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2006年第26卷第12期

Critical Role of Inducible Costimulator Signaling in the Development of Arteriosclerosis

来源:《动脉硬化血栓血管生物学杂志》
摘要:Induciblecostimulator(ICOS)isamajorcostimulatorofTcellactivation。Induciblecostimulator(ICOS)isamemberoftheCD28/CTLA4familyandisexpressedonTcellsafteractivation。...

点击显示 收起

【摘要】  Objective- Proliferation and migration of smooth muscle cells (SMCs) and migration and accumulation of monocytes and T cells are landmark events in the development of arteriosclerosis. SMC proliferation in the intima induces interruption of blood flow and results in ischemia and graft rejection. Inducible costimulator (ICOS) is a major costimulator of T cell activation. However, the effect of costimulatory molecules on the formation of neointimal hyperplasia has not been fully elucidated. We examined the role of the ICOS pathway in SMC proliferation.

Methods and Results- ICOS ligand (ICOSL) was detected in SMCs stimulated by interleukin (IL)-1ß, and coculture of stimulated SMCs and activated T cells induced SMC proliferation. Inhibition of the ICOS pathway resulted in inhibition of SMC proliferation. In models of transplantation and vascular injury, ICOSL was induced in SMCs in the neointima. Expression of IL-1ß, a key inducer of ICOSL expression, was significantly reduced in mice treated with anti-ICOS antibody or soluble form of ICOS (ICOSIg) and in ICOS-deficient mice. Inhibition of the ICOS pathway significantly suppressed neointimal thickening.

Conclusions- These results indicate that ICOS on activated T cells contributes to neointimal formation through the regulation of SMC proliferation. These findings provide insights into new therapeutic strategies for arteriosclerosis.

The role of the ICOS pathway in SMC proliferation has not yet been evaluated. We demonstrate that blockade of the ICOS pathway significantly suppressed SMC proliferation and neointimal thickening.

【关键词】  arteriosclerosis costimulation neointima formation smooth muscle cells


Introduction


Cardiovascular disease continues to be the major cause of death in developed countries. 1,2 Neointimal hyperplasia, characterized by graft vasculopathy after transplantation and restenosis after vascular injury, involves infiltration by mononuclear cells including T cells and macrophages. In addition, proliferation of smooth muscle cells (SMCs) plays important roles in neointimal formation, processes that are influenced by multiple cytokines and can be predicted by endothelial damage. 3-5 SMC proliferation in the intima induces interruption of blood flow and results in ischemia and graft rejection.


T cell activation requires 2 distinct signals through T cell receptors and costimulatory molecules. Inducible costimulator (ICOS) is a member of the CD28/CTLA4 family and is expressed on T cells after activation. Stimulation of the ICOS pathway promotes secretion of IFN-, IL-4, and IL-10. 4 Inhibition of the ICOS pathway with anti-ICOS antibody or the soluble form of ICOS (ICOSIg) prolonged cardiac allograft survival in a murine model, and combined treatment with anti-ICOS antibody and cyclosporin A 6 or ICOSIg and CTLA4Ig 7 prolonged allograft survival indefinitely and prevented the development of graft vasculopathy. Treatment with anti-ICOS antibody in addition to blockade of the CD40 ligand (CD40L)/CD40 pathway also attenuated the development of graft vasculopathy. 6


ICOS ligand (ICOSL), also known as B7-related protein 1 (B7RP-1), is expressed constitutively on B cells and in peripheral lymphoid tissues. 8,9 In vitro studies have shown that ICOSL expression is induced on fibroblasts treated with tumor necrosis factor (TNF)- and that it is expressed constitutively on endothelial cells and is upregulated by treatment with IL-1ß or TNF-. 9,10 However, it is not known whether ICOSL is expressed on SMCs.


Numerous studies have indicated that inhibition of the costimulatory pathway not only prolongs allograft survival 11 but also attenuates atherogenesis and neointimal hyperplasia after transplantation or vascular injury. 12-15 We recently reported that SMCs stimulated by interferon (IFN)- express herpes virus entry mediator (HVEM), which binds to LIGHT, and proliferate by interaction with T cells through the LIGHT pathway. 16 Programmed death-ligand 1 (PD-L1) is expressed on SMCs stimulated by IFN- and is associated with SMC proliferation. 17 Although it has not been shown that other costimulatory molecules are expressed on SMCs, data suggest that the costimulatory pathway plays an important role in regulating not only immune responses but also SMC proliferation and subsequent arteriopathy.


Here, we show that ICOSL is expressed on SMCs in neointima formed by vascular injury. The ICOS pathway promotes SMC proliferation in these animal models. Furthermore, we show that inhibition of the ICOS pathway prevents the development of neointimal hyperplasia after transplantation or vascular injury.


Materials and Methods


For complete Materials and Methods, please see http://atvb.ahajournals.org.


Reagents


Anti-mouse ICOS monoclonal antibody (JMAb51) was obtained from JT Pharmaceutical Frontier Research Laboratory (Kamagawa, Japan). 18 ICOSIg was prepared as described previously. 7


Animals


Six- to 8-week-old male C57BL/6 (B/6, H-2 b ) mice were obtained from Japan Clea. Male B6.C-H-2 bm12 KhEg (Bm12, H-2 bm12 ) mice were obtained from The Jackson Laboratory (Bar Harbor, Me). Combinations of B/6 and Bm12 mice are class II mismatch. Male ICOS -/- mice on a C57BL/6 background were provided by JT Pharmaceutical Frontier Research Laboratory. 19 Animals were maintained in our animal facility and weighed 20 to 25 grams. The present study conformed to the Guide for the Care and Use of Laboratory Animals of Tokyo Medical and Dental University.


Cardiac Transplantation


Donor hearts were transplanted heterotopically into recipient mice as described previously. 7 Recipient mice were injected intraperitoneally with anti-ICOS antibody (100 µg per treatment, n=10), ICOSIg (50 µg per treatment, n=10), or isotype-matched control IgG (n=10) every 7 d for 8 weeks after transplantation. ICOS -/- mice were used as recipients (n=10). Allografts were harvested 8 weeks after transplantation.


Mouse Femoral Arterial Injury Model


Transluminal arterial injury of the femoral arteries of B/6 or ICOS -/- mice (n=8) was induced by inserting a straight spring wire as described previously. 20 B/6 mice were injected intraperitoneally with anti-ICOS antibody (100 µg per treatment, n=8), ICOSIg (50 µg per treatment, n=8), or isotype-matched control IgG (n=8) on days 0, 2, 4, 6, 8, and 10 after injury. At 4 weeks after injury, the femoral arteries were excised.


Immunohistochemistry


Immunohistochemistry and immunofluorescence double staining were performed as described previously. 16


Histological Evaluation


Grafts and arteries were analyzed by Elastica van Gieson staining. The areas within the internal elastic lamina (IEL) and the external elastic lamina (EEL) and the lumen were carefully traced, and the planimetric areas were calculated with an image analysis system (Scion Image beta 4.0.2; Scion).


Coculture of SMCs and T cells


Primary SMCs were obtained from the thoracic aortas of B/6, Bm12, or ICOS -/- mice by an explant technique as described previously. 16


Splenocyte suspensions were obtained by disrupting spleens between sterile glass slides. Red blood cells were lysed by ammonium chloride lysis. Cells were washed, and T cells were obtained by depletion of non-T cells (B cells, NK cells, dendritic cells, macrophages, granulocytes, and erythroid cells) by magnetic cell sorting (Miltenyi Biotec). The purity of separated T cells 95% as assessed by flow cytometry.


SMCs were trypsinized and seeded onto 96-well plates. At confluence, SMCs were arrested in medium with 0.4% fetal bovine serum for 5 days. SMCs were then stimulated with recombinant mouse IL-1ß (10 ng/mL) for 48 hours. After a wash in phosphate-buffered saline, mitomycin-C-inactivated T cells (total 5 x 10 5 ) and anti-ICOS antibody, ICOSIg (10 µg/mL), or PD98059 (10 µg/mL) were added to each well for 24 hours. After washing in phosphate-buffered saline, SMCs were incubated with BrdU (Roche) according to the manufacturer?s instructions. Incorporated BrdU was measured by a microplate-imaging system (BioRad). SMC proliferation is expressed as the optical density.


Reverse-Transcription Polymerase Chain Reaction Analysis


Total RNA was extracted from SMCs after 24 hours of cytokine stimulation. cDNA was prepared from 5 µg of RNA by reverse transcription. cDNA (10 µL) was amplified. Polymerase chain reaction products were analyzed by ethidium bromide staining of 1.5% agarose gels.


Western Blot Analysis


Western blot was performed as described previously. 17


Fluorescence-Activated Cell Sorter analysis


Arrested SMCs were stimulated with IL-1ß (10 ng/mL) for 24 or 48 hours. SMCs with and without IL-1ß stimulation were incubated with anti-ICOSL antibody and stained with biotinylated isotype-matched control IgG or goat IgG. Cells were then stained with fluorescein isothiocyanate (FITC)-conjugated anti- -smooth muscle actin (SMA) antibody and streptavidin-phycoerythrin. Cells were then analyzed by flow cytometry on a fluorescence-activated cell sorter Calibur (Becton Dickinson).


Statistical Analysis


All data are expressed as mean±SEM. Differences between 2 groups were analyzed by Student t test. Differences in data between multiple groups were subjected to 1-way ANOVA followed by Scheffe test. P <0.05 was considered statistically significant.


Results


Expression of ICOSL on SMCs


We analyzed the expression of ICOSL on SMCs by reverse-transcription polymerase chain reaction and flow cytometry. Treatment with IL-1ß induced ICOSL expression on SMCs from B/6 or Bm12 mice ( Figure 1 and supplemental Figure I, available at http://atvb.ahajournals.org). ICOSL was not detected on untreated SMCs ( Figure 1 and supplemental Figure I) or on SMCs treated with TNF-, IFN-, or IL-4 (data not shown).


Figure 1. Expression of ICOSL in SMCs by reverse-transcription polymerase chain reaction. cDNA generated from untreated or IL-1ß-treated SMCs was amplified with primers for ICOSL. An ICOSL cDNA product of the expected size (300 bp) was detected in IL-1ß-treated SMCs from B/6 and Bm12 mice.


SMC Proliferation Induced by Interaction With T Cells


Naive T cells and T cells activated after transplantation were isolated from splenocytes of B/6 or ICOS -/- mice. Naiive T cells or activated T cells from B/6 mice did not induce proliferation of unstimulated SMCs from Bm12 mice ( Figure 2A and 2 B). Naiive T cells from B/6 mice also did not stimulate proliferation of SMCs from Bm12 mice treated with IL-1ß ( Figure 2 A). However, activated T cells from B/6 mice induced significant proliferation of SMCs from Bm12 mice treated with IL-1ß ( Figure 2 B). This proliferation was inhibited by addition of anti-ICOS antibody or ICOSIg ( Figure 2 B). Interestingly, activated T cells from B/6 mice induced significant proliferation of IL-1ß-treated SMCs from B/6 mice, and addition of anti-ICOS antibody or ICOSIg suppressed SMC proliferation (supplemental Figure IIA and IIB).


Figure 2. Coculture of T cells from B/6 mice and SMCs from Bm12 mice and Western blotting. Naiive T cells (A) did not induce proliferation of IL-1ß-untreated or IL-1ß-treated SMCs. Activated T cells (B) induced proliferation of IL-1ß-untreated SMCs and significantly stimulated proliferation of IL-1ß-treated SMCs. Phosphorylation of ERK 1/2 in SMCs stimulated by IL-1ß was detected after 15 minutes from addition of activated T cells, and inhibition of the ICOS pathway inhibited ERK 1/2 phosphorylation. Results are representative of three independent experiments. Blot density was defined as ratio to p-ERK of untreated SMCs. Data are expressed as mean±SEM of 3 independent experiments. * P <0.05. C and D, Inhibition of SMC proliferation by a MEK inhibitor PD98059. Data are expressed as mean±SEM of 3 independent experiments. * P <0.05. E and F, Coculture of T cells from ICOS -/- mice and SMCs from Bm12 mice and Western blotting. Naiive and activated T cells did not induce proliferation of unstimulated or IL-1ß-stimulated SMCs from Bm12 mice. Phosphorylation of ERK 1/2 was inhibited in SMCs. Results are representative of 3 independent experiments. Blot density was defined as ratio to p-ERK of untreated SMCs. Data are expressed as mean±SEM of 3 independent experiments. * P <0.05.


We tested whether this effect on SMC proliferation involved activation of mitogen-activated protein kinase (MAPK) signaling. Increased extracellular signal regulated kinase (ERK) 1 and 2 (ERK 1/2) phosphorylation in SMCs was detected in cocultures of IL-1ß-stimulated SMCs and activated T cells ( Figure 2A and 2 B; supplemental Figure IIA and IIB). To clarify the involvement of ERK1/2 in SMC proliferation, we used a MEK inhibitor PD98059. PD98059 significantly suppressed SMC proliferation induced by interaction with activated T cells ( Figure 2C and 2 D; supplemental Figure IIC and IID). However, Western blot analysis showed no effect on the phosphorylation of c-Jun-N-terminal kinase (JNK) or p38 MAPK (data not shown). Neither naive nor activated T cells from ICOS -/- mice induced proliferation of unstimulated or IL-1ß-stimulated SMCs from Bm12 mice ( Figure 2E and 2 F). Western blot analysis showed no effect on the phosphorylation of ERK 1/2, JNK or p38 MAPK in SMCs from Bm12 mice ( Figure 2E and 2 F, data not shown).


Prevention of Graft Vasculopathy by Inhibition of the ICOS Pathway


We used B/6 or ICOS -/- mice as recipients and Bm12 mice as donors. ICOS staining was detected in graft infiltrating cells (supplemental Figure IIIA) and on cells in thickened neointima (supplemental Figure IIIB). Although ICOS expression was not detected on naive CD4 + and CD8 + cells, it was induced on activated CD4 + and CD8 + cells (supplemental Figure IIIC). In comparison to those in control IgG-treated allografts, the numbers of SMCs in neointima and the numbers of CD4 +, CD8 +, and CD11b + cells infiltration in allografts treated with anti-ICOS antibody or ICOSIg or in ICOS -/- mice was significantly decreased (supplemental Figure IIID and IIIE).


Allografts in control IgG-treated mice at 8 weeks after transplantation showed severe neointimal hyperplasia (luminal occlusion: 70.1±5.5%; Figure 3A and 3 F). Interestingly, ICOSL staining was detected on SMCs in grafts with vasculopathic change, but no ICOSL staining was detected on SMCs in native hearts (supplemental Figure IIIF). In comparison to control IgG-treated allografts, allografts treated with anti-ICOS antibody or ICOSIg at 8 weeks after transplantation showed significantly less severe neointimal hyperplasia (8.3±4.8%, 10.1±5.6%, respectively; Figure 3B, 3C, and 3 F). Furthermore, sections from allografts of ICOS -/- recipient mice and isografts showed no intimal thickening by 8 weeks after transplantation (11.8±4.5%, 0.0%; Figure 3D, 3E, and 3 F).


Figure 3. Photomicrographs of paraffin sections of cardiac allografts at 8 weeks after transplantation stained with Elastica van Gieson stain. Representative vessels from allografts treated with control IgG (A), anti-ICOS antibody (B), or ICOSIg (C), an allograft from an ICOS -/- recipient mouse (D), and an isograft (E) are shown (original magnification, 400 x ). F, The degree of graft vasculopathy in each group was quantified. Data are expressed as mean±SEM of 10 mice in each group. * P <0.05.


To examine cytokine expression in cardiac allografts, we performed Western blot analysis on native hearts and cardiac allografts at 8 weeks after transplantation. Expression of IL-1ß, IL-6, and IFN- was detected in allografts from control IgG-treated mice (supplemental Figure IIIG). However, expression of these cytokines was significantly suppressed in allografts from anti-ICOS antibody-treated mice, from ICOSIg-treated mice, and from ICOS -/- mice (supplemental Figure IIIG). Expression of IL-4 and IL-10 was not altered (supplemental Figure IIIG).


Prevention of Neointimal Hyperplasia After Vascular Injury by Inhibition of the ICOS Pathway


Femoral arteries of B/6 mice developed severe intimal hyperplasia at 4 weeks after injury (intima-media ratio: 2.2±0.3, Figure 4B and 4 F). Although ICOS expression was not detected on naive CD4 + and CD8 + cells, it was induced on activated CD4 + and CD8 + cells (supplemental Figure IVA). ICOS-positive cells were detected in the neointima (supplemental Figure IVB). Immunofluorescence showed ICOSL staining on SMCs in neointimal hyperplasia at 4 weeks after injury; no staining was detected on SMCs in uninjured arteries (supplemental Figure IVC). In comparison to that in control IgG-treated mice, neointimal formation was significantly attenuated in mice treated with anti-ICOS antibody or ICOSIg and in ICOS -/- mice (I/M ratio: 1.1±0.2, 1.0±0.2, 0.9±0.2, respectively) ( Figure 4 A to 4E). In comparison to those in control IgG-treated mice, the numbers of SMCs in neointima and the numbers of CD4 +, CD8 +, and CD11b + cells infiltration was significantly reduced in mice treated with anti-ICOS antibody or ICOSIg and in ICOS -/- mice (supplemental Figure IVD and IVE) and IL-1ß staining was suppressed on SMCs in mice treated with anti-ICOS antibody or ICOSIg and in ICOS -/- mice (supplemental Figure IVF).


Figure 4. Photomicrographs of paraffin sections of femoral arteries at 4 weeks after vascular injury stained with Elastica van Gieson stain. Representative vessels from arteries treated with control IgG (A), anti-ICOS antibody (B), or ICOSIg (C), and arteries from ICOS -/- mice (D) are shown (original magnification, 400 x ). E, Quantitative analysis of I/M ratio in each group is shown. Data are expressed as mean±SEM of 8 mice in each group. * P <0.05.


Discussion


Acute coronary syndromes and chronic rejection of transplanted organs induce disruption of plaque and neointimal hyperplasia, leading to loss of blood flow. These illnesses are associated with immune responses, and SMC proliferation plays a central role in the progression of neointimal hyperplasia. Cytokine stimulation and costimulatory signaling induce SMC proliferation 16 and neointimal hyperplasia 17,21; however, the interactions between T cells and SMCs that result in signaling of the costimulatory pathway and in SMC proliferation have not been fully elucidated. To the best of our knowledge, the present study is the first to show that inhibition of the ICOS pathway reduces proliferation of SMCs cultured with activated T cells through abrogation of ERK 1/2 phosphorylation and inhibits the development of intimal hyperplasia after transplantation or vascular injury.


ICOSL is expressed on endothelial cells and is upregulated by various inflammatory stimuli. 9,10 We found that ICOSL expression was induced on SMCs treated with IL-1ß. It has been reported that cytokines 16 and growth factors 22 stimulate proliferation of SMCs. Our present data showed that IL-1ß stimulated SMC proliferation ( Figure 2 ). A previous study showed that T cells affect SMC proliferation, 23 and we recently reported that costimulatory molecules were expressed on SMCs and that the costimulatory pathway was associated with SMC proliferation. 16,17 Therefore, we examined whether the ICOS pathway was associated with SMC proliferation. SMCs proliferated in response to IL-1ß stimulation; however, synergistic proliferation was induced only when SMCs treated with IL-1ß were cocultured with T cells activated after transplantation. This proliferation was decreased by inhibition of the ICOS pathway with anti-ICOS antibody or ICOSIg. These results indicate that SMC proliferation in vitro is induced by signaling through the ICOS pathway, and activated T cells from B/6 mice stimulate proliferation not only of SMCs from Bm12 mice but also SMCs from B/6 mice.


MAPKs are known to play roles in regulating cell proliferation, survival, and differentiation. ERK 1/2 regulates cell proliferation and differentiation, and p38 MAPK and JNK are activated by cellular stresses such as inflammation and apoptosis. In this study, phosphorylation of ERK 1/2 in SMCs stimulated by IL-1ß was detected after addition of activated T cells, and inhibition of the ICOS pathway inhibited ERK 1/2 phosphorylation. Phosphorylation of p38 MAPK and JNK in IL-1ß-stimulated SMCs was not detected after addition of activated T cells. These data suggest that SMC proliferation is induced by activation of ERK 1/2 signaling in the ICOS pathway.


On the basis of in vitro data, we examined the relation between SMC proliferation and the ICOS pathway in vivo. Although survival early after transplantation has been improved by advances in immunosuppressive therapy, 24 graft vasculopathy remains a major problem limiting long-term survival of cardiac transplant recipients. T cells and macrophages are activated and result in the secretion of proinflammatory cytokines. Although these cytokines affect SMC proliferation, 16 it has been reported that costimulatory pathways such as those involving CD28/B7 and CD40L/CD40 are involved in the development of graft vasculopathy. 4,25 Neointimal thickening after vascular injury results from the infiltration of leukocytes, deposition of extracellular matrix, and excessive proliferation of SMCs. Previous studies have shown high plasma levels of soluble CD40L in apparently healthy women at high risk for cardiovascular events 26 and in patients with unstable angina. 27 Increased levels of soluble CD40L are also associated with the development of restenosis after balloon angioplasty. 28 However, the role of costimulatory pathways in neointimal hyperplasia after transplantation or vascular injury remains unknown. Immunofluorescence staining indicated that ICOSL expression was induced on SMCs in thickened neointima after transplantation or vascular injury. Therefore, we examined whether the development of neointimal hyperplasia is associated with the ICOS pathway. Compared with treatment with control IgG, treatment with anti-ICOS antibody or ICOSIg significantly reduced the development of neointimal hyperplasia after transplantation or vascular injury. Neointimal hyperplasia was also significantly attenuated in allografts and arteries from ICOS -/- mice. Compared with mice treated with control IgG, proinflammatory cytokines, including IL-1ß, an important regulator of ICOSL expression, were significantly suppressed in mice after transplantation or vascular injury treated by inhibition of the ICOS pathway. There is the limitation to this study. It is not clear how the blockade of the ICOS pathway inhibits the development of arteriosclerosis in vivo. The infiltration of T cells and the expression of proinflammatory cytokines were significantly suppressed by inhibition of the ICOS pathway. Cytokine stimulation plays critical roles in SMC proliferation and the development of neointimal hyperplasia after transplantation or vascular injury. T cell activation and differentiation are defective in the absence of ICOS. 29-31 Therefore, it is probable that the development of arteriosclerosis is inhibited by the attenuation of inflammatory responses. However, interaction between activated T cells and SMCs may be involved with SMC proliferation in vitro and in vivo. Our data also suggest that interaction between homotypic activated T cells and SMCs synergistically induces SMC proliferation and may contribute to restenosis after injury. Further studies are necessary to clarify the mechanism by which the interaction between T cells and SMCs inhibits arteriosclerosis.


Afek et al reported that blockade of the ICOS pathway increased atherosclerotic lesions in apoE knockout mice. 32 The data of Afeck et al are based on one side that ICOSIg fusion protein alone was administrated to block the ICOS pathway. However, we used not only ICOSIg fusion protein but also anti-ICOS antibody or ICOS -/- mice. It is possible that how to block the costimulatory pathways induces different actions. Also, the model of vasculopathy was used in our study, although the model of atherosclerosis was used in Afeck et al study. The ICOS pathway may play different roles in vasculopathy and atherosclerosis. Although acquired immunity is related to the development of vasculopathy, the role of acquired immunity in atherosclerosis is unclear. These points could explain the discrepancy between our data and Afek et al study.


In conclusion, the present study shows that ICOSL is expressed on SMCs stimulated by IL-1ß and that the ICOS pathway plays a pivotal role in regulating SMC proliferation in vitro and in vivo. Reduction of neointimal hyperplasia occurs not only by modulation of immune responses but also by suppression of SMC proliferation through the ICOS pathway. Therefore, the ICOS pathway may comprise an important target for the prevention of neointimal formation.


Acknowledgments


We thank Dr Ryo Gotoh, Noriko Tamura, and Masahito Ogawa for excellent technical assistance.


Sources of Funding


This study was supported by grants from grants-in-aid from the Japanese Ministry of Education, Culture, Sports, Science, and Technology and the Organization for Pharmaceutical Safety and Research.


Disclosures


None.


J.S. and G.H. contributed equally to this work.


Original received February 12, 2006; final version accepted August 26, 2006.

【参考文献】
  Ross R. Atherosclerosis-an inflammatory disease. N Engl J Med. 1999; 340: 115-126.

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

Ross R, Glomset JA. Atherosclerosis and the arterial smooth muscle cell: proliferation of smooth muscle is a key event in the genesis of the lesions of atherosclerosis. Science. 1973; 180: 1332-1339.

Russell ME, Hancock WW, Akalin E, Wallace AF, Glysing-Jensen T, Willett TA, Sayegh MH. Chronic cardiac rejection in the LEW to F344 rat model. Blockade of CD28-B7 costimulation by CTLA4Ig modulates T cell and macrophage activation and attenuates arteriosclerosis. J Clin Invest. 1996; 97: 833-838.

Suzuki J, Isobe M, Morishita R, Aoki M, Horie S, Okubo Y, Kaneda Y, Sawa Y, Matsuda H, Ogihara T, Sekiguchi M. Prevention of graft coronary arteriosclerosis by antisense cdk2 kinase oligonucleotide. Nat Med. 1997; 3: 900-903.

Ozkaynak E, Gao W, Shemmeri N, Wang C, Gutierrez-Ramos JC, Amaral J, Qin S, Rottman JB, Coyle AJ, Hancock WW. Importance of ICOS-B7RP-1 costimulation in acute and chronic allograft rejection. Nat Immunol. 2001; 2: 591-596.

Kosuge H, Suzuki J, Gotoh R, Koga N, Ito H, Isobe M, Inobe M, Uede T. Induction of immunologic tolerance to cardiac allograft by simultaneous blockade of inducible co-stimulator and cytotoxic T-lymphocyte antigen 4 pathway. Transplantation. 2003; 75: 1374-1379.

Yoshinaga SK, Whoriskey JS, Khare SD, Sarmiento U, Guo J, Horan T, Shih G, Zhang M, Coccia MA, Kohno T, Tafuri-Bladt A, Brankow D, Campbell P, Chang D, Chiu L, Dai T, Duncan G, Elliott GS, Hui A, McCabe SM, Scully S, Shahinian A, Shaklee CL, Van G, Mak TW, Senaldi G. T-cell co-stimulation through B7RP-1 and ICOS. Nature. 1999; 402: 827-832.

Swallow MM, Wallin JJ, Sha WC. B7h, a novel costimulatory homolog of B7.1 and B7.2, is induced by TNFalpha. Immunity. 1999; 11: 423-432.

Khayyamian S, Hutloff A, Buchner K, Grafe M, Henn V, Kroczek RA, Mages HW. ICOS-ligand, expressed on human endothelial cells, costimulates Th1 and Th2 cytokine secretion by memory CD4+ T cells. Proc Natl Acad Sci U S A. 2002; 99: 6198-6203.

Isobe M, Yagita H, Okumura K, Ihara A. Specific acceptance of cardiac allograft after treatment with antibodies to ICAM-1 and LFA-1. Science. 1992; 255: 1125-1127.

Mach F, Schonbeck U, Sukhova GK, Atkinson E, Libby P. Reduction of atherosclerosis in mice by inhibition of CD40 signalling. Nature. 1998; 394: 200-203.

Buono C, Pang H, Uchida Y, Libby P, Sharpe AH, Lichtman AH. B7-1/B7-2 costimulation regulates plaque antigen-specific T-cell responses and atherogenesis in low-density lipoprotein receptor-deficient mice. Circulation. 2004; 109: 2009-2015.

Kim KS, Denton MD, Chandraker A, Knoflach A, Milord R, Waaga AM, Turka LA, Russell ME, Peach R, Sayegh MH. CD28-B7-mediated T cell costimulation in chronic cardiac allograft rejection: differential role of B7-1 in initiation versus progression of graft arteriosclerosis. Am J Pathol. 2001; 158: 977-986.

Wang CY, Mazer SP, Minamoto K, Takuma S, Homma S, Yellin M, Chess L, Fard A, Kalled SL, Oz MC, Pinsky DJ. Suppression of murine cardiac allograft arteriopathy by long-term blockade of CD40-CD154 interactions. Circulation. 2002; 105: 1609-1614.

Kosuge H, Suzuki J, Kakuta T, Haraguchi G, Koga N, Futamatsu H, Gotoh R, Inobe M, Isobe M, Uede T. Attenuation of graft arterial disease by manipulation of the LIGHT pathway. Arterioscler Thromb Vasc Biol. 2004; 24: 1409-1415.

Koga N, Suzuki J, Kosuge H, Haraguchi G, Onai Y, Futamatsu H, Maejima Y, Gotoh R, Saiki H, Tsushima F, Azuma M, Isobe M. Blockade of the interaction between PD-1 and PD-L1 accelerates graft arterial disease in cardiac allografts. Arterioscler Thromb Vasc Biol. 2004; 24: 2057-2062.

Sakamoto S, Tezuka K, Tsuji T, Hori N, Tamatani T. AILIM/ICOS: its expression and functional analysis with monoclonal antibodies. Hybrid Hybridomics. 2001; 20: 293-303.

Iiyama R, Kanai T, Uraushihara K, Totsuka T, Nakamura T, Miyata T, Yagita H, Kushi A, Suzuki K, Tezuka K, Watanabe M. The role of inducible co-stimulator (ICOS)/B7-related protein-1 (B7RP-1) interaction in the functional development of Peyer?s patches. Immunol Lett. 2003; 88: 63-70.

Sata M, Maejima Y, Adachi F, Fukino K, Saiura A, Sugiura S, Aoyagi T, Imai Y, Kurihara H, Kimura K, Omata M, Makuuchi M, Hirata Y, Nagai R. A mouse model of vascular injury that induces rapid onset of medial cell apoptosis followed by reproducible neointimal hyperplasia. J Mol Cell Cardiol. 2000; 32: 2097-2104.

Rectenwald JE, Moldawer LL, Huber TS, Seeger JM, Ozaki CK. Direct evidence for cytokine involvement in neointimal hyperplasia. Circulation. 2000; 102: 1697-1702.

Agrotis A, Kanellakis P, Kostolias G, Di Vitto G, Wei C, Hannan R, Jennings G, Bobik A. Proliferation of neointimal smooth muscle cells after arterial injury. Dependence on interactions between fibroblast growth factor receptor-2 and fibroblast growth factor-9. J Biol Chem. 2004; 279: 42221-42229.

Rolfe BE, Campbell JH, Smith NJ, Cheong MW, Campbell GR. T lymphocytes affect smooth muscle cell phenotype and proliferation. Arterioscler Thromb Vasc Biol. 1995; 15: 1204-1210.

Taylor DO, Edwards LB, Boucek MM, Trulock EP, Keck BM, Hertz MI. The registry of the International Society for Heat and Lung Transplantation: twenty-first official adult heart transplant report-2004. J Heart Lung Transplant. 2004; 23: 796-803.

Larsen CP, Elwood ET, Alexander DZ, Ritchie SC, Hendrix R, Tucker-Burden C, Cho HR, Aruffo A, Hollenbaugh D, Linsley PS, Winn KJ, Pearson TC. Long-term acceptance of skin and cardiac allografts after blocking CD40 and CD28 pathways. Nature. 1996; 381: 434-438.

Schonbeck U, Varo N, Libby P, Buring J, Ridker PM. Soluble CD40L and cardiovascular risk in women. Circulation. 2001; 104: 2266-2268.

Aukrust P, Muller F, Ueland T, Berget T, Aaser E, Brunsvig A, Solum NO, Forfang K, Froland SS, Gullestad L. Enhanced levels of soluble and membrane-bound CD40 ligand in patients with unstable angina. Possible reflection of T lymphocyte and platelet involvement in the pathogenesis of acute coronary syndromes. Circulation. 1999; 100: 614-620.

Cipollone F, Ferri C, Desideri G, Paloscia L, Materazzo G, Mascellanti M, Fazia M, Iezzi A, Cuccurullo C, Pini B, Bucci M, Santucci A, Cuccurullo F, Mezzetti A. Preprocedural level of soluble CD40L is predictive of enhanced inflammatory response and restenosis after coronary angioplasty. Circulation. 2003; 108: 2776-2782.

Dong C, Juedes AE, Temann U, Sherasta S, Allison JP, Ruddle NH, Flavell RA. ICOS co-stimulatory receptor is essential for T-cell activation and function. Nature. 2001; 409: 97-101.

Park H, Li Z, Yang XO, Chang SH, Nurieva R, Wang Y, Wang Y, Hood L, Zhu Z, Tian Q, Dong C. A distinct lineage of CD4 T cells regulates tissue inflammation by producing interleukin 17. Nat Immunol. 2005; 6: 1133-1141.

Tafuri A, Shahinian A, Bladt F, Yoshinaga SK, Jordana M, Wakeham A, Boucher L, Bouchard D, Chan VS, Duncan G, Odermatt B, Ho A, Itie A, Horan T, Whoriskey JS, Pawson T, Penninger JM, Ohashi PS, Mak TW. ICOS is essential for effective T-helper-cell responces. Nature. 2001; 409: 105-109.

Afek AHD, Roth A, Keren G, George J. A functional role for inducible costimulator (ICOS) in atherosclerosis. Atherosclerosis. 2005; 183: 57-63.


作者单位:Department of Cardiovascular Medicine (H.K., J.S., G.H., N.K., Y.M., M. Isobe), Tokyo Medical and Dental University, Bunkyo-ku, Tokyo, Japan; and the Division of Molecular Immunology (M. Inobe, T.U.), Institute for Genetic Medicine, Hokkaido University, Kita-ku, Sapporo, Japan

作者: Hisanori Kosuge; Jun-ichi Suzuki; Go Haraguchi; No
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