Literature
首页医源资料库在线期刊动脉硬化血栓血管生物学杂志2007年第27卷第7期

Enhanced Immune System Activation and Arterial Inflammation Accelerates Atherosclerosis in Lupus-Prone Mice

来源:《动脉硬化血栓血管生物学杂志》
摘要:EnhancedatherogenesiswasassociatedwithenhancedactivationofbothBandTlymphocytesandwitharterialinflammationinvolvingendothelialcellactivation,monocyterecruitment,andaccumulationofapoptoticdebris,macrophages,andCD4Tcells,butwasindependentofplasmalipidlevelsand......

点击显示 收起

【摘要】  Objective- Premature atherosclerosis is a characteristic feature of systemic lupus erythematosus, a prototypic autoimmune disease. The principle cellular and molecular mechanisms which underlie such accelerated atherosclerosis are indeterminate.

Methods and Results- The pathophysiology of lupus-mediated atherogenesis was evaluated in a novel animal model involving transplantation of bone marrow cells from the lupus prone strain gld into Ldl-r -/- mice. Diet-induced atherogenesis in lethally-irradiated Ldl-r -/- mice transplanted with gld bone marrow cells resulted in accelerated atherosclerosis (+65%) as compared with control mice transplanted with wild-type marrow cells. Enhanced atherogenesis was associated with enhanced activation of both B and T lymphocytes and with arterial inflammation involving endothelial cell activation, monocyte recruitment, and accumulation of apoptotic debris, macrophages, and CD4 T cells, but was independent of plasma lipid levels and renal function.

Conclusions- Our data support the contention that despite the absence of both disturbed cholesterol homeostasis and renal dysfunction in autoimmune gld Ldl-r -/- mice, lupus disease induces enhanced activation of the immune system and acts locally on the vasculature to induce inflammation, together with accumulation of apoptotic debris, macrophages, and CD4 T cells, thereby accelerating plaque progression.

Premature atherosclerosis is a feature of autoimmune disease but the mechanisms that underlie such accelerated atherosclerosis are indeterminate. Our experimental findings highlight enhanced immune system activation, aortic inflammation, and lesional accumulation of apoptotic cells, macrophages, and CD4 T cells as central mechanisms in the progression of atherosclerosis in autoimmune mice.

【关键词】  atherosclerosis lupus arterial inflammation immune system apoptotic cells


Introduction


The risk of cardiovascular disease (CVD) is significantly increased in the prototypic autoimmune disease, systemic lupus erythematosus (SLE). 1,2 The mechanisms underlying premature CVD in SLE may be related to accelerated atherosclerosis. 3,4 Such accelerated atherosclerosis potentially involves a combination of autoimmune-specific mechanisms and traditional cardiovascular risk factors (dyslipidemia, renal failure, and inflammation), although the central mechanisms involved are indeterminate. 2 Moreover, as most SLE patients are on active therapy (corticosteroids and other pharmacological agents), such agents might interfere with the atherogenic process 5 and lead to confounding findings. Therefore, identification of the mechanisms that contribute to disease progression might allow optimization of therapeutic approaches for prevention of CVD in SLE patients. In this setting, development of murine models may facilitate evaluation of the specific impact of autoimmunity on atherosclerosis progression and of the underlying mechanisms involved.


Genetic studies of various strains of lupus-prone mice have identified at least 30 chromosomal regions of interest reflecting the multifactorial aspect of SLE. 6 In this context the use of various strains of lupus-prone mice and the study of their impact on atherogenesis may highlight the key molecular mechanisms that promote autoimmune-accelerated atherosclerosis. Mouse strains defective for the Fas / Fas L pathway ( gld, lpr ) present lupus-like autoimmune disorders comparable to those of human SLE. 6,7 Indeed, these mice are characterized by a deficit in apoptotic cell clearance 8 which induces the production of a repertoire of autoantibodies directed against neoantigens derived from apoptotic cells (nucleosome, dsDNA). In addition, these mice progressively develop renal dysfunction with age. 6,7


When gld mice are crossed with atherosclerosis-susceptible Apoe -/- animals, they develop accelerated atherogenesis associated with defective phagocytosis of apoptotic cells in lymph nodes and glomerulonephritis. 8 These findings suggest that defective apoptotic cell clearance, a feature of autoimmunity, might potentiate atherosclerosis. Impaired clearance of apoptotic cells is equally observed in Apoe -/- mice 9 and could synergize with the gld mutation to exacerbate both autoimmunity and atherogenesis. Moreover, the renal dysfunction observed in gld Apoe -/- mice might potentiate atherosclerosis as renal dysfunction is associated with accelerated atherogenesis in mice and humans. 10,11,12


To further evaluate the relationship between autoimmune disease and accelerated atherogenesis, we generated an atherosclerosis-susceptible autoimmune mouse model by transfer of bone marrow cells from the gld strain to lethally irradiated Ldl-r -/-. In this new murine model, we demonstrated both an enhanced activation of B and T cells and a vascular inflammatory profile that results in accelerated atherosclerosis. Such enhanced atherogenesis was independent of plasma lipid levels and renal dysfunction.


Materials and Methods


Please see the supplemental data section at http://atvb.ahajournals.org for detailed Methods.


Mice, Bone Marrow Transplantation, and Study Design


Ldl-r -/- and gld mice ( FasL -deficient mice) on the C57BL/6J background were obtained from Jackson Laboratories. Males Ldl-r -/- mice (8- to 9-week-old) were subjected to medullar aplasia with 10 Gray lethal total body irradiation. The next day, femurs were isolated from donor gld or wt mice and 2.5 x 10 6 bone marrow cells were injected via the retrorbital vein into the irradiated mice to rescue their hematopoietic systems. Mice were housed in cages under air-filtered conditions for 4 weeks to allow the hematopoietic system to reconstitute, after which they were fed a diet consisting of 0.15% cholesterol and 20% saturated fat (SAFE) for 12 weeks.


Assessment of Chimerism and Analysis of Gene Expression by Q-Polymerase Chain Reaction (PCR)


Real time quantitative PCR was performed using a LightCycler PCR System (Roche) as previously described. 13 The specific primers are described in supplemental Table I available online at http://atvb.ahajournals.org


Plasma Lipid Analyses and Lipoprotein Profile


Blood samples were collected and analyzed as previously described. 13


Flow Cytometry, Antibody Measurements, and Serum Cytokine Levels


Please see the supplemental data section at http://atvb.ahajournals.org for details.


Analysis of Atherosclerotic Plaques, Immunohistochemistry, and Terminal Deoxynucleotidyl Transferase-Mediated dUTP Nick End-Labeling (TUNEL) Staining


Atherosclerotic lesions quantification and Immunohistochemistry were performed as previously described. 13,14 TUNEL staining was performed according to the manufacturer?s instructions (In situ Cell Death Detection Kit, Roche Applied Science)


Statistical Analysis


The statistical significance of the differences between groups was evaluated using the Mann-Whitney U test for unpaired comparisons. P <0.05 was considered significant. Values are expressed as mean±SEM.


Results


Generation of Chimeric Mice With gld or wt Bone Marrow-Derived Cells


Irradiated male Ldl-r -/- mice were reconstituted with bone marrow cells from gld mice (n=18) or wild-type controls (n=14). After 12 weeks of a Western-style diet, Ldl-r -/- mice which had received gld bone marrow cells displayed similar body weight as compared with those receiving wt marrows (21.6±1.0 versus 22.2±1.2 g, respectively). The hematocrit, leukocyte count, and relative levels of T cells, B cells, and monocytes were not significantly different between the two groups (data not shown). The efficacy of transplantation was attested by the significant splenomegaly observed in gld Ldl-r -/-, a feature of the gld model, and by the fact that we detected less than 5% of FasL wt alleles in bone marrow cells from these mice (supplemental Figure I, available online at http://atvb.ahajournals.org). Clearly, the chimerism for FasL in our gld Ldl-r -/- was in the range of 95 to 100%.


Quantification of Atherosclerotic Lesions


As shown in Figure 1 A, the surface of ORO-positive areas in the aortic root of Ldl-r -/- recipients reconstituted with gld marrow cells was significantly greater (+65%) than that observed for their wt reconstituted controls (170454±64391 versus 104134±31709 µm 2 respectively; P <0.0005).


Figure 1. Increased atherosclerosis, apoptotic material deposition, and macrophage accumulation in gld Ldl-r -/- mice as compared with wt Ldl-r -/- mice. A, The degree of atherosclerosis was determined by ORO staining of aortic root sections. Each symbol represents the mean lesion area in a single mouse. The horizontal bar indicates the mean value for the respective group. B, Atherosclerotic lesions were stained by the TUNEL method and the number of apoptotic cells and bodies counted. The ratio of TUNEL-positive cells or bodies to ORO-positive staining was calculated for both groups. Each symbol represents the ratio for a single mouse. C, Atherosclerotic lesions were immunostained for the macrophage CD68 antigen and the degree of macrophage accumulation was determined. Each symbol represents the mean lesion area in a single mouse. Photomicrographs illustrate macrophage abundance in aortic root sections from both groups of mice (magnification x 100). Nuclei are visualized in blue with DAPI staining. *, **, and *** indicate statistically significant differences between the 2 groups: P <0.05, P <0.01, and P <0.0005, respectively.


Plasma Lipid and Lipoprotein Profile


To assess whether the effects of gld marrow cells on atherosclerosis in Ldl-r -/- mice were attributable to changes in lipid or lipoprotein metabolism, we analyzed serum lipids and lipoproteins. Ldl-r -/- mice transplanted with gld marrow cells displayed, as compared with controls, similar plasma total cholesterol (481±67 versus 511±99 mg/dL, respectively), free cholesterol (148±23 versus 150±24 mg/dL, respectively), and triglyceride (206±61 versus 227±64 mg/dL, respectively) levels. Analysis of plasma lipoproteins by gel filtration showed no difference in cholesterol distribution across the VLDL, LDL, and HDL lipoprotein classes between gld Ldl-r -/- and wt Ldl-r -/- mice (data not shown).


Systemic Autoimmunity in gld Ldl-r -/-


The spleen/body weight ratio in gld Ldl-r -/- mice was increased almost 2-fold as compared with wt Ldl-r -/- mice ( Table 1 ). The enhanced lymphoproliferation and autoimmunity seen both in Fas or FasL -deficient mice and Fas mutant human subjects is attributable predominantly to the expansion of an unusual T cell population of CD3 + cells that lacks both CD4 and CD8 (double negative T cells, DNTC). Therefore, we performed quantitative analysis of DNTC in spleen and blood samples from each animal. Significant expansion of DNTC was seen in both spleen and blood of gld Ldl-r -/- animals compared with their wt Ldl-r -/- controls ( Table 1 ). Lupus is associated with the appearance of specific subsets of autoantibodies directed against nuclear materials. Thus, we measured the levels of antibodies directed against double-strand DNA (dsDNA) and nuclear materials (ANA) and showed that they were significantly elevated in gld Ldl-r -/- mice as compared with the wt Ldl-r -/- animals ( Table 1 ). Moreover, autoimmune gld Ldl-r -/- mice presented a global hypergammaglobulinemia as observed by significantly increased serum IgG levels compared with controls. Therefore, these data demonstrated the development of autoimmune disease in gld Ldl-r -/- mice.


TABLE 1. Autoimmune Parameters and Renal Function in wt Ldl-r -/- and gld Ldl-r -/- Mice After 12 Weeks of Diet


Renal Function in Autoimmune Mice


As autoimmune disease is frequently associated with renal dysfunction, histological analysis was performed on renal tissue from both groups of mice. Glomerular cellularity in gld Ldl-r -/- mice was similar to that observed in wt Ldl-r -/- mice (data not shown). Accordingly, urinary protein and serum and urine creatinine levels were unchanged ( Table 1 ). Moreover, at the time of sacrifice, kidney weight to body M r S did not reveal differences between the two groups of mice ( Table 1 ). Collectively, these data argue for the absence of specific renal impairment in autoimmune gld Ldl-r -/- mice.


Systemic Inflammatory Cytokines


As immunoinflammatory cytokines play a significant role in atherosclerosis progression, we performed an exhaustive analysis of systemic cytokine profile. Serum levels of cytokines including interleukin (IL)-12, IFN, IL-1ß, IL-2, IL-4, and IL-6 were significantly decreased (up to 9-fold) in gld Ldl-r -/- mice as compared with wt Ldl-r -/- mice (supplemental Table II). No significant changes were observed for IL-10, IL-1, GM-colony stimulating factor (CSF), and tumor necrosis factor (TNF)- between the two groups. This particular profile observed in gld Ldl-r -/- mice was not associated with changes in IL-12p35, IL-1ß, IL-4, nor in TNF- mRNA levels in the spleen of these mice as compared with wt Ldl-r -/- mice (data not shown), thereby indicating that tissues other than spleen might contribute to the circulating cytokine pool.


Anti-MDA LDL Antibody Levels, IgG2a/IgG1 Ratio, and B Cell Activation in Autoimmune Mice


As levels of antibodies directed against oxidized-LDL are correlated with lesion size, 15 we sought to associate the serum titers of anti-MDA-LDL antibodies with the increased extent of lesions in gld Ldl-r -/- mice. Thus, we observed significantly elevated titers of IgG and IgM to MDA-LDL in gld Ldl-r -/- mice ( Figure 2A and 2 B, respectively). Next, as an indicator of Th1 versus Th2 polarization, we measured the serum IgG2a to IgG1 ratio. As this ratio is similar in wt Ldl-r -/- and gld Ldl-r -/- mice, it revealed no bias toward a Th1 or a Th2 response ( Figure 2 C). Finally, B cell activation was revealed by a significant increase in the proportion of CD86-positive cells among the B cell population of gld Ldl-r -/- mice as compared with controls ( Figure 2 D).


Figure 2. Increased anti-MDA-LDL antibody levels and B cell activation in autoimmune mice. Anti-MDA-LDL IgG (A) and IgM (B) levels were determined by ELISA in the serum of both wt Ldl-r -/- and gld Ldl-r -/- mice. The horizontal bar indicates the mean value from 12 animals per group. C, The IgG1 and IgG2a antibody subclass levels were determined by ELISA and the IgG1/IgG2a ratio is shown. Values represent the mean±SEM of 12 animals analyzed per group. D, Expression of the activation marker CD86 was determined on CD19-positive B cells by flow cytometry. *, **, and *** indicate statistically significant differences between the two groups: P <0.05, P <0.005, and P <0.0001, respectively.


Activation of CD4 T Cells and Accumulation in Atherosclerotic Plaques


CD4-positive T cells are key players in both atherosclerosis and autoimmunity, and therefore we analyzed both their activation status and presence in atherosclerotic plaques. Analysis of splenic CD4 T cells demonstrated that gld Ldl-r -/- mice displayed increased proportions of both activated CD69-positive T cells ( Figure 3 A, P <0.005) and memory CD44-positive T cells ( Figure 3 B, P <0.005); by contrast there was no change in regulatory CD25/Foxp3 double positive T cells (12.1±1.4 in gld Ldl-r -/- mice versus 11.0±1.5 in wt Ldl-r -/- control mice). The increase in the activated and memory phenotype of CD4 T cells observed in lupus-prone mice was associated with their increased accumulation within the lesions ( Figure 3 C, P <0.05).


Figure 3. CD4 T cell activation and accumulation in atherosclerotic lesions. A, Expression of the activation marker CD69 was determined on CD4-positive T cells by flow cytometry. B, Expression of the memory marker CD44 was determined on CD4-positive T cells by flow cytometry. C, Atherosclerotic lesions were immunostained for the lymphocyte T CD4 antigen and the number of CD4 T cells per mm 2 was determined. Values represent the mean±SEM of 9 animals analyzed per group. Photomicrographs illustrate CD4 T cell abundance in aortic root sections from both groups of mice (magnification x 100). Nuclei are visualized in blue by DAPI staining. * and ** indicate statistically significant differences between the 2 groups: P <0.05 and P <0.0001, respectively.


Apoptotic Structures in Atherosclerotic Lesions


As defective apoptotic cell clearance is observed in lymphoid organs of autoimmune mice, we quantified the number of cells in terminal stages of apoptosis in the aortic sinus of both groups of mice. Late stage apoptotic cells and apoptotic bodies were stained by fluorescein isothiocyanate (FITC)-dUTP TUNEL whereas nuclei were counterstained with DAPI. Discrimination of apoptotic cells and apoptotic bodies by DAPI staining was based on nuclear size and morphology (normal or fragmented nuclei respectively). The frequencies of apoptotic cells and apoptotic bodies in aortic tissue were 15-fold and 3-fold higher in gld Ldl-r -/- mice as compared with wt Ldl-r -/- animals respectively ( Figure 1 B, P <0.05).


Macrophage Accumulation and Recruitment of Monocytes in Atherosclerotic Lesions.


Aortic root sections from mice on the Western diet were stained with a macrophage-specific antibody directed against CD68. Quantitative analysis revealed a significant 2-fold increase in CD68-staining lesions of gld Ldl-r -/- mice as compared with wt Ldl-r -/- controls ( Figure 1 C, P <0.01). Macrophages were abundant in the subendothelial space of gld Ldl-r -/- mice and constituted the majority of the cells in the lesion ( Figure 1 C). To further document macrophage accumulation in atherosclerotic lesions, we extracted mRNA from descending aortas of mice to perform quantitative real-time PCR. Levels of mRNA for CD68 and F4/80 were significantly elevated (2- and 4-fold respectively) in the arch and abdominal areas of the aorta in gld Ldl-r -/- mice as compared with wt Ldl-r -/- controls ( Table 2 ). To investigate the potential molecular mechanisms underlying macrophage accumulation, we next evaluated expression of adhesion molecules and chemokines in the descending aorta. As shown in Table 2, the expression levels of MCP-1, P-selectin, and intercellular adhesion molecule-1 (ICAM-1) mRNA were significantly increased (3.5-, 1.5-, and 2.5-fold, respectively) in gld Ldl-r -/- mice as compared with their wt Ldl-r -/- controls, while the levels of vascular cell adhesion molecule (VCAM)-1 and CX3CL1 mRNA were comparable in both groups ( Table 2 ). Concomitantly, we observed that expression of the monocyte-associated genes CD115, CD11b, and CCR2 was elevated (1.5-, 2.5-, and 4-fold, respectively) in gld Ldl-r -/- aortas as compared with their wt Ldl-r -/- controls, whereas CX3CR1 expression was also increased but did not reach statistical significance ( Table 2 ).


TABLE 2. Gene Expression Profile in the Arterial Wall of wt Ldl-r -/- and gld Ldl-r -/- Mice After 12 Weeks of Diet


Discussion


We have developed a new animal model susceptible to concomitant development of SLE and atherogenesis. SLE prone-mice exhibited accelerated progression of atherosclerosis, although plasma lipid levels, lipoprotein-cholesterol distribution, and renal function were similar to wild-type controls. Aortic lesions in autoimmune mice were characterized by accumulation of apoptotic cells, consistent with the defective apoptotic cell clearance associated with SLE. 16 In addition, enhanced immune system activation, arterial inflammation, and recruitment of both lymphocytes and monocytes were prominent aspects of accelerated atherosclerosis in gld Ldl-r -/- lupus-prone animals. These data extend our comprehension of the mechanisms that can contribute to accelerated atherosclerosis in lupus patients as observed in prospective cohorts. 3,4


Chronic renal dysfunction associated with glomerulonephritis develops in the course of lupus. 8,17 In addition, several studies have provided evidence that renal dysfunction promotes atherosclerosis in mice, 10,11 but equally represents an independent risk factor for atherogenesis in man. 12 However, as we did not observe renal dysfunction within the time frame of our study (creatinine levels and proteinuria were unchanged in gld Ldl-r -/- mice), this mechanism does not appear to account for the accelerated atherosclerosis observed in our murine model.


Another prominent feature of autoimmunity in SLE patients concerns abnormal T and B-cell responses manifested by their activation status and the dysregulation in systemic cytokine profile. 18 In our mouse SLE model, serum levels of IL-12, IFN- (Th1 cytokines), IL-4 (Th2 cytokine), IL-1ß, IL-6 (inflammatory cytokines), and the T-cell growth factor IL-2 were significantly decreased whereas those of IL-10, TNF-, and GM-CSF remained unchanged. These findings are consistent with clinical investigations in SLE patients, in which attenuated expression of IL-12, 19,20,21 IFN-, 19,22 IL-4, 19 and IL-2 22 have been documented. Similarly, in the lupus-prone autoimmune strain NZB/WF1, IL-12, IL-1, IL-2, IL-4, and IL-6 cytokine levels were decreased, whereas those of the antiinflammatory cytokine IL-10 and the proinflammatory cytokine TNF- 18,23,24 were unchanged as equally observed in our model. Overall, the impact of reduced cytokine levels (ie, Th1 and inflammatory cytokines) might appear primarily antiatherogenic. Indeed, in mouse models of atherosclerosis, substantial data support a proatherogenic role for IFN-, 25,26 IL-12, 27,28 IL-4, 27,29 and IL-1ß 30, whereas IL-10 plays an antiatherogenic role. 31


Surprisingly, we observed an enhanced activation of the immune system consistent with increased B and T cell activation, increased memory phenotype of CD4 T cells, and concomitant elevation of anti-MDA-LDL IgG and IgM levels in gld Ldl-r -/- mice as compared with controls. Similar findings concerning T cell activation were reported in a recent study based on transplantation of bone marrow cells from congenic mice expressing a susceptibility locus for SLE in Ldl-r -/- mice. 17 In addition, it is important to note that the proportion of atheroprotective type 1 regulatory T cells did not differ between gld Ldl-r -/- and wt Ldl-r -/- mice. In our model, despite lower proinflammatory cytokine levels, we report higher activation of both B and T cells. These results are not contradictory as a diminished threshold of activation for both T and B cells could favor the onset of autoimmunity. These defects appear to be key players in the accelerated atherosclerosis typical of lupus. In this setting, the elevated degree of CD4 T cell activation, and memory status in gld Ldl-r -/- mice, which is associated with increased lesional T cell numbers could accelerate atherogenesis and is consistent with a highly inflammatory plaque phenotype.


Marked accumulation of apoptotic cells and bodies occurred in aortic sinus tissue of gld Ldl-r -/- mice, thereby corroborating the observation that increased amounts of apoptotic material were deposited in the lymph nodes of gld Apoe -/- mice. 8 Several studies have demonstrated that the impaired clearance of apoptotic cells by macrophages is a feature of SLE, 16 and that a defective complement system may play an active role in this process. 32 However, we did not observe decreased mRNA expression of the main complement components C1q, C2, C3, and C4 in the liver of gld Ldl-r -/- animals (data not shown). Additionally, the observed elevation in titers of autoantibodies to dsDNA and ANA directed against nuclear material suggest that elevated levels of circulating nucleosomes may occur in gld Ldl-r -/- mice. Such nucleosomes may partly account for defective apoptotic cell clearance as suggested by Laderach and colleagues. 33 In this context, the accumulation of TUNEL-positive bodies in the aortic sinus is indicative of an ineffective clearance of primary apoptotic cells, ie, cells exposing phosphatidylserine in the outer plasma membrane leaflet, rather than the result of increased rates of apoptosis. Such defective apoptotic cell clearance observed in atherosclerotic lesions suggests that apoptotic cells evolve to necrotic cells, the latter triggering proinflammatory and immunostimulatory responses. 34,35 In turn, impaired clearance of dying cells may promote arterial inflammation and contribute to lesion development. 8,34,36


In our model, plaque progression was potentiated by local inflammation in the arterial wall of gld Ldl-r -/- mice as demonstrated by significant upregulation of mRNAs indicative of activation of endothelial cells (MCP-1, P-selectin, and VCAM-1) and macrophages (MCP-1) as compared with controls. This finding was associated with significantly elevated expression of mRNAs encoding genes associated with monocytes (CCR2, CD11b, and CD115) and macrophages (CD68 and F4/80) in the descending aorta of gld Ldl-r -/- mice. Endothelial cell activation might be induced by elevated circulating IgG levels among which specific IgGs and/or specific immune complexes might interact with the endothelium to activate endothelial cells inducing endothelial dysfunction. 37,38,39,40 Elevated endothelial permeability typical of endothelium dysfunction facilitates enhanced lipoprotein access, retention, and deposition in the intimal space, a classical feature of fatty streak formation. In this setting, the adhesion molecules and chemokines whose expression is upregulated in the aortas of gld Ldl-r -/- mice are known to be key players in the recruitment of monocytes into the subendothelial space, thus favoring lesion progression. 41,42 Indeed, increase in the expression of CD11b may reflect the entry of newly recruited monocytes, 9,14 thereby leading to the accumulation of macrophages in the arterial wall. However, we cannot exclude the possibility that macrophages with the gld mutation could display higher resistance to apoptosis, a feature equally reported in another mouse model for SLE, 43 and which, in turn, favors their accumulation in the aortic sinus of gld Ldl-r -/- mice. Nevertheless, the similar proportions of splenic F4/80 + macrophages observed in wt Ldl-r -/- and gld Ldl-r -/- mice (4.8±1.1 versus 3.3±0.5%, respectively) suggest that the half life of gld macrophages is not affected and argues that higher resistance to apoptosis may not be operative in aortic lesions.


In our innovative gld Ldl-r -/- mouse model, we provide new insight into the molecular mechanisms that may underlie accelerated atherosclerosis in autoimmune disease. Indeed, our experimental findings highlight enhanced activation of the immune system, aortic inflammation, and endothelial cell activation, together with lesional accumulation of apoptotic cells, macrophages, and CD4 T cells as being central to the development and progression of atherosclerosis in autoimmune mice. These findings may lead to development of innovative therapeutic strategies in SLE patients at high cardiovascular risk.


Acknowledgments


We are indebted to Dr B. Lelongt for technical assistance with renal morphology and Pr J.J. Mazeron and Dr G. Boisserie of the Radiotherapy Unit for advice with mouse irradiation.


Sources of Funding


This work was supported by INSERM and by an Award from The Fondation de France to P.L. E.G. was supported by a Fellowship from the Ministère de la Recherche and the Fondation pour la Recherche Médicale. M.J.C. and P.L. gratefully acknowledge the award of a "Contrat d?Interface" by the Assistance Publique - Hôpitaux de Paris/INSERM.


Disclosures


None.

【参考文献】
  Urowitz MB, Bookman AA, Koehler BE, Gordon DA, Smythe HA, Ogryzlo MA. The bimodal mortality pattern of systemic lupus erythematosus. Am J Med. 1976; 60: 221-225.

Frostegard J. Atherosclerosis in patients with autoimmune disorders. Arterioscler Thromb Vasc Biol. 2005; 25: 1776-1785.

Roman MJ, Shanker BA, Davis A, Lockshin MD, Sammaritano L, Simantov R, Crow MK, Schwartz JE, Paget SA, Devereux RB, Salmon JE. Prevalence and correlates of accelerated atherosclerosis in systemic lupus erythematosus. N Engl J Med. 2003; 349: 2399-2406.

Asanuma Y, Oeser A, Shintani AK, Turner E, Olsen N, Fazio S, Linton MF, Raggi P, Stein CM. Premature coronary-artery atherosclerosis in systemic lupus erythematosus. N Engl J Med. 2003; 349: 2407-2415.

del Rincon I, O?Leary DH, Haas RW, Escalante A. Effect of glucocorticoids on the arteries in rheumatoid arthritis. Arthritis Rheum. 2004; 50: 3813-3822.

Liu K, Mohan C. What do mouse models teach us about human SLE? Clin Immunol. 2006; 119: 123-130.

Cohen PL, Eisenberg RA. Lpr and gld: single gene models of systemic autoimmunity and lymphoproliferative disease. Annu Rev Immunol. 1991; 9: 243-269.

Aprahamian T, Rifkin I, Bonegio R, Hugel B, Freyssinet JM, Sato K, Castellot JJ, Jr., Walsh K. Impaired clearance of apoptotic cells promotes synergy between atherogenesis and autoimmune disease. J Exp Med. 2004; 199: 1121-1131.

Grainger DJ, Reckless J, McKilligin E. Apolipoprotein E modulates clearance of apoptotic bodies in vitro and in vivo, resulting in a systemic proinflammatory state in apolipoprotein E-deficient mice. J Immunol. 2004; 173: 6366-6375.

Bro S, Bentzon JF, Falk E, Andersen CB, Olgaard K, Nielsen LB. Chronic renal failure accelerates atherogenesis in apolipoprotein E-deficient mice. J Am Soc Nephrol. 2003; 14: 2466-2474.

Ivanovski O, Szumilak D, Nguyen-Khoa T, Ruellan N, Phan O, Lacour B, Descamps-Latscha B, Drueke TB, Massy ZA. The antioxidant N-acetylcysteine prevents accelerated atherosclerosis in uremic apolipoprotein E knockout mice. Kidney Int. 2005; 67: 2288-2294.

Go AS, Chertow GM, Fan D, McCulloch CE, Hsu CY. Chronic kidney disease and the risks of death, cardiovascular events, and hospitalization. N Engl J Med. 2004; 351: 1296-1305.

Huby T, Doucet C, Dachet C, Ouzilleau B, Ueda Y, Afzal V, Rubin E, Chapman MJ, Lesnik P Knockdown expression and hepatic deficiency reveal an atheroprotective role for SR-BI in liver and peripheral tissues. J Clin Invest. 2006.

Lesnik P, Haskell CA, Charo IF. Decreased atherosclerosis in CX3CR1-/- mice reveals a role for fractalkine in atherogenesis. J Clin Invest. 2003; 111: 333-340.

Palinski W, Tangirala RK, Miller E, Young SG, Witztum JL. Increased autoantibody titers against epitopes of oxidized LDL in LDL receptor-deficient mice with increased atherosclerosis. Arterioscler Thromb Vasc Biol. 1995; 15: 1569-1576.

Munoz LE, Gaipl US, Franz S, Sheriff A, Voll RE, Kalden JR, Herrmann M. SLE-a disease of clearance deficiency? Rheumatology (Oxford). 2005; 44: 1101-1107.

Stanic AK, Stein CM, Morgan AC, Fazio S, Linton MF, Wakeland EK, Olsen NJ, Major AS. Immune dysregulation accelerates atherosclerosis and modulates plaque composition in systemic lupus erythematosus. Proc Natl Acad Sci U S A. 2006; 103: 7018-7023.

Theofilopoulos AN, Lawson BR Tumour necrosis factor and other cytokines in murine lupus. Ann Rheum Dis. 1999; 58 Suppl 1: I49-55.

Jones BM, Liu T, Wong RW. Reduced in vitro production of interferon-gamma, interleukin-4 and interleukin-12 and increased production of interleukin-6, interleukin-10 and tumour necrosis factor-alpha in systemic lupus erythematosus. Weak correlations of cytokine production with disease activity. Autoimmunity. 1999; 31: 117-124.

Hagiwara E, Gourley MF, Lee S, Klinman DK. Disease severity in patients with systemic lupus erythematosus correlates with an increased ratio of interleukin-10:interferon-gamma-secreting cells in the peripheral blood. Arthritis Rheum. 1996; 39: 379-385.

Horwitz DA, Gray JD, Behrendsen SC, Kubin M, Rengaraju M, Ohtsuka K, Trinchieri G. Decreased production of interleukin-12 and other Th1-type cytokines in patients with recent-onset systemic lupus erythematosus. Arthritis Rheum. 1998; 41: 838-844. <a href="/cgi/external_ref?access_num=10.1002/1529-0131(199805)41:5

Richaud-Patin Y, Alcocer-Varela J, Llorente L. High levels of TH2 cytokine gene expression in systemic lupus erythematosus. Rev Invest Clin. 1995; 47: 267-272.

Lin LC, Chen YC, Chou CC, Hsieh KH, Chiang BL. Dysregulation of T helper cell cytokines in autoimmune prone NZB x NZW F1 mice. Scand J Immunol. 1995; 42: 466-472.

Alleva DG, Kaser SB, Beller DI. Aberrant cytokine expression and autocrine regulation characterize macrophages from young MRL+/+ and NZB/W F1 lupus-prone mice. J Immunol. 1997; 159: 5610-5619.

Gupta S, Pablo AM, Jiang X, Wang N, Tall AR, Schindler C. IFN-gamma potentiates atherosclerosis in ApoE knock-out mice. J Clin Invest. 1997; 99: 2752-2761.

Buono C, Come CE, Stavrakis G, Maguire GF, Connelly PW, Lichtman AH. Influence of interferon-gamma on the extent and phenotype of diet-induced atherosclerosis in the LDLR-deficient mouse. Arterioscler Thromb Vasc Biol. 2003; 23: 454-460.

Davenport P, Tipping PG. The role of interleukin-4 and interleukin-12 in the progression of atherosclerosis in apolipoprotein E-deficient mice. Am J Pathol. 2003; 163: 1117-1125.

Hauer AD, Uyttenhove C, de Vos P, Stroobant V, Renauld JC, van Berkel TJ, van Snick J, Kuiper J. Blockade of interleukin-12 function by protein vaccination attenuates atherosclerosis. Circulation. 2005; 112: 1054-1062.

King VL, Szilvassy SJ, Daugherty A. Interleukin-4 deficiency decreases atherosclerotic lesion formation in a site-specific manner in female LDL receptor-/- mice. Arterioscler Thromb Vasc Biol. 2002; 22: 456-461.

Kirii H, Niwa T, Yamada Y, Wada H, Saito K, Iwakura Y, Asano M, Moriwaki H, Seishima M. Lack of interleukin-1beta decreases the severity of atherosclerosis in ApoE-deficient mice. Arterioscler Thromb Vasc Biol. 2003; 23: 656-660.

Mallat Z, Besnard S, Duriez M, Deleuze V, Emmanuel F, Bureau MF, Soubrier F, Esposito B, Duez H, Fievet C, Staels B, Duverger N, Scherman D, Tedgui A. Protective role of interleukin-10 in atherosclerosis. Circ Res. 1999; 85: e17-24.

Manderson AP, Botto M, Walport MJ. The role of complement in the development of systemic lupus erythematosus. Annu Rev Immunol. 2004; 22: 431-456.

Laderach D, Bach JF, Koutouzov S. Nucleosomes inhibit phagocytosis of apoptotic thymocytes by peritoneal macrophages from MRL+/+ lupus-prone mice. J Leukoc Biol. 1998; 64: 774-780.

Tabas I. Consequences and therapeutic implications of macrophage apoptosis in atherosclerosis: the importance of lesion stage and phagocytic efficiency. Arterioscler Thromb Vasc Biol. 2005; 25: 2255-2264.

Albert ML. Death-defying immunity: do apoptotic cells influence antigen processing and presentation? Nat Rev Immunol. 2004; 4: 223-231.

Boisvert WA, Rose DM, Boullier A, Quehenberger O, Sydlaske A, Johnson KA, Curtiss LK, Terkeltaub R. Leukocyte transglutaminase 2 expression limits atherosclerotic lesion size. Arterioscler Thromb Vasc Biol. 2006; 26: 563-569.

Binstadt BA, Patel PR, Alencar H, Nigrovic PA, Lee DM, Mahmood U, Weissleder R, Mathis D, Benoist C. Particularities of the vasculature can promote the organ specificity of autoimmune attack. Nat Immunol. 2006; 7: 284-292.

Cho CS, Cho ML, Chen PP, Min SY, Hwang SY, Park KS, Kim WU, Min DJ, Min JK, Park SH, Kim HY. Antiphospholipid antibodies induce monocyte chemoattractant protein-1 in endothelial cells. J Immunol. 2002; 168: 4209-4215.

Papa ND, Raschi E, Moroni G, Panzeri P, Borghi MO, Ponticelli C, Tincani A, Balestrieri G, Meroni PL. Anti-endothelial cell IgG fractions from systemic lupus erythematosus patients bind to human endothelial cells and induce a pro-adhesive and a pro-inflammatory phenotype in vitro. Lupus. 1999; 8: 423-429.

Carvalho D, Savage CO, Isenberg D, Pearson JD. IgG anti-endothelial cell autoantibodies from patients with systemic lupus erythematosus or systemic vasculitis stimulate the release of two endothelial cell-derived mediators, which enhance adhesion molecule expression and leukocyte adhesion in an autocrine manner. Arthritis Rheum. 1999; 42: 631-640. <a href="/cgi/external_ref?access_num=10.1002/1529-0131(199904)42:4

Libby P. Inflammation in atherosclerosis. Nature. 2002; 420: 868-874.

Hansson GK. Inflammation, atherosclerosis, and coronary artery disease. N Engl J Med. 2005; 352: 1685-1695.

Zhu J, Liu X, Xie C, Yan M, Yu Y, Sobel ES, Wakeland EK, Mohan C. T cell hyperactivity in lupus as a consequence of hyperstimulatory antigen-presenting cells. J Clin Invest. 2005; 115: 1869-1878.


作者单位:From INSERM Unit 55 UPMC-Paris Dyslipoproteinemia and Atherosclerosis Research Unit, Hôpital de la Pitié, Paris, France.

作者: Emmanuel L. Gautier; Thierry Huby; Betty Ouzilleau
医学百科App—中西医基础知识学习工具
  • 相关内容
  • 近期更新
  • 热文榜
  • 医学百科App—健康测试工具