and proangiogenic factors in diabetic patients. Mol Vis. 2008;14:637–43.

    CAS  PubMed  PubMed Central  Google Scholar 

  22. Ryden A, Faresjo M. Altered immune profile from pre-diabetes to manifestation of type 1 diabetes. Diab Res Clin Pract. 2013;100(1):74–84.

    Article  CAS  Google Scholar 

  23. Tatara Y, Ohishi M, Yamamoto K, Shiota A, Hayashi N, Iwamoto Y, Takeda M, Takagi T, Katsuya T, Ogihara T, et al. Macrophage inflammatory protein-1β induced cell adhesion with increased intracellular reactive oxygen species. J Mol Cell Cardiol. 2009;47(1):104–11.

    Article  CAS  PubMed  Google Scholar 

  24. Mirabelli-Badenier M, Braunersreuther V, Viviani GL, Dallegri F, Quercioli A, Veneselli E, Mach F, Montecucco F. CC and CXC chemokines are pivotal mediators of cerebral injury in ischaemic stroke. Thromb Haemost. 2011;105(3):409–20.

    Article  CAS  PubMed  Google Scholar 

  25. DeVries ME, Ran L, Kelvin DJ. On the edge: the physiological and pathophysiological role of chemokines during inflammatory and immunological responses. Semin Immunol. 1999;11(2):95–104.

    Article  CAS  PubMed  Google Scholar 

  26. Benoist C, Mathis D. Cell death mediators in autoimmune diabetes–no shortage of suspects. Cell. 1997;89(1):1–3.

    Article  CAS  PubMed  Google Scholar 

  27. Barbe-Tuana FM, Klein D, Ichii H, Berman DM, Coffey L, Kenyon NS, Ricordi C, Pastori RL. CD40-CD40 ligand interaction activates proinflammatory pathways in pancreatic islets. Diabetes. 2006;55(9):2437–45.

    Article  CAS  PubMed  Google Scholar 

  28. Mita A, Ricordi C, Miki A, Barker S, Haertter R, Hashikura Y, Miyagawa S, Burke GW 3rd, Inverardi L, Ichii H. Anti-proinflammatory effects of sirolimus on human islet preparations. Transplantation. 2008;86(1):46–53.

    Article  PubMed  PubMed Central  Google Scholar 

  29. Pham MN, Hawa MI, Roden M, Schernthaner G, Pozzilli P, Buzzetti R, Scherbaum WA, Seissler J, Hunter S, Leslie RD, et al. Increased serum concentrations of adhesion molecules but not of chemokines in patients with type 2 diabetes compared with patients with type 1 diabetes and latent autoimmune diabetes in adult age: action LADA 5. Diab Med. 2012;29(4):470–8.

    Article  CAS  Google Scholar 

  30. Purohit S, Sharma A, Hopkins D, Steed L, Bode B, Anderson SW, Reed JC, Steed RD, Yang T, She JX. Large-scale discovery and validation studies demonstrate significant reductions in circulating levels of IL8, IL-1Ra, MCP-1, and MIP-1β in patients with type 1 diabetes. J Clin Endocrinol Metab. 2015;100(9):E1179–87.

    Article  PubMed  Google Scholar 

  31. Neumeier M, Bauer S, Bruhl H, Eisinger K, Kopp A, Abke S, Walter R, Schaffler A, Buechler C. Adiponectin stimulates release of CCL2, -3, -4 and -5 while the surface abundance of CCR2 and -5 is simultaneously reduced in primary human monocytes. Cytokine. 2011;56(3):573–80.

    Article  CAS  PubMed  Google Scholar 

  32. Dandona P, Aljada A, Mohanty P, Ghanim H, Hamouda W, Assian E, Ahmad S. Insulin inhibits intranuclear nuclear factor κB and stimulates IκB in mononuclear cells in obese subjects: evidence for an anti-inflammatory effect? J Clin Endocrinol Metab. 2001;86(7):3257–65.

    CAS  PubMed  Google Scholar 

  33. Aljada A, Ghanim H, Saadeh R, Dandona P. Insulin inhibits NFκB and MCP-1 expression in human aortic endothelial cells. J Clin Endocrinol Metab. 2001;86(1):450–3.

    CAS  PubMed  Google Scholar 

  34. Ghanim H, Korzeniewski K, Sia CL, Abuaysheh S, Lohano T, Chaudhuri A, Dandona P. Suppressive effect of insulin infusion on chemokines and chemokine receptors. Diab Care. 2010;33(5):1103–8.

    Article  CAS  Google Scholar 

  35. Galkina EV, Butcher M, Keller SR, Goff M, Bruce A, Pei H, Sarembock IJ, Sanders JM, Nagelin MH, Srinivasan S, et al. Accelerated atherosclerosis in Apoe-/- mice heterozygous for the insulin receptor and the insulin receptor substrate-1. Arterioscler Thromb Vasc Biol. 2012;32(2):247–56.

    Article  CAS  PubMed  Google Scholar 

  36. Shamir R, Shehadeh N, Rosenblat M, Eshach-Adiv O, Coleman R, Kaplan M, Hamoud S, Lischinsky S, Hayek T. Oral insulin supplementation attenuates atherosclerosis progression in apolipoprotein E-deficient mice. Arterioscler Thromb Vasc Biol. 2003;23(1):104–10.

    Article  CAS  PubMed  Google Scholar 

  37. Pfleger C, Kaas A, Hansen L, Alizadeh B, Hougaard P, Holl R, Kolb H, Roep BO, Mortensen HB, Schloot NC. Relation of circulating concentrations of chemokine receptor CCR5 ligands to C-peptide, proinsulin and HbA1c and disease progression in type 1 diabetes. Clin Immunol. 2008;128(1):57–65.

    Article  CAS  PubMed  Google Scholar 

  38. Delovitch TL, Singh B. The nonobese diabetic mouse as a model of autoimmune diabetes: immune dysregulation gets the NOD. Immunity. 1997;7(6):727–38.

    Article  CAS  PubMed  Google Scholar 

  39. Bradley LM, Asensio VC, Schioetz LK, Harbertson J, Krahl T, Patstone G, Woolf N, Campbell IL, Sarvetnick N. Islet-specific Th1, but not Th2, cells secrete multiple chemokines and promote rapid induction of autoimmune diabetes. J Immunol. 1999;162(5):2511–20.

    CAS  PubMed  Google Scholar 

  40. Cameron MJ, Arreaza GA, Waldhauser L, Gauldie J, Delovitch TL. Immunotherapy of spontaneous type 1 diabetes in nonobese diabetic mice by systemic interleukin-4 treatment employing adenovirus vector-mediated gene transfer. Gene Ther. 2000;7(21):1840–6.

    Article  CAS  PubMed  Google Scholar 

  41. Goudy KS, Burkhardt BR, Wasserfall C, Song S, Campbell-Thompson ML, Brusko T, Powers MA, Clare-Salzler MJ, Sobel ES, Ellis TM, et al. Systemic overexpression of IL-10 induces CD4+ CD25+ cell populations in vivo and ameliorates type 1 diabetes in nonobese diabetic mice in a dose-dependent fashion. J Immunol. 2003;171(5):2270–8.

    Article  CAS  PubMed  Google Scholar 

  42. Fujihira K, Nagata M, Moriyama H, Yasuda H, Arisawa K, Nakayama M, Maeda S, Kasuga M, Okumura K, Yagita H, et al. Suppression and acceleration of autoimmune diabetes by neutralization of endogenous interleukin-12 in NOD mice. Diabetes. 2000;49(12):1998–2006.

    Article  CAS  PubMed  Google Scholar 

  43. Poulin M, Haskins K. Induction of diabetes in nonobese diabetic mice by Th2 T cell clones from a TCR transgenic mouse. J Immunol. 2000;164(6):3072–8.

    Article  CAS  PubMed  Google Scholar 

  44. O’Shea JJ, Ma A, Lipsky P. Cytokines and autoimmunity. Nat Rev Immunol. 2002;2(1):37–45.

    Article  PubMed  CAS  Google Scholar 

  45. Patterson SJ, Pesenacker AM, Wang AY, Gillies J, Mojibian M, Morishita K, Tan R, Kieffer TJ, Verchere CB, Panagiotopoulos C, et al. T regulatory cell chemokine production mediates pathogenic T cell attraction and suppression. J Clin Invest. 2016;126(3):1039–51.

    Article  PubMed  PubMed Central  Google Scholar 

  46. Cameron MJ, Arreaza GA, Grattan M, Meagher C, Sharif S, Burdick MD, Strieter RM, Cook DN, Delovitch TL. Differential expression of CC chemokines and the CCR5 receptor in the pancreas is associated with progression to type I diabetes. J Immunol. 2000;165(2):1102–10.

    Article  CAS  PubMed  Google Scholar 

  47. Youssef S, Wildbaum G, Maor G, Lanir N, Gour-Lavie A, Grabie N, Karin N. Long-lasting protective immunity to experimental autoimmune encephalomyelitis following vaccination with naked DNA encoding C–C chemokines. J Immunol. 1998;161(8):3870–9.

    CAS  PubMed  Google Scholar 

  48. Bergerot I, Arreaza GA, Cameron MJ, Burdick MD, Strieter RM, Chensue SW, Chakrabarti S, Delovitch TL. Insulin B-chain reactive CD4+ regulatory T-cells induced by oral insulin treatment protect from type 1 diabetes by blocking the cytokine secretion and pancreatic infiltration of diabetogenic effector T-cells. Diabetes. 1999;48(9):1720–9.

    Article  CAS  PubMed  Google Scholar 

  49. Meagher C, Beilke J, Arreaza G, Mi QS, Chen W, Salojin K, Horst N, Cruikshank WW, Delovitch TL. Neutralization of interleukin-16 protects nonobese diabetic mice from autoimmune type 1 diabetes by a CCL4-dependent mechanism. Diabetes. 2010;59(11):2862–71.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Chen W, Salojin KV, Mi QS, Grattan M, Meagher TC, Zucker P, Delovitch TL. Insulin-like growth factor (IGF)-I/IGF-binding protein-3 complex: therapeutic efficacy and mechanism of protection against type 1 diabetes. Endocrinology. 2004;145(2):627–38.

    Article  CAS  PubMed  Google Scholar 

  51. Meagher C, Arreaza G, Peters A, Strathdee CA, Gilbert PA, Mi QS, Santamaria P, Dekaban GA, Delovitch TL. CCL4 protects from type 1 diabetes by altering islet beta-cell-targeted inflammatory responses. Diabetes. 2007;56(3):809–17.

    Article  CAS  PubMed  Google Scholar 

  52. King AJF. The use of animal models in diabetes research. Br J Pharmacol. 2012;166(3):877–94.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Shoda LKM, Young DL, Ramanujan S, Whiting CC, Atkinson MA, Bluestone JA, Eisenbarth GS, Mathis D, Rossini AA, Campbell SE, et al. A comprehensive review of interventions in the NOD mouse and implications for translation. Immunity. 2005;23(2):115–26.

    Article  CAS  PubMed  Google Scholar 

  54. Yang XD, Tisch R, Singer SM, Cao ZA, Liblau RS, Schreiber RD, McDevitt HO. Effect of tumor necrosis factor α on insulin-dependent diabetes mellitus in NOD mice. I. The early development of autoimmunity and the diabetogenic process. J Exp Med. 1994;180(3):995–1004.

    Article  CAS  PubMed  Google Scholar 

  55. Yamada K, Nonaka K, Hanafusa T, Miyazaki A, Toyoshima H, Tarui S. Preventive and therapeutic effects of large-dose nicotinamide injections on diabetes associated with insulitis. An observation in nonobese diabetic (NOD) mice. Diabetes. 1982;31(9):749–53.

    Article  CAS  PubMed  Google Scholar 

  56. Zhang ZJ, Davidson L, Eisenbarth G, Weiner HL. Suppression of diabetes in nonobese diabetic mice by oral administration of porcine insulin. Proc Natl Acad Sci USA. 1991;88(22):10252–6.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  57. Hermitte L, Vialettes B, Atlef N, Payan MJ, Doll N, Scheimann A, Vague P. High dose nicotinamide fails to prevent diabetes in BB rats. Autoimmunity. 1989;5(1–2):79–86.

    Article  CAS  PubMed  Google Scholar 

  58. Mordes JP, Schirf B, Roipko D, Greiner DL, Weiner H, Nelson P, Rossini AA. Oral insulin does not prevent insulin-dependent diabetes mellitus in BB rats. Ann NY Acad Sci. 1996;778:418–21.

    Article  CAS  PubMed  Google Scholar 

  59. Chen TC, Chien SJ, Kuo HC, Huang WS, Sheen JM, Lin TH, Yen CK, Sung ML, Chen CN. High glucose-treated macrophages augment E-selectin expression in endothelial cells. J Biol Chem. 2011;286(29):25564–73.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  60. Boulbou MS, Koukoulis GN, Makri ED, Petinaki EA, Gourgoulianis KI, Germenis AE. Circulating adhesion molecules levels in type 2 diabetes mellitus and hypertension. Int J Cardiol. 2005;98(1):39–44.

    Article  PubMed  Google Scholar 

  61. Hwang SJ, Ballantyne CM, Sharrett AR, Smith LC, Davis CE, Gotto AM Jr, Boerwinkle E. Circulating adhesion molecules VCAM-1, ICAM-1, and E-selectin in carotid atherosclerosis and incident coronary heart disease cases: the atherosclerosis risk in communities (ARIC) study. Circulation. 1997;96(12):4219–25.

    Article  CAS  PubMed  Google Scholar 

  62. De Caterina R, Ghiadoni L, Taddei S, Virdis A, Almerigogna F, Basta G, Lazzerini G, Bernini W, Salvetti A. Soluble E-selectin in essential hypertension: a correlate of vascular structural changes. Am J Hypertens. 2001;14(3):259–66.

    Article  PubMed  Google Scholar 

  63. Matsumoto K, Sera Y, Nakamura H, Ueki Y, Miyake S. Serum concentrations of soluble adhesion molecules are related to degree of hyperglycemia and insulin resistance in patients with type 2 diabetes mellitus. Diab Res Clin Pract. 2002;55(2):131–8.

    Article  CAS  Google Scholar 

  64. Bala M, Kopp A, Wurm S, Buchler C, Scholmerich J, Schaffler A. Type 2 diabetes and lipoprotein metabolism affect LPS-induced cytokine and chemokine release in primary human monocytes. Exp Clin Endocrinol Diab. 2011;119(6):370–6.

    Article  CAS  Google Scholar 

  65. Dobaczewski M, Xia Y, Bujak M, Gonzalez-Quesada C, Frangogiannis NG. CCR5 signaling suppresses inflammation and reduces adverse remodeling of the infarcted heart, mediating recruitment of regulatory T cells. Am J Pathol. 2010;176(5):2177–87.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  66. Dewald O, Ren G, Duerr GD, Zoerlein M, Klemm C, Gersch C, Tincey S, Michael LH, Entman ML, Frangogiannis NG. Of mice and dogs: species-specific differences in the inflammatory response following myocardial infarction. Am J Pathol. 2004;164(2):665–77.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  67. Reape TJ, Groot PH. Chemokines and atherosclerosis. Atherosclerosis. 1999;147(2):213–25.

    Article  CAS  PubMed  Google Scholar 

  68. Montecucco F, Lenglet S, Gayet-Ageron A, Bertolotto M, Pelli G, Palombo D, Pane B, Spinella G, Steffens S, Raffaghello L, et al. Systemic and intraplaque mediators of inflammation are increased in patients symptomatic for ischemic stroke. Stroke. 2010;41(7):1394–404.

    Article  PubMed  Google Scholar 

  69. Nomura S, Shouzu A, Omoto S, Nishikawa M, Iwasaka T. Long-term treatment with nifedipine modulates procoagulant marker and C–C chemokine in hypertensive patients with type 2 diabetes mellitus. Thromb Res. 2005;115(4):277–85.

    Article  CAS  PubMed  Google Scholar 

  70. Samson M, Labbe O, Mollereau C, Vassart G, Parmentier M. Molecular cloning and functional expression of a new human CC-chemokine receptor gene. Biochemistry. 1996;35(11):3362–7.

    Article  CAS  PubMed  Google Scholar 

  71. Murphy PM, Baggiolini M, Charo IF, Hebert CA, Horuk R, Matsushima K, Miller LH, Oppenheim JJ, Power CA. International union of pharmacology. XXII. Nomenclature for chemokine receptors. Pharmacol Rev. 2000;52(1):145–76.

    CAS  PubMed  Google Scholar 

  72. Locati M, Bonecchi R, Corsi MM. Chemokines and their receptors: roles in specific clinical conditions and measurement in the clinical laboratory. Am J Clin Pathol. 2005;123(Suppl):S82–95.

    CAS  PubMed  Google Scholar 

  73. Balistreri CR, Caruso C, Grimaldi MP, Listi F, Vasto S, Orlando V, Campagna AM, Lio D, Candore G. CCR5 receptor: biologic and genetic implications in age-related diseases. Ann NY Acad Sci. 2007;1100:162–72.

    Article  CAS  PubMed  Google Scholar 

  74. Jones KL, Maguire JJ, Davenport AP. Chemokine receptor CCR5: from AIDS to atherosclerosis. Br J Pharmacol. 2011;162(7):1453–69.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  75. Kalev I, Oselin K, Parlist P, Zilmer M, Rajasalu T, Podar T, Mikelsaar AV. CC-chemokine receptor CCR5-del32 mutation as a modifying pathogenetic factor in type I diabetes. J Diab Complicat. 2003;17(6):387–91.

    Article  Google Scholar 

  76. Muntinghe FL, Gross S, Bakker SJ, Landman GW, van der Harst P, Bilo HJ, Navis G, Zuurman MW. CCR5∆32 genotype is associated with outcome in type 2 diabetes mellitus. Diab Res Clin Pract. 2009;86(2):140–5.

    Article  CAS  Google Scholar 

  77. Li G, Xu H, Zhu S, Xu W, Qin S, Liu S, Tu G, Peng H, Qiu S, Yu S, et al. Effects of neferine on CCL5 and CCR5 expression in SCG of type 2 diabetic rats. Brain Res Bull. 2013;90:79–87.

    Article  CAS  PubMed  Google Scholar 

  78. Ota T. CCR5: a novel player in the adipose tissue inflammation and insulin resistance? Adipocyte. 2013;2(2):99–103.

    Article  PubMed  PubMed Central  Google Scholar 

  79. Kitade H, Sawamoto K, Nagashimada M, Inoue H, Yamamoto Y, Sai Y, Takamura T, Yamamoto H, Miyamoto K, Ginsberg HN, et al. CCR5 plays a critical role in obesity-induced adipose tissue inflammation and insulin resistance by regulating both macrophage recruitment and M1/M2 status. Diabetes. 2012;61(7):1680–90.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  80. Solomon M, Balasa B, Sarvetnick N. CCR2 and CCR5 chemokine receptors differentially influence the development of autoimmune diabetes in the NOD mouse. Autoimmunity. 2010;43(2):156–63.

    Article  CAS  PubMed  Google Scholar 

  81. Li G, Zhu G, Gao Y, Xiao W, Xu H, Liu S, Tu G, Peng H, Zheng C, Liang S. Neferine inhibits the upregulation of CCL5 and CCR5 in vascular endothelial cells during chronic high glucose treatment. Inflammation. 2013;36(2):300–8.

    Article  CAS  PubMed  Google Scholar 

  82. Yang B, Houlberg K, Millward A, Demaine A. Polymorphisms of chemokine and chemokine receptor genes in type 1 diabetes mellitus and its complications. Cytokine. 2004;26(3):114–21.

    Article  CAS  PubMed  Google Scholar 

  83. Mlynarski WM, Placha GP, Wolkow PP, Bochenski JP, Warram JH, Krolewski AS. Risk of diabetic nephropathy in type 1 diabetes is associated with functional polymorphisms in RANTES receptor gene (CCR5): a sex-specific effect. Diabetes. 2005;54(11):3331–5.

    Article  CAS  PubMed  Google Scholar 

  84. Nakajima K, Tanaka Y, Nomiyama T, Ogihara T, Ikeda F, Kanno R, Iwashita N, Sakai K, Watada H, Onuma T, et al. RANTES promoter genotype is associated with diabetic nephropathy in type 2 diabetic subjects. Diab Care. 2003;26(3):892–8.

    Article  CAS  Google Scholar 

  85. Ahluwalia TS, Khullar M, Ahuja M, Kohli HS, Bhansali A, Mohan V, Venkatesan R, Rai TS, Sud K, Singal PK. Common variants of inflammatory cytokine genes are associated with risk of nephropathy in type 2 diabetes among Asian Indians. PLoS One. 2009;4(4):e5168.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  86. Buraczynska M, Zukowski P, Wacinski P, Berger-Smyka B, Dragan M, Mozul S. Chemotactic cytokine receptor 5 gene polymorphism: relevance to microvascular complications in type 2 diabetes. Cytokine. 2012;58(2):213–7.

    Article  CAS  PubMed  Google Scholar 

  87. Vielhauer V, Anders HJ, Mack M, Cihak J, Strutz F, Stangassinger M, Luckow B, Grone HJ, Schlondorff D. Obstructive nephropathy in the mouse: progressive fibrosis correlates with tubulointerstitial chemokine expression and accumulation of CC chemokine receptor 2- and 5-positive leukocytes. J Am Soc Nephrol. 2001;12(6):1173–87.

    CAS  PubMed  Google Scholar 

  88. Benigni A, Remuzzi G. How renal cytokines and growth factors contribute to renal disease progression. Am J Kidney Dis. 2001;37(1 Suppl 2):S21–4.

    Article  CAS  PubMed  Google Scholar 

  89. Mezzano S, Droguett A, Burgos ME, Ardiles LG, Flores CA, Aros CA, Caorsi I, Vio CP, Ruiz-Ortega M, Egido J. Renin-angiotensin system activation and interstitial inflammation in human diabetic nephropathy. Kidney Int Suppl. 2003;86:S64–70.

    Article  CAS  PubMed  Google Scholar 

  90. Verzola D, Cappuccino L, D’Amato E, Villaggio B, Gianiorio F, Mij M, Simonato A, Viazzi F, Salvidio G, Garibotto G. Enhanced glomerular toll-like receptor 4 expression and signaling in patients with type 2 diabetic nephropathy and microalbuminuria. Kidney Int. 2014;86(6):1229–43.

    Article  CAS  PubMed  Google Scholar 

  91. Mantovani A. The chemokine system: redundancy for robust outputs. Immunol Today. 1999;20(6):254–7.

    Article  CAS  PubMed  Google Scholar 

  92. Dytfeld J, Bogdanski P, Pupek-Musialik D, Jagodzinski PP, Bryl W, Kujawa A. Expression of chemokine receptor CCR5 in patients with type 2 diabetes. Pol Merkur Lekarski. 2006;20(116):195–8.

    CAS  PubMed  Google Scholar 

  93. Tacke F, Alvarez D, Kaplan TJ, Jakubzick C, Spanbroek R, Llodra J, Garin A, Liu J, Mack M, van Rooijen N, et al. Monocyte subsets differentially employ CCR2, CCR5, and CX3CR1 to accumulate within atherosclerotic plaques. J Clin Invest. 2007;117(1):185–94.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  94. Afzal AR, Kiechl S, Daryani YP, Weerasinghe A, Zhang Y, Reindl M, Mayr A, Weger S, Xu Q, Willeit J. Common CCR5-del32 frameshift mutation associated with serum levels of inflammatory markers and cardiovascular disease risk in the Bruneck population. Stroke. 2008;39(7):1972–8.

    Article  CAS  PubMed  Google Scholar 

  95. Dinh KM, Pedersen OB, Petersen MS, Sorensen E, Sorensen CJ, Kaspersen KA, Larsen MH, Moller B, Hjalgrim H, Ullum H, et al. The impact of CCR5-∆32 deletion on C-reactive protein levels and cardiovascular disease: results from the Danish Blood Donor Study. Atherosclerosis. 2015;242(1):222–5.

    Article  CAS  PubMed  Google Scholar 

  96. Pai JK, Kraft P, Cannuscio CC, Manson JE, Rexrode KM, Albert CM, Hunter D, Rimm EB. Polymorphisms in the CC-chemokine receptor-2 (CCR2) and -5 (CCR5) genes and risk of coronary heart disease among US women. Atherosclerosis. 2006;186(1):132–9.

    Article  CAS  PubMed  Google Scholar 

  97. Gonzalez P, Alvarez R, Batalla A, Reguero JR, Alvarez V, Astudillo A, Cubero GI, Cortina A, Coto E. Genetic variation at the chemokine receptors CCR5/CCR2 in myocardial infarction. Genes Immun. 2001;2(4):191–5.

    Article  CAS  PubMed  Google Scholar 

  98. Krinninger P, Ensenauer R, Ehlers K, Rauh K, Stoll J, Krauss-Etschmann S, Hauner H, Laumen H. Peripheral monocytes of obese women display increased chemokine receptor expression and migration capacity. J Clin Endocrinol Metab. 2014;99(7):2500–9.

    Article  CAS  PubMed  Google Scholar 

  99. Simeoni E, Winkelmann BR, Hoffmann MM, Fleury S, Ruiz J, Kappenberger L, Marz W, Vassalli G. Association of RANTES G-403A gene polymorphism with increased risk of coronary arteriosclerosis. Eur Heart J. 2004;25(16):1438–46.

    Article  CAS  PubMed  Google Scholar 

  100. Petrkova J, Cermakova Z, Lukl J, Petrek M. CC chemokine receptor 5 (CCR5) deletion polymorphism does not protect Czech males against early myocardial infarction. J Intern Med. 2005;257(6):564–6.

    Article  CAS  PubMed  Google Scholar 

  101. Apostolakis S, Baritaki S, Kochiadakis GE, Igoumenidis NE, Panutsopulos D, Spandidos DA. Effects of polymorphisms in chemokine ligands and receptors on susceptibility to coronary artery disease. Thromb Res. 2007;119(1):63–71.

    Article  CAS  PubMed  Google Scholar 

  102. Sharda S, Gilmour A, Harris V, Singh VP, Sinha N, Tewari S, Ramesh V, Agrawal S, Mastana S. Chemokine receptor 5 (CCR5) deletion polymorphism in North Indian patients with coronary artery disease. Int J Cardiol. 2008;124(2):254–8.

    Article  CAS  PubMed  Google Scholar 

  103. Golbus JR, Stitziel NO, Zhao W, Xue C, Farrall M, McPherson R, Erdmann J, Deloukas P, Watkins H, Schunkert H, et al. Common and rare genetic variation in CCR2, CCR5, or CX3CR1 and risk of atherosclerotic coronary heart disease and glucometabolic traits. Circ Cardiovasc Genet. 2016;9(3):250–8.

    Article  CAS  PubMed  Google Scholar 

  104. Schober A, Manka D, von Hundelshausen P, Huo Y, Hanrath P, Sarembock IJ, Ley K, Weber C. Deposition of platelet RANTES triggering monocyte recruitment requires P-selectin and is involved in neointima formation after arterial injury. Circulation. 2002;106(12):1523–9.

    Article  CAS  PubMed  Google Scholar 

  105. Veillard NR, Kwak B, Pelli G, Mulhaupt F, James RW, Proudfoot AE, Mach F. Antagonism of RANTES receptors reduces atherosclerotic plaque formation in mice. Circ Res. 2004;94(2):253–61.

    Article  CAS  PubMed  Google Scholar 

  106. Kuziel WA, Dawson TC, Quinones M, Garavito E, Chenaux G, Ahuja SS, Reddick RL, Maeda N. CCR5 deficiency is not protective in the early stages of atherogenesis in apoE knockout mice. Atherosclerosis. 2003;167(1):25–32.

    Article  CAS  PubMed  Google Scholar 

  107. Braunersreuther V, Zernecke A, Arnaud C, Liehn EA, Steffens S, Shagdarsuren E, Bidzhekov K, Burger F, Pelli G, Luckow B, et al. Ccr5 but not Ccr1 deficiency reduces development of diet-induced atherosclerosis in mice. Arterioscler Thromb Vasc Biol. 2007;27(2):373–9.

    Article  CAS  PubMed  Google Scholar 

  108. Quinones MP, Martinez HG, Jimenez F, Estrada CA, Dudley M, Willmon O, Kulkarni H, Reddick RL, Fernandes G, Kuziel WA, et al. CC chemokine receptor 5 influences late-stage atherosclerosis. Atherosclerosis. 2007;195(1):e92–103.

    Article  CAS  PubMed  Google Scholar 

  109. Li J, McArdle S, Gholami A, Kimura T, Wolf D, Gerhardt T, Miller J, Weber C, Ley K. CCR5+ T-bet + FoxP3 + effector CD4 T cells drive atherosclerosis. Circ Res. 2016;118(10):1540–52.

    Article  CAS  PubMed  Google Scholar 

  110. Papaspyridonos M, Smith A, Burnand KG, Taylor P, Padayachee S, Suckling KE, James CH, Greaves DR, Patel L. Novel candidate genes in unstable areas of human atherosclerotic plaques. Arterioscler Thromb Vasc Biol. 2006;26(8):1837–44.

    Article  CAS  PubMed  Google Scholar 

  111. Luehmann HP, Pressly ED, Detering L, Wang C, Pierce R, Woodard PK, Gropler RJ, Hawker CJ, Liu Y. PET/CT imaging of chemokine receptor CCR5 in vascular injury model using targeted nanoparticle. J Nucl Med. 2014;55(4):629–34.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  112. Maguire JJ, Jones KL, Kuc RE, Clarke MC, Bennett MR, Davenport AP. The CCR5 chemokine receptor mediates vasoconstriction and stimulates intimal hyperplasia in human vessels in vitro. Cardiovasc Res. 2014;101(3):513–21.

    Article  CAS  PubMed  Google Scholar 

  113. Kelly KM, Tocchetti CG, Lyashkov A, Tarwater PM, Bedja D, Graham DR, Beck SE, Metcalf Pate KA, Queen SE, Adams RJ, et al. CCR5 inhibition prevents cardiac dysfunction in the SIV/macaque model of HIV. J Am Heart Assoc. 2014;3(2):e000874.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  114. He J, Chen Y, Farzan M, Choe H, Ohagen A, Gartner S, Busciglio J, Yang X, Hofmann W, Newman W, et al. CCR3 and CCR5 are co-receptors for HIV-1 infection of microglia. Nature. 1997;385(6617):645–9.

    Article  CAS  PubMed  Google Scholar 

  115. Crowe SM, Westhorpe CL, Mukhamedova N, Jaworowski A, Sviridov D, Bukrinsky M. The macrophage: the intersection between HIV infection and atherosclerosis. J Leukoc Biol. 2010;87(4):589–98.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  116. Hsu DC, Ma YF, Hur S, Li D, Rupert A, Scherzer R, Kalapus SC, Deeks S, Sereti I, Hsue PY. Plasma IL-6 levels are independently associated with atherosclerosis and mortality in HIV-infected individuals on suppressive ART. AIDS. 2016;30(13):2065–74.

  117. Cipriani S, Francisci D, Mencarelli A, Renga B, Schiaroli E, D’Amore C, Baldelli F, Fiorucci S. Efficacy of the CCR5 antagonist maraviroc in reducing early, ritonavir-induced atherogenesis and advanced plaque progression in mice. Circulation. 2013;127(21):2114–24.

    Article  CAS  PubMed  Google Scholar 

  118. Carter PH. Chemokine receptor antagonism as an approach to anti-inflammatory therapy: ‘just right’ or plain wrong? Curr Opin Chem Biol. 2002;6(4):510–25.

    Article  CAS  PubMed  Google Scholar 

  119. Drechsler M, de Jong R, Rossaint J, Viola JR, Leoni G, Wang JM, Grommes J, Hinkel R, Kupatt C, Weber C, et al. Annexin A1 counteracts chemokine-induced arterial myeloid cell recruitment. Circ Res. 2015;116(5):827–35.

    Article  CAS  PubMed  Google Scholar 

Download references

Authors’ contributions

TTC contributed to study conception and design, literature review and preparation of the manuscript. JWC contributed to study conception and design, drafted the manuscript and revised it critically for important intellectual content, and gave final approval of the version to be sent. Both authors read and approved the final manuscript.

Competing interests

The authors declare that they have no competing interests.

Funding

This work was supported by Taipei Veterans General Hospital (V101E1-013, V102C-155, V102E1-006, and V103C-151 to JW Chen), Taiwan’s National Science Council (NSC 101-2314-B-010-058-MY3 to JW Chen), and the UST-UCSD International Center of Excellence in Advanced Bio-engineering sponsored by the Taiwan National Science Council I-RiCE Program (NSC-99-2911-I-009-101 to JW Chen). The funding body has no role in the design of the study and collection, analysis, and interpretation of data and in writing the manuscript.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Jaw-Wen Chen.

Rights and permissions

Open Access This article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://creativecommons.org/licenses/by/4.0/), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made. 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.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Chang, TT., Chen, JW. Emerging role of chemokine CC motif ligand 4 related mechanisms in diabetes mellitus and cardiovascular disease: friends or foes?. Cardiovasc Diabetol 15, 117 (2016). https://doi.org/10.1186/s12933-016-0439-9

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s12933-016-0439-9

Keywords