Adipose-derived cells improve left ventricular diastolic function and increase microvascular perfusion in advanced age

Natia Q Kelm, Jason E Beare, Fangping Yuan, Monika George, Charles M Shofner, Bradley B Keller, James B Hoying, Amanda J LeBlanc, Natia Q Kelm, Jason E Beare, Fangping Yuan, Monika George, Charles M Shofner, Bradley B Keller, James B Hoying, Amanda J LeBlanc

Abstract

An early manifestation of coronary artery disease in advanced age is the development of microvascular dysfunction leading to deficits in diastolic function. Our lab has previously shown that epicardial treatment with adipose-derived stromal vascular fraction (SVF) preserves microvascular function following coronary ischemia in a young rodent model. Follow-up studies showed intravenous (i.v.) delivery of SVF allows the cells to migrate to the walls of small vessels and reset vasomotor tone. Therefore we tested the hypothesis that the i.v. cell injection of SVF would reverse the coronary microvascular dysfunction associated with aging in a rodent model. Fischer 344 rats were divided into 4 groups: young control (YC), old control (OC), old + rat aortic endothelial cells (O+EC) and old + GFP+ SVF cells (O+SVF). After four weeks, cardiac function and coronary flow reserve (CFR) were measured via echocardiography, and hearts were explanted either for histology or isolation of coronary arterioles for vessel reactivity studies. In a subgroup of animals, microspheres were injected during resting and dobutamine-stimulated conditions to measure coronary blood flow. GFP+ SVF cells engrafted and persisted in the myocardium and coronary vasculature four weeks following i.v. injection. Echocardiography showed age-related diastolic dysfunction without accompanying systolic dysfunction; diastolic function was improved in old rats after SVF treatment. Ultrasound and microsphere data both showed increased stimulated coronary blood flow in O+SVF rats compared to OC and O+EC, while isolated vessel reactivity was mostly unchanged. I.v.-injected SVF cells were capable of incorporating into the vasculature of the aging heart and are shown in this study to improve CFR and diastolic function in a model of advanced age. Importantly, SVF injection did not lead to arrhythmias or increased mortality in aged rats. SVF cells provide an autologous cell therapy option for treatment of microvascular and cardiac dysfunction in aged populations.

Conflict of interest statement

The authors have declared that no competing interests exist.

Figures

Fig 1. Diastolic function assessment using echocardiography.
Fig 1. Diastolic function assessment using echocardiography.
The graphical representation of E/A ratio shows significant reduction of diastolic function in OC animals, which was rescued by SVF treatment;(A). The graphical representation of IVRT shows a significant increase of IVRT in OC and O+EC animals, which was rescued by SVF treatment; (B) P ≤ 0.05 vs. Old+SVF (*) and vs. Young Control (#); Data are presented as mean ± SD, analyzed with one-way ANOVA followed by Bonferroni post hoc test of following number of animals in each group: YC n = 9, OC n = 12, O+EC n = 10, O+SVF n = 8.
Fig 2. Measurement of coronary flow using…
Fig 2. Measurement of coronary flow using Doppler echocardiography in rats.
Graphical representation of CFR calculation (A), Doppler velocity waveforms from the LAD were traced and peak velocity was measured during rest (scale bar = 850 mm/s) and during stress (Dobutamine infusion, scale bar = 1065 mm/s) (B). CFR was calculated from the peak velocities in our experimental groups (C). LAD peak velocity during rest (D) and Dobutamine-induced stress (E). P ≤ 0.05 vs. Old+SVF (*) and vs. Young Control (#); Data are presented as mean ± SD, analyzed with one-way ANOVA followed by Bonferroni post hoc test of following number of animals in each group: YC n = 10, OC n = 9, O+EC n = 10, O+SVF n = 9.
Fig 3. Measurement of heart perfusion using…
Fig 3. Measurement of heart perfusion using microspheres.
Coronary blood flow was evaluated by the injection of two different stable isotope-labeled microspheres during rest and dobutamine infusion. After explant, the LV was divided into 4 rings above the apex up to papillary region, then rings were separated by septum and free LV wall. Percent blood flow above resting for each section is depicted on the left, P ≤ 0.05 vs. O+SVF (*) and vs. OC (#) (A). Absolute LV blood flow (ml/min-1.g-1, averaged across all sections of one heart) during rest was significantly higher in OC compared to YC, p < .05 (#); following dobutamine challenge, O+SVF showed increased blood flow compared to OC and O+EC, p < .05 (*) (B). Data are presented as mean ± SD, analyzed with one-way ANOVA followed by Bonferroni post hoc test of the following number of animals in each group: YC n = 11, OC n = 11, O+EC n = 4, O+SVF n = 5.
Fig 4. Isolated coronary arteriole vasoreactivity.
Fig 4. Isolated coronary arteriole vasoreactivity.
Endothelin-induced vasoconstriction in coronary arterioles was similar between all groups (A). Coronary arterioles from O+EC or O+SVF decreased sensitivity to the low doses of bradykinin (O+EC group in 1e-13 and 1e-12 concentrations and O+SVF in 1e-12 and 1e-11 concentrations). Main group effect, P < 0.001, P ≤ 0.05 (YC and OC) vs. O+EC (*) vs O+SVF (#) (B). Myogenic response was higher in O+EC group during the 15 and 30mm Hg intramural pressure. Main group effect, P < 0.001, P ≤ 0.05 YC vs. O+EC (*) (C). Data are presented as mean ± SEM, analyzed with repeated measures ANOVA followed by Bonferroni post hoc test of the following number of animals in each group: Endothelin response: YC n = 9, OC n = 24, O+EC n = 9, O+SVF n = 15; Bradykinin response: YC n = 8, OC n = 18, O+EC n = 6, O+SVF n = 14; Myogenic response: YC n = 12, OC n = 23, O+EC n = 8, O+SVF n = 17.
Fig 5. Representative H&E staining of heart…
Fig 5. Representative H&E staining of heart sections and trichrome analysis.
Examples of cross-sectional view of the whole hearts at the papillary level from the experimental groups, scale bar = 1mm (A). All old groups exhibited increased wall thickness compared to YC. SVF treatment significantly decreased wall thickness compared to OC (B). Main group effect P P < 0.05 vs. Young Control (*) and vs. Old Control (#). H&E data are analyzed with one-way ANOVA followed by Bonferroni post hoc test of following number of animals in each group: YC n = 4, OC n = 3, O+EC n = 3, O+SVF n = 7. Masson’s trichrome staining revealed an increase in % collagen content in OC compared to YC (C). Main group effect P = 0.010, P < 0.05 vs. Young Control (*). Trichrome data are analyzed with Kruskall-Wallis one-way ANOVA, YC: n = 3, OC n = 3, O+SVF n = 4. Data are presented as mean ± SD.
Fig 6. GFP positive cells in different…
Fig 6. GFP positive cells in different organs four weeks following GFP+ SVF cell injection.
GFP+ cells were found in aorta, carotid artery, lung, brain and heart, scale bar = 100μm (A). Graphical representation of percentage of GFP+ cells to total number of nucleated cells (DAPI, blue) in different organs (B). Co-localization of GFP (green), ED1 (macrophage marker, red), and NG2 (pericyte marker, orange) in heart, scale bar = 50μm and 20μm in the insert image (C). YC n = 4, OC n = 3, O+EC n = 3, O+SVF n = 7.

References

    1. Reis SE, Holubkov R, Conrad Smith AJ, Kelsey SF, Sharaf BL, Reichek N, et al. Coronary microvascular dysfunction is highly prevalent in women with chest pain in the absence of coronary artery disease: results from the NHLBI WISE study. Am Heart J. 2001;141(5):735–41. Epub 2001/04/26. .
    1. Zhao Z, Wang H, Jessup JA, Lindsey SH, Chappell MC, Groban L. Role of estrogen in diastolic dysfunction. American Journal of Physiology—Heart and Circulatory Physiology. 2014;306(5):H628–H40. 10.1152/ajpheart.00859.2013 PMC3949059.
    1. Kitzman DW. DIASTOLIC DYSFUNCTION IN THE ELDERLY: Genesis and Diagnostic and Therapeutic Implications. Cardiology Clinics. 2000;18(3):597–617. 10.1016/S0733-8651(05)70164-8.
    1. Taqueti VR, Everett BM, Murthy VL, Gaber M, Foster CR, Hainer J, et al. Interaction of impaired coronary flow reserve and cardiomyocyte injury on adverse cardiovascular outcomes in patients without overt coronary artery disease. Circulation. 2015;131(6):528–35. Epub 2014/12/07. 10.1161/CIRCULATIONAHA.114.009716 ; PubMed Central PMCID: PMCPMC4323995.
    1. Taqueti VR, Solomon SD, Shah AM, Desai AS, Groarke JD, Osborne MT, et al. Coronary microvascular dysfunction and future risk of heart failure with preserved ejection fraction. European Heart Journal. 2017:ehx721–ehx. 10.1093/eurheartj/ehx721
    1. Neely CF, DiPierro FV, Kong M, Greelish JP, Gardner TJ. A1 adenosine receptor antagonists block ischemia-reperfusion injury of the heart. Circulation. 1996;94(9 Suppl):Ii376–80. Epub 1996/11/01. .
    1. Briones AM, Aras-Lopez R, Alonso MJ, Salaices M. Small artery remodeling in obesity and insulin resistance. Current vascular pharmacology. 2014;12(3):427–37. Epub 2014/05/23. .
    1. Hill MA, Meininger GA, Davis MJ, Laher I. Therapeutic potential of pharmacologically targeting arteriolar myogenic tone. Trends Pharmacol Sci. 2009;30(7):363–74. Epub 2009/06/23. 10.1016/j.tips.2009.04.008 .
    1. Marinescu MA, Loffler AI, Ouellette M, Smith L, Kramer CM, Bourque JM. Coronary Microvascular Dysfunction, Microvascular Angina, and Treatment Strategies. JACC Cardiovascular imaging. 2015;8(2):210–20. Epub 2015/02/14. 10.1016/j.jcmg.2014.12.008 .
    1. Zimmerlin L, Donnenberg VS, Pfeifer ME, Meyer EM, Peault B, Rubin JP, et al. Stromal vascular progenitors in adult human adipose tissue. Cytometry Part A: the journal of the International Society for Analytical Cytology. 2010;77(1):22–30. Epub 2009/10/24. 10.1002/cyto.a.20813 .
    1. James AW, Zara JN, Zhang X, Askarinam A, Goyal R, Chiang M, et al. Perivascular stem cells: a prospectively purified mesenchymal stem cell population for bone tissue engineering. Stem cells translational medicine. 2012;1(6):510–9. Epub 2012/12/01. 10.5966/sctm.2012-0002 ; PubMed Central PMCID: PMC3659717.
    1. Feisst V, Meidinger S, Locke MB. From bench to bedside: use of human adipose-derived stem cells. Stem Cells and Cloning: Advances and Applications. 2015;8:149–62. 10.2147/SCCAA.S64373 .
    1. Gir P, Oni G, Brown SA, Mojallal A, Rohrich RJ. Human adipose stem cells: current clinical applications. Plast Reconstr Surg. 2012;129(6):1277–90. Epub 2012/05/29. 10.1097/PRS.0b013e31824ecae6 .
    1. Leblanc AJ, Nguyen QT, Touroo JS, Aird AL, Chang RC, Ng CK, et al. Adipose-derived cell construct stabilizes heart function and increases microvascular perfusion in an established infarct. Stem cells translational medicine. 2013;2(11):896–905. Epub 2013/10/10. 10.5966/sctm.2013-0046 ; PubMed Central PMCID: PMC3808204.
    1. Leblanc AJ, Touroo JS, Hoying JB, Williams SK. Adipose stromal vascular fraction cell construct sustains coronary microvascular function after acute myocardial infarction. American journal of physiology Heart and circulatory physiology. 2012;302(4):H973–82. Epub 2011/12/06. 10.1152/ajpheart.00735.2011 .
    1. Taqueti VR, Di Carli MF. Clinical significance of noninvasive coronary flow reserve assessment in patients with ischemic heart disease. Current opinion in cardiology. 2016;31(6):662–9. Epub 2016/10/18. 10.1097/HCO.0000000000000339 .
    1. Gould KL, Johnson NP, Bateman TM, Beanlands RS, Bengel FM, Bober R, et al. Anatomic versus physiologic assessment of coronary artery disease. Role of coronary flow reserve, fractional flow reserve, and positron emission tomography imaging in revascularization decision-making. Journal of the American College of Cardiology. 2013;62(18):1639–53. Epub 2013/08/21. 10.1016/j.jacc.2013.07.076 .
    1. LeBlanc AJ, Reyes R, Kang LS, Dailey RA, Stallone JN, Moningka NC, et al. Estrogen replacement restores flow-induced vasodilation in coronary arterioles of aged and ovariectomized rats. Am J Physiol Regul Integr Comp Physiol. 2009;297(6):R1713–23. Epub 2009/10/09. 10.1152/ajpregu.00178.2009 ; PubMed Central PMCID: PMCPMC2803626.
    1. Kang LS, Chen B, Reyes RA, Leblanc AJ, Teng B, Mustafa SJ, et al. Aging and estrogen alter endothelial reactivity to reactive oxygen species in coronary arterioles. American journal of physiology Heart and circulatory physiology. 2011;300(6):H2105–15. Epub 2011/03/29. 10.1152/ajpheart.00349.2010 ; PubMed Central PMCID: PMC3119103.
    1. Hachamovitch R, Wicker P, Capasso JM, Anversa P. Alterations of coronary blood flow and reserve with aging in Fischer 344 rats. Am J Physiol. 1989;256(1 Pt 2):H66–73. 10.1152/ajpheart.1989.256.1.H66 .
    1. Walker EM, Nillas MS Jr., Mangiarua EI, Cansino S, Morrison RG, Perdue RR, et al. Age-associated changes in hearts of male Fischer 344/Brown Norway F1 rats. Ann Clin Lab Sci. 2006;36(4):427–38. Epub 2006/11/28. 36/4/427 [pii]. .
    1. Morris ME, Beare JE, Reed RM, Dale JR, LeBlanc AJ, Kaufman CL, et al. Systemically delivered adipose stromal vascular fraction cells disseminate to peripheral artery walls and reduce vasomotor tone through a CD11b+ cell-dependent mechanism. Stem cells translational medicine. 2015;4(4):369–80. 10.5966/sctm.2014-0252 25722428; PubMed Central PMCID: PMC4367510.
    1. Aird AL, Nevitt CD, Christian K, Williams SK, Hoying JB, LeBlanc AJ. Adipose–derived stromal vascular fraction cells isolated from old animals exhibit reduced capacity to support the formation of microvascular networks. Experimental gerontology. 2015;63:18–26. 10.1016/j.exger.2015.01.044
    1. Ferreira RG, Brandon MW, Esper SA, Swaminathan M. Quantitative and Semiquantitative Echocardiography: Ventricular and Valvular Physiology A2—Reich, David L. In: Fischer GW, editor. Perioperative Transesophageal Echocardiography Philadelphia: W.B. Saunders; 2014. p. 90–106.
    1. Hassan EA, Torad FA. Two-Dimensional and M-Mode Echocardiographic Measurements in the Healthy Donkey (Equus asinus). Journal of Equine Veterinary Science. 2015;35(4):283–9. 10.1016/j.jevs.2015.01.016.
    1. Lang RM, Bierig M, Devereux RB, Flachskampf FA, Foster E, Pellikka PA, et al. Recommendations for chamber quantification: a report from the American Society of Echocardiography's Guidelines and Standards Committee and the Chamber Quantification Writing Group, developed in conjunction with the European Association of Echocardiography, a branch of the European Society of Cardiology. Journal of the American Society of Echocardiography: official publication of the American Society of Echocardiography. 2005;18(12):1440–63. Epub 2005/12/27. 10.1016/j.echo.2005.10.005 .
    1. Songstad NT, Serrano MC, Sitras V, Johansen D, Ytrehus K, Acharya G. Coronary flow reserve in pregnant rats with increased left ventricular afterload. PloS one. 2014;9(7):e102147 10.1371/journal.pone.0102147 ; PubMed Central PMCID: PMCPMC4089919.
    1. Pantos C, Mourouzis I, Tzeis S, Moraitis P, Malliopoulou V, Cokkinos DD, et al. Dobutamine administration exacerbates postischaemic myocardial dysfunction in isolated rat hearts: an effect reversed by thyroxine pretreatment. Eur J Pharmacol. 2003;460(2–3):155–61. Epub 2003/02/01. S0014299902029278 [pii]. .
    1. Chilian WM, Eastham CL, Marcus ML. Microvascular distribution of coronary vascular resistance in beating left ventricle. Am J Physiol. 1986;251(4 Pt 2):H779–88. 10.1152/ajpheart.1986.251.4.H779 .
    1. Anwar MA, Ford WR, Herbert AA, Broadley KJ. Signal transduction and modulating pathways in tryptamine-evoked vasopressor responses of the rat isolated perfused mesenteric bed. Vascular Pharmacology. 2013;58(1–2):140–9. 10.1016/j.vph.2012.10.007 PMC3884126.
    1. Yuill KH, Yarova P, Kemp-Harper BK, Garland CJ, Dora KA. A novel role for HNO in local and spreading vasodilatation in rat mesenteric resistance arteries. Antioxidants & redox signaling. 2011;14(9):1625–35. Epub 2010/07/10. 10.1089/ars.2010.3279 ; PubMed Central PMCID: PMCPMC3070002.
    1. Yuan FP, Lin DX, Rao CV, Lei ZM. Cryptorchidism in LhrKO animals and the effect of testosterone-replacement therapy. Human reproduction (Oxford, England). 2006;21(4):936–42. Epub 2005/12/20. 10.1093/humrep/dei433 .
    1. Liu H, Ledingham JM, Mullaney I, Laverty R. Endothelial function in mesenteric resistance arteries from the genetically hypertensive rat. Clinical and experimental pharmacology & physiology. 2002;29(5–6):405–11. Epub 2002/05/16. .
    1. Hofmann M, Wollert KC, Meyer GP, Menke A, Arseniev L, Hertenstein B, et al. Monitoring of bone marrow cell homing into the infarcted human myocardium. Circulation. 2005;111(17):2198–202. Epub 2005/04/27. 01.CIR.0000163546.27639.AA [pii] 10.1161/01.CIR.0000163546.27639.AA. 10.1161/01.CIR.0000163546.27639.AA .
    1. Cheng K, Malliaras K, Li TS, Sun B, Houde C, Galang G, et al. Magnetic enhancement of cell retention, engraftment, and functional benefit after intracoronary delivery of cardiac-derived stem cells in a rat model of ischemia/reperfusion. Cell transplantation. 2012;21(6):1121–35. Epub 2012/03/13. 10.3727/096368911X627381 ; PubMed Central PMCID: PMCPMC4323149.
    1. Taqueti VR, Solomon SD, Shah AM, Desai AS, Groarke JD, Osborne MT, et al. Coronary microvascular dysfunction and future risk of heart failure with preserved ejection fraction. Eur Heart J. 2018;39(10):840–9. Epub 2018/01/03. 10.1093/eurheartj/ehx721 .
    1. Dryer K, Gajjar M, Narang N, Lee M, Paul J, Shah AP, et al. Coronary Microvascular Dysfunction in Patients with Heart Failure with Preserved Ejection Fraction. American journal of physiology Heart and circulatory physiology. 2018. Epub 2018/02/10. 10.1152/ajpheart.00680.2017 .
    1. Lee JM, Jung JH, Hwang D, Park J, Fan Y, Na SH, et al. Coronary Flow Reserve and Microcirculatory Resistance in Patients With Intermediate Coronary Stenosis. Journal of the American College of Cardiology. 2016;67(10):1158–69. Epub 2016/03/12. 10.1016/j.jacc.2015.12.053 .
    1. Czernin J, Muller P, Chan S, Brunken RC, Porenta G, Krivokapich J, et al. Influence of age and hemodynamics on myocardial blood flow and flow reserve. Circulation. 1993;88(1):62–9. .
    1. Leblanc AJ, Chen B, Dougherty PJ, Reyes RA, Shipley RD, Korzick DH, et al. Divergent effects of aging and sex on vasoconstriction to endothelin in coronary arterioles. Microcirculation. 2013;20(5):365–76. Epub 2012/12/04. 10.1111/micc.12028 ; PubMed Central PMCID: PMC3594502.
    1. Nevitt C, McKenzie G, Christian K, Austin J, Hencke S, Hoying J, et al. Physiological levels of thrombospondin-1 decrease NO-dependent vasodilation in coronary microvessels from aged rats. American journal of physiology Heart and circulatory physiology. 2016;310(11):H1842–50. 10.1152/ajpheart.00086.2016 .
    1. Rengo G, Perrone-Filardi P, Femminella GD, Liccardo D, Zincarelli C, de Lucia C, et al. Targeting the beta-adrenergic receptor system through G-protein-coupled receptor kinase 2: a new paradigm for therapy and prognostic evaluation in heart failure: from bench to bedside. Circulation Heart failure. 2012;5(3):385–91. Epub 2012/05/17. 10.1161/CIRCHEARTFAILURE.112.966895 .
    1. Larson DF, Yang B, Shi J, Gorman M, Watson RR. Senescent ventricular dysfunction: issues related to cardiopulmonary bypass. J Extra Corpor Technol. 2000;32(3):142–7. Epub 2001/01/09. .
    1. Hirleman ED, Yu Q, Larson DF. Lusitropic effects of dobutamine in young and aged mice in vivo. J Extra Corpor Technol. 2008;40(1):10–5. ; PubMed Central PMCID: PMCPMC4680650.
    1. Bourin P, Bunnell BA, Casteilla L, Dominici M, Katz AJ, March KL, et al. Stromal cells from the adipose tissue-derived stromal vascular fraction and culture expanded adipose tissue-derived stromal/stem cells: a joint statement of the International Federation for Adipose Therapeutics and Science (IFATS) and the International Society for Cellular Therapy (ISCT). Cytotherapy. 2013;15(6):641–8. Epub 2013/04/11. 10.1016/j.jcyt.2013.02.006 ; PubMed Central PMCID: PMCPMC3979435.
    1. Hsiao ST, Lokmic Z, Peshavariya H, Abberton KM, Dusting GJ, Lim SY, et al. Hypoxic conditioning enhances the angiogenic paracrine activity of human adipose-derived stem cells. Stem cells and development. 2013;22(10):1614–23. Epub 2013/01/04. 10.1089/scd.2012.0602 ; PubMed Central PMCID: PMC3653395.
    1. Suga H, Glotzbach JP, Sorkin M, Longaker MT, Gurtner GC. Paracrine Mechanism of Angiogenesis in Adipose-Derived Stem Cell Transplantation. Ann Plast Surg. 2013. Epub 2013/05/03. 10.1097/SAP.0b013e318264fd6a ; PubMed Central PMCID: PMC3740067.
    1. De Barros S, Dehez S, Arnaud E, Barreau C, Cazavet A, Perez G, et al. Aging-related decrease of human ASC angiogenic potential is reversed by hypoxia preconditioning through ROS production. Molecular therapy: the journal of the American Society of Gene Therapy. 2013;21(2):399–408. Epub 2012/10/17. 10.1038/mt.2012.213 ; PubMed Central PMCID: PMC3594015.
    1. Hanley FL, Messina LM, Grattan MT, Hoffman IE. The effect of coronary inflow pressure on coronary vascular resistance in the isolated dog heart. Circ Res. 1984;54(6):760–72. .

Source: PubMed

3
購読する