Expansion of Umbilical Cord Blood Aldehyde Dehydrogenase Expressing Cells Generates Myeloid Progenitor Cells that Stimulate Limb Revascularization

David M Putman, Tyler T Cooper, Stephen E Sherman, Ayesh K Seneviratne, Mark Hewitt, Gillian I Bell, David A Hess, David M Putman, Tyler T Cooper, Stephen E Sherman, Ayesh K Seneviratne, Mark Hewitt, Gillian I Bell, David A Hess

Abstract

Uncompromised by chronic disease-related comorbidities, human umbilical cord blood (UCB) progenitor cells with high aldehyde dehydrogenase activity (ALDHhi cells) stimulate blood vessel regeneration after intra-muscular transplantation. However, implementation of cellular therapies using UCB ALDHhi cells for critical limb ischemia, the most severe form of severe peripheral artery disease, is limited by the rarity (<0.5%) of these cells. Our goal was to generate a clinically-translatable, allogeneic cell population for vessel regenerative therapies, via ex vivo expansion of UCB ALDHhi cells without loss of pro-angiogenic potency. Purified UCB ALDHhi cells were expanded >18-fold over 6-days under serum-free conditions. Consistent with the concept that ALDH-activity is decreased as progenitor cells differentiate, only 15.1% ± 1.3% of progeny maintained high ALDH-activity after culture. However, compared to fresh UCB cells, expansion increased the total number of ALDHhi cells (2.7-fold), CD34+ /CD133+ cells (2.8-fold), and hematopoietic colony forming cells (7.7-fold). Remarkably, injection of expanded progeny accelerated recovery of perfusion and improved limb usage in immunodeficient mice with femoral artery ligation-induced limb ischemia. At 7 or 28 days post-transplantation, mice transplanted with expanded ALDHhi cells showed augmented endothelial cell proliferation and increased capillary density compared to controls. Expanded cells maintained pro-angiogenic mRNA expression and secreted angiogenesis-associated growth factors, chemokines, and matrix modifying proteins. Coculture with expanded cells augmented human microvascular endothelial cell survival and tubule formation under serum-starved, growth factor-reduced conditions. Expanded UCB-derived ALDHhi cells represent an alternative to autologous bone marrow as an accessible source of pro-angiogenic hematopoietic progenitor cells for the refinement of vascular regeneration-inductive therapies. Stem Cells Translational Medicine 2017;6:1607-1619.

Keywords: Aldehyde dehydrogenase; Angiogenesis; Hematopoietic progenitor cell expansion; Peripheral artery disease; Transplantation; Umbilical cord blood.

© 2017 The Authors Stem Cells Translational Medicine published by Wiley Periodicals, Inc. on behalf of AlphaMed Press.

Figures

Figure 1
Figure 1
Purification and expansion of human UCB ALDHhi cells. Representative flow cytometry plots showing the purification of ALDHhi cells from human UCB. (A): Lin– cells treated with DEAB established selection criteria for ALDHlo (R2) and ALDHhi gates (R3). ALDHhi cells represented a rare subset (2.4% ± 0.4%) of Lin– cells. (B): Representative flow cytometry plots showing expanded progeny with low versus high ALDH‐activity after culture for 3, 6, or 9 days. (C): The frequency of cells retaining high ALDH‐activity diminished as culture time progressed. (D): Fold increase in total cells, ALDHhi cells, and ALDHlo cells was calculated at days 3, 6, or 9 compared to the number of cells seeded at day 0. Cell progeny at day 6 demonstrated significant expansion (2.7 ± 0.4‐fold) of total ALDHhi cells. Data represent mean ± SEM from 15 UCB samples. Statistical analyses were performed by ANOVA with Tukey's multiple comparisons test relative to cells seeded at day 0 (*, p < .05; **, p < .01; ***, p < .001). Abbreviations: DEAB, diethylaminobenzaldehyde; ALDH, aldehyde dehydrogenase; ALDHhi, high aldehyde dehydrogenase activity; ALDHlo, low aldehyde dehydrogenase activity; Lin–, Lineage depleted; UCB, umbilical cord blood.
Figure 2
Figure 2
Expanded cells with high ALDH‐activity coexpressed primitive surface markers. (A): Representative flow cytometry plots showing CD34 and CD133 coexpression on expanded cells at 3, 6, or 9 days culture. (B): The frequency of cells that coexpressed CD34/CD133 was significantly increased in the ALDHhi cell subset. (C): Expansion kinetics for total CD34+ cells, CD133+ cells, or CD34+/CD133+ cells during culture. CD34+, CD133+, and CD34+/CD133+ cell content was significantly increased in total cells at days 6 and 9 compared to Day 0 controls. At day 6, CD34+/CD133+ cells were almost exclusively contained within the ALDHhi cell subset. However, within the ALDHhi subset there was no increase in total CD34+, CD133+, or CD34+/CD133+ cells after day 6. Data represent mean ± SEM from five umbilical cord blood samples. Statistical analyses were performed by ANOVA with Tukey's multiple comparison test (*, p < .05; **, p < .01). Abbreviations: ALDHhi, high aldehyde dehydrogenase activity; ALDHlo, low aldehyde dehydrogenase activity.
Figure 3
Figure 3
Total hematopoietic progenitor cell number was increased after expansion. Fresh umbilical cord blood ALDHhi cells or expanded cells harvested at day 6 or day 9 were plated at limiting dilution in methylcellulose media to compare hematopoietic CFU production. (A): The frequency of CFU‐G and CFU‐M colonies was decreased at 6 or 9 days culture. (B): This resulted in significantly reduced total colony formation from 1 CFU in 3.1 ALDHhi cells fresh cells (Day 0), to 1 CFU in 7.3 expanded cells at day 6, or 1 CFU in 9.4 expanded cells at day 9. (C): Taking into account total cell number was increased 18.1 ± 1.7‐fold at day 6, and 38.9 ± 6.0‐fold at day 9, total CFU content compared to fresh ALDHhi cells was increased 7.7 ± 0.7‐fold after 6 days culture and 12.8 ± 2.0‐fold after 9 days culture. Data represent mean ± SEM from 5 UCB samples. Statistical analyses were performed by ANOVA with Tukey's multiple comparison test (*, p < .05; **, p < .01). Abbreviations: ALDHhi, high aldehyde dehydrogenase; BFU, E, blast forming unit‐ erythrocyte; CFU‐G, colony forming unit‐ granulocyte; CFU‐M, colony forming unit‐ macrophage; CFU‐GM, colony forming unit‐ granulocyte and macrophage; CFU‐mixed, colony forming unit containing erythrocytes, granulocytes, and macorphages.
Figure 4
Figure 4
Transplantation of day 6 expanded cells accelerated the recovery of perfusion and improved usage of the ischemic limb. (A–D): Representative LDPI following femoral artery ligation and i.m.‐injection of PBS (n = 10), 2 × 105 fresh umbilical cord blood (UCB) ALDHhi cells (n = 9), or 5 × 105 expanded cells harvested at day 6 (n = 7) or day 9 (n = 8). (E): Transplantation of fresh ALDHhi cells or day 6 expanded cells accelerated the recovery perfusion compared to PBS‐injected controls. However, transplantation of day 9 expanded cells showed perfusion ratios equivalent to PBS‐injected controls. (F): Noldus catwalk paw print intensity at 7 days post‐transplantation showed mice transplanted with day 6 expanded cells (n = 6) regained use of their ischemic leg faster than PBS‐injected controls (n = 6). Data represent mean ± SEM from 6 UCB samples. Statistical analyses were performed by 2‐way ANOVA with Bonferroni's multiple comparisons test (*, p < .05; ***, p < .001). Abbreviation: PBS, phosphate buffered saline.
Figure 5
Figure 5
Transplantation of day 6 expanded cells increased capillary density in the ischemic hind limb. (A, B): Representative photomicrographs of CD31 staining in ischemic adductor muscle sections taken 28 days after femoral artery ligation and intramuscular injection of PBS or day 6 expanded cells. (C, D): Summary of vessel density in the ischemic or nonischemic limb at 7 or 28 days after transplantation. Mice transplanted with fresh umbilical cord blood (UCB) ALDHhi cells or day 6 expanded cells demonstrated increased capillary density in the surgical hind limb. Data represent mean ± SEM from 5 UCB samples. Statistical analyses were performed by ANOVA with Tukey's multiple comparison test (*, p < .05). Abbreviations: ALDHhi, high aldehyde dehydrogenase; PBS, phosphate buffered saline.
Figure 6
Figure 6
Transplantation of day 6 expanded cells increased endothelial cell proliferation in the ischemic hind limb. (A, B): Representative photomicrographs of CD31 cell proliferation within the ischemic adductor muscle at 7 days post‐transplantation. Mouse CD31+EdU+ cells (arrows) were detected in ischemic muscle of mice transplanted with day 6 expanded cells. (C): Compared to mice injected with PBS or day 9 expanded cell, mice transplanted with day 6 expanded cells showed increased endothelial cell proliferation. Data represent mean ± SEM from 4 umbilical cord blood (UCB) samples. Statistical analyses were performed by ANOVA with Tukey's multiple comparison test (*, p < .05; **, p < .01). Abbreviations: EdU, 5‐ethynyl‐2′‐deoxyuridine; HPC, hematopoietic progenitor cell; PBS, phosphate buffered saline.
Figure 7
Figure 7
Coculture with day 6 expanded cells increased HMVEC tubule formation and survival under serum‐free, growth factor reduced conditions. (A): Contact coculture with day 6 expanded cells in growth factor reduced Matrigel significantly increased tubule formation by HMVEC at 6 and 24 hours. (B): Noncontact (transwell) coculture with day 6 expanded cells promoted HMVEC survival under growth factor‐free, serum‐starved conditions. (C–E): Representative flow cytometry plots of apoptotic HMVEC using 7AAD and Annexin V staining. Noncontact coculture with day 6 expanded cells decreased the frequency of dead (F): and apoptotic (G): HMVEC after 24 hours under serum and growth factor‐free conditions. Data represent mean ± SEM from 4 umbilical cord blood samples. Statistical analyses were performed by ANOVA with Tukey's multiple comparison test (*, p < .05; **, p < .01). Abbreviations: EBM2, endothelial basal media; EGM2, endothelial growth media; HMVEC, human microvascular endothelial cells.

References

    1. Lackland DT, Roccella EJ, Deutsch AF et al. Factors influencing the decline in stroke mortality: A statement from the American Heart Association/American Stroke Association. Stroke 2014;45:315–353.
    1. Dotsenko O. Stem/Progenitor cells, atherosclerosis and cardiovascular regeneration. Open Cardiovasc Med J 2010;4:97–104.
    1. Asahara T, Murohara T, Sullivan A et al. Isolation of putative progenitor endothelial cells for angiogenesis. Science 1997;275:964–967.
    1. Dimmeler S, Zeiher AM, Schneider MD. Unchain my heart: The scientific foundations of cardiac repair. J Clin Invest 2005;115:572–583.
    1. Rafii S, Lyden D. Therapeutic stem and progenitor cell transplantation for organ vascularization and regeneration. Nat Med 2003;9:702–712.
    1. Michler RE. Stem cell therapy for heart failure. Cardiol Rev 2014;22:105–116.
    1. Gupta NK, Armstrong EJ, Parikh SA. The current state of stem cell therapy for peripheral artery disease. Curr Cardiol Rep 2014;16:447.
    1. Botham CM, Bennett WL, Cooke JP. Clinical trials of adult stem cell therapy for peripheral artery disease. Methodist DeBakey Cardiovasc J 2013;9:201–205.
    1. Orlic D, Kajstura J, Chimenti S et al. Bone marrow cells regenerate infarcted myocardium. Nature 2001;410:701–705.
    1. Murray CE, Soonpas MH, Reinecke H et al. Haematopoietic stem cells do not transdifferentiate into cardiac myocytes in myocardial infarcts. Nature 2004;664–668.
    1. Balsam LB, Wagers AJ, Christensen LJ et al. Hematopoietic stem cells adopt mature hematopoietic fates in ischemic myocardium. Nature 2004;428:668–673.
    1. Rosenzweig A. Cardiac cell therapy–mixed results from mixed cells. N Engl J Med 2006;355:1274–1277.
    1. Janssens S, Dubois C, Bogaert J et al. Autologous bone marrow‐derived stem‐cell transfer in patients with ST‐segment elevation myocardial infarction: Double‐blind, randomised controlled trial. Lancet 2006;367:113–121.
    1. Wollert KC, Meyer GP, Lotz J et al. Intracoronary autologous bone‐marrow cell transfer after myocardial infarction: The BOOST randomised controlled clinical trial. Lancet 2004;364:141–148.
    1. Schachinger V, Erbs S, Elsässer A et al. Intracoronary bone marrow‐derived progenitor cells in acute myocardial infarction. N Engl J Med 2006;355:1210–1221.
    1. Tateishi‐Yuyama E, Matsubara H, Murohara T et al. Therapeutic angiogenesis for patients with limb ischaemia by autologous transplantation of bone‐marrow cells: A pilot study and a randomised controlled trial. Lancet 2002;360:427–435.
    1. Higashi Y, Kimura M, Hara K et al. Autologous bone‐marrow mononuclear cell implantation improves endothelium‐dependent vasodilation in patients with limb ischemia. Circulation 2004;109:1215–1218.
    1. Peichev M, Naiyer AJ, Pereira D et al. Expression of VEGFR‐2 and AC133 by circulating human CD34(+) cells identifies a population of functional endothelial precursors. Blood 2000;95:952–958.
    1. Schatteman GC, Hanlon HD, Jiao C et al. Blood‐derived angioblasts accelerate blood‐flow restoration in diabetic mice. J Clin Invest 2000;106:571–578.
    1. Urbich C, Heeschen C, Aicher A et al. Relevance of monocytic features for neovascularization capacity of circulating endothelial progenitor cells. Circulation 2003;108:2511–2516.
    1. Yoder MC, Mead LE, Prater D et al. Redefining endothelial progenitor cells via clonal analysis and hematopoietic stem/progenitor cell principals. Blood 2007;109:1801–1809.
    1. Capoccia BJ, Robson DL, Levac KD et al. Revascularization of ischemic limbs after transplantation of human bone marrow cells with high aldehyde dehydrogenase activity. Blood 2009;113:5340–5351.
    1. Hess DA, Craft TP, Wirthlin L et al. Widespread nonhematopoietic tissue distribution by transplanted human progenitor cells with high aldehyde dehydrogenase activity. Stem Cells 2008;26:611–620.
    1. Wara AK, Croce K, Foo S et al. Bone marrow‐derived CMPs and GMPs represent highly functional proangiogenic cells: Implications for ischemic cardiovascular disease. Blood 2011;118:6461–6464.
    1. Okuno Y, Nakamura‐Ishizu A, Kishi K et al. Bone marrow‐derived cells serve as proangiogenic macrophages but not endothelial cells in wound healing. Blood 2011;117:5264–5272.
    1. Ohtani K, Vlachojannis GJ, Koyanagi M et al. Epigenetic regulation of endothelial lineage committed genes in pro‐angiogenic hematopoietic and endothelial progenitor cells. Circ Res 2011;109:1219–1229.
    1. Grunewald M, Avraham I, Dor Y et al. VEGF‐induced adult neovascularization: Recruitment, retention, and role of accessory cells. Cell 2006;124:175–189.
    1. Putman DM, Liu KY, Broughton HC et al. Umbilical cord blood‐derived aldehyde dehydrogenase‐expressing progenitor cells promote recovery from acute ischemic injury. Stem Cells 2012;30:2248–2260.
    1. Perin EC, Silva G, Gahremanpour A et al. A randomized, controlled study of autologous therapy with bone marrow‐derived aldehyde dehydrogenase bright cells in patients with critical limb ischemia. Catheter Cardiovasc Interv 2011;78:1060–1067.
    1. Vasa M, Fichtlscherer S, Aicher A et al. Number and migratory activity of circulating endothelial progenitor cells inversely correlate with risk factors for coronary artery disease. Circ Res 2001;89:E1–E7.
    1. Tepper OM, Carr J, Allen RJ Jr. et al. Decreased circulating progenitor cell number and failed mechanisms of stromal cell‐derived factor‐1alpha mediated bone marrow mobilization impair diabetic tissue repair. Diabetes 2010;59:1974–1983.
    1. Fadini GP, Sartore S, Albiero M et al. Number and function of endothelial progenitor cells as a marker of severity for diabetic vasculopathy. Arterioscler Thromb Vasc Biol 2006;26:2140–2146.
    1. Jialal I, Devaraj S, Singh U et al. Decreased number and impaired functionality of endothelial progenitor cells in subjects with metabolic syndrome: Implications for increased cardiovascular risk. Atherosclerosis 2010. ; 211:297–302.
    1. Perin EC, Murphy M, Cooke JP et al. Rationale and design for PACE: Patients with Intermittent claudication injected with ALDH bright cells. American Heart J 2014;168:667–673.
    1. Bhatia M, Bonnet D, Kapp U et al. Quantitative analysis reveals expansion of human hematopoietic repopulating cells after short‐term expansion. J.Exp Med 1997;186:619–624.
    1. Chute JP, Muramoto GG, Whitesides J et al. Inhibition of aldehyde dehydrogenase and retinoid signaling induces the expansion of human hematopoietic stem cells. Proc Natl Acad Sci USA 2006;103:11707–11712.
    1. Boitano AE, Wang J, Romeo R et al. Aryl hydrocarbon receptor antagonist promotes the expansion of human hematopoietic stem cells. Science 2011;329:1345–1348.
    1. Csaszar E, Kirouac DC, Yu M et al. Rapid expansion of human hematopoietic stem cells by automated control of inhibitory feedback signaling. Cell Stem Cell 2012;10:218–229.
    1. Dahlberg A, Delaney C, Bernstein ID. Ex vivo expansion of human hematopoietic stem and progenitor cells. Blood 2011;117:6083–6090.
    1. Seneviratne AK, Bell GI, Sherman SE et al. Expanded hematopoietic progenitor cells with high aldehyde dehydrogenase activity demonstrate islet regenerative functions. Stem Cells 2016;34:873–887.
    1. Perin EC, Silva GV, Zheng Y et al. Randomized, double‐blind pilot study of transendocardial injection of autologous aldehyde dehydrogenase‐bright stem cells in patients with ischemic heart failure. Am. Heart J 2012;163:415–21–421.e1.
    1. Berezin AE, Kremzer AA. Circulating endothelial progenitor cells as markers for severity of ischemic chronic heart failure. J Card Fail 2014;6:438–447.
    1. Eklund L, Saharinen P. Angiopoietin signaling in the vasculature. Exp Cell Res 2013;319:1271–1280.
    1. Carmeliet P, Jain RK. Molecular mechanisms and clinical applications of angiogenesis. Nature 2011;473:298–307.
    1. Zhang F, Tang Z, Hou X et al. VEGF‐B is dispensable for blood vessel growth but critical for their survival, and VEGF‐B targeting inhibits pathological angiogenesis. Proc Natl Acad Sci USA 2009;106:6152–6157.
    1. Holash J. Vessel cooption, regression, and growth in tumors mediated by angiopoietins and VEGF. Science 1999;284:1994–1998.
    1. Stanic B, Pandey D, Fulton DJ et al. Increased epidermal growth factor‐like ligands are associated with elevated vascular nicotinamide adenine dinucleotide phosphate oxidase in a primate model of atherosclerosis. Arterioscler Thromb Vasc Biol 2012;32:2452–2460.
    1. Kato M, Inazu T, Kawai Y et al. Amphiregulin is a potent mitogen for the vascular smooth muscle cell line, A7r5. Biochem Biophys Res Commun 2003;301:1109–1115.
    1. Dreux AC, Lamb DJ, Modjtahedi H et al. The epidermal growth factor receptors and their family of ligands: Their putative role in atherogenesis. Atherosclerosis 2006;186:38–53.
    1. Makki N, Thiel K, Miller F. The epidermal growth factor receptor and its ligands in cardiovascular disease. IJMS 2013;14:20597–20613.
    1. Fiedler U, Scharpfenecker M, Koidl S et al. The Tie‐2 ligand angiopoietin‐2 is stored in and rapidly released upon stimulation from endothelial cell Weibel‐Palade bodies. Blood 2004;103:4150–4156.
    1. Fiedler U, Augustin HG. Angiopoietins: A link between angiogenesis and inflammation. Trends Immunol 2006;27:552–558.
    1. Crola Da Silva C, Lamerant‐Fayel N et al. Selective human endothelial cell activation by chemokines as a guide to cell homing. Immunology 2009;126:394–404.

Source: PubMed

3
Abonnieren