Informacja

Drogi użytkowniku, aplikacja do prawidłowego działania wymaga obsługi JavaScript. Proszę włącz obsługę JavaScript w Twojej przeglądarce.

Tytuł pozycji:

Costimulatory and coinhibitory molecules of B7-CD28 family in cardiovascular atherosclerosis: A review.

Tytuł:
Costimulatory and coinhibitory molecules of B7-CD28 family in cardiovascular atherosclerosis: A review.
Autorzy:
Yang M; Department of Cardiology, Electrophysiological Center of Cardiology, The Fourth Affiliated Hospital of Harbin Medical University, Harbin, China.
Tian S; Basic Medicine College, Harbin Medical University, Harbin, China.
Lin Z; State Key Laboratory of Medicinal Chemical Biology and College of Pharmacy, Nankai University, Tianjin, China.
Fu Z; Basic Medicine College, Harbin Medical University, Harbin, China.; State Key Laboratory of Medicinal Chemical Biology and College of Pharmacy, Nankai University, Tianjin, China.; Department of Immunology, Wu Lien-Teh Institute, Heilongjiang Provincial Key Laboratory for Infection and Immunity, Harbin Medical University, Heilongjiang Academy of Medical Science, Harbin, China.
Li C; State Key Laboratory of Medicinal Chemical Biology and College of Pharmacy, Nankai University, Tianjin, China.
Źródło:
Medicine [Medicine (Baltimore)] 2022 Nov 11; Vol. 101 (45), pp. e31667.
Typ publikacji:
Review; Journal Article
Język:
English
Imprint Name(s):
Original Publication: Hagerstown, Md : Lippincott Williams & Wilkins
MeSH Terms:
Atherosclerosis*/drug therapy
Atherosclerosis*/immunology
Cardiovascular System*/immunology
CD28 Antigens*/immunology
Animals ; Mice ; Inflammation/immunology ; T-Lymphocytes/immunology
References:
Libby P. Inflammation in atherosclerosis. Arterioscler Thromb Vasc Biol. 2012;32:2045–51.
Andersson J, Libby P, Hansson GK. Adaptive immunity and atherosclerosis. Clin Immunol. 2010;134:33–46.
Sadat U, Jaffer FA, van Zandvoort MA, et al. Inflammation and neovascularization intertwined in atherosclerosis. imaging of structural and molecular imaging targets. Circulation. 2014;130:786–94.
Wildgruber M, Swirski FK, Zernecke A. Molecular imaging of inflammation in atherosclerosis. Theranostics. 2013;3:865–84.
Viola J, Soehnlein O. Atherosclerosis - A matter of unresolved inflammation. Semin Immunol. 2015;27:184–93.
Reardon CA, Blachowicz L, White T, et al. Effect of immune deficiency on lipoproteins and atherosclerosis in male apolipoprotein E-deficient mice. Arterioscler Thromb Vasc Biol. 2001;21:1011–6.
Buono C, Come CE, Stavrakis G, et al. Influence of interferon-gamma on the extent and phenotype of diet-induced atherosclerosis in the LDLR-deficient mouse. Arterioscler Thromb Vasc Biol. 2003;23:454–60.
Bruen R, Curley S, Kajani S, et al. Liraglutide attenuates preestablished atherosclerosis in apolipoprotein E-deficient mice via regulation of immune cell phenotypes and proinflammatory mediators. J Pharmacol Exp Ther. 2019;370:447–58.
Weber C, Zernecke A, Libby P. The multifaceted contributions of leukocyte subsets to atherosclerosis. lessons from mouse models. Nat Rev Immunol. 2008;8:802–15.
Robbins CS, Hilgendorf I, Weber GF, et al. Local proliferation dominates lesional macrophage accumulation in atherosclerosis. Nat Med. 2013;19:1166–72.
Millonig G, Niederegger H, Rabl W, et al. Network of vascular-associated dendritic cells in intima of healthy young individuals. Arterioscler Thromb Vasc Biol. 2001;21:503–8.
Moore KJ, Tabas I. Macrophages in the pathogenesis of atherosclerosis. Cell. 2011;145:341–55.
Watts TH, DeBenedette MA. T cell co-stimulatory molecules other than CD28. Curr Opin Immunol. 1999;11:286–93.
Abbas AK, Lichtman AH, Pillai S. Cellular and molecular immunology. 7th edn. Philadephia: Elsevier Saunders, 2011.
Croft M. Co-stimulatory members of the TNFR family. keys to effective T-cell immunity? Nat Rev Immunol. 2003;3:609–20.
Podojil JR, Miller SD. Targeting the B7 family of co-stimulatory molecules. successes and challenges. BioDrugs. 2013;27:1–13.
Sharpe AH, Freeman GJ. The B7-CD28 superfamily. Nat Rev Immunol. 2002;2:116–26.
Wang S, Chen L. T lymphocyte co-signaling pathways of the B7-CD28 family. Cell Mol Immunol. 2004;1:37–42.
Sharpe AH. Mechanisms of co-stimulation. Immunol Rev. 2009;229:5–11.
Carreno BM, Collins M. The B7 family of ligands and its receptors. new pathways for co-stimulation and inhibition of immune responses. Annu Rev Immunol. 2002;20:29–53.
Krzyzak L, Seitz C, Urbat A, et al. CD83 modulates B cell activation and germinal center responses. J Immunol. 2016;196:3581–94.
Fujimoto Y, Tu L, Miller AS, et al. CD83 expression influences CD4+ T cell development in the thymus. Cell. 2002;108:755–67.
von Rohrscheidt J, Petrozziello E, Nedjic J, et al. Thymic CD4 T cell selection requires attenuation of March8-mediated MHCII turnover in cortical epithelial cells through CD83. J Exp Med. 2016;213:1685–94.
Chen L, Zhu Y, Zhang G, et al. CD83-stimulated monocytes suppress T-cell immune responses through production of prostaglandin E2. Proc Natl Acad Sci USA. 2011;108:18778–83.
Horvatinovich JM, Grogan EW, Norris M, et al. Soluble CD83 inhibits T cell activation by binding to the TLR4/MD-2 complex on CD14(+) monocytes. J Immunol. 2017;198:2286–301.
Hutloff A, Dittrich AM, Beier KC, et al. ICOS is an inducible T-cell co-stimulator structurally and functionally related to CD28. Nature. 1999;397:263–6.
Yoshinaga SK, Whoriskey JS, Khare SD, et al. T-cell co-stimulation through B7RP-1 and ICOS. Nature. 1999;402:827–32.
Wang S, Zhu G, Chapoval AI, et al. co-stimulation of T cells by B7-H2, a B7-like molecule that binds ICOS. Blood. 2000;96:2808–13.
van Berkel ME, Oosterwegel MA. CD28 and ICOS. similar or separate co-stimulators of T cells? Immunol Lett. 2006;105:115–22.
Swallow MM, Wallin JJ, Sha WC. B7h, a novel co-stimulatory homolog of B7.1 and B7.2, is induced by TNFalpha. Immunity. 1999;11:423–32.
Simpson TR, Quezada SA, Allison JP. Regulation of CD4 T cell activation and effector function by inducible co-stimulator (ICOS). Curr Opin Immunol. 2010;22:326–32.
Smith KM, Brewer JM, Webb P, et al. Inducible co-stimulatory molecule-B7-related protein 1 interactions are important for the clonal expansion and B cell helper functions of naive, Th1, and Th2 T cells. J Immunol. 2003;170:2310–5.
Khayyamian S, Hutloff A, Buchner K, et al. ICOS-ligand, expressed on human endothelial cells, co-stimulates Th1 and Th2 cytokine secretion by memory CD4+ T cells. Proc Natl Acad Sci USA. 2002;99:6198–203.
Lohning M, Hutloff A, Kallinich T, et al. Expression of ICOS in vivo defines CD4+ effector T cells with high inflammatory potential and a strong bias for secretion of interleukin 10. J Exp Med. 2003;197:181–93.
Vieira PL, Wassink L, Smith LM, et al. ICOS-mediated signaling regulates cytokine production by human T cells and provides a unique signal to selectively control the clonal expansion of Th2 helper cells. Eur J Immunol. 2004;34:1282–90.
Herman AE, Freeman GJ, Mathis D, et al. CD4+CD25+ T regulatory cells dependent on ICOS promote regulation of effector cells in the prediabetic lesion. J Exp Med. 2004;199:1479–89.
Nie X, Cai G, Zhang W, et al. Lipopolysaccharide mediated mast cells induce IL-10 producing regulatory T cells through the ICOSL/ICOS axis. Clin Immunol. 2012;142:269–79.
Chapoval AI, Ni J, Lau JS, et al. B7-H3. a co-stimulatory molecule for T cell activation and IFN-gamma production. Nat Immunol. 2001;2:269–74.
Saatian B, Yu XY, Lane AP, et al. Expression of genes for B7-H3 and other T cell ligands by nasal epithelial cells during differentiation and activation. Am J Physiol Lung Cell Mol Physiol. 2004;287:L217–225.
Suh WK, Wang SX, Jheon AH, et al. The immune regulatory protein B7-H3 promotes osteoblast differentiation and bone mineralization. Proc Natl Acad Sci USA. 2004;101:12969–73.
Stanciu LA, Bellettato CM, Laza-Stanca V, et al. Expression of programmed death-1 ligand (PD-L) 1, PD-L2, B7-H3, and inducible co-stimulator ligand on human respiratory tract epithelial cells and regulation by respiratory syncytial virus and type 1 and 2 cytokines. J Infect Dis. 2006;193:404–12.
Wu CP, Jiang JT, Tan M, et al. Relationship between co-stimulatory molecule B7-H3 expression and gastric carcinoma histology and prognosis. World J Gastroenterol. 2006;12:457–9.
Zhou YH, Chen YJ, Ma ZY, et al. 4IgB7-H3 is the major isoform expressed on immunocytes as well as malignant cells. Tissue Antigens. 2007;70:96–104.
Roth TJ, Sheinin Y, Lohse CM, et al. B7-H3 ligand expression by prostate cancer. a novel marker of prognosis and potential target for therapy. Cancer Res. 2007;67:7893–900.
Zang X, Thompson RH, Al-Ahmadie HA, et al. B7-H3 and B7x are highly expressed in human prostate cancer and associated with disease spread and poor outcome. Proc Natl Acad Sci USA. 2007;104:19458–63.
Luo L, Chapoval AI, Flies DB, et al. B7-H3 enhances tumor immunity in vivo by co-stimulating rapid clonal expansion of antigen-specific CD8+ cytolytic T cells. J Immunol. 2004;173:5445–50.
Lupu CM, Eisenbach C, Kuefner MA, et al. An orthotopic colon cancer model for studying the B7-H3 antitumor effect in vivo. J Gastrointest Surg. 2006;10:635–45.
Ma L, Luo L, Qiao H, et al. Complete eradication of hepatocellular carcinomas by combined vasostatin gene therapy and B7H3-mediated immunotherapy. J Hepatol. 2007;46:98–106.
Hashiguchi M, Kobori H, Ritprajak P, et al. Triggering receptor expressed on myeloid cell-like transcript 2 (TLT-2) is a counter-receptor for B7-H3 and enhances T cell responses. Proc Natl Acad Sci USA. 2008;105:10495–500.
Kobori H, Hashiguchi M, Piao J, et al. Enhancement of effector CD8+ T-cell function by tumour-associated B7-H3 and modulation of its counter-receptor triggering receptor expressed on myeloid cell-like transcript 2 at tumour sites. Immunology. 2010;130:363–73.
Brandt CS, Baratin M, Yi EC, et al. The B7 family member B7-H6 is a tumor cell ligand for the activating natural killer cell receptor NKp30 in humans. J Exp Med. 2009;206:1495–503.
Matta J, Baratin M, Chiche L, et al. Induction of B7-H6, a ligand for the natural killer cell-activating receptor NKp30, in inflammatory conditions. Blood. 2013;122:394–404.
Zhu Y, Yao S, Iliopoulou BP, et al. B7-H5 co-stimulates human T cells via CD28H. Nat Commun. 2013;4:2043.
Takahashi T, Tagami T, Yamazaki S, et al. Immunologic self-tolerance maintained by CD25(+)CD4(+) regulatory T cells constitutively expressing cytotoxic T lymphocyte-associated antigen 4. J Exp Med. 2000;192:303–10.
Teft WA, Kirchhof MG, Madrenas J. A molecular perspective of CTLA-4 function. Annu Rev Immunol. 2006;24:65–97.
Krummey SM, Cheeseman JA, Conger JA, et al. High CTLA-4 expression on Th17 cells results in increased sensitivity to CTLA-4 co-inhibition and resistance to belatacept. Am J Transplant. 2014;14:607–14.
Ishida Y, Agata Y, Shibahara K, et al. Induced expression of PD-1, a novel member of the immunoglobulin gene superfamily, upon programmed cell death. EMBO J. 1992;11:3887–95.
Freeman GJ, Long AJ, Iwai Y, et al. Engagement of the PD-1 immunoinhibitory receptor by a novel B7 family member leads to negative regulation of lymphocyte activation. J Exp Med. 2000;192:1027–34.
Chemnitz JM, Parry RV, Nichols KE, et al. SHP-1 and SHP-2 associate with immunoreceptor tyrosine-based switch motif of programmed death 1 upon primary human T cell stimulation, but only receptor ligation prevents T cell activation. J Immunol. 2004;173:945–54.
Chen L. Co-inhibitory molecules of the B7-CD28 family in the control of T-cell immunity. Nat Rev Immunol. 2004;4:336–47.
Yao S, Wang S, Zhu Y, et al. PD-1 on dendritic cells impedes innate immunity against bacterial infection. Blood. 2009;113:5811–8.
Said EA, Dupuy FP, Trautmann L, et al. Programmed death-1-induced interleukin-10 production by monocytes impairs CD4+ T cell activation during HIV infection. Nat Med. 2010;16:452–9.
Yao S, Chen L. PD-1 as an immune modulatory receptor. Cancer J. 2014;20:262–4.
Lemke D, Pfenning PN, Sahm F, et al. co-stimulatory protein 4IgB7H3 drives the malignant phenotype of glioblastoma by mediating immune escape and invasiveness. Clin Cancer Res. 2012;18:105–17.
Chen C, Shen Y, Qu QX, et al. Induced expression of B7-H3 on the lung cancer cells and macrophages suppresses T-cell mediating anti-tumor immune response. Exp Cell Res. 2013;319:96–102.
Liu C, Liu J, Wang J, et al. B7-H3 expression in ductal and lobular breast cancer and its association with IL-10. Mol Med Rep. 2013;7:134–8.
Choi IH, Zhu G, Sica GL, et al. Genomic organization and expression analysis of B7-H4, an immune inhibitory molecule of the B7 family. J Immunol. 2003;171:4650–4.
Sica GL, Choi IH, Zhu G, et al. B7-H4, a molecule of the B7 family, negatively regulates T cell immunity. Immunity. 2003;18:849–61.
Zang X, Loke P, Kim J, et al. B7x. a widely expressed B7 family member that inhibits T cell activation. Proc Natl Acad Sci USA. 2003;100:10388–92.
Zhu G, Augustine MM, Azuma T, et al. B7-H4-deficient mice display augmented neutrophil-mediated innate immunity. Blood. 2009;113:1759–67.
Compaan DM, Gonzalez LC, Tom I, et al. Attenuating lymphocyte activity. the crystal structure of the BTLA-HVEM complex. J Biol Chem. 2005;280:39553–61.
Sedy JR, Gavrieli M, Potter KG, et al. B and T lymphocyte attenuator regulates T cell activation through interaction with herpesvirus entry mediator. Nat Immunol. 2005;6:90–8.
Watanabe N, Gavrieli M, Sedy JR, et al. BTLA is a lymphocyte inhibitory receptor with similarities to CTLA-4 and PD-1. Nat Immunol. 2003;4:670–9.
Carreno BM, Collins M. BTLA. a new inhibitory receptor with a B7-like ligand. Trends Immunol. 2003;24:524–7.
Shui JW, Steinberg MW, Kronenberg M. Regulation of inflammation, autoimmunity, and infection immunity by HVEM-BTLA signaling. J Leukoc Biol. 2011;89:517–23.
Bai Y, Shi Y, Li Y, et al. Sirolimus-based regimen promotes inhibitory co-stimulatory signal of HVEM/BTLA/CD160/LIGHT pathway in allo-renal recipients. Transpl Immunol. 2013;28:38–47.
Krieg C, Han P, Stone R, et al. Functional analysis of B and T lymphocyte attenuator engagement on CD4+ and CD8+ T cells. J Immunol. 2005;175:6420–7.
Wang L, Rubinstein R, Lines JL, et al. VISTA, a novel mouse Ig superfamily ligand that negatively regulates T cell responses. J Exp Med. 2011;208:577–92.
Lines JL, Pantazi E, Mak J, et al. VISTA is an immune checkpoint molecule for human T cells. Cancer Res. 2014;74:1924–32.
Flies DB, Han X, Higuchi T, et al. Coinhibitory receptor PD-1H preferentially suppresses CD4⁺T cell-mediated immunity. J Clin Invest. 2014;124:1966–75.
Zhao R, Chinai JM, Buhl S, et al. HHLA2 is a member of the B7 family and inhibits human CD4 and CD8 T-cell function. Proc Natl Acad Sci USA. 2013;110:9879–84.
Glass CK, Witztum JL. Atherosclerosis. The road ahead. Cell. 2001;104:503–16.
Tabas I, Williams KJ, Boren J. Subendothelial lipoprotein retention as the initiating process in atherosclerosis. update and therapeutic implications. Circulation. 2007;116:1832–44.
Zhu SN, Chen M, Jongstra-Bilen J, et al. GM-CSF regulates intimal cell proliferation in nascent atherosclerotic lesions. J Exp Med. 2009;206:2141–9.
Miller YI, Choi SH, Wiesner P, et al. Oxidation-specific epitopes are danger-associated molecular patterns recognized by pattern recognition receptors of innate immunity. Circ Res. 2011;108:235–48.
van Gils JM, Derby MC, Fernandes LR, et al. The neuroimmune guidance cue netrin-1 promotes atherosclerosis by inhibiting the emigration of macrophages from plaques. Nat Immunol. 2012;13:136–43.
Wanschel A, Seibert T, Hewing B, et al. Neuroimmune guidance cue Semaphorin 3E is expressed in atherosclerotic plaques and regulates macrophage retention. Arterioscler Thromb Vasc Biol. 2013;33:886–93.
Ramkhelawon B, Yang Y, van Gils JM, et al. Hypoxia induces netrin-1 and Unc5b in atherosclerotic plaques. mechanism for macrophage retention and survival. Arterioscler Thromb Vasc Biol. 2013;33:1180–8.
Koltsova EK, Garcia Z, Chodaczek G, et al. Dynamic T cell-APC interactions sustain chronic inflammation in atherosclerosis. J Clin Invest. 2012;122:3114–26.
Peled M, Fisher EA. Dynamic aspects of macrophage polarization during atherosclerosis progression and regression. Front Immunol. 2014;5:579.
Nickel T, Schmauss D, Hanssen H, et al. oxLDL uptake by dendritic cells induces upregulation of scavenger-receptors, maturation and differentiation. Atherosclerosis. 2009;205:442–50.
Perrin-Cocon L, Coutant F, Agaugue S, et al. Oxidized low-density lipoprotein promotes mature dendritic cell transition from differentiating monocyte. J Immunol. 2001;167:3785–91.
Wu H, Gower RM, Wang H, et al. Functional role of CD11c+ monocytes in atherogenesis associated with hypercholesterolemia. Circulation. 2009;119:2708–17.
Gautier EL, Huby T, Saint-Charles F, et al. Conventional dendritic cells at the crossroads between immunity and cholesterol homeostasis in atherosclerosis. Circulation. 2009;119:2367–75.
Paulson KE, Zhu SN, Chen M, et al. Resident intimal dendritic cells accumulate lipid and contribute to the initiation of atherosclerosis. Circ Res. 2010;106:383–90.
Buono C, Pang H, Uchida Y, et al. B7-1/B7-2 co-stimulation regulates plaque antigen-specific T-cell responses and atherogenesis in low-density lipoprotein receptor-deficient mice. Circulation. 2004;109:2009–15.
de Boer OJ, Hirsch F, van der Wal AC, et al. co-stimulatory molecules in human atherosclerotic plaques. an indication of antigen specific T lymphocyte activation. Atherosclerosis. 1997;133:227–34.
Afek A, Harats D, Roth A, et al. Evidence for the involvement of T cell co-stimulation through the B-7/CD28 pathway in atherosclerotic plaques from apolipoprotein E knockout mice. Exp Mol Pathol. 2004;76:219–23.
Dopheide JF, Sester U, Schlitt A, et al. Monocyte-derived dendritic cells of patients with coronary artery disease show an increased expression of co-stimulatory molecules CD40, CD80 and CD86 in vitro. Coron Artery Dis. 2007;18:523–31.
Muller A, Mu L, Meletta R, et al. Towards non-invasive imaging of vulnerable atherosclerotic plaques by targeting co-stimulatory molecules. Int J Cardiol. 2014;174:503–15.
Ait-Oufella H, Salomon BL, Potteaux S, et al. Natural regulatory T cells control the development of atherosclerosis in mice. Nat Med. 2006;12:178–80.
Doesch AO, Zhao L, Gleissner CA, et al. Inhibition of B7-1 (CD80) by RhuDex(R) reduces lipopolysaccharide-mediated inflammation in human atherosclerotic lesions. Drug Des Devel Ther. 2014;8:447–57.
Meletta R, Steier L, Borel N, et al. CD80 is upregulated in a mouse model with shear stress-induced atherosclerosis and allows for evaluating CD80-targeting PET tracers. Mol Imaging Biol. 2017;19:90–9.
Rohm I, Atiskova Y, Drobnik S, et al. Decreased regulatory T cells in vulnerable atherosclerotic lesions. imbalance between pro- and anti-inflammatory cells in atherosclerosis. Mediators Inflamm. 2015;2015:364710.
Afek A, Harats D, Roth A, Keren G, et al. A functional role for inducible co-stimulator (ICOS) in atherosclerosis. Atherosclerosis. 2005;183:57–63.
Kim WJ, Kang YJ, Suk K, et al. Comparative analysis of the expression patterns of various TNFSF/TNFRSF in atherosclerotic plaques. Immunol Invest. 2008;37:359–73.
Niinisalo P, Oksala N, Levula M, et al. Activation of indoleamine 2,3-dioxygenase-induced tryptophan degradation in advanced atherosclerotic plaques. Tampere vascular study. Ann Med. 2010;42:55–63.
Gotsman I, Grabie N, Gupta R, et al. Impaired regulatory T-cell response and enhanced atherosclerosis in the absence of inducible co-stimulatory molecule. Circulation. 2006;114:2047–55.
Ghourbani Gazar S, Andalib A, Hashemi M, et al. CD4(+)Foxp3(+) Treg and its ICOS(+) subsets in patients with myocardial infarction. Iran J Immunol. 2012;9:53–60.
Gotsman I, Sharpe AH, Lichtman AH. T-cell co-stimulation and co-inhibition in atherosclerosis. Circ Res. 2008;103:1220–31.
Clement M, Guedj K, Andreata F, et al. Control of the T follicular helper-germinal center B-cell axis by CD8(+) regulatory T cells limits atherosclerosis and tertiary lymphoid organ development. Circulation. 2015;13:560–70.
Zhong Z, Zhang Q, Tan L, et al. T cell co-stimulator inducible co-stimulatory (ICOS) exerts potential anti-atherosclerotic roles through downregulation of vascular smooth muscle phagocytosis and proliferation. Ann Transl Med. 2020;8:1597.
Bonaccorsi I, Spinelli D, Cantoni C, et al. Symptomatic carotid atherosclerotic plaques are associated with increased infiltration of natural killer (NK) cells and higher serum levels of NK activating receptor ligands. Front Immunol. 2019;10:1503.
Choi SH, Kim J, Gonen A, et al. MD-2 binds cholesterol. Biochem Biophys Res Commun. 2016;470:877–80.
Zimmer S, Grebe A, Latz E. Danger signaling in atherosclerosis. Circ Res. 2015;116:323–40.
Chen T, Huang W, Qian J, et al. Macrophage-derived myeloid differentiation protein 2 plays an essential role in ox-LDL-induced inflammation and atherosclerosis. EBioMedicine. 2020;53:102706.
Chen S, Crother TR. MD-2 as a possible therapeutic target for atherosclerosis. EBioMedicine. 2020;55:102760.
Gotsman I, Grabie N, Dacosta R, et al. Proatherogenic immune responses are regulated by the PD-1/PD-L pathway in mice. J Clin Invest. 2007;117:2974–82.
Ewing MM, Karper JC, Abdul S, et al. T-cell co-stimulation by CD28-CD80/86 and its negative regulator CTLA-4 strongly influence accelerated atherosclerosis development. Int J Cardiol. 2013;168:1965–74.
Ma K, Lv S, Liu B, et al. CTLA4-IgG ameliorates homocysteine-accelerated atherosclerosis by inhibiting T-cell overactivation in apoE(-/-) mice. Cardiovasc Res. 2013;97:349–59.
Matsumoto T, Sasaki N, Yamashita T, et al. Overexpression of cytotoxic T-lymphocyte-associated antigen-4 prevents atherosclerosis in Mice. Arterioscler Thromb Vasc Biol. 2016;36:1141–51.
Vinh A, Chen W, Blinder Y, et al. Inhibition and genetic ablation of the B7/CD28 T-cell co-stimulation axis prevents experimental hypertension. Circulation. 2010;122:2529–37.
Rodig N, Ryan T, Allen JA, et al. Endothelial expression of PD-L1 and PD-L2 down-regulates CD8+ T cell activation and cytolysis. Eur J Immunol. 2003;33:3117–26.
Grabie N, Gotsman I, DaCosta R, et al. Endothelial programmed death-1 ligand 1 (PD-L1) regulates CD8+ T-cell mediated injury in the heart. Circulation. 2007;116:2062–71.
Day CL, Kaufmann DE, Kiepiela P, et al. PD-1 expression on HIV-specific T cells is associated with T-cell exhaustion and disease progression. Nature. 2006;443:350–4.
Bu DX, Tarrio M, Maganto-Garcia E, et al. Impairment of the programmed cell death-1 pathway increases atherosclerotic lesion development and inflammation. Arterioscler Thromb Vasc Biol. 2011;31:1100–7.
Qiu MK, Wang SC, Dai YX, et al. PD-1 and Tim-3 pathways regulate CD8+ T cells function in atherosclerosis. PLoS One. 2015;10:e0128523.
Weyand CM, Berry GJ, Goronzy JJ. The immunoinhibitory PD-1/PD-L1 pathway in inflammatory blood vessel disease. J Leukoc Biol. 2018;103:565–75.
Ding R, Gao W, He Z, et al. Circulating CD4(+)CXCR5(+) T cells contribute to proinflammatory responses in multiple ways in coronary artery disease. Int Immunopharmacol. 2017;52:318–23.
Eppihimer MJ, Gunn J, Freeman GJ, et al. Expression and regulation of the PD-L1 immunoinhibitory molecule on microvascular endothelial cells. Microcirculation. 2002;9:133–45.
Lee J, Zhuang Y, Wei X, et al. Contributions of PD-1/PD-L1 pathway to interactions of myeloid DCs with T cells in atherosclerosis. J Mol Cell Cardiol. 2009;46:169–76.
Shi B, Du X, Wang Q, et al. Increased PD-1 on CD4(+)CD28(-) T cell and soluble PD-1 ligand-1 in patients with T2DM. association with atherosclerotic macrovascular diseases. Metabolism. 2013;62:778–85.
Dietel B, Cicha I, Voskens CJ, et al. Decreased numbers of regulatory T cells are associated with human atherosclerotic lesion vulnerability and inversely correlate with infiltrated mature dendritic cells. Atherosclerosis. 2013;230:92–9.
Chen WJ, Hu XF, Yan M, et al. Human umbilical vein endothelial cells promote the inhibitory activation of CD4(+)CD25(+)Foxp3(+) regulatory T cells via PD-L1. Atherosclerosis. 2016;244:108–12.
Cochain C, Chaudhari SM, Koch M, et al. Programmed cell death-1 deficiency exacerbates T cell activation and atherogenesis despite expansion of regulatory T cells in atherosclerosis-prone mice. PLoS One. 2014;9:e93280.
Sandberg WJ, Halvorsen B, Yndestad A, et al. Inflammatory interaction between LIGHT and proteinase-activated receptor-2 in endothelial cells. potential role in atherogenesis. Circ Res. 2009;104:60–8.
Holven KB, Narverud I, Lindvig HW, et al. Subjects with familial hypercholesterolemia are characterized by an inflammatory phenotype despite long-term intensive cholesterol lowering treatment. Atherosclerosis. 2014;233:561–7.
Douna H, Amersfoort J, Schaftenaar FH, et al. B- and T-lymphocyte attenuator stimulation protects against atherosclerosis by regulating follicular B cells. Cardiovasc Res. 2020;116:295–305.
Mallat Z, Taleb S, Ait-Oufella H, et al. The role of adaptive T cell immunity in atherosclerosis. J Lipid Res. 2009;50(Suppl):S364–9.
Ketelhuth DF, Gistera A, Johansson DK, et al. T cell-based therapies for atherosclerosis. Curr Pharm Des. 2013;19:5850–8.
Zeller I, Srivastava S. Macrophage functions in atherosclerosis. Circ Res. 2014;115:e83–5.
Colin S, Chinetti-Gbaguidi G, Staels B. Macrophage phenotypes in atherosclerosis. Immunol Rev. 2014;262:153–66.
Chinetti-Gbaguidi G, Colin S, Staels B. Macrophage subsets in atherosclerosis. Nat Rev Cardiol. 2015;12:10–7.
Taghavie-Moghadam PL, Butcher MJ, et al. The dynamic lives of macrophage and dendritic cell subsets in atherosclerosis. Ann N Y Acad Sci. 2014;1319:19–37.
Alberts-Grill N, Denning TL, Rezvan A, et al. The role of the vascular dendritic cell network in atherosclerosis. Am J Physiol Cell Physiol. 2013;305:C1–21.
Gerli R, Schillaci G, Giordano A, et al. CD4+CD28- T lymphocytes contribute to early atherosclerotic damage in rheumatoid arthritis patients. Circulation. 2004;109:2744–8.
Liuzzo G, Biasucci LM, Trotta G, et al. Unusual CD4+CD28null T lymphocytes and recurrence of acute coronary events. J Am Coll Cardiol. 2007;50:1450–8.
Meletta R, Muller Herde A, Dennler P, et al. Preclinical imaging of the co-stimulatory molecules CD80 and CD86 with indium-111-labeled belatacept in atherosclerosis. EJNMMI Res. 2016;6:1.
Ford ML, Larsen CP. Translating co-stimulation blockade to the clinic. lessons learned from three pathways. Immunol Rev. 2009;229:294–306.
Shen Y, Xu LR, Tang X, et al. Identification of potential therapeutic targets for atherosclerosis by analysing the gene signature related to different immune cells and immune regulators in atheromatous plaques. BMC Med Genomics. 2021;14:145.
Substance Nomenclature:
0 (CD28 Antigens)
Entry Date(s):
Date Created: 20221118 Date Completed: 20221128 Latest Revision: 20230103
Update Code:
20240104
PubMed Central ID:
PMC9666218
DOI:
10.1097/MD.0000000000031667
PMID:
36397436
Czasopismo naukowe
Accumulating evidence supports the active involvement of vascular inflammation in atherosclerosis pathogenesis. Vascular inflammatory events within atherosclerotic plaques are predominated by innate antigen-presenting cells (APCs), including dendritic cells, macrophages, and adaptive immune cells such as T lymphocytes. The interaction between APCs and T cells is essential for the initiation and progression of vascular inflammation during atherosclerosis formation. B7-CD28 family members that provide either costimulatory or coinhibitory signals to T cells are important mediators of the cross-talk between APCs and T cells. The balance of different functional members of the B7-CD28 family shapes T cell responses during inflammation. Recent studies from both mouse and preclinical models have shown that targeting costimulatory molecules on APCs and T cells may be effective in treating vascular inflammatory diseases, especially atherosclerosis. In this review, we summarize recent advances in understanding how APC and T cells are involved in the pathogenesis of atherosclerosis by focusing on B7-CD28 family members and provide insight into the immunotherapeutic potential of targeting B7-CD28 family members in atherosclerosis.
Competing Interests: The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as potential conflicts of interest. The authors have no conflicts of interest to disclose.
(Copyright © 2022 the Author(s). Published by Wolters Kluwer Health, Inc.)

Ta witryna wykorzystuje pliki cookies do przechowywania informacji na Twoim komputerze. Pliki cookies stosujemy w celu świadczenia usług na najwyższym poziomie, w tym w sposób dostosowany do indywidualnych potrzeb. Korzystanie z witryny bez zmiany ustawień dotyczących cookies oznacza, że będą one zamieszczane w Twoim komputerze. W każdym momencie możesz dokonać zmiany ustawień dotyczących cookies