Reactive oxygen species adversely impacts bone marrow microenvironment in diabetes

Giuseppe Mangialardi, Gaia Spinetti, Carlotta Reni, Paolo Madeddu, Giuseppe Mangialardi, Gaia Spinetti, Carlotta Reni, Paolo Madeddu

Abstract

Significance: Patients with diabetes mellitus suffer an excess of cardiovascular complications and recover worse from them as compared with their nondiabetic peers. It is well known that microangiopathy is the cause of renal damage, blindness, and heart attacks in patients with diabetes. This review highlights molecular deficits in stem cells and a supporting microenvironment, which can be traced back to oxidative stress and ultimately reduce stem cells therapeutic potential in diabetic patients.

Recent advances: New research has shown that increased oxidative stress contributes to inducing microangiopathy in bone marrow (BM), the tissue contained inside the bones and the main source of stem cells. These precious cells not only replace old blood cells but also exert an important reparative function after acute injuries and heart attacks.

Critical issues: The starvation of BM as a consequence of microangiopathy can lead to a less efficient healing in diabetic patients with ischemic complications. Furthermore, stem cells from a patient's BM are the most used in regenerative medicine trials to mend hearts damaged by heart attacks.

Future directions: A deeper understanding of redox signaling in BM stem cells will lead to new modalities for preserving local and systemic homeostasis and to more effective treatments of diabetic cardiovascular complications.

Figures

FIG. 1.
FIG. 1.
BM microenvironment organization. The BM is spatially organized into different niches. The osteoblastic niche (left) resides in the inner part of BM, lining on the endosteum. It includes the most primitive stem cells and a frame of stromal cells, principally osteoblasts. The vascular niche (right) is constituted by committed progenitor elements that are embedded in a frame of several kinds of stromal cells (ECs, macrophages, perycites, etc.). In both niches, stem and progenitor cells establish cell–cell contacts that are instrumental to reciprocal control of cell function. Ang-1, angiopoietin 1; BM, bone marrow; cKit, stem cell factor receptor; EC, endothelial cell; MMP-9, metalloproteinase 9; N-Cad, N-cadherin; SCF, also known as kit-ligand, stem cell factor; SDF-1, stromal cell-derived factor-1; SDF-1 receptor, CXCR4, C-X-C chemokine receptor type 4; Tie2, angiopoietin receptor 2; VCAM-1, integrin receptor vascular cell adhesion molecule-1; very late antigen-4, Vla-4, integrin alpha4beta1. To see this illustration in color, the reader is referred to the web version of this article at www.liebertpub.com/ars
FIG. 2.
FIG. 2.
Oxidative stress network in BM microenvironment. In BM, the sources of ROS are principally represented by the mitochondria and NADPH oxidase catalytic subunit NOX. Superoxide produced is then converted by superoxide dismutase into hydrogen peroxide and scavenged by catalase or glutathione peroxidase. ROS can enhance or decrease stem cell functions by activating or inhibiting different signaling pathways. For example, ROS induce differentiation and migration through the PTEN/PI3K/Akt pathway, sustain quiescence via FOXOs and ATM, and induce senescence through p38 MAPK pathway. ATM, ataxia telangiectasia mutated protein; FOXOs, forkhead box transcription factors; MAPK, mitogen-activated protein kinase; PI3K, phosphatidylinositide 3-kinase; PTEN, phosphatase and tensin homolog; ROS, reactive oxygen species. To see this illustration in color, the reader is referred to the web version of this article at www.liebertpub.com/ars
FIG. 3.
FIG. 3.
Proposed model of microenvironmental alteration in diabetic BM. Representation of endosteal and vascular niches in healthy and diabetic conditions. Different colors identify HSCs (blue), HPCs (green), MSCs (beige), and ECs (brown). Erythrocytes inside the marrow vessels are represented in red. Excessive production of ROS in the marrow microenvironment tends to cause a disruption of stem and progenitor cell homeostasis. The ROS gradient is lost, thus triggering stem cell proliferation and differentiation in areas of the marrow that are devoted to maintaining stem cell quiescence. Furthermore, failure of sinusoidal barrier function causes unselective mobilization, skewed toward inflammatory cells instead of regenerative cells. In addition, microvascular rarefaction causes hypoperfusion and deprives stem cells of necessary trophic inputs, leading to stem cells apoptosis. Altogether, these changes jeopardize the regenerative capacity of BM with consequences for peripheral complications. HPCs, hematopoietic progenitor cells; HSCs, hematopoietic stem cells; MSCs, mesenchymal stem cells. To see this illustration in color, the reader is referred to the web version of this article at www.liebertpub.com/ars
FIG. 4.
FIG. 4.
Influence of ROS on microRNA generation. Recent evidence indicates that miRs control several functions of stem cells. ROS can influence the regulation of gene expression through interference of miRs. This influence is exerted at different levels of miR generation and processing. miR, microRNA. To see this illustration in color, the reader is referred to the web version of this article at www.liebertpub.com/ars
FIG. 5.
FIG. 5.
Diabetes-induced mobilopathy. Stem and progenitor cell mobilization from BM is a multistep process triggered by a gradient of chemokines generated from ischemic tissues. Chemokines also promote proteolytic activity in the marrow environment, thus favoring stem and progenitor cell relocation from the endosteal niche to the vascular niche and then the passage to the bloodstream. Oxidative stress generated in stem cells by activation of NOX2 acts as a mediator of those chemokines. However, sustained and excessive oxidative stress can lead to eNOS uncoupling, thereby compromising stem cell mobilization. eNOS, endothelial nitric oxide synthase. To see this illustration in color, the reader is referred to the web version of this article at www.liebertpub.com/ars

References

    1. Abbas HA, Maccio DR, Coskun S, Jackson JG, Hazen AL, Sills TM, You MJ, Hirschi KK, and Lozano G. Mdm2 is required for survival of hematopoietic stem cells/progenitors via dampening of ROS-induced p53 activity. Cell Stem Cell 7: 606–617, 2010
    1. Abraham RT. and Tibbetts RS. Cell biology. Guiding ATM to broken DNA. Science 308: 510–511, 2005
    1. Ago T, Kitazono T, Ooboshi H, Iyama T, Han YH, Takada J, Wakisaka M, Ibayashi S, Utsumi H, and Iida M. Nox4 as the major catalytic component of an endothelial NAD(P)H oxidase. Circulation 109: 227–233, 2004
    1. Ahlqvist KJ, Hamalainen RH, Yatsuga S, Uutela M, Terzioglu M, Gotz A, Forsstrom S, Salven P, Angers-Loustau A, Kopra OH, Tyynismaa H, Larsson NG, Wartiovaara K, Prolla T, Trifunovic A, and Suomalainen A. Somatic progenitor cell vulnerability to mitochondrial DNA mutagenesis underlies progeroid phenotypes in Polg mutator mice. Cell Metab 15: 100–109, 2012
    1. Amadesi S, Reni C, Katare R, Meloni M, Oikawa A, Beltrami AP, Avolio E, Cesselli D, Fortunato O, Spinetti G, Ascione R, Cangiano E, Valgimigli M, Hunt SP, Emanueli C, and Madeddu P. Role for substance p-based nociceptive signaling in progenitor cell activation and angiogenesis during ischemia in mice and in human subjects. Circulation 125: 1774–1786, S1–S19, 2012
    1. Assmus B, Urbich C, Aicher A, Hofmann WK, Haendeler J, Rossig L, Spyridopoulos I, Zeiher AM, and Dimmeler S. HMG-CoA reductase inhibitors reduce senescence and increase proliferation of endothelial progenitor cells via regulation of cell cycle regulatory genes. Circ Res 92: 1049–1055, 2003
    1. Barbagallo I, Vanella A, Peterson SJ, Kim DH, Tibullo D, Giallongo C, Vanella L, Parrinello N, Palumbo GA, Di Raimondo F, Abraham NG, and Asprinio D. Overexpression of heme oxygenase-1 increases human osteoblast stem cell differentiation. J Bone Miner Metab 28: 276–288, 2010
    1. Bedard K. and Krause KH. The NOX family of ROS-generating NADPH oxidases: physiology and pathophysiology. Physiol Rev 87: 245–313, 2007
    1. Beerman I, Bock C, Garrison BS, Smith ZD, Gu H, Meissner A, and Rossi DJ. Proliferation-dependent alterations of the DNA methylation landscape underlie hematopoietic stem cell aging. Cell Stem Cell 12: 413–425, 2013
    1. Bjelakovic G, Nikolova D, Gluud LL, Simonetti RG, and Gluud C. Mortality in randomized trials of antioxidant supplements for primary and secondary prevention: systematic review and meta-analysis. JAMA 297: 842–857, 2007
    1. Boueiz A. and Hassoun PM. Regulation of endothelial barrier function by reactive oxygen and nitrogen species. Microvasc Res 77: 26–34, 2009
    1. Boyle JP, Thompson TJ, Gregg EW, Barker LE, and Williamson DF. Projection of the year 2050 burden of diabetes in the US adult population: dynamic modeling of incidence, mortality, and prediabetes prevalence. Popul Health Metr 8: 29, 2010
    1. Chandra S, Romero MJ, Shatanawi A, Alkilany AM, Caldwell RB, and Caldwell RW. Oxidative species increase arginase activity in endothelial cells through the RhoA/Rho kinase pathway. Br J Pharmacol 165: 506–519, 2012
    1. Chen C, Liu Y, Liu R, Ikenoue T, Guan KL, and Zheng P. TSC-mTOR maintains quiescence and function of hematopoietic stem cells by repressing mitochondrial biogenesis and reactive oxygen species. J Exp Med 205: 2397–2408, 2008
    1. Dagan LN, Jiang X, Bhatt S, Cubedo E, Rajewsky K, and Lossos IS. miR-155 regulates HGAL expression and increases lymphoma cell motility. Blood 119: 513–520, 2012
    1. De Falco E, Avitabile D, Totta P, Straino S, Spallotta F, Cencioni C, Torella AR, Rizzi R, Porcelli D, Zacheo A, Di Vito L, Pompilio G, Napolitano M, Melillo G, Capogrossi MC, and Pesce M. Altered SDF-1-mediated differentiation of bone marrow-derived endothelial progenitor cells in diabetes mellitus. J Cell Mol Med 13: 3405–3414, 2009
    1. Dernbach E, Urbich C, Brandes RP, Hofmann WK, Zeiher AM, and Dimmeler S. Antioxidative stress-associated genes in circulating progenitor cells: evidence for enhanced resistance against oxidative stress. Blood 104: 3591–3597, 2004
    1. Diebold I, Djordjevic T, Petry A, Hatzelmann A, Tenor H, Hess J, and Gorlach A. Phosphodiesterase 2 mediates redox-sensitive endothelial cell proliferation and angiogenesis by thrombin via Rac1 and NADPH oxidase 2. Circ Res 104: 1169–1177, 2009
    1. DiPersio JF. Diabetic stem-cell “mobilopathy”. N Engl J Med 365: 2536–2538, 2011
    1. Donahue RE, Jin P, Bonifacino AC, Metzger ME, Ren J, Wang E, and Stroncek DF. Plerixafor (AMD3100) and granulocyte colony-stimulating factor (G-CSF) mobilize different CD34+ cell populations based on global gene and microRNA expression signatures. Blood 114: 2530–2541, 2009
    1. Du X, Matsumura T, Edelstein D, Rossetti L, Zsengeller Z, Szabo C, and Brownlee M. Inhibition of GAPDH activity by poly(ADP-ribose) polymerase activates three major pathways of hyperglycemic damage in endothelial cells. J Clin Invest 112: 1049–1057, 2003
    1. El-Remessy AB, Tawfik HE, Matragoon S, Pillai B, Caldwell RB, and Caldwell RW. Peroxynitrite mediates diabetes-induced endothelial dysfunction: possible role of Rho kinase activation. Exp Diabetes Res 2010: 247861, 2010
    1. Engelgau MM, Geiss LS, Saaddine JB, Boyle JP, Benjamin SM, Gregg EW, Tierney EF, Rios-Burrows N, Mokdad AH, Ford ES, Imperatore G, and Narayan KM. The evolving diabetes burden in the United States. Ann Intern Med 140: 945–950, 2004
    1. Fadini GP, Boscaro E, de Kreutzenberg S, Agostini C, Seeger F, Dimmeler S, Zeiher A, Tiengo A, and Avogaro A. Time course and mechanisms of circulating progenitor cell reduction in the natural history of type 2 diabetes. Diabetes Care 33: 1097–1102, 2010
    1. Fadini GP, de Kreutzenberg SV, Boscaro E, Albiero M, Cappellari R, Krankel N, Landmesser U, Toniolo A, Bolego C, Cignarella A, Seeger F, Dimmeler S, Zeiher A, Agostini C, and Avogaro A. An unbalanced monocyte polarisation in peripheral blood and bone marrow of patients with type 2 diabetes has an impact on microangiopathy. Diabetologia 56: 1856–1866, 2013
    1. Fadini GP, Losordo D, and Dimmeler S. Critical reevaluation of endothelial progenitor cell phenotypes for therapeutic and diagnostic use. Circ Res 110: 624–637, 2012
    1. Fadini GP, Miorin M, Facco M, Bonamico S, Baesso I, Grego F, Menegolo M, de Kreutzenberg SV, Tiengo A, Agostini C, and Avogaro A. Circulating endothelial progenitor cells are reduced in peripheral vascular complications of type 2 diabetes mellitus. J Am Coll Cardiol 45: 1449–1457, 2005
    1. Fadini GP, Sartore S, Albiero M, Baesso I, Murphy E, Menegolo M, Grego F, Vigili de Kreutzenberg S, Tiengo A, Agostini C, and Avogaro A. Number and function of endothelial progenitor cells as a marker of severity for diabetic vasculopathy. Arterioscler Thromb Vasc Biol 26: 2140–2146, 2006
    1. Fadini GP, Sartore S, Schiavon M, Albiero M, Baesso I, Cabrelle A, Agostini C, and Avogaro A. Diabetes impairs progenitor cell mobilisation after hindlimb ischaemia-reperfusion injury in rats. Diabetologia 49: 3075–3084, 2006
    1. Falanga V. Wound healing and its impairment in the diabetic foot. Lancet 366: 1736–1743, 2005
    1. Ferraro F, Lymperi S, Mendez-Ferrer S, Saez B, Spencer JA, Yeap BY, Masselli E, Graiani G, Prezioso L, Rizzini EL, Mangoni M, Rizzoli V, Sykes SM, Lin CP, Frenette PS, Quaini F, and Scadden DT. Diabetes impairs hematopoietic stem cell mobilization by altering niche function. Sci Transl Med 3: 104ra101, 2011
    1. Fraser PA. The role of free radical generation in increasing cerebrovascular permeability. Free Radic Biol Med 51: 967–977, 2011
    1. Fujisaki J, Wu J, Carlson AL, Silberstein L, Putheti P, Larocca R, Gao W, Saito TI, Lo Celso C, Tsuyuzaki H, Sato T, Cote D, Sykes M, Strom TB, Scadden DT, and Lin CP. In vivo imaging of Treg cells providing immune privilege to the haematopoietic stem-cell niche. Nature 474: 216–219, 2011
    1. Gadau S, Emanueli C, Van Linthout S, Graiani G, Todaro M, Meloni M, Campesi I, Invernici G, Spillmann F, Ward K, and Madeddu P. Benfotiamine accelerates the healing of ischaemic diabetic limbs in mice through protein kinase B/Akt-mediated potentiation of angiogenesis and inhibition of apoptosis. Diabetologia 49: 405–420, 2006
    1. Gao L. and Mann GE. Vascular NAD(P)H oxidase activation in diabetes: a double-edged sword in redox signalling. Cardiovasc Res 82: 9–20, 2009
    1. Garrison BS. and Rossi DJ. Reactive oxygen species resulting from mitochondrial mutation diminishes stem and progenitor cell function. Cell Metab 15: 2–3, 2012
    1. Georgantas RW, 3rd, Hildreth R, Morisot S, Alder J, Liu CG, Heimfeld S, Calin GA, Croce CM, and Civin CI. CD34+ hematopoietic stem-progenitor cell microRNA expression and function: a circuit diagram of differentiation control. Proc Natl Acad Sci U S A 104: 2750–2755, 2007
    1. Golan K, Vagima Y, Ludin A, Itkin T, Cohen-Gur S, Kalinkovich A, Kollet O, Kim C, Schajnovitz A, Ovadya Y, Lapid K, Shivtiel S, Morris AJ, Ratajczak MZ, and Lapidot T. S1P promotes murine progenitor cell egress and mobilization via S1P1-mediated ROS signaling and SDF-1 release. Blood 119: 2478–2488, 2012
    1. Gorlach A, Brandes RP, Nguyen K, Amidi M, Dehghani F, and Busse R. A gp91phox containing NADPH oxidase selectively expressed in endothelial cells is a major source of oxygen radical generation in the arterial wall. Circ Res 87: 26–32, 2000
    1. Haddad P, Dussault S, Groleau J, Turgeon J, Michaud SE, Menard C, Perez G, Maingrette F, and Rivard A. Nox2-containing NADPH oxidase deficiency confers protection from hindlimb ischemia in conditions of increased oxidative stress. Arterioscler Thromb Vasc Biol 29: 1522–1528, 2009
    1. Hazra S, Jarajapu YP, Stepps V, Caballero S, Thinschmidt JS, Sautina L, Bengtsson N, Licalzi S, Dominguez J, Kern TS, Segal MS, Ash JD, Saban DR, Bartelmez SH, and Grant MB. Long-term type 1 diabetes influences haematopoietic stem cells by reducing vascular repair potential and increasing inflammatory monocyte generation in a murine model. Diabetologia 56: 644–653, 2013
    1. He J, Xu Q, Jing Y, Agani F, Qian X, Carpenter R, Li Q, Wang XR, Peiper SS, Lu Z, Liu LZ, and Jiang BH. Reactive oxygen species regulate ERBB2 and ERBB3 expression via miR-199a/125b and DNA methylation. EMBO Rep 13: 1116–1122, 2012
    1. Hidalgo I. and Gonzalez S. New epigenetic pathway for stemness maintenance mediated by the histone methyltransferase Ezh1. Cell Cycle 12: 383–384, 2013
    1. Hilenski LL, Clempus RE, Quinn MT, Lambeth JD, and Griendling KK. Distinct subcellular localizations of Nox1 and Nox4 in vascular smooth muscle cells. Arterioscler Thromb Vasc Biol 24: 677–683, 2004
    1. Hochmuth CE, Biteau B, Bohmann D, and Jasper H. Redox regulation by Keap1 and Nrf2 controls intestinal stem cell proliferation in Drosophila. Cell Stem Cell 8: 188–199, 2011
    1. Iino K, Iwase M, Sonoki K, Yoshinari M, and Iida M. Combination treatment of vitamin C and desferrioxamine suppresses glomerular superoxide and prostaglandin E production in diabetic rats. Diabetes Obes Metab 7: 106–109, 2005
    1. Ishikawa ET. and Cancelas JA. Lack of communication rusts and ages stem cells. Cell Cycle 11: 3149–3150, 2012
    1. Ito K, Hirao A, Arai F, Matsuoka S, Takubo K, Hamaguchi I, Nomiyama K, Hosokawa K, Sakurada K, Nakagata N, Ikeda Y, Mak TW, and Suda T. Regulation of oxidative stress by ATM is required for self-renewal of haematopoietic stem cells. Nature 431: 997–1002, 2004
    1. Ito K, Hirao A, Arai F, Takubo K, Matsuoka S, Miyamoto K, Ohmura M, Naka K, Hosokawa K, Ikeda Y, and Suda T. Reactive oxygen species act through p38 MAPK to limit the lifespan of hematopoietic stem cells. Nat Med 12: 446–451, 2006
    1. Jang YY. and Sharkis SJ. A low level of reactive oxygen species selects for primitive hematopoietic stem cells that may reside in the low-oxygenic niche. Blood 110: 3056–3063, 2007
    1. Juarez JG, Harun N, Thien M, Welschinger R, Baraz R, Pena AD, Pitson SM, Rettig M, DiPersio JF, Bradstock KF, and Bendall LJ. Sphingosine-1-phosphate facilitates trafficking of hematopoietic stem cells and their mobilization by CXCR4 antagonists in mice. Blood 119: 707–716, 2012
    1. Juntilla MM, Patil VD, Calamito M, Joshi RP, Birnbaum MJ, and Koretzky GA. AKT1 and AKT2 maintain hematopoietic stem cell function by regulating reactive oxygen species. Blood 115: 4030–4038, 2010
    1. Kalyanaraman B, Darley-Usmar V, Davies KJ, Dennery PA, Forman HJ, Grisham MB, Mann GE, Moore K, Roberts LJ, 2nd, and Ischiropoulos H. Measuring reactive oxygen and nitrogen species with fluorescent probes: challenges and limitations. Free Radic Biol Med 52: 1–6, 2012
    1. Katare R, Oikawa A, Cesselli D, Beltrami AP, Avolio E, Muthukrishnan D, Munasinghe PE, Angelini G, Emanueli C, and Madeddu P. Boosting the pentose phosphate pathway restores cardiac progenitor cell availability in diabetes. Cardiovasc Res 97: 55–65, 2013
    1. Kharas MG, Okabe R, Ganis JJ, Gozo M, Khandan T, Paktinat M, Gilliland DG, and Gritsman K. Constitutively active AKT depletes hematopoietic stem cells and induces leukemia in mice. Blood 115: 1406–1415, 2010
    1. Kiel MJ, Acar M, Radice GL, and Morrison SJ. Hematopoietic stem cells do not depend on N-cadherin to regulate their maintenance. Cell Stem Cell 4: 170–179, 2009
    1. Kiel MJ, Radice GL, and Morrison SJ. Lack of evidence that hematopoietic stem cells depend on N-cadherin-mediated adhesion to osteoblasts for their maintenance. Cell Stem Cell 1: 204–217, 2007
    1. Kobayashi H, Butler JM, O'Donnell R, Kobayashi M, Ding BS, Bonner B, Chiu VK, Nolan DJ, Shido K, Benjamin L, and Rafii S. Angiocrine factors from Akt-activated endothelial cells balance self-renewal and differentiation of haematopoietic stem cells. Nat Cell Biol 12: 1046–1056, 2010
    1. Kops GJ, Dansen TB, Polderman PE, Saarloos I, Wirtz KW, Coffer PJ, Huang TT, Bos JL, Medema RH, and Burgering BM. Forkhead transcription factor FOXO3a protects quiescent cells from oxidative stress. Nature 419: 316–321, 2002
    1. Kubo M, Li TS, Suzuki R, Ohshima M, Qin SL, and Hamano K. Short-term pretreatment with low-dose hydrogen peroxide enhances the efficacy of bone marrow cells for therapeutic angiogenesis. Am J Physiol Heart Circ Physiol 292: H2582–H2588, 2007
    1. Lassegue B. and Clempus RE. Vascular NAD(P)H oxidases: specific features, expression, and regulation. Am J Physiol Regul Integr Comp Physiol 285: R277–R297, 2003
    1. Lei H. and Quelle FW. FOXO transcription factors enforce cell cycle checkpoints and promote survival of hematopoietic cells after DNA damage. Mol Cancer Res 7: 1294–1303, 2009
    1. Li H. and Forstermann U. Uncoupling of endothelial NO synthase in atherosclerosis and vascular disease. Curr Opin Pharmacol 13: 161–167, 2013
    1. Li TS. and Marban E. Physiological levels of reactive oxygen species are required to maintain genomic stability in stem cells. Stem Cells 28: 1178–1185, 2010
    1. Lin J, Bierhaus A, Bugert P, Dietrich N, Feng Y, Vom Hagen F, Nawroth P, Brownlee M, and Hammes HP. Effect of R-(+)-alpha-lipoic acid on experimental diabetic retinopathy. Diabetologia 49: 1089–1096, 2006
    1. Liu J, Cao L, Chen J, Song S, Lee IH, Quijano C, Liu H, Keyvanfar K, Chen H, Cao LY, Ahn BH, Kumar NG, Rovira II, Xu XL, van Lohuizen M, Motoyama N, Deng CX, and Finkel T. Bmi1 regulates mitochondrial function and the DNA damage response pathway. Nature 459: 387–392, 2009
    1. Liu J, van Mil A, Vrijsen K, Zhao J, Gao L, Metz CH, Goumans MJ, Doevendans PA, and Sluijter JP. MicroRNA-155 prevents necrotic cell death in human cardiomyocyte progenitor cells via targeting RIP1. J Cell Mol Med 15: 1474–1482, 2011
    1. Loomans CJ, de Koning EJ, Staal FJ, Rookmaaker MB, Verseyden C, de Boer HC, Verhaar MC, Braam B, Rabelink TJ, and van Zonneveld AJ. Endothelial progenitor cell dysfunction: a novel concept in the pathogenesis of vascular complications of type 1 diabetes. Diabetes 53: 195–199, 2004
    1. Mangialardi G, Katare R, Oikawa A, Meloni M, Reni C, Emanueli C, and Madeddu P. Diabetes causes bone marrow endothelial barrier dysfunction by activation of the rhoa-rho-associated kinase signaling pathway. Arterioscler Thromb Vasc Biol 33: 555–564, 2013
    1. Mangialardi G, Monopoli A, Ongini E, Spinetti G, Fortunato O, Emanueli C, and Madeddu P. Nitric oxide-donating statin improves multiple functions of circulating angiogenic cells. Br J Pharmacol 164: 570–583, 2011
    1. Masaki S, Ohtsuka R, Abe Y, Muta K, and Umemura T. Expression patterns of microRNAs 155 and 451 during normal human erythropoiesis. Biochem Biophys Res Commun 364: 509–514, 2007
    1. Mendez-Ferrer S, Michurina TV, Ferraro F, Mazloom AR, Macarthur BD, Lira SA, Scadden DT, Ma'ayan A, Enikolopov GN, and Frenette PS. Mesenchymal and haematopoietic stem cells form a unique bone marrow niche. Nature 466: 829–834, 2010
    1. Miyamoto K, Araki KY, Naka K, Arai F, Takubo K, Yamazaki S, Matsuoka S, Miyamoto T, Ito K, Ohmura M, Chen C, Hosokawa K, Nakauchi H, Nakayama K, Nakayama KI, Harada M, Motoyama N, Suda T, and Hirao A. Foxo3a is essential for maintenance of the hematopoietic stem cell pool. Cell Stem Cell 1: 101–112, 2007
    1. Muller-Sieburg CE, Sieburg HB, Bernitz JM, and Cattarossi G. Stem cell heterogeneity: implications for aging and regenerative medicine. Blood 119: 3900–3907, 2012
    1. Naka K, Muraguchi T, Hoshii T, and Hirao A. Regulation of reactive oxygen species and genomic stability in hematopoietic stem cells. Antioxid Redox Signal 10: 1883–1894, 2008
    1. Nance DM. and Sanders VM. Autonomic innervation and regulation of the immune system (1987–2007). Brain Behav Immun 21: 736–745, 2007
    1. Naveiras O, Nardi V, Wenzel PL, Hauschka PV, Fahey F, and Daley GQ. Bone-marrow adipocytes as negative regulators of the haematopoietic microenvironment. Nature 460: 259–263, 2009
    1. Ohshima M, Li TS, Kubo M, Qin SL, and Hamano K. Antioxidant therapy attenuates diabetes-related impairment of bone marrow stem cells. Circ J 73: 162–166, 2009
    1. Oikawa A, Siragusa M, Quaini F, Mangialardi G, Katare RG, Caporali A, van Buul JD, van Alphen FP, Graiani G, Spinetti G, Kraenkel N, Prezioso L, Emanueli C, and Madeddu P. Diabetes mellitus induces bone marrow microangiopathy. Arterioscler Thromb Vasc Biol 30: 498–508, 2010
    1. Orlandi A, Chavakis E, Seeger F, Tjwa M, Zeiher AM, and 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 105: 703–712, 2010
    1. Parmar K, Mauch P, Vergilio JA, Sackstein R, and Down JD. Distribution of hematopoietic stem cells in the bone marrow according to regional hypoxia. Proc Natl Acad Sci U S A 104: 5431–5436, 2007
    1. Piccoli C, Ria R, Scrima R, Cela O, D'Aprile A, Boffoli D, Falzetti F, Tabilio A, and Capitanio N. Characterization of mitochondrial and extra-mitochondrial oxygen consuming reactions in human hematopoietic stem cells. Novel evidence of the occurrence of NAD(P)H oxidase activity. J Biol Chem 280: 26467–26476, 2005
    1. Raaijmakers MH. and Scadden DT. Evolving concepts on the microenvironmental niche for hematopoietic stem cells. Curr Opin Hematol 15: 301–306, 2008
    1. Saito H, Yamamoto Y, and Yamamoto H. Diabetes alters subsets of endothelial progenitor cells that reside in blood, bone marrow, and spleen. Am J Physiol Cell Physiol 302: C892–C901, 2012
    1. Sattler M, Winkler T, Verma S, Byrne CH, Shrikhande G, Salgia R, and Griffin JD. Hematopoietic growth factors signal through the formation of reactive oxygen species. Blood 93: 2928–2935, 1999
    1. Scadden DT. The stem-cell niche as an entity of action. Nature 441: 1075–1079, 2006
    1. Scadden DT. Circadian rhythms: stem cells traffic in time. Nature 452: 416–417, 2008
    1. Schroder K, Kohnen A, Aicher A, Liehn EA, Buchse T, Stein S, Weber C, Dimmeler S, and Brandes RP. NADPH oxidase Nox2 is required for hypoxia-induced mobilization of endothelial progenitor cells. Circ Res 105: 537–544, 2009
    1. Schroder K, Schutz S, Schloffel I, Batz S, Takac I, Weissmann N, Michaelis UR, Koyanagi M, and Brandes RP. Hepatocyte growth factor induces a proangiogenic phenotype and mobilizes endothelial progenitor cells by activating Nox2. Antioxid Redox Signal 15: 915–923, 2011
    1. Spindler V, Schlegel N, and Waschke J. Role of GTPases in control of microvascular permeability. Cardiovasc Res 87: 243–253, 2010
    1. Spinetti G, Cordella D, Fortunato O, Sangalli E, Losa S, Gotti A, Carnelli F, Rosa F, Riboldi S, Sessa F, Avolio E, Beltrami AP, Emanueli C, and Madeddu P. Global remodeling of the vascular stem cell niche in bone marrow of diabetic patients: implication of the microRNA-155/FOXO3a signaling pathway. Circ Res 112: 510–522, 2013
    1. Spinetti G, Fortunato O, Caporali A, Shantikumar S, Marchetti M, Meloni M, Descamps B, Floris I, Sangalli E, Vono R, Faglia E, Specchia C, Pintus G, Madeddu P, and Emanueli C. MicroRNA-15a and microRNA-16 impair human circulating proangiogenic cell functions and are increased in the proangiogenic cells and serum of patients with critical limb ischemia. Circ Res 112: 335–346, 2013
    1. Spradling A, Drummond-Barbosa D, and Kai T. Stem cells find their niche. Nature 414: 98–104, 2001
    1. Stahl M, Dijkers PF, Kops GJ, Lens SM, Coffer PJ, Burgering BM, and Medema RH. The forkhead transcription factor FoxO regulates transcription of p27Kip1 and Bim in response to IL-2. J Immunol 168: 5024–5031, 2002
    1. Storz P. Forkhead homeobox type O transcription factors in the responses to oxidative stress. Antioxid Redox Signal 14: 593–605, 2011
    1. Takubo K, Ohmura M, Azuma M, Nagamatsu G, Yamada W, Arai F, Hirao A, and Suda T. Stem cell defects in ATM-deficient undifferentiated spermatogonia through DNA damage-induced cell-cycle arrest. Cell Stem Cell 2: 170–182, 2008
    1. Takubo K. and Suda T. Roles of the hypoxia response system in hematopoietic and leukemic stem cells. Int J Hematol 95: 478–483, 2012
    1. Taniguchi Ishikawa E, Gonzalez-Nieto D, Ghiaur G, Dunn SK, Ficker AM, Murali B, Madhu M, Gutstein DE, Fishman GI, Barrio LC, and Cancelas JA. Connexin-43 prevents hematopoietic stem cell senescence through transfer of reactive oxygen species to bone marrow stromal cells. Proc Natl Acad Sci U S A 109: 9071–9076, 2012
    1. Tesio M, Golan K, Corso S, Giordano S, Schajnovitz A, Vagima Y, Shivtiel S, Kalinkovich A, Caione L, Gammaitoni L, Laurenti E, Buss EC, Shezen E, Itkin T, Kollet O, Petit I, Trumpp A, Christensen J, Aglietta M, Piacibello W, and Lapidot T. Enhanced c-Met activity promotes G-CSF-induced mobilization of hematopoietic progenitor cells via ROS signaling. Blood 117: 419–428, 2011
    1. Thallas-Bonke V, Thorpe SR, Coughlan MT, Fukami K, Yap FY, Sourris KC, Penfold SA, Bach LA, Cooper ME, and Forbes JM. Inhibition of NADPH oxidase prevents advanced glycation end product-mediated damage in diabetic nephropathy through a protein kinase C-alpha-dependent pathway. Diabetes 57: 460–469, 2008
    1. Thum T, Fraccarollo D, Schultheiss M, Froese S, Galuppo P, Widder JD, Tsikas D, Ertl G, and Bauersachs J. Endothelial nitric oxide synthase uncoupling impairs endothelial progenitor cell mobilization and function in diabetes. Diabetes 56: 666–674, 2007
    1. Tojo T, Ushio-Fukai M, Yamaoka-Tojo M, Ikeda S, Patrushev N, and Alexander RW. Role of gp91phox (Nox2)-containing NAD(P)H oxidase in angiogenesis in response to hindlimb ischemia. Circulation 111: 2347–2355, 2005
    1. Tothova Z. and Gilliland DG. FoxO transcription factors and stem cell homeostasis: insights from the hematopoietic system. Cell Stem Cell 1: 140–152, 2007
    1. Tothova Z, Kollipara R, Huntly BJ, Lee BH, Castrillon DH, Cullen DE, McDowell EP, Lazo-Kallanian S, Williams IR, Sears C, Armstrong SA, Passegue E, DePinho RA, and Gilliland DG. FoxOs are critical mediators of hematopoietic stem cell resistance to physiologic oxidative stress. Cell 128: 325–339, 2007
    1. Touyz RM, Chen X, Tabet F, Yao G, He G, Quinn MT, Pagano PJ, and Schiffrin EL. Expression of a functionally active gp91phox-containing neutrophil-type NAD(P)H oxidase in smooth muscle cells from human resistance arteries: regulation by angiotensin II. Circ Res 90: 1205–1213, 2002
    1. Trumpower BL. The protonmotive Q cycle. Energy transduction by coupling of proton translocation to electron transfer by the cytochrome bc1 complex. J Biol Chem 265: 11409–11412, 1990
    1. Tsai JJ, Dudakov JA, Takahashi K, Shieh JH, Velardi E, Holland AM, Singer NV, West ML, Smith OM, Young LF, Shono Y, Ghosh A, Hanash AM, Tran HT, Moore MA, and van den Brink MR. Nrf2 regulates haematopoietic stem cell function. Nat Cell Biol 15: 309–316, 2013
    1. Turgeon J, Haddad P, Dussault S, Groleau J, Maingrette F, Perez G, and Rivard A. Protection against vascular aging in Nox2-deficient mice: impact on endothelial progenitor cells and reparative neovascularization. Atherosclerosis 223: 122–129, 2012
    1. Urao N, Inomata H, Razvi M, Kim HW, Wary K, McKinney R, Fukai T, and Ushio-Fukai M. Role of nox2-based NADPH oxidase in bone marrow and progenitor cell function involved in neovascularization induced by hindlimb ischemia. Circ Res 103: 212–220, 2008
    1. van der Heijden M, Versteilen AM, Sipkema P, van Nieuw Amerongen GP, Musters RJ, and Groeneveld AB. Rho-kinase-dependent F-actin rearrangement is involved in the inhibition of PI3-kinase/Akt during ischemia-reperfusion-induced endothelial cell apoptosis. Apoptosis 13: 404–412, 2008
    1. Vanella L, Sanford C, Jr., Kim DH, Abraham NG, and Ebraheim N. Oxidative stress and heme oxygenase-1 regulated human mesenchymal stem cells differentiation. Int J Hypertens 2012: 890671, 2012
    1. Wang L, Li Q, Du J, Chen B, Huang X, Guo X, and Huang Q. Advanced glycation end products induce moesin phosphorylation in murine retinal endothelium. Acta Diabetol 49: 47–55, 2012
    1. Westerweel PE, Teraa M, Rafii S, Jaspers JE, White IA, Hooper AT, Doevendans PA, and Verhaar MC. Impaired endothelial progenitor cell mobilization and dysfunctional bone marrow stroma in diabetes mellitus. PLoS One 8: e60357, 2013
    1. Wiesen JL. and Tomasi TB. Dicer is regulated by cellular stresses and interferons. Mol Immunol 46: 1222–1228, 2009
    1. Winkler IG, Sims NA, Pettit AR, Barbier V, Nowlan B, Helwani F, Poulton IJ, van Rooijen N, Alexander KA, Raggatt LJ, and Levesque JP. Bone marrow macrophages maintain hematopoietic stem cell (HSC) niches and their depletion mobilizes HSCs. Blood 116: 4815–4828, 2010
    1. Witkowski S. and Hagberg JM. Progenitor cells and age: can we fight aging with exercise? J Appl Physiol 102: 834–835, 2007
    1. Wojakowski W, Landmesser U, Bachowski R, Jadczyk T, and Tendera M. Mobilization of stem and progenitor cells in cardiovascular diseases. Leukemia 26: 23–33, 2012
    1. Xu Y, Morse LR, da Silva RA, Odgren PR, Sasaki H, Stashenko P, and Battaglino RA. PAMM: a redox regulatory protein that modulates osteoclast differentiation. Antioxid Redox Signal 13: 27–37, 2010
    1. Yamamoto M, Kondo E, Takeuchi M, Harashima A, Otani T, Tsuji-Takayama K, Yamasaki F, Kumon H, Kibata M, and Nakamura S. miR-155, a modulator of FOXO3a protein expression, is underexpressed and cannot be upregulated by stimulation of HOZOT, a line of multifunctional Treg. PLoS One 6: e16841, 2011
    1. Yang YH, Wang Y, Lam KS, Yau MH, Cheng KK, Zhang J, Zhu W, Wu D, and Xu A. Suppression of the Raf/MEK/ERK signaling cascade and inhibition of angiogenesis by the carboxyl terminus of angiopoietin-like protein 4. Arterioscler Thromb Vasc Biol 28: 835–840, 2008
    1. Yao D. and Brownlee M. Hyperglycemia-induced reactive oxygen species increase expression of the receptor for advanced glycation end products (RAGE) and RAGE ligands. Diabetes 59: 249–255, 2010
    1. Yilmaz OH, Valdez R, Theisen BK, Guo W, Ferguson DO, Wu H, and Morrison SJ. Pten dependence distinguishes haematopoietic stem cells from leukaemia-initiating cells. Nature 441: 475–482, 2006
    1. Zherebitskaya E, Akude E, Smith DR, and Fernyhough P. Development of selective axonopathy in adult sensory neurons isolated from diabetic rats: role of glucose-induced oxidative stress. Diabetes 58: 1356–1364, 2009

Source: PubMed

3
Tilaa