Advances in Biological Regulation xxx (2014) 1e11

Contents lists available at ScienceDirect

Advances in Biological Regulation journal homepage: www.elsevier.com/locate/jbior

Role of formyl peptide receptor 2 in homing of endothelial progenitor cells and therapeutic angiogenesis Il Ho Jang a, Soon Chul Heo a, Yang Woo Kwon a, Eun Jung Choi a, Jae Ho Kim a, b, * a

Department of Physiology, School of Medicine, Pusan National University, Yangsan 626-870, Republic of Korea b Research Institute of Convergence Biomedical Science and Technology, Pusan National University Yangsan Hospital, Yangsan 626-770, Gyeongsangnam-do, Republic of Korea

a b s t r a c t Keywords: Endothelial progenitor cells Formyl peptide receptor 2 Homing Angiogenesis

Endothelial progenitor cells (EPCs) hold a great promise as a therapeutic mediator in treatment of ischemic disease conditions. The discovery of EPCs in adult blood has been a cause of significant enthusiasm in the field of endothelial cell research and numerous clinical trials have been expedited. After more than a decade of research in basic science and clinical applications, limitations and new strategies of EPC therapeutics have emerged. With various phenotypes, vague definitions, and uncertain distinction from hematopoietic cells, understanding EPC biology remains challenging. However, EPCs, still hold great hope for treatment of critical ischemic injury as low concern regarding safety can accelerate the clinical applications from basic findings. This review provides an introduction to EPC as cellular therapeutics, which highlights a recent finding that EPC homing was promoted through FPR2 signaling. © 2014 Elsevier Ltd. All rights reserved.

* Corresponding author. Department of Physiology, School of Medicine, Pusan National University, Yangsan 626-870, Gyeongsangnam-do, Republic of Korea. Tel.: þ82 51 510 8073; fax: þ82 51 510 8076. E-mail address: [email protected] (J.H. Kim).

http://dx.doi.org/10.1016/j.jbior.2014.09.011 2212-4926/© 2014 Elsevier Ltd. All rights reserved.

Please cite this article in press as: Jang IH, et al., Role of formyl peptide receptor 2 in homing of endothelial progenitor cells and therapeutic angiogenesis, Advances in Biological Regulation (2014), http:// dx.doi.org/10.1016/j.jbior.2014.09.011

2

I.H. Jang et al. / Advances in Biological Regulation xxx (2014) 1e11

Introduction Endothelial progenitor cells (EPCs) are immature cells with a capacity for proliferation, migration, and differentiation into endothelial cells (ECs) (Ribatti, 2007). Unlike ECs, EPCs show clonal expansion and stemness characteristics, including high proliferation and resistance to stress, and lack mature EC markers (Dernbach et al., 2004; He et al., 2004; Urbich and Dimmeler, 2004). Discovery of EPCs in the adult circulation led to a new concept that vasculogenesis may occur in postnatal life as angiogenesis was previously regarded as the sole mechanism of vascular growth after embryonic development (Asahara et al., 1997). Historically, detection of angioblastic activity from circulating cells dates back to 1932, as Hueper and Russel observed capillary-like formation in cultures of leukocytes (Hueper and Russell, 1932). In 1994, suspended polytetrafluoroethylene pledget retrieved from the aorta of dogs were covered with ECs and capillary-like structures (Scott et al., 1994). In 1997, Asahara et al. isolated EPCs from human peripheral blood using the surface antigen CD34 or VEGFR-2 (KDR) and showed the incorporation of EPCs into sites of active angiogenesis after transplantation into ischemic animal models (Asahara et al., 1997). Since then, various types of EPCs have been reported from different tissues using distinct markers, however, there is still no clear and unambiguous definition of EPC and EPC phenotype (Alev et al., 2011; Asahara et al., 2011). EPCs based on source tissue EPCs can be isolated from the circulation and solid tissue. EPCs in the adult circulation are believed to originate from bone marrow (BM) and the frequency of EPCs in the circulation increases with a mobilization stimulant such as granulocyte colony stimulating factor (G-CSF) (Kawamoto et al., 2009). EPCs can be harvested directly from BM or umbilical cord blood (UCB) (Ingram et al., 2004). In contrast to circulating EPCs, which are floating and nonadhesive, tissue EPCs are adhesive and are believed to originate from circulating EPCs or tissue-resident stem cells (Beltrami et al., 2003; Ii et al., 2009; Kovacic and Boehm, 2009). Endothelial colony-forming cells (ECFCs), also termed endothelial outgrowth cells (EOCs), are adherent cells generated from the culture of circulating EPCs and are regarded as homed-down tissue-resident EPCs (Critser and Yoder, 2010; Ingram et al., 2004). However, ECFCs can also be derived from primary culture of human umbilical vein and adult aortic endothelial cells (Ingram et al., 2005; Yoder, 2010), thus the exact origin of ECFC remains ambiguous. The cellular origins of circulating and tissue-resident EPCs have also not been identified. EPCs based on culture method Putative human EPCs in culture have been assigned different names and show different characteristics and function depending on the culture methods (Basile and Yoder, 2014). Circulating angiogenic cells (CACs) are derived from culture of human peripheral blood mononuclear cells (hPBMNCs) on a fibronectin-coated plate with endothelial differentiation media containing various growth factors such as vascular endothelial growth factor (VEGF), fibroblast growth factor 2 (FGF2), insulin-like growth factor I (IGFI), and epidermal growth factor (EGF) (Kalka et al., 2000; Rehman et al., 2003). After 4 days of culture, non-adherent cells are washed off and persisting adherent cells show characteristics of EPC such as taking up acetylated low-density lipoprotein (ac-LDL) and binding to Ulex europaeus agglgutinin-I lectin (UEA-I). CACs, however, exhibit activities of contaminating platelets and monocytes and are thus regarded as an inaccurate method for identification of EPCs (Prokopi et al., 2009; Rohde et al., 2011). Heterogeneity in the culture can be reduced by maintaining EPCs through colony forming assays. Colony forming unit-Hill (CFU-Hill) cells are generated by culturing adherent hPBMNCs on fibronectin-coated plates at low-density, in which clusters of spindle-shaped cells emerge in 4e9 days (Hill et al., 2003). CFU-Hill cells show expression of surface antigens, similar to ECs, and the gene expression is distinct from that of ECs but indistinguishable from that of CACs (Ahrens et al., 2011; Desai et al., 2009; Medina et al., 2010b). However, CFU-Hill cells, like CACs, exhibit low proliferation potential and do not possess replating potential or contribute to in vivo de novo vessel formation, suggesting that EPCs cannot be identified using CFU-Hill assay (Hirschi et al., 2008; Yoder et al., 2007). ECFCs are derived from the culture of hPBMNCs or UCB MNCs on type I collagen-coated plate (Ingram Please cite this article in press as: Jang IH, et al., Role of formyl peptide receptor 2 in homing of endothelial progenitor cells and therapeutic angiogenesis, Advances in Biological Regulation (2014), http:// dx.doi.org/10.1016/j.jbior.2014.09.011

I.H. Jang et al. / Advances in Biological Regulation xxx (2014) 1e11

3

et al., 2004). Adherent and cobblestone-looking colonies appear in 5 or 22 days of culture and the emerging colonies can be replated and expanded. Clonal analysis of the emerging colonies showed two different types of cells; endothelial cell colony-forming units (CFU-ECs), which show low proliferative potential and do not form vessels in vivo, and ECFCs, which show high proliferative potential and form blood vessels when transplanted in immunodeficient mice (Yoder et al., 2007). ECFCs exhibit high telomerase activity and robust replating potential, and infusion of ECFCs into various ischemia animal models such as hindlimb ischemia (Yoon et al., 2005), myocardial infarction (Dubois et al., 2010), and retinal ischemia (Medina et al., 2010a) enhances vascular recovery with incorporation of ECFCs into newly-formed vessels. Thus, ECFCs present many features of EPCs by definition. However, ECFCs appear to belong to a substantially-differentiated stage as ECFCs lack immature stem/progenitor cell-related marker expression and the cellular origin of ECFCs, whether vessel walls or non-hematopoietic BM cells, remains ambiguous (Alev et al., 2011; Yoder, 2010). Recently developed EPC colony forming assay (EPC-CFA) using semisolid medium and CD34þ or CD133þ cells from UB MNCs, hPBMNCs, or BM MNCs can identify two different types of colonies; primitive EPCs with small cells and highly-proliferative property and definitive EPCs with large cells with a vasculogenic property with developmental hierarchy from primitive EPCs to definitive EPCs and eventually to ECs (Masuda et al., 2011). In addition, EPC-CFA indicates that a single CD34þ or CD133þ cell can develop into EPCs and hematopoietic cells and suggests that the origin of EPC can be hematopoietic stem cells (HSCs). Characteristics of human EPCs according to different culture methods are summarized in Table 1. In identification of putative murine EPCs, the surface markers used are noticeably different from those used in identification of human ones. The applications of surface markers, such as Sca-1, c-Kit, Flk-1, Flt-1, CD31, Tie-2, CXCR4, CD133, or CD144, in the pursuit of murine EPCs are highly variable among individual studies (Asahara et al., 1999; Gallagher et al., 2007; Heeschen et al., 2003; Iwakura et al., 2003; Khakoo and Finkel, 2005; Krishnamurthy et al., 2011; Lyden et al., 2001; Nakajima et al., 2006; Patschan et al., 2006; Thal et al., 2012; Wang et al., 2006; Westerweel et al., 2013). Murine CACs can be identified by culturing PBMNC or BM cells on fibronectin-coated plates, however, these cells, like human CACs, are mostly pro-angiogenic myeloid cells (Basile and Yoder, 2014; Chen et al., 2012; Coffelt et al., 2010). Murine ECFCs do not appear to be present in the circulating blood at an individual animal level as the detection requires pooling of blood from 4 to 6 mice (Somani et al., 2007). EPC-CFA with murine c-KitþSca-1þlineage BM cells can identify small primitive EPCs and large

Table 1 EPCs according to culture method. Plate coating

Cell source

Culture method

Media

References In vivo tube formation

Fibronectin

PBMNC

(Rehman et al., 2003)

PBMNC

No

(Desai et al., 2009; Hill et al., 2003)

ECFC

PBMNC, CBMNC

EGM-2 with VEGF, bFGF, IGF-1, EGF, 5e20% FBS M199 with 20% FCS; Endocult Liquid Medium kit complete EGM-2

No

CFUeHill Fibronectin

At 4 day, nonadherent cells are washed off, adherent cells are kept for further culture At 2 day, nonadherent cells are transferred for the further culture - > spindle cell colony

Yes

(Ingram et al., 2004; Yoder et al., 2007)

CAC

collagen I

At 24 h, nonadhrerent cells are washed off, adherent cells are kept for further culture - > cobblestone colony EPCeCFA Primaria/semi-solid CD34/133 þ PBMNC, 18 days for one cell to media CBMNC form colony; 10 days for 2nd colony - > Hematopoietic/ Endothelial cells

Yes Methocult SF H4236 with SCF, VEGF, bFGF, EGF, IGF-1, IL-3, Heparin, 305 FBS

(Masuda et al., 2011; Masuda and Asahara, 2013)

Please cite this article in press as: Jang IH, et al., Role of formyl peptide receptor 2 in homing of endothelial progenitor cells and therapeutic angiogenesis, Advances in Biological Regulation (2014), http:// dx.doi.org/10.1016/j.jbior.2014.09.011

4

I.H. Jang et al. / Advances in Biological Regulation xxx (2014) 1e11

definitive EPCs, thus showing consistency between human and mouse (Kwon et al., 2008; Tanaka et al., 2008). However, a hierarchical relationship of primitive EPCs, definitive EPCs, and HSCs remains to be examined in a murine system. Hematopoietic connection Endothelial cells and hematopoietic cells have intimate relationship from the beginning during embryo development as EPCs (angioblasts) and HSCs are believed to originate from the common precursor, hemangioblast, a bi-potential cell that produces endothelial and hematopoietic systems (Choi et al., 1998; Huber et al., 2004; Kennedy et al., 1997). The surface markers used for isolation of EPCs in the adult show significant overlap with HSC markers; CD34, CD133, KDR, CXCR4, and CD105 for human and c-Kit, Sca-1, and CD34 in combination with Flk-1 for mouse (Timmermans et al., 2009). It is interesting to note that recent advancement in hematopoiesis has shown that development of hemangioblasts occurs through formation of hemogenic endothelium, an endothelium that generates hematopoietic cells, and the lineage tracing in mice indicates that most hematopoietic cells including HSCs originate from hemogenic endothelium during embryo development (Chen et al., 2009; Lancrin et al., 2009). Although hemogenic endothelium is believed to be present transiently during embryo development, EPC-CFA has demonstrated the existence of a single cell that generates hematopoietic and endothelial cells in the adult (Jaffredo et al., 2013; Masuda et al., 2011; Masuda and Asahara, 2013). Currently there is still no surface marker combination that can distinguish EPCs from HSCs. Treatment of ischemic injury with endothelial progenitor cells Since the discovery of EPCs, the regenerative potential of EPCs has been widely tested in preclinical and clinical trials for more than a decade (Ribatti, 2007). EPCs have been shown to contribute to neovascularization directly through incorporation into newly formed vessels or indirectly through secretion of angiogenic factors. Transplantation of BM cells from transgenic mice in which LacZ is expressed under the control of Flk-1 or Tie-2 promoter into wild type control mice has shown that LacZ-positive cells are detected in vascular structures in cardiac ischemia, corneal neovascularization, and wound healing models (Bauer et al., 2006; Iwakura et al., 2006; Murayama et al., 2002). In cases where direct vasculogenic contributions of EPCs are not detected, EPCs migrate to the ischemic injury sites and remain in the interstitial tissue and stimulate migration and proliferation of pre-existing ECs by providing paracrine factors such as VEGF, hepatocyte growth factor, angiopoietin-1, stromal cellderived factor-1a, insulin-like growth factor-1, and nitric oxide (Dai et al., 2008; Ii et al., 2005; Jujo et al., 2008; Miyamoto et al., 2007; Urbich et al., 2005). Thus, EPCs participate in tissue protection and regeneration by two different mechanisms. Regenerative potential of human EPCs was first tested in animal models. Intravenous injection of cultured hPBMNC into immunodeficient mice of a hindlimb ischemia model resulted in the restoration of blood flow at the ischemic injury site and salvation of the hindlimb (Kalka et al., 2000). Intravenous injection of cultured hPBMNC into nude rats of myocardial ischemia model resulted in the incorporation of transplanted EPCs into myocardial neovascularization and improved cardiac function (Kawamoto et al., 2001). The therapeutic potential in promoting the recovery from ischemic injury increased substantially by use of CB MNCs and enrichment of EPCs by selection of CD34þ cells (Iwasaki et al., 2006; Kawamoto et al., 2006; Kocher et al., 2001; Murohara et al., 2000). CD133 has also been a frequently-chosen surface marker for enrichment of EPCs in improving vascular recovery (Friedrich et al., 2006; Senegaglia et al., 2008, 2010). However, a detailed understanding of the contribution mechanism, for example the quantification of how much was contributed by direct incorporation through vasculogenesis and how much was contributed by angiogenic paracrine factors and a standardized method for such measurement has not yet emerged. Clinical trials with human EPCs for treatment of ischemic conditions started in the early 21st century, and 225 trials are currently listed in the U.S. National Institutes of Health. According to Asahara et al. clinical trials with human EPCs can be categorized to three generations (Asahara et al., 2011). In the first generation trials, whole MNCs from PB or BM were transplanted and showed relatively lowdegree contributions to angiogenesis and vasculogenesis with complications such as inflammation and Please cite this article in press as: Jang IH, et al., Role of formyl peptide receptor 2 in homing of endothelial progenitor cells and therapeutic angiogenesis, Advances in Biological Regulation (2014), http:// dx.doi.org/10.1016/j.jbior.2014.09.011

I.H. Jang et al. / Advances in Biological Regulation xxx (2014) 1e11

5

fibrosis (Assmus et al., 2006; Dobert et al., 2004; Erbs et al., 2005; Li et al., 2007; Schachinger et al., 2004; Tatsumi et al., 2007) The second generation trials have been conducted with sorted EPCs for CD34 or CD133 expression and have been reported to show better angiogenesis and vasculogenesis with fewer complications (Ahmadi et al., 2007; Bartunek et al., 2005; Boyle et al., 2006; Kawamoto et al., 2009; Lara-Hernandez et al., 2010; Losordo et al., 2007; Stamm et al., 2007, 2003). The third generation trials are comprised of transplanting cultured and expanded CD34 þ or CD133 þ cells and are expected to yield much better outcomes based on animal experiments (Kamei et al., 2013; Kawakami et al., 2012; Masuda et al., 2014; Senegaglia et al., 2010) although evaluations in a clinical setting are necessary. Clinical trials with human EPCs can also be categorized to three groups depending on the method of introducing EPCs (Resch et al., 2012). The least invasive method involves the mobilization of the patient's own BM EPCs into the circulation by administration of G-CSF, expecting the homing of mobilized EPCs to the injury sites (Ohtsuka et al., 2004). Pilot studies for promoting myocardial regeneration showed improvement of left ventricular (LV) function, however, serious side effects, including increased in-stent restenosis and re-infarction, were also observed (Ince et al., 2005; Syed and Beeching, 2005; Tongers and Losordo, 2007; Valgimigli et al., 2005). The more invasive method consists of injection of EPCs into the circulation. Intracoronary infusion of BM EPCs to patients with myocardiac infarction showed positive LV function (Assmus et al., 2002; FernandezAviles et al., 2004; Strauer et al., 2002), followed by two phase II studies, the TOPCARE-AMI and BOOST, which showed a significant increase of LV function 4e6 months after administration of BM EPCs (Leistner et al., 2011; Schachinger et al., 2004; Wollert et al., 2004). Ten major clinical studies confirmed the safety of the procedure and the clinical benefit in more than half of studies, however, the benefits were transient and weak (Wei et al., 2009). The most invasive method involves the direct injection of EPCs into the target tissue. Intramuscular injections of autologous BM EPCs in patients with chronic limb ischemia resulted in immediate improvement and lowered amputation rates in three-year followup assessment (Matoba et al., 2008; Tateishi-Yuyama et al., 2002). Studies with patients of ischemic heart disease showed the improved ventricular function, however, larger studies are required in order to prove the beneficial effects (Hamano et al., 2001; Hattori and Matsubara, 2004; Perin et al., 2003; Tse et al., 2003). Although EPCs have been a subject of accelerated investigation since their discovery with the vision of rapid clinical applications, as more insights are gained through basic research on EPC biology, more demanding views are taking over the fervor of the pioneering period. Calculation based on the accumulated data in animal and human studies suggests that a blood volume of as much as 12 L will be necessary for isolation of EPCs for culture in order to achieve a satisfactory recovery in treatment of critical limb ischemia (Alev et al., 2011; Asahara et al., 2011). Thus, basic research such as selective mobilization of endogenous EPCs, enhancing EPC function with genetic modifications, generating EPCs from alternative sources like ES/iPS cells, and identifying/tracking the development of EPCs during embryogenesis will be necessary in order to bring EPCs to effective clinical applications. EPC homing and FPR2 A number of studies in animal models have reported homing of transplanted EPCs to sites of ischemic injury (Kalka et al., 2000; Kocher et al., 2001; Shi et al., 1998). Although the precise mechanisms of EPC homing are not completely understood, the interaction between surface molecules on EPCs and their ligands upregulated at the injury site is regarded as playing a central role (Avci-Adali et al., 2010). The increased expression of P-selectin glycoprotein ligand-1 (PSGL-1) or Eselectin on the EPC surface enhanced vascular recovery in the ischemic limb after transplanting EPCs (Foubert et al., 2007; Oh et al., 2007). Activation of b2-integrins on the EPC surface for the interaction with ICAM-1 in the ischemic muscle enhanced the homing of EPCs and EPC-induced neovascularization (Chavakis et al., 2005; Yoon et al., 2006). a4b1 integrin (very late antigen-4, VLA-4), which interacts with vascular cell adhesion molecule (VCAM) and fibronectin, also participate in homing of EPCs to ischemic tissue (Duan et al., 2006; Jin et al., 2006). Chemokines, which play important roles in angiogenesis and arteriogenesis, contribute to EPC homing (Schober and Zernecke, 2007; Weber et al., 2004). Chemokines are involved in mobilization, recruitment and adhesion of EPCs through the interaction of angiogenic chemokines such as CCL2, CXCL1, CXCL7, and Please cite this article in press as: Jang IH, et al., Role of formyl peptide receptor 2 in homing of endothelial progenitor cells and therapeutic angiogenesis, Advances in Biological Regulation (2014), http:// dx.doi.org/10.1016/j.jbior.2014.09.011

6

I.H. Jang et al. / Advances in Biological Regulation xxx (2014) 1e11

Fig. 1. WKYMVm promotes homing of EPCs to ischemic injury site through FPR2.

CXCL12 (SDF-1a) at the injury site and their corresponding receptors on the EPC surface such as CCR2, CXCR2, and CXCR4 (Ceradini et al., 2004; Hristov et al., 2007; Weber et al., 1999). The interaction of CCL2/CCR2 and SDF1-a/CXCR4 is also known to direct EPCs to tumor neoangiogenesis (Guleng et al., 2005; Spring et al., 2005). As the efficient homing of EPCs to the site of therapeutic focus is a critical step in the successful treatment of ischemic conditions, further understanding of homing mechanisms of EPCs and development of a new strategy for directing EPC homing will significantly improve the therapeutic potential of EPCs. In a recent report, Heo et al. showed that activation of formyl peptide receptor 2 (FPR2) on EPC by injection of synthetic WKYMVm peptide near the ischemic injury site promoted homing of transplanted human EPCs and enhanced the vascular repair (Heo et al., 2014). FPRs, belonging to the G protein-coupled receptor family, play important roles in host defense and inflammatory regulation through promotion of chemotaxis and activation of leukocytes and phagocytes (Le et al., 2002). EPCs were generated from 7 to 10 day culture of human CB MNCs and shown to express FPR2 and FPR3. WKYMVm, a synthetic peptide identified from a library screen for activating lymphocytes, is involved in chemoattraction of human phagocytes (Bae et al., 1999; Baek et al., 1996). WKYMVm has a higher affinity for FPR2, which induces proliferation, migration, and sprouting activity of endothelial cells (Cattaneo et al., 2013; Lee et al., 2006). Heo et al. transplanted human EPCs by trail vein injection into nude mice with hindlimb ischemia and showed that repeated injection of WKYMVm into ischemic limbs led to increased homing of EPCs to the injury site and enhanced the recovery of vasculature and blood flow (Fig. 1). In addition, knockdown of FPR2 in EPCs significantly decreased the therapeutic effect of WKYMVm. EPCs can be promising cellular therapeutics with relatively low concerns regarding safety issues, however, they still show a limitation in effectiveness. Further understanding of EPC biology and development of new tools for improvement of EPC therapy, such as WKYMVm-FPR2 directed homing, will expedite the arrival of EPC therapeutics to clinical beds. Acknowledgment This work was supported for two years by Pusan National University Research Grant. Please cite this article in press as: Jang IH, et al., Role of formyl peptide receptor 2 in homing of endothelial progenitor cells and therapeutic angiogenesis, Advances in Biological Regulation (2014), http:// dx.doi.org/10.1016/j.jbior.2014.09.011

I.H. Jang et al. / Advances in Biological Regulation xxx (2014) 1e11

7

References Ahmadi H, Baharvand H, Ashtiani SK, Soleimani M, Sadeghian H, Ardekani JM, et al. Safety analysis and improved cardiac function following local autologous transplantation of CD133(þ) enriched bone marrow cells after myocardial infarction. Curr Neurovascular Res 2007;4:153e60. Ahrens I, Domeij H, Topcic D, Haviv I, Merivirta RM, Agrotis A, et al. Successful in vitro expansion and differentiation of cord blood derived CD34þ cells into early endothelial progenitor cells reveals highly differential gene expression. PloS One 2011; 6:e23210. Alev C, Ii M, Asahara T. Endothelial progenitor cells: a novel tool for the therapy of ischemic diseases. Antioxid Redox Signal 2011;15:949e65. Asahara T, Kawamoto A, Masuda H. Concise review: circulating endothelial progenitor cells for vascular medicine. Stem Cells 2011;29:1650e5. Asahara T, Murohara T, Sullivan A, Silver M, van der Zee R, Li T, et al. Isolation of putative progenitor endothelial cells for angiogenesis. Science 1997;275:964e7. Asahara T, Takahashi T, Masuda H, Kalka C, Chen D, Iwaguro H, et al. VEGF contributes to postnatal neovascularization by mobilizing bone marrow-derived endothelial progenitor cells. EMBO J 1999;18:3964e72. Assmus B, Honold J, Schachinger V, Britten MB, Fischer-Rasokat U, Lehmann R, et al. Transcoronary transplantation of progenitor cells after myocardial infarction. N Engl J Med 2006;355:1222e32. Assmus B, Schachinger V, Teupe C, Britten M, Lehmann R, Dobert N, et al. Transplantation of progenitor cells and regeneration enhancement in acute myocardial infarction (TOPCARE-AMI). Circulation 2002;106:3009e17. Avci-Adali M, Ziemer G, Wendel HP. Induction of EPC homing on biofunctionalized vascular grafts for rapid in vivo selfendothelializationea review of current strategies. Biotechnol Adv 2010;28:119e29. Bae YS, Kim Y, Kim Y, Kim JH, Suh PG, Ryu SH. Trp-Lys-Tyr-Met-Val-D-Met is a chemoattractant for human phagocytic cells. J Leukoc Biol 1999;66:915e22. Baek SH, Seo JK, Chae CB, Suh PG, Ryu SH. Identification of the peptides that stimulate the phosphoinositide hydrolysis in lymphocyte cell lines from peptide libraries. J Biol Chem 1996;271:8170e5. Bartunek J, Vanderheyden M, Vandekerckhove B, Mansour S, De Bruyne B, De Bondt P, et al. Intracoronary injection of CD133positive enriched bone marrow progenitor cells promotes cardiac recovery after recent myocardial infarction: feasibility and safety. Circulation 2005;112:I178e83. Basile DP, Yoder MC. Circulating and tissue resident endothelial progenitor cells. J Cell Physiol 2014;229:10e6. Bauer SM, Goldstein LJ, Bauer RJ, Chen H, Putt M, Velazquez OC. The bone marrow-derived endothelial progenitor cell response is impaired in delayed wound healing from ischemia. J Vasc Surg 2006;43:134e41. Beltrami AP, Barlucchi L, Torella D, Baker M, Limana F, Chimenti S, et al. Adult cardiac stem cells are multipotent and support myocardial regeneration. Cell 2003;114:763e76. Boyle AJ, Whitbourn R, Schlicht S, Krum H, Kocher A, Nandurkar H, et al. Intra-coronary high-dose CD34þ stem cells in patients with chronic ischemic heart disease: a 12-month follow-up. Int J Cardiol 2006;109:21e7. Cattaneo F, Parisi M, Ammendola R. Distinct signaling cascades elicited by different formyl Peptide receptor 2 (FPR2) agonists. Int J Mol Sci 2013;14:7193e230. Ceradini DJ, Kulkarni AR, Callaghan MJ, Tepper OM, Bastidas N, Kleinman ME, et al. Progenitor cell trafficking is regulated by hypoxic gradients through HIF-1 induction of SDF-1. Nat Med 2004;10:858e64. Chavakis E, Aicher A, Heeschen C, Sasaki K, Kaiser R, El Makhfi N, et al. Role of beta2-integrins for homing and neovascularization capacity of endothelial progenitor cells. J Exp Med 2005;201:63e72. Chen JY, Feng L, Zhang HL, Li JC, Yang XW, Cao XL, et al. Differential regulation of bone marrow-derived endothelial progenitor cells and endothelial outgrowth cells by the Notch signaling pathway. PloS One 2012;7:e43643. Chen MJ, Yokomizo T, Zeigler BM, Dzierzak E, Speck NA. Runx1 is required for the endothelial to haematopoietic cell transition but not thereafter. Nature 2009;457:887e91. Choi K, Kennedy M, Kazarov A, Papadimitriou JC, Keller G. A common precursor for hematopoietic and endothelial cells. Development 1998;125:725e32. Coffelt SB, Tal AO, Scholz A, De Palma M, Patel S, Urbich C, et al. Angiopoietin-2 regulates gene expression in TIE2-expressing monocytes and augments their inherent proangiogenic functions. Cancer Res 2010;70:5270e80. Critser PJ, Yoder MC. Endothelial colony-forming cell role in neoangiogenesis and tissue repair. Curr Opin Organ Transplant 2010;15:68e72. Dai Y, Ashraf M, Zuo S, Uemura R, Dai YS, Wang Y, et al. Mobilized bone marrow progenitor cells serve as donors of cytoprotective genes for cardiac repair. J Mol Cell Cardiol 2008;44:607e17. Dernbach E, Urbich C, Brandes RP, Hofmann WK, Zeiher AM, Dimmeler S. Antioxidative stress-associated genes in circulating progenitor cells: evidence for enhanced resistance against oxidative stress. Blood 2004;104:3591e7. Desai A, Glaser A, Liu D, Raghavachari N, Blum A, Zalos G, et al. Microarray-based characterization of a colony assay used to investigate endothelial progenitor cells and relevance to endothelial function in humans. Arterioscler, Thromb, Vasc Biol 2009;29:121e7. Dobert N, Britten M, Assmus B, Berner U, Menzel C, Lehmann R, et al. Transplantation of progenitor cells after reperfused acute myocardial infarction: evaluation of perfusion and myocardial viability with FDG-PET and thallium SPECT. Eur J Nucl Med Mol Imaging 2004;31:1146e51. Duan H, Cheng L, Sun X, Wu Y, Hu L, Wang J, et al. LFA-1 and VLA-4 involved in human high proliferative potential-endothelial progenitor cells homing to ischemic tissue. Thromb Haemost 2006;96:807e15. Dubois C, Liu X, Claus P, Marsboom G, Pokreisz P, Vandenwijngaert S, et al. Differential effects of progenitor cell populations on left ventricular remodeling and myocardial neovascularization after myocardial infarction. J Am Coll Cardiol 2010;55: 2232e43. Erbs S, Linke A, Adams V, Lenk K, Thiele H, Diederich KW, et al. Transplantation of blood-derived progenitor cells after recanalization of chronic coronary artery occlusion: first randomized and placebo-controlled study. Circ Res 2005;97: 756e62.

Please cite this article in press as: Jang IH, et al., Role of formyl peptide receptor 2 in homing of endothelial progenitor cells and therapeutic angiogenesis, Advances in Biological Regulation (2014), http:// dx.doi.org/10.1016/j.jbior.2014.09.011

8

I.H. Jang et al. / Advances in Biological Regulation xxx (2014) 1e11

Fernandez-Aviles F, San Roman JA, Garcia-Frade J, Fernandez ME, Penarrubia MJ, de la Fuente L, et al. Experimental and clinical regenerative capability of human bone marrow cells after myocardial infarction. Circ Res 2004;95:742e8. Foubert P, Silvestre JS, Souttou B, Barateau V, Martin C, Ebrahimian TG, et al. PSGL-1-mediated activation of EphB4 increases the proangiogenic potential of endothelial progenitor cells. J Clin Investig 2007;117:1527e37. Friedrich EB, Walenta K, Scharlau J, Nickenig G, Werner N. CD34/CD133þ/VEGFR-2þ endothelial progenitor cell subpopulation with potent vasoregenerative capacities. Circ Res 2006;98:e20e5. Gallagher KA, Liu ZJ, Xiao M, Chen H, Goldstein LJ, Buerk DG, et al. Diabetic impairments in NO-mediated endothelial progenitor cell mobilization and homing are reversed by hyperoxia and SDF-1 alpha. J Clin Investig 2007;117:1249e59. Guleng B, Tateishi K, Ohta M, Kanai F, Jazag A, Ijichi H, et al. Blockade of the stromal cell-derived factor-1/CXCR4 axis attenuates in vivo tumor growth by inhibiting angiogenesis in a vascular endothelial growth factor-independent manner. Cancer Res 2005;65:5864e71. Hamano K, Nishida M, Hirata K, Mikamo A, Li TS, Harada M, et al. Local implantation of autologous bone marrow cells for therapeutic angiogenesis in patients with ischemic heart disease: clinical trial and preliminary results. Jpn Circ J 2001;65: 845e7. Hattori R, Matsubara H. Therapeutic angiogenesis for severe ischemic heart diseases by autologous bone marrow cells transplantation. Mol Cell Biochem 2004;264:151e5. He T, Peterson TE, Holmuhamedov EL, Terzic A, Caplice NM, Oberley LW, et al. Human endothelial progenitor cells tolerate oxidative stress due to intrinsically high expression of manganese superoxide dismutase. Arterioscler, Thromb, Vasc Biol 2004;24:2021e7. Heeschen C, Aicher A, Lehmann R, Fichtlscherer S, Vasa M, Urbich C, et al. Erythropoietin is a potent physiologic stimulus for endothelial progenitor cell mobilization. Blood 2003;102:1340e6. Heo SC, Kwon YW, Jang IH, Jeong GO, Yoon JW, Kim CD, et al. WKYMVm-induced activation of formyl peptide receptor 2 stimulates ischemic neovasculogenesis by promoting homing of endothelial colony-forming cells. Stem cells 2014;32: 779e90. Hill JM, Zalos G, Halcox JP, Schenke WH, Waclawiw MA, Quyyumi AA, et al. Circulating endothelial progenitor cells, vascular function, and cardiovascular risk. N Engl J Med 2003;348:593e600. Hirschi KK, Ingram DA, Yoder MC. Assessing identity, phenotype, and fate of endothelial progenitor cells. Arterioscler, Thromb, Vasc Biol 2008;28:1584e95. Hristov M, Zernecke A, Bidzhekov K, Liehn EA, Shagdarsuren E, Ludwig A, et al. Importance of CXC chemokine receptor 2 in the homing of human peripheral blood endothelial progenitor cells to sites of arterial injury. Circ Res 2007;100:590e7. Huber TL, Kouskoff V, Fehling HJ, Palis J, Keller G. Haemangioblast commitment is initiated in the primitive streak of the mouse embryo. Nature 2004;432:625e30. Hueper WC, Russell MA. Capillary-like formations in tissue cultures of leukocytes. Arch Exp Zellforsch 1932;12:407e24. Ii M, Nishimura H, Iwakura A, Wecker A, Eaton E, Asahara T, et al. Endothelial progenitor cells are rapidly recruited to myocardium and mediate protective effect of ischemic preconditioning via “imported” nitric oxide synthase activity. Circulation 2005;111:1114e20. Ii M, Nishimura H, Sekiguchi H, Kamei N, Yokoyama A, Horii M, et al. Concurrent vasculogenesis and neurogenesis from adult neural stem cells. Circ Res 2009;105:860e8. Ince H, Petzsch M, Kleine HD, Eckard H, Rehders T, Burska D, et al. Prevention of left ventricular remodeling with granulocyte colony-stimulating factor after acute myocardial infarction: final 1-year results of the Front-Integrated Revascularization and Stem Cell Liberation in Evolving Acute Myocardial Infarction by Granulocyte Colony-Stimulating Factor (FIRSTLINEAMI) Trial. Circulation 2005;112:I73e80. Ingram DA, Mead LE, Moore DB, Woodard W, Fenoglio A, Yoder MC. Vessel wall-derived endothelial cells rapidly proliferate because they contain a complete hierarchy of endothelial progenitor cells. Blood 2005;105:2783e6. Ingram DA, Mead LE, Tanaka H, Meade V, Fenoglio A, Mortell K, et al. Identification of a novel hierarchy of endothelial progenitor cells using human peripheral and umbilical cord blood. Blood 2004;104:2752e60. Iwakura A, Luedemann C, Shastry S, Hanley A, Kearney M, Aikawa R, et al. Estrogen-mediated, endothelial nitric oxide synthasedependent mobilization of bone marrow-derived endothelial progenitor cells contributes to reendothelialization after arterial injury. Circulation 2003;108:3115e21. Iwakura A, Shastry S, Luedemann C, Hamada H, Kawamoto A, Kishore R, et al. Estradiol enhances recovery after myocardial infarction by augmenting incorporation of bone marrow-derived endothelial progenitor cells into sites of ischemia-induced neovascularization via endothelial nitric oxide synthase-mediated activation of matrix metalloproteinase-9. Circulation 2006;113:1605e14. Iwasaki H, Kawamoto A, Ishikawa M, Oyamada A, Nakamori S, Nishimura H, et al. Dose-dependent contribution of CD34positive cell transplantation to concurrent vasculogenesis and cardiomyogenesis for functional regenerative recovery after myocardial infarction. Circulation 2006;113:1311e25. Jaffredo T, Lempereur A, Richard C, Bollerot K, Gautier R, Canto PY, et al. Dorso-ventral contributions in the formation of the embryonic aorta and the control of aortic hematopoiesis. Blood cells, Mol Dis 2013;51:232e8. Jin H, Aiyer A, Su J, Borgstrom P, Stupack D, Friedlander M, et al. A homing mechanism for bone marrow-derived progenitor cell recruitment to the neovasculature. J Clin Investig 2006;116:652e62. Jujo K, Ii M, Losordo DW. Endothelial progenitor cells in neovascularization of infarcted myocardium. J Mol Cell Cardiol 2008;45: 530e44. Kalka C, Masuda H, Takahashi T, Kalka-Moll WM, Silver M, Kearney M, et al. Transplantation of ex vivo expanded endothelial progenitor cells for therapeutic neovascularization. Proc Natl Acad Sci USA 2000;97:3422e7. Kamei N, Kwon SM, Alev C, Nakanishi K, Yamada K, Masuda H, et al. Ex-vivo expanded human blood-derived CD133þ cells promote repair of injured spinal cord. J Neurol Sci 2013;328:41e50. Kawakami Y, Ii M, Alev C, Kawamoto A, Matsumoto T, Kuroda R, et al. Local transplantation of ex vivo expanded bone marrowderived CD34-positive cells accelerates fracture healing. Cell Transplant 2012;21:2689e709. Kawamoto A, Gwon HC, Iwaguro H, Yamaguchi JI, Uchida S, Masuda H, et al. Therapeutic potential of ex vivo expanded endothelial progenitor cells for myocardial ischemia. Circulation 2001;103:634e7.

Please cite this article in press as: Jang IH, et al., Role of formyl peptide receptor 2 in homing of endothelial progenitor cells and therapeutic angiogenesis, Advances in Biological Regulation (2014), http:// dx.doi.org/10.1016/j.jbior.2014.09.011

I.H. Jang et al. / Advances in Biological Regulation xxx (2014) 1e11

9

Kawamoto A, Iwasaki H, Kusano K, Murayama T, Oyamada A, Silver M, et al. CD34-positive cells exhibit increased potency and safety for therapeutic neovascularization after myocardial infarction compared with total mononuclear cells. Circulation 2006;114:2163e9. Kawamoto A, Katayama M, Handa N, Kinoshita M, Takano H, Horii M, et al. Intramuscular transplantation of G-CSF-mobilized CD34(þ) cells in patients with critical limb ischemia: a phase I/IIa, multicenter, single-blinded, dose-escalation clinical trial. Stem Cells 2009;27:2857e64. Kennedy M, Firpo M, Choi K, Wall C, Robertson S, Kabrun N, et al. A common precursor for primitive erythropoiesis and definitive haematopoiesis. Nature 1997;386:488e93. Khakoo AY, Finkel T. Endothelial progenitor cells. Annu Rev Med 2005;56:79e101. Kocher AA, Schuster MD, Szabolcs MJ, Takuma S, Burkhoff D, Wang J, et al. Neovascularization of ischemic myocardium by human bone-marrow-derived angioblasts prevents cardiomyocyte apoptosis, reduces remodeling and improves cardiac function. Nat Med 2001;7:430e6. Kovacic JC, Boehm M. Resident vascular progenitor cells: an emerging role for non-terminally differentiated vessel-resident cells in vascular biology. Stem cell Res 2009;2:2e15. Krishnamurthy P, Thal M, Verma S, Hoxha E, Lambers E, Ramirez V, et al. Interleukin-10 deficiency impairs bone marrowderived endothelial progenitor cell survival and function in ischemic myocardium. Circ Res 2011;109:1280e9. Kwon SM, Eguchi M, Wada M, Iwami Y, Hozumi K, Iwaguro H, et al. Specific Jagged-1 signal from bone marrow microenvironment is required for endothelial progenitor cell development for neovascularization. Circulation 2008;118:157e65. Lancrin C, Sroczynska P, Stephenson C, Allen T, Kouskoff V, Lacaud G. The haemangioblast generates haematopoietic cells through a haemogenic endothelium stage. Nature 2009;457:892e5. Lara-Hernandez R, Lozano-Vilardell P, Blanes P, Torreguitart-Mirada N, Galmes A, Besalduch J. Safety and efficacy of therapeutic angiogenesis as a novel treatment in patients with critical limb ischemia. Ann Vasc Surg 2010;24:287e94. Le Y, Murphy PM, Wang JM. Formyl-peptide receptors revisited. Trends Immunol 2002;23:541e8. Lee MS, Yoo SA, Cho CS, Suh PG, Kim WU, Ryu SH. Serum amyloid A binding to formyl peptide receptor-like 1 induces synovial hyperplasia and angiogenesis. J Immunol 2006;177:5585e94. Leistner DM, Fischer-Rasokat U, Honold J, Seeger FH, Schachinger V, Lehmann R, et al. Transplantation of progenitor cells and regeneration enhancement in acute myocardial infarction (TOPCARE-AMI): final 5-year results suggest long-term safety and efficacy. Clin Res Cardiol: Off J Ger Card Soc 2011;100:925e34. Li ZQ, Zhang M, Jing YZ, Zhang WW, Liu Y, Cui LJ, et al. The clinical study of autologous peripheral blood stem cell transplantation by intracoronary infusion in patients with acute myocardial infarction (AMI). Int J Cardiol 2007;115:52e6. Losordo DW, Schatz RA, White CJ, Udelson JE, Veereshwarayya V, Durgin M, et al. Intramyocardial transplantation of autologous CD34þ stem cells for intractable angina: a phase I/IIa double-blind, randomized controlled trial. Circulation 2007;115: 3165e72. Lyden D, Hattori K, Dias S, Costa C, Blaikie P, Butros L, et al. Impaired recruitment of bone-marrow-derived endothelial and hematopoietic precursor cells blocks tumor angiogenesis and growth. Nat Med 2001;7:1194e201. Masuda H, Alev C, Akimaru H, Ito R, Shizuno T, Kobori M, et al. Methodological development of a clonogenic assay to determine endothelial progenitor cell potential. Circ Res 2011;109:20e37. Masuda H, Asahara T. Clonogenic assay of endothelial progenitor cells. Trends Cardiovasc Med 2013;23:99e103. Masuda H, Tanaka R, Fujimura S, Ishikawa M, Akimaru H, Shizuno T, et al. Vasculogenic conditioning of peripheral blood mononuclear cells promotes endothelial progenitor cell expansion and phenotype transition of anti-inflammatory macrophage and T lymphocyte to cells with regenerative potential. J Am Heart Assoc 2014;3:e000743. Matoba S, Tatsumi T, Murohara T, Imaizumi T, Katsuda Y, Ito M, et al. Long-term clinical outcome after intramuscular implantation of bone marrow mononuclear cells (Therapeutic Angiogenesis by Cell Transplantation [TACT] trial) in patients with chronic limb ischemia. Am Heart J 2008;156:1010e8. Medina RJ, O'Neill CL, Humphreys MW, Gardiner TA, Stitt AW. Outgrowth endothelial cells: characterization and their potential for reversing ischemic retinopathy. Investig Ophthalmol Vis Sci 2010a;51:5906e13. Medina RJ, O'Neill CL, Sweeney M, Guduric-Fuchs J, Gardiner TA, Simpson DA, et al. Molecular analysis of endothelial progenitor cell (EPC) subtypes reveals two distinct cell populations with different identities. BMC Med Genomics 2010b;3:18. Miyamoto Y, Suyama T, Yashita T, Akimaru H, Kurata H. Bone marrow subpopulations contain distinct types of endothelial progenitor cells and angiogenic cytokine-producing cells. J Mol Cell Cardiol 2007;43:627e35. Murayama T, Tepper OM, Silver M, Ma H, Losordo DW, Isner JM, et al. Determination of bone marrow-derived endothelial progenitor cell significance in angiogenic growth factor-induced neovascularization in vivo. Exp Hematol 2002;30:967e72. Murohara T, Ikeda H, Duan J, Shintani S, Sasaki K, Eguchi H, et al. Transplanted cord blood-derived endothelial precursor cells augment postnatal neovascularization. J Clin Investig 2000;105:1527e36. Nakajima M, Ogawa M, Shimoda Y, Hiraoka S, Iida M, Koseki H, et al. Presenilin-1 controls the growth and differentiation of endothelial progenitor cells through its beta-catenin-binding region. Cell Biol Int 2006;30:239e43. Oh IY, Yoon CH, Hur J, Kim JH, Kim TY, Lee CS, et al. Involvement of E-selectin in recruitment of endothelial progenitor cells and angiogenesis in ischemic muscle. Blood 2007;110:3891e9. Ohtsuka M, Takano H, Zou Y, Toko H, Akazawa H, Qin Y, et al. Cytokine therapy prevents left ventricular remodeling and dysfunction after myocardial infarction through neovascularization. FASEB J: off Publ Fed Am Soc Exp Biol 2004;18:851e3. Patschan D, Krupincza K, Patschan S, Zhang Z, Hamby C, Goligorsky MS. Dynamics of mobilization and homing of endothelial progenitor cells after acute renal ischemia: modulation by ischemic preconditioning. Am J Physiol Ren Physiol 2006;291: F176e85. Perin EC, Dohmann HF, Borojevic R, Silva SA, Sousa AL, Mesquita CT, et al. Transendocardial, autologous bone marrow cell transplantation for severe, chronic ischemic heart failure. Circulation 2003;107:2294e302. Prokopi M, Pula G, Mayr U, Devue C, Gallagher J, Xiao Q, et al. Proteomic analysis reveals presence of platelet microparticles in endothelial progenitor cell cultures. Blood 2009;114:723e32. 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;107:1164e9.

Please cite this article in press as: Jang IH, et al., Role of formyl peptide receptor 2 in homing of endothelial progenitor cells and therapeutic angiogenesis, Advances in Biological Regulation (2014), http:// dx.doi.org/10.1016/j.jbior.2014.09.011

10

I.H. Jang et al. / Advances in Biological Regulation xxx (2014) 1e11

Resch T, Pircher A, Kahler CM, Pratschke J, Hilbe W. Endothelial progenitor cells: current issues on characterization and challenging clinical applications. Stem Cell Rev 2012;8:926e39. Ribatti D. The discovery of endothelial progenitor cells. An historical review. Leukemia Res 2007;31:439e44. Rohde E, Schallmoser K, Reinisch A, Hofmann NA, Pfeifer T, Frohlich E, et al. Pro-angiogenic induction of myeloid cells for therapeutic angiogenesis can induce mitogen-activated protein kinase p38-dependent foam cell formation. Cytotherapy 2011;13:503e12. Schachinger V, Assmus B, Britten MB, Honold J, Lehmann R, Teupe C, et al. Transplantation of progenitor cells and regeneration enhancement in acute myocardial infarction: final one-year results of the TOPCARE-AMI Trial. J Am Coll Cardiol 2004;44: 1690e9. Schober A, Zernecke A. Chemokines in vascular remodeling. Thromb haemost 2007;97:730e7. Scott SM, Barth MG, Gaddy LR, Ahl Jr ET. The role of circulating cells in the healing of vascular prostheses. J Vasc Surg 1994;19: 585e93. Senegaglia AC, Barboza LA, Dallagiovanna B, Aita CA, Hansen P, Rebelatto CL, et al. Are purified or expanded cord blood-derived CD133þ cells better at improving cardiac function? Exp Biol Med 2010;235:119e29. Senegaglia AC, Brofman PR, Aita CA, Dallagiovanna B, Rebelatto CL, Hansen P, et al. In vitro formation of capillary tubules from human umbilical cord blood cells with perspectives for therapeutic application. Rev Bras Cir Cardiovasc Orgao of Soc Bras Cir Cardiovasc 2008;23:467e73. Shi Q, Rafii S, Wu MH, Wijelath ES, Yu C, Ishida A, et al. Evidence for circulating bone marrow-derived endothelial cells. Blood 1998;92:362e7. Somani A, Nguyen J, Milbauer LC, Solovey A, Sajja S, Hebbel RP. The establishment of murine blood outgrowth endothelial cells and observations relevant to gene therapy. Transl Res: J Lab Clin Med 2007;150:30e9. Spring H, Schuler T, Arnold B, Hammerling GJ, Ganss R. Chemokines direct endothelial progenitors into tumor neovessels. Proc Natl Acad Sci USA 2005;102:18111e6. Stamm C, Kleine HD, Choi YH, Dunkelmann S, Lauffs JA, Lorenzen B, et al. Intramyocardial delivery of CD133þ bone marrow cells and coronary artery bypass grafting for chronic ischemic heart disease: safety and efficacy studies. J Thorac Cardiovasc Surg 2007;133:717e25. Stamm C, Westphal B, Kleine HD, Petzsch M, Kittner C, Klinge H, et al. Autologous bone-marrow stem-cell transplantation for myocardial regeneration. Lancet 2003;361:45e6. Strauer BE, Brehm M, Zeus T, Kostering M, Hernandez A, Sorg RV, et al. Repair of infarcted myocardium by autologous intracoronary mononuclear bone marrow cell transplantation in humans. Circulation 2002;106:1913e8. Syed FF, Beeching NJ. Lower-limb deep-vein thrombosis in a general hospital: risk factors, outcomes and the contribution of intravenous drug use. QJM: Mon J Assoc Physicians 2005;98:139e45. Tanaka R, Wada M, Kwon SM, Masuda H, Carr J, Ito R, et al. The effects of flap ischemia on normal and diabetic progenitor cell function. Plastic Reconstr Surg 2008;121:1929e42. Tateishi-Yuyama E, Matsubara H, Murohara T, Ikeda U, Shintani S, Masaki H, 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;360:427e35. Tatsumi T, Ashihara E, Yasui T, Matsunaga S, Kido A, Sasada Y, et al. Intracoronary transplantation of non-expanded peripheral blood-derived mononuclear cells promotes improvement of cardiac function in patients with acute myocardial infarction. Circ J: off J Jpn Circ Soc 2007;71:1199e207. Thal MA, Krishnamurthy P, Mackie AR, Hoxha E, Lambers E, Verma S, et al. Enhanced angiogenic and cardiomyocyte differentiation capacity of epigenetically reprogrammed mouse and human endothelial progenitor cells augments their efficacy for ischemic myocardial repair. Circ Res 2012;111:180e90. Timmermans F, Plum J, Yoder MC, Ingram DA, Vandekerckhove B, Case J. Endothelial progenitor cells: identity defined? J Cell Mol Med 2009;13:87e102. Tongers J, Losordo DW. Frontiers in nephrology: the evolving therapeutic applications of endothelial progenitor cells. J Am Soc Nephrol: JASN 2007;18:2843e52. Tse HF, Kwong YL, Chan JK, Lo G, Ho CL, Lau CP. Angiogenesis in ischaemic myocardium by intramyocardial autologous bone marrow mononuclear cell implantation. Lancet 2003;361:47e9. Urbich C, Aicher A, Heeschen C, Dernbach E, Hofmann WK, Zeiher AM, et al. Soluble factors released by endothelial progenitor cells promote migration of endothelial cells and cardiac resident progenitor cells. J Mol Cell Cardiol 2005;39:733e42. Urbich C, Dimmeler S. Endothelial progenitor cells functional characterization. Trends Cardiovasc Med 2004;14:318e22. Valgimigli M, Rigolin GM, Cittanti C, Malagutti P, Curello S, Percoco G, et al. Use of granulocyte-colony stimulating factor during acute myocardial infarction to enhance bone marrow stem cell mobilization in humans: clinical and angiographic safety profile. Eur Heart J 2005;26:1838e45. Wang CH, Verma S, Hsieh IC, Chen YJ, Kuo LT, Yang NI, et al. Enalapril increases ischemia-induced endothelial progenitor cell mobilization through manipulation of the CD26 system. J Mol Cell Cardiol 2006;41:34e43. Weber C, Schober A, Zernecke A. Chemokines: key regulators of mononuclear cell recruitment in atherosclerotic vascular disease. Arterioscler, Thromb, Vasc Biol 2004;24:1997e2008. Weber KS, Nelson PJ, Grone HJ, Weber C. Expression of CCR2 by endothelial cells : implications for MCP-1 mediated wound injury repair and in vivo inflammatory activation of endothelium. Arterioscler, Thromb, Vasc Biol 1999;19:2085e93. Wei HM, Wong P, Hsu LF, Shim W. Human bone marrow-derived adult stem cells for post-myocardial infarction cardiac repair: current status and future directions. Singap Med J 2009;50:935e42. Westerweel PE, Teraa M, Rafii S, Jaspers JE, White IA, Hooper AT, et al. Impaired endothelial progenitor cell mobilization and dysfunctional bone marrow stroma in diabetes mellitus. PloS One 2013;8:e60357. Wollert KC, Meyer GP, Lotz J, Ringes-Lichtenberg S, Lippolt P, Breidenbach C, et al. Intracoronary autologous bone-marrow cell transfer after myocardial infarction: the BOOST randomised controlled clinical trial. Lancet 2004;364:141e8. Yoder MC. Is endothelium the origin of endothelial progenitor cells? Arterioscler, Thromb, Vasc Biol 2010;30:1094e103. Yoder MC, Mead LE, Prater D, Krier TR, Mroueh KN, Li F, et al. Redefining endothelial progenitor cells via clonal analysis and hematopoietic stem/progenitor cell principals. Blood 2007;109:1801e9.

Please cite this article in press as: Jang IH, et al., Role of formyl peptide receptor 2 in homing of endothelial progenitor cells and therapeutic angiogenesis, Advances in Biological Regulation (2014), http:// dx.doi.org/10.1016/j.jbior.2014.09.011

I.H. Jang et al. / Advances in Biological Regulation xxx (2014) 1e11

11

Yoon CH, Hur J, Oh IY, Park KW, Kim TY, Shin JH, et al. Intercellular adhesion molecule-1 is upregulated in ischemic muscle, which mediates trafficking of endothelial progenitor cells. Arterioscler, Thromb, Vasc Biol 2006;26:1066e72. Yoon CH, Hur J, Park KW, Kim JH, Lee CS, Oh IY, et al. Synergistic neovascularization by mixed transplantation of early endothelial progenitor cells and late outgrowth endothelial cells: the role of angiogenic cytokines and matrix metalloproteinases. Circulation 2005;112:1618e27.

Please cite this article in press as: Jang IH, et al., Role of formyl peptide receptor 2 in homing of endothelial progenitor cells and therapeutic angiogenesis, Advances in Biological Regulation (2014), http:// dx.doi.org/10.1016/j.jbior.2014.09.011

Role of formyl peptide receptor 2 in homing of endothelial progenitor cells and therapeutic angiogenesis.

Endothelial progenitor cells (EPCs) hold a great promise as a therapeutic mediator in treatment of ischemic disease conditions. The discovery of EPCs ...
477KB Sizes 5 Downloads 10 Views