Circulating Pro-Vascular Progenitor Cell Depletion During Type 2 Diabetes: Translational Insights Into the Prevention of Ischemic Complications in Diabetes

Daniella C Terenzi, Mohammed Al-Omran, Adrian Quan, Hwee Teoh, Subodh Verma, David A Hess, Daniella C Terenzi, Mohammed Al-Omran, Adrian Quan, Hwee Teoh, Subodh Verma, David A Hess

Abstract

Detection of vascular regenerative cell exhaustion is required to combat ischemic complications during type 2 diabetes mellitus (T2D). We used high aldehyde dehydrogenase (ALDH) activity and surface marker co-expression to develop a high-throughput flow cytometry-based assay to quantify circulating proangiogenic and proinflammatory cell content in the peripheral blood of individuals with T2D. Circulating proangiogenic monocytes expressing anti-inflammatory M2 markers were decreased in patients with T2D. Individuals with longer duration of T2D exhibited reduced frequencies of circulating proangiogenic ALDHhiCD34+ progenitor cells with primitive (CD133) and migratory (CXCR4) phenotypes. This approach consistently detected increased inflammatory cell burden and decreased provascular progenitor content in individuals with T2D.

Keywords: ALDH, aldehyde dehydrogenase; BM, bone marrow; HbA1c, glycosylated hemoglobin; ROS, reactive oxygen species; SSC, side scatter; T2D, type 2 diabetes mellitus; Wnt, wingless related integration site; aldehyde dehydrogenase; angiogenesis; ischemia; progenitor cells; type 2 diabetes.

Figures

Graphical abstract
Graphical abstract
Figure 1
Figure 1
Circulating Progenitor Cell Subpopulations Are Discerned According to High ALDH-Activity and SSC Properties (A and B) Representative flow cytometry plots using N,N-diethylaminobenzaldehyde (DEAB) to inhibit aldehyde dehydrogenase (ALDH) activity establishing gates for low versus high ALDH-activity. Without inhibition, cells with high ALDH-activity exhibit increased fluorescence intensity (right shift) and detect primitive cells with a self-protective progenitor cell phenotype. High ALDH-activity combined with side scatter (SSC) properties selects for a progenitor cell subpopulation with low intracellular complexity (R1 = ALDHhiSSClow cells), a monocyte subpopulation with intermediate intracellular complexity (R2 = ALDHhiSSCmid cells), and a granulocytic subpopulation with high intracellular complexity (R3 = ALDHhiSSChi cells). (C and D) Representative flow cytometry plots showing the frequencies of each population in control subjects and patients with type 2 diabetes mellitus (T2D). Compared with control subjects, patients with T2D exhibited an increased frequency of ALDHhi cells within the granulocyte subpopulation (R3) and an equal frequency of circulating ALDHhi cells with low (R1) and intermediate (R2) complexity. Values are mean ± SEM. ***p < 0.001 with the Student's t-test.
Figure 2
Figure 2
Circulating ALDHhiSSClow Cells With Primitive, Myeloid, and Migratory Phenotypes Are Decreased in Patients with T2D (A–C) The frequency of circulating ALDHhiSSClow progenitor cells with primitive cell phenotype (CD34+CD133+) was reduced in patients with T2D compared with control subjects. (D–F) The frequency of circulating ALDHhiSSClow progenitor cells with early myeloid cell phenotype (CD34+CD33+) was reduced in patients with T2D compared with control subjects. (G–I) The frequency of circulating ALDHhiSSClow progenitor cells with migratory phenotype (CD34+CXCR4+) was reduced in patients with T2D compared with control subjects. Values are mean ± SEM. ***p < 0.01 with the Student's t-test. Abbreviations as in Figure 1.
Figure 3
Figure 3
Circulating ALDHhiSSClow Cells With Endothelial Cell and Pericyte-associated Adhesive Phenotypes Are Decreased in Patients With T2D (A–C) The frequency of circulating ALDHhiSSClow progenitor cells co-expressing CD34 with hematopoietic/endothelial cell marker platelet endothelial cell adhesion molecule (PECAM) (CD31) was equivalent in patients with T2D compared with control subjects. (D–F) The frequency of circulating ALDHhiSSClow cells in progenitor cells co-expressing CD34 with the endothelial cell–associated marker vascular endothelial (VE)-cadherin (CD144) was decreased in patients with T2D compared with control subjects. (G–I) The frequency of circulating ALDHhiSSClow progenitor cells co-expressing CD34 with the endothelial/pericytes marker melanoma cell adhesion molecule (MCAM) (CD146) was decreased in patients with T2D compared with control subjects. Values are mean ± SEM. ∗p < 0.001 with the Student's t-test. Other abbreviations as in Figure 1.
Figure 4
Figure 4
Circulating ALDHhiSSCmid Cells With M2 Phenotype Are Decreased in Patients With T2D (A to C) The frequency of circulating ALDHhiSSCmid cells co-expressing CD34 with the macrophage scavenger receptor (CD68) was decreased in patients with T2D compared with control subjects. (D to F) The frequency of circulating ALDHhiSSCmid cells co-expressing CD68 with the M1 macrophage–associated marker CD80 was equal in patients with T2D compared with control subjects. (G to I) The frequency of circulating ALDHhiSSCmid progenitor cells co-expressing CD68 with the M2 macrophage–associated marker CD163 was decreased in patients with T2D compared with control subjects. Values are mean ± SEM. *p < 0.05 with the Student's t-test. Other abbreviations as in Figure 1.
Figure 5
Figure 5
Circulating ALDHhiSSChi Inflammatory Cells Are Decreased in Patients Taking Insulin (A and B) The frequency of cells with high ALDH-activity and high SSC properties was decreased in patients with T2D taking insulin. (C and D) The frequency of primitive and migratory progenitor cells was equivalent in patients with T2D despite insulin therapy. Values are mean ± SEM. *p < 0.05 with the Student's t-test. Abbreviations as in Figure 1.
Figure 6
Figure 6
Circulating ALDHhiSSClow Cells With Primitive and Migratory Progenitor Cell Phenotypes Decreased With Longer Duration of Diabetes (A and B) In patients with T2D, the frequency of cells with high ALDH-activity was equivalent in patients ≤70 years of age compared with patients >70 years of age with T2D. (C and D) The frequency of circulating primitive progenitor cells (CD34+/CD133+) and migratory progenitor cells (CD34+/CXCR4+) was equivalent in patients ≤70 years of age compared with patients >70 years of age. (E and F) In patients with T2D, the frequency of cells with high ALDH-activity was equivalent in patients with diabetes duration ≤13 years compared with patients with diabetes duration >13 years. (G and H) However, the frequency of circulating primitive progenitor cells (CD34+/CD133+) and migratory progenitor cells was decreased in patients with diabetes duration ≤13 years compared with patients with diabetes duration >13 years. Values are mean ± SEM. *p < 0.05 with the Student's t-test. Abbreviations as in Figure 1.

References

    1. American Diabetes Association 2. Classification and diagnosis of diabetes: standards of medical care in diabetes-2018. Diabetes Care. 2018;41:S13–S27.
    1. Diabetes Canada Clinical Practice Guidelines Expert Committee. Punthakee Z., Goldenberg R., Katz P. Definition, classification and diagnosis of diabetes, prediabetes and metabolic syndrome. Can J Diabetes. 2018;42 Suppl 1:S10–S15.
    1. International Diabetes Federation . International Diabetes Federation; Brussels, Belgium: 2017. IDF Diabetes Atlas—Eight Edition.
    1. Qadura M., Terenzi D.C., Verma S., Al-Omran M., Hess D.A. Concise review: cell therapy for critical limb ischemia: an integrated review of preclinical and clinical studies. Stem Cells. 2018;36:161–171.
    1. Zinman B., Wanner C., Lachin J.M. Empagliflozin, cardiovascular outcomes, and mortality in type 2 diabetes. N Engl J Med. 2015;373:2117–2128.
    1. Verma S., Mazer C.D., Fitchett D. Empagliflozin reduces cardiovascular events, mortality and renal events in participants with type 2 diabetes after coronary artery bypass graft surgery: subanalysis of the EMPA-REG OUTCOME(R) randomised trial. Diabetologia. 2018;61:1712–1723.
    1. Verma S., Mazer C.D., Al-Omran M. Cardiovascular outcomes and safety of empagliflozin in patients with type 2 diabetes mellitus and peripheral artery disease: a subanalysis of EMPA-REG OUTCOME. Circulation. 2018;137:405–407.
    1. Verma S., Bhatt D.L., Bain S.C. Effect of liraglutide on cardiovascular events in patients with type 2 diabetes mellitus and polyvascular disease: results of the LEADER trial. Circulation. 2018;137:2179–2183.
    1. Mann J.F., Orsted D.D., Brown-Frandsen K. Liraglutide and renal outcomes in type 2 diabetes. N Engl J Med. 2017;377:839–848.
    1. Marso S.P., Daniels G.H., Brown-Frandsen K. Liraglutide and cardiovascular outcomes in type 2 diabetes. N Engl J Med. 2016;375:311–322.
    1. Verma S., Leiter L.A., Mazer C.D. Liraglutide reduces cardiovascular events and mortality in type 2 diabetes mellitus independently of baseline low-density lipoprotein cholesterol levels and statin use. Circulation. 2018;138:1605–1607.
    1. Verma S., Poulter N.R., Bhatt D.L. Effects of liraglutide on cardiovascular outcomes in patients with type 2 diabetes with or without history of myocardial infarction or stroke: a post hoc analysis from the LEADER trial. Circulation. 2018;137:2179–2183.
    1. Sherman S.E., Bell G.I., Teoh H. Canagliflozin improves the recovery of blood flow in an experimental model of severe limb ischemia. J Am Coll Cardiol Basic Trans Science. 2018;3:327–329.
    1. Verma S., Rawat S., Ho K.L. Empagliflozin increases cardiac energy production in diabetes. Novel translational insights into the heart failure benefits of SGLT2 inhibitors. J Am Coll Cardiol Basic Trans Science. 2018;3:575–587.
    1. Wanner C., Inzucchi S.E., Lachin J.M. Empagliflozin and progression of kidney disease in type 2 diabetes. N Engl J Med. 2016;375:323–334.
    1. Prospective Studies Collaboration, Asia Pacific Cohort Studies Collaboration Sex-specific relevance of diabetes to occlusive vascular and other mortality: a collaborative meta-analysis of individual data from 980 793 adults from 68 prospective studies. Lancet Diabetes Endocrinol. 2018;6:538–546.
    1. Hess D.A., Meyerrose T.E., Wirthlin L. Functional characterization of highly purified human hematopoietic repopulating cells isolated according to aldehyde dehydrogenase activity. Blood. 2004;104:1648–1655.
    1. Chambers S.E., O'Neill C.L., O'Doherty T.M., Medina R.J., Stitt A.W. The role of immune-related myeloid cells in angiogenesis. Immunobiology. 2013;218:1370–1375.
    1. Norgren L., Hiatt W.R., Dormandy J.A. Inter-society consensus for the management of peripheral arterial disease. Int Angiol. 2007;26:81–157.
    1. Bergers G., Song S. The role of pericytes in blood-vessel formation and maintenance. Neuro Oncol. 2005;7:452–464.
    1. Buschmann I., Heil M., Jost M., Schaper W. Influence of inflammatory cytokines on arteriogenesis. Microcirculation. 2003;10:371–379.
    1. King A., Balaji S., Keswani S.G., Crombleholme T.M. The role of stem cells in wound angiogenesis. Adv Wound Care (New Rochelle) 2014;3:614–625.
    1. Fadini G.P., Ferraro F., Quaini F., Asahara T., Madeddu P. Concise review: diabetes, the bone marrow niche, and impaired vascular regeneration. Stem Cells Transl Med. 2014;3:949–957.
    1. Putman D.M., Liu K.Y., Broughton H.C., Bell G.I., Hess D.A. Umbilical cord blood-derived aldehyde dehydrogenase-expressing progenitor cells promote recovery from acute ischemic injury. Stem Cells. 2012;30:2248–2260.
    1. Putman D.M., Cooper T.T., Sherman S.E. Expansion of umbilical cord blood aldehyde dehydrogenase expressing cells generates myeloid progenitor cells that stimulate limb revascularization. Stem Cells Transl Med. 2017;6:1607–1619.
    1. Patel R.S., Li Q., Ghasemzadeh N. Circulating CD34+ progenitor cells and risk of mortality in a population with coronary artery disease. Circ Res. 2015;116:289–297.
    1. Bigarella C.L., Liang R., Ghaffari S. Stem cells and the impact of ROS signaling. Development. 2014;141:4206–4218.
    1. Mangialardi G., Spinetti G., Reni C., Madeddu P. Reactive oxygen species adversely impacts bone marrow microenvironment in diabetes. Antioxid Redox Signal. 2014;21:1620–1633.
    1. Loomans C.J., van Haperen R., Duijs J.M. Differentiation of bone marrow-derived endothelial progenitor cells is shifted into a proinflammatory phenotype by hyperglycemia. Mol Med. 2009;15:152–159.
    1. Mackie A.R., Losordo D.W. CD34-positive stem cells: in the treatment of heart and vascular disease in human beings. Tex Heart Inst J. 2011;38:474–485.
    1. Jetten N., Verbruggen S., Gijbels M.J., Post M.J., De Winther M.P., Donners M.M. Anti-inflammatory M2, but not pro-inflammatory M1 macrophages promote angiogenesis in vivo. Angiogenesis. 2014;17:109–118.
    1. Mathiyalagan P., Liang Y., Kim D. Angiogenic mechanisms of human CD34(+) stem cell exosomes in the repair of ischemic hindlimb. Circ Res. 2017;120:1466–1476.
    1. Urbich C., Heeschen C., Aicher A., Dernbach E., Zeiher A.M., Dimmeler S. Relevance of monocytic features for neovascularization capacity of circulating endothelial progenitor cells. Circulation. 2003;108:2511–2516.
    1. Capoccia B.J., Robson D.L., Levac K.D. Revascularization of ischemic limbs after transplantation of human bone marrow cells with high aldehyde dehydrogenase activity. Blood. 2009;113:5340–5351.
    1. Asahara T., Murohara T., Sullivan A. Isolation of putative progenitor endothelial cells for angiogenesis. Science. 1997;275:964–967.
    1. Asahara T., Masuda H., Takahashi T. Bone marrow origin of endothelial progenitor cells responsible for postnatal vasculogenesis in physiological and pathological neovascularization. Circ Res. 1999;85:221–228.
    1. Hess D.A., Wirthlin L., Craft T.P. Selection based on CD133 and high aldehyde dehydrogenase activity isolates long-term reconstituting human hematopoietic stem cells. Blood. 2006;107:2162–2169.
    1. Siemerink M.J., Klaassen I., Vogels I.M., Griffioen A.W., Van Noorden C.J., Schlingemann R.O. CD34 marks angiogenic tip cells in human vascular endothelial cell cultures. Angiogenesis. 2012;15:151–163.
    1. Fadini G.P., Miorin M., Facco M. Circulating endothelial progenitor cells are reduced in peripheral vascular complications of type 2 diabetes mellitus. J Am Coll Cardiol. 2005;45:1449–1457.
    1. Yoder M.C., Mead L.E., Prater D. Redefining endothelial progenitor cells via clonal analysis and hematopoietic stem/progenitor cell principals. Blood. 2007;109:1801–1809.
    1. Travnickova J., Tran Chau V., Julien E. Primitive macrophages control HSPC mobilization and definitive haematopoiesis. Nat Commun. 2015;6:6227.
    1. Jaipersad A.S., Lip G.Y., Silverman S., Shantsila E. The role of monocytes in angiogenesis and atherosclerosis. J Am Coll Cardiol. 2014;63:1–11.
    1. Dalton H.J., Armaiz-Pena G.N., Gonzalez-Villasana V., Lopez-Berestein G., Bar-Eli M., Sood A.K. Monocyte subpopulations in angiogenesis. Cancer Res. 2014;74:1287–1293.
    1. Heil M., Schaper W. Influence of mechanical, cellular, and molecular factors on collateral artery growth (arteriogenesis) Circ Res. 2004;95:449–458.
    1. Peiser L., Gordon S. The function of scavenger receptors expressed by macrophages and their role in the regulation of inflammation. Microbes Infect. 2001;3:149–159.
    1. Kovacic J.C., Moreno P., Hachinski V., Nabel E.G., Fuster V. Cellular senescence, vascular disease, and aging: part 1 of a 2-part review. Circulation. 2011;123:1650–1660.
    1. Dykstra B., de Haan G. Hematopoietic stem cell aging and self-renewal. Cell Tissue Res. 2008;331:91–101.
    1. Fadini G.P., Ciciliot S., Albiero M. Concise review: perspectives and clinical implications of bone marrow and circulating stem cell defects in diabetes. Stem Cells. 2017;35:106–116.
    1. Werner N., Kosiol S., Scheigel T. Circulating endothelial progenitor cells and cardiovascular outcomes. N Engl J Med. 2005;355:999–1007.
    1. Barcelos L.S., Duplaa C., Krankel N. Human CD133+ progenitor cells promote the healing of diabetic ischemic ulcers by paracrine stimulation of angiogenesis and activation of Wnt signaling. Circ Res. 2009;104:1095–1102.
    1. Rosales C. Neutrophil: a cell with many roles in inflammation or several cell types? Front Physiol. 2018;9:113.
    1. Wong S.L., Demers M., Martinod K. Diabetes primes neutrophils to undergo NETosis, which impairs wound healing. Nat Med. 2015;21:815–819.
    1. Martinez F.O., Gordon S. The M1 and M2 paradigm of macrophage activation: time for reassessment. F1000Prime Rep. 2014;6:13.
    1. Sata M. Role of circulating vascular progenitors in angiogenesis, vascular healing, and pulmonary hypertension: lessons from animal models. Arterioscler Thromb Vasc Biol. 2006;26:1008–1014.
    1. Xu H., Barnes G.T., Yang Q. Chronic inflammation in fat plays a crucial role in the development of obesity-related insulin resistance. J Clin Invest. 2003;112:1821–1830.
    1. Perin E.C., Murphy M., Cooke J.P. Rationale and design for PACE: patients with intermittent claudication injected with ALDH bright cells. Am Heart J. 2014;168:667–673.
    1. Gupta R., Losordo D.W. Cell therapy for critical limb ischemia: moving forward one step at a time. Circ Cardiovasc Interv. 2011;4:2–5.
    1. Losordo D.W., Henry T.D., Davidson C. Intramyocardial, autologous CD34+ cell therapy for refractory angina. Circ Res. 2011;109:428–436.
    1. Lee W.J., Tateya S., Cheng A.M. M2 macrophage polarization mediates anti-inflammatory effects of endothelial nitric oxide signaling. Diabetes. 2015;64:2836–2846.
    1. Liu Y.C., Zou X.B., Chai Y.F., Yao Y.M. Macrophage polarization in inflammatory diseases. Int J Biol Sci. 2014;10:520–529.
    1. Tchkonia T., Zhu Y., van Deursen J., Campisi J., Kirkland J.L. Cellular senescence and the senescent secretory phenotype: therapeutic opportunities. J Clin Invest. 2013;123:966–972.
    1. Freund A., Orjalo A.V., Desprez P.Y., Campisi J. Inflammatory networks during cellular senescence: causes and consequences. Trends Mol Med. 2010;16:238–246.
    1. Porto M.L., Rodrigues B.P., Menezes T.N. Reactive oxygen species contribute to dysfunction of bone marrow hematopoietic stem cells in aged C57BL/6 J mice. J Biomed Sci. 2015;22:97.
    1. Shao L., Li H., Pazhanisamy S.K., Meng A., Wang Y., Zhou D. Reactive oxygen species and hematopoietic stem cell senescence. Int J Hematol. 2011;94:24–32.
    1. Shinohara A., Imai Y., Nakagawa M., Takahashi T., Ichikawa M., Kurokawa M. Intracellular reactive oxygen species mark and influence the megakaryocyte-erythrocyte progenitor fate of common myeloid progenitors. Stem Cells. 2014;32:548–557.
    1. Rauscher F.M., Goldschmidt-Clermont P.J., Davis B.H. Aging, progenitor cell exhaustion, and atherosclerosis. Circulation. 2003;108:457–463.
    1. Williams A.R., Hare J.M. Mesenchymal stem cells: biology, pathophysiology, translational findings, and therapeutic implications for cardiac disease. Circ Res. 2011;109:923–940.
    1. Kfoury Y., Scadden D.T. Mesenchymal cell contributions to the stem cell niche. Cell Stem Cell. 2015;16:239–253.
    1. Caplan A.I., Correa D. The MSC: an injury drugstore. Cell Stem Cell. 2011;9:11–15.
    1. Colter D.C., Sekiya I., Prockop D.J. Identification of a subpopulation of rapidly self-renewing and multipotential adult stem cells in colonies of human marrow stromal cells. Proc Natl Acad Sci U S A. 2001;98:7841–7845.
    1. Zoungas S., Woodward M., Li Q. Impact of age, age at diagnosis and duration of diabetes on the risk of macrovascular and microvascular complications and death in type 2 diabetes. Diabetologia. 2014;57:2465–2474.
    1. Duncan A.W., Rattis F.M., DiMascio L.N. Integration of Notch and Wnt signaling in hematopoietic stem cell maintenance. Nat Immunol. 2005;6:314–322.

Source: PubMed

3
Subskrybuj