Diabetes impairs stem cell and proangiogenic cell mobilization in humans

Gian Paolo Fadini, Mattia Albiero, Saula Vigili de Kreutzenberg, Elisa Boscaro, Roberta Cappellari, Mariacristina Marescotti, Nicol Poncina, Carlo Agostini, Angelo Avogaro, Gian Paolo Fadini, Mattia Albiero, Saula Vigili de Kreutzenberg, Elisa Boscaro, Roberta Cappellari, Mariacristina Marescotti, Nicol Poncina, Carlo Agostini, Angelo Avogaro

Abstract

Objective: Diabetes mellitus (DM) increases cardiovascular risk, at least in part, through shortage of vascular regenerative cells derived from the bone marrow (BM). In experimental models, DM causes morphological and functional BM alterations, but information on BM function in human DM is missing. Herein, we sought to assay mobilization of stem and proangiogenic cells in subjects with and without DM.

Research design and methods: In a prospective trial (NCT01102699), we tested BM responsiveness to 5 μg/kg human recombinant granulocyte colony-stimulating factor (hrG-CSF) in 24 individuals with DM (10 type 1 and 14 type 2) and 14 individuals without DM. Before and 24 h after hrG-CSF, we quantified circulating stem/progenitor cells and total and differential white blood cell counts. We also evaluated in vivo the proangiogenic capacity of peripheral blood mononuclear cells using the Matrigel plug assay.

Results: In response to hrG-CSF, levels of CD34(+) cells and other progenitor cell phenotypes increased in subjects without DM. Patients with DM had significantly impaired mobilization of CD34(+), CD133(+), and CD34(+)CD133(+) hematopoietic stem cells and CD133(+)KDR(+) endothelial progenitors, independently of potential confounders. The in vivo angiogenic capacity of peripheral blood mononuclear cells significantly increased after hrG-CSF in control subjects without DM, but not in patients with DM. DM was also associated with the inability to upregulate CD26/DPP-4 on CD34(+) cells, which is required for the mobilizing effect of granulocyte colony-stimulating factor.

Conclusions: Stem and proangiogenic cell mobilization in response to hrG-CSF is impaired in DM, possibly because of maladaptive CD26/DPP-4 regulation. These alterations may hamper tissue repair and favor the development of cardiovascular complications.

Figures

Figure 1
Figure 1
CD34+ cell mobilization after hrG-CSF. A: Absolute pre-G-CSF and post-G-CSF CD34+ cell count in all DM and all non–DM control (Ctrl) subjects (study primary end point). *P < 0.05 vs. baseline. Absolute pre-G-CSF and post-G-CSF CD34+ cell count in type 1 DM (T1D) compared with young control subjects (B) and in type 2 DM (T2D) compared with age-matched control subjects (C). *P < 0.05 vs. baseline. D: Changes in the absolute levels of circulating CD34+ cell counts. *P < 0.05 vs. DM.
Figure 2
Figure 2
Mobilization of other progenitor cell phenotypes after hrG-CSF. AE: Absolute pre-G-CSF and post-G-CSF cell counts of circulating hematopoietic stem cells (CD133+, CD34+CD133+) and endothelial progenitor cells (CD34+KDR+, CD133+KDR+, and CD34+CD133+KDR+) in DM and non–DM patients. *P < 0.05 vs. baseline. F: Changes in the absolute levels of stem/progenitor cell phenotypes in DM and non–DM patients. *P < 0.05 vs. non–DM control subjects.
Figure 3
Figure 3
Effects of hrG-CSF on CD26/DPP-4 expression. Percentage CD26/DPP-4 expression on CD34+ cells was significantly (*P < 0.05) increased in non–DM control subjects, whereas it was reduced in DM patients.
Figure 4
Figure 4
In vivo proangiogenic cell function. Patients’ PBMCs before collected pre-G-CSF and post-G-CSF were embedded into Matrigel plugs and implanted in immunodeficient mice. Representative gross appearance of the plugs (scale bar = 1 cm) and histological sections (scale bar = 100 μm) are shown. In the bottom, quantification of hemoglobin content, a quantitative surrogate of perfusion, adjusted for mobilized monocytes is shown. *P < 0.05 vs. pre-G-CSF. †P < 0.05 vs. non–DM.

References

    1. Fadini GP, Avogaro A. It is all in the blood: the multifaceted contribution of circulating progenitor cells in diabetic complications. Exp Diabetes Res 2012;2012:742976
    1. Fadini GP. An underlying principle for the study of circulating progenitor cells in diabetes and its complications. Diabetologia 2008;51:1091–1094
    1. Fadini GP, Sartore S, Schiavon M, et al. Diabetes impairs progenitor cell mobilisation after hindlimb ischaemia-reperfusion injury in rats. Diabetologia 2006;49:3075–3084
    1. Oikawa A, Siragusa M, Quaini F, et al. Diabetes mellitus induces bone marrow microangiopathy. Arterioscler Thromb Vasc Biol 2010;30:498–508
    1. Busik JV, Tikhonenko M, Bhatwadekar A, et al. Diabetic retinopathy is associated with bone marrow neuropathy and a depressed peripheral clock. J Exp Med 2009;206:2897–2906
    1. Orlandi A, Chavakis E, Seeger F, Tjwa M, Zeiher AM, Dimmeler S. Long-term diabetes impairs repopulation of hematopoietic progenitor cells and dysregulates the cytokine expression in the bone marrow microenvironment in mice. Basic Res Cardiol 2010;105:703–712
    1. Ferraro F, Lymperi S, Mendez-Ferrer S, et al. Diabetes impairs hematopoietic stem cell mobilization by altering niche function. Sci Transl Med 2011;3:104ra101
    1. DiPersio JF. Diabetic stem-cell “mobilopathy”. N Engl J Med 2011;365:2536–2538
    1. Fadini GP, Boscaro E, de Kreutzenberg S, et al. Time course and mechanisms of circulating progenitor cell reduction in the natural history of type 2 diabetes. Diabetes Care 2010;33:1097–1102
    1. Roberts AWG-CSF. a key regulator of neutrophil production, but that’s not all! Growth Factors 2005;23:33–41
    1. Fadini GP, Albiero M, Menegazzo L, de Kreutzenberg SV, Avogaro A. The increased dipeptidyl peptidase-4 activity is not counteracted by optimized glucose control in type 2 diabetes, but is lower in metformin-treated patients. Diabetes Obes Metab 2012;14:518–522
    1. Fadini GP. Is bone marrow another target of diabetic complications? Eur J Clin Invest 2011;41:457–463
    1. van Der Auwera P, Platzer E, Xu ZX, et al. Pharmacodynamics and pharmacokinetics of single doses of subcutaneous pegylated human G-CSF mutant (Ro 25-8315) in healthy volunteers: comparison with single and multiple daily doses of filgrastim. Am J Hematol 2001;66:245–251
    1. Fadini GP, de Kreutzenberg SV, Coracina A, et al. Circulating CD34+ cells, metabolic syndrome, and cardiovascular risk. Eur Heart J 2006;27:2247–2255
    1. Rehman J, Li J, Orschell CM, March KL. Peripheral blood “endothelial progenitor cells” are derived from monocyte/macrophages and secrete angiogenic growth factors. Circulation 2003;107:1164–1169
    1. Pucci F, Venneri MA, Biziato D, et al. A distinguishing gene signature shared by tumor-infiltrating Tie2-expressing monocytes, blood “resident” monocytes, and embryonic macrophages suggests common functions and developmental relationships. Blood 2009;114:901–914
    1. Christopherson KW, 2nd, Uralil SE, Porecha NK, Zabriskie RC, Kidd SM, Ramin SM. G-CSF- and GM-CSF-induced upregulation of CD26 peptidase downregulates the functional chemotactic response of CD34+CD38- human cord blood hematopoietic cells. Exp Hematol 2006;34:1060–1068
    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. Christopherson KW, 2nd, Cooper S, Broxmeyer HE. Cell surface peptidase CD26/DPPIV mediates G-CSF mobilization of mouse progenitor cells. Blood 2003;101:4680–4686
    1. Nishimura Y, Ii M, Qin G, et al. CXCR4 antagonist AMD3100 accelerates impaired wound healing in diabetic mice. J Invest Dermatol 2012;132:711–720
    1. Fiorina P, Jurewicz M, Vergani A, et al. Targeting the CXCR4-CXCL12 axis mobilizes autologous hematopoietic stem cells and prolongs islet allograft survival via programmed death ligand 1. J Immunol 2011;186:121–131
    1. Du XL, Edelstein D, Dimmeler S, Ju Q, Sui C, Brownlee M. Hyperglycemia inhibits endothelial nitric oxide synthase activity by posttranslational modification at the Akt site. J Clin Invest 2001;108:1341–1348
    1. Aicher A, Heeschen C, Mildner-Rihm C, et al. Essential role of endothelial nitric oxide synthase for mobilization of stem and progenitor cells. Nat Med 2003;9:1370–1376
    1. de Resende MM, Huw LY, Qian HS, Kauser K. Role of endothelial nitric oxide in bone marrow-derived progenitor cell mobilization. Handb Exp Pharmacol 2007;(108):37–44
    1. Fadini GP, Boscaro E, Albiero M, et al. The oral dipeptidyl peptidase-4 inhibitor sitagliptin increases circulating endothelial progenitor cells in patients with type 2 diabetes: possible role of stromal-derived factor-1alpha. Diabetes Care 2010;33:1607–1609
    1. Boodhwani M, Sodha NR, Mieno S, et al. Functional, cellular, and molecular characterization of the angiogenic response to chronic myocardial ischemia in diabetes. Circulation 2007;116(Suppl):I31–I37
    1. Caporali A, Meloni M, Völlenkle C, et al. Deregulation of microRNA-503 contributes to diabetes mellitus-induced impairment of endothelial function and reparative angiogenesis after limb ischemia. Circulation 2011;123:282–291
    1. Fadini GP, Maruyama S, Ozaki T, et al. Circulating progenitor cell count for cardiovascular risk stratification: a pooled analysis. PLoS ONE 2010;5:e11488.
    1. Loomans CJ, de Koning EJ, Staal FJ, et al. Endothelial progenitor cell dysfunction: a novel concept in the pathogenesis of vascular complications of type 1 diabetes. Diabetes 2004;53:195–199
    1. Tepper OM, Galiano RD, Capla JM, et al. Human endothelial progenitor cells from type II diabetics exhibit impaired proliferation, adhesion, and incorporation into vascular structures. Circulation 2002;106:2781–2786
    1. Fadini GP, Agostini C, Avogaro A. Autologous stem cell therapy for peripheral arterial disease meta-analysis and systematic review of the literature. Atherosclerosis 2010;209:10–17

Source: PubMed

3
Abonnieren