Chonnam Med J.  2018 Jan;54(1):10-16. 10.4068/cmj.2018.54.1.10.

Functional Relevance of Macrophage-mediated Inflammation to Cardiac Regeneration

Affiliations
  • 1Biomedical Research Institute, Chonnam National University Hospital, Gwangju, Korea.
  • 2Cell Regeneration Research Center, Chonnam National University Hospital, Gwangju, Korea. cecilyk@hanmail.net
  • 3Department of Cardiology, Chonnam National University Hospital, Gwangju, Korea.

Abstract

Cardiovascular disease remains the leading cause of death worldwide and regenerative medicine is a promising therapeutic option for this disease. We have developed various techniques to attenuate the cardiac remodeling and to regenerate cardiovascular systems via stem cell application. Besides cell therapy, we are interested in the modulation of pathological inflammation mediated by macrophages in the damaged heart tissue to arouse endogenous reparative responses with biocompatible small molecules. Certainly, current understanding of mechanisms of tissue regeneration will lead to the development of innovative regenerative medicine for cardiovascular disease.

Keyword

Cardiovascular Diseases; Regeneration; Stem Cells; Macrophages; Inflammation

MeSH Terms

Cardiovascular Diseases
Cardiovascular System
Cause of Death
Cell- and Tissue-Based Therapy
Heart
Inflammation*
Macrophages
Regeneration*
Regenerative Medicine
Stem Cells

Figure

  • FIG. 1 Prevalence of myocardial infarction and the incidence rate of heart failure in Korea. AMI: acute myocardial infarction, IHD: ischemic heart disease, HF: heart failure, yr: years, mo: months.

  • FIG. 2 The sources of stem cells.

  • FIG. 3 (A) Representative images of immunocytofluorescence staining showed the cardiogenic MSCs expressed cardiac-specific proteins GATA4 (green) and cTnI (red). Nuclei were stained with DAPI (blue). (B) Treatment of MSCs with differentiation inducer upregulated cardiac marker expressions such as MEF2C, cTnI, and GATA4. MSCs: mesenchymal stem cells, MEF2C: myocyte enhancer factor 2c, cTnI: cardiac troponin I.

  • FIG. 4 Mapping images of left ventricle in a porcine myocardial infarction model. red: infarct zone, yellow: peri-infarct zone, green: viable myocardium. Stem cells are injected into the myocardium endocardially by using MYOSTAR injection catheter.

  • FIG. 5 Phenotype changes of infiltrated macrophages in the infarcted myocardium over time. M1 type represents pro-inflammatory macrophages, while M2 represents anti-inflammatory subtype. MI: myocardial infarction, iNOS: inducible nitric oxide synthase, IL-1β: interleukin-1β, TNF-α: tumor necrosis factor-α, Arg1: arginase 1.

  • FIG. 6 Stem cells and small molecules such as 5-azacytidine and BIO modulate multiple aspects of the cardiac microenvironment to improve cardiac remodeling after injury. Cardiomyocytes are protected from cell death to provide a therapeutic opportunity to replace a portion of the dead cardiomyocytes, whereas proliferation of cardiac myofibroblasts was inhibited. Macrophage phenotype was shifted toward anti-inflammatory in the cardiac microenvironment.


Reference

1. Blau HM, Brazelton TR, Weimann JM. The evolving concept of a stem cell: entity or function? Cell. 2001; 105:829–841.
2. Lim SY, Kim YS, Ahn Y, Jeong MH, Hong MH, Joo SY, et al. The effects of mesenchymal stem cells transduced with Akt in a porcine myocardial infarction model. Cardiovasc Res. 2006; 70:530–542.
Article
3. Kim YS, Kwon JS, Hong MH, Kim J, Song CH, Jeong MH, et al. Promigratory activity of oxytocin on umbilical cord blood-derived mesenchymal stem cells. Artif Organs. 2010; 34:453–461.
Article
4. Kim YS, Ahn Y, Kwon JS, Cho YK, Jeong MH, Cho JG, et al. Priming of mesenchymal stem cells with oxytocin enhances the cardiac repair in ischemia/reperfusion injury. Cells Tissues Organs. 2012; 195:428–442.
Article
5. Cho DI, Kang WS, Hong MH, Kang HJ, Kim MR, Kim MC, et al. The optimization of cell therapy by combinational application with apicidin-treated mesenchymal stem cells after myocardial infarction. Oncotarget. 2017; 8:44281–44294.
Article
6. Kim MC, Kim YS, Kang WS, Lee KH, Cho M, Hong MH, et al. Intramyocardial injection of stem Cells in pig myocardial infarction model: the first trial in Korea. J Korean Med Sci. 2017; 32:1708–1712.
Article
7. Lefer DJ, Marbán E. Is cardioprotection dead? Circulation. 2017; 136:98–109.
Article
8. Nahrendorf M, Swirski FK, Aikawa E, Stangenberg L, Wurdinger T, Figueiredo JL, et al. The healing myocardium sequentially mobilizes two monocyte subsets with divergent and complementary functions. J Exp Med. 2007; 204:3037–3047.
Article
9. Takahashi K. Development and differentiation of macrophages and their related cells. Hum Cell. 1994; 7:109–115.
10. Yona S, Kim KW, Wolf Y, Mildner A, Varol D, Breker M, et al. Fate mapping reveals origins and dynamics of monocytes and tissue macrophages under homeostasis. Immunity. 2013; 38:79–91.
Article
11. Epelman S, Lavine KJ, Beaudin AE, Sojka DK, Carrero JA, Calderon B, et al. Embryonic and adult-derived resident cardiac macrophages are maintained through distinct mechanisms at steady state and during inflammation. Immunity. 2014; 40:91–104.
Article
12. Bain CC, Bravo-Blas A, Scott CL, Perdiguero EG, Geissmann F, Henri S, et al. Constant replenishment from circulating monocytes maintains the macrophage pool in the intestine of adult mice. Nat Immunol. 2014; 15:929–937.
Article
13. Perdiguero EG, Geissmann F. The development and maintenance of resident macrophages. Nat Immunol. 2016; 17:2–8.
Article
14. Dutta P, Sager HB, Stengel KR, Naxerova K, Courties G, Saez B, et al. Myocardial infarction activates CCR2(+) hematopoietic stem and progenitor cells. Cell Stem Cell. 2015; 16:477–487.
Article
15. Gordon S. Alternative activation of macrophages. Nat Rev Immunol. 2003; 3:23–35.
Article
16. Frantz S, Hofmann U, Fraccarollo D, Schäfer A, Kranepuhl S, Hagedorn I, et al. Monocytes/macrophages prevent healing defects and left ventricular thrombus formation after myocardial infarction. FASEB J. 2013; 27:871–881.
Article
17. Galis ZS, Sukhova GK, Kranzhöfer R, Clark S, Libby P. Macrophage foam cells from experimental atheroma constitutively produce matrix-degrading proteinases. Proc Natl Acad Sci U S A. 1995; 92:402–406.
Article
18. Clinton SK, Underwood R, Hayes L, Sherman ML, Kufe DW, Libby P. Macrophage colony-stimulating factor gene expression in vascular cells and in experimental and human atherosclerosis. Am J Pathol. 1992; 140:301–316.
19. Randolph GJ. Emigration of monocyte-derived cells to lymph nodes during resolution of inflammation and its failure in atherosclerosis. Curr Opin Lipidol. 2008; 19:462–468.
Article
20. Melián A, Geng YJ, Sukhova GK, Libby P, Porcelli SA. CD1 expression in human atherosclerosis. A potential mechanism for T cell activation by foam cells. Am J Pathol. 1999; 155:775–786.
21. Gordon S, Taylor PR. Monocyte and macrophage heterogeneity. Nat Rev Immunol. 2005; 5:953–964.
Article
22. Gordon S, Plüddemann A, Martinez Estrada F. Macrophage heterogeneity in tissues: phenotypic diversity and functions. Immunol Rev. 2014; 262:36–55.
Article
23. Gordon S, Plüddemann A. Tissue macrophages: heterogeneity and functions. BMC Biol. 2017; 15:53.
Article
24. Tsujioka H, Imanishi T, Ikejima H, Kuroi A, Takarada S, Tanimoto T, et al. Impact of heterogeneity of human peripheral blood monocyte subsets on myocardial salvage in patients with primary acute myocardial infarction. J Am Coll Cardiol. 2009; 54:130–138.
Article
25. Ben-Mordechai T, Holbova R, Landa-Rouben N, Harel-Adar T, Feinberg MS, Abd Elrahman I, et al. Macrophage subpopulations are essential for infarct repair with and without stem cell therapy. J Am Coll Cardiol. 2013; 62:1890–1901.
Article
26. Cho DI, Kim MR, Jeong HY, Jeong HC, Jeong MH, Yoon SH, et al. Mesenchymal stem cells reciprocally regulate the M1/M2 balance in mouse bone marrow-derived macrophages. Exp Mol Med. 2014; 46:e70.
Article
27. Degousee N, Simpson J, Fazel S, Scholich K, Angoulvant D, Angioni C, et al. Lack of microsomal prostaglandin E(2) synthase-1 in bone marrow-derived myeloid cells impairs left ventricular function and increases mortality after acute myocardial infarction. Circulation. 2012; 125:2904–2913.
Article
28. Hilgendorf I, Gerhardt LM, Tan TC, Winter C, Holderried TA, Chousterman BG, et al. Ly-6Chigh monocytes depend on Nr4a1 to balance both inflammatory and reparative phases in the infarcted myocardium. Circ Res. 2014; 114:1611–1622.
Article
29. Cella M, Sallusto F, Lanzavecchia A. Origin, maturation and antigen presenting function of dendritic cells. Curr Opin Immunol. 1997; 9:10–16.
Article
30. Swirski FK, Libby P, Aikawa E, Alcaide P, Luscinskas FW, Weissleder R, et al. Ly-6Chi monocytes dominate hypercholesterolemia-associated monocytosis and give rise to macrophages in atheromata. J Clin Invest. 2007; 117:195–205.
Article
31. Chistiakov DA, Sobenin IA, Orekhov AN, Bobryshev YV. Myeloid dendritic cells: development, functions, and role in atherosclerotic inflammation. Immunobiology. 2015; 220:833–844.
Article
32. Choo EH, Lee JH, Park EH, Park HE, Jung NC, Kim TH, et al. Infarcted Myocardium-primed dendritic cells improve remodeling and cardiac function after myocardial infarction by modulating the regulatory T cell and macrophage polarization. Circulation. 2017; 135:1444–1457.
Article
33. Soehnlein O, Weber C, Lindbom L. Neutrophil granule proteins tune monocytic cell function. Trends Immunol. 2009; 30:538–546.
Article
34. Carbone F, Nencioni A, Mach F, Vuilleumier N, Montecucco F. Pathophysiological role of neutrophils in acute myocardial infarction. Thromb Haemost. 2013; 110:501–514.
Article
35. Jung K, Kim P, Leuschner F, Gorbatov R, Kim JK, Ueno T, et al. Endoscopic time-lapse imaging of immune cells in infarcted mouse hearts. Circ Res. 2013; 112:891–899.
Article
36. Horckmans M, Ring L, Duchene J, Santovito D, Schloss MJ, Drechsler M, et al. Neutrophils orchestrate post-myocardial infarction healing by polarizing macrophages towards a reparative phenotype. Eur Heart J. 2017; 38:187–197.
Article
37. Jeong HY, Kang WS, Hong MH, Jeong HC, Shin MG, Jeong MH, et al. 5-Azacytidine modulates interferon regulatory factor 1 in macrophages to exert a cardioprotective effect. Sci Rep. 2015; 5:15768.
Article
38. Kim YS, Kang WS, Kwon JS, Hong MH, Jeong HY, Jeong HC, et al. Protective role of 5-azacytidine on myocardial infarction is associated with modulation of macrophage phenotype and inhibition of fibrosis. J Cell Mol Med. 2014; 18:1018–1027.
Article
39. Kim YS, Jeong HY, Kim AR, Kim WH, Cho H, Um J, et al. Natural product derivative BIO promotes recovery after myocardial infarction via unique modulation of the cardiac microenvironment. Sci Rep. 2016; 6:30726.
Article
40. Sager HB, Hulsmans M, Lavine KJ, Moreira MB, Heidt T, Courties G, et al. Proliferation and recruitment contribute to myocardial macrophage expansion in chronic heart failure. Circ Res. 2016; 119:853–864.
Article
41. Menasché P, Alfieri O, Janssens S, McKenna W, Reichenspurner H, Trinquart L, et al. The Myoblast Autologous Grafting in Ischemic Cardiomyopathy (MAGIC) trial: first randomized placebo-controlled study of myoblast transplantation. Circulation. 2008; 117:1189–1200.
Article
42. Hare JM, Fishman JE, Gerstenblith G, DiFede Velazquez DL, Zambrano JP, Suncion VY, et al. Comparison of allogeneic vs autologous bone marrow–derived mesenchymal stem cells delivered by transendocardial injection in patients with ischemic cardiomyopathy: the POSEIDON randomized trial. JAMA. 2012; 308:2369–2379.
Article
43. Bolli R, Chugh AR, D'Amario D, Loughran JH, Stoddard MF, Ikram S, et al. Cardiac stem cells in patients with ischaemic cardiomyopathy (SCIPIO): initial results of a randomised phase 1 trial. Lancet. 2011; 378:1847–1857.
Article
44. Bartunek J, Behfar A, Dolatabadi D, Vanderheyden M, Ostojic M, Dens J, et al. Cardiopoietic stem cell therapy in heart failure: the C-CURE (Cardiopoietic stem Cell therapy in heart failURE) multicenter randomized trial with lineage-specified biologics. J Am Coll Cardiol. 2013; 61:2329–2338.
45. Makkar RR, Smith RR, Cheng K, Malliaras K, Thomson LE, Berman D, et al. Intracoronary cardiosphere-derived cells for heart regeneration after myocardial infarction (CADUCEUS): a prospective, randomised phase 1 trial. Lancet. 2012; 379:895–904.
Article
46. Heldman AW, DiFede DL, Fishman JE, Zambrano JP, Trachtenberg BH, Karantalis V, et al. Transendocardial mesenchymal stem cells and mononuclear bone marrow cells for ischemic cardiomyopathy: the TAC-HFT randomized trial. JAMA. 2014; 311:62–73.
Article
47. Karantalis V, DiFede DL, Gerstenblith G, Pham S, Symes J, Zambrano JP, et al. Autologous mesenchymal stem cells produce concordant improvements in regional function, tissue perfusion, and fibrotic burden when administered to patients undergoing coronary artery bypass grafting: The Prospective Randomized Study of Mesenchymal Stem Cell Therapy in Patients Undergoing Cardiac Surgery (PROMETHEUS) trial. Circ Res. 2014; 114:1302–1310.
Article
48. Fischer-Rasokat U, Assmus B, Seeger FH, Honold J, Leistner D, Fichtlscherer S, et al. A pilot trial to assess potential effects of selective intracoronary bone marrow-derived progenitor cell infusion in patients with nonischemic dilated cardiomyopathy: final 1-year results of the transplantation of progenitor cells and functional regeneration enhancement pilot trial in patients with nonischemic dilated cardiomyopathy. Circ Heart Fail. 2009; 2:417–423.
Article
49. Strauer BE, Yousef M, Schannwell CM. The acute and long-term effects of intracoronary Stem cell Transplantation in 191 patients with chronic heARt failure: the STAR-heart study. Eur J Heart Fail. 2010; 12:721–729.
Article
Full Text Links
  • CMJ
Actions
Cited
CITED
export Copy
Close
Share
  • Twitter
  • Facebook
Similar articles
Copyright © 2024 by Korean Association of Medical Journal Editors. All rights reserved.     E-mail: koreamed@kamje.or.kr