Systemic delivery of menstrual blood stem cells is more effective in preventing remote organ injuries following myocardial infarction in comparison with bone marrow stem cells in rat

Document Type : Original Article

Authors

1 Nanobiotechnology Research Center, Avicenna Research Institute, ACECR, Tehran, Iran

2 Rajaie Cardiovascular Medical and Research Center, Iran University of Medical Sciences, Tehran, Iran

3 Physiology Research Center, Iran University of Medical Sciences, Tehran, Iran

4 Department of Physiology, Faculty of Medicine, Iran University of Medical Sciences, Tehran, Iran

Abstract

Objective(s): Remote organ injury is a phenomenon that could happen following myocardial infarction (MI). We evaluated the potency of menstrual blood stromal (stem) cells (MenSCs) and bone marrow stem cells (BMSCs) to alleviate remote organ injuries following MI in rats.
Materials and Methods: 2 × 106 MenSCs or BMSCs were administrated seven days after MI induction via the tail vein. Four weeks after cell therapy, activities of aspartate aminotransferase (AST), urea, creatinine, and Blood Urea Nitrogen (BUN) were evaluated. The level of tumor necrosis factor-α (TNF-α), interleukin (IL)-1β, and IL-6 were determined by ELISA assay. The expression of Nuclear Factor-κB (NF-κB) was evaluated by immunohistochemical staining. Apoptosis activity and tissue damage were also determined by TUNEL and H&E staining, respectively.
Results: MenSCs and BMSCs administration caused a significant reduction in AST, urea, and BUN levels compared with the MI group. In addition, systemic injection of MenSCs significantly decreased the IL-1β level compared with BMSCs and MI groups (P<0.05 and P<0.01 respectively). Apoptosis in injured kidneys was noticeably diminished in MenSCs-treated rats compared with BMSCs administrated and MI groups (P<0.05 and P<0.05, respectively). In hepatic tissue, limited numbers of TUNEL-positive cells were detected in all groups. Interestingly, MenSCs therapy evoked inhibition of NF-κB in the kidney strikingly. Although, no significant NF-κB expression was observed in hepatic tissue in any group (P>0.05).
Conclusion: MenSCs are probably more protective than BMSCs on remote organ injuries following MI via decreasing cell death and immunoregulatory properties.

Keywords

Main Subjects


1. Benjamin EJ, Muntner P, Alonso A, Bittencourt MS, Callaway CW, Carson AP, et al. Heart disease and stroke statistics—2019 update: A report from the American Heart Association. Circulation 2019; 139:e56-e528.
2. Takahama H, Kitakaze M. Pathophysiology of cardiorenal syndrome in patients with heart failure: Potential therapeutic targets. Am J Physiol Heart Circ Physiol 2017; 313:H715-H721.
3. Krag A, Gluud LL. Cross-talk between the liver, heart and kidney-another piece in the puzzle. J Gastrointestin Liver Dis 2014; 23:119-121.
4. Kazory A, Ronco C. Hepatorenal syndrome or hepatocardiorenal syndrome: revisiting basic concepts in view of emerging data. Cardiorenal Med 2019; 9:1-7.
5. Harjola VP, Mullens W, Banaszewski M, Bauersachs J, Brunner‐La Rocca HP, Chioncel O, et al. Organ dysfunction, injury and failure in acute heart failure: from pathophysiology to diagnosis and management. A review on behalf of the Acute Heart Failure Committee of the Heart Failure Association (HFA) of the European Society of Cardiology (ESC). Eur J Heart Fail 2017; 19:821-836.
6. Husain-Syed F, McCullough PA, Birk H-W, Renker M, Brocca A, Seeger W, et al. Cardio-pulmonary-renal interactions: a multidisciplinary approach. J Am Coll Cardiol 2015; 65:2433-2448.
7. Taverne YJ, Bogers AJ, Duncker DJ, Merkus D. Reactive oxygen species and the cardiovascular system. Oxid Med Cell Longev 2013; 2013:862423.
8. Moris D, Spartalis M, Spartalis E, Karachaliou G-S, Karaolanis GI, Tsourouflis G, et al. The role of reactive oxygen species in the pathophysiology of cardiovascular diseases and the clinical significance of myocardial redox. Ann Transl Med 2017; 5:326-336.
9. Naschitz JE, Slobodin G, Lewis RJ, Zuckerman E, Yeshurun D. Heart diseases affecting the liver and liver diseases affecting the heart. Am Heart J 2000; 140:111-120.
10. Jahng JWS, Song E, Sweeney G. Crosstalk between the heart and peripheral organs in heart failure. Exp Mol Med 2016; 48:e217-e217.
11. Pittenger MF, Discher DE, Péault BM, Phinney DG, Hare JM, Caplan AI. Mesenchymal stem cell perspective: cell biology to clinical progress. NPJ Regen Med 2019; 4:1-15.
12. Miao C, Lei M, Hu W, Han S, Wang Q. A brief review: The therapeutic potential of bone marrow mesenchymal stem cells in myocardial infarction. Stem Cell Res Ther 2017; 8:1-6.
13. Ullah I, Subbarao RB, Rho GJ. Human mesenchymal stem cells-current trends and future prospective. Biosci Rep 2015; 35: e00191.
14. Chen L, Qu J, Xiang C. The multi-functional roles of menstrual blood-derived stem cells in regenerative medicine. Stem Cell Res Ther 2019; 10:1-10.
15. Chen L, Zhang C, Chen L, Wang X, Xiang B, Wu X, et al. Human menstrual blood‐derived stem cells ameliorate liver fibrosis in mice by targeting hepatic stellate cells via paracrine mediators. Stem Cells Transl Med  2017; 6:272-284.
16. Azedi F, Kazemnejad S, Zarnani AH, Soleimani M, Shojaei A, Arasteh S. Comparative capability of menstrual blood versus bone marrow derived stem cells in neural differentiation. Mol Biol Rep 2017; 44:169-182.
17. Lin J, Xiang D, Zhang J-l, Allickson J, Xiang C. Plasticity of human menstrual blood stem cells derived from the endometrium. J Zhejiang Univ Sci B 2011; 12:372-380.
18. Darzi S, Zarnani AH, Jeddi-Tehrani M, Entezami K, Mirzadegan E, Akhondi MM, et al. Osteogenic differentiation of stem cells derived from menstrual blood versus bone marrow in the presence of human platelet releasate. Tissue Eng Part A 2012; 18:1720-1728.
19. Chen L, Qu J, Cheng T, Chen X, Xiang C. Menstrual blood-derived stem cells: Toward therapeutic mechanisms, novel strategies, and future perspectives in the treatment of diseases. Stem Cell Res Ther 2019; 10:1-12.
20. Nikoo S, Ebtekar M, Jeddi-Tehrani M, Bozorgmehr M, Zarnani A-H. Culture density of menstrual blood-derived stromal/stem cells determines the quality of T cell responses: An experimental study. Int J Reprod Biomed 2021; 19:75-86.
21. Liu Y, Niu R, Yang F, Yan Y, Liang S, Sun Y, et al. Biological characteristics of human menstrual blood‐derived endometrial stem cells. J Cell Mol Med 2018; 22:1627-1639.
22. Shokri M-R, Bozorgmehr M, Ghanavatinejad A, Falak R, Aleahmad M, Kazemnejad S, et al. Human menstrual blood-derived stromal/stem cells modulate functional features of natural killer cells. Scientific Reports 2019; 9:1-13.
23. Liu Y, Niu R, Li W, Lin J, Stamm C, Steinhoff G, et al. Therapeutic potential of menstrual blood-derived endometrial stem cells in cardiac diseases. Cell Mol Life Sci 2019; 76:1681-1695.
24. Alcayaga-Miranda F, Cuenca J, Luz-Crawford P, Aguila-Díaz C, Fernandez A, Figueroa FE, et al. Characterization of menstrual stem cells: angiogenic effect, migration and hematopoietic stem cell support in comparison with bone marrow mesenchymal stem cells. Stem Cell Res Ther 2015; 6:1-14.
25. Bozorgmehr M, Gurung S, Darzi S, Nikoo S, Kazemnejad S, Zarnani A-H, et al. Endometrial and menstrual blood mesenchymal stem/stromal cells: Biological properties and clinical application. Front Cell Dev Biol 2020; 8:497-523.
26. Manshori M, Kazemnejad S, Naderi N, Darzi M, Aboutaleb N, Golshahi H. Greater angiogenic and immunoregulatory potency of bFGF and 5-aza-2’-deoxycytidine pre-treated menstrual blood stem cells in compare to bone marrow stem cells in rat model of myocardial infarction. BBMC Cardiovasc Disord  2022; 22:578-589.
27. van Dokkum RP, Eijkelkamp WB, Kluppel AC, Henning RH, van Goor H, Citgez M, et al. Myocardial infarction enhances progressive renal damage in an experimental model for cardio-renal interaction. J Am Soc Nephrol 2004; 15:3103-3110.
28. Anzai A, Anzai T, Naito K, Kaneko H, Mano Y, Jo Y, et al. Prognostic significance of acute kidney injury after reperfused ST-elevation myocardial infarction: Synergistic acceleration of renal dysfunction and left ventricular remodeling. J Card Fail 2010; 16:381-389.
29. Ma T, Sun J, Zhao Z, Lei W, Chen Y, Wang X, et al. A brief review: Adipose-derived stem cells and their therapeutic potential in cardiovascular diseases. Stem Cell Res Ther 2017; 8:124-131.
30. Møller S, Bernardi M. Interactions of the heart and the liver. Eur Heart J 2013; 34:2804-2811.
31.Xanthopoulos A, Starling RC, Kitai T, Triposkiadis F. Heart failure and liver disease: cardiohepatic interactions. JACC Heart Fail 2019; 7:87-97.
32. Cho E, Kim M, Ko YS, Lee HY, Song M, Kim MG, et al. Role of inflammation in the pathogenesis of cardiorenal syndrome in a rat myocardial infarction model. Nephrol Dial Transplant 2013; 28:2766-2778.
33. Lekawanvijit S, Kompa AR, Zhang Y, Wang BH, Kelly DJ, Krum H. Myocardial infarction impairs renal function, induces renal interstitial fibrosis, and increases renal KIM-1 expression: implications for cardiorenal syndrome. Am J Physiol Heart Circ Physiol 2012; 302:H1884-H1893.
34. Lu J, Wang X, Wang W, Muniyappa H, Deshmukh A, Hu C, et al. Abrogation of lectin-like oxidized LDL receptor-1 attenuates acute myocardial ischemia-induced renal dysfunction by modulating systemic and local inflammation. Kidney Int 2012; 82:436-444.
35. Ruparelia N, Digby JE, Jefferson A, Medway DJ, Neubauer S, Lygate CA, et al. Myocardial infarction causes inflammation and leukocyte recruitment at remote sites in the myocardium and in the renal glomerulus. Inflamm Res 2013; 62:515-525.
36. Virzì GM, Torregrossa R, Cruz DN, Chionh CY, De Cal M, Soni SS, et al. Cardiorenal syndrome type 1 may be immunologically mediated: A pilot evaluation of monocyte apoptosis. Cardiorenal Med 2012; 2:33-42.
37. Aleahmad M, Bozorgmehr M, Nikoo S, Ghanavatinejad A, Shokri M-R, Montazeri S, et al. Endometrial mesenchymal stem/stromal cells: The enigma to code messages for generation of functionally active regulatory T cells. Stem Cell Res Ther 2021; 12: 536-550.
38. Liu S, Liu F, Zhou Y, Jin B, Sun Q, Guo S. Immunosuppressive property of MSCs mediated by cell surface receptors. Front Immunol 2020; 11:1076-1090.
39.Lemos DR, McMurdo M, Karaca G, Wilflingseder J, Leaf IA, Gupta N, et al. Interleukin-1β activates a MYC-dependent metabolic switch in kidney stromal cells necessary for progressive tubulointerstitial fibrosis. J Am Soc Nephrol 2018; 29:1690-1705.
40. Lei Y, Devarapu SK, Motrapu M, Cohen CD, Lindenmeyer MT, Moll S, et al. Interleukin-1β inhibition for chronic kidney disease in obese mice with type 2 diabetes. Front Immunol 2019; 10:1223-1230.
41. Peron J, Jazedje T, Brandao W, Perin P, Maluf M, Evangelista L, et al. Human endometrial-derived mesenchymal stem cells suppress inflammation in the central nervous system of EAE mice. Stem Cell Rev Rep 2012; 8:940-952.
42. Martínez-Aguilar R, Romero-Pinedo S, Ruiz-Magaña MJ, Olivares EG, Ruiz-Ruiz C, Abadía-Molina AC. Menstrual blood-derived stromal cells modulate functional properties of mouse and human macrophages. Scientific reports 2020; 10:1-14.
43. Afsar B, Ortiz A, Covic A, Solak Y, Goldsmith D, Kanbay M. Focus on renal congestion in heart failure. Clin Kidney J 2016; 9:39-47.
44. Irazabal MV, Torres VE. Reactive oxygen species and redox signaling in chronic kidney disease. Cells 2020; 9:1-17.
45. Song N, Thaiss F, Guo L. NFκB and kidney injury. Front Immunol 2019; 10:1-12.
46. D’Ignazio L, Rocha S. Hypoxia induced NF-κB. Cells 2016; 5:1-8.
47. Kumar D, Singla SK, Puri V, Puri S. The restrained expression of NF-kB in renal tissue ameliorates folic acid induced acute kidney injury in mice. PLoS One 2015; 10:e115947.
48. Sun L, Zhu M, Feng W, Lin Y, Yin J, Jin J, et al. Exosomal miRNA Let-7 from menstrual blood-derived endometrial stem cells alleviates pulmonary fibrosis through regulating mitochondrial DNA damage. Oxid Med Cell Longev 2019; 2019: 4506303.
49. Vescovo G, Castellani C, Fedrigo M, Virzì GM, Vescovo GM, Tavano R, et al. Stem cells transplantation positively modulates the heart-kidney cross talk in cardiorenal syndrome type II. Int J Cardiol 2019; 275:136-144.
50. Cun Y, Diao B, Zhang Z, Wang G, Yu J, Ma L, et al. Role of the stromal cell derived factor‑1 in the biological functions of endothelial progenitor cells and its underlying mechanisms. Exp Ther Med 2021; 21:1-9.
51. Mayorga ME, Kiedrowski M, McCallinhart P, Forudi F, Ockunzzi J, Weber K, et al. Role of SDF‐1: CXCR4 in impaired post‐myocardial infarction cardiac repair in diabetes. Stem Cells Transl Med 2018; 7:115-124.