Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2014 Dec 1.
Published in final edited form as: Acta Biomater. 2013 Aug 17;9(12):9258–9269. doi: 10.1016/j.actbio.2013.08.012

Covalent immobilization of SCF and SDF1α for in vitro culture of hematopoietic progenitor cells

Maude L Cuchiara 1, Kelsey L Horter 1, Omar A Banda 1, Jennifer L West 1,2
PMCID: PMC3972068  NIHMSID: NIHMS516772  PMID: 23958779

Abstract

Hematopoietic stem cells (HSCs) are currently utilized in the treatment of blood diseases, but widespread application of HSC therapeutics has been hindered by the limited availability of HSCs. With a better understanding of the HSC microenvironment and the ability to precisely recapitulate its components, we may be able to gain control of HSC behavior. In this work we developed a novel, biomimetic PEG hydrogel material as a substrate for this purpose and tested its potential with an anchorage independent hematopoietic cell line, 32D clone 3 cells. We immobilized a fibronectin-derived adhesive peptide sequence, RGDS; a cytokine critical in HSC self-renewal, stem cell factor (SCF); and a chemokine important in HSC homing and lodging, stromal derived factor 1α (SDF1α), onto the surfaces of poly(ethylene glycol) (PEG) hydrogels. To evaluate the system’s capabilities, we observed the effects of the biomolecules on 32D cell adhesion and morphology. We demonstrated that the incorporation of RGDS onto the surfaces promotes 32D cell adhesion in a dose dependent fashion. We also observed an additive response in adhesion on surfaces with RGDS in combination with either SCF or SDF1α. In addition, the average cell area increased and circularity decreased on gel surfaces containing immobilized SCF or SDF1α, indicating enhanced cell spreading. By recapitulating aspects of the HSC microenvironment using a PEG hydrogel scaffold, we have shown the ability to control the adhesion and spreading of the 32D cells and demonstrated the potential of the system for the culture of primary hematopoietic cell populations.

Keywords: hydrogel, poly(ethylene glycol), hematopoietic cell, stem cell factor, stromal derived factor 1α

1. Introduction

Hematopoietic stem cells (HSCs) have been successfully transplanted into humans for fifty years to treat blood diseases such as leukemia [13]. The creation of a robust system to culture HSCs ex vivo would aid in the optimization of current treatment regimens and facilitate the development of new HSC therapeutics. In vivo, HSCs reside in a microenvironment, or niche, within the marrow of long bones. Complex interactions between HSCs, stromal cells, the extracellular matrix, and signaling molecules help to maintain an intricate balance between HSC quiescence, self-renewal, and differentiation. One of the key regulators in the niche is adhesion to ECM proteins. HSCs adhere to fibronectin (FN) and other extracellular matrix proteins through integrins such as VLA-4 (very late antigen-4, α4β1), VLA-5 (very late antigen-5, α5β1), αVβ3, LFA1, [46]. Several integrins can bind to the essential amino acid sequence, RGD, of FN. Because of its specificity, this short oligopeptide can be utilized to mimic some of the adhesive properties of intact FN proteins. These interactions are not only important in cell adhesion and spreading but are also implicated in many signaling pathways within the cells, such as those that regulate hematopoiesis and proliferation [79].

Other molecules critical to maintaining HSCs in the niche include stem cell factor (SCF) and stromal derived factor 1α (SDF1α). SCF binds to and activates the c-kit receptor expressed on the surfaces of HSCs and exists in both soluble and membrane-bound forms [1012]. Membrane- or surface-bound SCF has been shown to prolong c-kit activation [1316], an interaction that is critical in HSC maintenance and self-renewal [17]. SDF1α binds to C-X-C chemokine receptor type 4 (CXCR4) on HSC membranes. SDF1α, similarly to SCF, can be found in both membrane-bound and soluble forms in the niche [18]. It plays an integral role in the homing of HSCs to the bone marrow as well as mobilization of the cells to injury sites [1921]. Signaling between surface bound SDF1α and CXCR4 was shown to maintain a pool of HSCs in the niche, leading to the proposal that this protein supports self-renewal of stem cell populations [22].

Incorporating niche molecules such as these into biomaterials, we can begin to culture HSCs in vitro. Several groups have begun to explore different bioactive scaffolds for the culture of HSCs. RGD, LDV (a peptide sequence that can bind the α4β1 integrin), and FN covalently conjugated to PET films showed successful ex vivo expansion of human CD34+ cells [8,23]. Others have focused on the effects of the mechanical properties on hematopoietic cell behavior cultured on substrates like FN-functionalized poly(ethylene glycol) diacrylate (PEG-DA) hydrogels, collagen, or collagen-functionalized poly(acrylamide) [24,25].

Another approach is the fabrication of biomaterial wells for HSC culture. This is advantageous because it allows containment of anchorage independent HSCs and facilitates interactions between HSC surface receptors and molecules presented on the well surface. Kurth et al. (2009) and Kurth et al. (2011) have immobilized ECM molecules onto poly(dimethylsiloxane) (PDMS) microcavities to study the relationship between these molecules and HSC fate [26,27]. Kobel et al. and Lutolf et al. have demonstrated the ability to generate poly(ethylene glycol) hydrogel well surfaces to study single HSC proliferation kinetics [28,29]. Lutolf et al. used microcontact printing to functionalize the well surfaces with a variety of proteins to investigate the effects of specific molecules on HSC division and engraftment.

One disadvantage of the system described by Lutolf et al. is the manner in which the wells are functionalized. The PEG prepolymer solution is molded against PDMS pillars inked with PEG-modified Protein A to functionalize the entire well surface. A chimeric protein is then added to the wells, binding to Protein A via its Fc fragment [28]. While the need to PEGylate proteins does potentially impact bioactivity, a photopolymerization strategy would enable direct patterning of PEGylated biomolecules on the well surfaces [3034]. Previous work has shown spatial presentation of specific adhesive ligands or niche proteins to HSCs to be critical [35]. The need to use chimeric proteins in Lutolf’s strategy also limits the molecules that can be incorporated onto the well surfaces. Finally, Kobel et al. and Lutolf et al. used the wells as a tool to gain a better understanding of the kinetics of HSC proliferation and the effects of cell division on engraftment potential as opposed to generating therapeutic populations of HSCs.

We have built on the work of Kobel et al. and Lutolf et al. by using photopolymerizable PEG-DA hydrogel wells as a substrate for the development of an ex vivo HSC niche. Unmodified PEG-DA hydrogels are biologically inert though the polymer matrix can easily by modified with bioactive elements such as adhesive peptide sequences, degradable elements, and whole proteins [3646] The ability to selectively incorporate these biomolecules in the matrix allows for significant control over the cell microenvironment in both two and three-dimensions. To recapitulate aspects of the HSC niche in the current work, RGDS, SCF, and SDF-1α were covalently immobilized onto the surfaces of PEG-DA hydrogels that were fabricated into culture wells. To evaluate the efficacy of the newly designed materials, we observed the adhesion and morphology of 32D cells, an interleukin-3 dependent myeloid hematopoietic progenitor cell line that expresses integrins binding RGD [47,48] as well as both c-kit and CXCR4 (Figure S1). Through the incorporation of RGDS, SCF, and SDF1α onto the substrate surface we were able to influence 32D cell adhesion and total cell area on the hydrogel surfaces and believe that further optimization of the system will result in the ability to support HSC adhesion and growth during in vitro culture.

2. Materials and Methods

All chemicals were obtained from Sigma-Aldrich (St. Louis, Missouri) unless noted.

2.1 Synthesis of PEG-DA

PEG-DA was synthesized as described previously [34,43,45,46,49,50]. Briefly, 6 kDa poly(ethylene glycol) (PEG) was reacted with acryloyl chloride at a molar ratio of 4:1 (PEG:acryloyl chloride) and triethylamine (TEA) at a molar ratio of 2:1 (PEG:TEA) in anhydrous dichloromethane (DCM). The resulting product was rinsed with K2CO3 and allowed to phase separate overnight. The organic layer containing PEG-DA was collected and dried with MgSO4, and then rotary evaporated and precipitated in diethyl ether. The precipitate was filtered and lyophilized. A sample of the resultant polymer was dissolved in chloroform, and acrylation was confirmed with proton nuclear magnetic resonance analysis.

2.2 Synthesis of PEG-RGDS

3400 MW acrylate PEG-succinidimidyl carboxymethyl (PEG-SCM, Laysan, Arab, AL) was reacted with RGDS peptide (American Peptide, Sunnyvale, CA) at a molar ratio of 1:1.1 (PEG-SCM:RGDS) in dimethylsulfoxide with N,N-Diisopropylethylamine overnight at 4°C as previously described [34,44]. The resulting product was purified by dialysis and lyophilized. The conjugation was verified using gel permeation chromatography.

2.3 Synthesis of PEG-SCF and PEG-SDF1α

An alternative PEG derivative compatible with aqueous conjugation was used to PEGylate SCF and SDF1α. 3400 MW acrylate PEG-succinimidyl valerate (PEG-SVA, Laysan) was reacted with carrier-free murine SCF (R&D, Minneapolis, MN) at a molar ratio of 42:1 (PEG-SVA:SCF) and with carrier-free murine SDF1α (R&D) at a molar ratio of 18:1 (PEG-SVA:SDF1α). The reactions were performed in phosphate buffered saline (PBS) overnight at 4°C, pH 8.0. To confirm conjugation, a Western blot was performed on the PEGylated and unmodified proteins using a 15% Tris-HCl precast polyacrylamide gel (BioRad, Hercules, CA) as previously described [4346]. Primary antibodies included a rabbit polyclonal SCF and a rabbit polyclonal SDF1α (Abcam, Cambridge, MA). The secondary antibody used was a goat polyclonal rabbit IgG conjugated to horseradish peroxidase (Abcam). For detection, the ECL chemiluminescent Western blotting analysis system (GE Healthcare, Chalfont St. Giles, UK) was employed.

2.4 Cell culture

Murine 32D clone 3 cells (32D cells, ATCC, Manassas, VA), an IL-3 dependent myeloid progenitor line, were cultured in RPMI-1640 media (Invitrogen, Carlsbad, CA) supplemented with 2 mM L-glutamine, 10% IL-3 culture supplement (Becton Dickinson, Franklin Lakes, NJ), 10% heat-inactivated fetal bovine serum (FBS), 100 U/L penicillin, and 100 mg/L streptomycin. Cells were maintained at 37 °C at 5% CO2.

2.5 Bioactivity of PEGylated proteins

To evaluate PEG-SCF bioactivity, 32D cells were seeded into tissue culture polystyrene plates at a density of 5000 cells/cm2 in three formulations of media: control media (RPMI-1640 with 2 mM L-glutamine, 10% IL-3 culture supplement, 10% heat-inactivated FBS, 100 U/L penicillin, and 100 mg/L streptomycin), control media with SCF (200 ng/ml) and control media with PEG-SCF (200 ng SCF/ml). After 24 h, the wells were imaged, and cell number was calculated using ImageJ software (NIH, Bethesda, MD).

To evaluate the bioactivity of PEGylated SDF1α, a migration assay utilizing transwells with 3 μm pores was performed. One of three media formulations was added to the well plate below the transwell: control media (RPMI 1640 containing 10% heat-inactivated FBS, 10% IL-3 culture supplement, and 1% penicillin/streptomycin), control media with SDF1α (100 ng/ml), or control media with PEG-SDF1α (100 ng SDF1α/ml). 32D cells were seeded into the top of the transwell chamber in control media at 6000 cells/cm2, and the number of cells that migrated to the bottom well was determined after 72 hr.

2.6 Fabrication of PEG-DA culture wells

To create PEG-DA hydrogel wells for cell culture, we first used microfabrication techniques to fabricate photoresist pillars as previously described [51]. The resulting photoresist masters had 500 μm high pillars with pillar diameters of 5.34 mm. To form hydrogel wells, PEG-DA was dissolved in HEPES-buffered saline (HBS, 10% w/v) at pH 7.4, and 10 μl/ml of photoinitiator (2,2-dimethoxy-2-phenyl acetophenone: 300 mg/ml in n-vinylpyrrolidone, NVP) were added to the polymer solution. The PEG solution was then sterilized via filtration (0.2 μm). The hydrogel mold consisted of two SigmaCote treated glass slides, one modified with the SU-8 pillars, that were separated with a 1 mm PTFE spacer and clamped together. The polymer solution was injected into the mold and crosslinked using long wavelength UV light (365 nm, 10 mW/cm2) for 45 s/3.75 cm2 creating a PEG-DA base hydrogel with wells. A schematic of the fabrication is shown in Figure 1A. The PEG-DA hydrogel wells were soaked in sterile PBS with 0.1% NaN3 overnight to allow swelling and maintain sterility.

Figure 1. Microfabrication and Functionalization of PEG-DA hydrogel wells.

Figure 1

A. To generate PEG-DA wells, SU-8 2100 photoresist was spin-coated onto glass slides. The photoresist slab was then exposed to UV light through a high resolution photomask. After removing the unexposed photoresist, PEG-DA wells were molded between a glass slides and the slide containing the SU-8 pillars. B. The bottom surfaces of the wells were biofunctionalized by pipetting an aqueous solution containing PEGylated biomolecules and a photoinitiator into the wells and exposing the material to UV light for 3 minutes. (Note: not to scale)

2.7 Surface immobilization of RGDS, SCF, and SDF1α

A PEG-RGDS solution (or PEG-RGDS/PEG-SCF or PEG-RGDS/PEG-SDF1α mixture) containing 10 μl/ml of photoinitiator (described above) was added to each well of a hydrogel containing 4 wells. The hydrogel wells were then exposed to UV light for 3 min. to covalently attach the RGDS (and SCF or SDF1α if included). The gels were then rinsed and soaked in sterile PBS containing 0.1% NaN3. A schematic of the immobilization process is displayed in Figure 1B. To remove any NaN3 before cell seeding, gels were rinsed twice in PBS for 1 h at 37°C and once in culture media for 1 h at 37°C. The mesh size of a 10%, 6 kDa PEG-DA hydrogel can be approximated as 3.3 nm (A description of how mesh size was determined can be found in the Supplemental Material). The minimum hydronamic radius of PEGylated protein can be estimated using the formula Rmin=0.066*MW1/3 [52]. For SCF this was 2.6 nm and for SDF1α it was 2.4 nm. Due to the similarity in mesh size and hydrodynamic radii and the short exposure time to the solution (~ 3 minutes), we do not anticipate that a significant amount of PEGylated protein diffuses into the gel.

2.8 Quantification of SCF and SDF1α on surfaces

To confirm surface protein conjugation on hydrogels, varying concentrations of SCF and SDF1α (100, 200, and 400 ng/cm2) in combination with PEG-RGDS (25 μg/cm2) were reacted with surfaces of hydrogel wells. After UV exposure, the solution was removed from each hydrogel well, and gels were soaked in PBS. A sandwich ELISA was performed on the removed reactant solution and soak solutions using Quantikine ELISA kits for SCF and SDF1α (R&D) [45].

To quantitatively determine the amount of SCF and SDF1α adsorbed to tissue culture polystyrene (TCPS) wells, the coating solution was collected and assessed with Quantikine ELISA kits. An ELISA was also performed on a the well plate to confirm adsorption to the plate surface.

2.9 Seeding cells in hydrogel and TCPS wells

32D cells were seeded into all wells at 5000 cells/cm2. The experimental groups consisted of hydrogel wells with surface immobilized: RGDS (2.5, 25, and 250 μg/cm2); RGDS (25 μg/cm2) and SCF (400 ng/cm2); and RGDS (25 μg/cm2) and SDF1α (400 ng/cm2). PEG-DA wells (0 μg/cm2 RGDS) served as controls. Each group consisted of 8 gel wells. Cells were also seeded into tissue culture wells coated with the following solutions: FN (1 μg/cm2); FN (1 μg/cm2) and SCF (400 ng/cm2); FN (1 μg/cm2) and SDF1α (400 ng/cm2); SCF (400 ng/cm2); and SDF1α (400 ng/cm2). Each group consisted of 4 wells. Cells were maintained in culture for 6 days at 37°C with 5% CO2.

2.10 Evaluation of cell adhesion

To evaluate 32D cell adhesion, hydrogel and TCPS wells were rinsed by the addition of fresh media after 48 h in culture. Phase contrast images of each well were captured in randomly selected locations using a Zeiss Axiovert 135 inverted microscope (Zeiss, Oberkochen, Germany) and Jenoptik ProgRes C5 charge-coupled device (CCD) camera (Jenoptik, Jena, Germany). Cells were counted using ImageJ software.

2.11 Evaluation of cell spreading

Cells were fixed with 4% paraformaldehyde, permeabilized with 0.1% Triton X-100, and blocked in PBS containing 1% bovine serum albumin. Cells were then incubated with Alexa-Fluor 488 phalloidin (Invitrogen), which stains actin filaments, and counterstained with DAPI to visualize cell nuclei. Cells were imaged using epifluorescent microscopy on a Zeiss Axiovert 135 inverted microscope. Cell area and circularity were determined using ImageJ software.

2.12 Statistical analysis

One-way ANOVA and Tukey’s post-hoc analyses were performed to evaluate statistical differences between groups using a 95% probability level (p<0.05).

3. Results

3.1 PEGylation of Proteins

The conjugations of SCF and SDF1α to PEG were confirmed with a Western blot (Figure 2). In the PEG-SCF lane, a protein smear beginning at 28 kDa demonstrated successful conjugation to the polymer by an increase over the molecular weight of the soluble cytokine (Figure 2A). A faint band at 18 kDa in the PEG-SCF lane signified a low fraction of unconjugated protein in the final product. Rather than forming a single band on the blot, these types of reactions generate a product with a range of molecular sizes due to the polydispersity of the PEG reactant and also because multiple polymer chains will attach to the proteins [4446]. Similar results for PEG-SDF1α are shown in Figure 2B, where the conjugated PEG-protein product showed as a smear beginning at 14 kDa, a significant shift from the 8 kDa unmodified protein.

Figure 2. Western blot confirming PEGylation of SCF and SDF1α.

Figure 2

A. In the SCF lane, a band at 18 kDa corresponds to the molecular weight (MW) of the extracellular domain of SCF. In the PEG-SCF lane, there is a smear beginning at a MW of ~28 kDa. The increase in MW confirms the addition of PEG chains to the SCF molecule, and the resulting smear indicates that some SCF molecules have multiple PEG chains. B. There is a band present in the SDF1α lane near 8 kDa. In the PEG-SDF1α lane you can see a smear beginning around 14 kDa indicating the protein has been successfully conjugated to PEG chains.

3.2 SCF and SDF1α bioactivity after PEGylation

The addition of large or multiple PEG chains to a protein can alter the bioactivity of a protein particularly if a high number of chains are covalently attached to the protein or the PEG chains are tethered at the protein’s reactive site [5356]. In order to ensure that SCF and SDF1α remained bioactive after PEGylation, we exposed 32D cells to soluble forms of PEG-SCF and PEG-SDF1α and evaluated the effects on proliferation and migration, respectively. The addition of both unmodified SCF and PEG-SCF to culture media resulted in significant increases in cell numbers compared to control wells without any form of the protein (Figure 3A). The addition of SDF1α and PEG-SDF1α to the media promoted significant 32D cell migration through the transwell membrane (Figure 3B), though migration in response to PEG-SDF1α was significantly reduced compared to the native protein.

Figure 3. Bioactivity of PEGylated SCF and SDF1α.

Figure 3

The bioactivity of the PEGylated proteins was evaluated by culturing 32D cells in media supplemented with either no protein, the natural occurring form of the protein, or the PEGylated version of the protein. For SCF, the effects on 32D cell proliferation and for SDF1α the effects on 32D cell migration were evaluated. A. In the SCF bioactivity assay, the percent change in 32D cell number after 5 days was significantly higher when SCF or PEG-SCF was added to the media. We saw no significant difference between the PEGylated and unPEGylated versions of the protein (n=8). B. In the migration assay, when SDF1α or PEG-SDF1α was added to the media, we saw an increase in 32D cell migration. However, the PEG chains appeared to interfere with the protein function as the migration of 32D cells in the presence of PEG-SDF1α was significantly reduced compared to that demonstrated with SDF1α (n=54). Data reported as average ± standard deviation, * denotes significance compared to control, and # denotes significance compared to PEG-SDF1α (p<0.05).

3.3 Biomolecule incorporation onto hydrogel and TCPS surfaces

To confirm that the proteins were covalently immobilized on well surfaces, ELISAs were conducted. The results indicated that approximately 80% of the PEG-SCF available for reaction was conjugated to the hydrogel with the PEG-SDF1α immobilization slightly higher at 98%. The surface concentrations of SCF and SDF1α used in experiments (400 ng/cm2) resulted in 73.5 ± 6.7 ng/well and 88.3 ± 0.5 ng/well, respectively.

On TCPS wells, over 99 % of the proteins were adsorbed to the plate surface. The coating solution used (400 ng/cm2) resulted in 127.997 ± 0.0093 ng/well (SCF), 127.104 ± 0.0891 ng/well (SCF + FN), 127.992 ± 0.0014 ng/well (SDF1α), and 127.983 ± 0.0059 ng/well (FN SDF1α). ELISAS on the coated TCPS plates confirmed the presence of SCF and SDF1α on the TCPS surfaces.

3.4 32D cell adhesion on RGDS modified hydrogels

Figure 4 displays the average number of adherent 32D cells per square centimeter on the PEG-RGDS hydrogels after 48 h, with increases in RGDS concentration corresponding to higher levels of attachment. On hydrogels with no bioactive components, 874 ± 316 cells/cm2 were adherent. Upon addition of 2.5 μg/cm2 RGDS, 1,572 ± 1,194 cells adhered, and with 25 μg/cm2 RGDS the adherent cell number increased to 4,102 ± 3,196 cells/cm2 though neither of these changes was significant. At 250 μg/cm2 RGDS, there was a significant increase in 32D cell adhesion to 10,299 ± 1,328 cells/cm2. These numbers were found to be statistically similar to 32D cell adhesion to FN coated TCPS plates (Figure S2).

Figure 4. Adherent 32D cells on hydrogel well surfaces functionalized with biomolecules.

Figure 4

32D cells were cultured on surfaces with covalently immobilized RGDS, SCF, and SDF1α. As the concentration of RGDS was increased, there was a corresponding significant increase in cell adhesion. With the addition of SCF (400 ng/cm2) or SDF1α (400 ng/cm2) to the surface of gels with medium RGDS concentrations (25 μg/cm2), there was a significant increase in cell adhesion compared to the peptide alone. However, with 250 μg/cm2 RGDS on the surface, the addition of SCF or SDF1α did not similarly increase the number of adherent cells. Data presented as average ± standard deviation, * indicates statistical significance compared to 0 μg/cm2 RGDS, 2.5 μg/cm2 RGDS, and 25 μg/cm2 RGDS (n=4–8, p<0.05).

3.5 32D cell adhesion on hydrogels with surface immobilized SCF and SDF1α

Compared to the samples containing the intermediate concentration of RGDS (25 μg/cm2) alone, gels also presenting surface immobilized SCF had significantly more adherent 32Ds after 48 h: 4,102.0 ± 3,196.1 cells/cm2 on RGDS hydrogels versus 12,236 ± 2,389 cells/cm2 with the SCF and RGDS combination. We also observed a significant increase in adherent 32D cells when SDF1α (400 ng/cm2) was added to the surfaces of gels already modified with 25 μg/cm2 RGDS (Figure 4). After 48 hours, the number of cells on gels with SDF1α was comparable to that on SCF surfaces at 11,839 ± 4,722 cells/cm2. At the higher concentration of RGDS (250 μg/cm2), the effects of SCF and SDF1α were masked by those of the adhesive peptide with no further increase in cell number with addition of the proteins. These were statistically similar to 32D cell adhesion levels on SCF- and SDF1α-coated TCPS plates (Figure S2).

3.6 32D cell morphology on hydrogel surfaces functionalized with SCF and SDF1α

The morphology of 32D cells on hydrogel surfaces was quantified in fluorescent images obtained following DAPI/phalloidin staining (Figure 5). On gels with RGDS alone, the cells appeared round and exhibited minimal spreading (Figure 5B–D), similar to those seen on the unmodified PEG-DA gels (Figure 5A). A definitive morphological change was observed in cells when SCF or SDF1α was present on the surface (Figure 5E–F) as compared to gels containing RGDS alone. In these experimental groups, 32D cells were well dispersed across the entire surface of the gels and more spread with many exhibiting distinct filopodia as indicated by the white arrows in Figure 5. While there were 32D cells present on TCPS wells with adsorbed SCF or SDF1α, the cells remained small and rounded compared to hydrogels with covalently immobilized SCF or SDF1α (Figure 6).

Figure 5. 32D cell morphology in bioactive PEG hydrogel wells.

Figure 5

Representative images of 32D cells on hydrogel surfaces. Actin filaments are stained with phalloidin (green) and nuclei are stained with DAPI (blue). Higher concentrations of RGDS on the surface resulted in more adherent 32D cells on the gels. With the covalent incorporation of SCF and SDF1α on the surfaces, the cells appeared more spread, and distinct filopodia can be seen extending from many of the cells (denoted by white arrows). (A. PEG-DA, B. 2.5 μg/cm2 RGDS, C. 25 μg/cm2 RGDS, D. 250 μg/cm2 RGDS, E. 25 μg/cm2 RGDS + 400 ng/cm2 SCF, F. 25 μg/cm2 RGDS + 400 ng/cm2 SDF1α; Scale bars = 25 μm).

Figure 6. 32D cell morphology in TCPS wells with physioadsorbed proteins.

Figure 6

Representative images of 32D cells on hydrogel surfaces. Actin filaments are stained with phalloidin (green) and nuclei are stained with DAPI (blue). Cells appeared rounded and small in all groups. (A. 1 μg/cm2 FN, B. 1 μg/cm2 FN + 400 ng/cm2 SCF, C. 1 μg/cm2 FN + 400 ng/cm2 SDF1α, D. 400 ng/cm2 SCF, E. 400 ng/cm2 SDF1α; Scale bars = 25 μm).

A quantification of the cell images revealed that the average cell area varies widely within each group (Figure 7). This large distribution of cell size can be attributed to the varying degrees of spreading seen in the 32D population regardless of culture surface. When the proteins were covalently immobilized to hydrogel surfaces, there were significant increases in average cell size from 261 ± 175 μm2 with 250 μg/cm2 RGDS to 450 ± 275 μm2 and 367 ± 247 μm2 for SCF and SDF1α, respectively. The data was also plotted as a histogram to obtain a better sense of the distribution of cell sizes (Figure 8A). A similar cell size distribution was seen between all three RGDS concentrations with most cell sizes falling between 0–300 μm2. With the addition of SCF and SDF1α, the cell size distribution broadened and the average cell size centered on 300–400 μm2. There was also an increase in the percentage of cells that were larger than 500 μm2 (30% for SCF and 20% for SDF1α) when compared to gels containing only or no RGDS (less than 10% for these groups). In contrast, 32D cells cultured in TCPS wells with physioadsorbed proteins were significantly less spread (Figure 7). The histogram shows that the 32D cell area centered around 200 μm2 (Figure 8B). There was also a shift of the spread to the left with more than 99% of the cells smaller than 500 μm2 in all groups.

Figure 7. Quantification of 32D cell morphology in hydrogel and TCPS wells.

Figure 7

A) 32D cells spread to a greater extent on hydrogels modified with RGDS (25 μg/cm2) and SCF (400 ng/cm2) or SDF1α (400 ng/cm2) compared to RGDS alone gels and TCPS wells with physioadsorbed proteins. The wide distribution of cell size is due to the heterogeneity of 32D cells. (Black bars are hydrogel wells while grey bars are TCPS wells. Bars represent mean ± standard deviation. Bars that do no share a common letter are significantly different, n=105 (0 μg/cm2 RGDS), 193 (2.5 μg/cm2 RGDS), 2454 (25 μg/cm2 RGDS), 863 (250 μg/cm2 RGDS), 617 (SCF), 631 (SDF1α), 622 (1 μg/cm2 FN), 1177(1 μg/cm2 FN +SCF), 839(1 μg/cm2 FN +SDF1α), 881(SCF), 472(SDF1α), p < 0.05).

Figure 8. 32D cell area distribution.

Figure 8

A. Cell area distribution on hydrogel wells. On all three RGDS concentrations most cells were between 100 – 200 μm2. On hydrogel surfaces with SCF or SDF1α there was a shift to the right and a broadening of the cell area distribution compared to RGDS alone. B. Cell area distribution on TCPS wells. On physioadsorbed biomolecules, cell area was centered around 100 – 200 μm2. The distribution of cell sizes is not as wide as on hydrogel surfaces with most cells smaller than 400 μm2.

Measurements of circularity were also generated to determine cell morphology (Figure 9). 32D cells on surfaces with low concentrations of RGDS (0 or 2.5 μg/cm2) retained a round shape as denoted by circularity values of 0.82 ± 0.12 or 0.82 ± 0.11 respectively. As the RGDS concentration increased from 2.5 to 25 μg/cm2 and from 25 to 250 μg/cm2, circularity decreased significantly (0.77 ± 0.15 and 0.72 ± 0.13 respectively) indicating a higher degree of cell spreading as more RGDS was immobilized. Upon addition of SCF or SDF1α onto gel surfaces, circularity again dropped significantly (0.64 ± 0.13 and 0.68 ± 0.12 respectively) demonstrating the effect of these proteins on hematopoietic cell behavior. Cells cultured on TCPS wells with physioadsorbed proteins, were significantly rounder that cells cultured on hydrogel wells functionalized with RGDS and SCF or RGDS and SDF1α. All circularity values were greater than 0.71: 0.71 ± 0.11 (1 μg/cm2 FN), 0.77 ± 0.10 (1 μg/cm2 FN +SCF), 0.75 ± 0.12 (1 μg/cm2 FN + SDF1α), 0.74 ± 0.11 (SCF), and 0.76 ± 0.11 (SDF1α).

Figure 9. Circularity of 32D cells in PEG hydrogel and TCPS wells.

Figure 9

32D cells on samples with no RGDS present retained a round shape. 32D cells on bioactive hydrogels were less circular indicating spreading. 32D cells on TCPS coated plates were rounder and less spread than cells on hydrogels with covalently immobilized SCF or SDF1α. Black bars are hydrogel wells while grey bars are TCPS wells. Bars that do no share a common letter are significantly different. n=105 (0 μg/cm2 RGDS), 185 (2.5 μg/cm2 RGDS), 2454 (25 μg/cm2 RGDS), 699 (250 μg/cm2 RGDS), 497 (25 μg/cm2 RGDS+SCF), 458 (25 μg/cm2 RGDS+SDF1α), 482 (1 μg/cm2 FN), 861 (1 μg/cm2 FN +SCF), 617 (1 μg/cm2 FN +SDF1α), 625 (SCF), 388 (SDF1α) (p < 0.05).

4. Discussion

HSCs have proved successful in clinical applications to treat blood diseases and cancers, but they remain difficult to study for applications in other types of treatments due to their limited availability and inability to be expanded effectively in vitro. There is a need to engineer the HSC microenvironment to better control and understand HSC self-renewal and differentiation [57]. Several attempts have been made to expand the cells using biofunctional materials to mimic the complex HSC microenvironment [8,2329,5862]. However, a system that provides for both HSC expansion and long-term maintenance of their multipotent state has not been developed.

PEG-DA hydrogels are ideal materials for the culture of HSCs because we can easily control and modulate the biochemical components and mechanical properties of the scaffold [24,25,63]. In this work the niche components RGDS, SCF, and SDF1α were PEGylated and immobilized on the surface of PEG-DA hydrogel well surfaces and assayed for their effects on cell adhesion and morphology of a model cell line, 32D cells, in an effort to demonstrate the feasibility of this approach in directing hematopoietic progenitor cell behavior. PEGylation has previously been shown to extend the half-lives of biomolecules [53,56], and our group has PEGylated several proteins critical in angiogenesis and used these molecules to promote and support blood vessel formation within PEG hydrogels for several weeks [4346,64].

To ensure that the PEGylated versions of SCF and SDF1α maintained their bioactivity, we observed the effects of SCF on 32D cell proliferation and SDF1α on 32D cell migration. There was no significant difference between the PEGylated and unmodified forms of SCF suggesting that the PEG chains do not substantially affect the bioactivity of the protein. However, the presence of the PEG chains appeared to diminish SDF1α bioactivity as demonstrated by a statistical decrease in cell migration in the PEG-SDF1α experimental group when compared to the positive control. Nonetheless, substantial bioactivity was retained, and this was sufficient for the desired studies.

PEG-RGDS was immobilized onto the hydrogel surfaces to mimic the in vivo interactions between HSCs and ECM proteins in the stem cell niche, since adhesive interactions of this nature have been shown to retain HSCs in the niche and affect their fate [35,65,66]. In previous studies, RGD tethered to PEGylated glass surfaces via avidin-biotin binding successfully promoted hematopoietic cell adhesion [9], and a cyclic form of the peptide was similarly effective in inducing progenitor cell adhesion when incorporated into solid lipid monolayers [67,68]. RGD has also been shown to promote proliferation of HSCs when conjugated to poly(ethylene teraphthalate) substrates [8].

32D cells cultured on PEG-DA substrates, containing no biofunctional components, demonstrated minimal adhesion. When PEG-RGDS was incorporated onto the surfaces of the hydrogel wells, we observed a dose dependent response. Previous work with a variety of cell types has shown adhesion to be sensitive to surface ligand concentration [42,6971]. For example, Gonzalez et al. utilized PEG-DA hydrogels with RGDS peptide incorporated into the bulk to culture neutrophils and showed a similar increase in adhesion when increasing the peptide concentration [42]. Our work demonstrates a similar ability to affect hematopoietic cell adhesion by altering the RGDS concentration in hydrogel wells. At the low peptide concentration (2.5 μg/cm2), minimal cell adhesion, comparable to that seen on un-modified PEG-DA hydrogels, indicated that this amount of RGDS is insufficient to enable surface adhesion. By increasing the RGDS surface concentration from 25 to 250 μg/cm2, we saw a correlative increase in 32D cell adhesion reaching a similar number of adherent cells as on a FN-coated TCPS plate. Further, the significant increase in cell adhesion with 250 μg/cm2 RGDS compared to lower surface RGDS concentrations (2.5 and 25 μg/cm2) and unmodified hydrogel controls suggested that 32D cells require binding multiple RGDS molecules to adhere to gel surfaces with enough strength to withstand rinsing. In the niche, hematopoietic cells express integrins, which bind specifically to the RGDS sequence. Integrin activation by RGDS can act as a positive feedback loop resulting in the expression of additional integrins and enhanced cell adhesion [28]. Adhesive interactions retain cells in the HSC niche and help maintain them in a multipotent state. As cells differentiate they lose the ability to adhere to FN and travel out of the niche to sites of injury [7276]. Thus, the ability to promote cell adhesion to our synthetic niche is critical to the successful expansion of clinically relevant HSCs. To exert finer control of HSC behavior, RGDS ligands could be patterned on the hydrogel surface [30,34]. Previous work has shown that the spatial presentation of cyclic RGD ligands significantly affects HSC adhesion, integrin distribution, and signal transduction [35].

Fibronectin works with various cytokines in the niche to promote cell adhesion [7]. When an RGDS concentration of 25 μg/cm2 was immobilized on hydrogel well surfaces with SCF and SDF1α, 32D adhesion increased significantly. As described in Section 1, SCF is a cytokine that promotes HSC self-renewal and maintenance while SDF1α is a chemokine that recruits circulating HSCs and is thought to maintain a pool of undifferentiated HSCs [16,22,7780]. Several studies have shown that SCF can enhance adhesion of hematopoietic cells to FN [81,82]. Additional evidence suggests that the binding of SCF to the c-kit receptor activates integrins on the cell surface to promote adhesion to RGDS [78]. Furthermore, Lévesque et al. found that in several minimally adherent hematopoietic progenitor cell lines, exposure to SCF resulted in increased avidity of integrins to FN [83]. We hypothesize that SCF is activating additional integrins on the 32D cells to enable a greater number of cells to adhere. In addition, due to the covalent immobilization of the SCF in this synthetic niche, the prolonged maintenance of 32D cell adhesion may be due to sustained activation of the c-kit receptor as bound SCF cannot be internalized [1315,84]. In previous work, SCF was non-covalently immobilized onto microparticle coated plates and successfully stimulated hematopoietic cell proliferation; however, it was gradually released from the particles over time with a half-life of five days [85]. The immobilization strategy utilized in this work can potentially increase the half-life of SCF and thus allow longer activation of the c-kit receptor and maintenance of cell adhesion.

Activation of the CXCR4 receptor on HSC surfaces by immobilized SDF1α has been shown to activate integrins and help retain HSCs in the niche [86]. We hypothesize that immobilization of SDF1α onto the gel surface upregulated the expression of these integrins in 32D cells. Similar to SCF, the increase in integrins allows more 32D cells to bind to RGDS molecules presented on the gel surface. The immobilization of SDF1α on the gel surface is critical, as soluble SDF1α has previously been shown to be less effective in promoting hematopoietic cell adhesion [87]. Again, we believe that our functionalization methods can increase the half-life of SDF1α and in turn prolong cell adhesion.

When compared to hydrogel wells with the highest concentration of RGDS (250 μg/cm2), the addition of SCF and SDF1α to hydrogel well surfaces—with either 25 μg/cm2 or 250 μg/cm2 of RGDS—did not significantly affect 32D cell adhesion. This is likely because the final result of each of these factors is activation of a common integrin. With a finite number of integrins available for cell surface display, direct activation of integrins in the presence of high levels of adhesive peptide could be saturating the response, thus negating any potential effect via c-kit or CXCR4. Similarly, when comparing the covalently immobilized proteins in hydrogel wells to proteins coated on TCPS plates, there was not a significant difference in 32D cell adhesion. This was not surprising as the 32D cells express integrins that bind to FN and can also adhere to serum proteins adsorbed to TCPS plates [47,48]. In addition, cell adhesion was evaluated at an early timepoint (48 h) before significant turnover of the proteins on the TCPS plate. Though these proteins did not significantly affect cell adhesion when presented in combination with high RGDS or FN concentrations, SCF and SDF1α did influence 32D cell morphology.

32D cells on surfaces with either SCF or SDF1α were significantly more spread than cells on surfaces with RGDS alone, indicating a greater degree of cell spreading. Cells on surfaces with no biofunctional components or RGDS alone also had a significantly rounder shape and fewer filopodia when compared to 32D cells on hydrogel substrates with SCF or SDF1α. As mentioned above, it is known that binding to these molecules activates multiple integrins on the cell surface [78,8688]. The integrins then cluster and can connect the RGD molecules to the cells’ actin cytoskeleton allowing the cells to migrate and spread on the gel surfaces [89]. Integrin activation acts in a positive feedback loop causing a sustained upregulation of integrin expression and resulting in larger cell spread areas [90].

Many of the 32D cells on gels functionalized with SCF or SDF1α also had a motile morphology indicated by filopodia extending outward from the cell centers. The binding of c-kit to SCF is known to promote cell motility [9193], and previously, Fruehauf et al. observed podia formation and associated 32D cell motility in response to stimulation with SDF1α on FN coated glass surfaces [18]. SDF1α is also known to be a major modulator of HSC migration through its interaction with the CXCR4 receptor on HSC surfaces, and recent work has shown that stimulation with SDF1α induces actin polymerization and migration in human CD34+ cells [94]. This migration depends on the activation of the VLA4 and VLA5 integrins which bind to FN in the HSC niche [86]. The 32D podia formation seen on our surface modified gels suggests that SCF and SDF1α have not only upregulated cell adhesion but may have also triggered the 32D cells to migrate across the hydrogel surfaces via the interactions between integrins and the RGD peptide. This migratory phenotype could be beneficial in the culture of primary HSCs, as HSCs that are used in clinical applications must be able to home to the bone marrow environment and engraft to successfully repopulate the immune system. A migratory morphology in vitro can be indicative of the potential mobility of cells upon implantation. Previous work has shown that HSCs that were more migratory in vitro displayed higher degrees of engraftment [94,95].

Interestingly, the 32D cells did not respond similarly to physioadsorbed SCF and SDF1α on TCPS well plates. The 32D cells were significantly less spread and more circular than 32D cells cultured on the hydrogels with covalently immobilized proteins. This could be due to the fact that covalent immobilization of SCF and SDF1α can lead to sustained activation of c-kit and CXCR4 respectively and subsequent upregulation of integrins which allows the cells to spread and migrate [78,83,8688]. Alternatively, the differences in the surfaces’ mechanical properties may be influencing cell morphology [24,25,63]. Previous work has shown elastic modulus to influence the behavior of 32D cells, and the softer hydrogel environment could support cell spreading.

Combining these results with the cell adhesion data, we have demonstrated the ability to stimulate specific hematopoietic cell behavior via covalent immobilization of biomolecules on the hydrogel surface. We selected HSC niche components known to influence hematopoietic cell behavior to evaluate the possibility of designing PEG hydrogels to function as synthetic HSC microenvironments. We believe our findings validate the further development of bioactive PEG hydrogel wells for primary HSC expansion.

5. Conclusions

In this work, we designed biomimetic PEG-DA hydrogels for the in vitro culture of hematopoietic progenitor cells. By incorporating RGDS, SCF, and SDF1α onto hydrogel surfaces, we were able to recapitulate important properties of the HSC microenvironment and alter 32D cell adhesion and spreading. This same strategy can be used to incorporate other niche molecules into the system to more finely tune the material to match properties of the in vivo HSC microenvironment. Currently, HSCs cannot be maintained in culture for extended periods of time without proceeding to differentiation or apoptosis. The development of synthetic scaffolds to control HSC behavior and growth will be critical in exploring the potential of HSC in novel therapeutics, which require large numbers of these multipotent cells. Biomimetic hydrogels are optimal platforms for such a culture system because we can easily render them bioactive and control cell interactions by selecting specific molecules to include in the matrix. Future work will investigate the use and optimization of this system for the expansion of primary HSCs, and the effectiveness of the scaffold will finally be determined by evaluating the differentiation potential of the cells after culture in the synthetic niche.

Supplementary Material

01

Acknowledgments

This work was supported by an NSF GRF (MLC) and NIH R01 EB005173. Thank you to Dr. Melissa McHale for reviewing the manuscript.

Footnotes

Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final citable form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

References

  • 1.McGovern J, Russell P, Atkins L, Webster E. Treatment of terminal leukemic relapse by total-body irradiation and intravenous infusion of stored autologous bone marrow obtained during remission. N Engl J Med. 1959;260:675–83. doi: 10.1056/NEJM195904022601401. [DOI] [PubMed] [Google Scholar]
  • 2.Mathe G, Amiel J, Schwarzenberg L, Cattan A, Schneider M. Adoptive immunotherapy of acute leukemia: experimental and clinical results. Cancer Res. 1965;25:1525. [PubMed] [Google Scholar]
  • 3.Thomas ED, Lochte HL, Jr, Cannon JH, Sahler OD, Ferrebee JW. Supralethal whole body irradiation and isologous marrow transplantation in man. J Clin Invest. 1959;38:1709. doi: 10.1172/JCI103949. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Rosemblatt M, Vuillet-Gaugler MH, Leroy C, Coulombel L. Coexpression of two fibronectin receptors, VLA-4 and VLA-5, by immature human erythroblastic precursor cells. J Clin Invest. 1991;87:6–11. doi: 10.1172/JCI115002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Teixidó J, Hemler ME, Greenberger JS, Anklesaria P. Role of beta 1 and beta 2 integrins in the adhesion of human CD34hi stem cells to bone marrow stroma. J Clin Invest. 1992;90:358–67. doi: 10.1172/JCI115870. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Kerst JM, et al. Alpha 4 beta 1 and alpha 5 beta 1 are differentially expressed during myelopoiesis and mediate the adherence of human CD34+ cells to fibronectin in an activation-dependent way. Blood. 1993;81:344–51. [PubMed] [Google Scholar]
  • 7.Yokota T, et al. Growth-supporting activities of fibronectin on hematopoietic stem/progenitor cells in vitro and in vivo: structural requirement for fibronectin activities of CS1 and cell-binding domains. Blood. 1998;91:3263–72. [PubMed] [Google Scholar]
  • 8.Jiang X-S, Chai C, Zhang Y, Zhuo R-X, Mao H-Q, Leong KW. Surface-immobilization of adhesion peptides on substrate for ex vivo expansion of cryopreserved umbilical cord blood CD34+ cells. Biomaterials. 2006;27:2723–32. doi: 10.1016/j.biomaterials.2005.12.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Gunawan RC, King Ja, Lee BP, Messersmith PB, Miller WM. Surface presentation of bioactive ligands in a nonadhesive background using DOPA-tethered biotinylated poly(ethylene glycol) Langmuir. 2007;23:10635–43. doi: 10.1021/la701415z. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Anderson DM, et al. Molecular cloning of mast cell growth factor, a hematopoietin that is active in both membrane bound and soluble forms. Cell. 1990;63:235–43. doi: 10.1016/0092-8674(90)90304-w. [DOI] [PubMed] [Google Scholar]
  • 11.McNiece IK, Briddell RA. Stem cell factor. J Leukocyte Biol. 1995;58:14–22. doi: 10.1002/jlb.58.1.14. [DOI] [PubMed] [Google Scholar]
  • 12.Toksoz D, et al. Support of human hematopoiesis in long-term bone marrow cultures by murine stromal cells selectively expressing the membrane-bound and secreted forms of the human homolog of the steel gene product, stem cell factor. Proc Natl Acad Sci USA. 1992;89:7350–4. doi: 10.1073/pnas.89.16.7350. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Miyazawa K, Toyama K, Gotoh A, Hendrie PC, Mantel C, Broxmeyer HE. Ligand-dependent polyubiquitination of c-kit gene product: a possible mechanism of receptor down modulation in M07e cells. Blood. 1994;83:137–45. [PubMed] [Google Scholar]
  • 14.Yee NS, Langen H, Besmer P. Mechanism of kit ligand, phorbol ester, and calcium-induced down-regulation of c-kit receptors in mast cells. J Biol Chem. 1993;268:14189–201. [PubMed] [Google Scholar]
  • 15.Yee NS, Hsiau CW, Serve H, Vosseller K, Besmer P. Mechanism of down-regulation of c-kit receptor. Roles of receptor tyrosine kinase, phosphatidylinositol 3′-kinase, and protein kinase C. J Biol Chem. 1994;269:31991–8. [PubMed] [Google Scholar]
  • 16.Miyazawa K, Williams DA, Gotoh A, Nishimaki J, Broxmeyer HE, Toyama K. Membrane-bound Steel factor induces more persistent tyrosine kinase activation and longer life span of c-kit gene-encoded protein than its soluble form. Blood. 1995;85:641–9. [PubMed] [Google Scholar]
  • 17.McCarthy KF, Ledney GD, Mitchell R. A deficiency of hematopoietic stem cells in steel mice. Cell Tissue Kinet. 1977;10:121–6. doi: 10.1111/j.1365-2184.1977.tb00137.x. [DOI] [PubMed] [Google Scholar]
  • 18.Fruehauf S, Srbic K, Seggewiss R, Topaly J, Ho A. Functional characterization of podia formation in normal and malignant hematopoietic cells. J Leukocyte Biol. 2002;71:425. [PubMed] [Google Scholar]
  • 19.Aiuti A, Webb I, Bleul C, Springer T. The Chemokine SDF-1 Is a Chemoattractant for Human CD34+ hematopoietic progenitor cells and Provides a New Mechanism to Explain the Mobilization of CD34+ Progenitors to Peripheral Blood. J Exp Med. 1997;185:111–120. doi: 10.1084/jem.185.1.111. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Bleul CC, Fuhlbrigge RC, Casasnovas JM, Aiuti A, Springer Ta. A highly efficacious lymphocyte chemoattractant, stromal cell-derived factor 1 (SDF-1) J Exp Med. 1996;184:1101–9. doi: 10.1084/jem.184.3.1101. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Netelenbos T, Zuijderduijn S, Van Den Born J, Kessler FL, Zweegman S, Huijgens PC, Dra AM. Proteoglycans guide SDF-1-induced migration of hematopoietic progenitor cells progenitor cell (HPC) trafficking to the bone mar-Cell cultures. J Leukocyte Biol. 2002;72:353–362. [PubMed] [Google Scholar]
  • 22.Sugiyama T, Kohara H, Noda M, Nagasawa T. Maintenance of the hematopoietic stem cell pool by CXCL12-CXCR4 chemokine signaling in bone marrow stromal cell niches. Immunity. 2006;25:977–88. doi: 10.1016/j.immuni.2006.10.016. [DOI] [PubMed] [Google Scholar]
  • 23.Feng Q, Chai C, Jiang XS, Leong KW, Mao HQ. Expansion of engrafting human hematopoietic stem/progenitor cells in three-dimensional scaffolds with surface-immobilized fibronectin. J Biomed Mater Res Part A. 2006;78:781–791. doi: 10.1002/jbm.a.30829. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Choi JS, Harley BAC. The combined influence of substrate elasticity and ligand density on the viability and biophysical properties of hematopoietic stem and progenitor cells. Biomaterials. 2012;33:4460–8. doi: 10.1016/j.biomaterials.2012.03.010. [DOI] [PubMed] [Google Scholar]
  • 25.Lee-Thedieck C, Rauch N, Fiammengo R, Klein G, Spatz JP. Impact of substrate elasticity on human hematopoietic stem and progenitor cell adhesion and motility. J Cell Sci. 2012;125:3765–75. doi: 10.1242/jcs.095596. [DOI] [PubMed] [Google Scholar]
  • 26.Kurth I, Franke K, Pompe T, Bornhäuser M, Werner C. Hematopoietic stem and progenitor cells in adhesive microcavities. Integr Biol. 2009;1:427–34. doi: 10.1039/b903711j. [DOI] [PubMed] [Google Scholar]
  • 27.Kurth I, Franke K, Pompe T, Bornhäuser M, Werner C. Extracellular matrix functionalized microcavities to control hematopoietic stem and progenitor cell fate. Macromol Biosci. 2011;11:739–47. doi: 10.1002/mabi.201000432. [DOI] [PubMed] [Google Scholar]
  • 28.Lutolf MP, Doyonnas R, Havenstrite K, Koleckar K, Blau HM. Perturbation of single hematopoietic stem cell fates in artificial niches. Integr Biol. 2009;1:59–69. doi: 10.1039/b815718a. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Kobel S, Limacher M, Gobaa S, Laroche T, Lutolf MP. Micropatterning of hydrogels by soft embossing. Langmuir. 2009;25:8774–9. doi: 10.1021/la9002115. [DOI] [PubMed] [Google Scholar]
  • 30.Hahn MS, Taite LJ, Moon JJ, Rowland MC, Ruffino KA, West JL. Photolithographic patterning of polyethylene glycol hydrogels. Biomaterials. 2006;27:2519–24. doi: 10.1016/j.biomaterials.2005.11.045. [DOI] [PubMed] [Google Scholar]
  • 31.Lee S-H, Moon JJ, West JL. Three-dimensional micropatterning of bioactive hydrogels via two-photon laser scanning photolithography for guided 3D cell migration. 2008;29:2962–8. doi: 10.1016/j.biomaterials.2008.04.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Liu Tsang V, et al. Fabrication of 3D hepatic tissues by additive photopatterning of cellular hydrogels. FASEB J. 2007;21:790–801. doi: 10.1096/fj.06-7117com. [DOI] [PubMed] [Google Scholar]
  • 33.Miller JS, Béthencourt MI, Hahn M, Lee TR, West JL. Laser-scanning lithography (LSL) for the soft lithographic patterning of cell-adhesive self-assembled monolayers. Biotechnol Bioeng. 2006;93:1060–1068. doi: 10.1002/bit.20809. [DOI] [PubMed] [Google Scholar]
  • 34.Hoffmann JC, West JL. Three-dimensional photolithographic patterning of multiple bioactive ligands in poly(ethylene glycol) hydrogels. Soft Matter. 2010;6:5056. [Google Scholar]
  • 35.Altrock E, Muth CA, Klein G, Spatz JP, Lee-Thedieck C. The significance of integrin ligand nanopatterning on lipid raft clustering in hematopoietic stem cells. Biomaterials. 2012;33:3107–18. doi: 10.1016/j.biomaterials.2012.01.002. [DOI] [PubMed] [Google Scholar]
  • 36.Temenoff JS, Mikos AG. Injectable biodegradable materials for orthopedic tissue engineering. Biomaterials. 2000;21:2405–12. doi: 10.1016/s0142-9612(00)00108-3. [DOI] [PubMed] [Google Scholar]
  • 37.West JL, Hubbell JA. Polymeric Biomaterials with Degradation Sites for Proteases Involved in Cell Migration. Macromolecules. 1999;32:241–244. [Google Scholar]
  • 38.Hern DL, Hubbell JA. Incorporation of adhesion peptides into nonadhesive hydrogels useful for tissue resurfacing. J Biomed Mater Res. 1998;39:266–76. doi: 10.1002/(sici)1097-4636(199802)39:2<266::aid-jbm14>3.0.co;2-b. [DOI] [PubMed] [Google Scholar]
  • 39.Mann BK, West JL. Cell adhesion peptides alter smooth muscle cell adhesion, proliferation, migration, and matrix protein synthesis on modified surfaces and in polymer scaffolds. J Biomed Mater Res. 2002;60:86–93. doi: 10.1002/jbm.10042. [DOI] [PubMed] [Google Scholar]
  • 40.Burdick JA, Anseth KS. Photoencapsulation of osteoblasts in injectable RGD-modified PEG hydrogels for bone tissue engineering. Biomaterials. 2002;23:4315–23. doi: 10.1016/s0142-9612(02)00176-x. [DOI] [PubMed] [Google Scholar]
  • 41.Burdick JA, Khademhosseini A, Langer R. Fabrication of gradient hydrogels using a microfluidics/photopolymerization process. Langmuir. 2004;20:5153–6. doi: 10.1021/la049298n. [DOI] [PubMed] [Google Scholar]
  • 42.Gonzalez AL, Gobin AS, West JL, McIntire LV, Smith CW. Integrin interactions with immobilized peptides in polyethylene glycol diacrylate hydrogels. Tissue Eng. 2004;10:1775–86. doi: 10.1089/ten.2004.10.1775. [DOI] [PubMed] [Google Scholar]
  • 43.Moon JJ, Lee S-H, West JL. Synthetic biomimetic hydrogels incorporated with ephrin-A1 for therapeutic angiogenesis. Biomacromolecules. 2007;8:42–9. doi: 10.1021/bm060452p. [DOI] [PubMed] [Google Scholar]
  • 44.Saik JE, Gould DJ, Watkins EM, Dickinson ME, West JL. Covalently Immobilized Platelet Derived Growth Factor-BB Promotes Angiogenesis in Biomimetic Poly(ethylene glycol) Hydrogels. Acta biomater. 2010;7:133–143. doi: 10.1016/j.actbio.2010.08.018. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Leslie-Barbick JE, Moon JJ, West JL. Covalently-immobilized vascular endothelial growth factor promotes endothelial cell tubulogenesis in poly(ethylene glycol) diacrylate hydrogels. J Biomater Sci Polym Ed. 2009;20:1763–79. doi: 10.1163/156856208X386381. [DOI] [PubMed] [Google Scholar]
  • 46.DeLong SA, Moon JJ, West JL. Covalently immobilized gradients of bFGF on hydrogel scaffolds for directed cell migration. Biomaterials. 2005;26:3227–34. doi: 10.1016/j.biomaterials.2004.09.021. [DOI] [PubMed] [Google Scholar]
  • 47.Bazzoni G, Carlesso N, Griffin JD, Hemler ME. Bcr/Abl expression stimulates integrin function in hematopoietic cell lines. J Clin Invest. 1996;98:521–8. doi: 10.1172/JCI118820. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Krämer A, Hörner S, Willer A, Fruehauf S, Hochhaus A, Hallek M, Hehlmann R. Adhesion to fibronectin stimulates proliferation of wild-type and bcr/abl-transfected murine hematopoietic cells. Proc Natl Acad Sci USA. 1999;96:2087–92. doi: 10.1073/pnas.96.5.2087. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Cruise GM, Scharp DS, Hubbell JA. Characterization of permeability and network structure of interfacially photopolymerized poly(ethylene glycol) diacrylate hydrogels. Biomaterials. 1998;19:1287–1294. doi: 10.1016/s0142-9612(98)00025-8. [DOI] [PubMed] [Google Scholar]
  • 50.Hahn MS, Miller JS, West JL. Laser Scanning Lithography for Surface Micropatterning on Hydrogels. Adv Mater. 2005;17:2939–2942. [Google Scholar]
  • 51.Cuchiara MP, Allen ACB, Chen TM, Miller JS, West JL. Multilayer microfluidic PEGDA hydrogels. Biomaterials. 2010;31:5491–7. doi: 10.1016/j.biomaterials.2010.03.031. [DOI] [PubMed] [Google Scholar]
  • 52.Erickson HP. Size and shape of protein molecules at the nanometer level determined by sedimentation, gel filtration, and electron microscopy. Biol Proced Online. 2009;11:32–51. doi: 10.1007/s12575-009-9008-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Veronese FM, Mero A. The impact of PEGylation on biological therapies. BioDrugs. 2008;22:315–29. doi: 10.2165/00063030-200822050-00004. [DOI] [PubMed] [Google Scholar]
  • 54.Bailon P, Berthold W. Polyethylene glycol-conjugated pharmaceutical proteins. Pharm Sci Technol Today. 1998;1:352–356. [Google Scholar]
  • 55.Veronese FM. Peptide and protein PEGylation: a review of problems and solutions. Biomaterials. 2001;22:405–17. doi: 10.1016/s0142-9612(00)00193-9. [DOI] [PubMed] [Google Scholar]
  • 56.Roberts MJ, Bentley MD, Harris JM. Chemistry for peptide and protein PEGylation. Adv Drug Delivery Rev. 2002;54:459–76. doi: 10.1016/s0169-409x(02)00022-4. [DOI] [PubMed] [Google Scholar]
  • 57.DiMaggio N, Piccinini E, Jaworski M, Trumpp A, Wendt DJ, Martin I. Toward modeling the bone marrow niche using scaffold-based 3D culture systems. Biomaterials. 2011;32:321–9. doi: 10.1016/j.biomaterials.2010.09.041. [DOI] [PubMed] [Google Scholar]
  • 58.Chua K, Chai C, Lee P, Ramakrishna S, Leong K, Mao H. Functional nanofiber scaffolds with different spacers modulate adhesion and expansion of cryopreserved umbilical cord blood hematopoietic stem/progenitor cells. Exp Hematol. 2007;35:771–781. doi: 10.1016/j.exphem.2007.02.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Chua K-N, Chai C, Lee P-C, Tang Y-N, Ramakrishna S, Leong KW, Mao H-Q. Surface-aminated electrospun nanofibers enhance adhesion and expansion of human umbilical cord blood hematopoietic stem/progenitor cells. Biomaterials. 2006;27:6043–51. doi: 10.1016/j.biomaterials.2006.06.017. [DOI] [PubMed] [Google Scholar]
  • 60.Bagley J, Rosenzweig M, Marks DF, Pykett MJ. Extended culture of multipotent hematopoietic progenitors without cytokine augmentation in a novel three-dimensional device. Exp Hematol. 1999;27:496–504. doi: 10.1016/s0301-472x(98)00053-8. [DOI] [PubMed] [Google Scholar]
  • 61.Ma K, Chan CK, Liao S, Hwang WYK, Feng Q, Ramakrishna S. Electrospun nanofiber scaffolds for rapid and rich capture of bone marrow-derived hematopoietic stem cells. Biomaterials. 2008;29:2096–2103. doi: 10.1016/j.biomaterials.2008.01.024. [DOI] [PubMed] [Google Scholar]
  • 62.Lee-Thedieck C, Spatz JP. Artificial niches: biomimetic materials for hematopoietic stem cell culture. Macromol Rapid Commun. 2012;33:1432–8. doi: 10.1002/marc.201200219. [DOI] [PubMed] [Google Scholar]
  • 63.Holst J, et al. Substrate elasticity provides mechanical signals for the expansion of hemopoietic stem and progenitor cells. Nat Biotechnol. 2010;28:1123–8. doi: 10.1038/nbt.1687. [DOI] [PubMed] [Google Scholar]
  • 64.Saik JE, Gould DJ, Keswani AH, Dickinson ME, West JL. Biomimetic hydrogels with immobilized ephrinA1 for therapeutic angiogenesis. Biomacromolecules. 2011;12:2715–22. doi: 10.1021/bm200492h. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Forsberg EC, Smith-Berdan S. Parsing the niche code: the molecular mechanisms governing hematopoietic stem cell adhesion and differentiation. Haematologica. 2009;94:1477–81. doi: 10.3324/haematol.2009.013730. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Ellis SJ, Tanentzapf G. Integrin-mediated adhesion and stem-cell-niche interactions. Cell Tissue Res. 2010;339:121–30. doi: 10.1007/s00441-009-0828-4. [DOI] [PubMed] [Google Scholar]
  • 67.Garcia AS, Dellatore SM, Messersmith PB, Miller WM. Effects of supported lipid monolayer fluidity on the adhesion of hematopoietic progenitor cell lines to fibronectin-derived peptide ligands for alpha5beta1 and alpha4beta1 integrins. Langmuir. 2009;25:2994–3002. doi: 10.1021/la802772y. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Jensen TW, Hu B-H, Delatore SM, Garcia AS, Messersmith PB, Miller WM. Lipopeptides incorporated into supported phospholipid monolayers have high specific activity at low incorporation levels. J Am Chem Soc. 2004;126:15223–30. doi: 10.1021/ja048684o. [DOI] [PubMed] [Google Scholar]
  • 69.Chollet C, et al. The effect of RGD density on osteoblast and endothelial cell behavior on RGD-grafted polyethylene terephthalate surfaces. Biomaterials. 2009;30:711–20. doi: 10.1016/j.biomaterials.2008.10.033. [DOI] [PubMed] [Google Scholar]
  • 70.Tosatti S, et al. RGD-containing peptide GCRGYGRGDSPG reduces enhancement of osteoblast differentiation by poly(L-lysine)-graft-poly(ethylene glycol)-coated titanium surfaces. J Biomed Mater Res Part A. 2004;68:458–72. doi: 10.1002/jbm.a.20082. [DOI] [PubMed] [Google Scholar]
  • 71.Boateng SY, Lateef SS, Mosley W, Hartman TJ, Hanley L, Russell B. RGD and YIGSR synthetic peptides facilitate cellular adhesion identical to that of laminin and fibronectin but alter the physiology of neonatal cardiac myocytes. Am J Physiol. 2005;288:C30–8. doi: 10.1152/ajpcell.00199.2004. [DOI] [PubMed] [Google Scholar]
  • 72.Vuillet-Gaugler MH, Breton-Gorius J, Vainchenker W, Guichard J, Leroy C, Tchernia G, Coulombel L. Loss of attachment to fibronectin with terminal human erythroid differentiation. Blood. 1990;75:865–73. [PubMed] [Google Scholar]
  • 73.Patel VP, Ciechanover A, Platt O, Lodish HF. Mammalian reticulocytes lose adhesion to fibronectin during maturation to erythrocytes. Proc Natl Acad Sci USA. 1985;82:440–4. doi: 10.1073/pnas.82.2.440. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.Lemoine FM, Dedhar S, Lima GM, Eaves CJ. Transformation-associated alterations in interactions between pre-B cells and fibronectin. Blood. 1990;76:2311–20. [PubMed] [Google Scholar]
  • 75.Verfaillie CM, McCarthy JB, McGlave PB. Differentiation of primitive human multipotent hematopoietic progenitors into single lineage clonogenic progenitors is accompanied by alterations in their interaction with fibronectin. J Exp Med. 1991;174:693–703. doi: 10.1084/jem.174.3.693. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Weinstein R, Riordan Ma, Wenc K, Kreczko S, Zhou M, Dainiak N. Dual role of fibronectin in hematopoietic differentiation. Blood. 1989;73:111–6. [PubMed] [Google Scholar]
  • 77.Lyman SD, Jacobsen SE. c-kit ligand and Flt3 ligand: stem/progenitor cell factors with overlapping yet distinct activities. Blood. 1998;91:1101–34. [PubMed] [Google Scholar]
  • 78.Kovach NL, Lin N, Yednock T, Harlan JM, Broudy VC. Stem cell factor modulates avidity of alpha 4 beta 1 and alpha 5 beta 1 integrins expressed on hematopoietic cell lines. Blood. 1995;85:159–67. [PubMed] [Google Scholar]
  • 79.Long MW, Briddell R, Walter AW, Bruno E, Hoffman R. Human hematopoietic stem cell adherence to cytokines and matrix molecules. J Clin Invest. 1992;90:251–5. doi: 10.1172/JCI115844. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Avraham H, Scadden DT, Chi S, Broudy VC, Zsebo KM, Groopman JE. Interaction of human bone marrow fibroblasts with megakaryocytes: role of the c-kit ligand. Blood. 1992;80:1679–84. [PubMed] [Google Scholar]
  • 81.Dastych J, Metcalfe DD. Stem cell factor induces mast cell adhesion to fibronectin. J Immunol. 1994;152:213–9. [PubMed] [Google Scholar]
  • 82.Bendall LJ, Makrynikola V, Hutchinson A, Bianchi AC, Bradstock KF, Gottlieb DJ. Stem cell factor enhances the adhesion of AML cells to fibronectin and augments fibronectin-mediated anti-apoptotic and proliferative signals. Leukemia. 1998;12:1375–82. doi: 10.1038/sj.leu.2401136. [DOI] [PubMed] [Google Scholar]
  • 83.Lévesque JP, Leavesley DI, Niutta S, Vadas M, Simmons PJ. Cytokines increase human hemopoietic cell adhesiveness by activation of very late antigen (VLA)-4 and VLA-5 integrins. J Exp Med. 1995;181:1805–15. doi: 10.1084/jem.181.5.1805. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84.Doran MR, Markway BD, Aird IA, Rowlands AS, George Pa, Nielsen LK, Cooper-White JJ. Surface-bound stem cell factor and the promotion of hematopoietic cell expansion. Biomaterials. 2009;30:4047–52. doi: 10.1016/j.biomaterials.2009.04.043. [DOI] [PubMed] [Google Scholar]
  • 85.Kishimoto S, et al. Immobilization, stabilization, and activation of human stem cell factor (SCF) on fragmin/protamine microparticle (F/P MP)-coated plates. J Biomed Mater Res Part B. 2010;92:32–9. doi: 10.1002/jbm.b.31486. [DOI] [PubMed] [Google Scholar]
  • 86.Peled A, et al. The chemokine SDF-1 activates the integrins LFA-1, VLA-4, and VLA-5 on immature human CD34(+) cells: role in transendothelial/stromal migration and engraftment of NOD/SCID mice. Blood. 2000;95:3289–96. [PubMed] [Google Scholar]
  • 87.Peled A, et al. The chemokine SDF-1 stimulates integrin-mediated arrest of CD34(+) cells on vascular endothelium under shear flow. J Clin Invest. 1999;104:1199–211. doi: 10.1172/JCI7615. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88.Hidalgo A, et al. Chemokine stromal cell-derived factor-1alpha modulates VLA-4 integrin-dependent adhesion to fibronectin and VCAM-1 on bone marrow hematopoietic progenitor cells. Exp Hematol. 2001;29:345–55. doi: 10.1016/s0301-472x(00)00668-8. [DOI] [PubMed] [Google Scholar]
  • 89.Brakebusch C, Fässler R. The integrin-actin connection, an eternal love affair. EMBO J. 2003;22:2324–33. doi: 10.1093/emboj/cdg245. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90.Hersel U, Dahmen C, Kessler H. RGD modified polymers: biomaterials for stimulated cell adhesion and beyond. Biomaterials. 2003;24:4385–4415. doi: 10.1016/s0142-9612(03)00343-0. [DOI] [PubMed] [Google Scholar]
  • 91.Tan BL, Yazicioglu MN, Ingram D, McCarthy J, Borneo J, Williams DA, Kapur R. Genetic evidence for convergence of c-Kit- and alpha4 integrin-mediated signals on class IA PI-3kinase and the Rac pathway in regulating integrin-directed migration in mast cells. Blood. 2003;101:4725–32. doi: 10.1182/blood-2002-08-2521. [DOI] [PubMed] [Google Scholar]
  • 92.Schofield KP, Rushton G, Humphries MJ, Dexter TM, Gallagher JT. Influence of interleukin-3 and other growth factors on alpha4beta1 integrin-mediated adhesion and migration of human hematopoietic progenitor cells. Blood. 1997;90:1858–66. [PubMed] [Google Scholar]
  • 93.Nilsson G, Butterfield JH, Nilsson K, Siegbahn A. Stem cell factor is a chemotactic factor for human mast cells. J Immunol. 1994;153:3717–23. [PubMed] [Google Scholar]
  • 94.Voermans C, Anthony EC, Mul E, Van der Schoot E, Hordijk P. SDF-1α induced actin polymerization and migration in human hematopoietic progenitor cells. Exp Hematol. 2001;29:1456–64. doi: 10.1016/s0301-472x(01)00740-8. [DOI] [PubMed] [Google Scholar]
  • 95.Peled A, et al. Dependence of human stem cell engraftment and repopulation of NOD/SCID mice on CXCR4. Science. 1999;283:845–8. doi: 10.1126/science.283.5403.845. [DOI] [PubMed] [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Supplementary Materials

01

RESOURCES