951
|
|
952
|
Wilensky RL, Shi Y, Mohler ER, Hamamdzic D, Burgert ME, Li J, Postle A, Fenning RS, Bollinger JG, Hoffman BE, Pelchovitz DJ, Yang J, Mirabile RC, Webb CL, Zhang L, Zhang P, Gelb MH, Walker MC, Zalewski A, Macphee CH. Inhibition of lipoprotein-associated phospholipase A2 reduces complex coronary atherosclerotic plaque development. Nat Med 2008; 14:1059-66. [PMID: 18806801 PMCID: PMC2885134 DOI: 10.1038/nm.1870] [Citation(s) in RCA: 286] [Impact Index Per Article: 17.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2008] [Accepted: 08/21/2008] [Indexed: 12/13/2022]
Abstract
Increased lipoprotein-associated phospholipase A(2) (Lp-PLA(2)) activity is associated with increased risk of cardiac events, but it is not known whether Lp-PLA(2) is a causative agent. Here we show that selective inhibition of Lp-PLA(2) with darapladib reduced development of advanced coronary atherosclerosis in diabetic and hypercholesterolemic swine. Darapladib markedly inhibited plasma and lesion Lp-PLA(2) activity and reduced lesion lysophosphatidylcholine content. Analysis of coronary gene expression showed that darapladib exerted a general anti-inflammatory action, substantially reducing the expression of 24 genes associated with macrophage and T lymphocyte functioning. Darapladib treatment resulted in a considerable decrease in plaque area and, notably, a markedly reduced necrotic core area and reduced medial destruction, resulting in fewer lesions with an unstable phenotype. These data show that selective inhibition of Lp-PLA(2) inhibits progression to advanced coronary atherosclerotic lesions and confirms a crucial role of vascular inflammation independent from hypercholesterolemia in the development of lesions implicated in the pathogenesis of myocardial infarction and stroke.
Collapse
Affiliation(s)
- Robert L Wilensky
- Hospital of the University of Pennsylvania, 3400 Spruce Street, 9 Gates, Philadelphia, Pennsylvania 19104, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
953
|
Loppnow H, Werdan K, Buerke M. Vascular cells contribute to atherosclerosis by cytokine- and innate-immunity-related inflammatory mechanisms. Innate Immun 2008; 14:63-87. [PMID: 18713724 DOI: 10.1177/1753425908091246] [Citation(s) in RCA: 62] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Cardiovascular diseases are the human diseases with the highest death rate and atherosclerosis is one of the major underlying causes of cardiovascular diseases. Inflammatory and innate immune mechanisms, employing monocytes, innate receptors, innate cytokines, or chemokines are suggested to be involved in atherogenesis. Among the inflammatory pathways the cytokines are central players. Plasma levels of cytokines and related proteins, such as CRP, have been investigated in cardiovascular patients, tissue mRNA expression was analyzed and correlations to vascular diseases established. Consistent with these findings the generation of cytokine-deficient animals has provided direct evidence for a role of cytokines in atherosclerosis. In vitro cell culture experiments further support the suggestion that cytokines and other innate mechanisms contribute to atherogenesis. Among the initiation pathways of atherogenesis are innate mechanisms, such as toll-like-receptors (TLRs), including the endotoxin receptor TLR4. On the other hand, innate cytokines, such as IL-1 or TNF, or even autoimmune triggers may activate the cells. Cytokines potently activate multiple functions relevant to maintain or spoil homeostasis within the vessel wall. Vascular cells, not least smooth muscle cells, can actively contribute to the inflammatory cytokine-dependent network in the blood vessel wall by: (i) production of cytokines; (ii) response to these potent cell activators; and (iii) cytokine-mediated interaction with invading cells, such as monocytes, T-cells, or mast cells. Activation of these pathways results in accumulation of cells and increased LDL- and ECM-deposition which may serve as an 'immunovascular memory' resulting in an ever-growing response to subsequent invasions. Thus, vascular cells may potently contribute to the inflammatory pathways involved in development and acceleration of atherosclerosis.
Collapse
Affiliation(s)
- Harald Loppnow
- Martin-Luther-Universität Halle-Wittenberg, Universitätsklinik und Poliklinik für Innere Medizin , Halle (Saale), Germany.
| | | | | |
Collapse
|
954
|
Libby P, Nahrendorf M, Pittet MJ, Swirski FK. Diversity of denizens of the atherosclerotic plaque: not all monocytes are created equal. Circulation 2008; 117:3168-70. [PMID: 18574058 DOI: 10.1161/circulationaha.108.783068] [Citation(s) in RCA: 56] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
955
|
Korosoglou G, Weiss RG, Kedziorek DA, Walczak P, Gilson WD, Schär M, Sosnovik DE, Kraitchman DL, Boston RC, Bulte JWM, Weissleder R, Stuber M. Noninvasive detection of macrophage-rich atherosclerotic plaque in hyperlipidemic rabbits using "positive contrast" magnetic resonance imaging. J Am Coll Cardiol 2008; 52:483-91. [PMID: 18672170 DOI: 10.1016/j.jacc.2008.03.063] [Citation(s) in RCA: 103] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2007] [Revised: 02/12/2008] [Accepted: 03/19/2008] [Indexed: 11/16/2022]
Abstract
OBJECTIVES This study was designed to identify macrophage-rich atherosclerotic plaque noninvasively by imaging the tissue uptake of long-circulating superparamagnetic nanoparticles with a positive contrast off-resonance imaging sequence (inversion recovery with ON-resonant water suppression [IRON]). BACKGROUND The sudden rupture of macrophage-rich atherosclerotic plaques can trigger the formation of an occlusive thrombus in coronary vessels, resulting in acute myocardial infarction. Therefore, a noninvasive technique that can identify macrophage-rich plaques and thereby assist with risk stratification of patients with atherosclerosis would be of great potential clinical utility. METHODS Experiments were conducted on a clinical 3-T magnetic resonance imaging (MRI) scanner in 7 heritable hyperlipidemic and 4 control rabbits. Monocrystalline iron-oxide nanoparticles (MION)-47 were administrated intravenously (2 doses of 250 mumol Fe/kg), and animals underwent serial IRON-MRI before injection of the nanoparticles and serially after 1, 3, and 6 days. RESULTS After administration of MION-47, a striking signal enhancement was found in areas of plaque only in hyperlipidemic rabbits. The magnitude of enhancement on magnetic resonance images had a high correlation with the number of macrophages determined by histology (p < 0.001) and allowed for the detection of macrophage-rich plaque with high accuracy (area under the curve: 0.92, SE: 0.04, 95% confidence interval: 0.84 to 0.96, p < 0.001). No significant signal enhancement was measured in remote areas without plaque by histology and in control rabbits without atherosclerosis. CONCLUSIONS Using IRON-MRI in conjunction with superparamagnetic nanoparticles is a promising approach for the noninvasive evaluation of macrophage-rich, vulnerable plaques.
Collapse
|
956
|
Waterhouse DF, Cahill RA, Sheehan F, McCreery C. Prediction of calculated future cardiovascular disease by monocyte count in an asymptomatic population. Vasc Health Risk Manag 2008; 4:177-87. [PMID: 18629357 PMCID: PMC2464752 DOI: 10.2147/vhrm.2008.04.01.177] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
INTRODUCTION Although atherogenesis is clearly entwined with systemic inflammation, the risk-predictive relationship between preclinical and overt cardiovascular disease (CVD) and systemic white blood cell (WBC) subtypes remains unclear. Implication of an association would greatly facilitate cardiac risk prediction, assessment and monitoring. METHODS 1383 asymptomatic individuals (795 men, 588 women) attending for executive health screening were examined clinically as well as with phlebotomy and exercise stress testing to determine their ten-year risk of developing overt cardiovascular disease (as estimated by both Framingham and SCORE calculations). The significance of their association with overall WBC and subtypes were determined using both univariate and multiple regression modeling. RESULTS OF ALL WBC SUBTYPES, MONOCYTE COUNT WAS FOUND TO HAVE THE STRONGEST, INDEPENDENT RELATIONSHIP WITH OVERALL CVD RISK BY BACKWARDS LINEAR REGRESSION MODELING (FRAMINGHAM: beta = 0.057; p = 0.03; SCORE: beta = 0.128; p = <0.0005). Independent associations with BMI (beta = 5.214; p = <0.0005), waist circumference (beta = 21.866; p = <0.0005), systolic blood pressure (beta = 10.738; p = 0.003), HDL cholesterol (beta = -0.639; p = <0.0005) and triglyceride concentrations (beta = 0.787; p = <0.0005) were also evident. Overall WBC along with neutrophils, lymphocytes and basophil subfractions were variably (but less strongly) associated with such dependents and outcome measures. CONCLUSIONS In conclusion, monocyte count, a simple inexpensive test, may provide useful predictive cardiovascular risk information in asymptomatic individuals to inform and guide attempts at interrupting CVD development at a preclinical stage.
Collapse
Affiliation(s)
- Deirdre F Waterhouse
- Department of Cardiovascular Medicine, St Vincent's University Hospital, Dublin, Ireland.
| | | | | | | |
Collapse
|
957
|
Jakubzick C, Tacke F, Ginhoux F, Wagers AJ, van Rooijen N, Mack M, Merad M, Randolph GJ. Blood monocyte subsets differentially give rise to CD103+ and CD103- pulmonary dendritic cell populations. THE JOURNAL OF IMMUNOLOGY 2008; 180:3019-27. [PMID: 18292524 DOI: 10.4049/jimmunol.180.5.3019] [Citation(s) in RCA: 185] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
There are two major myeloid pulmonary dendritic cell (DC) populations: CD103+ DCs and CD11bhigh DCs. In this study, we investigated in detail the origins of both myeloid DC pools using multiple experimental approaches. We show that, in resting lung, Ly-6ChighCCR2high monocytes repopulated CD103+ DCs using a CCR2-dependent mechanism, and these DCs preferentially retained residual CCR2 in the lung, whereas, conversely, Ly-6ClowCCR2low monocytes repopulated CD11bhigh DCs. CX3CR1 was required to generate normal numbers of pulmonary CD11bhigh DCs, possibly because Ly-6Clow monocytes in the circulation, which normally express high levels of CX3CR1, failed to express bcl-2 and may have diminished survival in the circulation in the absence of CX3CR1. Overall, these data demonstrate that the two circulating subsets of monocytes give rise to distinct tissue DC populations.
Collapse
Affiliation(s)
- Claudia Jakubzick
- Department of Gene and Cell Medicine, Icahn Research Institute, Mount Sinai School of Medicine, New York, NY 10029, USA
| | | | | | | | | | | | | | | |
Collapse
|
958
|
Johnson JL, Sala-Newby GB, Ismail Y, Aguilera CM, Newby AC. Low tissue inhibitor of metalloproteinases 3 and high matrix metalloproteinase 14 levels defines a subpopulation of highly invasive foam-cell macrophages. Arterioscler Thromb Vasc Biol 2008; 28:1647-53. [PMID: 18566294 DOI: 10.1161/atvbaha.108.170548] [Citation(s) in RCA: 60] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
OBJECTIVE An excess of metalloproteinases (MMPs) over tissue inhibitors of metalloproteinases (TIMPs) may favor atherosclerotic plaque rupture. We compared TIMP levels in nonfoamy and foam-cell macrophages (FCM) generated in vivo. METHODS AND RESULTS In vivo generated rabbit FCM exhibited 84% reduced TIMP-3 protein compared to nonfoamy macrophages, and immunocytochemistry revealed a TIMP-3 negative subset (28%). Strikingly, only TIMP-3 negative FCM invaded a synthetic basement membrane, and invasion was inhibited by exogenous TIMP-3. TIMP-3 negative FCM also had increased proliferation and apoptosis rates compared to TIMP-3 positive cells, which were retarded by exogenous TIMP-3; this also reduced gelatinolytic activity. TIMP-3 negative FCM were found at the base of advanced rabbit plaques and in the rupture-prone shoulders of human plaques. To explain the actions of low TIMP-3 we observed a 26-fold increase in MT1-MMP (MMP-14) protein in FCM. Adding an MT1-MMP neutralizing antibody reduced foam-cell invasion, apoptosis, and gelatinolytic activity. Furthermore, MT1-MMP overexpressing and TIMP-3 negative FCM were found at the same locations in atherosclerotic plaques. CONCLUSIONS These results demonstrate that TIMP-3 is downregulated in a distinct subpopulation of FCM which have increased MMP-14. These cells are highly invasive and have increased proliferation and apoptosis, all properties expected to destabilise atherosclerotic plaques.
Collapse
|
959
|
Wang HW, Liu PY, Oyama N, Rikitake Y, Kitamoto S, Gitlin J, Liao JK, Boisvert WA. Deficiency of ROCK1 in bone marrow-derived cells protects against atherosclerosis in LDLR-/- mice. FASEB J 2008; 22:3561-70. [PMID: 18556458 DOI: 10.1096/fj.08-108829] [Citation(s) in RCA: 60] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Rho kinases (ROCKs) are serine-threonine protein kinases that regulate the actin cytoskeleton. Recent studies suggest that ROCKs also play an important role in cardiovascular disease. However, the isoform- and tissue-specific role of ROCKs in mediating this process is unknown. Using homologous recombination, we generated mutant mice harboring alleles with homozygous deletion of ROCK1 (ROCK1(-/-)). Most ROCK1(-/-) mice die perinatally. However, a few ROCK1(-/-) mice survive to adulthood, are phenotypically normal, and have no apparent compensatory changes in ROCK2. Using these ROCK1(-/-) mice, we show that ROCK1 in bone marrow-derived macrophages is critical to the development of atherosclerosis, in part, by mediating foam cell formation and macrophage chemotaxis. Lipid accumulation and atherosclerotic lesions were reduced in atherosclerosis-prone LDLR(-/-) mice, whose bone marrows have been replaced with bone marrows derived from ROCK1(-/-) mice. Bone marrow-derived ROCK1-deficient macrophages exhibited impaired chemotaxis to monocyte chemotactic protein-1 and showed reduced ability to take up lipids and to develop into foam cells when exposed to modified low-density lipoprotein. These findings indicate that ROCK1 in bone marrow-derived cells is a critical mediator of atherogenesis and suggest that macrophage ROCK1 may be an important therapeutic target for vascular inflammation and atherosclerosis.
Collapse
Affiliation(s)
- Hong-Wei Wang
- Vascular Medicine Research Unit, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts 02139, USA
| | | | | | | | | | | | | | | |
Collapse
|
960
|
An G, Wang H, Tang R, Yago T, McDaniel JM, McGee S, Huo Y, Xia L. P-selectin glycoprotein ligand-1 is highly expressed on Ly-6Chi monocytes and a major determinant for Ly-6Chi monocyte recruitment to sites of atherosclerosis in mice. Circulation 2008; 117:3227-37. [PMID: 18519846 DOI: 10.1161/circulationaha.108.771048] [Citation(s) in RCA: 133] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
BACKGROUND Ly-6C(hi) monocytes are key contributors to atherosclerosis in mice. However, the manner in which Ly-6C(hi) monocytes selectively accumulate in atherosclerotic lesions is largely unknown. Monocyte homing to sites of atherosclerosis is primarily initiated by rolling on P- and E-selectin expressed on endothelium. We hypothesize that P-selectin glycoprotein ligand-1 (PSGL-1), the common ligand of P- and E-selectin on leukocytes, contributes to the preferential homing of Ly-6C(hi) monocytes to atherosclerotic lesions. METHODS AND RESULTS To test this hypothesis, we examined the expression and function of PSGL-1 on Ly-6C(hi) and Ly-6C(lo) monocytes from wild-type mice, ApoE(-/-) mice, and mice lacking both ApoE and PSGL-1 genes (ApoE(-/-)/PSGL-1(-/-)). We found that Ly-6C(hi) monocytes expressed a higher level of PSGL-1 and had enhanced binding to fluid-phase P- and E-selectin compared with Ly-6C(lo) monocytes. Under in vitro flow conditions, more Ly-6C(hi) monocytes rolled on P-, E-, and L-selectin at slower velocities than Ly-6C(lo) cells. In an ex vivo perfused carotid artery model, Ly-6C(hi) monocytes interacted preferentially with atherosclerotic endothelium compared with Ly-6C(lo) monocytes in a PSGL-1-dependent manner. In vivo, ApoE(-/-) mice lacking PSGL-1 had impaired Ly-6C(hi) monocyte recruitment to atherosclerotic lesions. Moreover, ApoE(-/-)/PSGL-1(-/-) mice exhibited significantly reduced monocyte infiltration in wire injury-induced neointima and in atherosclerotic lesions. ApoE(-/-)/PSGL-1(-/-) mice also developed smaller neointima and atherosclerotic plaques. CONCLUSIONS These data indicate that PSGL-1 is a new marker for Ly-6C(hi) monocytes and a major determinant for Ly-6C(hi) cell recruitment to sites of atherosclerosis in mice.
Collapse
Affiliation(s)
- Guangyu An
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK 73104, USA
| | | | | | | | | | | | | | | |
Collapse
|
961
|
Abstract
The link between inflammation and metabolism was apparent already early last century, but has recently been revitalized following molecular studies of atherosclerosis, obesity and insulin resistance. A growing list of nuclear receptors, pivotal players in lipid, xenobiotic and energy metabolism has been identified as having immunomodulatory functions. These receptors might hold the key to some of the questions pertinent to chronic inflammation, and can lend themselves to be manipulated as therapeutic agents. This review will attempt to appraise the importance of such mediators in the pathophysiology of chronic inflammation in the colon.
Collapse
Affiliation(s)
- V Arulampalam
- Department of Microbiology Tumorbiology & Cell Biology, Karolinska Institutet, Stockholm, Sweden.
| |
Collapse
|
962
|
Abstract
Our understanding of the biologic effects (including toxicity) of nanomaterials is incomplete. In vivo animal studies remain the gold standard; however, widespread testing remains impractical, and the development of in vitro assays that correlate with in vivo activity has proven challenging. Here, we demonstrate the feasibility of analyzing in vitro nanomaterial activity in a generalizable, systematic fashion. We assessed nanoparticle effects in a multidimensional manner, using multiple cell types and multiple assays that reflect different aspects of cellular physiology. Hierarchical clustering of these data identifies nanomaterials with similar patterns of biologic activity across a broad sampling of cellular contexts, as opposed to extrapolating from results of a single in vitro assay. We show that this approach yields robust and detailed structure-activity relationships. Furthermore, a subset of nanoparticles were tested in mice, and nanoparticles with similar activity profiles in vitro exert similar effects on monocyte number in vivo. These data suggest a strategy of multidimensional characterization of nanomaterials in vitro that can inform the design of novel nanomaterials and guide studies of in vivo activity.
Collapse
|
963
|
Rader DJ, Daugherty A. Translating molecular discoveries into new therapies for atherosclerosis. Nature 2008; 451:904-13. [PMID: 18288179 DOI: 10.1038/nature06796] [Citation(s) in RCA: 363] [Impact Index Per Article: 22.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Atherosclerosis is characterized by the thickening of the arterial wall and is the primary cause of coronary artery disease and cerebrovascular disease, two of the most common causes of illness and death worldwide. Clinical trials have confirmed that certain lipoproteins and the renin-angiotensin-aldosterone system are important in the pathogenesis of atherosclerotic cardiovascular disease, and that interventions targeted towards these are beneficial. Furthermore, efforts to understand how risk factors such as high blood pressure, dysregulated blood lipids and diabetes contribute to atherosclerotic disease, as well as to understand the molecular pathogenesis of atherosclerotic plaques, are leading to new targets for therapy.
Collapse
Affiliation(s)
- Daniel J Rader
- Cardiovascular Institute and Institute for Translational Medicine and Therapeutics, University of Pennsylvania School of Medicine, 654 BRBII/III, 421 Curie Boulevard, Philadelphia, Pennsylvania 19104, USA.
| | | |
Collapse
|
964
|
Combadière C, Potteaux S, Rodero M, Simon T, Pezard A, Esposito B, Merval R, Proudfoot A, Tedgui A, Mallat Z. Combined inhibition of CCL2, CX3CR1, and CCR5 abrogates Ly6C(hi) and Ly6C(lo) monocytosis and almost abolishes atherosclerosis in hypercholesterolemic mice. Circulation 2008; 117:1649-57. [PMID: 18347211 DOI: 10.1161/circulationaha.107.745091] [Citation(s) in RCA: 529] [Impact Index Per Article: 33.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
BACKGROUND Monocytes are critical mediators of atherogenesis. Deletion of individual chemokines or chemokine receptors leads to significant but only partial inhibition of lesion development, whereas deficiency in other signals such as CXCL16 or CCR1 accelerates atherosclerosis. Evidence that particular chemokine pathways may cooperate to promote monocyte accumulation into inflamed tissues, particularly atherosclerotic arteries, is still lacking. METHODS AND RESULTS Here, we show that chemokine-mediated signals critically determine the frequency of monocytes in the blood and bone marrow under both noninflammatory and atherosclerotic conditions. Particularly, CCL2-, CX3CR1-, and CCR5-dependent signals differentially alter CD11b(+) Ly6G(-) 7/4(hi) (also known as Ly6C(hi)) and CD11b(+) Ly6G(-) 7/4(lo) (Ly6C(lo)) monocytosis. Combined inhibition of CCL2, CX3CR1, and CCR5 in hypercholesterolemic, atherosclerosis-susceptible apolipoprotein E-deficient mice leads to abrogation of bone marrow monocytosis and to additive reduction in circulating monocytes despite persistent hypercholesterolemia. These effects are associated with a marked and additive 90% reduction in atherosclerosis. Interestingly, lesion size highly correlates with the number of circulating monocytes, particularly the CD11b(+) Ly6G(-) 7/4(lo) subset. CONCLUSIONS CCL2, CX3CR1, and CCR5 play independent and additive roles in atherogenesis. Signals mediated through these pathways critically determine the frequency of circulating monocyte subsets and thereby account for almost all macrophage accumulation into atherosclerotic arteries.
Collapse
Affiliation(s)
- Christophe Combadière
- Institut National de la Santé et de la Recherche Médicale, Unit 543, Université Pierre et Marie Curie, Paris 6, Paris, France
| | | | | | | | | | | | | | | | | | | |
Collapse
|
965
|
|
966
|
Liao B, McCall E, Cox K, Lee CW, Huang S, Higgs RE, Chio LC, Zhen E, Hale JE, Jackson NK, Rutherford PG, Huang XD, Gifford-Moore D, Hui K, Duffin K, Gould KE, Rekhter M. Circulating Markers Reflect Both Anti- and Pro-Atherogenic Drug Effects in ApoE-Deficient Mice. Biomark Insights 2008; 3:147-157. [PMID: 19578502 PMCID: PMC2688353 DOI: 10.4137/bmi.s632] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
Abstract
Background Current drug therapy of atherosclerosis is focused on treatment of major risk factors, e.g. hypercholesterolemia while in the future direct disease modification might provide additional benefits. However, development of medicines targeting vascular wall disease is complicated by the lack of reliable biomarkers. In this study, we took a novel approach to identify circulating biomarkers indicative of drug efficacy by reducing the complexity of the in vivo system to the level where neither disease progression nor drug treatment was associated with the changes in plasma cholesterol. Results ApoE−/− mice were treated with an ACE inhibitor ramipril and HMG-CoA reductase inhibitor simvastatin. Ramipril significantly reduced the size of atherosclerotic plaques in brachiocephalic arteries, however simvastatin paradoxically stimulated atherogenesis. Both effects occurred without changes in plasma cholesterol. Blood and vascular samples were obtained from the same animals. In the whole blood RNA samples, expression of MMP9, CD14 and IL-1RN reflected pro-and anti-atherogenic drug effects. In the plasma, several proteins, e.g. IL-1β, IL-18 and MMP9 followed similar trends while protein readout was less sensitive than RNA analysis. Conclusion In this study, we have identified inflammation-related whole blood RNA and plasma protein markers reflecting anti-atherogenic effects of ramipril and pro-atherogenic effects of simwastatin in a mouse model of atherosclerosis. This opens an opportunity for early, non-invasive detection of direct drug effects on atherosclerotic plaques in complex in vivo systems.
Collapse
|
967
|
Ogawa H. [Coronary artery disease and inflammation]. NIHON NAIKA GAKKAI ZASSHI. THE JOURNAL OF THE JAPANESE SOCIETY OF INTERNAL MEDICINE 2008; 97:524-532. [PMID: 22675752 DOI: 10.2169/naika.97.524] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/01/2023]
|
968
|
Waldo SW, Li Y, Buono C, Zhao B, Billings EM, Chang J, Kruth HS. Heterogeneity of human macrophages in culture and in atherosclerotic plaques. THE AMERICAN JOURNAL OF PATHOLOGY 2008; 172:1112-26. [PMID: 18321997 DOI: 10.2353/ajpath.2008.070513] [Citation(s) in RCA: 183] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Research suggests that monocytes differentiate into unique lineage-determined macrophage subpopulations in response to the local cytokine environment. The present study evaluated the atherogenic potential of two divergent lineage-determined human monocyte-derived macrophage subpopulations. Monocytes were differentiated for 7 days in the presence of alternative macrophage development cytokines: granulocyte-macrophage colony-stimulating factor to produce granulocyte-macrophage-CSF macrophages (GM-Mac), or macrophage colony-stimulating factor (M-CSF) to produce M-Mac. Gene chip analyses of three monocyte donors demonstrated differential expression of inflammatory and cholesterol homeostasis genes in the macrophage subpopulations. Quantitative PCR confirmed a fivefold elevation in the expression of genes that promote reverse cholesterol transport (PPAR-gamma, LXR-alpha, and ABCG1) and macrophage emigration from lesions (CCR7) in GM-Mac compared to that in M-Mac. Immunocytochemistry confirmed enhanced expression of the proinflammatory marker CD14 in M-Mac relative to GM-Mac. M-Mac spontaneously accumulated cholesterol when incubated with unmodified low-density lipoprotein whereas GM-Mac only accumulated similar levels of cholesterol after protein kinase C activation. Immunostained human coronary arteries showed that macrophages with similar antigen expression to that of M-Mac (CD68(+)/CD14(+)) were predominant within atherosclerotic lesions whereas macrophages with antigen expression similar to GM-Mac (CD68(+)/CD14(-)) were predominant in areas devoid of disease. The identification of macrophage subpopulations with different gene expression patterns and, thus, different potentials for promoting atherosclerosis has important experimental and clinical implications and could prove to be a valuable finding in developing therapeutic interventions in diseases dependent on macrophage function.
Collapse
Affiliation(s)
- Stephen W Waldo
- Section of Experimental Atherosclerosis, National Heart, Lung, and Blood Institute, National Institutes of Health, 10 Center Drive, Bethesda, MD 20892-1422, USA
| | | | | | | | | | | | | |
Collapse
|
969
|
Abstract
The initiation and progression of vascular inflammation are driven by the retention of cholesterol in the artery wall, where its modification by oxidation and/or enzymes triggers the innate immune host response. Although previously considered a broad, primitive defense mechanism against invading pathogens, it has become clear that pattern recognition receptors of the innate immune system can cooperate to precisely regulate signaling pathways essential for the proper initiation of both innate and acquired immunity. Recent evidence suggests that these pattern recognition receptors may orchestrate the host response to modified endogenous ligands involved in sterile chronic inflammatory syndromes, including atherosclerosis. In this review we will summarize the current understanding of innate immune receptors and the putative ligands that regulate the numerous responses that promote this disease, including monocyte recruitment, macrophage cholesterol uptake, and pro-inflammatory signaling cascades. Specific emphasis will be placed on the potential of these innate immune targets for therapeutic interventions to retard the progression of atherosclerosis or to induce its regression.
Collapse
Affiliation(s)
- Kathryn J Moore
- Lipid Metabolism Unit, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114
| | | |
Collapse
|
970
|
Bourlier V, Zakaroff-Girard A, Miranville A, De Barros S, Maumus M, Sengenes C, Galitzky J, Lafontan M, Karpe F, Frayn K, Bouloumié A. Remodeling Phenotype of Human Subcutaneous Adipose Tissue Macrophages. Circulation 2008; 117:806-15. [DOI: 10.1161/circulationaha.107.724096] [Citation(s) in RCA: 274] [Impact Index Per Article: 17.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Affiliation(s)
- V. Bourlier
- From the Institut National de la Santé et de la Recherche Médicale (INSERM), Toulouse, France (V.B., A.Z.-G., S.D.B., M.M., C.S., J.G., M.L., A.B.); Université Toulouse III Paul-Sabatier, Institut de Médecine Moléculaire de Rangueil, Equipe No. 1 AVENIR, Toulouse, France (V.B., A.Z.-G., S.D.B., M.M., C.S., J.G., M.L., A.B.); Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Oxford, UK (F.K., K.N.F); and Department of Cardiovascular Physiology, J.-W. Goethe
| | - A. Zakaroff-Girard
- From the Institut National de la Santé et de la Recherche Médicale (INSERM), Toulouse, France (V.B., A.Z.-G., S.D.B., M.M., C.S., J.G., M.L., A.B.); Université Toulouse III Paul-Sabatier, Institut de Médecine Moléculaire de Rangueil, Equipe No. 1 AVENIR, Toulouse, France (V.B., A.Z.-G., S.D.B., M.M., C.S., J.G., M.L., A.B.); Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Oxford, UK (F.K., K.N.F); and Department of Cardiovascular Physiology, J.-W. Goethe
| | - A. Miranville
- From the Institut National de la Santé et de la Recherche Médicale (INSERM), Toulouse, France (V.B., A.Z.-G., S.D.B., M.M., C.S., J.G., M.L., A.B.); Université Toulouse III Paul-Sabatier, Institut de Médecine Moléculaire de Rangueil, Equipe No. 1 AVENIR, Toulouse, France (V.B., A.Z.-G., S.D.B., M.M., C.S., J.G., M.L., A.B.); Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Oxford, UK (F.K., K.N.F); and Department of Cardiovascular Physiology, J.-W. Goethe
| | - S. De Barros
- From the Institut National de la Santé et de la Recherche Médicale (INSERM), Toulouse, France (V.B., A.Z.-G., S.D.B., M.M., C.S., J.G., M.L., A.B.); Université Toulouse III Paul-Sabatier, Institut de Médecine Moléculaire de Rangueil, Equipe No. 1 AVENIR, Toulouse, France (V.B., A.Z.-G., S.D.B., M.M., C.S., J.G., M.L., A.B.); Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Oxford, UK (F.K., K.N.F); and Department of Cardiovascular Physiology, J.-W. Goethe
| | - M. Maumus
- From the Institut National de la Santé et de la Recherche Médicale (INSERM), Toulouse, France (V.B., A.Z.-G., S.D.B., M.M., C.S., J.G., M.L., A.B.); Université Toulouse III Paul-Sabatier, Institut de Médecine Moléculaire de Rangueil, Equipe No. 1 AVENIR, Toulouse, France (V.B., A.Z.-G., S.D.B., M.M., C.S., J.G., M.L., A.B.); Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Oxford, UK (F.K., K.N.F); and Department of Cardiovascular Physiology, J.-W. Goethe
| | - C. Sengenes
- From the Institut National de la Santé et de la Recherche Médicale (INSERM), Toulouse, France (V.B., A.Z.-G., S.D.B., M.M., C.S., J.G., M.L., A.B.); Université Toulouse III Paul-Sabatier, Institut de Médecine Moléculaire de Rangueil, Equipe No. 1 AVENIR, Toulouse, France (V.B., A.Z.-G., S.D.B., M.M., C.S., J.G., M.L., A.B.); Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Oxford, UK (F.K., K.N.F); and Department of Cardiovascular Physiology, J.-W. Goethe
| | - J. Galitzky
- From the Institut National de la Santé et de la Recherche Médicale (INSERM), Toulouse, France (V.B., A.Z.-G., S.D.B., M.M., C.S., J.G., M.L., A.B.); Université Toulouse III Paul-Sabatier, Institut de Médecine Moléculaire de Rangueil, Equipe No. 1 AVENIR, Toulouse, France (V.B., A.Z.-G., S.D.B., M.M., C.S., J.G., M.L., A.B.); Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Oxford, UK (F.K., K.N.F); and Department of Cardiovascular Physiology, J.-W. Goethe
| | - M. Lafontan
- From the Institut National de la Santé et de la Recherche Médicale (INSERM), Toulouse, France (V.B., A.Z.-G., S.D.B., M.M., C.S., J.G., M.L., A.B.); Université Toulouse III Paul-Sabatier, Institut de Médecine Moléculaire de Rangueil, Equipe No. 1 AVENIR, Toulouse, France (V.B., A.Z.-G., S.D.B., M.M., C.S., J.G., M.L., A.B.); Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Oxford, UK (F.K., K.N.F); and Department of Cardiovascular Physiology, J.-W. Goethe
| | - F. Karpe
- From the Institut National de la Santé et de la Recherche Médicale (INSERM), Toulouse, France (V.B., A.Z.-G., S.D.B., M.M., C.S., J.G., M.L., A.B.); Université Toulouse III Paul-Sabatier, Institut de Médecine Moléculaire de Rangueil, Equipe No. 1 AVENIR, Toulouse, France (V.B., A.Z.-G., S.D.B., M.M., C.S., J.G., M.L., A.B.); Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Oxford, UK (F.K., K.N.F); and Department of Cardiovascular Physiology, J.-W. Goethe
| | - K.N. Frayn
- From the Institut National de la Santé et de la Recherche Médicale (INSERM), Toulouse, France (V.B., A.Z.-G., S.D.B., M.M., C.S., J.G., M.L., A.B.); Université Toulouse III Paul-Sabatier, Institut de Médecine Moléculaire de Rangueil, Equipe No. 1 AVENIR, Toulouse, France (V.B., A.Z.-G., S.D.B., M.M., C.S., J.G., M.L., A.B.); Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Oxford, UK (F.K., K.N.F); and Department of Cardiovascular Physiology, J.-W. Goethe
| | - A. Bouloumié
- From the Institut National de la Santé et de la Recherche Médicale (INSERM), Toulouse, France (V.B., A.Z.-G., S.D.B., M.M., C.S., J.G., M.L., A.B.); Université Toulouse III Paul-Sabatier, Institut de Médecine Moléculaire de Rangueil, Equipe No. 1 AVENIR, Toulouse, France (V.B., A.Z.-G., S.D.B., M.M., C.S., J.G., M.L., A.B.); Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Oxford, UK (F.K., K.N.F); and Department of Cardiovascular Physiology, J.-W. Goethe
| |
Collapse
|
971
|
Type 1 diabetes promotes disruption of advanced atherosclerotic lesions in LDL receptor-deficient mice. Proc Natl Acad Sci U S A 2008; 105:2082-7. [PMID: 18252823 DOI: 10.1073/pnas.0709958105] [Citation(s) in RCA: 69] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Cardiovascular disease, largely because of disruption of atherosclerotic lesions, accounts for the majority of deaths in people with type 1 diabetes. Recent mouse models have provided insights into the accelerated atherosclerotic lesion initiation in diabetes, but it is unknown whether diabetes directly worsens more clinically relevant advanced lesions. We therefore used an LDL receptor-deficient mouse model, in which type 1 diabetes can be induced at will, to investigate the effects of diabetes on preexisting lesions. Advanced lesions were induced by feeding mice a high-fat diet for 16 weeks before induction of diabetes. Diabetes, independently of lesion size, increased intraplaque hemorrhage and plaque disruption in the brachiocephalic artery of mice fed low-fat or high-fat diets for an additional 14 weeks. Hyperglycemia was not sufficient to induce plaque disruption. Furthermore, diabetes resulted in increased accumulation of monocytic cells positive for S100A9, a proinflammatory biomarker for cardiovascular events, and for a macrophage marker protein, without increasing lesion macrophage content. S100A9 immunoreactivity correlated with intraplaque hemorrhage. Aggressive lowering primarily of triglyceride-rich lipoproteins prevented both plaque disruption and the increased S100A9 in diabetic atherosclerotic lesions. Conversely, oleate promoted macrophage differentiation into an S100A9-positive population in vitro, thereby mimicking the effects of diabetes. Thus, diabetes increases plaque disruption, independently of effects on plaque initiation, through a mechanism that requires triglyceride-rich lipoproteins and is associated with an increased accumulation of S100A9-positive monocytic cells. These findings indicate an important link between diabetes, plaque disruption, and the innate immune system.
Collapse
|
972
|
Randolph GJ, Jakubzick C, Qu C. Antigen presentation by monocytes and monocyte-derived cells. Curr Opin Immunol 2008; 20:52-60. [PMID: 18160272 PMCID: PMC2408874 DOI: 10.1016/j.coi.2007.10.010] [Citation(s) in RCA: 167] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2007] [Accepted: 10/31/2007] [Indexed: 01/18/2023]
Abstract
Monocytes are circulating mononuclear phagocytes with a fundamental capacity to differentiate into macrophages. This differentiation can, in the presence of the right environmental cues, be re-directed instead to dendritic cells (DCs). Recent advances have been made in understanding the role of monocytes and their derivatives in presenting antigen to drive immune responses, and we review this topic herein. We briefly discuss the heterogeneity of monocytes in the blood and subsequently raise the possibility that one of the major monocyte phenotypes in the blood corresponds with a population of 'blood DCs' previously proposed to drive T-independent antibody reactions in the spleen. Then we evaluate the role of monocytes in T-dependent immunity, considering their role in acquiring antigens for presentation before exiting the bloodstream and their ability to differentiate into macrophages versus antigen-presenting DCs. Finally, we review recent literature on the role of monocyte-derived cells in cross-presentation and discuss the possibility that monocyte-derived cells participate critically in processing antigen for cross-priming, even if they do not present that antigen to T cells themselves.
Collapse
Affiliation(s)
- Gwendalyn J Randolph
- Department of Gene and Cell Medicine and the Immunology Institute, Mount Sinai School of Medicine, New York, NY 10029, USA.
| | | | | |
Collapse
|
973
|
Granulocytes do not express but acquire monocyte-derived tissue factor in whole blood: evidence for a direct transfer. Blood 2008; 111:1208-16. [DOI: 10.1182/blood-2007-08-107698] [Citation(s) in RCA: 92] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
Abstract
AbstractUnlike unanimous opinion on tissue factor (TF) expression in monocytes, the quest for TF presence in granulocytes has been going on for decades. To study the cell origin and track the blood-borne TF, we assessed TF activity and protein levels, knocked-down endogenous TF expression with small interfering RNA (siRNA), and overexpressed TF–yellow fluorescent protein (TF-YFP) fusion in immunologically isolated human monocytes and granulocytes. Monocytes and, to a much lesser extent, granulocytes isolated from lipopolysaccharide (LPS)/phorbol 12-myristate-13-acetate (PMA)–stimulated whole blood contained active TF antigen. However, only monocytes possessed significant TF activity and protein levels when stimulated with LPS/PMA in suspension. Reintroduction of TF-silenced monocytes to whole blood led to a profound reduction of LPS/PMA-stimulated TF activity in both mononuclear cell (MNC) and granulocyte fractions. No reduction in TF activity in MNC and granulocyte fractions was observed when TF-silenced granulocytes were reintroduced to whole blood. As shown by immunoblotting, flow cytometry, and confocal microscopy, granulocytes became positive for TF-YFP when isolated from whole blood reconstituted with TF-YFP–expressing monocytes. Together, we pinpoint monocytes as a major source of TF and provide solid experimental evidence for a direct transfer of TF protein from the monocytes to granulocytes in the blood.
Collapse
|
974
|
Kircher MF, Grimm J, Swirski FK, Libby P, Gerszten RE, Allport JR, Weissleder R. Noninvasive in vivo imaging of monocyte trafficking to atherosclerotic lesions. Circulation 2008; 117:388-95. [PMID: 18172031 DOI: 10.1161/circulationaha.107.719765] [Citation(s) in RCA: 93] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND Monocytes play a key role in atherogenesis, but their participation has been discerned largely via ex vivo analyses of atherosclerotic lesions. We sought to establish a noninvasive technique to determine monocyte trafficking to atherosclerotic lesions in live animals. METHODS AND RESULTS Using a micro-single-photon emission computed tomography small-animal imaging system and a Food and Drug Administration-approved radiotracer ([indium 111] oxyquinoline, (111)In-oxine), we demonstrate here that monocyte recruitment to atherosclerotic lesions can be visualized in a noninvasive, dynamic, and 3-dimensional fashion in live animals. We show in vivo that monocytes are recruited avidly to plaques within days of adoptive transfer. Using micro-single-photon emission computed tomography imaging as a screening tool, we were able to investigate modulatory effects on monocyte recruitment in live animals. We found that 3-hydroxy-3-methylglutaryl coenzyme A reductase inhibitors rapidly and substantially reduce monocyte recruitment to existing atherosclerotic lesions, as imaged here in vivo. CONCLUSIONS This novel approach to track monocytes to atherosclerotic plaques in vivo should have broad applications and create new insights into the pathogenesis of atherosclerosis and other inflammatory diseases.
Collapse
Affiliation(s)
- Moritz F Kircher
- Center for Molecular Imaging Research, Massachusetts General Hospital and Harvard Medical School, Charlestown, Mass, USA
| | | | | | | | | | | | | |
Collapse
|
975
|
Packard RRS, Libby P. Inflammation in atherosclerosis: from vascular biology to biomarker discovery and risk prediction. Clin Chem 2008; 54:24-38. [PMID: 18160725 DOI: 10.1373/clinchem.2007.097360] [Citation(s) in RCA: 607] [Impact Index Per Article: 37.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Recent investigations of atherosclerosis have focused on inflammation, providing new insight into mechanisms of disease. Inflammatory cytokines involved in vascular inflammation stimulate the generation of endothelial adhesion molecules, proteases, and other mediators, which may enter the circulation in soluble form. These primary cytokines also induce production of the messenger cytokine interleukin-6, which stimulates the liver to increase production of acute-phase reactants such as C-reactive protein. In addition, platelets and adipose tissue can generate inflammatory mediators relevant to atherothrombosis. Despite the irreplaceable utility of plasma lipid profiles in assessment of atherosclerotic risk, these profiles provide an incomplete picture. Indeed, many cardiovascular events occur in individuals with plasma cholesterol concentrations below the National Cholesterol Education Program thresholds of 200 mg/dL for total cholesterol and 130 mg/dL for low-density lipoprotein (LDL) cholesterol. The concept of the involvement of inflammation in atherosclerosis has spurred the discovery and adoption of inflammatory biomarkers for cardiovascular risk prediction. C-reactive protein is currently the best validated inflammatory biomarker; in addition, soluble CD40 ligand, adiponectin, interleukin 18, and matrix metalloproteinase 9 may provide additional information for cardiovascular risk stratification and prediction. This review retraces the biology of atherothrombosis and the evidence supporting the role of inflammatory biomarkers in predicting primary cardiovascular events in this biologic context.
Collapse
Affiliation(s)
- René R S Packard
- Leducq Center for Cardiovascular Research, Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, 77 Avenue Louis Pasteur, Boston, MA 02115, USA
| | | |
Collapse
|
976
|
Saederup N, Chan L, Lira SA, Charo IF. Fractalkine deficiency markedly reduces macrophage accumulation and atherosclerotic lesion formation in CCR2-/- mice: evidence for independent chemokine functions in atherogenesis. Circulation 2007; 117:1642-8. [PMID: 18165355 DOI: 10.1161/circulationaha.107.743872] [Citation(s) in RCA: 175] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
BACKGROUND Monocyte-derived foam cells are the hallmark of early atherosclerosis, and recent evidence indicates that chemokines play important roles in directing monocyte migration from the blood to the vessel wall. Genetic deletions of monocyte chemoattractant protein-1 (MCP-1, CCL2), fractalkine (CX3CL1), or their cognate receptors, CCR2 and CX3CR1, markedly reduce atherosclerotic lesion size in murine models of atherosclerosis. The aim of this study was to determine whether these 2 chemokines act independently or redundantly in promoting atherogenesis. METHODS AND RESULTS We crossed CX3CL1(-/-)ApoE(-/-) and CCR2(-/-)ApoE(-/-) mice to create CX3CL1(-/-)CCR2(-/-)ApoE(-/-) triple knockouts and performed a 4-arm atherosclerosis study. Here, we report that deletion of CX3CL1 in CCR2(-/-) mice dramatically reduced macrophage accumulation in the artery wall and the subsequent development of atherosclerosis. Deletion of CX3CL1 did not reduce the number of circulating monocytes in either "wild-type" ApoE(-/-) mice or CCR2(-/-)ApoE(-/-) mice, which suggests a role for CX3CL1 in the direct recruitment and/or capture of CCR2-deficient monocytes. CONCLUSIONS These data provide the first in vivo evidence for independent roles for CCR2 and CX3CL1 in macrophage accumulation and atherosclerotic lesion formation and suggest that successful therapeutic strategies may need to target multiple chemokines or chemokine receptors.
Collapse
Affiliation(s)
- Noah Saederup
- Gladstone Institute of Cardiovascular Disease, 1650 Owens St, San Francisco, CA 94158, USA
| | | | | | | |
Collapse
|
977
|
Ritchlin C. Psoriatic disease--from skin to bone. ACTA ACUST UNITED AC 2007; 3:698-706. [PMID: 18037929 DOI: 10.1038/ncprheum0670] [Citation(s) in RCA: 64] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2007] [Accepted: 09/24/2007] [Indexed: 01/18/2023]
Abstract
Psoriatic arthritis is an inflammatory joint disease that is heterogeneous in presentation and clinical course. Evidence that this disease is distinct from rheumatoid arthritis and other spondyloarthropathies is based on data derived from characteristic clinical features, histopathologic analyses, immunogenetic associations and musculoskeletal imaging. Emphasis has centered previously on a dominant role for the T lymphocyte in the inflammatory process; however, studies provide support for a major contribution from monocyte-macrophages in the initiation and perpetuation of joint and skin inflammation. The occurrence of arthritis in the absence of psoriasis in a minority of patients with psoriatic arthritis, coupled with divergent genetic risk factors, indicates that psoriatic arthritis is distinct from psoriatic skin inflammation. A new terminology, psoriatic disease, has emerged that encompasses the various manifestations of tissue and organ involvement observed in many psoriasis patients, including inflammation in the joint, eye and gut. Moreover, adverse cardiovascular and metabolic outcomes in patients with psoriasis or psoriatic arthritis might be directly linked to the cutaneous and musculoskeletal manifestations of these diseases via subsets of circulating monocytes and tissue macrophages activated by inflammatory cytokine networks that arise in the skin and possibly the joint.
Collapse
Affiliation(s)
- Christopher Ritchlin
- University of Rochester School of Medicine and Dentistry, 601 Elmwood Avenue, Box 695, Rochester, NY 14642, USA.
| |
Collapse
|
978
|
Nofer JR. High-density lipoprotein, sphingosine 1-phosphate, and atherosclerosis. J Clin Lipidol 2007; 2:4-11. [PMID: 21291709 DOI: 10.1016/j.jacl.2007.11.002] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2007] [Accepted: 11/26/2007] [Indexed: 11/27/2022]
Abstract
Numerous epidemiologic and interventional studies have revealed an inverse relationship between plasma concentrations of high-density lipoprotein (HDL) and coronary risk. There are several well-documented HDL functions, which may account for the antiatherogenic effects of this lipoprotein. Recent studies document that HDL serves as a carrier for the bioactive lysosphingolipid sphingosine 1-phosphate (S1P), which determines its functional properties. Generally available databases (eg, PubMed) were used, as well as our own results. An increasing body of evidence indicates that S1P is a mediator of many of the atheroprotective effects of HDL, including the ability to promote vasodilation and angiogenesis and protection against ischemia/reperfusion injury. These latter effects are believed to involve S1P-mediated retardation or suppression of inflammatory processes, such as endothelial expression of adhesion molecules, production of proinflammatory chemokines and cytokines, generation of reactive oxygen species, and cardiomyocyte apoptosis after myocardial infarction. This review article summarizes the evidence that S1P is a component of HDL contributing to the antiatherogenic and cardioprotective potential attributed to this lipoprotein.
Collapse
Affiliation(s)
- Jerzy-Roch Nofer
- Center for Laboratory Medicine, University Hospital Münster, and Leibniz Institute for Arteriosclerosis Research, University of Münster, Albert Schweizer Str. 33, D-48129 Münster, Germany
| |
Collapse
|
979
|
Nahrendorf M, Swirski FK, Aikawa E, Stangenberg L, Wurdinger T, Figueiredo JL, Libby P, Weissleder R, Pittet MJ. The healing myocardium sequentially mobilizes two monocyte subsets with divergent and complementary functions. ACTA ACUST UNITED AC 2007; 204:3037-47. [PMID: 18025128 PMCID: PMC2118517 DOI: 10.1084/jem.20070885] [Citation(s) in RCA: 1710] [Impact Index Per Article: 100.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Healing of myocardial infarction (MI) requires monocytes/macrophages. These mononuclear phagocytes likely degrade released macromolecules and aid in scavenging of dead cardiomyocytes, while mediating aspects of granulation tissue formation and remodeling. The mechanisms that orchestrate such divergent functions remain unknown. In view of the heightened appreciation of the heterogeneity of circulating monocytes, we investigated whether distinct monocyte subsets contribute in specific ways to myocardial ischemic injury in mouse MI. We identify two distinct phases of monocyte participation after MI and propose a model that reconciles the divergent properties of these cells in healing. Infarcted hearts modulate their chemokine expression profile over time, and they sequentially and actively recruit Ly-6Chi and -6Clo monocytes via CCR2 and CX3CR1, respectively. Ly-6Chi monocytes dominate early (phase I) and exhibit phagocytic, proteolytic, and inflammatory functions. Ly-6Clo monocytes dominate later (phase II), have attenuated inflammatory properties, and express vascular–endothelial growth factor. Consequently, Ly-6Chi monocytes digest damaged tissue, whereas Ly-6Clo monocytes promote healing via myofibroblast accumulation, angiogenesis, and deposition of collagen. MI in atherosclerotic mice with chronic Ly-6Chi monocytosis results in impaired healing, underscoring the need for a balanced and coordinated response. These observations provide novel mechanistic insights into the cellular and molecular events that regulate the response to ischemic injury and identify new therapeutic targets that can influence healing and ventricular remodeling after MI.
Collapse
Affiliation(s)
- Matthias Nahrendorf
- Center for Systems Biology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA 02129, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
980
|
Microglia in the adult brain arise from Ly-6ChiCCR2+ monocytes only under defined host conditions. Nat Neurosci 2007; 10:1544-53. [PMID: 18026096 DOI: 10.1038/nn2015] [Citation(s) in RCA: 822] [Impact Index Per Article: 48.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2007] [Accepted: 10/25/2007] [Indexed: 12/24/2022]
Abstract
Microglia are crucially important myeloid cells in the CNS and constitute the first immunological barrier against pathogens and environmental insults. The factors controlling microglia recruitment from the blood remain elusive and the direct circulating microglia precursor has not yet been identified in vivo. Using a panel of bone marrow chimeric and adoptive transfer experiments, we found that circulating Ly-6C(hi)CCR2(+) monocytes were preferentially recruited to the lesioned brain and differentiated into microglia. Notably, microglia engraftment in CNS pathologies, which are not associated with overt blood-brain barrier disruption, required previous conditioning of brain (for example, by direct tissue irradiation). Our results identify Ly-6C(hi)CCR2(+) monocytes as direct precursors of microglia in the adult brain and establish the importance of local factors in the adult CNS for microglia engraftment.
Collapse
|
981
|
Zernecke A, Bot I, Djalali-Talab Y, Shagdarsuren E, Bidzhekov K, Meiler S, Krohn R, Schober A, Sperandio M, Soehnlein O, Bornemann J, Tacke F, Biessen EA, Weber C. Protective role of CXC receptor 4/CXC ligand 12 unveils the importance of neutrophils in atherosclerosis. Circ Res 2007; 102:209-17. [PMID: 17991882 DOI: 10.1161/circresaha.107.160697] [Citation(s) in RCA: 304] [Impact Index Per Article: 17.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
The CXC ligand (CXCL)12/CXC receptor (CXCR)4 chemokine-receptor axis controls hematopoiesis, organ development, and angiogenesis, but its role in the inflammatory pathogenesis of atherosclerosis is unknown. Here we show that interference with Cxcl12/Cxcr4 by a small-molecule antagonist, genetic Cxcr4 deficiency, or lentiviral transduction with Cxcr4 degrakine in bone marrow chimeras aggravated diet-induced atherosclerosis in apolipoprotein E-deficient (Apoe-/-) or LDL receptor-deficient (Ldlr-/-) mice. Chronic blockade of Cxcr4 caused leukocytosis and an expansion of neutrophils and increased neutrophil content in plaques, associated with apoptosis and a proinflammatory phenotype. Whereas circulating neutrophils were recruited to atherosclerotic lesions, depletion of neutrophils reduced plaque formation and prevented its exacerbation after blocking Cxcr4. Disrupting Cxcl12/Cxcr4 thus promotes lesion formation through deranged neutrophil homeostasis, indicating that Cxcl12/Cxcr4 controls the important contribution of neutrophils to atherogenesis in mice.
Collapse
Affiliation(s)
- Alma Zernecke
- Institute for Molecular Cardiovascular Research, Rheinisch-Westfälische Technische Hochschule, Aachen University, Germany
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
982
|
Klingenberg R, Nofer JR, Rudling M, Bea F, Blessing E, Preusch M, Grone HJ, Katus HA, Hansson GK, Dengler TJ. Sphingosine-1-Phosphate Analogue FTY720 Causes Lymphocyte Redistribution and Hypercholesterolemia in ApoE-Deficient Mice. Arterioscler Thromb Vasc Biol 2007; 27:2392-9. [PMID: 17761943 DOI: 10.1161/atvbaha.107.149476] [Citation(s) in RCA: 56] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Objective—
Resident immune cells are a hallmark of atherosclerotic lesions. The sphingolipid analogue drug FTY720 mediates retrafficking of immune cells and inhibits their homing to inflammatory sites. We have evaluated the effect of FTY720 on atherogenesis and lipid metabolism.
Methods and Results—
ApoE
−/−
mice on a normal laboratory diet received oral FTY720 for 12 weeks, which led to a 2.4-fold increase in serum cholesterol (largely VLDL fraction) and a 1.8-fold increase in hepatic HMGCoA reductase mRNA. FTY720 increased plasma sphingosine-1-phosphate and induced marked peripheral blood lymphopenia. A discoordinate modulation of B, T and monocyte cell numbers was found in peripheral lymphoid organs. Overall depletion of T cells was accompanied by a relative (2-fold) increase in regulatory T cell content paralleled by a similar increase in effector memory T cells (CD4+CD44hiCD62lo) as absolute numbers of both subpopulations remained essentially unchanged. Lymphocyte function was unaltered as indicated by anti-OxLDL antibodies and T cell proliferation. There were no changes in atherosclerotic lesions in early and established atherosclerosis.
Conclusions—
FTY720 mediated peripheral lymphocyte depletion and retrafficking without altering function and overall balance of pro- and antiatherogenic lymphocyte populations. A net decrease in lymphocyte numbers occurred concomitantly with a more proatherogenic hypercholesterolemia resulting in unaltered atherogenesis.
Collapse
|
983
|
Schneider JG, Zhu Y, Coleman T, Semenkovich CF. Macrophage beta3 integrin suppresses hyperlipidemia-induced inflammation by modulating TNFalpha expression. Arterioscler Thromb Vasc Biol 2007; 27:2699-706. [PMID: 17951320 DOI: 10.1161/atvbaha.107.153650] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
OBJECTIVE High-fat, cholesterol-containing diets contribute to hyperlipidemia. Both high-fat diets and hyperlipidemia are associated with chronic inflammatory diseases like atherosclerosis. Integrins, heterodimeric mediators of inflammatory cell recruitment, are not generally thought to be affected by diet. However, high-fat feeding promotes inflammation, atherosclerosis, and death in hyperlipidemic mice with beta3 integrin deficiency, and treatment of humans from Western populations with oral beta3 integrin inhibitors increases mortality. The mechanisms responsible for these beta3 integrin-associated events are unknown. METHODS AND RESULTS Here we show that diet-induced death in beta3 integrin-deficient mice is a TNFalpha-dependent process mediated by bone marrow-derived cells. In 2 different hyperlipidemic models, apoE-null and LDL receptor-null mice, beta3-replete animals transplanted with beta3-deficient marrow died with Western-type high-fat feeding whereas beta3-deficient animals transplanted with beta3-replete marrow were rescued from diet-induced death. Transplantation with beta3-deficient marrow also increased atherosclerosis. TNFalpha [corrected] expression was increased in beta3-deficient macrophages and normalized by either retroviral or adenoviral reconstitution of beta3 integrin expression. Treatment with the anti-TNFalpha antibody infliximab rescued beta3 integrin-deficient mice from Western diet-induced death, directly implicating TNFalpha in the pathophysiology triggered by diet-induced hyperlipidemia. CONCLUSIONS These findings suggest that macrophage beta3 integrin, acting through TNFalpha, suppresses inflammation caused by hyperlipidemia attributable to high-fat feeding.
Collapse
Affiliation(s)
- Jochen G Schneider
- Campus Box 8127, Washington University School of Medicine, 660 South Euclid Avenue, St. Louis, Missouri 63110, USA
| | | | | | | |
Collapse
|
984
|
Rao RM, Yang L, Garcia-Cardena G, Luscinskas FW. Endothelial-dependent mechanisms of leukocyte recruitment to the vascular wall. Circ Res 2007; 101:234-47. [PMID: 17673684 DOI: 10.1161/circresaha.107.151860b] [Citation(s) in RCA: 292] [Impact Index Per Article: 17.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Inflammation is a fundamental process that protects organisms by removing or neutralizing injurious agents. A key event in the inflammatory response is the localized recruitment of various leukocyte subsets. Here we address the cellular and regulatory mechanisms of leukocyte recruitment to the vessel wall in cardiovascular disease and discuss our evolving understanding of the role of the vascular endothelium in this process. The vascular endothelium is the continuous single-cell lining of the cardiovascular system that forms a critical interface between the blood and its components on one side and the tissues and organs on the other. It is heterogeneous and has many synthetic and metabolic functions including secretion of platelet-derived growth factor, von Willebrand factor, prostacyclin, NO, endothelin-1, and chemokines and the expression of adhesion molecules. It also acts as a nonthrombogenic and selective permeable barrier. Endothelial cells also interact closely with the extracellular matrix and with adjacent cells including pericytes and smooth muscle cells within the vessel wall. A central question in vascular biology is the role of the endothelium in the initiation of inflammatory response, the extent of its "molecular conversations" with recruited leukocytes, and its influence on the extent and/or outcome of this response.
Collapse
Affiliation(s)
- Ravi M Rao
- Vascular Science, National Heart and Lung Institute, Imperial College School of Medicine, Hammersmith Hospital, London, UK
| | | | | | | |
Collapse
|
985
|
Bellini A, Mattoli S. The role of the fibrocyte, a bone marrow-derived mesenchymal progenitor, in reactive and reparative fibroses. J Transl Med 2007; 87:858-70. [PMID: 17607298 DOI: 10.1038/labinvest.3700654] [Citation(s) in RCA: 317] [Impact Index Per Article: 18.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Human fibrocytes are mesenchymal progenitors that exhibit mixed morphological and molecular characteristics of hematopoietic stem cells, monocytes and fibroblasts. They likely represent the obligate intermediate stage of differentiation into mature mesenchymal cells of a bone marrow-derived precursor of the monocyte lineage under permissive conditions. On in vitro stimulation with pro-fibrotic cytokines and growth factors, human fibrocytes produce large quantities of extracellular matrix components and further differentiate into cells identical to the contractile myofibroblasts that emerge at the tissue sites during repair processes and in some fibrotic lesions. Studies in various animal models of wound healing or fibrotic diseases have confirmed the ability of fibrocytes to differentiate into mature mesenchymal cells in vivo and have suggested a causal link between fibrocyte accumulation and ongoing tissue fibrogenesis or vascular remodeling in response to tissue damage or hypoxia. Fibrocytes synthesizing new collagen or acquiring myofibroblast markers have been detected in human hypertrophic scars, in the skin of patients affected by nephrogenic systemic fibrosis, in human atherosclerotic lesions, and in pulmonary diseases characterized by repeated cycles of inflammation and repair, like asthma. The presence of fibrocyte-like cells has been reported in human chronic pancreatitis and chronic cystitis. Similar cells also populate the stroma surrounding human benign tumors. The available data indicate that human fibrocytes serve as a source of mature mesenchymal cells during reparative processes and in fibrotic disorders or stromal reactions predominantly associated with a persistent inflammatory infiltrate or with the selective recruitment of monocytes induced by ischemic changes and tumor development. A deeper understanding of the mechanisms involved in fibrocyte differentiation in these pathological conditions may lead to the development of novel therapies for preventing detrimental tissue or vascular remodeling and metastatic progression of invasive tumors.
Collapse
|
986
|
Abstract
Numerous reports document the role of vascular adhesion molecules in the development and progression of atherosclerosis. Recent novel findings in the field of adhesion molecules require an updated summary of current research. In this review, we highlight the role of vascular adhesion molecules including selectins, vascular cell adhesion molecule (VCAM)-1, intercellular adhesion molecule1 (ICAM-1), PECAM-1, JAMs, and connexins in atherosclerosis. The immune system is important in atherosclerosis, and significant efforts are under way to understand the vascular adhesion molecule-dependent mechanisms of immune cell trafficking into healthy and atherosclerosis-prone arterial walls. This review focuses on the role of vascular adhesion molecules in the regulation of immune cell homing during atherosclerosis and discusses future directions that will lead to better understanding of this disease.
Collapse
Affiliation(s)
- Elena Galkina
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA 22908, USA
| | | |
Collapse
|
987
|
Bouhlel MA, Derudas B, Rigamonti E, Dièvart R, Brozek J, Haulon S, Zawadzki C, Jude B, Torpier G, Marx N, Staels B, Chinetti-Gbaguidi G. PPARgamma activation primes human monocytes into alternative M2 macrophages with anti-inflammatory properties. Cell Metab 2007; 6:137-43. [PMID: 17681149 DOI: 10.1016/j.cmet.2007.06.010] [Citation(s) in RCA: 1028] [Impact Index Per Article: 60.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/09/2007] [Revised: 06/13/2007] [Accepted: 06/27/2007] [Indexed: 01/03/2023]
Abstract
Th1 cytokines promote monocyte differentiation into proatherogenic M1 macrophages, while Th2 cytokines lead to an "alternative" anti-inflammatory M2 macrophage phenotype. Here we show that in human atherosclerotic lesions, the expression of M2 markers and PPARgamma, a nuclear receptor controlling macrophage inflammation, correlate positively. Moreover, PPARgamma activation primes primary human monocytes into M2 differentiation, resulting in a more pronounced anti-inflammatory activity in M1 macrophages. However, PPARgamma activation does not influence M2 marker expression in resting or M1 macrophages, nor does PPARgamma agonist treatment influence the expression of M2 markers in atherosclerotic lesions, indicating that only native monocytes can be primed by PPARgamma activation to an enhanced M2 phenotype. Furthermore, PPARgamma activation significantly increases expression of the M2 marker MR in circulating peripheral blood mononuclear cells. These data demonstrate that PPARgamma activation skews human monocytes toward an anti-inflammatory M2 phenotype.
Collapse
Affiliation(s)
- M Amine Bouhlel
- Institut Pasteur de Lille, F-59019 Lille, France; Inserm, U545, F-59019 Lille, France
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
988
|
|
989
|
Yamashita T, Kawashima S, Hirase T, Shinohara M, Takaya T, Sasaki N, Takeda M, Tawa H, Inoue N, Hirata KI, Yokoyama M. Xenogenic macrophage immunization reduces atherosclerosis in apolipoprotein E knockout mice. Am J Physiol Cell Physiol 2007; 293:C865-73. [PMID: 17553935 DOI: 10.1152/ajpcell.00117.2007] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Atherosclerosis is a complex chronic inflammatory disease in which macrophages play a critical role, and the intervention of the inflammatory process in atherogenesis could be a therapeutic strategy. In this study, we investigated the efficacy of xenogenic macrophage immunization on the atherosclerotic lesion formation in a model of murine atherosclerosis. Apolipoprotein E knockout (apoE-KO) mice were repeatedly immunized with formaldehyde-fixed cultured human macrophages (phorbol ester-stimulated THP-1 cells), using human serum albumin as a control protein or HepG2 cells as human control cells, once a week for four consecutive weeks. The vehicle phosphate-buffered saline was injected in the nonimmunized controls. THP-1 immunization induced antibodies that are immunoreactive with mouse macrophages. Although the plasma lipid levels were unchanged by the immunization, the atherosclerotic lesion area in the aortic root was significantly reduced by >50% in 16-wk-old THP-1-immunized apoE-KO mice compared with that in control mice. THP-1 immunization reduced in vivo macrophage infiltration, reduced in vitro macrophage adhesion, and changed cytokine production by macrophages to the antiatherogenic phenotype. Xenogenic macrophage immunization protects against the development of atherosclerosis in apoE-KO mice by modulating macrophage function in which antibodies induced by the immunization are likely to be involved. This method is a novel and potentially useful cell-mediated immune therapeutic technique against atherosclerosis.
Collapse
Affiliation(s)
- Tomoya Yamashita
- Division of Cardiovascular Medicine, Dept. of Internal Medicine, Kobe Univ. Graduate School of Medicine, 7-5-1 Kusunoki-cho, Chuo-ku, Kobe 650-0017, Japan.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
990
|
Abstract
We discuss the anatomy, physiology, and pathophysiology of epicardial adipose tissue and its relationship to coronary atherosclerosis. Epicardial fat stores triglyceride to supply free fatty acids for myocardial energy production and produces adipokines. It shares a common embryological origin with mesenteric and omental fat. Like visceral abdominal fat, epicardial fat thickness, measured by echocardiography, is increased in obesity. Epicardial fat could influence coronary atherogenesis and myocardial function because there is no fibrous fascial layer to impede diffusion of free fatty acids and adipokines between it and the underlying vessel wall as well as the myocardium. Segments of coronary arteries lacking epicardial fat or separated from it by a bridge of myocardial tissue are protected against the development of atherosclerosis in those segments. However, when epicardial fat is totally absent in congenital generalized lipodystrophy, coronary atherosclerosis can still occur. Macrophages are more numerous and densely packed in the periadventitial fat of human atherosclerotic coronary arteries with lipid cores than in that of fibrocalcific or nonatherosclerotic coronary arteries. In obese patients with multiple cardiovascular risk factors, epicardial fat around atheromatous coronaries secretes several proinflammatory cytokines and is infiltrated by macrophages, lymphocytes, and basophils. Epicardial adipokine expression in obesity without coronary atherosclerosis has not been determined. In nonobese patients, epicardial fat around atheromatous coronary arteries expresses proinflammatory cytokines but produces either less adiponectin, a vasoprotective adipokine, than fat around nonatheromatous coronaries or a similar amount compared with thoracic subcutaneous fat. Further studies should be done to test the hypothesis that adipokines produced by and released from human epicardial adipose tissue might contribute locally to the pathogenesis of coronary atherosclerosis.
Collapse
Affiliation(s)
- Harold S Sacks
- Division of Endocrinology and Metabolism, University of Tennessee, and Baptist Hospital Heart Institute, Memphis, TN, USA.
| | | |
Collapse
|
991
|
Arnold L, Henry A, Poron F, Baba-Amer Y, van Rooijen N, Plonquet A, Gherardi RK, Chazaud B. Inflammatory monocytes recruited after skeletal muscle injury switch into antiinflammatory macrophages to support myogenesis. ACTA ACUST UNITED AC 2007; 204:1057-69. [PMID: 17485518 PMCID: PMC2118577 DOI: 10.1084/jem.20070075] [Citation(s) in RCA: 1486] [Impact Index Per Article: 87.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Macrophages (MPs) are important for skeletal muscle regeneration in vivo and may exert beneficial effects on myogenic cell growth through mitogenic and antiapoptotic activities in vitro. However, MPs are highly versatile and may exert various, and even opposite, functions depending on their activation state. We studied monocyte (MO)/MP phenotypes and functions during skeletal muscle repair. Selective labeling of circulating MOs by latex beads in CX3CR1(GFP/+) mice showed that injured muscle recruited only CX3CR1(lo)/Ly-6C(+) MOs from blood that exhibited a nondividing, F4/80(lo), proinflammatory profile. Then, within muscle, these cells switched their phenotype to become proliferating antiinflammatory CX3CR1(hi)/Ly-6C(-) cells that further differentiated into F4/80(hi) MPs. In vitro, phagocytosis of muscle cell debris induced a switch of proinflammatory MPs toward an antiinflammatory phenotype releasing transforming growth factor beta1. In co-cultures, inflammatory MPs stimulated myogenic cell proliferation, whereas antiinflammatory MPs exhibited differentiating activity, assessed by both myogenin expression and fusion into myotubes. Finally, depletion of circulating MOs in CD11b-diphtheria toxin receptor mice at the time of injury totally prevented muscle regeneration, whereas depletion of intramuscular F4/80(hi) MPs at later stages reduced the diameter of regenerating fibers. In conclusion, injured skeletal muscle recruits MOs exhibiting inflammatory profiles that operate phagocytosis and rapidly convert to antiinflammatory MPs that stimulate myogenesis and fiber growth.
Collapse
Affiliation(s)
- Ludovic Arnold
- Institut National de la Santé et de la Recherche Médicale, Unité 841, Institut Mondor de Recherche Biomédicale, Cell Interactions in the Neuromuscular System Team, Université Paris 12 Val-de-Marne, Créteil, France
| | | | | | | | | | | | | | | |
Collapse
|
992
|
Abstract
Macrophages are present as resident cells in adipose tissue, and blood monocytes are recruited in increased numbers to sites of lipid accumulation in atherosclerosis, a modified form of inflammation in the arterial wall. Recent findings reported by 3 separate groups in this issue of the JCI provide evidence for distinct monocyte subsets, differential chemokine receptor usage, and phenotypic modulation of macrophages in murine models of genetic and high-fat diet-induced disease (see the related articles beginning on pages 175, 185, and 195). These studies raise prospects for selective therapeutic targets to ameliorate macrophage hyperinflammatory responses, while sparing host defense and repair mechanisms.
Collapse
Affiliation(s)
- Siamon Gordon
- Sir William Dunn School of Pathology, University of Oxford, Oxford, United Kingdom.
| |
Collapse
|
993
|
Leavy O. Fat gets picky with macrophages. Nat Rev Immunol 2007. [DOI: 10.1038/nri2026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
|