We have shown that stromal O-sulfated heparan sulfate glycosaminoglycans (O-S-GAGs) regulate primitive human hematopoietic progenitor cell (HPC) growth and differentiation by colocalizing heparin-binding cytokines and matrix proteins with HPC in stem cell “niches” in the marrow microenvironment. We now show that long-term culture-initiating cells (LTC-IC) are maintained for 5 weeks in the absence of stroma when O-S-GAGs are added to IL-3 and either MIP-1 or PF4 (LTC-IC maintenance without GAGs, 32 ± 2%; with GAGs, 95 ± 7%; P < .001). When cultured with 5 additional cytokines, O-S-GAGs, IL-3, and MIP-1, LTC-IC expanded 2- to 4-fold at 2 weeks, and 92 ± 8% LTC-IC were maintained at 5 weeks. Similar results were seen when PF4 replaced MIP-1. Although O-S-GAG omission did not affect 2-week expansion, only 20% LTC-IC were maintained for 5 weeks. When O-S-heparin was replaced by completely desulfated-, N-sulfated (O-desulfated), or unmodified heparins, LTC-IC maintenance at week 5 was not better than with cytokines alone. Unmodified- and O-S-heparin, but not desulfated- or N-sulfated heparin, bound to MIP-1, IL-3, PF4, VEGF, thrombospondin, and fibronectin. However, the affinity of heparin for thrombospondin and PF4, and the association and dissociation rates of heparin for PF4, were higher than those of O-S-heparin. We conclude that (i) although cytokines may suffice to induce early expansion, adult human LTC-IC maintenance for longer than 1 month requires O-S-GAGs, and (ii) HPC support may depend not only on the ability of GAGs to bind proteins, but also on optimal affinity and kinetics of interactions that affect presentation of proteins in a biologically active manner to progenitors. (Blood. 2000;95:147-155)

Stem cells may exist in “stem cell niches” in the marrow microenvironment in vivo, where stromal cells, specific extracellular matrix (ECM) components, and cytokines bound to stromal cell surfaces or to ECM macromolecules provide the appropriate balance of signals that preserves the stem cell pool while permitting controlled proliferation and differentiation.1-4 A number of cytokines, chemokines, and matrix components considered to be essential for the localization, conservation, and proliferation of primitive hematopoietic progenitors cells (HPC) are heparin-binding proteins, including platelet factor 4 (PF4), basic fibroblast growth factor (bFGF), hepatocyte growth factor (HGF), interleukin-3 (IL-3), vascular endothelial growth factor (VEGF), macrophage inflammatory protein-1α (MIP-1α), thrombospondin (TSP), and fibronectin (FN).5-20 Specific sulfation patterns of heparan sulfate (HS) are required for binding and modulation of the activity of cytokines that have activity on hematopoietic progenitors.6,8,21,22 Additionally, the ability of HS to modulate cytokine activity on target cells may depend on the affinity and kinetics of association (“on” rate) between cytokines and HS.23 O-sulfation of glycosaminoglycans (GAGs) is required for binding of several growth factors, including PF4 and platelet-derived growth factor.24,25 We have recently shown that highly O-sulfated stromal cell-derived HS GAGs help to mediate the formation of such “stem cell niches” by colocalizing specific heparin-binding proteins with HPC, thereby orchestrating the controlled growth and differentiation of stem cells.26 

Of the numerous cytokines that modulate human HPC survival and proliferation in vitro, the heparin-binding cytokine IL-3 and chemokines MIP-1α or PF4 are particularly critical. We have recently demonstrated that the human long-term culture-initiating cell (LTC-IC)-maintaining capability of stromal supernatant27,28is improved by the addition of nanogram concentrations of IL-3 + MIP-1α.29 In these stromal supernatant cultures, MIP-1α can be replaced by PF4,30 a GAG-binding chemokine that has effects similar to MIP-1α on hematopoietic progenitors.31Moreover, IL-3 and MIP-1α are required for the ex vivo survival of human HPC with myeloid and lymphoid differentiation potential.29,32,33 HS proteoglycans may be essential for the localization of both MIP-1α and IL-3 within the marrow stromal microenvironment9,26,34-36 and may interact with IL-3 to augment adhesion and colocalization of HPC within the microenvironment.37 

Thrombospondin is a matrix protein present in serum, which is synthesized by both hematopoietic and stromal cells, which directly mediates the adhesion of HPC.38 TSP in soluble or immobilized form also modulates cytokine activity, synergizing with stem cell factor (SCF) to stimulate HPC adhesion and proliferation but inhibiting IL-3–mediated HPC proliferation.5 TSP has a high affinity for binding to heparin and interacts with stromal HS proteoglycans to increase HPC adhesion.37 HS GAGs also influence the growth-modulatory activity of TSP, because low molecular weight heparin neutralizes TSP-mediated inhibition of multilineage colony-forming units-granulocyte, erythroid, macrophage, megakaryocyte (CFU-GEMM) and CFU-megakaryocyte (CFU-MK) growth that is induced by cytokines including IL-3, SCF, granulocyte-macrophage colony-stimulating factor (GM-CSF), bFGF, and thrombopoietin.39 

We hypothesized that in the absence of stroma, specifically sulfated HS GAGs are required for IL-3 + chemokine (MIP-1α or PF4)-mediated LTC-IC maintenance and that this biological activity is dependent on optimal binding characteristics of the GAGs for early-acting chemokines (eg, PF4) and matrix components (eg, TSP). In the present studies, we have examined the ability of purified, differentially sulfated heparin GAGs to support IL-3 + chemokine-mediated LTC-IC maintenance and the binding characteristics of these GAGs for cytokines and matrix components essential for HPC localization and survival.

Cell separation

Bone marrow was aspirated in preservative-free heparin from the posterior iliac crest of healthy young volunteers after informed consent. Bone marrow mononuclear cells were separated by Ficoll-Hypaque centrifugation (s.g., 1.077), and CD34+ enriched cells were obtained using Ceprate® LC CD34 selection columns (CellPro Inc, Bothell, WA). CD34+/HLA-DRcells were obtained by flow cytometry, using a FACStar Plus® flow cytometry system equipped with a Consort 32 computer as previously described.26,34 

Stromal feeders and conditioned media

Human primary bone marrow stromal feeders were established from human bone marrow mononuclear cells as previously described, irradiated at 1250 rad when confluent, and maintained in long-term bone marrow culture (LTBMC) medium.26,34 Supernatant from irradiated stromal cultures was harvested 2 to 3 days after a half-medium change, centrifuged to remove cell debris, and frozen at −70°C until use.

Cytokines

Recombinant human cytokines used in long-term cultures included 500 pg/mL granulocyte colony-stimulating factor (G-CSF) (Neupogen®; Amgen, Thousand Oaks, CA), 50 pg/mL GM-CSF (Immunex Corp, Seattle, WA), 200 pg/mL SCF (a kind gift from Amgen), 50 pg/mL leukemia inhibitory factor (LIF; R & D Systems Inc, Minneapolis, MN), 200 pg/mL MIP-1α (R & D Systems) and 2 ng/mL IL-6 (a kind gift from Dr G. Wong, Genetics Institute).

As indicated, medium in some long-term cultures was supplemented with G-CSF, GM-CSF, SCF, LIF, and IL-6 in the same concentrations as described above, along with 5 ng/mL IL-3 (a kind gift from Dr G. Wong) and either 10 ng/mL MIP-1α or 200 ng/mL PF4 (purified from human platelets).

Glycosaminoglycans

HS, chondroitin sulfate, and heparin were obtained from Sigma (St. Louis, MO). The differentially sulfated heparins used in this study, derived from the same parent unmodified heparin molecule by N-desulfation and N-reacetylation (O-sulfated heparin), N- and O-desulfation and N-reacetylation (completely desulfated heparin), or N- and O-desulfation and N-resulfation, were obtained from Seikagaku America (Falmouth, MA). Disaccharide compositional analysis of the unmodified heparin and differentially sulfated heparins (Seikagaku America, personal communication) is shown in the Table. The completely desulfated heparin was largely composed of desulfated disaccharides. The O-sulfated heparin was largely composed of O-disulfated disaccharides {-UA(2-OSO3)-GlcNAc(6-OSO3)-} and 6-O-sulfated disaccharides {-UA-GlcNAc(6-OSO3)-} (where the UA was mainly iduronic acid) and contained only trace amounts (<1%) of the trisulfated disaccharides {-IdoA (2-OSO3)-GlcNS (6-OSO3)-}, whereas unmodified heparin was mainly composed of trisulfated disaccharides, with a smaller proportion of O-disulfated units. The N-sulfated heparin was depleted of O-sulfated units (not shown). For several growth factors, the regions of transition between highly sulfated GlcNS-containing blocks and unsulfated GlcNAc-containing blocks are believed to be important for binding and biological activity of HS. Because of the specificity of the sulfotransferase enzymes, the terminal GlcNAc (adjacent to GlcNS) in such blocks may be 6-O-sulfated in such regions.40,41 Disaccharides at such transition points thus are 6-sulfated, with or without 2-O sulfation of the hexuronic acid: Most disaccharides in O-sulfated heparin are identical to such units and more closely resemble sulfation patterns of HS rather than heparin.

Disaccharide composition of unmodified and desulfated heparins

Sulfated Disaccharide Positions of Sulfation on Disaccharides Percent of Total Disaccharides
O-sulfated Heparin Unmodified Heparin
UA-GlcNAc-  None  6.8  3.8  
-UA-GlcNSO3N  1.1  2.2  
-UA-GlcNAc(6-OSO3)-  10.8  3.1 
-UA(2-OSO3)-GlcNAc- 12.4 1.9 
-UA(2-OSO3)-GlcNSO32, N  0.8  9.1 
-UA(2-OSO3)-GlcNAc(6-OSO3)-  2, 6 67.3  8.1 
-UA(2-OSO3)-GlcNSO3(6-OSO3)- 2, N, 6  0.8 71.7 
Sulfated Disaccharide Positions of Sulfation on Disaccharides Percent of Total Disaccharides
O-sulfated Heparin Unmodified Heparin
UA-GlcNAc-  None  6.8  3.8  
-UA-GlcNSO3N  1.1  2.2  
-UA-GlcNAc(6-OSO3)-  10.8  3.1 
-UA(2-OSO3)-GlcNAc- 12.4 1.9 
-UA(2-OSO3)-GlcNSO32, N  0.8  9.1 
-UA(2-OSO3)-GlcNAc(6-OSO3)-  2, 6 67.3  8.1 
-UA(2-OSO3)-GlcNSO3(6-OSO3)- 2, N, 6  0.8 71.7 

Derived from data from Seikagaku America, Inc (personal communication).

Highly (“default”) sulfated disaccharide; UA indicates hexuronic acid (iduronic acid or glucuronic acid); GlcN, glucosamine.

The GAGs were added to long-term cultures at a final concentration of 5 μg/mL; we have previously shown this concentration to be in the optimal range for LTC-IC maintenance.26 

Preparation of synthetic proteoglycan-like molecules

GAGs and either ovalbumin or human albumin were dissolved (2 mg each) in 1 mL distilled water, and 20 mg of 1-ethyl-3-(3-dimethylaminopropyl)-carbodiimide (EDC) in 50 μL water was added (all chemicals from Sigma). The mixture was shaken at 4°C for 12 hours to couple GAGs to ovalbumin or albumin via the carboxyl groups by amide bonds.42 Low molecular weight by-products were removed by dialysis in 12 000-14 000 molecular weight cut-off (MWCO) dialysis tubing against phosphate-buffered saline (PBS). For use in cultures, the protein-coupled GAGs (final concentration 5 μg/mL) were dissolved in fresh LTBMC medium and sterile-filtered.

Long-term cultures

LTBMC medium.

The medium consisted of Iscove's modified Dulbecco's medium with 12.5% fetal calf serum, 12.5% horse serum, 2 mM L-glutamine, 1000 U/mL penicillin, 100 U/mL streptomycin, and 10−6mol/L hydrocortisone.43 

Conditioned medium cultures.

A total of 10 × 103 to 14 × 103DR cells were suspended in a transwell insert with a microporous 0.4-μm membrane placed in stroma-free wells of 6- or 24-well plates. Media in the lower well were replaced 5 times weekly with 3 mL (for 6-well plates) or 0.8 mL (for 24-well plates) fresh unsupplemented LTBMC medium, LTBMC medium supplemented with cytokines alone or in combination with cell line–derived or artificially synthesized PGs or GAG chains, or conditioned medium from M2-10B4 or human stromal feeders. After 2 or 5 weeks of culture, progeny of DR cells were recovered from the transwells by vigorous washing and counted on a hemocytometer. Cells were plated in methylcellulose cultures to enumerate colony forming cells (CFC) and in limiting dilution analysis to determine the absolute number of LTC-IC present, as described.28,44 All cultures were maintained at 37°C in humidified 5% CO2 atmosphere.

Short-term methylcellulose cultures.

Day 0 DR cells and the progeny of DR cells recovered at 2 or 5 weeks from long-term cultures were plated in methylcellulose-containing medium supplemented with 30% fetal calf serum (FCS), 3 IU/mL erythropoietin, and 5 ng/mL IL-3 and CFC enumerated at day 14. All cultures were maintained at 37°C in humidified 5% CO2 atmosphere.

Enumeration of the absolute number of LTC-IC by limited dilution analysis.

Day 0 DR cells (22 replicates per concentration) or progeny of DR cells recovered from long-term cultures were plated at limiting dilutions onto irradiated M2-10B4 feeders in 96-well plates, as described.26,28,34 Cultures were fed by weekly replacement of 75 μL fresh LTBMC medium. After 5 weeks, all the medium was removed and the wells overlaid with methylcellulose-containing medium supplemented with 30% FCS, 3 IU/mL erythropoietin, and 5 ng/mL IL-3. All cultures were maintained at 37°C in humidified 5% CO2 atmosphere. Wells were scored for the presence or absence of secondary CFC at day 14. The absolute number of LTC-IC was calculated as the reciprocal of the concentration of test cells that yields 37% negative wells, using Poisson statistics and the weighted mean method.28,44 

Iodination of differentially sulfated heparins

Heparins were coupled with tyramine14,15 and were iodinated using carrier-free Na2125I (Amersham, Arlington Heights, IL) and the Iodo-gen iodination reagent (Pierce, Rockford, IL).14 Free 125I was removed from the labeled HS by separation on a Sephadex G-25 column, followed by exhaustive dialysis. The specific activity was 5 to 70 μCi/μg for the iodinated GAGs.

Affinity coelectrophoresis (ACE)

Binding of iodinated heparins to cytokines including IL-3 (R & D Systems), MIP-1α, bFGF (R & D Systems), and VEGF (isoforms VEGF165 and VEGF121; kind gifts from Dr S. Ramakrishnan, University of Minnesota) and to ECM molecules including TSP (a kind gift from Dr Robert Hebbel, University of Minnesota) and FN (Sigma) was examined using ACE. ACE was performed as described.26,45 Briefly, a 1% agarose gel (low melting point Seaplaque; FMC, Rockland, MD) in 50 mM sodium MOPSO (Sigma), pH 7.0; 125 mM NaCl; 0.5% CHAPS buffer was cast with a strip well (for HS) and a perpendicular 8-lane comb (for proteins). Protein samples (TSP, bFGF or IL-3) prepared at twice the desired concentration in MOPSO/NaCl/CHAPS electrophoresis buffer were mixed with an equal volume of 2% agarose and allowed to gel in appropriate wells created by the 8-lane comb. The heparin preparation in MOPSO/NaCl/CHAPS electrophoresis buffer containing bromophenol blue and sucrose was added to the strip well. Electrophoresis was performed in a Hoefer electrophoresis apparatus in MOPSO/NaCl running buffer prepared without CHAPS at 330 mA, 45 to 60 V for 2.5 to 2.75 hours at 20°C to 25°C. Gels were air-dried and autoradiographed at −80°C. For VEGF, FN, and MIP-1α, the gels were cast in 50 mM MOPSO, pH 7.0; 50 mM sodium acetate (NaAc). Protein samples and the heparins were also prepared in the same buffer. Electrophoresis was performed as above, for 2 to 2.5 hours.

Determination of half-maximal binding (EC50).

125I-labeled heparin or O-sulfated heparin was electrophoresed through varying concentrations of TSP (1-100 nM) in the affinity coelectrophoresis system. The retardation coefficient45 was calculated as R = (M0 − M)/M0, where M0 was the mobility of free GAGs (taken as mobility in presence of ovalbumin), and M was the mobility of GAGs through the TSP-containing lanes. Mobility was measured as the distance from the loading well at the top of the gel to the upper end (most retarded) of the GAG smear. Because retardation is a function of the binding affinity of the GAG to TSP, a plot of retardation (binding) versus concentration of TSP was used to calculate half-maximal binding concentration, as a measure of the affinity of the 2 GAGs for TSP.

Surface plasmon resonance.

The binding of GAGs to PF4 was examined on a BIAcore®biosensor equipment (Pharmacia, Uppsala, Sweden) using surface plasmon resonance, which is highly sensitive and monitors real-time binding of an analyte (GAG) to a ligand (PF4) immobilized on a sensor chip.46,47 This is measured in resonance units (RU), which correlates with the amount of analyte bound (1 RU = 1 pg/mm2). Binding of GAGs in solution to immobilized PF4 was examined as described.26 The KD was calculated using BIAevaluation 2.1 software (Pharmacia Biosensor, Uppsala, Sweden) from the average of ka (association rate, “run-on” phase) and kd (dissociation rate, “wash-out” phase) kinetics: KD = av. kd/ka (ks).

Statistics.

Results of data are reported as the mean ± SEM. Levels of significance were determined by the 2-sided Student's t test.

HS in combination with IL-3 and MIP-1 supports long-term maintenance of LTC-IC

In stroma-free cultures supplemented with IL-3 + MIP-1α, only 22 ± 1% of LTC-IC were recovered after 5 weeks (P < .001 vs day 0 LTC-IC frequency; Figure1), even though 89 ± 3% of day-0 LTC-IC were maintained for 2 weeks. In cultures where 5 μg/mL HS was added together with IL-3 + MIP-1α, 121 ± 10% of input numbers of LTC-IC were maintained for up to 5 weeks (P = .002 vs cultures with IL-3 + MIP-1α alone). A lower concentration of HS (1 μg/mL) was less effective (LTC-IC maintenance 73 ± 5% at 5 weeks; P = .001 vs 5 μg/mL HS). High concentrations of GAGs (20 μg/mL HS) inhibited cell proliferation and LTC-IC maintenance (data not shown). Further, maintenance of LTC-IC at week 5 in HS + IL-3 + MIP-1α cultures was similar to that seen in cultures supplemented with stroma-conditioned medium + IL-3 + MIP-1α (134 ± 12%). LTC-IC expanded 4 ± 0.8-fold at 2 weeks in stroma-conditioned medium-supplemented cultures, whereas LTC-IC expansion in HS + IL-3 + MIP-1α cultures was only 1.3 ± 0.1-fold at week 2. These results suggest that, in the presence of HS, long-term in vitro maintenance of LTC-IC can be obtained by addition of a single growth-promoting cytokine (IL-3) and a single growth-inhibitory chemokine (MIP-1α). LTC-IC expansion, in contrast, may require the presence of additional cytokines and/or factors present in stroma-conditioned medium.

Fig. 1.

LTC-IC maintenance in the presence of HS with IL-3 and MIP-1.

A total of 10 000 to 20 000 DR cells were plated in 0.4-μm transwell inserts in 6-well tissue culture clusters. Medium in the lower chambers of the wells was replaced daily by either stroma-conditioned medium (Stroma CM) or long-term bone marrow culture (LTBMC) medium supplemented with or without 5 μg/mL HS. A total of 5 ng/mL IL-3 and 10 ng/mL MIP-1α were added to all cultures, including Stroma CM. Cultures were harvested after 2 weeks (A) or 5 weeks (B) and cells replated at limiting dilutions for estimation of LTC-IC frequency, as described in Methods. Numbers within the bars indicate the number of experiments. Comparison between LTBMC medium only and other conditions: *P < .005.

Fig. 1.

LTC-IC maintenance in the presence of HS with IL-3 and MIP-1.

A total of 10 000 to 20 000 DR cells were plated in 0.4-μm transwell inserts in 6-well tissue culture clusters. Medium in the lower chambers of the wells was replaced daily by either stroma-conditioned medium (Stroma CM) or long-term bone marrow culture (LTBMC) medium supplemented with or without 5 μg/mL HS. A total of 5 ng/mL IL-3 and 10 ng/mL MIP-1α were added to all cultures, including Stroma CM. Cultures were harvested after 2 weeks (A) or 5 weeks (B) and cells replated at limiting dilutions for estimation of LTC-IC frequency, as described in Methods. Numbers within the bars indicate the number of experiments. Comparison between LTBMC medium only and other conditions: *P < .005.

Close modal

Cytokines alone induce short-term expansion of LTC-IC, but O-sulfated GAGs are required for long-term LTC-IC maintenance

We have demonstrated that stroma-conditioned medium can be replaced by a combination of low-dose cytokines (CK) + HS.34 We therefore cultured DR cells for 2 to 5 weeks in stroma-free cultures supplemented with IL-3 (5 ng/mL) and MIP-1α (10 ng/mL) with or without a combination of CK in the concentrations detected in stroma-conditioned medium and with or without bovine kidney HS. The combination of IL-3 + MIP-1α + CK but no HS induced a 2 ± 0.5-fold expansion of LTC-IC at 2 weeks (Figure2). However, the majority of LTC-IC was lost at 5 weeks (LTC-IC recovery 32 ± 2%). The addition of HS to the combination of IL-3 + MIP-1α + CK resulted in a 2.8 ± 0.6-fold expansion of LTC-IC at 2 weeks. In contrast to IL-3 + MIP-1α + CK, cultures supplemented with IL-3 + MIP-1α + CK + HS resulted in long-term maintenance of a significantly greater proportion of LTC-IC (76 ± 7%) at 5 weeks (P < .001 vs IL-3 + MIP-1α + CK cultures).

Fig. 2.

Modulation of LTC-IC maintenance by differentially sulfated GAGs in the presence of multiple growth-promoting cytokines with IL-3 and MIP-1.

A total of 10 000 to 20 000 DR cells were plated in 0.4-μm transwell inserts in 6-well tissue culture clusters. Medium in the lower chambers of the wells was replaced daily by either stroma-conditioned medium (Stroma CM) or by LTBMC medium supplemented with or without a combination of cytokines (500 pg/mL G-CSF, 50 pg/mL GM-CSF, 200 pg/mL SCF, 50 pg/mL LIF, and 2 ng/nl IL-6) and with or without 5 μg/mL each of GAGs. A total of 5 ng/mL IL-3 and 10 ng/mL MIP-1α were added to all cultures, including Stroma CM. Cultures were harvested after 2 weeks (A) or 5 weeks (B). The equivalent of 500 to 1000 DR cells plated at day 0 were replated after harvesting in methylcellulose cultures for estimation of CFC generation, and the remaining cells were replated at limiting dilutions for estimation of LTC-IC frequency, as described in Methods. Numbers within the bars indicate the number of experiments. Comparison between cytokines only and other conditions: *P < .002; comparison between desulfated heparin and other conditions: §P < .001.

Fig. 2.

Modulation of LTC-IC maintenance by differentially sulfated GAGs in the presence of multiple growth-promoting cytokines with IL-3 and MIP-1.

A total of 10 000 to 20 000 DR cells were plated in 0.4-μm transwell inserts in 6-well tissue culture clusters. Medium in the lower chambers of the wells was replaced daily by either stroma-conditioned medium (Stroma CM) or by LTBMC medium supplemented with or without a combination of cytokines (500 pg/mL G-CSF, 50 pg/mL GM-CSF, 200 pg/mL SCF, 50 pg/mL LIF, and 2 ng/nl IL-6) and with or without 5 μg/mL each of GAGs. A total of 5 ng/mL IL-3 and 10 ng/mL MIP-1α were added to all cultures, including Stroma CM. Cultures were harvested after 2 weeks (A) or 5 weeks (B). The equivalent of 500 to 1000 DR cells plated at day 0 were replated after harvesting in methylcellulose cultures for estimation of CFC generation, and the remaining cells were replated at limiting dilutions for estimation of LTC-IC frequency, as described in Methods. Numbers within the bars indicate the number of experiments. Comparison between cytokines only and other conditions: *P < .002; comparison between desulfated heparin and other conditions: §P < .001.

Close modal

We have recently shown that chemically differentially sulfated heparins have different LTC-IC supportive capabilities, such that LTC-IC maintenance is supported by O-sulfated heparin in combination with low concentrations of CK but not by desulfated-, N-sulfated- or unmodified-heparin.26 We therefore examined the effect of completely desulfated and O-sulfated heparins on IL-3 + MIP-1α–mediated LTC-IC maintenance (Figure 2). Addition of O-sulfated heparin to IL-3 + MIP-1α + CK resulted in 1.7 ± 0.2-fold LTC-IC expansion at week 2, which was lower than with HS. However, 95 ± 7% of LTC-IC were maintained at week 5. When desulfated heparin was combined with IL-3 + MIP-1α + CK, 2.5 ± 0.4-fold LTC-IC expansion was seen at week 2, but LTC-IC recovery at week 5 (23 ± 6%) was no better than with cytokines alone.

When PF4 (200 ng/mL) was substituted for MIP-1α (10 ng/mL) in stroma-free cultures, LTC-IC maintenance was similar to that seen in the presence of MIP-1α (Figure 3). Most of the LTC-IC were maintained in cultures supplemented with IL-3 + PF4 + CK in the presence of either HS or O-sulfated heparin (LTC-IC supportive GAGs) but not in the presence of desulfated heparin (LTC-IC nonsupportive GAG). Thus, both the heparin-binding chemokines MIP-1α and PF4 require the sequences present in O-sulfated HS to support LTC-IC maintenance.

Fig. 3.

Efficacy of differentially sulfated GAGs to modulate LTC-IC maintenance in the presence of PF4.

A total of 10 000 to 20 000 DR cells were plated in 0.4-μm transwell inserts in 6-well tissue culture clusters. Medium in the lower chambers of the wells was replaced daily by LTBMC medium supplemented with or without a combination of cytokines (500 pg/mL G-CSF, 50 pg/mL GM-CSF, 200 pg/mL SCF, 50 pg/mL LIF, 2 ng/mL IL-6, and 200 pg/mL MIP-1α) with 5 ng/mL IL-3 and 200 ng/mL PF4 and with or without 5 μg/mL each of GAGs. Cultures were harvested after 5 weeks and replated at limiting dilutions for estimation of LTC-IC frequency, as described in Methods. Numbers within the bars indicate the number of experiments. Comparison between day 0 and other conditions: *P < .002.

Fig. 3.

Efficacy of differentially sulfated GAGs to modulate LTC-IC maintenance in the presence of PF4.

A total of 10 000 to 20 000 DR cells were plated in 0.4-μm transwell inserts in 6-well tissue culture clusters. Medium in the lower chambers of the wells was replaced daily by LTBMC medium supplemented with or without a combination of cytokines (500 pg/mL G-CSF, 50 pg/mL GM-CSF, 200 pg/mL SCF, 50 pg/mL LIF, 2 ng/mL IL-6, and 200 pg/mL MIP-1α) with 5 ng/mL IL-3 and 200 ng/mL PF4 and with or without 5 μg/mL each of GAGs. Cultures were harvested after 5 weeks and replated at limiting dilutions for estimation of LTC-IC frequency, as described in Methods. Numbers within the bars indicate the number of experiments. Comparison between day 0 and other conditions: *P < .002.

Close modal

The generation of CFC from primitive progenitors is induced by cytokines and does not require GAGs

A 2-fold expansion in the numbers of CFC generated was sustained for 2 to 5 weeks in cultures supplemented with IL-3 + MIP-1α (Figure4). The addition of HS to IL-3 + MIP-1α did not affect CFC numbers. However, CFC generation was significantly higher in the presence of stroma-conditioned medium + IL-3 + MIP-1α at both 2 and 5 weeks (P < .001 and P = .02, respectively, vs HS + IL-3 + MIP-1α). This suggests that the increased CFC generation in the latter condition is due to the presence of low concentrations of cytokines present in stroma-conditioned medium. Indeed, when low-dose cytokines (known to be present in stroma-conditioned medium) were added to IL-3 + MIP-1α in stroma-free cultures, the generation of CFC at 2 to 5 weeks became comparable to that seen in the presence of stroma-conditioned medium + IL-3 + MIP-1α, although the size of the colonies generated in the presence of stroma-conditioned medium was significantly larger. This may be due to the presence in stroma-conditioned medium of additional cytokines, such as flt-3 ligand, IL-11, and thrombopoietin, which were not added to the stroma-free cultures. Overall, the presence of CK + IL-3 + MIP-1α induced a 10- to12-fold increase in CFC at 2 weeks and 6-fold increase in CFC at 5 weeks, compared to CFC present in the input population at day 0. CFC numbers were not affected by the further addition of HS, O-sulfated heparin, or desulfated heparin to CK + IL-3 + MIP-1α.

Fig. 4.

Generation of CFC.

Number of CFC in the starting DR population at day 0 and after 2 weeks (A) or 5 weeks (B) in culture. All cultures were supplemented with 5 ng/mL IL-3 and 10 ng/mL MIP-1α. The indicated cultures were additionally supplemented with the low-dose cytokine combination. Numbers within the bars indicate the number of experiments. Comparison between HS and Stroma CM: *P < .001 (at 2 weeks); §P = .02 (at 5 weeks).

Fig. 4.

Generation of CFC.

Number of CFC in the starting DR population at day 0 and after 2 weeks (A) or 5 weeks (B) in culture. All cultures were supplemented with 5 ng/mL IL-3 and 10 ng/mL MIP-1α. The indicated cultures were additionally supplemented with the low-dose cytokine combination. Numbers within the bars indicate the number of experiments. Comparison between HS and Stroma CM: *P < .001 (at 2 weeks); §P = .02 (at 5 weeks).

Close modal

As for CFC generation, the addition of HS or O-sulfated heparin to IL-3 + MIP-1α + CK did not affect cell expansion (the total numbers of terminally differentiated mature cells present in culture) at 2 to 5 weeks (data not shown). Cell expansion in cultures supplemented with IL-3 + MIP-1α + CK was comparable to that seen in cultures supplemented with stroma-conditioned medium + IL-3 + MIP-1α (500- to 800-fold expansion at 5 weeks).

Cytokine-binding profiles of hematopoietically supportive GAGs

Because O-sulfated heparin but not the other modified heparins or unmodified heparin supported LTC-IC, we next examined the cytokine- and matrix component–binding capabilities of these heparins to examine if differences in this capability correlated with their ability to support LTC-IC maintenance. Desulfated heparin and N-sulfated heparin, which did not support LTC-IC maintenance, showed no binding to MIP-1α, IL-3, VEGF, FN, or TSP by ACE (Figure 5). N-sulfated heparin, but not desulfated heparin, bound bFGF to a small extent. O-sulfated heparin, which supports LTC-IC maintenance, bound to IL-3, bFGF, FN, TSP and, to a lesser extent, to VEGF and MIP-1α. Unmodified heparin, which possesses a high degree of both N- and O-sulfation, also bound to all these proteins. We have already shown,26 using surface plasmon resonance, that O-sulfated heparin and unmodified heparin, but not desulfated heparin or N-sulfated heparin, bind PF4.

Fig. 5.

Binding of modified heparins to proteins by affinity coelectrophoresis (ACE).

125I-labeled GAGs (HEP, unmodified heparin; De-S HEP, completely desulfated heparin; N-S HEP, N-sulfated heparin; O-S HEP, O-sulfated heparin) were electrophoresed through the proteins (cytokines and matrix components) cast in agarose gels. Results are shown for binding of the GAGs to 100 nM TSP, 25 nM bFGF, and 500 nM each of VEGF, FN, IL-3, and MIP-1α. Ovalbumin (500 nM) was used as a negative control protein. The NaCl electrophoresis buffer was used for TSP, bFGF, and IL-3, and the sodium acetate buffer was used for VEGF, FN, and MIP-1α. Migration of unbound GAGs that are not bound to proteins is dependent on both size and charge. The average size of all 4 GAGs was comparable (8 to 12 kd). Because the negative charge on De-S HEP is the least, its migration was slowest. N-S HEP has an intermediate charge and migrated faster than De-S HEP. O-S HEP and HEP, which are the most highly charged, migrated at comparable rates. Binding to proteins was seen as retardation of the relative rate of migration of the GAGs through the protein lanes, compared to its migration through ovalbumin.

Fig. 5.

Binding of modified heparins to proteins by affinity coelectrophoresis (ACE).

125I-labeled GAGs (HEP, unmodified heparin; De-S HEP, completely desulfated heparin; N-S HEP, N-sulfated heparin; O-S HEP, O-sulfated heparin) were electrophoresed through the proteins (cytokines and matrix components) cast in agarose gels. Results are shown for binding of the GAGs to 100 nM TSP, 25 nM bFGF, and 500 nM each of VEGF, FN, IL-3, and MIP-1α. Ovalbumin (500 nM) was used as a negative control protein. The NaCl electrophoresis buffer was used for TSP, bFGF, and IL-3, and the sodium acetate buffer was used for VEGF, FN, and MIP-1α. Migration of unbound GAGs that are not bound to proteins is dependent on both size and charge. The average size of all 4 GAGs was comparable (8 to 12 kd). Because the negative charge on De-S HEP is the least, its migration was slowest. N-S HEP has an intermediate charge and migrated faster than De-S HEP. O-S HEP and HEP, which are the most highly charged, migrated at comparable rates. Binding to proteins was seen as retardation of the relative rate of migration of the GAGs through the protein lanes, compared to its migration through ovalbumin.

Close modal

However, the binding profile of O-sulfated heparin was different than that of heparin. At the concentrations examined, unmodified heparin bound homogeneously to TSP, bFGF, VEGF, and FN, whereas O-sulfated heparin bound heterogeneously to these proteins, resulting in a smear in the protein lanes. Binding of both heparin and O-sulfated heparin to IL-3 was seen as a smear. When labeled heparin was eluted from the upper and lower portions of the smear and reelectrophoresed into a separate IL-3–containing gel (see ref. 14 for method), labeled heparin from the upper portion bound to IL-3, whereas that from the lowest portion did not bind to IL-3 (data not shown). Thus, the uppermost portion of the smear (most retarded) is indicative of the most avidly bound subset of labeled molecules in the O-sulfated heparin, whereas the lowest portion of the smear (least retarded) is indicative of unbound O-sulfated heparin molecules, because it migrated at the same rate as O-sulfated heparin in the ovalbumin lane. Thus, O-sulfated heparin may contain GAG chains that bind to proteins over a range of affinities, unlike the highly sulfated unmodified heparin, and suggests that at least part of the binding requirements are provided by N-sulfate groups.

The VEGF165 isoform contains a heparin-binding domain in exon 7. VEGF121 isoform lacks this exon.18,48,49 Both O-sulfated heparin and unmodified heparin bound to VEGF165 but not to VEGF121. This indicates that the binding of both GAGs to VEGF165, as seen on ACE, is dependent on the presence of the heparin-binding domain in this protein.

Recent studies demonstrate that the modulation of the biological activity of cytokines by HS depends on the affinity and kinetics of association (“on” rate) between cytokines and HS.50 We observed that both O-sulfated heparin and heparin bound all the proteins examined above, but only O-sulfated heparin supported LTC-IC maintenance. We therefore examined the relative affinity and kinetics of binding of these GAGs for proteins required for HPC localization and maintenance. The affinity of heparin for TSP, a matrix protein, was 4-fold higher than the affinity of O-sulfated heparin (half maximal binding {EC50} 4 nM vs 17 nM, respectively; Figure 6).

Fig. 6.

Affinity of unmodified and O-sulfated heparins for thrombospondin.

The affinity of unmodified or O-sulfated heparin for TSP was examined using affinity coelectrophoresis. 125I-labeled GAGs were electrophoresed through varying concentrations of TSP (0 to 100 nM). Half-maximal binding (EC50; a measure of affinity) was calculated as the concentration of TSP (in nM) at which retardation of migration of the GAGs (R; a function of binding) was 50%.

Fig. 6.

Affinity of unmodified and O-sulfated heparins for thrombospondin.

The affinity of unmodified or O-sulfated heparin for TSP was examined using affinity coelectrophoresis. 125I-labeled GAGs were electrophoresed through varying concentrations of TSP (0 to 100 nM). Half-maximal binding (EC50; a measure of affinity) was calculated as the concentration of TSP (in nM) at which retardation of migration of the GAGs (R; a function of binding) was 50%.

Close modal

Because the chemokine PF4 is effective in supporting maintenance of LTC-IC in vitro, its binding was also investigated. By ACE, the affinity of heparin for PF4 was higher than the affinity of O-sulfated heparin (not shown). We therefore further examined the real-time binding kinetics between PF4 and unmodified or O-sulfated heparin using surface plasmon resonance (Figure 7). These studies demonstrated that the affinity of heparin for PF4 was 5-fold higher than the affinity of O-sulfated heparin (KD, 17 nM vs 90 nM). Moreover, the rates of association (“on” rate; ka) and dissociation (“off” rate; kd) of heparin were 2- to 2.5-fold higher than those of O-sulfated heparin, for PF4.

Fig. 7.

Binding of unmodified and O-sulfated heparins to PF4.

After a stable baseline tracing was established by perfusion of unsupplemented buffer over biotinylated PF4 immobilized on a streptavidin chip in a BIAcore® biosensor equipment, a range of concentrations of unmodified heparin (HEP) or O-sulfated heparin (O-S HEP) were perfused over the chip. Binding of each GAG was measured in resonance units (1 RU = 1 pg/mm2) in triplicate at each concentration at 5 different concentrations. Representative binding curves are shown for the following concentrations of GAGs: 20.8 μmol/L HEP and 31.3 μmol/L O-S HEP. The perfusion of GAGs resulted in a shift in the tracing due to binding of the GAGs to PF4 (association phase; shaded area labeled A). When GAG perfusion was stopped and perfusion with unsupplemented buffer resumed, the tracing returned downward as the bound GAGs dissociated from PF4 (dissociation phase; shaded area labeled D). Perfusion of heparin resulted in a sharp up-slope, indicating rapid association, and a sharp down-slope following cessation of heparin perfusion, indicating rapid dissociation. In contrast, perfusion of O-sulfated heparin resulted in more gradual up-slope and down-slope, indicating slower association and dissociation from PF4. The table shows the median values for KD, ka (association rate) and kd(dissociation rate).

Fig. 7.

Binding of unmodified and O-sulfated heparins to PF4.

After a stable baseline tracing was established by perfusion of unsupplemented buffer over biotinylated PF4 immobilized on a streptavidin chip in a BIAcore® biosensor equipment, a range of concentrations of unmodified heparin (HEP) or O-sulfated heparin (O-S HEP) were perfused over the chip. Binding of each GAG was measured in resonance units (1 RU = 1 pg/mm2) in triplicate at each concentration at 5 different concentrations. Representative binding curves are shown for the following concentrations of GAGs: 20.8 μmol/L HEP and 31.3 μmol/L O-S HEP. The perfusion of GAGs resulted in a shift in the tracing due to binding of the GAGs to PF4 (association phase; shaded area labeled A). When GAG perfusion was stopped and perfusion with unsupplemented buffer resumed, the tracing returned downward as the bound GAGs dissociated from PF4 (dissociation phase; shaded area labeled D). Perfusion of heparin resulted in a sharp up-slope, indicating rapid association, and a sharp down-slope following cessation of heparin perfusion, indicating rapid dissociation. In contrast, perfusion of O-sulfated heparin resulted in more gradual up-slope and down-slope, indicating slower association and dissociation from PF4. The table shows the median values for KD, ka (association rate) and kd(dissociation rate).

Close modal

The present studies demonstrate that, in the absence of stroma, a combination of specific HS GAGs with IL-3 and MIP-1α or PF4 is sufficient and necessary for the maintenance of the majority of input numbers of human LTC-IC for up to 5 weeks. LTC-IC expansion requires the presence of additional growth-promoting cytokines that are detectable in stroma-conditioned medium.

Results presented here are consistent with our recent observation that stroma-derived, O-sulfated HS GAGs improve LTC-IC maintenance in stroma-free cultures.26,34 In support of the importance of such O-sulfated GAGs for hematopoiesis, HS from the AFT024 stromal cell line, which supports multipotential and transplantable progenitors,2-4,51-53 is also enriched in such O-sulfated HS.54 Use of either HS or HS coupled to ovalbumin or human serum albumin in combination with IL-3 + MIP-1α ± CK resulted in equivalent LTC-IC maintenance, CFC generation, and cell expansion (data not shown). This indicates that (i) the LTC-IC maintaining activity is attributable to the HS side chains of HS proteoglycans and (ii) that the multimeric structure formed by coupling of HS chains to a “core” protein may not be more active than single HS chains in solution. Consistent with previous studies from our group, GAGs were not required for the cytokine-induced generation of mature blood cells and committed progenitors, or for short-term expansion of more primitive LTC-IC.

Both 2-week expansion and 5-week maintenance of LTC-IC were slightly superior in stroma-conditioned medium + IL-3 + MIP-1α cultures, compared to stroma-free cultures supplemented with cytokines and GAGs. This suggests that additional cytokines, such as flt-3 ligand or thrombopoietin, or additional unidentified stromal factors may further enhance support of primitive hematopoietic progenitors.3,51,52 

Of note, we demonstrate that IL-3, MIP-1α, and PF4 require O-sulfated GAGs for their LTC-IC–maintaining capability. Other investigators have shown that the reconstituting ability of stem cells is lost when cultured in vitro with high concentrations of IL-3 in the absence of stroma.55,56 In contrast, microenvironmentally presented IL-3 produced by genetically engineered stromal cells does not exhaust primitive progenitors.57 Thus, whereas IL-3 alone may be detrimental to the in vitro maintenance of primitive stem cells, in the presence of O-sulfated GAGs and/or other matrix molecules (added to the cultures or produced by stromal feeders), IL-3 supports HPC in vitro.

It is still not completely clear how GAGs modulate LTC-IC maintenance in vitro. We have previously hypothesized that GAGs may colocalize cytokines and matrix proteins with HPC and/or modulate the activity of cytokines.26,34 Although IL-3 is not detectable in soluble form in stromal supernatants,58 stromal HS proteoglycans bind and present IL-3 in a biologically active form to hematopoietic cells9,35,59 and augment the localization of clonogenic progenitors with IL-3.37 These studies, and our observation that HPC-supportive O-sulfated heparin directly binds IL-3 and MIP-1α, support our colocalization hypothesis. GAGs may also modulate the activity of localized cytokines on target cells.8,21,22,60 Previous studies have shown that heparin-induced dimerization of FGF molecules induces receptor activation and cell proliferation61 and that dimers of IL-3 may be required for high-affinity receptor binding.62 The interaction of IL-3 with HPC-supportive GAGs may thus promote or modulate the biological activity of the cytokine via diverse mechanisms. Highly sulfated heparin also directly inhibits expression of cytokines and cell cycle proteins in target cells,63suggesting yet another mechanism for GAG-mediated modulation of the proliferation and differentiation-inducing effects of exogenously added cytokines on HPC.

Structural features of O-sulfated heparin and unmodified heparin (Table) may account for the observed differences in their affinity, kinetics, and profiles of protein binding. These differences may be responsible for our observation that O-sulfated heparin, but not unmodified heparin, supports LTC-IC maintenance, even though both GAGs bind the same cytokines and matrix molecules. Nearly all the individual GAG chains in unmodified heparin possess an adequate number of highly sulfated (“default” sulfated)64 disaccharide units with sulfation at N-, 2-O- and 6-O- positions {-IdoA (2−OSO3)-GlcNS (6−OSO3)-} (Table) for the various proteins to bind to the GAG chains at the concentrations examined. In contrast, O-sulfated heparin chains infrequently possess such “default” sulfated disaccharides (Table). The binding of proteins to O-sulfated heparin chains is likely to occur to more “selective” protein-binding regions related to the O-sulfated disaccharides, which are 6-sulfated, with or without 2-O sulfation: The majority of disaccharides in O-sulfated heparin are identical to such units and resemble sulfation pattern at the “transition points” of HS. The variability in the ability of subsets of O-sulfated heparin chains to bind proteins, as seen on ACE, might be indicative of the presence or absence of such binding regions on individual O-sulfated heparin chains. Consistent with this notion, the slow “on” time (association rate) seen for binding of PF4 suggests that proteins may take time to find such binding sites on O-sulfated heparin, and the slow “off” time (dissociation rate) suggests that, once bound, proteins may not come off rapidly.

Recent studies have demonstrated that HS possessing fast “on” rate and high-affinity binding sites for bFGF do not activate the biological activity of bFGF on target cells, whereas HS with a slow “on” rate, low-affinity binding sites have this capability.50 Separation of the 2 types of binding sites in microheterogenous populations of HS, by enzymatic digestion with heparinase that digests the high-affinity sites, restored the ability of the HS to activate bFGF. We demonstrate that the O-sulfated heparin, which optimally supports HPC maintenance and expansion, binds relevant growth factors and matrix molecules with a lower affinity and a slower “on” rate than unmodified heparin. This suggests that the higher degree of sulfation in heparin64 is detrimental to the LTC-IC–maintaining ability of GAGs. O-sulfated heparin represents an “average” structure and sulfation pattern, required for binding of multiple cytokines and matrix proteins. The lower protein-binding affinity of O-sulfated heparin than of the more highly sulfated unmodified heparin may be essential for optimal delivery of proteins to cell surface GAGs or receptors, and the slow dissociation rate may help mediate accumulation of bound proteins in close proximity to the progenitors.50,65 Taken together, differences between unmodified heparin and O-sulfated heparin are consistent with the concept that GAG-mediated support of human HPC in vitro (and likely other target cells) depends not only on the ability of GAGs to bind proteins, but also on optimal binding affinity and kinetics of interactions with early-acting cytokines and matrix components.

The authors acknowledge the excellent technical assistance of Brad Anderson and of the Biomedical Imaging and Processing Laboratory, University of Minnesota, and thank Dr James D. San Antonio (Jefferson Medical College, Philadelphia, PA) for helpful discussions.

Supported by the US Department of Veterans Affairs, Washington, DC; grants R01-HL-48738, P01-CA-65493, and AR-32372 from the National Institutes of Health, Bethesda, MD; the Minnesota Medical Foundation, the University of Minnesota Bone Marrow Transplant Research Fund, and the University of Minnesota Hospital and Clinic, Minneapolis, MN.

C.M.V. is a scholar of the Leukemia Society of America.

Reprints:Pankaj Gupta, Hematology/Oncology Section 111E, VA Medical Center, 1 Veterans Drive, Minneapolis, MN 55417; e-mail: gupta013@gold.tc.umn.edu.

The publication costs of this article were defrayed in part by page charge payment. Therefore, and solely to indicate this fact, this article is hereby marked “advertisement” in accordance with 18 U.S.C. section 1734.

1
Lowry
PA
Deacon
D
Whitefield
P
McGrath
HE
Quesenberry
PJ
Stem cell factor induction of in vitro murine hematopoietic colony formation by “subliminal” cytokine combinations: the role of “anchor factors.”
Blood.
80
1992
663
669
2
Lemischka
IR
Microenvironmental regulation of hematopoietic stem cells.
Stem Cells.
15
1997
63
68
3
Moore
KA
Pytowski
B
Witte
L
Hicklin
D
Lemischka
IR
Hematopoietic activity of a stromal cell transmembrane protein containing epidermal growth factor-like repeat motifs.
Proc Natl Acad Sci U S A.
94
1997
4011
4016
4
Moore
KA
Ema
H
Lemischka
IR
In vitro maintenance of highly purified, transplantable hematopoietic stem cells.
Blood.
89
1997
4337
4347
5
Long
MW
Briddell
R
Walter
AW
Bruno
E
Hoffman
R
Human hematopoietic stem cell adherence to cytokines and matrix molecules.
J Clin Invest.
90
1992
251
255
6
Lyon
M
Deakin
JA
Mizuno
K
Nakamura
T
Gallagher
JT
Interaction of hepatocyte growth factor with heparan sulfate: elucidation of the major heparan sulfate structural determinants.
J Biol Chem.
269
1994
11,216
11,223
7
Maccarana
M
Lindahl
U
Mode of interaction between platelet factor 4 and heparin.
Glycobiology.
3
1993
271
277
8
Ornitz
DM
Herr
AB
Nilsson
M
Westman
J
Svahn
C-M
Waksman
G
FGF binding and FGF receptor activation by synthetic heparan-derived di- and trisaccharides.
Science.
268
1995
432
436
9
Roberts
R
Gallagher
J
Spooncer
E
Allen
TD
Bloomfield
F
Dexter
RM
Heparan sulphate bound growth factors: a mechanism for stromal cell mediated haemopoiesis.
Nature.
332
1998
376
378
10
Schon P, Vischer P, Volker W, Schmidt A, Faber V. Cell-associated proteoheparan sulfate mediates binding and uptake of thrombospondin in cultured porcine vascular endothelial cells. Eur J Cell Biol. 59:329-339.
11
Sun
X
Kaesberg
PR
Choay
J
Harenberg
J
Ershler
WB
Mosher
DF
Effects of sized heparin oligosaccharides on the interactions of Chinese hamster ovary cells with thrombospondin.
Semin Thromb Hemost.
18
1992
243
251
12
Corless
CL
Mendoza
A
Collins
T
Lawler
J
Colocalization of thrombospondin and syndecan during murine development.
Dev Dyn.
193
1992
346
358
13
Brunner
G
Gabrilove
J
Rifkin
DB
Wilson
EL
Phospholipase C release of basic fibroblast growth factor from human bone marrow cultures as a biologically active complex with a phosphatidylinositol-anchored heparan sulfate proteoglycan.
J Cell Biol.
114
1991
1275
1283
14
San Antonio
JD
Slover
J
Lawler
J
Karnovsky
MJ
Lander
AD
Specificity in the interactions of extracellular matrix proteins with subpopulations of the glycosaminoglycan heparin.
Biochemistry.
32
1993
4746
4755
15
Lee
KB
al-Hakim
A
Loganathan
D
Linhardt
RJ
A new method for sequencing linear oligosaccharides on gels using charged, fluorescent conjugates.
Carbohydr Res.
214
1991
155
168
16
Graham
GJ
Wright
EG
Hewick
R
et al
Identification and characterization of an inhibitor of haemopoietic stem cell proliferation.
Nature.
344
1990
442
444
17
Kennedy
M
Firpo
M
Choi
K
et al
A common precursor for primitive erythropoiesis and definitive haematopoiesis.
Nature.
386
1997
488
493
18
Fiebich
BL
Jager
B
Schollmann
C
et al
Synthesis and assembly of functionally active human vascular endothelial growth factor homodimers in insect cells.
Eur J Biochem.
211
1993
19
26
19
Gitay-Goren
H
Soker
S
Vlodavsky
I
Neufeld
G
The binding of vascular endothelial growth factor to its receptors is dependent on cell surface-associated heparin-like molecules.
J Biol Chem.
267
1992
6093
6098
20
Nakayama
N
Fang
I
Elliott
G
Natural killer and B-lymphoid potential in CD34+ cells derived from embryonic stem cells differentiated in the presence of vascular endothelial growth factor.
Blood.
91
1998
2283
2295
21
Faham
S
Hileman
RE
Fromm
JR
Linhardt
RJ
Rees
DC
Heparin structure and interactions with basic fibroblast growth factor.
Science.
271
1996
1116
1120
22
Kato
M
Wang
H
Kainulainen
V
et al
Physiological degradation converts the soluble syndecan-1 ectodomain from an inhibitor to a potent activator of FGF-2.
Nature Med.
4
1998
691
697
23
Rahmoune
H
Rudland
PS
Gallagher
JT
Fernig
DG
Hepatocyte growth factor/scatter factor has distinct classes of binding site in heparan sulfate from mammary cells.
Biochemistry.
37
1998
6003
6008
24
Stringer
SE
Gallagher
JT
Specific binding of the chemokine platelet factor 4 to heparan sulfate.
J Biol Chem.
272
1997
20,508
20,514
25
Feyzi
E
Saldeen
T
Larsson
E
Lindahl
U
Salmivirta
M
Age-dependent modulation of heparan sulfate structure and function.
J Biol Chem.
273
1998
13,395
13,398
26
Gupta
P
Oegema
TR
Brazil
JJ
Dudek
AZ
Slungaard
A
Verfaillie
CM
Structurally specific heparan sulfates support primitive human hematopoiesis by formation of a multimolecular stem cell niche.
Blood.
92
1998
4641
4651
27
Verfaillie
CM
Soluble factor(s) produced by human bone marrow stroma increase cytokine-induced proliferation and maturation of primitive hematopoietic progenitors while preventing their terminal differentiation.
Blood.
82
1993
2045
2053
28
Verfaillie
CM
Direct contact between human primitive hematopoietic progenitors and bone marrow stroma is not required for long-term in vitro hematopoiesis.
Blood.
79
1992
2821
2826
29
Verfaillie
CM
Catanzarro
PM
Li
W
Macrophage inflammatory protein-1α, interleukin 3 and diffusible marrow stromal factors maintain human hematopoietic stem cells for at least eight weeks in vitro.
J Exp Med.
179
1994
643
649
30
Bhatia
R
McGlave
PB
Miller
JS
Wissink
S
Lin
WN
Verfaillie
CM
A clinically suitable ex vivo expansion culture system for LTC-IC and CFC using stroma-conditioned medium.
Exp Hematol.
25
1997
980
991
31
Broxmeyer
HE
Sherry
B
Cooper
S
et al
Comparative analysis of the human macrophage inflammatory protein family of cytokines (chemokines) on proliferation of human myeloid progenitor cells: interacting effects involving suppression, synergistic suppression, and blocking of suppression.
J Immunol.
150
1993
3448
3458
32
Miller
JS
McCullar
V
Verfaillie
CM
Ex vivo culture of CD34+/Lin-/DR- cells in stroma-derived soluble factors, interleukin-3, and macrophage inflammatory protein-1alpha maintains not only myeloid but also lymphoid progenitors in a novel switch culture assay.
Blood.
91
1998
4516
4522
33
Leary
AG
Zeng
HQ
Clark
SC
Ogawa
M
Growth factor requirements for survival in G0 and entry into the cell cycle of primitive human hematopoietic progenitors.
Proc Natl Acad Sci U S A.
89
1992
4013
4017
34
Gupta
P
McCarthy
JB
Verfaillie
CM
Stromal fibroblast heparan sulfate is required for cytokine-mediated ex vivo maintenance of human long-term culture-initiating cells.
Blood.
87
1996
3229
3236
35
Gordon
MY
Riley
GP
Watt
SM
Greaves
MF
Compartmentalization of a haematopoietic growth factor (GM-CSF) by glycosaminoglycans in the bone marrow microenvironment.
Nature.
326
1987
403
405
36
Alvarez-Silva
M
Borojevic
R
GM-CSF and IL-3 activities in schistosomal liver granulomas are controlled by stroma-associated heparan sulfate proteoglycans.
J Leukoc Biol.
59
1996
435
441
37
Bruno
E
Luikart
SD
Long
MW
Hoffman
R
Marrow-derived heparan sulfate proteoglycan mediates the adhesion of hematopoietic progenitor cells to cytokines.
Exp Hematol.
23
1995
1212
1217
38
Long
MW
Dixit
VM
Thrombospondin functions as a cytoadhesion molecule for human hematopoietic progenitor cells.
Blood.
75
1990
2311
2318
39
Chen
YZ
Incardona
F
Legrand
C
Momeux
L
Caen
J
Han
ZC
Thrombospondin, a negative modulator of megakaryocytopoiesis.
J Lab Clin Med.
129
1997
231
238
40
Sanderson
PN
Nieduszynski
IA
Huckerby
TN
Very-high-field nmr studies of bovine lung heparan sulfate oligosaccharides produced by nitrous acid deaminative cleavage.
Biochem J.
211
1983
677
682
41
Maccarana
M
Sakura
Y
Tawada
A
Yoshida
K
Lindahl
U
Domain structure of heparan sulfates from bovine organs.
J Biol Chem.
271
1996
17,804
17,810
42
Danishefsky
I
Tzeng
F
Ahrens
M
Klein
S
Synthesis of heparin-sepharoses and their binding with thrombin and antithrombin-heparin cofactor.
Thromb Res.
8
1976
131
140
43
Verfaillie
CM
Blakolmer
K
McGlave
P
Purified primitive human hematopoietic progenitor cells with long-term in vitro repopulating capacity adhere selectively to irradiated bone marrow stroma.
J Exp Med.
172
1990
509
512
44
Taswell
C
Limiting dilution assays for the determination of immunocompetent cell frequencies. I. Data analysis.
J Immunol.
126
1981
1614
1619
45
Lee
MK
Lander
AD
Analysis of affinity and structural selectivity in the binding of proteins to glycosaminoglycans: development of a sensitive electrophoretic approach.
Proc Natl Acad Sci U S A.
88
1991
2768
2772
46
Mach
H
Volkin
DB
Burke
CJ
et al
Nature of the interaction of heparin with acidic fibroblast growth factor.
Biochemistry.
32
1993
5480
5489
47
Fisher
RJ
Fivash
M
Surface plasmon resonance based methods for measuring the kinetics and binding affinities of biomolecular interactions.
Curr Opin Biotechnol.
5
1994
389
395
48
Gitay-Goren
H
Cohen
T
Tessler
S
et al
Selective binding of VEGF121 to one of the three vascular endothelial growth factor receptors of vascular endothelial cells.
J Biol Chem.
271
1996
5519
5523
49
Park
JE
Keller
GA
Ferrara
N
The vascular endothelial growth factor (VEGF) isoforms: differential deposition into the subepithelial extracellular matrix and bioactivity of extracellular matrix-bound VEGF.
Mol Biol Cell.
4
1993
1317
1326
50
Rahmoune
H
Chen
HL
Gallagher
JT
Rudland
PS
Fernig
DG
Interaction of heparan sulfate from mammary cells with acidic fibroblast growth factor (FGF) and basic FGF. Regulation of the activity of basic FGF by high and low affinity binding sites in heparan sulfate.
J Biol Chem.
273
1998
7303
7310
51
Punzel
M
Miller
JS
Moore
KA
Lemischka
IR
Verfaillie
CM
Culture of human CD34+/HLA-DR-/LIN- cells for 4 weeks on the AFT024 stromal cell line results in extensive amplification of CD34+ cells with 3-100 fold expansion of LTC-IC, CFC and primitive lymphoid progenitors [abstract].
Blood.
90
10 suppl 1
1997
161a
52
Punzel
M
Wissink
S
Asleson
K
et al
Development of an in vitro assay that can enumerate generative multilineage-initiating cells (ML-IC) in adult human BM [abstract].
Exp Hematol.
26
1998
693
53
Thiemann
FT
Moore
KA
Smogorzewska
EM
Lemischka
IR
Crooks
GM
The murine stromal cell line AFT024 acts specifically on human CD34+CD38- progenitors to maintain primitive function and immunophenotype in vitro.
Exp Hematol.
26
1998
612
619
54
Punzel
M
Gupta
P
Roodell
M
Mortari
F
Verfaillie
CM
Factor(s) secreted by AFT024 fetal liver cells following stimulation with human cytokines, are important for human LTC-IC growth.
Leukemia
7
1999
1079
1084
55
Matsunaga
T
Hirayama
F
Yonemura
Y
Murray
R
Ogawa
M
Negative regulation by interleukin-3 (IL-3) of mouse early B-cell progenitors and stem cells in culture: transduction of the negative signals by betac and betaIL-3 proteins of IL-3 receptor and absence of negative regulation by granulocyte-macrophage colony-stimulating factor.
Blood.
92
1998
901
907
56
Yonemura
Y
Ku
H
Hirayama
F
Souza
LM
Ogawa
M
Interleukin 3 or interleukin 1 abrogates the reconstituting ability of hematopoietic stem cells.
Proc Natl Acad Sci U S A.
93
1996
4040
4044
57
Otsuka
T
Thacker
JD
Eaves
CJ
Hogge
DE
Differential effects of microenvironmentally presented interleukin 3 versus soluble growth factor on primitive human hematopoietic cells.
J Clin Invest.
88
1991
417
422
58
Burroughs
J
Gupta
P
Blazar
BR
Verfaillie
CM
Diffusible factors from the murine cell line M2-10B4 support human in vitro hematopoiesis.
Exp Hematol.
22
1994
1095
1101
59
Kittler
EL
McGrath
H
Temeles
D
Crittenden
RB
Kister
VK
Quesenberry
PJ
Biologic significance of constitutive and subliminal growth factor production by bone marrow stroma.
Blood.
79
1992
3168
3178
60
Modrowski
D
Lomri
A
Marie
PJ
Glycosaminoglycans bind granulocyte-macrophage colony-stimulating factor and modulate its mitogenic activity and signaling in human osteoblastic cells.
J Cell Physiol.
177
1998
187
195
61
Spivak-Kroizman
T
Lemmon
MA
Dikic
I
et al
Heparin-induced oligomerization of FGF molecules is responsible for FGF receptor dimerization, activation, and cell proliferation.
Cell.
79
1994
1015
1024
62
Muther
H
Kuhlcke
K
Gessner
A
Abdallah
S
Lother
H
Homodimeric murine interleukin-3 agonists indicate that ligand dimerization is important for high-affinity receptor complex formation.
Growth Factors.
10
1994
17
27
63
Yang
L
Yang
Y-C
Heparin inhibits the expression of interleukin-11 and granulocyte-macrophage colony-stimulating factor in primate bone marrow stromal fibroblasts through mRNA destabilization.
Blood.
86
1995
2526
2533
64
Salmivirta
M
Lidholt
K
Lindahl
U
Heparan sulfate: a piece of information.
FASEB J.
10
1996
1270
1279
65
Wang
H
Toida
T
Kim
YS
et al
Glycosaminoglycans can influence fibroblast growth factor-2 mitogenicity without significant growth factor binding.
Biochem Biophys Res Commun.
235
1997
369
373
Sign in via your Institution