Abstract

From signaling mediators in stem cells to markers of differentiation and lineage commitment to facilitators for the entry of viruses, such as HIV-1, cell surface heparan sulfate (HS) glycans with distinct modification patterns play important roles in hematopoietic biology. In this review, we provide an overview of the importance of HS and the proteoglycans (HSPGs) to which they are attached within the major cellular subtypes of the hematopoietic system. We summarize the roles of HSPGs, HS, and HS modifications within each main hematopoietic cell lineage of both myeloid and lymphoid arms. Lastly, we discuss the biological advances in the detection of HS modifications and their potential to further discriminate cell types within hematopoietic tissue.

Hematopoiesis is the process of blood formation in which hematopoietic stem and progenitor cells (HSPCs) self-renew to maintain the hematopoietic pool as well as differentiate and commit toward specialized lineages that carry out a host of cell-type–specific functions. Hematopoietic tissue can be broadly divided into myeloid and lymphoid arms, each of which contains its own distinct partially committed progenitors. Master transcriptional regulators, distinct stromal niches, and specific cytokine cues are critical in the maintenance of HSPCs, as well as their differentiation toward fully mature, functional cell lineages. External cues such as cytokine/chemokine stimulation occur through the binding of ligands to their corresponding surface receptors, a process frequently facilitated by coreceptors, as well as molecular modifications.

Heparan sulfates (HSs) are linear polysaccharide chains attached to select surface proteins (HS proteoglycans [HSPGs]) that are present in all eukaryotic cells. They are part of a family of molecules known as glycosaminoglycans, which also include chondroitin/dermatan sulfate, keratan sulfate, and hyaluronic acid. HS chains are synthesized as repeating disaccharide units of hexuronic acid (glucuronic acid [GlcA] or iduronic acid [IdoA]) and N-acetylglucosamine (GlcNAc), which can be modified at various positions, allowing for enormous chain complexity. The synthesis of HS is a concerted process relying on multiple families of enzymes and occurs in the endoplasmic reticulum and/or golgi apparatus. First, a conserved tetra-saccharide linker (made of xylose-galactose-galactose-glucuronic acid) is sequentially added to serine residues of a core HSPG protein by specific glycosyltransferases. A GlcNAc residue is then added to initiate HS biosynthesis, a step that distinguishes HS biosynthesis from other types of glycosaminoglycan synthesis (ie, chondroitin sulfate). In the next step, which is known as chain polymerization, the exostoses family of enzymes adds repeating GlcA-GlcNAc units. The resulting 50 to 150 unit polysaccharide chains are additionally modified by sulfation (at O- and N-residues), acetylation, and epimerization at numerous points along the chain, which is carried out by an array of modification enzymes (Figure 1A).1 This elaborate process results in highly diverse modification patterns owing to near-limitless combinatorial possibilities. This molecular diversity contributes to cell-specific functionalities in many physiologic and developmental processes.2-4 

Figure 1.

Overview of HS synthesis and modification of HSPGs. (A) HS chain initiation begins with the sequential attachment of a core tetra-saccharide sequence to the serine residue of the HSPG. HS synthesis enzymes then facilitate the attachment of repeating GlcA-GlcNAc disaccharide moieties, creating a linear HS chain, whereas multiple HS-modifying enzymes alter the linear HS chain through distinct modifications of specific residues (including O-sulfation, N-sulfation, and epimerization). This sequential process produces a unique chain of modified HS attached to the core HSPG. Extracellular modification of HS also occurs through the removal of sulfates by sulf enzymes, as well as the removal of sections of the HS chain by heparanase. (B) Overview of the main HSPG subcategories, including syndecans, transmembrane proteins that contain a variable (V) domain, as well as 2 common domains (C1 and C2), of which C2 contains a PDZ binding motif, glypicans, which are glycophosphatidylinositol–linked proteins; and secreted or pericellular proteins such as perlecan and agrin. The cell membrane lipid bilayer is shown in black, and pink delineates the core HSPG protein with attached HS chains (in green) and chondroitin sulfate chains (in blue).

Figure 1.

Overview of HS synthesis and modification of HSPGs. (A) HS chain initiation begins with the sequential attachment of a core tetra-saccharide sequence to the serine residue of the HSPG. HS synthesis enzymes then facilitate the attachment of repeating GlcA-GlcNAc disaccharide moieties, creating a linear HS chain, whereas multiple HS-modifying enzymes alter the linear HS chain through distinct modifications of specific residues (including O-sulfation, N-sulfation, and epimerization). This sequential process produces a unique chain of modified HS attached to the core HSPG. Extracellular modification of HS also occurs through the removal of sulfates by sulf enzymes, as well as the removal of sections of the HS chain by heparanase. (B) Overview of the main HSPG subcategories, including syndecans, transmembrane proteins that contain a variable (V) domain, as well as 2 common domains (C1 and C2), of which C2 contains a PDZ binding motif, glypicans, which are glycophosphatidylinositol–linked proteins; and secreted or pericellular proteins such as perlecan and agrin. The cell membrane lipid bilayer is shown in black, and pink delineates the core HSPG protein with attached HS chains (in green) and chondroitin sulfate chains (in blue).

Close modal

HS chains are attached to core proteins called HS proteoglycans (HSPGs). These include 3 major classes of proteins: syndecans (SDCs), a class of transmembrane proteins (Sdc1-4 in mammals); glypicans, a class of glycophosphatidylinositol–linked proteins (Gpc1-6 in mammals); and secreted proteins such as agrin, perlecan, and serglycin among others (Figure 1B). Specific proteoglycan families play roles in distinct signaling pathways: glypicans, for example, facilitate Hedgehog, Wnt, and fibroblast growth factor (FGF) signaling, betaglycan (also known as transforming growth factor β receptor 3 [TGFBR3]) mediates transforming growth factor β (TGF-β) signaling, and the SDC family acts as coreceptors for many receptor tyrosine kinases, including FGF receptors among others.4,5 

Once on the cell surface, HS can be further altered through enzymatic removal of sulfation marks by sulfatases, Sulf1 and Sulf2, or cleaved by the enzyme heparanase (Figure 1A). These processes remodel the composition of HS and its modifications on the cell surface and facilitate the formation and reshaping of the extracellular matrix. Importantly, they influence cytokine/chemokine gradients, facilitate cell migration, and promote cellular signaling; all functions essential for the biology of hematopoietic cells.6-8 

The importance of HS in cells that interact with hematopoietic cells (ie, bone marrow [BM] stroma, endothelial cells, etc) has been extensively documented and reviewed, but less is known about the roles of HS in hematopoietic cells themselves.6,8 HS on stromal cells supports stem cell maintenance and hematopoietic cell differentiation, resulting in blood cell formation, and HS on the endothelial cell surface of tissues mediates leukocyte adhesion, trafficking, and migration. However, the surface of hematopoietic cells also contains HS, which confer unique cell-specific functionalities. There is a dearth of knowledge on the expression, function, and significance of HS that is expressed on many subtypes of hematopoietic cells when compared with cell types that comprise the hematopoietic niche (osteoblast/clasts, endothelial cells, etc). This review summarizes what is known about HS with regard to the regulation of hematopoiesis and blood cell function, focusing on each general cell type within hematopoietic tissue.

A multitude of cytokines and chemokines that are essential for hematopoietic cell differentiation and maintenance have been shown to bind to HS either in vitro or in vivo (Table 1). Many of these ligands are secreted by hematopoietic cells themselves and regulate normal hematopoiesis in an autocrine or paracrine manner.60 Binding of cytokines and chemokines to HS can facilitate oligomerization and cell signaling, protect against proteolytic degradation, and create concentration gradients that fine-tune the signaling processes. Although much remains to be elucidated, biochemical studies have shown specific requirements of HS modifications (both specific sulfation requirements, as well as regional sulfation/charge distribution requirements) for the binding of certain cytokines and chemokines to HS.61 Many interleukin molecules selectively bind to distinctly modified HS, highlighting the specificity of HS-cytokine interactions (Table 1). With the expression of HS on the surface of hematopoietic cells and the abundance of HS-binding cytokines and chemokines, there is still much to be discovered regarding the role of HS-chemokine/cytokine interactions in hematopoiesis.

Table 1.

Hematopoietic chemokines and cytokines that bind HS

Cytokine/chemokineMajor roles in hematopoiesisHS modifications facilitating bindingReference
IL-1 Myeloid differentiation of stem and progenitor cells
Response to infections
Inflammation/immune response 
N-sulfation (important, not required) 9  
IL-2 T-cell differentiation
NK-cell activation 
N-sulfation 9  
IL-3 HSPC maintenance 6-O-sulfation 7,10  
IL-4 Mature B- and T-cell development
B-cell class switching
Macrophage regulator 
IdoA, N-sulfation, O-sulfation 11,12  
IL-5 B-cell growth
Eosinophil growth and activation 
Heparin, N-sulfation 13  
IL-6 Neutrophil production
Fever and acute phase response
B-cell growth and development
T-cell antagonist 
Heparin, N-sulfation (important, not required), 2-O-sulfation 9,14  
IL-7 Stimulates lymphoid differentiation
Mature B- and T-cell development
Immune modulator (pro and antiinflammatory effects) 
Heparin, HS (unspecified) 15  
IL-8 Granulocyte chemotaxis and phagocytosis Broad sulfation (N-sulfation, 2-O-sulfation, 6-O-sulfation) 16-18  
IL-10 B-cell survival and growth
Anti-inflammatory response
Immune regulation 
Heparin, N-sulfation, 2-O-sulfation (less), 6-O-sulfation (less) 19,20  
IL-12 T-cell growth, function, and differentiation Heparin 21  
IL-18 Proinflammatory
Mature T-cell differentiation 
Unknown 22  
IL-21 B-cell and T-cell differentiation and maturation N-sulfation, 2-O-sulfation (less), 6-O-sulfation (less) 23  
MCP-1 (CCL2) Leukocyte activation and recruitment High sulfation (N-, 2-O-, 3-O-, 6-O-), 3-O-sulfation (preferred) 18,24-27  
MIP 1α (CCL3) Leukocyte activation and recruitment 6-O-sulfation 10  
MIP 1β (CCL4) Leukocyte activation and recruitment Unknown 28  
RANTES/CCL5 Leukocyte migration/trafficking
Mediator of viral infections and immune response (eg, HIV) 
Heparin, HS, 6-O-sulfation (more), 2-O-sulfation (less) 18,29  
MCP-3 (CCL7) Macrophage attractant High sulfation 18,26,30  
MCP-2 (CCL8) Leukocyte activation and recruitment Unknown 26,31  
Eotaxin-1 (CCL11) Granulocyte differentiation
Eosinophil migration 
Unknown 32,33  
MCP-4 (CCL13) Leukocyte chemotaxis High sulfation 18,33  
Eotaxin-3 (CCL26) Eosinophil migration Unknown 34  
Stem cell factor HSC maintenance, self-renewal, adhesion Heparin, N-sulfation, 2-O-sulfation, 6-O-sulfation 35  
Granulocyte/macrophage colony-stimulating factor Myeloid differentiation (granulocyte and monocyte)
Neutrophil migration 
Unknown 7,36  
Hepatocyte growth factor HSC maintenance and mobilization 6-O-sulfation 37  
Platelet factor 4 (CXCL4) Coagulation
HSC maintenance
Chemotaxis 
N-sulfation, 2-O-sulfation,6-O-sulfation, (3-O-sulfation decreases binding) 10,27,38  
Stromal derived factor-1 (CXCL12) HSC survival, self-renewal, homing, and engraftment
T-cell migration
B-cell survival and migration 
N-sulfation, 2-O-sulfation, 6-O-sulfation 39  
CXCL13 B-cell and T-cell organization in lymph nodes Unknown 40  
FGF-1 HSC maintenance
Lineage commitment
Erythropoiesis 
2-O-sulfation, 6-O-sulfation (required) 41  
Basic FGF (FGF2) HSPC proliferation N-sulfation, 2-O-sulfation, 6-O-sulfation (not required) 18,42  
Vascular endothelial growth factor
(only certain isoforms) 
HSPC survival
Erythropoiesis 
6-O-sulfation (more), N-sulfation (less), 2-O-sulfation (less) 43,44  
Heparin binding–epidermal growth factor-like growth factor Myeloid and lymphoid differentiation 6-O-sulfation 45  
Platelet–derived growth factor Megakaryocyte and platelet proliferation and development N-sulfation, 6-O-sulfation 46  
APRIL B-cell growth and survival
Plasma cell function 
C5-epimerization 47,48  
IFN-γ–inducible protein 10 (CXCL10) Leukocyte chemoattractant
T-cell proliferation and trafficking 
HS, 6-O-sulfation (more), 2-O-sulfation (less) 18,49  
IFN-γ HSC maintenance
Immune response mediator
Induces myelopoiesis
Suppression of erythropoiesis and B-cell lymphopoiesis 
N-sulfation 50,51  
Transforming growth factor-β (only certain isoforms) HSC quiescence
Myeloid vs lymphoid commitment 
Unknown 52,53  
BMP-2 HSC maintenance N-sulfation 54  
BMP-4 HSC maintenance
Differentiation
Thymopoiesis 
N-sulfation 54  
BMP-6 HSPC migration
B-cell suppression 
Unknown 55  
BMP-7 HSC maintenance Unknown 56  
Noggin Inhibition of BMP N-sulfation, 2-O-sulfation, 6-O-sulfation (required) 57,58  
von Willebrand factor Hemostasis and platelet adhesion Unknown 59  
Cytokine/chemokineMajor roles in hematopoiesisHS modifications facilitating bindingReference
IL-1 Myeloid differentiation of stem and progenitor cells
Response to infections
Inflammation/immune response 
N-sulfation (important, not required) 9  
IL-2 T-cell differentiation
NK-cell activation 
N-sulfation 9  
IL-3 HSPC maintenance 6-O-sulfation 7,10  
IL-4 Mature B- and T-cell development
B-cell class switching
Macrophage regulator 
IdoA, N-sulfation, O-sulfation 11,12  
IL-5 B-cell growth
Eosinophil growth and activation 
Heparin, N-sulfation 13  
IL-6 Neutrophil production
Fever and acute phase response
B-cell growth and development
T-cell antagonist 
Heparin, N-sulfation (important, not required), 2-O-sulfation 9,14  
IL-7 Stimulates lymphoid differentiation
Mature B- and T-cell development
Immune modulator (pro and antiinflammatory effects) 
Heparin, HS (unspecified) 15  
IL-8 Granulocyte chemotaxis and phagocytosis Broad sulfation (N-sulfation, 2-O-sulfation, 6-O-sulfation) 16-18  
IL-10 B-cell survival and growth
Anti-inflammatory response
Immune regulation 
Heparin, N-sulfation, 2-O-sulfation (less), 6-O-sulfation (less) 19,20  
IL-12 T-cell growth, function, and differentiation Heparin 21  
IL-18 Proinflammatory
Mature T-cell differentiation 
Unknown 22  
IL-21 B-cell and T-cell differentiation and maturation N-sulfation, 2-O-sulfation (less), 6-O-sulfation (less) 23  
MCP-1 (CCL2) Leukocyte activation and recruitment High sulfation (N-, 2-O-, 3-O-, 6-O-), 3-O-sulfation (preferred) 18,24-27  
MIP 1α (CCL3) Leukocyte activation and recruitment 6-O-sulfation 10  
MIP 1β (CCL4) Leukocyte activation and recruitment Unknown 28  
RANTES/CCL5 Leukocyte migration/trafficking
Mediator of viral infections and immune response (eg, HIV) 
Heparin, HS, 6-O-sulfation (more), 2-O-sulfation (less) 18,29  
MCP-3 (CCL7) Macrophage attractant High sulfation 18,26,30  
MCP-2 (CCL8) Leukocyte activation and recruitment Unknown 26,31  
Eotaxin-1 (CCL11) Granulocyte differentiation
Eosinophil migration 
Unknown 32,33  
MCP-4 (CCL13) Leukocyte chemotaxis High sulfation 18,33  
Eotaxin-3 (CCL26) Eosinophil migration Unknown 34  
Stem cell factor HSC maintenance, self-renewal, adhesion Heparin, N-sulfation, 2-O-sulfation, 6-O-sulfation 35  
Granulocyte/macrophage colony-stimulating factor Myeloid differentiation (granulocyte and monocyte)
Neutrophil migration 
Unknown 7,36  
Hepatocyte growth factor HSC maintenance and mobilization 6-O-sulfation 37  
Platelet factor 4 (CXCL4) Coagulation
HSC maintenance
Chemotaxis 
N-sulfation, 2-O-sulfation,6-O-sulfation, (3-O-sulfation decreases binding) 10,27,38  
Stromal derived factor-1 (CXCL12) HSC survival, self-renewal, homing, and engraftment
T-cell migration
B-cell survival and migration 
N-sulfation, 2-O-sulfation, 6-O-sulfation 39  
CXCL13 B-cell and T-cell organization in lymph nodes Unknown 40  
FGF-1 HSC maintenance
Lineage commitment
Erythropoiesis 
2-O-sulfation, 6-O-sulfation (required) 41  
Basic FGF (FGF2) HSPC proliferation N-sulfation, 2-O-sulfation, 6-O-sulfation (not required) 18,42  
Vascular endothelial growth factor
(only certain isoforms) 
HSPC survival
Erythropoiesis 
6-O-sulfation (more), N-sulfation (less), 2-O-sulfation (less) 43,44  
Heparin binding–epidermal growth factor-like growth factor Myeloid and lymphoid differentiation 6-O-sulfation 45  
Platelet–derived growth factor Megakaryocyte and platelet proliferation and development N-sulfation, 6-O-sulfation 46  
APRIL B-cell growth and survival
Plasma cell function 
C5-epimerization 47,48  
IFN-γ–inducible protein 10 (CXCL10) Leukocyte chemoattractant
T-cell proliferation and trafficking 
HS, 6-O-sulfation (more), 2-O-sulfation (less) 18,49  
IFN-γ HSC maintenance
Immune response mediator
Induces myelopoiesis
Suppression of erythropoiesis and B-cell lymphopoiesis 
N-sulfation 50,51  
Transforming growth factor-β (only certain isoforms) HSC quiescence
Myeloid vs lymphoid commitment 
Unknown 52,53  
BMP-2 HSC maintenance N-sulfation 54  
BMP-4 HSC maintenance
Differentiation
Thymopoiesis 
N-sulfation 54  
BMP-6 HSPC migration
B-cell suppression 
Unknown 55  
BMP-7 HSC maintenance Unknown 56  
Noggin Inhibition of BMP N-sulfation, 2-O-sulfation, 6-O-sulfation (required) 57,58  
von Willebrand factor Hemostasis and platelet adhesion Unknown 59  

The table summarizes hematopoietic cytokines shown to bind HS, their general roles in hematopoietic cell function, and what is currently known about HS characteristics that are required for or facilitate/impede binding.

APRIL, a proliferation-inducing ligand; BMP, bone morphogenic protein; IFN-γ, interferon gamma; MCP, monocyte chemoattractant protein; MIP, macrophage inflammatory protein; RANTES/CCL5, regulated upon activation, normal T-cell expressed and presumably secreted.

Perhaps the best-characterized HS interaction relevant to the hematologic system is the interaction between heparin (an on average shorter HS polysaccharide chain with higher sulfation and predominance of L-iduronic acid instead of D-glucuronic acid) and antithrombin, which mechanistically defines the anticoagulant properties of heparin. The binding of heparin activates antithrombin, which substantially increases the rate of inactivation of key coagulation factors, including thrombin, factor Xa, and factor IXa. It was initially observed that only a fraction of the isolated heparin molecules had high-affinity binding to antithrombin, and this small portion accounted for most of the anticoagulant activity of heparin.62,63 Molecular studies later identified the pentasaccharide sequence with specific modifications, GlcNAc/NS(6S)-GlcA-GlcNS(3S,6S)-IdoA(2S)-GlcNS(6S), with a unique and necessary 3-O-sulfation, as the primary ligand for antithrombin.64 The heparin-antithrombin interaction is the exemplar of the specificity requirements for functionally relevant HS ligand binding.

Numerous animal models deficient in specific HS-related genes have been developed, many of which show embryonic lethality and drastic phenotypes in nonhematopoietic tissues, such as skeletal, cardiac, and lung defects. Table 2 summarizes the hematologic findings (if relevant) of knockout animal models of HS-related genes. Other existing HS–related gene knockout models have either not been generated (eg, Gpc5 and Hs3st5), have not been viable for analysis, and no conditional models have been generated to date (eg, Gpc6), are viable but have not had their hematopoietic compartment studied (eg, Gpc1 and Gpc2), or show no overt hematopoietic phenotype (eg, Hs3st1 and Ndst4), revealing a substantial gap in knowledge. The fact that HS–related gene knockouts lead to hematologic phenotypes in animals highlights the importance of HS in hematopoietic development and maintenance in both the myeloid and lymphoid arms.

Table 2.

Knockout animal models of HS-related genes and their associated hematopoietic phenotypes

HS geneSpeciesType of knockoutSignificant/notable hematologic effect(s)References
HSPGs     
Agrn Mouse Musk-L;Agrn−/−
Whole body KO (with expression of Musk-L in skeletal tissue to rescue embryonic lethality) 
Global absolute leukocyte reduction
Reduction in monocytic lineage
Severe anemia
Disrupted erythroblastic islands 
65,66  
Srgn Mouse Whole body KO (viable) Age-related enlargement of lymphoid tissue
Age-related decrease in splenic CD4+ cells, peritoneal macrophages
Increase in splenic naïve lymphocytes
Mast cell secretory granule defects
Cytotoxic T-cell defects
Platelet, megakaryocyte, and neutrophil abnormalities 
67-70  
Gpc3 Mouse Whole body KO (perinatal lethality) Poor HSC retention in BM
Increased circulation of HSPCs
Reduction in PMN, BM CD11b+, and GR-1+ cells 
71,72  
Sdc1 Mouse Whole body KO (prevention of surface expression of SDC1) Slight increase in lineage-negative cells
Impaired immune responses 
73  
Sdc2 Zebrafish Maternal zygotic deletion sdc2–/– Immature/defective erythroid and neutrophil populations
Poor response to infection 
74  
Sdc4 Mouse Whole body KO Reduced B-cell number, migration, and deficient lymph-node germinal center formation in arthritis-induced model 75  
Hspg2 (perlecan) Drosophila Trol (perlecan homolog) Reduction in HSPC proliferation
Precocious hematopoietic differentiation 
76  
Tgfbr3 Mouse Tgfbr3-LckCre (T cell) Enhanced T-cell–dependent antibody response
Increased Th1 T-cell differentiation 
77  
HS synthesis enzymes     
Ext1 Mouse Ext1–/– (embryonic lethal)
Ext1+/− (viable) 
Normal endogenous hematopoiesis
Decreased HSPC engraftment upon transplant 
78  
Ext1 Mouse Ext1-CD19Cre (B cell) Slightly increased Pro-B-cell population
Reduced splenic B cells
Reduced FGF2 binding/activation
No change in antibody production/response 
79  
Extl3 Zebrafish Whole body KO Defective thymopoiesis 80  
HS-modifying enzymes     
Ndst1 Mouse NDST1-TekCre (Tie2) No abnormal hematopoietic/blood parameters
Slightly decreased bleeding time 
81  
Ndst1 Mouse NDST1-LckCre (T cell) Altered T-cell activation and proliferation 79  
Ndst2 Mouse Whole body KO Impaired mast cell development/function 82  
Ndst3 Mouse Whole body KO 
Reduced circulating lymphocytes
Reduced CD8+ T cells (spleen) 
83  
Hs2st1 Mouse Hs2st1-TekCre (Tie2), Hs2st1-LysMCre No major hematopoietic phenotypes
Enhanced neutrophil invasion and rolling 
84  
Hs6st1 Mouse Whole body KO Scarce embryo–derived nucleated red blood cell in the placenta
Mast cell granule defects 
85,86  
Hs6st2 Mouse Whole body KO Mast cell granule defects 85  
Glce Mouse GLCE–/– (fetal liver HSCs transplanted into Rag-2/IL2rg KO mice) Impaired B-lineage development.
Decreased plasma cells 
49  
Hpse Mouse Whole body KO Slightly reduced total WBC
Increased maturation of BM dendritic cells
Impaired DC migration 
87,88  
HS geneSpeciesType of knockoutSignificant/notable hematologic effect(s)References
HSPGs     
Agrn Mouse Musk-L;Agrn−/−
Whole body KO (with expression of Musk-L in skeletal tissue to rescue embryonic lethality) 
Global absolute leukocyte reduction
Reduction in monocytic lineage
Severe anemia
Disrupted erythroblastic islands 
65,66  
Srgn Mouse Whole body KO (viable) Age-related enlargement of lymphoid tissue
Age-related decrease in splenic CD4+ cells, peritoneal macrophages
Increase in splenic naïve lymphocytes
Mast cell secretory granule defects
Cytotoxic T-cell defects
Platelet, megakaryocyte, and neutrophil abnormalities 
67-70  
Gpc3 Mouse Whole body KO (perinatal lethality) Poor HSC retention in BM
Increased circulation of HSPCs
Reduction in PMN, BM CD11b+, and GR-1+ cells 
71,72  
Sdc1 Mouse Whole body KO (prevention of surface expression of SDC1) Slight increase in lineage-negative cells
Impaired immune responses 
73  
Sdc2 Zebrafish Maternal zygotic deletion sdc2–/– Immature/defective erythroid and neutrophil populations
Poor response to infection 
74  
Sdc4 Mouse Whole body KO Reduced B-cell number, migration, and deficient lymph-node germinal center formation in arthritis-induced model 75  
Hspg2 (perlecan) Drosophila Trol (perlecan homolog) Reduction in HSPC proliferation
Precocious hematopoietic differentiation 
76  
Tgfbr3 Mouse Tgfbr3-LckCre (T cell) Enhanced T-cell–dependent antibody response
Increased Th1 T-cell differentiation 
77  
HS synthesis enzymes     
Ext1 Mouse Ext1–/– (embryonic lethal)
Ext1+/− (viable) 
Normal endogenous hematopoiesis
Decreased HSPC engraftment upon transplant 
78  
Ext1 Mouse Ext1-CD19Cre (B cell) Slightly increased Pro-B-cell population
Reduced splenic B cells
Reduced FGF2 binding/activation
No change in antibody production/response 
79  
Extl3 Zebrafish Whole body KO Defective thymopoiesis 80  
HS-modifying enzymes     
Ndst1 Mouse NDST1-TekCre (Tie2) No abnormal hematopoietic/blood parameters
Slightly decreased bleeding time 
81  
Ndst1 Mouse NDST1-LckCre (T cell) Altered T-cell activation and proliferation 79  
Ndst2 Mouse Whole body KO Impaired mast cell development/function 82  
Ndst3 Mouse Whole body KO 
Reduced circulating lymphocytes
Reduced CD8+ T cells (spleen) 
83  
Hs2st1 Mouse Hs2st1-TekCre (Tie2), Hs2st1-LysMCre No major hematopoietic phenotypes
Enhanced neutrophil invasion and rolling 
84  
Hs6st1 Mouse Whole body KO Scarce embryo–derived nucleated red blood cell in the placenta
Mast cell granule defects 
85,86  
Hs6st2 Mouse Whole body KO Mast cell granule defects 85  
Glce Mouse GLCE–/– (fetal liver HSCs transplanted into Rag-2/IL2rg KO mice) Impaired B-lineage development.
Decreased plasma cells 
49  
Hpse Mouse Whole body KO Slightly reduced total WBC
Increased maturation of BM dendritic cells
Impaired DC migration 
87,88  

Summary of current animal models with genetic perturbations of HSPGs, HS synthesis enzymes, or HS-modifying enzymes and their hematopoietic phenotypes.

KO, knockout.

Specific HSPGs and HS-related genes (including specific HS modifications) have been functionally implicated in specific hematopoietic cell types (Figure 2A-B). Although cell-specific functions for HS have been described, some HS and HSPG functions are shared across multiple hematopoietic cell types. For example, secretory granules within various immune cells (eg, T cells, mast cells, and natural killer [NK] cells) are critical for immune function. Serglycin (once known as hematopoietic proteoglycan core protein), was initially characterized as a predominantly intracellular proteoglycan in the secretory granules of various immune cells as well as in platelets. Serglycin plays central roles in the integrity and content of granules within various hematopoietic cells, as specified in “Granulocyte (neutrophil/basophil/eosinophil) lineage,” “Mast cell lineage,” “Megakaryocyte lineage,” “The T-cell lineage,” and “NK cells.”

Figure 2.

Relevant HSPGs, HS-related genes, and HS-modification signatures in various hematopoietic cell lineages. (A) Sunburst plot of known functionally relevant HS-related genes within specific hematopoietic lineages. The specific HS-related gene or protein for which expression or function has been identified (outer circle) within the categories of HS-related genes, including HSPGs and HS synthesis enzymes (Synth.), and HS-modification enzymes (Mod.) (middle circle), are listed for each hematopoietic cell type (inner circle). (B) Sunburst plot of HS modifications identified as functionally relevant for each hematopoietic cell type. For each hematopoietic cell type (inner circle), the specific modification and residue (outer circle) among the possible types of modifications, including epimerization (Epi), N-sulfation (N-Sulf), and O-sulfation (O-Sulf) (middle circle) is listed.

Figure 2.

Relevant HSPGs, HS-related genes, and HS-modification signatures in various hematopoietic cell lineages. (A) Sunburst plot of known functionally relevant HS-related genes within specific hematopoietic lineages. The specific HS-related gene or protein for which expression or function has been identified (outer circle) within the categories of HS-related genes, including HSPGs and HS synthesis enzymes (Synth.), and HS-modification enzymes (Mod.) (middle circle), are listed for each hematopoietic cell type (inner circle). (B) Sunburst plot of HS modifications identified as functionally relevant for each hematopoietic cell type. For each hematopoietic cell type (inner circle), the specific modification and residue (outer circle) among the possible types of modifications, including epimerization (Epi), N-sulfation (N-Sulf), and O-sulfation (O-Sulf) (middle circle) is listed.

Close modal

CD44 is expressed in nearly all hematopoietic cell types and has isoforms with various glycosylation patterns due to differential splicing. The CD44v3 isoform uniquely contains motifs available for HS attachment.89 This specific isoform has been shown to contribute to key biological functions in monocytes, macrophages, T cells, B cells, and other hematopoietic cell types.

Hematopoietic stem and progenitor cells

HSPGs

Multiple proteoglycans have been identified in HSPC biology in various species. Early gene expression studies implicated SDC2 as a regulator of long-term repopulating HSCs (LT-HSCs), whereas SDC1 was differentially expressed in multipotent progenitors (MPPs).90 Recent studies have confirmed the role of SDC2 in hematopoietic stem cells in regulating stem cell quiescence and have shown surface SDC2 enriches for long-term repopulating HSCs.91 Glypican-3 expressed on mouse and human HSPCs appears to mediate HSPC residence in the BM as well as their trafficking through circulation, presumably by a mechanism of CD26 inhibition by tissue factor pathway inhibitor.71 Loss of the Drosophila homolog of perlecan (Trol) was found to diminish the proliferation of HSPCs and induce premature differentiation into mature blood cells, suggesting that pericellular perlecan is involved in HSPC maintenance and self-renewal (Table 2).76 However, to date, no studies looking at perlecan in HSPC biology have been performed on other organisms.

The ApoE receptor, which has been shown to regulate the proliferation of HSPCs, was found in a proteoglycan-rich pool on the HSPC cell surface, implicating HS in the function of cholesterol transport into HSPCs.92 The FDCP-1 cell line, which mimics HSPCs was observed to produce and secrete HS proteoglycans.93 

Gfi1b, a master hematopoietic regulator, helps dictate erythroid and megakaryocytic lineage commitment from common bipotent megakaryocyte-erythrocyte progenitor (MEP), partly through direct repression of TGFBR3 proteoglycan (also called betaglycan), resulting in regulated TGF-β signaling.94 

HS and HS modifications

Developmentally, the hematopoietic system in mammals arises from the mesodermal embryonic layer. Temporal expression of specifically modified HS has previously been shown to characterize the mesodermal cells that give rise to Flk1+ hemangioblasts, which ultimately contribute to the formation of the hematopoietic system.95 Correspondingly, the addition of specifically modified HS (mainly N- and 6-O-sulfated HS) rescued hematopoietic differentiation in murine embryonic stem cell cultures lacking HS (Ext1/ ES cells).96 The addition of O-sulfated HS along with canonical cytokines increased the long-term colony-initiating capacity (a measure of “stemness”) of human HSPCs in vitro.97 Interestingly, induced pluripotent stem cells (iPSCs) derived from patients with Extl3 deficiency and cultured to promote HSPC and lymphocyte differentiation displayed diminished expansion of HSPCs and reduced ability to generate lymphoid progenitors.80 These findings indicate that both the overall HS expression and temporal expression of distinct HS modifications have a significant impact on HSPC maintenance and differentiation ex vivo.

Moreover, the presence of distinct surface HS modification patterns can separate erythroid and megakaryocytic-primed MEPs, highlighting the expression of HS modifications as a contributor to hematopoietic cell fate decision, at least at the megakaryocyte-erythroid progenitor level.98 

Granulocyte (neutrophil/basophil/eosinophil) lineage

HSPGs

Serglycin expression has been shown in the Golgi and granules of promyelocytes and other developing immature neutrophil populations.99 The role and content of neutrophil granules seem to heavily rely on serglycin expression and function, including neutrophil elastase loading into granules and alpha-defensin retention during myelopoiesis.100,101 

HS and HS modifications

HS molecules on endothelial cells are critical for interaction with adhesion molecules such as L-selectin on neutrophils to promote migration and intravasation. Yet, much less is known about what roles HS might play on the neutrophil surface. Neutrophil migration in response to antithrombin III, but not fMLP (fMet-Leu-Phe) or interleukin-8 (IL-8) is mediated by HSPGs, suggesting the role of neutrophil HS in chemotaxis is ligand-specific.102 In mice, removal of 2-O-Sulfation on endothelial cells significantly reduced neutrophil migration in response to inflammatory triggers; however, removal of 2-O-Sulfation specifically on neutrophils caused no change in the inflammatory response, suggesting that HS (especially 2-O-Sulfation) on neutrophils is not a driving component in inflammation-mediated neutrophil migration.84 Heparanase is found inside tertiary granules of neutrophils and is secreted to mediate matrix degradation and facilitate intravasation.103,104 Although surface-HS on neutrophils seems to play a minor role in migration as compared with endothelial HS, HS on neutrophils facilitates their activation through binding to cytokines such as IL-8.16 

Eosinophil chemotaxis also heavily relies on HS, in part through the interaction of HS with the eotaxin-1 (CCL11) and eotaxin-3 (CCL26) ligands (Table 1).32,34 Eosinophil major basic protein, the predominant constituent of the crystalline core of eosinophilic granules and mediator of histamine release and protection against helminths and bacteria, not only binds HS but is also a natural inhibitor of heparanase activity (Table 1).105-107 

Although basophils express HS on their surface and their granules contain an abundance of heparin, little is known about the function of HS and HSPGs in these cells. Circulating basophils have been shown to capture viruses such as HIV-1 partially through HS.108 

Mast cell lineage

HSPGs

As with other granule-containing cells, serglycin is an essential component of mast cell integrity and storage and is the predominant HSPG expressed in mast cells.67 

HS and HS modifications

Multiple pieces of evidence suggest that specific HS modifications are needed for proper mast cell function. Deletion of Ndst2 in mice, for example, results in mice harboring mast cells with minimal amounts of granules, which themselves lack heparin and decreased levels of histamine and mast cell proteases.82,109 Furthermore, fetal skin-derived mast cells from mice deficient in 6-O-sulfation ability (Hs6st1/, Hs6st2/, or Hs6st1/Hs6st2/) show decreased protease activity and quantity of specific mast cell proteases (tryptase and carboxypeptidase A, but not chymase) at the protein level, suggesting that 6-O-sulfated HS prevents degradation of specific proteases.85 Mast cells also contain and release heparanase.110 

Monocyte/macrophage lineage

HSPGs

Primary human macrophages have been shown to shed SDC1 and SDC4 from their cell surface, a process accelerated by stromal derived factor-1 stimulation and mediated by matrix-metalloprotease 9 (MMP-9).111 However, while still on the cell surface, these proteoglycans have been shown to bind to the RANTES (regulated upon activation, normal T-cell expressed and presumably secreted) ligand to facilitate signaling.112 SDC2 is also expressed on the surface of macrophages but does not bind to RANTES. Instead, SDC2 regulates FGF2 signaling and the binding of other cytokines to facilitate macrophage–derived growth factor function.113 

Agrin proteoglycan plays a key role in monocyte/macrophage lineage commitment through cell-autonomous interaction with the α-dystroglycan receptor and is required for monocyte/macrophage lineage development.65 Serglycin is also secreted by macrophages and plays a role in tumor necrosis factor alpha (TNF-α) secretion in response to lipopolysaccharide.114 

The polarization of macrophages to either a proinflammatory or adaptive state results in drastic gene expression changes of proteoglycans and the HS machinery.115 Inflammatory macrophages induce the expression of CD44v3, which interacts with FGF2 to facilitate the immune response.116 

HS and HS modifications

Loss of N-sulfation in tissue-resident macrophages of soft tissues has been shown to promote atherosclerosis and obesity, among other biological processes.117 

Erythroid lineage

HSPGs

HS is present in cells of the erythroid lineage and has been shown to play many roles in the biology of erythropoiesis and red blood cell function. Early work on human cell lines identified the presence of HSPGs on the surface of erythroid progenitors.118 TGFBR3 (betaglycan) proteoglycan is expressed during erythropoiesis and distinguishes early and late burst-forming unit erythroid progenitors (BFU-Es).119 The expression of TGFBR3 (and the HS attached to it) likely modulates erythroid development by impacting local TGF-β concentrations. Agrin has been identified on the surface of erythroid cells and induces clustering of EphB1 receptors on erythroblasts. Moreover, Agrin-deficient mice display severe anemia and poorly developed erythroblastic islands among other hematopoietic findings (Table 2),66 suggesting a role in erythroid maturation.

HS and HS modifications

Similarly, mice deficient in HS6ST1 (which catalyzes 6-O-sulfation) are embryonically lethal, and only scarce numbers of embryonic-derived erythroid cells are present in placental tissue sections, indicating the involvement of 6-O-sulfated HS in erythroid production during embryogenesis (Table 2).86 In both mice and humans, erythroid cells display dynamic and temporal expression of specifically modified HS during terminal differentiation, implying that the levels of specific surface HS modifications (including 6-O-sulfation) vary depending on the stage of erythroid differentiation, which may be of functional significance (Figure 3).98 

Figure 3.

The proposed orthogonal application of HS glycotyping with canonical CD-marker flow cytometry in heterogenous populations and the differential and dynamic binding of an HS scFv within megakaryocytic and erythroid lineages. (A) The long-standing use of CD markers has helped identify and isolate hematopoietic cell subpopulations from within heterogenous pools (eg, MEP, CMP, and GMP from a mixed pool of hematopoietic progenitor cells). The addition of HS scFvs to this process can help further purify distinct populations. With nearly limitless possibilities of HS-modification patterns, the combinatorial use of HS scFvs holds immense potential in isolating rare subtypes of hematopoietic cells, possibly even at the single-cell level. (B) Schematic of HS3A8 binding during various stages of megakaryocyte and erythroid differentiation. HS3A8 scFv recognizes specific HS motifs containing sulfation marks (eg, 2-O-, 3-O-, and 6-O-sulfation) at certain residues. MEPs contain a variable level of HS3A8 binding, which when separated into high and low binding, helps delineate megakaryocyte vs erythroid lineage commitment. Furthermore, HS3A8 shows dynamic and temporal binding across the different stages of erythroid differentiation, whereas megakaryocytic cells show low HS3A8 binding regardless of the stage of maturation. Baso-E, basophilic erythroblast; CMP, common myeloid progenitors; GMP, granulocyte-monocyte progenitors; Mk, megakaryocyte; MkP, megakaryocyte progenitor; Ortho-E, orthochromatic erythroblast; Poly-E, polychromatophilic erythroblast; Pro-E, proerythroblast.

Figure 3.

The proposed orthogonal application of HS glycotyping with canonical CD-marker flow cytometry in heterogenous populations and the differential and dynamic binding of an HS scFv within megakaryocytic and erythroid lineages. (A) The long-standing use of CD markers has helped identify and isolate hematopoietic cell subpopulations from within heterogenous pools (eg, MEP, CMP, and GMP from a mixed pool of hematopoietic progenitor cells). The addition of HS scFvs to this process can help further purify distinct populations. With nearly limitless possibilities of HS-modification patterns, the combinatorial use of HS scFvs holds immense potential in isolating rare subtypes of hematopoietic cells, possibly even at the single-cell level. (B) Schematic of HS3A8 binding during various stages of megakaryocyte and erythroid differentiation. HS3A8 scFv recognizes specific HS motifs containing sulfation marks (eg, 2-O-, 3-O-, and 6-O-sulfation) at certain residues. MEPs contain a variable level of HS3A8 binding, which when separated into high and low binding, helps delineate megakaryocyte vs erythroid lineage commitment. Furthermore, HS3A8 shows dynamic and temporal binding across the different stages of erythroid differentiation, whereas megakaryocytic cells show low HS3A8 binding regardless of the stage of maturation. Baso-E, basophilic erythroblast; CMP, common myeloid progenitors; GMP, granulocyte-monocyte progenitors; Mk, megakaryocyte; MkP, megakaryocyte progenitor; Ortho-E, orthochromatic erythroblast; Poly-E, polychromatophilic erythroblast; Pro-E, proerythroblast.

Close modal

Finally, HS on the surface of erythrocytes has also been shown to bind and mediate the entry of Plasmodium falciparum.120,121 

Megakaryocyte lineage

HSPGs

Lysates from human megakaryocytes and megakaryocytic cell lines have been found to contain serglycin and betaglycan; however, the specific roles of these molecules in megakaryocyte biology are yet to be defined.122 Mass spectrometry analysis of platelet extracts identified betaglycan, serglycin, and SDC1, suggesting that proteoglycans produced in megakaryocytes are packaged into platelets.123 Serglycin-deficient mice were found to have normal megakaryocyte function; however, emperipolesis of neutrophils at all stages of maturing megakaryocytes was observed in serglycin-deficient (SG–/–) mice but not in wild-type mice.68 Although platelet counts and the number and histological morphology of granules inside platelets were similar between wild-type and SG–/– mice, mice lacking serglycin showed significantly diminished platelet activation, aggregation, and secretion.68 The diminished platelet content of key platelet factors, such as platelet factor 4, β-thromboglobulin, and platelet–derived growth factor in serglycin-deficient mice is partly due to improper loading into granules.68 SDC4 has been detected in platelets and impacts their aggregation.124 

HS and HS modifications

Although studies on HS in the megakaryocytic lineage are limited, it appears that the lack of or decreased surface HS is a characteristic of megakaryocyte differentiation and commitment (Figure 3).98,125 Consistent with this, transgenic mice overexpressing the heparanase enzyme show increased number of platelets, higher number and percentages of megakaryocytes in tissues, and an increased concentration of plasma thrombopoietin.126 However, these effects may be related to systemic heparanase expression and additional tissue-specific experiments are needed. The functional importance of the lack of or diminished expression of HS on the megakaryocyte cell surface, as well as the role of heparanase in megakaryopoiesis, has yet to be elucidated. Notwithstanding, megakaryocytes are dependent on interactions with HS. For example, the G6b-B receptor is vital for megakaryocyte activity and platelet production and has been shown to function by binding with the HS chains of pericellular perlecan as well as exogenous HS.127 Numerous studies have also demonstrated the importance of exogenous HS (especially sulfated HS) as a ligand for stimulating megakaryopoiesis and platelet activation in conjunction with canonical activators of megakaryopoiesis, such as thrombopoietin.128 

Heparanase is found in abundance in the lysosomes of platelets, where it is stored and enzymatically activated. Platelet heparanase levels are increased under pathogenic conditions, such as sepsis and cancer, and have been implicated in platelet adhesion and thrombogenicity.129 

The B cell/plasma cell lineage

HSPGs

HSPGs are expressed on the surface of B-lineage cells and their tight regulation is critical for numerous B-cell functions. Early pioneering work identified the transient and temporal expression of SDC1 (also known as CD138) within populations during B-cell development, providing the foundation for the use of CD138 as a marker for B-cell populations, including mature plasma cells.130 B-cell activation has also been shown to induce the expression of CD44v3, whose associated HS chains bind HGF and induce Met phosphorylation.131 

HS and HS modifications

HS expressed on the surface of mouse B-cell precursors mediates IL-7–driven B lymphopoiesis, suggesting that HS acts as a binding target for IL-7 and a contributor to B-cell differentiation.132 Moreover, the exogenous addition of HS (either soluble or surface coculture expression) enhances IL-7-mediated early B-cell differentiation in mice.132 Interestingly, however, early B-cell development in humans is IL-7-independent, which corresponds to a lack of SDC1 expression in early human B cells.133 

It has also been noted that expression of HS-modifying enzymes are differentially expressed during specific stages of B-cell development, suggesting that unique HS-modification patterns with potential functional relevance exist within different B-cell subtypes.134 Indeed, the presence of specific modification patterns, particularly epimerization by the C5-epimerase GLCE, has been shown to be dispensable in early B-cell development but critical in the transition between immature and mature B cells, primarily by the binding of APRIL (a proliferation-inducing ligand) to specifically modified HS (Table 1).47 Moreover, specific B-cell subtypes show differential HS-modification patterns. Naïve B cells and antibody-secreting B cells have been shown to express higher levels of N-sulfation than germinal center B cells, and the lack of N-sulfation on germinal center B cells prevents IL-21 binding (Table 1).23 These collective studies suggest that differential HS-modification patterns exist within unique B-cell subtypes that play cell-type–specific roles in hematopoiesis.

In addition to B-cell development, HS appears to also have roles in B-cell function. Surface HS expression in murine B cells was significantly upregulated upon infection by gammaherpesvirus or betaherpesvirus, as well as with stimulation by multiple potent inducers of inflammation (toll-like receptor ligands, B-cell receptor agonists, etc), an effect mediated by the type I interferon receptor.135 

The T-cell lineage

HSPGs

Fetal thymocytes lacking TGFBR3 exhibit higher apoptosis and a delay in T-cell development ex vivo, implicating betaglycan in the maturation and survival of T cells in the thymus.136 Additionally, betaglycan is differentially expressed in various T-cell populations, reduced in regulatory T cells, and is induced upon T-cell receptor activation.137 

The activation of T cells upon antigen presentation occurs at the immunological synapse (the interface between the T cell and antigen-presenting cell) and involves numerous interactions and structural organization. Agrin was found to be differentially expressed in resting and activated primary immune cells and shown to be involved in lipid raft reorganization during T-cell activation.138 Agrin functions at the immunologic synapse, similar to that in monocytes and macrophages, and involves its interaction with the α-dystroglycan receptor.139 Activated T cells also appear to upregulate the surface expression of CD44v3, suggesting HS-involvement in immune processes.140 In contrast, it seems that surface SDC4 facilitates the suppression of T-cell activation through its interactions with dendritic cell-associated HS proteoglycan–dependent integrin ligand on antigen-presenting cells.141 

Cytotoxic (killer) T cells carry toxic granules that facilitate cell destruction during the immune response. Not only is serglycin present in these granules, but it also plays a role in the secretory granule repertoire, regulating its composition.142 

HS and HS modifications

Several studies have suggested that the proliferation of T cells is at least partially dependent on surface HS, and proliferation cues through cytokine signaling are likely orchestrated by HS binding. In both in vitro and transplantation studies of Ext1-deficient fetal thymus, T cells were found to be significantly diminished in number, albeit with no perturbation of T-cell differentiation.143 IL-2-mediated control of T-cell responses and propagation is controlled by HS-bound IL-2, which localizes to the lymphoid organs.144,145 Consistent with this are recent findings that show regulatory T cells (FoxP3+) access local HS-bound IL-2 stores by secreting heparanase.146 

In mice whose T cells lack N-sulfation (through T-cell specific deletion of NDST1 and NDST2), development was not altered (Table 2). However, Ndst-deficient T cells were hyper-responsive to low-level activation, likely due to the release of sequestered IL-2 by surface HS and activation of the IL-2 receptor.79 

T-cell migration into tissues has been found to be heavily mediated by their secretion of heparanase.147 Extracellular matrix degradation of solid tissues by heparanase most likely facilitates T-cell migration.

The interaction between surface HS and T-cell-infecting viruses, including HIV-1, human T-lymphotropic virus 1 (HTLV-1), and severe acute respiratory syndrome coronavirus 2 (SARS-COV-2), is essential for efficient viral infection.148-150 In the case of HIV-1, it has been shown that a conserved arginine in the gp120 protein specifically interacts with 6-O-sulfated HS to facilitate viral entry.148 In addition, HIV-1 Tat protein has been shown to bind HS in a size-dependent manner and requires 2-O-, 6-O-, and N-sulfation for binding.151 HS also binds to HTLV-1 in CD4+ T cells, and desulfation of HS on the surface of T cells drastically reduces infectivity, suggesting that certain sulfation patterns may also be required for HTLV-1 entry.150 Recently, it was discovered that SARS-COV-2 can infect T lymphocytes in an ACE2-independent manner, suggesting alternative receptors for SARS-COV-2 on the surface of T cells, such as HS.152 Given the known interactions between SARS-COV-2 and HS in other cell types, it can be speculated that HS on the surface of T cells may also play a role in SARS-COV-2 infection.149 

NK cells

HSPGs

Similar to cytotoxic T cells and platelets, the secretory granule repertoire of NK cells also depends on the presence of serglycin.142 

HS and HS modifications

NK cells execute their function through germ line-encoded activating and inhibitory surface receptors, including natural cytotoxicity receptors (NCRs), killer-cell lectin-like receptors, and killer-cell immunoglobulin-like receptors. HS not only plays a key role on the surface of NK-cell targets by binding to NCRs, such as NKp30, NKp44, and NKp46153,154 but is also present on the surface of NK cells themselves. Importantly, fine specification of HS modifications on target cells is needed for NK-cell identification and activation. Interestingly, in-cis interactions between HS on the NK-cell surface and NCRs can act to dampen the trigger for NK-cell activation.155 

The orchestrated removal of HS has important implications for tissue homeostasis and function. Heparanase expression and secretion by neutrophils, platelets, T cells, mast cells, and macrophages have been reported.110,147,156-159 Heparanase secretion is required for macrophage activation as part of the inflammation process, both in normal immune function (likely through modulation of toll-like receptor function) and mediation of protumorigenic and prometastatic processes in cancer.156,160 NK cells significantly upregulate heparanase upon activation, which plays a key role in NK-cell invasion into tumors and thereby in tumor progression and metastases.161 Regulatory T cells access the local IL-2 needed for their survival and activity through the secretion of heparanase.146 As previously mentioned, heparanase expression may play a role in megakaryopoiesis and platelet maturation.126 HSC mobilization and engraftment have been shown to be impacted by the lack of HS in the stroma by either the absence of Ext1 or by overexpression of heparanase.162 These collective findings highlight the important balance and regulation of HS expression in hematopoietic cells as part of their biological functions.

The functions of key regulatory pathways that are indispensable for hematopoiesis, such as TGF-β, Wnt, bone morphogenic protein (BMP), and interleukin-driven pathways, as well as the accompanying cytokines, are dependent on HS and the proteoglycans they decorate (Table 1). HSPGs expressed in hematopoietic populations serve cell type-specific functions, including directly facilitating differentiation (eg, SDCs in B cells), establishing concentration gradients needed for differentiation (eg, betaglycan in erythroid populations), promoting migration (glypicans in HSPCs), and maintaining intracellular granule integrity (eg, serglycin in granulocytes, T cells, and mast cells). Although there is no doubt that HS is important for the proper function of hematopoietic tissues, only a few knockout mouse models of HS deficiency/defects show drastic hematopoietic phenotypes (Table 2). The embryonic lethality of many HS-related genes may hamper the investigation of the true importance of specific HS/HSPGs in hematopoiesis, and much more work is needed to elucidate these roles, potentially through the generation of inducible or conditional animal models or the development of specific HS-binding blocking agents.

It is becoming increasingly clear that the composition of HS-modification patterns on the surface of cells is unique and plays a role in defining cell-specific functions, including those within the hematopoietic system (Figure 2B). Barriers in the high-resolution detection and characterization of specific HS-modification patterns on cell surfaces, both in vitro and in vivo, have hindered the understanding of how distinct modifications may play detailed roles in cell-cell interactions, cytokine binding, viral entry, and other key biological functions within hematopoietic cells. Technologies such as mass spectrometry are becoming gradually more successful in identifying sequences of HS modifications, and increasingly complex and diverse arrays of HS oligosaccharides with distinct modification patterns are being created to test binding/interactions. Single-chain variable fragment antibodies (scFvs) against specific HS-modification patterns were isolated from a phage display library and have been used to identify the presence of specific HS-modification patterns in tissues.163-165 These studies revealed astounding specificity of cellular expression of HS-modification patterns, which in some instances appeared to be single-cell specific. Based on these observations, scFvs have recently been applied to the analysis of cells of the hematopoietic lineage. Specifically, they were incorporated into multiparameter flow cytometry in combination with established CD-marker gating schemes to establish cellular glycotypes within hematopoietic cells of mice and humans, providing a platform to characterize systematically HS-modification patterns in individual hematopoietic cell populations.98 Furthermore, this portable HS glycotyping system can serve as a tool to isolate living cells based on distinct HS patterns for downstream functional utilization and characterization. Although this can be applied to all tissues across species, the strong reliance on flow cytometry in hematopoietic tissues, such as bone marrow and blood, makes HS glycotyping a tool with immense potential. With near limitless possible combinations of HS-modification patterns, one can speculate that even more specialized and rare subpopulations of hematopoietic cells exist that currently cannot be defined or purified using existing technologies, as suggested by current destructive technologies such as single-cell RNA sequencing (Figure 3A). The detection of HS-modification patterns through glycotyping not only holds potential in further isolating distinct populations but also allows for the interrogation of the importance of HS modifications in hematopoietic functions, including lineage commitment. For example, HS3A8 scFv, which recognizes distinct combinations of HS modifications, not only helps separate MEPs from other progenitor types but also illustrates the dynamic HS-modification patterning throughout megakaryocyte and erythroid differentiation and development (Figure 3B).98 Deciphering the composition of HS modifications on the surface of cells through various approaches will play a transformative role in uncovering highly specialized and unique functions that contribute to the biology and pathobiology of hematopoietic and other tissues.

The authors acknowledge the Creative Services Department at the Albert Einstein College of Medicine, specifically Tatyana Harris, for assistance with figure illustrations.

Work conducted in the Bülow and Steidl laboratories is supported by grants from the National Institutes of Health (NIH), National Cancer Institute (U01CA241981 [H.E.B.], U01CA241981, and R35CA253127 [U.S.]), and the NIH, National Institute of Neurological Disorders and Stroke (R01NS125134 and R01NS129992 [H.E.B.]). U.S. holds the Edward P. Evans Endowed Professorship for Myelodysplastic Syndromes at Albert Einstein College of Medicine. The Endowed Professorship was supported by a grant from the Edward P. Evans Foundation.

Contribution: R.T.P., H.E.B., and U.S. wrote the manuscript.

Conflict-of-interest disclosure: The authors disclose the patent application “Antibody-based method to identify, purify, and manipulate cell types and processes” (US patent publication no.: US 2022/0227886 A1). U.S. has received research funding from GlaxoSmithKline, Bayer Healthcare, Aileron Therapeutics, and Novartis; has received compensation for consultancy services and for serving on scientific advisory boards from GlaxoSmithKline, Bayer Healthcare, Celgene, Aileron Therapeutics, Novartis, Stelexis Therapeutics, Pieris Pharmaceuticals, and Trillium Therapeutics; and has equity ownership in and serves on the board of directors of Stelexis Therapeutics.

Correspondence: Ulrich Steidl, Albert Einstein College of Medicine, Chanin Building Room #601-605, 1300 Morris Park Ave, Bronx, NY 10461; email: ulrich.steidl@einsteinmed.edu.

1.
Esko
JD
,
Selleck
SB
.
Order out of chaos: assembly of ligand binding sites in heparan sulfate
.
Annu Rev Biochem
.
2002
;
71
:
435
-
471
.
2.
Sarrazin
S
,
Lamanna
WC
,
Esko
JD
.
Heparan sulfate proteoglycans
.
Cold Spring Harb Perspect Biol
.
2011
;
3
(
7
):
a004952
.
3.
Bülow
HE
,
Hobert
O
.
The molecular diversity of glycosaminoglycans shapes animal development
.
Annu Rev Cell Dev Biol
.
2006
;
22
:
375
-
407
.
4.
Xu
D
,
Esko
JD
.
Demystifying heparan sulfate–protein interactions
.
Annu Rev Biochem
.
2014
;
83
:
129
-
157
.
5.
Iozzo
RV
,
Schaefer
L
.
Proteoglycan form and function: a comprehensive nomenclature of proteoglycans
.
Matrix Biol
.
2015
;
42
:
11
-
55
.
6.
Papy-Garcia
D
,
Albanese
P
.
Heparan sulfate proteoglycans as key regulators of the mesenchymal niche of hematopoietic stem cells
.
Glycoconj J
.
2017
;
34
(
3
):
377
-
391
.
7.
Roberts
R
,
Gallagher
J
,
Spooncer
E
,
Allen
TD
,
Bloomfield
F
,
Dexter
TM
.
Heparan sulphate bound growth factors: a mechanism for stromal cell mediated haemopoiesis
.
Nature
.
1988
;
332
(
6162
):
376
-
378
.
8.
Rodgers
KD
,
San Antonio
JD
,
Jacenko
O
.
Heparan sulfate proteoglycans: a GAGgle of skeletal-hematopoietic regulators
.
Dev Dyn
.
2008
;
237
(
10
):
2622
-
2642
.
9.
Ramsden
L
,
Rider
CC
.
Selective and differential binding of interleukin (IL)-1α, IL-1β, IL-2 and IL-6 to glycosaminoglycans
.
Eur J Immunol
.
1992
;
22
(
11
):
3027
-
3031
.
10.
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
.
1998
;
92
(
12
):
4641
-
4651
.
11.
Lortat-Jacob
H
,
Garrone
P
,
Banchereau
J
,
Grimaud
J-A
.
Human interleukin 4 is a glycosaminoglycan-binding protein
.
Cytokine
.
1997
;
9
(
2
):
101
-
105
.
12.
den Dekker
E
,
Grefte
S
,
Huijs
T
, et al
.
Monocyte cell surface glycosaminoglycans positively modulate IL-4-induced differentiation toward dendritic cells
.
J Immunol
.
2008
;
180
(
6
):
3680
-
3688
.
13.
Lipscombe
R J
,
Nakhoul
AM
,
Sanderson
CJ
,
Coombe
DR
.
Interleukin-5 binds to heparin/heparan sulfate. a model for an interaction with extracellular matrix
.
J Leukoc Biol
.
1998
;
63
(
3
):
342
-
350
.
14.
Mummery
RS
,
Rider
CC
.
Characterization of the heparin-binding properties of IL-6
.
J Immunol
.
2000
;
165
(
10
):
5671
-
5679
.
15.
Clarke
D
,
Katoh
O
,
Gibbs
R
,
Griffiths
S
,
Gordon
M
.
Interaction of interleukin 7 (IL-7) with glycosaminoglycans and its biological relevance
.
Cytokine
.
1995
;
7
(
4
):
325
-
330
.
16.
Webb
L
,
Ehrengruber
MU
,
Clark-Lewis
I
,
Baggiolini
M
,
Rot
A
.
Binding to heparan sulfate or heparin enhances neutrophil responses to interleukin 8
.
Proc Natl Acad Sci U S A
.
1993
;
90
(
15
):
7158
-
7162
.
17.
Pichert
A
,
Samsonov
SA
,
Theisgen
S
, et al
.
Characterization of the interaction of interleukin-8 with hyaluronan, chondroitin sulfate, dermatan sulfate and their sulfated derivatives by spectroscopy and molecular modeling
.
Glycobiology
.
2012
;
22
(
1
):
134
-
145
.
18.
Zong
C
,
Venot
A
,
Li
X
, et al
.
Heparan sulfate microarray reveals that heparan sulfate–protein binding exhibits different ligand requirements
.
J Am Chem Soc
.
2017
;
139
(
28
):
9534
-
9543
.
19.
Salek-Ardakani
S
,
Arrand
JR
,
Shaw
D
,
Mackett
M
.
Heparin and heparan sulfate bind interleukin-10 and modulate its activity
.
Blood
.
2000
;
96
(
5
):
1879
-
1888
.
20.
Künze
G
,
Gehrcke
J-P
,
Pisabarro
MT
,
Huster
D
.
NMR characterization of the binding properties and conformation of glycosaminoglycans interacting with interleukin-10
.
Glycobiology
.
2014
;
24
(
11
):
1036
-
1049
.
21.
Hasan
M
,
Najjam
S
,
Gordon
MY
,
Gibbs
RV
,
Rider
CC
.
IL-12 is a heparin-binding cytokine
.
J Immunol
.
1999
;
162
(
2
):
1064
-
1070
.
22.
Reeves
EP
,
Williamson
M
,
Byrne
B
, et al
.
IL-8 dictates glycosaminoglycan binding and stability of IL-18 in cystic fibrosis
.
J Immunol
.
2010
;
184
(
3
):
1642
-
1652
.
23.
Chen
Z
,
Cui
Y
,
Yao
Y
, et al
.
Heparan sulfate regulates IL-21 bioavailability and signal strength that control germinal center B cell selection and differentiation
.
Sci Immunol
.
2023
;
8
(
80
):
eadd1728
.
24.
Lau
EK
,
Paavola
CD
,
Johnson
Z
, et al
.
Identification of the glycosaminoglycan binding site of the CC chemokine, MCP-1: implications for structure and function in vivo
.
J Biol Chem
.
2004
;
279
(
21
):
22294
-
22305
.
25.
Sweeney
MD
,
Yu
Y
,
Leary
JA
.
Effects of sulfate position on heparin octa saccharide binding to CCL2 examined by tandem mass spectrometry
.
J Am Soc Mass Spectrom
.
2006
;
17
(
8
):
1114
-
1119
.
26.
Schenauer
MR
,
Yu
Y
,
Sweeney
MD
,
Leary
JA
.
CCR2 chemokines bind selectively to acetylated heparan sulfate octa saccharides
.
J Biol Chem
.
2007
;
282
(
35
):
25182
-
25188
.
27.
Gray
AL
,
Karlsson
R
,
Roberts
ARE
, et al
.
Chemokine CXCL4 interactions with extracellular matrix proteoglycans mediate widespread immune cell recruitment independent of chemokine receptors
.
Cell Rep
.
2023
;
42
(
1
):
111930
.
28.
Koopmann
W
,
Ediriwickrema
C
,
Krangel
MS
.
Structure and function of the glycosaminoglycan binding site of chemokine macrophage-inflammatory protein-1β
.
J Immunol
.
1999
;
163
(
4
):
2120
-
2127
.
29.
Mbemba
E
,
Slimani
H
,
Atemezem
A
,
Saffar
L
,
Gattegno
L
.
Glycans are involved in RANTES binding to CCR5 positive as well as to CCR5 negative cells
.
Biochim Biophys Acta
.
2001
;
1510
(
1-2
):
354
-
366
.
30.
Ali
S
,
Robertson
H
,
Wain
JH
,
Isaacs
JD
,
Malik
G
,
Kirby
JA
.
A non-glycosaminoglycan-binding variant of CC chemokine ligand 7 (monocyte chemoattractant protein-3) antagonizes chemokine-mediated inflammation
.
J Immunol
.
2005
;
175
(
2
):
1257
-
1266
.
31.
Cochran
S
,
Li
CP
,
Ferro
V
.
A surface plasmon resonance-based solution affinity assay for heparan sulfate-binding proteins
.
Glycoconj J
.
2009
;
26
(
5
):
577
-
587
.
32.
Ellyard
JI
,
Simson
L
,
Bezos
A
,
Johnston
K
,
Freeman
C
,
Parish
CR
.
Eotaxin selectively binds heparin: an interaction that protects eotaxin from proteolysis and potentiates chemotactic activity in vivo
.
J Biol Chem
.
2007
;
282
(
20
):
15238
-
15247
.
33.
Culley
FJ
,
Fadlon
EJ
,
Kirchem
A
,
Williams
TJ
,
Jose
PJ
,
Pease
JE
.
Proteoglycans are potent modulators of the biological responses of eosinophils to chemokines
.
Eur J Immunol
.
2003
;
33
(
5
):
1302
-
1310
.
34.
Pum
A
,
Ennemoser
M
,
Gerlza
T
,
Kungl
AJ
.
The role of heparan sulfate in CCL26-induced eosinophil chemotaxis
.
Int J Mol Sci
.
2022
;
23
(
12
):
6519
.
35.
Kishimoto
S
,
Nakamura
S
,
Hattori
H
, et al
.
Human stem cell factor (SCF) is a heparin-binding cytokine
.
J Biochem
.
2009
;
145
(
3
):
275
-
278
.
36.
Gordon
MY
,
Riley
GP
,
Watt
SM
,
Greaves
MF
.
Compartmentalization of a hematopoietic growth factor (GM-CSF) by glycosaminoglycans in the bone marrow microenvironment
.
Nature
.
1987
;
326
(
6111
):
403
-
405
.
37.
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
.
1994
;
269
(
15
):
11216
-
11223
.
38.
Karlsson
R
,
Chopra
P
,
Joshi
A
, et al
.
Dissecting structure-function of 3-O-sulfated heparin and engineered heparan sulfates
.
Sci Adv
.
2021
;
7
(
52
):
eabl6026
.
39.
Murphy
JW
,
Cho
Y
,
Sachpatzidis
A
,
Fan
C
,
Hodsdon
ME
,
Lolis
E
.
Structural and functional basis of CXCL12 (stromal cell-derived factor-1α) binding to heparin
.
J Biol Chem
.
2007
;
282
(
13
):
10018
-
10027
.
40.
Monneau
YR
,
Luo
L
,
Sankaranarayanan
NV
, et al
.
Solution structure of CXCL13 and heparan sulfate binding show that GAG binding site and cellular signaling rely on distinct domains
.
Open Biol
.
2017
;
7
(
10
):
170133
.
41.
Kreuger
J
,
Prydz
K
,
Pettersson
RF
,
Lindahl
U
,
Salmivirta
M
.
Characterization of fibroblast growth factor 1 binding heparan sulfate domain
.
Glycobiology
.
1999
;
9
(
7
):
723
-
729
.
42.
Yayon
A
,
Klagsbrun
M
,
Esko
JD
,
Leder
P
,
Ornitz
DM
.
Cell surface, heparin-like molecules are required for binding of basic fibroblast growth factor to its high affinity receptor
.
Cell
.
1991
;
64
(
4
):
841
-
848
.
43.
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
.
1992
;
267
(
9
):
6093
-
6098
.
44.
Robinson
CJ
,
Mulloy
B
,
Gallagher
JT
,
Stringer
SE
.
VEGF165-binding sites within heparan sulfate encompass two highly sulfated domains and can be liberated by K5 lyase
.
J Biol Chem
.
2006
;
281
(
3
):
1731
-
1740
.
45.
Higashiyama
S
,
Abraham
JA
,
Miller
J
,
Fiddes
JC
,
Klagsbrun
M
.
A heparin-binding growth factor secreted by macrophage-like cells that is related to EGF
.
Science
.
1991
;
251
(
4996
):
936
-
939
.
46.
Feyzi
E
,
Lustig
F
,
Fager
G
,
Spillmann
D
,
Lindahl
U
,
Salmivirta
M
.
Characterization of heparin and heparan sulfate domains binding to the long splice variant of platelet-derived growth factor A chain
.
J Biol Chem
.
1997
;
272
(
9
):
5518
-
5524
.
47.
Reijmers
RM
,
Groen
RWJ
,
Kuil
A
, et al
.
Disruption of heparan sulfate proteoglycan conformation perturbs B-cell maturation and APRIL-mediated plasma cell survival
.
Blood
.
2011
;
117
(
23
):
6162
-
6171
.
48.
Hendriks
J
,
Planelles
L
,
de Jong-Odding
J
, et al
.
Heparan sulfate proteoglycan binding promotes APRIL-induced tumor cell proliferation
.
Cell Death Differ
.
2005
;
12
(
6
):
637
-
648
.
49.
Luster
AD
,
Greenberg
SM
,
Leder
P
.
The IP-10 chemokine binds to a specific cell surface heparan sulfate site shared with platelet factor 4 and inhibits endothelial cell proliferation
.
J Exp Med
.
1995
;
182
(
1
):
219
-
231
.
50.
Lortat-Jacob
H
,
Grimaud
J-A
.
Interferon-γ binds to heparan sulfate by a cluster of amino acids located in the C-terminal part of the molecule
.
FEBS Lett
.
1991
;
280
(
1
):
152
-
154
.
51.
Lortat-Jacob
H
,
Grimaud
J-A
.
Binding of interferon-gamma to heparan sulfate is restricted to the heparin-like domains ad involves carboxylic-but not N-sulfated-groups
.
Biochim Biophys Acta
.
1992
;
1117
(
2
):
126
-
130
.
52.
Lyon
M
,
Rushton
G
,
Gallagher
JT
.
The interaction of the transforming growth factor-βs with heparin/heparan sulfate is isoform-specific
.
J Biol Chem
.
1997
;
272
(
29
):
18000
-
18006
.
53.
McCaffrey
TA
,
Falcone
DJ
,
Du
B
.
Transforming growth factor-β1 is a heparin-binding protein: identification of putative heparin-binding regions and isolation of heparins with varying affinity for TGF-β1
.
J Cell Physiol
.
1992
;
152
(
2
):
430
-
440
.
54.
Hu
Z
,
Wang
C
,
Xiao
Y
, et al
.
NDST1-dependent heparan sulfate regulates BMP signaling and internalization in lung development
.
J Cell Sci
.
2009
;
122
(
Pt 8
):
1145
-
1154
.
55.
Denardo
A
,
Elli
S
,
Federici
S
, et al
.
BMP6 binding to heparin and heparan sulfate is mediated by N-terminal and C-terminal clustered basic residues
.
Biochim Biophys Acta Gen Subj
.
2021
;
1865
(
2
):
129799
.
56.
Irie
A
,
Habuchi
H
,
Kimata
K
,
Sanai
Y
.
Heparan sulfate is required for bone morphogenetic protein-7 signaling
.
Biochem Biophys Res Commun
.
2003
;
308
(
4
):
858
-
865
.
57.
Paine-Saunders
S
,
Viviano
BL
,
Economides
AN
,
Saunders
S
.
Heparan sulfate proteoglycans retain Noggin at the cell surface: a potential mechanism for shaping bone morphogenetic protein gradients
.
J Biol Chem
.
2002
;
277
(
3
):
2089
-
2096
.
58.
Viviano
BL
,
Paine-Saunders
S
,
Gasiunas
N
,
Gallagher
J
,
Saunders
S
.
Domain-specific modification of heparan sulfate by Qsulf1 modulates the binding of the bone morphogenetic protein antagonist Noggin
.
J Biol Chem
.
2004
;
279
(
7
):
5604
-
5611
.
59.
Wang
Y
,
Liu
X
,
Obser
T
, et al
.
Heparan sulfate dependent binding of plasmatic von Willebrand factor to blood circulating melanoma cells attenuates metastasis
.
Matrix Biol
.
2022
;
111
:
76
-
94
.
60.
Majka
M
,
Janowska-Wieczorek
A
,
Ratajczak
J
, et al
.
Numerous growth factors, cytokines, and chemokines are secreted by human CD34+ cells, myeloblasts, erythroblasts, and megakaryoblasts and regulate normal hematopoiesis in an autocrine/paracrine manner
.
Blood
.
2001
;
97
(
10
):
3075
-
3085
.
61.
Lortat-Jacob
H
,
Grosdidier
A
,
Imberty
A
.
Structural diversity of heparan sulfate binding domains in chemokines
.
Proc Natl Acad Sci U S A
.
2002
;
99
(
3
):
1229
-
1234
.
62.
Hook
M
,
Bjork
I
,
Hopwood
J
,
Lindahl
U
.
Anticoagulant activity of heparin: separation of high-activity and low-activity heparin species by affinity chromatography on immobilized antithrombin
.
FEBS Lett
.
1976
;
66
(
1
):
90
-
93
.
63.
Lam
L
,
Silbert
J
,
Rosenberg
R
.
The separation of active and inactive forms of heparin
.
Biochem Biophys Res Commun
.
1976
;
69
(
2
):
570
-
577
.
64.
Lindahl
U
,
Bäckström
G
,
Thunberg
L
,
Leder
IG
.
Evidence for a 3-O-sulfated D-glucosamine residue in the antithrombin-binding sequence of heparin
.
Proc Natl Acad Sci U S A
.
1980
;
77
(
11
):
6551
-
6555
.
65.
Mazzon
C
,
Anselmo
A
,
Soldani
C
, et al
.
Agrin is required for survival and function of monocytic cells
.
Blood
.
2012
;
119
(
23
):
5502
-
5511
.
66.
Anselmo
A
,
Lauranzano
E
,
Soldani
C
, et al
.
Identification of a novel agrin-dependent pathway in cell signaling and adhesion within the erythroid niche
.
Cell Death Differ
.
2016
;
23
(
8
):
1322
-
1330
.
67.
Abrink
M
,
Grujic
M
,
Pejler
G
.
Serglycin is essential for maturation of mast cell secretory granule
.
J Biol Chem
.
2004
;
279
(
39
):
40897
-
40905
.
68.
Woulfe
DS
,
Lilliendahl
JK
,
August
S
, et al
.
Serglycin proteoglycan deletion induces defects in platelet aggregation and thrombus formation in mice
.
Blood
.
2008
;
111
(
7
):
3458
-
3467
.
69.
Grujic
M
,
Braga
T
,
Lukinius
A
, et al
.
Serglycin-deficient cytotoxic T lymphocytes display defective secretory granule maturation and granzyme B storage
.
J Biol Chem
.
2005
;
280
(
39
):
33411
-
33418
.
70.
Wernersson
S
,
Braga
T
,
Sawesi
O
, et al
.
Age-related enlargement of lymphoid tissue and altered leukocyte composition in serglycin-deficient mice
.
J Leukoc Biol
.
2009
;
85
(
3
):
401
-
408
.
71.
Khurana
S
,
Margamuljana
L
,
Joseph
C
,
Schouteden
S
,
Buckley
SM
,
Verfaillie
CM
.
Glypican-3–mediated inhibition of CD26 by TFPI: a novel mechanism in hematopoietic stem cell homing and maintenance
.
Blood
.
2013
;
121
(
14
):
2587
-
2595
.
72.
Viviano
BL
,
Silverstein
L
,
Pflederer
C
,
Paine-Saunders
S
,
Mills
K
,
Saunders
S
.
Altered hematopoiesis in glypican-3-deficient mice results in decreased osteoclast differentiation and a delay in endochondral ossification
.
Dev Biol
.
2005
;
282
(
1
):
152
-
162
.
73.
Spinler
K
,
Bajaj
J
,
Ito
T
, et al
.
A stem cell reporter-based platform to identify and target drug resistant stem cells in myeloid leukemia
.
Nat Commun
.
2020
;
11
:
5998
. 15.
74.
Lamichhane
BS
,
Bisgrove
BW
,
Su
Y-C
,
Demarest
BL
,
Yost
HJ
.
Syndecan 2 regulates hematopoietic lineages and infection resolution in zebrafish
.
bioRxiv
.
Preprint posted online 5 May 2020
.
75.
Endo
T
,
Ito
K
,
Morimoto
J
, et al
.
Syndecan 4 regulation of the development of autoimmune arthritis in mice by modulating B cell migration and germinal center formation
.
Arthritis Rheumatol
.
2015
;
67
(
9
):
2512
-
2522
.
76.
Grigorian
M
,
Liu
T
,
Banerjee
U
,
Hartenstein
V
.
The proteoglycan Trol controls the architecture of the extracellular matrix and balances proliferation and differentiation of blood progenitors in the Drosophila lymph gland
.
Dev Biol
.
2013
;
384
(
2
):
301
-
312
.
77.
Raman
C
,
Sestero
CM
,
Orlandella
RM
, et al
.
TGF-β receptor 3 (betaglycan) regulates Th1 differentiation and T dependent B cell responses
.
J Immunol
.
2016
;
196
(
suppl 1
):
189.12
.
78.
Shekels
LL
,
Buelt-Gebhardt
M
,
Gupta
P
.
Effect of systemic heparan sulfate haploinsufficiency on steady state hematopoiesis and engraftment of hematopoietic stem cells
.
Blood Cells Mol Dis
.
2015
;
55
(
1
):
3
-
9
.
79.
Garner
OB
,
Yamaguchi
Y
,
Esko
JD
,
Videm
V
.
Small changes in lymphocyte development and activation in mice through tissue-specific alteration of heparan sulphate
.
Immunology
.
2008
;
125
(
3
):
420
-
429
.
80.
Volpi
S
,
Yamazaki
Y
,
Brauer
PM
, et al
.
EXTL3 mutations cause skeletal dysplasia, immune deficiency, and developmental delay PID links heparan sulfate to thymopoiesis
.
J Exp Med
.
2017
;
214
(
3
):
623
-
637
.
81.
Wang
L
,
Fuster
M
,
Sriramarao
P
,
Esko
JD
.
Endothelial heparan sulfate deficiency impairs L-selectin-and chemokine-mediated neutrophil trafficking during inflammatory responses
.
Nat Immunol
.
2005
;
6
(
9
):
902
-
910
.
82.
Forsberg
E
,
Pejler
G
,
Ringvall
M
, et al
.
Abnormal mast cells in mice deficient in a heparin-synthesizing enzyme
.
Nature
.
1999
;
400
(
6746
):
773
-
776
.
83.
Pallerla
SR
,
Lawrence
R
,
Lewejohann
L
, et al
.
Altered heparan sulfate structure in mice with deleted NDST3 gene function
.
J Biol Chem
.
2008
;
283
(
24
):
16885
-
16894
.
84.
Axelsson
J
,
Xu
D
,
Kang
BN
, et al
.
Inactivation of heparan sulfate 2-O-sulfotransferase accentuates neutrophil infiltration during acute inflammation in mice
.
Blood
.
2012
;
120
(
8
):
1742
-
1751
.
85.
Anower-E-Khuda
MF
,
Habuchi
H
,
Nagai
N
,
Habuchi
O
,
Yokochi
T
,
Kimata
K
.
Heparan sulfate 6-O-sulfotransferase isoform-dependent regulatory effects of heparin on the activities of various proteases in mast cells and the biosynthesis of 6-O-sulfated heparin
.
J Biol Chem
.
2013
;
288
(
6
):
3705
-
3717
.
86.
Habuchi
H
,
Nagai
N
,
Sugaya
N
,
Atsumi
F
,
Stevens
RL
,
Kimata
K
.
Mice deficient in heparan sulfate 6-O-sulfotransferase-1 exhibit defective heparan sulfate biosynthesis, abnormal placentation, and late embryonic lethality
.
J Biol Chem
.
2007
;
282
(
21
):
15578
-
15588
.
87.
Benhamron
S
,
Reiner
I
,
Zcharia
E
, et al
.
Dissociation between mature phenotype and impaired transmigration in dendritic cells from heparanase-deficient mice
.
PLoS One
.
2012
;
7
(
5
):
e35602
.
88.
Poon
I K
,
Goodall
KJ
,
Phipps
S
, et al
.
Mice deficient in heparanase exhibit impaired dendritic cell migration and reduced airway inflammation
.
Eur J Immunol
.
2014
;
44
(
4
):
1016
-
1030
.
89.
Jackson
DG
,
Bell
JI
,
Dickinson
R
,
Timans
J
,
Shields
J
,
Whittle
N
.
Proteoglycan forms of the lymphocyte homing receptor CD44 are alternatively spliced variants containing the v3 exon
.
J Cell Biol
.
1995
;
128
(
4
):
673
-
685
.
90.
Forsberg
EC
,
Prohaska
SS
,
Katzman
S
,
Heffner
GC
,
Stuart
JM
,
Weissman
IL
.
Differential expression of novel potential regulators in hematopoietic stem cells
.
PLoS Genet
.
2005
;
1
(
3
):
e28
.
91.
Termini
CM
,
Pang
A
,
Li
M
,
Fang
T
,
Chang
VY
,
Chute
JP
.
Syndecan-2 enriches for hematopoietic stem cells and regulates stem cell repopulating capacity
.
Blood
.
2022
;
139
(
2
):
188
-
204
.
92.
Murphy
AJ
,
Akhtari
M
,
Tolani
S
, et al
.
ApoE regulates hematopoietic stem cell proliferation, monocytosis, and monocyte accumulation in atherosclerotic lesions in mice
.
J Clin Invest
.
2011
;
121
(
10
):
4138
-
4149
.
93.
Minguell
JJ
,
Tavassoli
M
.
Proteoglycan synthesis by hematopoietic progenitor cells
.
Blood
.
1989
;
73
(
7
):
1821
-
1827
.
94.
Randrianarison-Huetz
V
,
Laurent
B
,
Bardet
V
,
Blobe
GC
,
Huetz
F
,
Duménil
D
.
Gfi-1B controls human erythroid and megakaryocytic differentiation by regulating TGF-β signaling at the bipotent erythro-megakaryocytic progenitor stage
.
Blood
.
2010
;
115
(
14
):
2784
-
2795
.
95.
Baldwin
RJ
,
ten Dam
GB
,
van Kuppevelt
TH
, et al
.
A developmentally regulated heparan sulfate epitope defines a subpopulation with increased blood potential during mesodermal differentiation
.
Stem Cells
.
2008
;
26
(
12
):
3108
-
3118
.
96.
Holley
RJ
,
Pickford
CE
,
Rushton
G
, et al
.
Influencing hematopoietic differentiation of mouse embryonic stem cells using soluble heparin and heparan sulfate saccharides
.
J Biol Chem
.
2011
;
286
(
8
):
6241
-
6252
.
97.
Gupta
P
,
Oegema
TR
,
Brazil
JJ
,
Dudek
AZ
,
Slungaard
A
,
Verfaillie
CM
.
Human LTC-IC can be maintained for at least 5 weeks in vitro when interleukin-3 and a single chemokine are combined with O-sulfated heparan sulfates: requirement for optimal binding interactions of heparan sulfate with early-acting cytokines and matrix proteins
.
Blood
.
2000
;
95
(
1
):
147
-
155
.
98.
Piszczatowski
RT
,
Schwenger
E
,
Sundaravel
S
, et al
.
A glycan-based approach to cell characterization and isolation: hematopoiesis as a paradigm
.
J Exp Med
.
2022
;
219
(
11
):
e20212552
.
99.
Niemann
CU
,
Cowland
JB
,
Klausen
P
,
Askaa
J
,
Calafat
J
,
Borregaard
N
.
Localization of serglycin in human neutrophil granulocytes and their precursors
.
J Leukoc Biol
.
2004
;
76
(
2
):
406
-
415
.
100.
Niemann
CU
,
Abrink
M
,
Pejler
G
, et al
.
Neutrophil elastase depends on serglycin proteoglycan for localization in granules
.
Blood
.
2007
;
109
(
10
):
4478
-
4486
.
101.
Glenthøj
A
,
Cowland
JB
,
Heegaard
NH
,
Larsen
MT
,
Borregaard
N
.
Serglycin participates in retention of α-defensin in granules during myelopoiesis
.
Blood
.
2011
;
118
(
16
):
4440
-
4448
.
102.
Dunzendorfer
S
,
Kaneider
N
,
Rabensteiner
A
, et al
.
Cell-surface heparan sulfate proteoglycan–mediated regulation of human neutrophil migration by the serpin antithrombin III
.
Blood
.
2001
;
97
(
4
):
1079
-
1085
.
103.
Matzner
Y
,
Bar-Ner
M
,
Yahalom
J
,
Ishai-Michaeli
R
,
Fuks
Z
,
Vlodavsky
I
.
Degradation of heparan sulfate in the subendothelial extracellular matrix by a readily released heparanase from human neutrophils. possible role in invasion through basement membranes
.
J Clin Invest
.
1985
;
76
(
4
):
1306
-
1313
.
104.
MOLLINEDO
F
,
Nakajima
M
,
Llorens
A
, et al
.
Major co-localization of the extracellular-matrix degradative enzymes heparanase and gelatinase in tertiary granules of human neutrophils
.
Biochem J
.
1997
;
327
(
3
):
917
-
923
.
105.
Glerup
S
,
Kløverpris
S
,
Oxvig
C
.
The proform of the eosinophil major basic protein binds the cell surface through a site distinct from its C-type lectin ligand-binding region
.
J Biol Chem
.
2006
;
281
(
42
):
31509
-
31516
.
106.
Swaminathan
GJ
,
Myszka
DG
,
Katsamba
PS
,
Ohnuki
LE
,
Gleich
GJ
,
Acharya
KR
.
Eosinophil-granule major basic protein, a C-type lectin, binds heparin
.
Biochemistry
.
2005
;
44
(
43
):
14152
-
14158
.
107.
Temkin
V
,
Aingorn
H
,
Puxeddu
I
, et al
.
Eosinophil major basic protein: first identified natural heparanase-inhibiting protein
.
J Allergy Clin Immunol
.
2004
;
113
(
4
):
703
-
709
.
108.
Jiang
A-P
,
Jiang
JF
,
Guo
MG
,
Jin
YM
,
Li
YY
,
Wang
JH
.
Human blood-circulating basophils capture HIV-1 and mediate viral trans-infection of CD4+ T cells
.
J Virol
.
2015
;
89
(
15
):
8050
-
8062
.
109.
Humphries
DE
,
Wong
GW
,
Friend
DS
, et al
.
Heparin is essential for the storage of specific granule proteases in mast cells
.
Nature
.
1999
;
400
(
6746
):
769
-
772
.
110.
Bashkin
P
,
Razin
E
,
Eldor
A
,
Vlodavsky
I
.
Degranulating mast cells secrete an endoglycosidase that degrades heparan sulfate in subendothelial extracellular matrix
.
Blood
.
1990
;
75
(
11
):
2204
-
2212
.
111.
Brule
S
,
Charnaux
N
,
Sutton
A
, et al
.
The shedding of syndecan-4 and syndecan-1 from HeLa cells and human primary macrophages is accelerated by SDF-1/CXCL12 and mediated by the matrix metalloproteinase-9
.
Glycobiology
.
2006
;
16
(
6
):
488
-
501
.
112.
Slimani
H
,
Charnaux
N
,
Mbemba
E
, et al
.
Interaction of RANTES with syndecan-1 and syndecan-4 expressed by human primary macrophages
.
Biochim Biophys Acta
.
2003
;
1617
(
1-2
):
80
-
88
.
113.
Clasper
S
,
Vekemans
S
,
Fiore
M
, et al
.
Inducible expression of the cell surface heparan sulfate proteoglycan syndecan-2 (fibroglycan) on human activated macrophages can regulate fibroblast growth factor action
.
J Biol Chem
.
1999
;
274
(
34
):
24113
-
24123
.
114.
Zernichow
L
,
Åbrink
M
,
Hallgren
J
,
Grujic
M
,
Pejler
G
,
Kolset
SO
.
Serglycin is the major secreted proteoglycan in macrophages and has a role in the regulation of macrophage tumor necrosis factor-α secretion in response to lipopolysaccharide
.
J Biol Chem
.
2006
;
281
(
37
):
26792
-
26801
.
115.
Martinez
P
,
Denys
A
,
Delos
M
, et al
.
Macrophage polarization alters the expression and sulfation pattern of glycosaminoglycans
.
Glycobiology
.
2015
;
25
(
5
):
502
-
513
.
116.
Jones
M
,
Tussey
L
,
Athanasou
N
,
Jackson
DG
.
Heparan sulfate proteoglycan isoforms of the CD44 hyaluronan receptor induced in human inflammatory macrophages can function as paracrine regulators of fibroblast growth factor action
.
J Biol Chem
.
2000
;
275
(
11
):
7964
-
7974
.
117.
Gordts
PL
,
Foley
EM
,
Lawrence
R
, et al
.
Reducing macrophage proteoglycan sulfation increases atherosclerosis and obesity through enhanced type I interferon signaling
.
Cell Metab
.
2014
;
20
(
5
):
813
-
826
.
118.
Drzeniek
Z
,
Stöcker
G
,
Siebertz
B
, et al
.
Heparan sulfate proteoglycan expression is induced during early erythroid differentiation of multipotent hematopoietic stem cells
.
Blood
.
1999
;
93
(
9
):
2884
-
2897
.
119.
Gao
X
,
Lee
HY
,
da Rocha
EL
, et al
.
TGF-β inhibitors stimulate red blood cell production by enhancing self-renewal of BFU-E erythroid progenitors
.
Blood
.
2016
;
128
(
23
):
2637
-
2641
.
120.
Kobayashi
K
,
Kato
K
,
Sugi
T
, et al
.
Plasmodium falciparum BAEBL binds to heparan sulfate proteoglycans on the human erythrocyte surface
.
J Biol Chem
.
2010
;
285
(
3
):
1716
-
1725
.
121.
Vogt
AM
,
Winter
G
,
Wahlgren
M
,
Spillmann
D
.
Heparan sulphate identified on human erythrocytes: a Plasmodium falciparum receptor
.
Biochem J
.
2004
;
381
(
Pt 3
):
593
-
597
.
122.
Schick
BP
,
Jacoby
JA
.
Serglycin and betaglycan proteoglycans are expressed in the megakaryocytic cell line CHRF 288-11 and normal human megakaryocytes
.
J Cell Physiol
.
1995
;
165
(
1
):
96
-
106
.
123.
Lord
MS
,
Cheng
B
,
Farrugia
BL
,
McCarthy
S
,
Whitelock
JM
.
Platelet factor 4 binds to vascular proteoglycans and controls both growth factor activities and platelet activation
.
J Biol Chem
.
2017
;
292
(
10
):
4054
-
4063
.
124.
Kaneider
NC
,
Feistritzer
C
,
Gritti
D
, et al
.
Expression and function of syndecan-4 in human platelets
.
Thromb Haemost
.
2005
(
6
):
1120
-
1127
.
125.
Wan
LM
,
Zhang
SK
,
Li
SB
, et al
.
Heparanase facilitates PMA-induced megakaryocytic differentiation in K562 cells via interleukin 6/STAT3 pathway
.
Thromb Haemost
.
2020
;
120
(
4
):
647
-
657
.
126.
Tan
Y-X
,
Cui
H
,
Wan
LM
, et al
.
Overexpression of heparanase in mice promoted megakaryopoiesis
.
Glycobiology
.
2018
;
28
(
5
):
269
-
275
.
127.
Vögtle
T
,
Sharma
S
,
Mori
J
, et al
.
Heparan sulfates are critical regulators of the inhibitory megakaryocyte-platelet receptor G6b-B
.
Elife
.
2019
;
8
:
e46840
.
128.
Falet
H
,
Rivadeneyra
L
,
Hoffmeister
KM
.
Clinical impact of glycans in platelet and megakaryocyte biology
.
Blood
.
2022
;
139
(
22
):
3255
-
3263
.
129.
Eustes
AS
,
Campbell
RA
,
Middleton
EA
, et al
.
Heparanase expression and activity are increased in platelets during clinical sepsis
.
J Thromb Haemost
.
2021
;
19
(
5
):
1319
-
1330
.
130.
Sanderson
RD
,
Lalor
P
,
Bernfield
M
.
B lymphocytes express and lose syndecan at specific stages of differentiation
.
Cell Regul
.
1989
;
1
:
27
-
35
.
131.
van der Voort
R
,
Keehnen
RM
,
Beuling
EA
,
Spaargaren
M
,
Pals
ST
.
Regulation of cytokine signaling by B cell antigen receptor and CD40-controlled expression of heparan sulfate proteoglycans
.
J Exp Med
.
2000
;
192
(
8
):
1115
-
1124
.
132.
Borghesi
LA
,
Yamashita
Y
,
Kincade
PW
.
Heparan sulfate proteoglycans mediate interleukin-7–dependent B lymphopoiesis
.
Blood
.
1999
;
93
(
1
):
140
-
148
.
133.
Reijmers
RM
,
Spaargaren
M
,
Pals
ST
.
Heparan sulfate proteoglycans in the control of B cell development and the pathogenesis of multiple myeloma
.
FEBS J
.
2013
;
280
(
10
):
2180
-
2193
.
134.
Bret
C
,
Hose
D
,
Reme
T
, et al
.
Expression of genes encoding for proteins involved in heparan sulphate and chondroitin sulphate chain synthesis and modification in normal and malignant plasma cells
.
Br J Haematol
.
2009
;
145
(
3
):
350
-
368
.
135.
Jarousse
N
,
Trujillo
DL
,
Wilcox-Adelman
S
,
Coscoy
L
.
Virally-induced upregulation of heparan sulfate on B cells via the action of type I IFN
.
J Immunol
.
2011
;
187
(
11
):
5540
-
5547
.
136.
Aleman-Muench
GR
,
Mendoza
V
,
Stenvers
K
, et al
.
Betaglycan (TβRIII) is expressed in the thymus and regulates T cell development by protecting thymocytes from apoptosis
.
PLoS One
.
2012
;
7
:
e44217
.
137.
Ortega-Francisco
S
,
de la Fuente-Granada
M
,
Alvarez Salazar
EK
, et al
.
TβRIII is induced by TCR signaling and downregulated in FoxP3+ regulatory T cells
.
Biochem Biophys Res Commun
.
2017
;
494
(
1-2
):
82
-
87
.
138.
Khan
AA
,
Bose
C
,
Yam
LS
,
Soloski
MJ
,
Rupp
F
.
Physiological regulation of the immunological synapse by agrin
.
Science
.
2001
;
292
(
5522
):
1681
-
1686
.
139.
Zhang
J
,
Wang
Y
,
Chu
Y
, et al
.
Agrin is involved in lymphocytes activation that is mediated by α-dystroglycan
.
FASEB J
.
2006
;
20
(
1
):
50
-
58
.
140.
Forster-Horváth
C
,
Bocsi
J
,
Rásó
E
, et al
.
Constitutive intracellular expression and activation-induced cell surface up-regulation of CD44v3 in human T lymphocytes
.
Eur J Immunol
.
2001
;
31
(
2
):
600
-
608
.
141.
Chung
J-S
,
Dougherty
I
,
Cruz
PD
,
Ariizumi
K
.
Syndecan-4 mediates the coinhibitory function of DC-HIL on T cell activation
.
J Immunol
.
2007
;
179
(
9
):
5778
-
5784
.
142.
Sutton
VR
,
Brennan
AJ
,
Ellis
S
, et al
.
Serglycin determines secretory granule repertoire and regulates natural killer cell and cytotoxic T lymphocyte cytotoxicity
.
FEBS J
.
2016
;
283
(
5
):
947
-
961
.
143.
Hsu
H-P
,
Chen
YT
,
Chen
YY
, et al
.
Heparan sulfate is essential for thymus growth
.
J Biol Chem
.
2021
;
296
:
100419
.
144.
Wrenshall
LE
,
Platt
JL
.
Regulation of T cell homeostasis by heparan sulfate-bound IL-2
.
J Immunol
.
1999
;
163
(
7
):
3793
-
3800
.
145.
Wrenshall
LE
,
Platt
JL
,
Stevens
ET
,
Wight
TN
,
Miller
JD
.
Propagation and control of T cell responses by heparan sulfate-bound IL-2
.
J Immunol
.
2003
;
170
(
11
):
5470
-
5474
.
146.
Martinez
HA
,
Koliesnik
I
,
Kaber
G
, et al
.
FOXP3+ regulatory T cells use heparanase to access IL-2 bound to ECM in inflamed tissues
.
bioRxiv
.
Preprint published online 27 February 2023
.
147.
Naparstek
Y
,
Cohen
IR
,
Fuks
Z
,
Vlodavsky
I
.
Activated T lymphocytes produce a matrix-degrading heparan sulphate endoglycosidase
.
Nature
.
1984
;
310
(
5974
):
241
-
244
.
148.
de Parseval
A
,
Bobardt
MD
,
Chatterji
A
, et al
.
A highly conserved arginine in gp120 governs HIV-1 binding to both syndecans and CCR5 via sulfated motifs
.
J Biol Chem
.
2005
;
280
(
47
):
39493
-
39504
.
149.
Clausen
TM
,
Sandoval
DR
,
Spliid
CB
, et al
.
SARS-CoV-2 infection depends on cellular heparan sulfate and ACE2
.
Cell
.
2020
;
183
(
4
):
1043
-
1057.e15
.
150.
Jones
KS
,
Petrow-Sadowski
C
,
Bertolette
DC
,
Huang
Y
,
Ruscetti
FW
.
Heparan sulfate proteoglycans mediate attachment and entry of human T-cell leukemia virus type 1 virions into CD4+ T cells
.
J Virol
.
2005
;
79
(
20
):
12692
-
12702
.
151.
Rusnati
M
,
Coltrini
D
,
Oreste
P
, et al
.
Interaction of HIV-1 Tat protein with heparin: role of the backbone structure, sulfation, and size
.
J Biol Chem
.
1997
;
272
(
17
):
11313
-
11320
.
152.
Shen
X-R
,
Geng
R
,
Li
Q
, et al
.
ACE2-independent infection of T lymphocytes by SARS-CoV-2
.
Signal Transduct Target Ther
.
2022
;
7
:
83
.
153.
Hecht
M-L
,
Rosental
B
,
Horlacher
T
, et al
.
Natural cytotoxicity receptors NKp30, NKp44 and NKp46 bind to different heparan sulfate/heparin sequences
.
J Proteome Res
.
2009
;
8
(
2
):
712
-
720
.
154.
Zilka
A
,
Landau
G
,
Hershkovitz
O
, et al
.
Characterization of the heparin/heparan sulfate binding site of the natural cytotoxicity receptor NKp46
.
Biochemistry
.
2005
;
44
:
14477
-
14485
.
155.
Brusilovsky
M
,
Radinsky
O
,
Cohen
L
, et al
.
Regulation of natural cytotoxicity receptors by heparan sulfate proteoglycans in-cis: a lesson from NKp44
.
Eur J Immunol
.
2015
;
45
(
4
):
1180
-
1191
.
156.
Elkin
M
.
Role of heparanase in macrophage activation
.
Adv Exp Med Biol
.
2020
;
1221
:
445
-
460
.
157.
Ishai-Michaeli
R
,
Eldor
A
,
Vlodavsky
I
.
Heparanase activity expressed by platelets, neutrophils, and lymphoma cells releases active fibroblast growth factor from extracellular matrix
.
Cell Regul
.
1990
;
1
(
11
):
833
-
842
.
158.
Matzner
Y
,
Vlodavsky
I
,
Bar-Ner
M
,
Ishai-Michaeli
R
,
Tauber
A I
.
Subcellular localization of heparanase in human neutrophils
.
J Leukoc Biol
.
1992
;
51
(
6
):
519
-
524
.
159.
Vlodavsky
I
,
Eldor
A
,
Haimovitz-Friedman
A
, et al
.
Expression of heparanase by platelets and circulating cells of the immune system: possible involvement in diapedesis and extravasation
.
Invasion Metastasis
.
1992
;
12
(
2
):
112
-
127
.
160.
Gutter-Kapon
L
,
Alishekevitz
D
,
Shaked
Y
, et al
.
Heparanase is required for activation and function of macrophages
.
Proc Natl Acad Sci U S A
.
2016
;
113
(
48
):
E7808
-
E7817
.
161.
Putz
EM
,
Mayfosh
AJ
,
Kos
K
, et al
.
NK cell heparanase controls tumor invasion and immune surveillance
.
J Clin Invest
.
2017
;
127
(
7
):
2777
-
2788
.
162.
Saez
B
,
Ferraro
F
,
Yusuf
RZ
, et al
.
Inhibiting stromal cell heparan sulfate synthesis improves stem cell mobilization and enables engraftment without cytotoxic conditioning
.
Blood
.
2014
;
124
(
19
):
2937
-
2947
.
163.
Dennissen
MA
,
Jenniskens
GJ
,
Pieffers
M
, et al
.
Large, tissue-regulated domain diversity of heparan sulfates demonstrated by phage display antibodies
.
J Biol Chem
.
2002
;
277
(
13
):
10982
-
10986
.
164.
van Kuppevelt
TH
,
Dennissen
MA
,
van Venrooij
WJ
,
Hoet
RM
,
Veerkamp
JH
.
Generation and application of type-specific anti-heparan sulfate antibodies using phage display technology: further evidence for heparan sulfate heterogeneity in the kidney
.
J Biol Chem
.
1998
;
273
(
21
):
12960
-
12966
.
165.
Attreed
M
,
Desbois
M
,
Van Kuppevelt
TH
,
Bülow
HE
.
Direct visualization of specifically modified extracellular glycans in living animals
.
Nat Methods
.
2012
;
9
(
5
):
477
-
479
.
Sign in via your Institution