DOI QR코드

DOI QR Code

Role of vascular smooth muscle cell in the inflammation of atherosclerosis

  • Lim, Soyeon (Severance Integrative Research Institute for Cerebral & Cardiovascular Diseases, Yonsei University College of Medicine) ;
  • Park, Sungha (Severance Integrative Research Institute for Cerebral & Cardiovascular Diseases, Yonsei University College of Medicine)
  • Received : 2013.12.16
  • Published : 2014.01.31

Abstract

Atherosclerosis is a pathologic process occurring within the artery, in which many cell types, including T cell, macrophages, endothelial cells, and smooth muscle cells, interact, and cause chronic inflammation, in response to various inner- or outer-cellular stimuli. Atherosclerosis is characterized by a complex interaction of inflammation, lipid deposition, vascular smooth muscle cell proliferation, endothelial dysfunction, and extracellular matrix remodeling, which will result in the formation of an intimal plaque. Although the regulation and function of vascular smooth muscle cells are important in the progression of atherosclerosis, the roles of smooth muscle cells in regulating vascular inflammation are rarely focused upon, compared to those of endothelial cells or inflammatory cells. Therefore, in this review, we will discuss here how smooth muscle cells contribute or regulate the inflammatory reaction in the progression of atherosclerosis, especially in the context of the activation of various membrane receptors, and how they may regulate vascular inflammation.

Keywords

THE ROLES OF vSMC IN THE PROGRESSION OF ATHEROSCLEROSIS

Atherosclerosis is a complicated chronic inflammatory disease, characterized by the development of atheromatous plaque in the intimal layer. Currently, the progression of atherosclerosis is divided into three stages. The initial step is the initiation of atherosclerosis, due to the activation of endothelial cells by metabolic risk factors, such as hyperlipidemia, hypertension, or pro-inflammatory mediators. The resultant endothelial dysfunction allows for the blood monocytes to permeate the endothelial cell layer, and infiltrate into the intima and subintima, resulting in phagocytosis of the LDL cholesterol, or oxidized phospholipids and foam cell formation. Necrosis of the lipid containing foam cells will result in propagation of inflammation, as well as coalescence of the lipid contents, resulting in necrotic core formation. In the next step, SMCs migrate from the media to the intima, proliferate, and form plaque. Pidkova and Berliner demonstrated that oxidized phospholipids, called POVPC, lead to vSMC proliferation and apoptosis, by inhibiting cellular differentiation at high concentration (1-3). Migrated SMCs in intima can produce extracellular matrix (ECM), to make a fibrous cap. The last step is thrombosis, and rupture of the atherosclerotic plaque. Typically, the thin layer of the fibrous cap easily ruptures, in which few SMCs and abundant macrophages exist, because inflammatory cells cause the death of SMCs, which produce collagen for the fibrous cap as a main source (4). Macrophages are observed to induce apoptosis of SMCs by the TNF-α/NO signaling pathway, and decrease collagen synthesis by the secretion of MMPs, such as MMP-9 and MMP-12 (5-7). Subsequently, these series of reactions cause diseases, such as myocardial infarction, stroke, or congestive heart failure (8).

 

THE ROLE OF MEMBRANE RECEPTORS OF vSMC IN VASCULAR INFLAMMATION

AT1R (Angiotensin II type 1 receptor)

The role of the Renin-Angiotensin system in the pathophysiology of cardiovascular diseases has been extensively studied. Angiotensin II (Ang II), through their activation of the angiotensin type 1 receptor, has been known to play an important role in the development of atherosclerosis, as well as cardiac dysfunction (9,10). AT1R was first shown to be expressed in vascular smooth muscle cell in 1973 (11). Extensive researches have been done regarding the role of Ang II in promoting inflammation by increasing oxidative stress, as well as by upregulating various proinflammatory signals, such as RAGE, VCAM-1, ICAM-1, and MCP-1 in vSMCs (12). Moreover, Cai et al. showed that Ang II-induced proinflammatory signals allow monocytes to bind to vSMCs, indicating a highly significant association of vSMCs in inflammation (13). Ang II, besides the proinflammatory effect, also induces SMCs proliferation/migration and hypertrophic response, which can contribute to the development of atherosclerosis. However, there are conflicting reports on whether the presence of AT1aR activation in the endothelial cells or vSMCs is required for atherosclerosis development. In a recent study, AT1aR in endothelial or smooth muscle cell was not necessary to induce atherosclerosis and abdominal aortic aneurysm in mice having endothelial or smooth muscle cell-specific deletion of AT1aR (14). However, in another study, endothelial cell-specific deficiency of AT1aR showed the attenuation of Ang II-induced ascending aortic aneurysm in LDL receptor KO mice (15). ATRs have been extensively researched, and angiotensin type 1 receptor antagonists are being clinically used for the treatment of cardiovascular disease (16,17). However, more research needs to be done regarding the role of other receptors in the renin-angiotensin system, such as the MAS receptor and angiotensin II type 2 receptor, in the pathogenesis of atherosclerosis.

RAGE (Receptor for advanced glycation end products)

RAGE is a transmembrane receptor, and recognizes various endogenous ligands, including AGE, HMGB1 and S100 proteins, as a multi-ligand receptor (18-20). RAGE is a pattern recognizing receptor that is important in mediating innate immunity. RAGE is abdundantly expressed in various tissues, and are known to be expressed in endothelial cells and vSMCs (21). Since RAGE was first described and demonstrated from cardiovascular cells (22,23), many studies have focused on the proinflammatory effect of RAGE activation in diabetes and atherosclerosis. As direct evidence, soluble RAGE (sRAGE), a decoy receptor that blocks RAGE activation, inhibited atherosclerotic severity in streptozotocin-induced diabetic ApoE-null mice (24). Our recent study has shown that infusion of Angiotensin II in ApoE knockout mice results in increased atherosclerosis and overexpression of RAGE activation, a process that has been shown to be attenuated by RAGE inhibition (12). Studies have shown that ligands of RAGE, such as S100B, HMGB1 increase inflammation, and promote atherosclerosis. Hayakawa and colleagues showed that the treatment of S100B in RAGE-overexpressed SMC cell line was associated with the expression of proinflammatory genes, such as MCP-1, IL-6, and ICAM-1, and activates mitogen-activated protein kinase and the NFκ-B signaling pathway (25). HMGB1, a chromatin structural protein that is released extracellularly during tissue necrosis, has been shown to trigger inflammation during atherogenesis, and amplify inflammatory reaction, through increasing IL-1β, an inflammatory cytokine, in vSMCs (26). Recent studies suggest the involvement of new signaling pathways in HMGB1-induced inflammation, in which caspase-1 activation mediated by active inflammasome, such as NLRP3, results in increased IL-1β production and the propagation of inflammation. HMGB1 mediated inflammation is mediated through various pattern recognizing receptors, including TLR2, TLR4, and RAGE (27,28). Although these results were demonstrated in other cell types, the involvement of the novel signaling pathway in mediating vascular inflammation is highly expected. Since several receptors can interact under inflammatory stimuli, researchers have focused on the main receptors involved in mediating vascular inflammation. Kokkola compared the proinflammatory activity among TLR2, RAGE, and IL-1 receptor type I using KO mouse, and demonstrated that RAGE is the major receptor activated from stimuli in macrophages (29). Although RAGE activation is associated with increased oxidative stress and proinflammatory signaling activation through NFκB activation, the intracellular signaling cascasde for these processes is not well defined. For RAGE interacting adaptor proteins, few molecular mechanisms have been elucidated. However, a recent study by Schmidt et al. revealed that mDia1, a member of the formin family, binds to cytoplasmic domain of RAGE under the activation of S100B, which can induce oxidative stress, migration and vascular remodeling, through c-Src translocation and Rac1 activation, followed by NOX1-PI3K-AKT-GSK3β signaling pathway in SMCs, of the wire-injured mouse model (30). A new role of RAGE in vascular osteogenesis has recently been demonstrated, which showed that RAGE promotes vascular calcification, through the activation of Wnt/β-catenin signaling (31). Another article demonstrates that RAGE can play a crucial role in vascular calcification, in peritoneal dialysis patients (32). Although RAGE-ligand induced signaling has been implicated in the inflammation associated with chronic diseases, such as diabetes and atherogenic process, the physiological roles or signaling pathways in vSMCs have still not been fully elucidated.

LOX-1 (lectin-like oxidized LDL receptor)

The importance of LDLR in atherosclerosis has been demonstrated in the middle of the 1970s (33,34), from the accelerated atherosclerosis found in familial hypercholesterolemia patients lacking LDLR. The lack of LDL receptor is associated with decreased clearance of LDL in the circulation and hypercholesterolemia, resulting in the accelerated development of atherosclerosis. Since then, mouse models of atherosclerosis have been developed, by knocking out the LDL receptor in mice (35), and LDLR-knockout mice have been widely used to study several diseases or symptoms, including atherosclerosis, fibrosis, and nephrotic syndrome. In addition, LDLR is also known to regulate the amount of apolipoprotein E or amyloid β in the brain (36). However, the LDL receptor is not the main pathway for the uptake of cholesterol in the arteries, as evidenced by the accelerated atherosclerosis in familial hypercholesterolemia (34,37). In macrophage, LDL needs to be modified by oxidization or acetylation for phagocytosis (38). The modified LDLs are bound and internalized through LOX-1. In vSMCs, overexpression of LOX-1 levels in response to ox-LDL results in activation of inflammatory signals, such as NF-κB and JNK (39). LOX-1, identified as the major ox-LDL receptor in endothelial cells, may play a critical role in the initiation and progression of atherosclerosis, through their uptake of modified LDLs, and activation of the proinflammatory pathway in vSMCs (40). In vSMCs, proinflammatory cytokines, such as TGF-β, IL-1α, and IL-1β, increase LOX-1 expression; whereas, LOX-1 inhibition leads to significant reduction in the formation of the atherosclerotic lesions (41). Since ox-LDL uptake through LOX-1 is critical for foam cell formation, which is the hallmark of the atherosclerotic region, and vSMCs in intimal area highly express LOX-1, some pioneer studies have addressed the role of vSMC related to LOX-1. Although these studies did not present direct evidence, high possibilities are suggested for the transformation from SMCs into foam cells though LOX-1 (42,43). Statins such as pravastatin showed a new pleiotropic effect, in which pravastatin downregulated LOX-1 expressions in human vSMCs and atherosclerotic lesions, and reduced intimal media thickness and lipid core area (43). In a study using mice overexpressing LOX-1 from ApoE KO background, proatherosclerotic role of LOX-1 was demonstrated. Also, a synergistic role with LDLR was suggested from LOX-1 and LDL double knockout mice, in which the double knockout mice had a significantly reduced atherosclerotic lesion formation, compared to that of LDLR knockout mice (44).

TLRs (Toll-like receptors)

TLRs, one of the well-characterized-pattern-recognition receptors, are important receptors for mediating innate immunity. Around 13 different TLRs have been demonstrated in humans and mammals, and exogenous and endogenous ligands have been identified for 9 different types of TLRs, in monocytes/macrophages, dendritic cells, T and B lymphocytes, mast cells and vascular cells (45). Among the TLRs, increased expression levels of TLR 2 and 4 have been shown in atherosclerotic plaques, and their importance for initiation and progression have been demonstrated from several studies (46,47).

TLR2 is important for the recognition of bacterial lipoproteins, and known to promote inflammation in vSMCs, which has been demonstrated by previous studies, using apolipoprotein E-knockout (ApoE KO) mouse (48-50). Although researches related to TLR2 have been performed in vSMCs, most studies focus on the proliferation or migration mechanisms of vSMCs, rather than being related to inflammatory response. In the case of vSMCs proliferation, TLR2 and TLR4 signaling pathway are known to be involved in neointimal lesions under HSP60 stimulation (51). According to recent studies, TLR2 can activate p38 MAPK, ERK1/2, JNK1/2 and AKT, in vSMCs from TLR2 KO mouse. Also, cAMP response element-binding protein (CREB)-dependent IL-6 production was highly related to TLR2, not TLR4, in vSMCs migration (52). Interestingly, Lee et al. suggested a new role of TLR2 in vSMCs, where MMP2 and proinflammatory cytokines can be produced through TLR2-Nox1-dependent manner, leading to increasing monocyte-endothelial cell adhesion, and transendothelial migration of monocytic cells (53).

TLR4 is mainly stimulated by lipopolysaccharide (LPS) in vSMCs, and is known to induce inflammatory signals, as well as TLR2. Yang and colleagues showed that TLR4 stimulated by LPS activated phosphorylation of extracellular regulated kinase 1/2 (ERK1/2), induced monocyte-chemoattractant protein-1 (MCP-1) release and interleukin (IL)-6, and increased IL-1α expression (54). Another research group also examined increased TLR4 expression and nitric oxide (NO) production by LPS playing very important roles to induce inflammation in human aortic vascular SMCs (55). TLR4 can increase MMP-9 expression, which is known to be involved in the pathogenesis of atherosclerotic plaque instability, as well as abnormal arterial remodeling. Li et al. demonstrated that TLR4/NF-κB pathway, stimulated by LPS, was able to increase the expression of MMP-9 mRNA and protein for human aortic vascular SMCs migration, indicating a high possibility for the correlation between TLR4/NF-κΒ pathway, and pathogenesis of atherosclerosis (56). Up to date, studies have demonstrated that TLR2 and TLR4 share common pathways, or have synergetic effects in the inflammatory signaling. However, recent study suggested that TLR4 has a protective role in atherosclerosis progression, whereas TLR2 has a proinflammatory response, followed by periodontal pathogen (57). Therefore, further in-depth researches are needed to understand the differing roles of TLR4 in vascular inflammation.

NLRs (Nod-like receptors)

NLRs are another type of cytoplasmic pattern-recognition receptors, which play important roles in innate immunity (58). Among members of the NLRs family, NLRP3 is the receptor studied most extensively for its association with cardiovascular diseases, such as cardiomyopathy and heart failure (59). Microbial molecules or endogenous danger signals can activate NLRP3, and in turn, NLRP3 recruits and makes a complex with apoptosis-associated speck-like protein (ASC), and pro-caspase-1 (60). This complex is called an active NLRP3 inflammasome, which activates caspase-1, IL-1β, and IL-18. NLRP3 is also involved with autophagy, which is a quality control system by degradation or re-use of intracellular organelles and proteins, and is essential for cell survival (61). NLRP3 inflammasome was reported to co-localize with autophagosome, as autophagy mechanism can regulate inflammasome activation, through the interaction between autophagic adaptor p62 and ASC (62). Another study demonstrated the importance of this relationship, by showing that LPS induces more mortality, through the active NLRP3 inflammasome in autophagy-deficient (LC3B-deficient) mouse model (63). New roles of NLRP3 have been elucidated, with regard to the pathogenesis of cardiovascular diseases. In human coronary atherosclerosis, NLRP3 was reported to have a positive correlation with an increased epicardial adipose tissue volume, which has been shown to be associated with increased cytokine production and proatherogenesis (64). In heart from NLRP3 KO mice, the protective effect of ischemic preconditioning is increased against ischemia-reperfusion injury, through the reduced expression of STAT3 (65). There are some debates as to whether NLRP3 inflammasome is involved in the progression of atherosclerosis, or not. In the LDLR KO-diabetic mice model, activation of NLRP3 inflammasome with increased inflammatory responses, as well as IL-1β production, has been demonstrated in the atherosclerotic lesions (66). On the other hand, NLRP3 did not show any difference from ApoE KO and NLRP3-/ApoE-double KO mice study (68). Nevertheless, recent studies strongly suggest the involvement of NLRP3 inflammasome in atherosclerosis. Qial et al. showed that activated NF-κB due to TLR4 activation can bind to the NLRP3 promoter, resulting in the increase of NLRP3 expression from macrophages (67). Xiao et al. also demonstrated that sterol regulatory element binding protein 2 (SREBP2) overexpress NLRP3, which results in endothelial inflammation, and atherosclerosis formation. SREBP2, as an initial activator, was able to be activated in endothelial cells by atheroprone arterial blood flow, leading to NLRP3 inflammasome activated IL-1β signaling, followed by subsequent activation of NADPH oxidase2 (NOX2), in the area of atherosclerotic lesions (69). In a study regarding the role of NLRP3 within vSMCs in mediating inflammation during atherogenesis, Xiao et al. demonstrated that NLRP3 activation due to atheroprone blood flow is important for inflammation and progression of atherosclerosis. As disturbed arterial blood flow has been known to induce phenotypic changes in vSMCs that are highly involved in inflammatory reaction through the overexpression of receptors affecting inflammation and adhesion, we speculate that NLRP3 may have an important role in mediating activation of vSMCs in atherogenesis (70,71).

 

CONCLUSIONS

Until recently, vSMCs had been considered to passively transform or differentiate into form cells, by factors from macrophage stimulation. However, recent studies have focused on the active roles of vSMCs in propagating inflammation, during the development of atherosclerosis. In this review, we summarized the possible role of vSMCs in actively mediating inflammation, through their regulation of cytokine secretion and membrane receptors, during the pathogenesis of atherosclerosis (Fig. 1).

Fig. 1.Involvement of vSMCs in the inflammatory response of atherosclerosis. 1-5 briefly show LDL-induced inflammatory reactions, in which modified LDL particles are accumulated in the artery under various risk factors, resulting in the formation of form cells. This process is mediated by uptake of ox-LDL particles from the activation of scavenger receptors of macrophages, such as CD36, SRA1/2, LOX-1, and TLRs. In addition, SMCs can produce proinflammatory cytokines, through the activation of LOX-1, RAGE, TLR2, TLR4, AT1R, or NLRP3 by their specific agonists, and by their interaction with endothelial cells and macrophages.

References

  1. Pidkovka, N. A., Cherepanova, O. A., Yoshida, T., Alexander, M. R., Deaton, R. A., Thomas, J. A., Leitinger, N. and Owens, G. K. (2007) Oxidized phospholipids induce phenotypic switching of vascular smooth muscle cells in vivo and in vitro. Circ. Res. 101, 792-801. https://doi.org/10.1161/CIRCRESAHA.107.152736
  2. Berliner, J. A. and Watson, A. D. (2005) A role for oxidized phospholipids in atherosclerosis. N. Engl. J. Med. 353, 9-11. https://doi.org/10.1056/NEJMp058118
  3. Libby, P., Ridker, P. M. and Hansson, G. K. (2011) Progress and challenges in translating the biology of atherosclerosis. Nature 473, 317-325. https://doi.org/10.1038/nature10146
  4. Lacolley, P., Regnault, V., Nicoletti, A., Li, Z. and Michel, J. B. (2012) The vascular smooth muscle cell in arterial pathology: a cell that can take on multiple roles. Cardiovasc. Res. 95, 194-204. https://doi.org/10.1093/cvr/cvs135
  5. Boyle, J. J., Weissberg, P. L. and Bennett, M. R. (2003) Tumor necrosis factor-alpha promotes macrophage-induced vascular smooth muscle cell apoptosis by direct and autocrine mechanisms. Arterioscler. Thromb. Vasc. Biol. 23, 1553-1558. https://doi.org/10.1161/01.ATV.0000086961.44581.B7
  6. Gough, P. J., Gomez, I. G., Wille, P. T. and Raines, E. W. (2006) Macrophage expression of active MMP-9 induces acute plaque disruption in apoE-deficient mice. J. Clin. Invest. 116, 59-69.
  7. Johnson, J. L., Devel, L., Czarny, B., George, S. J., Jackson, C. L., Rogakos, V., Beau, F., Yiotakis, A., Newby, A. C. and Dive, V. (2011) A selective matrix metalloproteinase-12 inhibitor retards atherosclerotic plaque development in apolipoprotein E-knockout mice. Arterioscler. Thromb. Vasc. Biol. 31, 528-535. https://doi.org/10.1161/ATVBAHA.110.219147
  8. Mehta, P. K. and Griendling, K. K. (2007) Angiotensin II cell signaling: physiological and pathological effects in the cardiovascular system. Am. J. Physiol. Cell. Physiol. 292, 82-97.
  9. Williams, B. (2001) Angiotensin II and the pathophysiology of cardiovascular remodeling. Am. J. Cardiol. 87, 10-17. https://doi.org/10.1016/S0002-9149(01)01507-7
  10. Ferrario, C. M. (2006) Role of angiotensin II in cardiovascular disease therapeutic implications of more than a century of research. J. Renin Angiotensin Aldosterone Syst. 7, 3-14. https://doi.org/10.3317/jraas.2006.003
  11. Park, W. K., Regoli, D. and Rioux, F. (1973) Characterization of angiotensin receptors in vascular and intestinal smooth muscles. Br. J. Pharmacol. 48, 288-301. https://doi.org/10.1111/j.1476-5381.1973.tb06915.x
  12. Lee, D., Lee, K. H., Park, H., Kim, S. H., Jin, T., Cho, S., Chung, J. H., Lim, S. and Park, S. (2013) The effect of soluble RAGE on inhibition of angiotensin II-mediated atherosclerosis in apolipoprotein E deficient mice. PloS One 8, e69669. https://doi.org/10.1371/journal.pone.0069669
  13. Cai, Q., Lanting, L. and Natarajan, R. (2004) Growth factors induce monocyte binding to vascular smooth muscle cells: implications for monocyte retention in atherosclerosis. American journal of physiology. Am. J. Physiol. Cell Physiol. 287, 707-714.
  14. Rateri, D. L., Moorleghen, J. J., Knight, V., Balakrishnan, A., Howatt, D. A., Cassis, L. A. and Daugherty, A. (2012) Depletion of endothelial or smooth muscle cell-specific angiotensin II type 1a receptors does not influence aortic aneurysms or atherosclerosis in LDL receptor deficient mice. PloS One 7, e51483. https://doi.org/10.1371/journal.pone.0051483
  15. Rateri, D. L., Moorleghen, J. J., Balakrishnan, A., Owens, A. P. 3rd, Howatt, D. A., Subramanian, V., Poduri, A., Charnigo, R., Cassis, L. A. and Daugherty, A. (2011) Endothelial cell-specific deficiency of Ang II type 1a receptors attenuates Ang II-induced ascending aortic aneurysms in LDL receptor-/- mice. Circ. Res. 108, 574-581. https://doi.org/10.1161/CIRCRESAHA.110.222844
  16. Doggrell, S. A. (2002) Angiotensin AT-1 receptor antagonism: complementary or alternative to ACE inhibition in cardiovascular and renal disease? Expert Opin. Pharmacother. 11, 1543-1556.
  17. Schmidt, B., Drexler, H. and Schieffer, B. (2004) Therapeutic effects of angiotensin (AT1) receptor antagonists: potential contribution of mechanisms other than AT1 receptor blockade. Am. J. Cardiovasc. Drugs. 4, 361-368. https://doi.org/10.2165/00129784-200404060-00004
  18. Neeper, M., Schmidt, A. M., Brett, J., Yan, S. D., Wang, F., Pan, Y. C., Elliston, K., Stern, D. and Shaw, A. (1992) Cloning and expression of a cell surface receptor for advanced glycosylation end products of proteins. J. Biol. Chem. 267, 14998-15004.
  19. Hori, O., Brett, J., Slattery, T., Cao, R., Zhang, J., Chen, J. X., Nagashima, M., Lundh, E. R., Vijay, S., Nitecki, D., Morser, J., Stern, D. and Schmidt, A. M. (1995) The receptor for advanced glycation end products (RAGE) is a cellular binding site for amphoterin. Mediation of neurite outgrowth and co-expression of rage and amphoterin in the developing nervous system. J. Biol. Chem. 270, 25752-25761. https://doi.org/10.1074/jbc.270.43.25752
  20. Heizmann, C. W., Ackermann, G. E. and Galichet, A. (2007) Pathologies involving the S100 proteins and RAGE. Subcell. Biochem. 45, 93-138. https://doi.org/10.1007/978-1-4020-6191-2_5
  21. Brett, J., Schmidt, A. M., Yan, S. D., Zou, Y. S., Weidman, E., Pinsky, D., Nowygrod, R., Neeper, M., Przysiecki, C., Shaw, A., Migheli, A. and Stern, D. (1993) Survey of the distribution of a newly characterized receptor for advanced glycation end products in tissues. Am. J. Pathol. 143, 1699-1712.
  22. Kirstein, M., Brett, J., Radoff, S., Ogawa, S., Stern, D. and Vlassara, H. (1990) Advanced protein glycosylation induces transendothelial human monocyte chemotaxis and secretion of platelet-derived growth factor: role in vascular disease of diabetes and aging. Proc. Natl. Acad. Sci. U. S. A. 87, 9010-9014. https://doi.org/10.1073/pnas.87.22.9010
  23. Esposito, C., Gerlach, H., Brett, J., Stern, D. and Vlassara, H. (1989) Endothelial receptor-mediated binding of glucose-modified albumin is associated with increased monolayer permeability and modulation of cell surface coagulant properties. J. Exp. Med. 170, 1387-1407. https://doi.org/10.1084/jem.170.4.1387
  24. Hudson, B. I., Bucciarelli, L. G., Wendt, T., Sakaguchi, T., Lalla, E., Qu, W., Lu, Y., Lee, L., Stern, D. M., Naka, Y., Ramasamy, R., Yan, S. D., Yan, S. F., D'Agati, V. and Schmidt, A. M. (2003) Blockade of receptor for advanced glycation endproducts: a new target for therapeutic intervention in diabetic complications and inflammatory disorders. Arch. Biochem. Biophys. 419, 80-88. https://doi.org/10.1016/j.abb.2003.08.030
  25. Hayakawa, E., Yoshimoto, T., Sekizawa, N., Sugiyama, T. and Hirata, Y. (2012) Overexpression of receptor for advanced glycation end products induces monocyte chemoattractant protein-1 expression in rat vascular smooth muscle cell line. J. Atheroscler. Thromb. 19, 13-22. https://doi.org/10.5551/jat.9472
  26. Jaulmes, A., Thierry, S., Janvier, B., Raymondjean, M. and Marechal, V. (2006) Activation of sPLA2-IIA and PGE2 production by high mobility group protein B1 in vascular smooth muscle cells sensitized by IL-1beta. FASEB J. 20, 1727-1729. https://doi.org/10.1096/fj.05-5514fje
  27. Lippai, D., Bala, S., Petrasek, J., Csak, T., Levin, I., Kurt-Jones, E. A. and Szabo, G. (2013) Alcohol-induced IL-1beta in the brain is mediated by NLRP3/ASC inflammasome activation that amplifies neuroinflammation. J. leukoc. Biol. 94, 171-182. https://doi.org/10.1189/jlb.1212659
  28. Bae, J.-S. and Rezaie, A. R. (2013) Thrombin inhibits HMGB1-mediated proinflammatory signaling responses when endothelial protein C receptor is occupied by its natural ligand. BMB Rep. 46, 544-549. https://doi.org/10.5483/BMBRep.2013.46.11.056
  29. Kokkola, R., Andersson, A., Mullins, G., Ostberg, T., Treutiger, C. J., Arnold, B., Nawroth, P., Andersson, U., Harris, R. A. and Harris, H. E. (2005) RAGE is the major receptor for the proinflammatory activity of HMGB1 in rodent macrophages. Scand. J. Immunol. 61, 1-9. https://doi.org/10.1111/j.0300-9475.2005.01534.x
  30. Toure, F., Fritz, G., Li, Q., Rai, V., Daffu, G., Zou, Y. S., Rosario, R., Ramasamy, R., Alberts, A. S., Yan, S. F. and Schmidt, A. M. (2012) Formin mDia1 mediates vascular remodeling via integration of oxidative and signal transduction pathways. Circ. Res. 110, 1279-1293. https://doi.org/10.1161/CIRCRESAHA.111.262519
  31. Menini, S., Iacobini, C., Ricci, C., Blasetti Fantauzzi, C., Salvi, L., Pesce, C. M., Relucenti, M., Familiari, G., Taurino, M. and Pugliese, G. (2013) The galectin-3/RAGE dyad modulatesvascular osteogenesis in atherosclerosis. Cardiovasc. Res. 100, 472-480. https://doi.org/10.1093/cvr/cvt206
  32. Kim, J. K., Park, S., Lee, M. J., Song, Y. R., Han, S. H., Kim, S. G., Kang, S. W., Choi, K. H., Kim, H. J. and Yoo, T. H. (2012) Plasma levels of soluble receptor for advanced glycation end products (sRAGE) and proinflammatory ligand for RAGE (EN-RAGE) are associated with carotid atherosclerosis in patients with peritoneal dialysis. Atherosclerosis 220, 208-214. https://doi.org/10.1016/j.atherosclerosis.2011.07.115
  33. Goldstein, J. L. and Brown, M. S. (1975) Lipoprotein receptors, cholesterol metabolism, and atherosclerosis. Arch. Pathol. 99, 181-184.
  34. Nestel, P. J. (1980) Lipoprotein protein receptors and their relation to atherosclerosis. Circ. Res. 46, I106-109.
  35. Kowala, M. C., Recce, R., Beyer, S., Gu, C. and Valentine, M. (2000) Characterization of atherosclerosis in LDL receptor knockout mice: macrophage accumulation correlates with rapid and sustained expression of aortic MCP-1/JE. Atherosclerosis 149, 323-330. https://doi.org/10.1016/S0021-9150(99)00342-1
  36. Basak, J. M., Verghese, P. B., Yoon, H., Kim, J. and Holtzman, D. M. (2012) Low-density lipoprotein receptor represents an apolipoprotein E-independent pathway of Abeta uptake and degradation by astrocytes. J. Biol. Chem. 287, 13959-13971. https://doi.org/10.1074/jbc.M111.288746
  37. Naderi, G. A., Asgary, S., Sarraf-Zadegan, N. and Shirvany, H. (2003) Anti-oxidant effect of flavonoids on the susceptibility of LDL oxidation. Mol. Cell. Biochem. 246, 193-196. https://doi.org/10.1023/A:1023483223842
  38. Basu, S. K., Brown, M. S., Ho, Y. K. and Goldstein, J. L. (1979) Degradation of low density lipoprotein . dextran sulfate complexes associated with deposition of cholesteryl esters in mouse macrophages. J. Biol. Chem. 254, 7141-7146.
  39. Sun, Y. and Chen, X. (2011) Ox-LDL-induced LOX-1 expression in vascular smooth muscle cells: role of reactive oxygen species. Funda. Clin. Pharmacol. 25, 572-579. https://doi.org/10.1111/j.1472-8206.2010.00885.x
  40. Sawamura, T., Kume, N., Aoyama, T., Moriwaki, H., Hoshikawa, H., Aiba, Y., Tanaka, T., Miwa, S., Katsura, Y., Kita, T. and Masaki, T. (1997) An endothelial receptor for oxidized low-density lipoprotein. Nature 386, 73-77. https://doi.org/10.1038/386073a0
  41. Hofnagel, O., Luechtenborg, B., Stolle, K., Lorkowski, S., Eschert, H., Plenz, G. and Robenek, H. (2004) Proinflammatory cytokines regulate LOX-1 expression in vascular smooth muscle cells. Arterioscler. Thromb. Vasc. Biol. 24, 1789-1795. https://doi.org/10.1161/01.ATV.0000140061.89096.2b
  42. Limor, R., Kaplan, M., Sawamura, T., Sharon, O., Keidar, S., Weisinger, G., Knoll, E., Naidich, M. and Stern, N. (2005) Angiotensin II increases the expression of lectin-like oxidized low-density lipoprotein receptor-1 in human vascular smooth muscle cells via a lipoxygenase-dependent pathway. Am. J. Hypertens. 18, 299-307. https://doi.org/10.1016/j.amjhyper.2004.09.008
  43. Hofnagel, O., Luechtenborg, B., Eschert, H., Weissen-Plenz, G., Severs, N. J. and Robenek, H. (2006) Pravastatin inhibits expression of lectin-like oxidized low-density lipoprotein receptor-1 (LOX-1) in Watanabe heritable hyperlipidemic rabbits: a new pleiotropic effect of statins. Arterioscler. Thromb. Vasc. Biol. 26, 604-610.
  44. Morawietz, H. (2007) LOX-1 and atherosclerosis: proof of concept in LOX-1-knockout mice. Circ. Res. 100, 1534-1536. https://doi.org/10.1161/CIRCRESAHA.107.101105
  45. Cole, J. E., Georgiou, E. and Monaco, C. (2010) The expression and functions of toll-like receptors in atherosclerosis. Mediators Inflamm. 2010, 393946.
  46. Edfeldt, K., Swedenborg, J., Hansson, G. K. and Yan, Z. Q. (2002) Expression of toll-like receptors in human atherosclerotic lesions: a possible pathway for plaque activation. Circulation 105, 1158-1161.
  47. Curtiss, L. K. and Tobias, P. S. (2009) Emerging role of Toll-like receptors in atherosclerosis. J. Lipid Res. 50 (Suppl), 340-345. https://doi.org/10.1194/jlr.R800056-JLR200
  48. Yang, X., Coriolan, D., Schultz, K., Golenbock, D. T. and Beasley, D. (2005) Toll-like receptor 2 mediates persistent chemokine release by Chlamydia pneumoniae-infected vascular smooth muscle cells. Arterioscler. Thromb. Vasc. Biol. 25, 2308-2314. https://doi.org/10.1161/01.ATV.0000187468.00675.a3
  49. Schoneveld, A. H., Oude Nijhuis, M. M., van Middelaar, B., Laman, J. D., de Kleijn, D. P. and Pasterkamp, G. (2005) Toll-like receptor 2 stimulation induces intimal hyperplasia and atherosclerotic lesion development. Cardiovas. Res. 66, 162-169. https://doi.org/10.1016/j.cardiores.2004.12.016
  50. Madan, M. and Amar, S. (2008) Toll-like receptor-2 mediates diet and/or pathogen associated atherosclerosis: proteomic findings. PloS One 3, e3204. https://doi.org/10.1371/journal.pone.0003204
  51. de Graaf, R., Kloppenburg, G., Kitslaar, P. J., Bruggeman, C. A. and Stassen, F. (2006) Human heat shock protein 60 stimulates vascular smooth muscle cell proliferation through Toll-like receptors 2 and 4. Microbes Infect. 8, 1859-1865. https://doi.org/10.1016/j.micinf.2006.02.024
  52. Lee, G. L., Chang, Y. W., Wu, J. Y., Wu, M. L., Wu, K. K., Yet, S. F. and Kuo, C. C. (2012) TLR 2 induces vascular smooth muscle cell migration through cAMP response element-binding protein-mediated interleukin-6 production. Arterioscler. Thromb. Vasc. Biol. 32, 2751-2760. https://doi.org/10.1161/ATVBAHA.112.300302
  53. Lee, J. H., Joo, J. H., Kim, J., Lim, H. J., Kim, S., Curtiss, L., Seong, J. K., Cui, W., Yabe-Nishimura, C. and Bae, Y. S. (2013) Interaction of NADPH oxidase 1 with Toll-like receptor 2 induces migration of smooth muscle cells. Cardiovas. Res. 99, 483-493. https://doi.org/10.1093/cvr/cvt107
  54. Yang, X., Coriolan, D., Murthy, V., Schultz, K., Golenbock, D. T. and Beasley, D. (2005) Proinflammatory phenotype of vascular smooth muscle cells: role of efficient Toll-like receptor 4 signaling. Am. J. Physiol. Heart. Circ. Physiol. 289, 1069-1076. https://doi.org/10.1152/ajpheart.00143.2005
  55. Heo, S. K., Yun, H. J., Noh, E. K., Park, W. H. and Park, S. D. (2008) LPS induces inflammatory responses in human aortic vascular smooth muscle cells via Toll-like receptor 4 expression and nitric oxide production. Immunol. Lett. 120, 57-64. https://doi.org/10.1016/j.imlet.2008.07.002
  56. Li, H., Xu, H. and Sun, B. (2012) Lipopolysaccharide regulates MMP-9 expression through TLR4/NF-kappaB signaling in human arterial smooth muscle cells. Mol. Med. Rep. 6, 774-778. https://doi.org/10.3892/mmr.2012.1010
  57. Hayashi, C., Papadopoulos, G., Gudino, C. V., Weinberg, E. O., Barth, K. R., Madrigal, A. G., Chen, Y., Ning, H., LaValley, M., Gibson, F. C. 3rd, Hamilton, J. A. and Genco, C. A. (2012) Protective role for TLR4 signaling in atherosclerosis progression as revealed by infection with a common oral pathogen. J. Immunol. 189, 3681-3688. https://doi.org/10.4049/jimmunol.1201541
  58. Delbridge, L. M. and O'Riordan, M. X. (2007) Innate recognition of intracellular bacteria. Curr. Opin. Immunol. 19, 10-16. https://doi.org/10.1016/j.coi.2006.11.005
  59. Bracey, N. A., Beck, P. L., Muruve, D. A., Hirota, S. A., Guo, J., Jabagi, H., Wright, J. R. Jr., Macdonald, J. A., Lees-Miller, J. P., Roach, D., Semeniuk, L. M. and Duff, H. J. (2013) The Nlrp3 inflammasome promotes myocardial dysfunction in structural cardiomyopathy through interleukin-1beta. Exp. Physiol. 98, 462-472. https://doi.org/10.1113/expphysiol.2012.068338
  60. Srinivasula, S. M., Poyet, J. L., Razmara, M., Datta, P., Zhang, Z. and Alnemri, E. S. (2002) The PYRIN-CARD protein ASC is an activating adaptor for caspase-1. J. Biol. Chem. 277, 21119-21122. https://doi.org/10.1074/jbc.C200179200
  61. Behrends, C., Sowa, M. E., Gygi, S. P. and Harper, J. W. (2010) Network organization of the human autophagy system. Nature 466, 68-76. https://doi.org/10.1038/nature09204
  62. Shi, C. S., Shenderov, K., Huang, N. N., Kabat, J., Abu-Asab, M., Fitzgerald, K. A., Sher, A. and Kehrl, J. H. (2012) Activation of autophagy by inflammatory signals limits IL-1beta production by targeting ubiquitinated inflammasomes for destruction. Nat. Immunol. 13, 255-263. https://doi.org/10.1038/ni.2215
  63. Nakahira, K., Haspel, J. A., Rathinam, V. A., Lee, S. J., Dolinay, T., Lam, H. C., Englert, J. A., Rabinovitch, M., Cernadas, M., Kim, H. P., Fitzgerald, K. A., Ryter, S. W. and Choi, A. M. (2011) Autophagy proteins regulate innate immune responses by inhibiting the release of mitochondrial DNA mediated by the NALP3 inflammasome. Nat. Immunol. 12, 222-230.
  64. Shimabukuro, M., Hirata, Y., Tabata, M., Dagvasumberel, M., Sato, H., Kurobe, H., Fukuda, D., Soeki, T., Kitagawa, T., Takanashi, S. and Sata, M. (2013) Epicardial adipose tissue volume and adipocytokine imbalance are strongly linked to human coronary atherosclerosis. Arterioscler. Thromb. Vasc. Biol. 33, 1077-1084. https://doi.org/10.1161/ATVBAHA.112.300829
  65. Zuurbier, C. J., Jong, W. M., Eerbeek, O., Koeman, A., Pulskens, W. P., Butter, L. M., Leemans, J. C. and Hollmann, M. W. (2012) Deletion of the innate immune NLRP3 receptor abolishes cardiac ischemic preconditioning and is associated with decreased Il-6/STAT3 signaling. PloS One 7, e40643. https://doi.org/10.1371/journal.pone.0040643
  66. Li, Y., Xu, S., Jiang, B., Cohen, R. A. and Zang, M. (2013) Activation of sterol regulatory element binding protein and NLRP3 inflammasome in atherosclerotic lesion development in diabetic pigs. PloS One 8, e67532. https://doi.org/10.1371/journal.pone.0067532
  67. Menu, P., Pellegrin, M., Aubert, J. F., Bouzourene, K., Tardivel, A., Mazzolai, L. and Tschopp, J. (2011) Atherosclerosis in ApoE-deficient mice progresses independently of the NLRP3 inflammasome. Cell Death Dis. 2, e137. https://doi.org/10.1038/cddis.2011.18
  68. Qiao, Y., Wang, P., Qi, J., Zhang, L. and Gao, C. (2012) TLR-induced NF-kappaB activation regulates NLRP3 expression in murine macrophages. FEBS Lett. 586, 1022-1026. https://doi.org/10.1016/j.febslet.2012.02.045
  69. Xiao, H., Lu, M., Lin, T. Y., Chen, Z., Chen, G., Wang, W. C., Marin, T., Shentu, T. P., Wen, L., Gongol, B., Sun, W., Liang, X., Chen, J., Huang, H. D., Pedra, J. H., Johnson, D. A. and Shyy, J. Y. (2013) Sterol regulatory element binding protein 2 activation of NLRP3 inflammasome in endothelium mediates hemodynamic-induced atherosclerosis susceptibility. Circulation 128, 632-642. https://doi.org/10.1161/CIRCULATIONAHA.113.002714
  70. Shi, Z. D. and Tarbell, J. M. (2011) Fluid flow mechanotransduction in vascular smooth muscle cells and fibroblasts. Ann. Biomed. Eng. 39, 1608-1619. https://doi.org/10.1007/s10439-011-0309-2
  71. Tedgui, A. and Mallat, Z. (2001) Anti-inflammatory mechanisms in the vascular wall. Circ. Res. 88, 877-887. https://doi.org/10.1161/hh0901.090440

Cited by

  1. The Role of Protein Arginine Methyltransferases in Inflammatory Responses vol.2016, 2016, https://doi.org/10.1155/2016/4028353
  2. Proatherosclerotic Effect of the α1-Subunit of Soluble Guanylyl Cyclase by Promoting Smooth Muscle Phenotypic Switching vol.186, pp.8, 2016, https://doi.org/10.1016/j.ajpath.2016.04.010
  3. Maternal serum uric acid concentration is associated with the expression of tumour necrosis factor-α and intercellular adhesion molecule-1 in patients with preeclampsia vol.30, pp.7, 2016, https://doi.org/10.1038/jhh.2015.110
  4. Inflammation in arterial diseases vol.67, pp.1, 2015, https://doi.org/10.1002/iub.1344
  5. Gingerol Inhibits Serum-Induced Vascular Smooth Muscle Cell Proliferation and Injury-Induced Neointimal Hyperplasia by Suppressing p38 MAPK Activation vol.21, pp.2, 2016, https://doi.org/10.1177/1074248415598003
  6. PEP-1-FK506BP inhibits alkali burn-induced corneal inflammation on the rat model of corneal alkali injury vol.48, pp.11, 2015, https://doi.org/10.5483/BMBRep.2015.48.11.041
  7. Matrix Metalloproteinases and Subclinical Atherosclerosis in Chronic Kidney Disease: A Systematic Review vol.2016, 2016, https://doi.org/10.1155/2016/9498013
  8. The p-ERK–p-c-Jun–cyclinD1 pathway is involved in proliferation of smooth muscle cells after exposure to cigarette smoke extract vol.453, pp.3, 2014, https://doi.org/10.1016/j.bbrc.2014.09.062
  9. Circulating cell-free mitochondrial DNA as the probable inducer of early endothelial dysfunction in the prediabetic patient vol.69, 2015, https://doi.org/10.1016/j.exger.2015.05.010
  10. MicroRNA-26a prevents endothelial cell apoptosis by directly targeting TRPC6 in the setting of atherosclerosis vol.5, pp.1, 2015, https://doi.org/10.1038/srep09401
  11. Platelet-Derived Growth Factor Receptor-β Regulates Vascular Smooth Muscle Cell Phenotypic Transformation and Neuroinflammation After Intracerebral Hemorrhage in Mice vol.44, pp.6, 2016, https://doi.org/10.1097/CCM.0000000000001425
  12. Syk and IRAK1 Contribute to Immunopharmacological Activities of Anthraquinone-2-carboxlic Acid vol.21, pp.6, 2016, https://doi.org/10.3390/molecules21060809
  13. MAPK/AP-1-Targeted Anti-Inflammatory Activities of Xanthium strumarium vol.44, pp.06, 2016, https://doi.org/10.1142/S0192415X16500622
  14. Angiotensin-(1–7) counteracts the effects of Ang II on vascular smooth muscle cells, vascular remodeling and hemorrhagic stroke: Role of the NFкB inflammatory pathway vol.73, 2015, https://doi.org/10.1016/j.vph.2015.08.007
  15. Upregulation of TRPC1/6 may be involved in arterial remodeling in rat vol.195, pp.1, 2015, https://doi.org/10.1016/j.jss.2014.12.047
  16. Stigmasterol protects against Ang II-induced proliferation of the A7r5 aortic smooth muscle cell-line vol.6, pp.7, 2015, https://doi.org/10.1039/C5FO00031A
  17. Lipopolysaccharide induced vascular smooth muscle cells proliferation: A new potential therapeutic target for proliferative vascular diseases vol.50, pp.2, 2017, https://doi.org/10.1111/cpr.12332
  18. Icaritin Inhibits Collagen Degradation-Related Factors and Facilitates Collagen Accumulation in Atherosclerotic Lesions: A Potential Action for Plaque Stabilization vol.17, pp.2, 2016, https://doi.org/10.3390/ijms17020169
  19. Thymoquinone: An IRAK1 inhibitor with in vivo and in vitro anti-inflammatory activities vol.7, 2017, https://doi.org/10.1038/srep42995
  20. Human miR-221/222 in Physiological and Atherosclerotic Vascular Remodeling vol.2015, 2015, https://doi.org/10.1155/2015/354517
  21. Cyanidin-3-O-glucoside Induces Apoptosis and Inhibits Migration of Tumor Necrosis Factor-α-Treated Rat Aortic Smooth Muscle Cells vol.16, pp.3, 2016, https://doi.org/10.1007/s12012-015-9333-z
  22. Ginsenoside Rc from Panax ginseng exerts anti-inflammatory activity by targeting TANK-binding kinase 1/interferon regulatory factor-3 and p38/ATF-2 vol.41, pp.2, 2017, https://doi.org/10.1016/j.jgr.2016.02.001
  23. Analysis of Soluble Molecular Fibronectin-Fibrin Complexes and EDA-Fibronectin Concentration in Plasma of Patients with Atherosclerosis 2016, https://doi.org/10.1007/s10753-016-0336-0
  24. Ursolic acid induced anti-proliferation effects in rat primary vascular smooth muscle cells is associated with inhibition of microRNA-21 and subsequent PTEN/PI3K vol.781, 2016, https://doi.org/10.1016/j.ejphar.2016.04.001
  25. Vasoregression: A Shared Vascular Pathology Underlying Macrovascular And Microvascular Pathologies? vol.19, pp.12, 2015, https://doi.org/10.1089/omi.2015.0128
  26. In vivo treatment of rat arterial adventitia with interleukin-1β induces intimal proliferation via the JAK2/STAT3 signaling pathway vol.13, pp.4, 2016, https://doi.org/10.3892/mmr.2016.4982
  27. Atheroma Susceptible to Thrombosis Exhibit Impaired Endothelial Permeability In Vivo as Assessed by Nanoparticle-Based Fluorescence Molecular Imaging vol.10, pp.5, 2017, https://doi.org/10.1161/CIRCIMAGING.116.005813
  28. Vascular Smooth Muscle Cells and Arterial Stiffening: Relevance in Development, Aging, and Disease vol.97, pp.4, 2017, https://doi.org/10.1152/physrev.00003.2017
  29. PCSK9: A novel inflammation modulator in atherosclerosis? vol.234, pp.3, 2019, https://doi.org/10.1002/jcp.27254
  30. Lysosomal membrane permeabilization causes secretion of IL-1β in human vascular smooth muscle cells vol.67, pp.10, 2018, https://doi.org/10.1007/s00011-018-1178-z
  31. Puerarin inhibits vascular smooth muscle cells proliferation induced by fine particulate matter via suppressing of the p38 MAPK signaling pathway vol.18, pp.1, 2018, https://doi.org/10.1186/s12906-018-2206-9
  32. Dan-Lou Prescription Inhibits Foam Cell Formation Induced by ox-LDL via the TLR4/NF-κB and PPARγ Signaling Pathways vol.9, pp.1664-042X, 2018, https://doi.org/10.3389/fphys.2018.00590
  33. SHP2 inhibitor PHPS1 protects against atherosclerosis by inhibiting smooth muscle cell proliferation vol.18, pp.1, 2018, https://doi.org/10.1186/s12872-018-0816-2
  34. Overexpression of UHRF1 promoted the proliferation of vascular smooth cells via the regulation of Geminin protein levels vol.39, pp.2, 2019, https://doi.org/10.1042/BSR20181341