Myocardial infarction accelerates atherosclerosis

Partha Dutta, Gabriel Courties, Ying Wei, Florian Leuschner, Rostic Gorbatov, Clinton S Robbins, Yoshiko Iwamoto, Brian Thompson, Alicia L Carlson, Timo Heidt, Maulik D Majmudar, Felix Lasitschka, Martin Etzrodt, Peter Waterman, Michael T Waring, Adam T Chicoine, Anja M van der Laan, Hans W M Niessen, Jan J Piek, Barry B Rubin, Jagdish Butany, James R Stone, Hugo A Katus, Sabina A Murphy, David A Morrow, Marc S Sabatine, Claudio Vinegoni, Michael A Moskowitz, Mikael J Pittet, Peter Libby, Charles P Lin, Filip K Swirski, Ralph Weissleder, Matthias Nahrendorf, Partha Dutta, Gabriel Courties, Ying Wei, Florian Leuschner, Rostic Gorbatov, Clinton S Robbins, Yoshiko Iwamoto, Brian Thompson, Alicia L Carlson, Timo Heidt, Maulik D Majmudar, Felix Lasitschka, Martin Etzrodt, Peter Waterman, Michael T Waring, Adam T Chicoine, Anja M van der Laan, Hans W M Niessen, Jan J Piek, Barry B Rubin, Jagdish Butany, James R Stone, Hugo A Katus, Sabina A Murphy, David A Morrow, Marc S Sabatine, Claudio Vinegoni, Michael A Moskowitz, Mikael J Pittet, Peter Libby, Charles P Lin, Filip K Swirski, Ralph Weissleder, Matthias Nahrendorf

Abstract

During progression of atherosclerosis, myeloid cells destabilize lipid-rich plaques in the arterial wall and cause their rupture, thus triggering myocardial infarction and stroke. Survivors of acute coronary syndromes have a high risk of recurrent events for unknown reasons. Here we show that the systemic response to ischaemic injury aggravates chronic atherosclerosis. After myocardial infarction or stroke, Apoe-/- mice developed larger atherosclerotic lesions with a more advanced morphology. This disease acceleration persisted over many weeks and was associated with markedly increased monocyte recruitment. Seeking the source of surplus monocytes in plaques, we found that myocardial infarction liberated haematopoietic stem and progenitor cells from bone marrow niches via sympathetic nervous system signalling. The progenitors then seeded the spleen, yielding a sustained boost in monocyte production. These observations provide new mechanistic insight into atherogenesis and provide a novel therapeutic opportunity to mitigate disease progression.

Conflict of interest statement

COMPETING FINANCIAL INTERESTS

Marc S. Sabatine, David A. Morrow and Sabina A. Murphy received grant support from AstraZeneca and GSK. The remaining authors declare no competing financial interests.

Figures

Figure 1. Increased inflammation in atherosclerotic plaques…
Figure 1. Increased inflammation in atherosclerotic plaques after MI
a, Protease activity by FMT-CT before and 3 weeks after MI. Circles indicate aortic root (n = 10 per group). b, Protease activity in excised aortae by fluorescence reflectance imaging (FRI), expressed as target to background ratio (TBR, n = 10 per group). c, Flow cytometric quantification of myeloid cells and Ly-6Chigh monocytes in aorta (n = 5–9 per group). Dot plots 3 weeks after MI are shown. d, CD11b staining and lesion size (n = 9–10 per group). Scale bar represents 150 μm. Mean ± s.e.m., * P < 0.05, ** P < 0.01.
Figure 2. Elevated levels of progenitor cells…
Figure 2. Elevated levels of progenitor cells in the spleen of apoE−/− mice after MI
a, Quantification for HSPCs, MDPs, and GMPs at different time points after MI (n = 3–15 per group). The gating strategy is shown in Supplementary Fig. 10. b, Number of colony-forming units. Mean ± s.e.m., * P < 0.05, ** P < 0.01.
Figure 3. β 3 adrenergic receptor-mediated progenitor…
Figure 3. β3 adrenergic receptor-mediated progenitor release after MI
a, Flow cytometric analyses of HSPC in blood of C57BL/6 mice (n = 6–11 per group). b, Immunostaining for tyrosine hydroxylase (TH). Scale bar represents 10μm. Insets depict low magnification overview. Bar graph shows quantitation of TH+ area (n = 5 per group). c, Expression of HSPC retention factors (relative to Gapdh) in the bone marrow of C57BL/6 mice on day 4 after MI (n = 8 per group). Mean ± s.e.m., * P < 0.05, ** P < 0.01.
Figure 4. Serial intravital imaging of progenitor…
Figure 4. Serial intravital imaging of progenitor release from the bone marrow
a, DiD labelled-HSPC Flk2− cells were imaged in the skull bone marrow by intravital microscopy (IVM) before and then again 4 days after MI. DiD labelled-HSPC are white, blood pool red, and bone is blue. The scale bar represents 50 μm. b, Change of HSPC presence between 1st and 2nd IVM session (n = 3 per group). Mean ± s.e.m., * P < 0.05.
Figure 5. Splenic progenitor engraftment after MI
Figure 5. Splenic progenitor engraftment after MI
a, qPCR of SCF in spleen (n = 5–6 per group). b, Number of SCF+ cells in spleen of C57BL/6 mice 4 days after MI by immunofluorescence. c, Enumeration of adoptively transferred GFP+ HSPCs on day 4 after MI (n = 8 per group). d, Proliferation of endogenous HSPCs determined by BrdU incorporation (n = 8 per group). e, Paradigm. Mean ± s.e.m., * P < 0.05, ** P < 0.01.

References

    1. Goldstein JA, et al. Multiple complex coronary plaques in patients with acute myocardial infarction. N Engl J Med. 2000;343:915–922.
    1. Milonas C, et al. Effect of Angiotensin-converting enzyme inhibition on one-year mortality and frequency of repeat acute myocardial infarction in patients with acute myocardial infarction. Am J Cardiol. 2010;105:1229–1234.
    1. Libby P, Ridker PM, Hansson GK. Progress and challenges in translating the biology of atherosclerosis. Nature. 2011;473:317–325.
    1. Weber C, Noels H. Atherosclerosis: current pathogenesis and therapeutic options. Nat Med. 2011;17:1410–1422.
    1. Randolph GJ. The fate of monocytes in atherosclerosis. J Thromb Haemost. 2009;7 (Suppl 1):28–30.
    1. Charo IF, Ransohoff RM. The many roles of chemokines and chemokine receptors in inflammation. N Engl J Med. 2006;354:610–621.
    1. Galkina E, Ley K. Immune and inflammatory mechanisms of atherosclerosis (*) Annu Rev Immunol. 2009;27:165–197.
    1. Ernst E, Hammerschmidt DE, Bagge U, Matrai A, Dormandy JA. Leukocytes and the risk of ischemic diseases. JAMA. 1987;257:2318–2324.
    1. Sabatine MS, et al. Relationship between baseline white blood cell count and degree of coronary artery disease and mortality in patients with acute coronary syndromes: a TACTICS-TIMI 18 (Treat Angina with Aggrastat and determine Cost of Therapy with an Invasive or Conservative Strategy- Thrombolysis in Myocardial Infarction 18 trial)substudy. J Am Coll Cardiol. 2002;40:1761–1768.
    1. Nahrendorf M, et al. The healing myocardium sequentially mobilizes two monocyte subsets with divergent and complementary functions. J Exp Med. 2007;204:3037–3047.
    1. Nahrendorf M, Pittet MJ, Swirski FK. Monocytes: protagonists of infarct inflammation and repair after myocardial infarction. Circulation. 2010;121:2437–2445.
    1. Massa M, et al. Increased circulating hematopoietic and endothelial progenitor cells in the early phase of acute myocardial infarction. Blood. 2005;105:199–206.
    1. Galis ZS, Sukhova GK, Lark MW, Libby P. Increased expression of matrix metalloproteinases and matrix degrading activity in vulnerable regions of human atherosclerotic plaques. J Clin Invest. 1994;94:2493–2503.
    1. Libby P. Inflammation in atherosclerosis. Nature. 2002;420:868–874.
    1. Chen J, et al. In vivo imaging of proteolytic activity in atherosclerosis. Circulation. 2002;105:2766–2771.
    1. Nahrendorf M, et al. Hybrid in vivo FMT-CT imaging of protease activity in atherosclerosis with customized nanosensors. Arterioscler Thromb Vasc Biol. 2009;29:1444–1451.
    1. Tacke F, et al. Monocyte subsets differentially employ CCR2, CCR5, and CX3CR1 to accumulate within atherosclerotic plaques. J Clin Invest. 2007;117:185–194.
    1. Swirski FK, et al. Ly-6Chi monocytes dominate hypercholesterolemia-associated monocytosis and give rise to macrophages in atheromata. J Clin Invest. 2007;117:195–205.
    1. Robbins CS, et al. Extramedullary hematopoiesis generates Ly–6C(high) monocytes that infiltrate atherosclerotic lesions. Circulation. 2012;125:364–374.
    1. Leuschner F, et al. Rapid monocyte kinetics in acute myocardial infarction are sustained by extramedullary monocytopoiesis. J Exp Med. 2012;209:123–137.
    1. Swirski FK, et al. Identification of splenic reservoir monocytes and their deployment to inflammatory sites. Science. 2009;325:612–616.
    1. Psaltis PJ, et al. Identification of a monocyte-predisposed hierarchy of hematopoietic progenitor cells in the adventitia of postnatal murine aorta. Circulation. 2012;125:592–603.
    1. Kondo M, et al. Biology of hematopoietic stem cells and progenitors: implications for clinical application. Annu Rev Immunol. 2003;21:759–806.
    1. Geissmann F, et al. Development of monocytes, macrophages, and dendritic cells. Science. 2010;327:656–661.
    1. Zigmond RE, Ben-Ari Y. Electrical stimulation of preganglionic nerve increases tyrosine hydroxylase activity in sympathetic ganglia. Proc Natl Acad Sci U S A. 1977;74:3078–3080.
    1. Katayama Y, et al. Signals from the sympathetic nervous system regulate hematopoietic stem cell egress from bone marrow. Cell. 2006;124:407–421.
    1. Mendez-Ferrer S, et al. Mesenchymal and haematopoietic stem cells form a unique bone marrow niche. Nature. 2010;466:829–834.
    1. Mendez-Ferrer S, Lucas D, Battista M, Frenette PS. Haematopoietic stem cell release is regulated by circadian oscillations. Nature. 2008;452:442–447.
    1. Cannon CP, et al. Intensive versus moderate lipid lowering with statins after acute coronary syndromes. N Engl J Med. 2004;350:1495–1504.
    1. Hoffmann C, Leitz MR, Oberdorf-Maass S, Lohse MJ, Klotz KN. Comparative pharmacology of human beta-adrenergic receptor subtypes--characterization of stably transfected receptors in CHO cells. Naunyn Schmiedebergs Arch Pharmacol. 2004;369:151–159.
    1. Kruszewska B, Felten SY, Moynihan JAB. Alterations in cytokine and antibody production following chemical sympathectomy in two strains of mice. J Immunol. 1995;155:4613–4620.
    1. Lo Celso C, et al. Live–animal tracking of individual haematopoietic stem/progenitor cells in their niche. Nature. 2009;457:92–96.
    1. Ding L, Saunders TL, Enikolopov G, Morrison SJ. Endothelial and perivascular cells maintain haematopoietic stem cells. Nature. 2012;481:457–462.
    1. Williams DA, Rios M, Stephens C, Patel VP. Fibronectin and VLA-4 in haematopoietic stem cell-microenvironment interactions. Nature. 1991;352:438–441.
    1. Lo Celso C, Scadden DT. The haematopoietic stem cell niche at a glance. J Cell Sci. 2011;124:3529–3535.
    1. Libby P, Theroux P. Pathophysiology of coronary artery disease. Circulation. 2005;111:3481–3488.

Source: PubMed

3
S'abonner