Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 54
Filter
1.
Am J Physiol Cell Physiol ; 322(5): C896-C912, 2022 05 01.
Article in English | MEDLINE | ID: mdl-35319900

ABSTRACT

Receptor-ligand interactions play an important role in many biological processes by triggering specific cellular responses. These interactions are frequently regulated by coreceptors that facilitate, alter, or inhibit signaling. Coreceptors work in parallel with other specific and accessory molecules to coordinate receptor-ligand interactions. Cell surface heparan sulfate proteoglycans (HSPGs) function as unique coreceptors because they can bind to many ligands and receptors through their HS and core protein motifs. Cell surface HSPGs are typically expressed in abundance of the signaling receptors and, thus, are capable of mediating the initial binding of ligands to the cell surface. HSPG coreceptors do not possess kinase domains or intrinsic enzyme activities and, for the most part, binding to cell surface HSPGs does not directly stimulate intracellular signaling. Because of these features, cell surface HSPGs primarily function as coreceptors for many receptor-ligand interactions. Given that cell surface HSPGs are widely conserved, they likely serve fundamental functions to preserve basic physiological processes. Indeed, cell surface HSPGs can support specific cellular interactions with growth factors, morphogens, chemokines, extracellular matrix (ECM) components, and microbial pathogens and their secreted virulence factors. Through these interactions, HSPG coreceptors regulate cell adhesion, proliferation, migration, and differentiation, and impact the onset, progression, and outcome of pathophysiological processes, such as development, tissue repair, inflammation, infection, and tumorigenesis. This review seeks to provide an overview of the various mechanisms of how cell surface HSPGs function as coreceptors.


Subject(s)
Heparan Sulfate Proteoglycans , Signal Transduction , Cell Membrane/metabolism , Heparan Sulfate Proteoglycans/chemistry , Heparan Sulfate Proteoglycans/metabolism , Heparitin Sulfate/metabolism , Intercellular Signaling Peptides and Proteins , Ligands , Signal Transduction/physiology
2.
Methods Mol Biol ; 2303: 605-625, 2022.
Article in English | MEDLINE | ID: mdl-34626410

ABSTRACT

Heparan sulfate proteoglycans (HSPGs) are at the forefront of host-microbe interactions. Cell surface HSPGs are thought to promote infection as attachment and internalization receptors for many bacterial pathogens and as soluble inhibitors of host immunity when released from the cell surface by ectodomain shedding. However, the importance of HSPG-pathogen interactions in vivo has yet to be clearly established. Here we describe several representative methods to study the role of HSPGs in systemic bacterial infections, such as bacteremia and sepsis. The overall experimental strategy is to use mouse models to establish the physiological significance of HSPGs, to determine the identity of HSPGs that specifically promote infection, and to define key structural features of HSPGs that enhance bacterial virulence in systemic infections.


Subject(s)
Bacterial Infections , Animals , Cell Membrane , Disease Models, Animal , Heparan Sulfate Proteoglycans , Heparitin Sulfate , Mice , Sepsis
3.
JCI Insight ; 6(18)2021 09 22.
Article in English | MEDLINE | ID: mdl-34549726

ABSTRACT

Invariant NKT (iNKT) cells are potent immunomodulatory cells that acquire effector function during their development in the thymus. IL-17-producing iNKT cells are commonly referred to as NKT17 cells, and they are unique among iNKT cells to express the heparan sulfate proteoglycan CD138 and the transcription factor RORγt. Whether and how CD138 and RORγt contribute to NKT17 cell differentiation, and whether there is an interplay between RORγt and CD138 expression to control iNKT lineage fate, remain mostly unknown. Here, we showed that CD138 expression was only associated with and not required for the differentiation and IL-17 production of NKT17 cells. Consequently, CD138-deficient mice still generated robust numbers of IL-17-producing RORγt+ NKT17 cells. Moreover, forced expression of RORγt significantly promoted the generation of thymic NKT17 cells, but did not induce CD138 expression on non-NKT17 cells. These results indicated that NKT17 cell generation and IL-17 production were driven by RORγt, employing mechanisms that were independent of CD138. Therefore, our study effectively dissociated CD138 expression from the RORγt-driven molecular pathway of NKT17 cell differentiation.


Subject(s)
Cell Differentiation , Interleukin-17/metabolism , Natural Killer T-Cells/metabolism , Nuclear Receptor Subfamily 1, Group F, Member 3/metabolism , Syndecan-1/genetics , Syndecan-1/metabolism , Animals , CD4 Antigens/metabolism , CD8 Antigens/metabolism , CD8-Positive T-Lymphocytes/physiology , Cell Differentiation/genetics , Female , Granzymes/metabolism , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Natural Killer T-Cells/physiology , Nuclear Receptor Subfamily 1, Group F, Member 3/genetics , Phenotype , Pore Forming Cytotoxic Proteins/metabolism , Thymocytes/metabolism
4.
mBio ; 11(6)2020 12 08.
Article in English | MEDLINE | ID: mdl-33293379

ABSTRACT

Subversion of heparan sulfate proteoglycans (HSPGs) is thought to be a common virulence mechanism shared by many microbial pathogens. The prevailing assumption is that pathogens co-opt HSPGs as cell surface attachment receptors or as inhibitors of innate host defense. However, there are few data that clearly support this idea in vivo We found that deletion of syndecan-1 (Sdc1), a major cell surface HSPG of epithelial cells, causes a gain of function in a mouse model of scarified corneal infection, where Sdc1-/- corneas were significantly less susceptible to Streptococcus pneumoniae infection. Administration of excess Sdc1 ectodomains significantly inhibited S. pneumoniae corneal infection, suggesting that Sdc1 promotes infection as a cell surface attachment receptor. However, S. pneumoniae did not interact with Sdc1 and Sdc1 was shed upon S. pneumoniae infection, indicating that Sdc1 does not directly support S. pneumoniae adhesion. Instead, Sdc1 promoted S. pneumoniae adhesion by driving the assembly of fibronectin (FN) fibrils in the corneal basement membrane to which S. pneumoniae attaches when infecting injured corneas. S. pneumoniae specifically bound to corneal FN via PavA, and PavA deletion significantly attenuated S. pneumoniae virulence in the cornea. Excess Sdc1 ectodomains inhibited S. pneumoniae corneal infection by binding to the Hep II domain and interfering with S. pneumoniae PavA binding to FN. These findings reveal a previously unknown virulence mechanism of S. pneumoniae where key extracellular matrix (ECM) interactions and structures that are essential for host cell homeostasis are exploited for bacterial pathogenesis.IMPORTANCE Bacterial pathogens have evolved several ingenious mechanisms to subvert host cell biology for their pathogenesis. Bacterial attachment to the host ECM establishes a niche to grow and is considered one of the critical steps of infection. This pathogenic mechanism entails coordinated assembly of the ECM by the host to form the ECM structure and organization that are specifically recognized by bacteria for their adhesion. We serendipitously discovered that epithelial Sdc1 facilitates the assembly of FN fibrils in the corneal basement membrane and that this normal biological function of Sdc1 has detrimental consequences for the host in S. pneumoniae corneal infection. Our studies suggest that bacterial subversion of the host ECM is more complex than previously appreciated.


Subject(s)
Fibronectins/metabolism , Host-Pathogen Interactions , Keratitis/metabolism , Keratitis/microbiology , Streptococcus pneumoniae/physiology , Syndecan-1/metabolism , Animals , Bacterial Adhesion , Cornea/metabolism , Cornea/microbiology , Cornea/pathology , Disease Models, Animal , Extracellular Matrix/metabolism , Fluorescent Antibody Technique , Gain of Function Mutation , Gene Expression , Heparan Sulfate Proteoglycans/genetics , Heparan Sulfate Proteoglycans/metabolism , Host-Pathogen Interactions/genetics , Immunohistochemistry , Keratitis/pathology , Mice , Mice, Knockout , Syndecan-1/genetics
5.
Nat Commun ; 11(1): 5998, 2020 11 26.
Article in English | MEDLINE | ID: mdl-33243988

ABSTRACT

Intratumoral heterogeneity is a common feature of many myeloid leukemias and a significant reason for treatment failure and relapse. Thus, identifying the cells responsible for residual disease and leukemia re-growth is critical to better understanding how they are regulated. Here, we show that a knock-in reporter mouse for the stem cell gene Musashi 2 (Msi2) allows identification of leukemia stem cells in aggressive myeloid malignancies, and provides a strategy for defining their core dependencies. Specifically, we carry out a high throughput screen using Msi2-reporter blast crisis chronic myeloid leukemia (bcCML) and identify several adhesion molecules that are preferentially expressed in therapy resistant bcCML cells and play a key role in bcCML. In particular, we focus on syndecan-1, whose deletion triggers defects in bcCML growth and propagation and markedly improves survival of transplanted mice. Further, live imaging reveals that the spatiotemporal dynamics of leukemia cells are critically dependent on syndecan signaling, as loss of this signal impairs their localization, migration and dissemination to distant sites. Finally, at a molecular level, syndecan loss directly impairs integrin ß7 function, suggesting that syndecan exerts its influence, at least in part, by coordinating integrin activity in bcCML. These data present a platform for delineating the biological underpinnings of leukemia stem cell function, and highlight the Sdc1-Itgß7 signaling axis as a key regulatory control point for bcCML growth and dissemination.


Subject(s)
Blast Crisis/therapy , Leukemia, Myeloid, Acute/therapy , Neoplastic Stem Cells/pathology , RNA-Binding Proteins/genetics , Syndecan-1/antagonists & inhibitors , Animals , Antineoplastic Agents/therapeutic use , Blast Crisis/genetics , Blast Crisis/pathology , Chemoradiotherapy/methods , Disease Models, Animal , Drug Resistance, Neoplasm/drug effects , Gene Knock-In Techniques , Gene Knockout Techniques , Genes, Reporter/genetics , Green Fluorescent Proteins/chemistry , Green Fluorescent Proteins/genetics , High-Throughput Screening Assays , Humans , Imatinib Mesylate/pharmacology , Imatinib Mesylate/therapeutic use , Integrin beta Chains/metabolism , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/pathology , Mice, Transgenic , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/radiation effects , RNA-Seq , Signal Transduction/drug effects , Syndecan-1/genetics , Syndecan-1/metabolism
6.
PLoS Pathog ; 16(5): e1008497, 2020 05.
Article in English | MEDLINE | ID: mdl-32453780

ABSTRACT

Heparan sulfate proteoglycans (HSPGs) are at the forefront of host-microbe interactions. Molecular and cell-based studies suggest that HSPG-pathogen interactions promote pathogenesis by facilitating microbial attachment and invasion of host cells. However, the specific identity of HSPGs, precise mechanisms by which HSPGs promote pathogenesis, and the in vivo relevance of HSPG-pathogen interactions remain to be determined. HSPGs also modulate host responses to tissue injury and inflammation, but functions of HSPGs other than facilitating microbial attachment and internalization are understudied in infectious disease. Here we examined the role of syndecan-1 (Sdc1), a major cell surface HSPG of epithelial cells, in mouse models of Listeria monocytogenes (Lm) infection. We show that Sdc1-/- mice are significantly less susceptible to both intragastric and intravenous Lm infection compared to wild type (Wt) mice. This phenotype is not seen in Sdc3-/- or Sdc4-/- mice, indicating that ablation of Sdc1 causes a specific gain of function that enables mice to resist listeriosis. However, Sdc1 does not support Lm attachment or invasion of host cells, indicating that Sdc1 does not promote pathogenesis as a cell surface Lm receptor. Instead, Sdc1 inhibits the clearance of Lm before the bacterium gains access to its intracellular niche. Large intravascular aggregates of neutrophils and neutrophil extracellular traps (NETs) embedded with antimicrobial compounds are formed in Sdc1-/- livers, which trap and kill Lm. Lm infection induces Sdc1 shedding from the surface of hepatocytes in Wt livers, which is directly associated with the decrease in size of intravascular aggregated NETs. Furthermore, administration of purified Sdc1 ectodomains or DNase inhibits the formation of intravascular aggregated neutrophils and NETs and significantly increases the liver bacterial burden in Sdc1-/- mice. These data indicate that Lm induces Sdc1 shedding to subvert the activity of Sdc1 ectodomains to inhibit its clearance by intravascular aggregated NETs.


Subject(s)
Extracellular Traps/immunology , Listeria monocytogenes/immunology , Listeriosis/immunology , Neutrophils/immunology , Syndecan-1/immunology , Animals , Extracellular Traps/genetics , Hepatocytes/immunology , Hepatocytes/pathology , Listeria monocytogenes/pathogenicity , Listeriosis/genetics , Listeriosis/pathology , Mice , Mice, Inbred BALB C , Mice, Knockout , Neutrophils/pathology , Protein Domains , Syndecan-1/genetics
7.
Sci Transl Med ; 12(535)2020 03 18.
Article in English | MEDLINE | ID: mdl-32188725

ABSTRACT

Acetaminophen/paracetamol (APAP) overdose is the leading cause of drug-induced acute liver failure (ALF) in the United States and Europe. The progression of the disease is attributed to sterile inflammation induced by the release of high mobility group box 1 (HMGB1) and the interaction with receptor for advanced glycation end products (RAGE). A specific, effective, and safe approach to neutralize the proinflammatory activity of HMGB1 is highly desirable. Here, we found that a heparan sulfate (HS) octadecasaccharide (18-mer-HP or hepatoprotective 18-mer) displays potent hepatoprotection by targeting the HMGB1/RAGE axis. Endogenous HS proteoglycan, syndecan-1, is shed in response to APAP overdose in mice and humans. Furthermore, purified syndecan-1, but not syndecan-1 core protein, binds to HMGB1, suggesting that HMGB1 binds to HS polysaccharide side chains of syndecan-1. Last, we compared the protection effect between 18-mer-HP and N-acetyl cysteine, which is the standard of care to treat APAP overdose. We demonstrated that 18-mer-HP administered 3 hours after a lethal dose of APAP is fully protective; however, the treatment of N-acetyl cysteine loses protection. Therefore, 18-mer-HP may offer a potential therapeutic advantage over N-acetyl cysteine for late-presenting patients. Synthetic HS provides a potential approach for the treatment of APAP-induced ALF.


Subject(s)
Chemical and Drug Induced Liver Injury , Liver Failure, Acute , Acetaminophen/toxicity , Animals , Anti-Inflammatory Agents , Chemical and Drug Induced Liver Injury/drug therapy , Chemical and Drug Induced Liver Injury/prevention & control , Europe , Heparitin Sulfate , Humans , Liver , Liver Failure, Acute/chemically induced , Liver Failure, Acute/drug therapy , Liver Failure, Acute/prevention & control , Mice , Mice, Inbred C57BL
8.
Vet Res ; 49(1): 109, 2018 Oct 29.
Article in English | MEDLINE | ID: mdl-30373658

ABSTRACT

Streptococcus suis is a swine pathogen and zoonotic agent responsible for meningitis and septic shock. Although several putative virulence factors have been described, the initial steps of the S. suis pathogenesis remain poorly understood. While controversial results have been reported for a S. suis serotype 2 zinc metalloprotease (Zmp) regarding its IgA protease activity, recent phylogenetic analyses suggested that this protein is homologous to the ZmpC of Streptococcus pneumoniae, which is not an IgA protease. Based on the previously described functions of metalloproteases (including IgA protease and ZmpC), different experiments were carried out to study the activities of that of S. suis serotype 2. First, results showed that S. suis, as well as the recombinant Zmp, were unable to cleave human IgA1, confirming lack of IgA protease activity. Similarly, S. suis was unable to cleave P-selectin glycoprotein ligand-1 and to activate matrix metalloprotease 9, at least under the conditions tested. However, S. suis was able to partially cleave mucin 16 and syndecan-1 ectodomains. Experiments carried out with an isogenic Δzmp mutant showed that the Zmp protein was partially involved in such activities. The absence of a functional Zmp protein did not affect the ability of S. suis to adhere to porcine bronchial epithelial cells in vitro, or to colonize the upper respiratory tract of pigs in vivo. Taken together, our results show that S. suis serotype 2 Zmp is not a critical virulence factor and highlight the importance of independently confirming results on S. suis virulence by different teams.


Subject(s)
Metalloendopeptidases/metabolism , Streptococcus suis/enzymology , Animals , DNA, Bacterial/genetics , Gene Expression Regulation, Bacterial , Gene Expression Regulation, Enzymologic , Metalloendopeptidases/genetics , Mice , Protein Domains , Serine Endopeptidases/metabolism , Serogroup , Streptococcal Infections/microbiology , Streptococcus suis/genetics , Streptococcus suis/pathogenicity , Virulence
9.
J Immunol ; 201(6): 1651-1661, 2018 09 15.
Article in English | MEDLINE | ID: mdl-30045969

ABSTRACT

IL-17 is a potent proinflammatory cytokine that drives pathogenesis of multiple autoimmune diseases, including psoriasis. A major source of pathogenic IL-17 is a subset of γδ T cells (Tγδ17) that acquires the ability to produce IL-17 while developing in the thymus. The mechanisms that regulate homeostasis of Tγδ17 cells and their roles in psoriasis, however, are not fully understood. In this paper, we show that the heparan sulfate proteoglycan syndecan-1 (sdc1) plays a critical role in regulating homeostasis of Tγδ17 cells and modulating psoriasis-like skin inflammation in mice. sdc1 was predominantly expressed by Tγδ17 cells (but not IL-17- Tγδ cells) in the thymus, lymph nodes, and dermis. sdc1 deficiency significantly and selectively increased the frequency and absolute numbers of Tγδ17 cells by mechanisms that included increased proliferation and decreased apoptosis. Adoptive transfer experiments ruled out a significant role of sdc1 expressed on nonhematopoietic cells in halting expansion and proliferation of sdc1-deficient Tγδ17 cells. When subjected to imiquimod-induced psoriasiform dermatitis, Tγδ17 cells in sdc1KO mice displayed heightened responses accompanied by significantly increased skin inflammation than their wild-type counterparts. Furthermore, transferred sdc1-deficient γδ T cells caused more severe psoriasiform dermatitis than their sdc1-sufficient counterparts in TCR-ßδ KO hosts. The results uncover a novel role for sdc1 in controlling homeostasis of Tγδ17 cells and moderating host responses to psoriasis-like inflammation.


Subject(s)
Dermatitis/immunology , Interleukin-17/immunology , Psoriasis/immunology , Receptors, Antigen, T-Cell, gamma-delta/immunology , Syndecan-1/immunology , T-Lymphocytes/immunology , Animals , Dermatitis/genetics , Dermatitis/pathology , Disease Models, Animal , Inflammation/genetics , Inflammation/immunology , Inflammation/pathology , Interleukin-17/genetics , Mice , Mice, Knockout , Psoriasis/genetics , Psoriasis/pathology , Receptors, Antigen, T-Cell, gamma-delta/genetics , Syndecan-1/genetics , T-Lymphocytes/pathology
11.
Biochem Soc Trans ; 46(2): 371-377, 2018 04 17.
Article in English | MEDLINE | ID: mdl-29523771

ABSTRACT

Syndecan-1 (Sdc1) is a major cell surface heparan sulfate (HS) proteoglycan of epithelial cells, a cell type targeted by many bacterial pathogens early in their pathogenesis. Loss of Sdc1 in mice is a gain-of-function mutation that significantly decreases the susceptibility to several bacterial infections, suggesting that subversion of Sdc1 is an important virulence strategy. HS glycosaminoglycan (GAG) chains of cell surface Sdc1 promote bacterial pathogenesis by facilitating the attachment of bacteria to host cells. Engagement of cell surface Sdc1 HS chains by bacterial adhesins transmits signal through the highly conserved Sdc1 cytoplasmic domain, which can lead to uptake of intracellular bacterial pathogens. On the other hand, several bacteria that do not require Sdc1 for their attachment and invasion stimulate Sdc1 shedding and exploit the capacity of Sdc1 ectodomain HS GAGs to disarm innate defense mechanisms to evade immune clearance. Recent data suggest that select HS sulfate motifs, and not the overall charge of HS, are important in the inhibition of innate immune mechanisms. Here, we discuss several examples of Sdc1 subversion in bacterial infections.


Subject(s)
Bacterial Infections/metabolism , Glycomics , Syndecan-1/metabolism , Adhesins, Bacterial/metabolism , Animals , Glycosaminoglycans/metabolism , Heparitin Sulfate/metabolism , Mice , Mutation , Syndecan-1/genetics , Virulence
12.
Methods Cell Biol ; 143: 317-333, 2018.
Article in English | MEDLINE | ID: mdl-29310785

ABSTRACT

Syndecans comprise a major family of cell surface heparan sulfate proteoglycans (HSPGs). Syndecans are composed of sulfated glycosaminoglycans (GAGs), heparan sulfate (HS) or both HS and chondroitin sulfate (CS), attached covalently to core proteins. Syndecans regulate many cellular processes, such as adhesion, proliferation, and migration. Syndecans bind and regulate molecules primarily through their HS chains, but do not bind to all HS/heparin-binding molecules. Furthermore, mice ablated for the syndecan-1 or -4 gene do not show major developmental abnormalities, but they do show striking pathological phenotypes when challenged with infectious or inflammatory stimuli and conditions, suggesting that certain functions of syndecans are specific and cannot be compensated for by other syndecans or other HSPGs. These observations underscore the physiological importance of syndecans and indicate a need to study the activities of isolated native syndecans to define their molecular and cellular functions, and to establish their biological significance. Here we describe methods to isolate syndecans and several assays to analyze their functions.


Subject(s)
Cell Membrane/chemistry , Chromatography, Affinity/methods , Syndecans/isolation & purification , Animals , Cells, Cultured , Chondroitin Sulfates/chemistry , Chromatography, Affinity/instrumentation , Culture Media, Conditioned/chemistry , Epithelial Cells/chemistry , Epithelial Cells/cytology , Heparitin Sulfate/chemistry , Mice , Neutrophils/chemistry , Syndecans/analysis , Syndecans/chemistry
13.
Hepatology ; 66(5): 1601-1615, 2017 11.
Article in English | MEDLINE | ID: mdl-28543100

ABSTRACT

Accidental or intentional misuse of acetaminophen (APAP) is the leading cause of acute liver failure in the Western world. Although mechanisms that trigger APAP-induced liver injury (AILI) are well known, those that halt the progression of APAP liver disease and facilitate liver recovery are less understood. Heparan sulfate proteoglycans (HSPGs) bind to and regulate various tissue injury factors through their heparan sulfate (HS) chains, but the importance of HSPGs in liver injury in vivo remains unknown. Here, we examined the role of syndecan-1, the major cell-surface HSPG of hepatocytes, in AILI. Ablation of syndecan-1 in mice led to unopposed progression of liver injury upon APAP overdose. However, direct APAP hepatoxicity and liver injury at early times post-APAP overdose were unaffected by syndecan-1, suggesting that syndecan-1 influences later mechanisms that lead to liver repair. The exuberant liver injury phenotypes in syndecan-1 null (Sdc1-/- ) mice were traced to a deficiency in protein kinase B (Akt) activation in hepatocytes, which led to a delayed increase in glycogen synthase kinase-3ß (GSK-3ß)-mediated hepatocyte apoptosis. Inhibition of Akt worsened, whereas inhibition of GSK-3ß and caspases protected mice from AILI. Moreover, administration of purified syndecan-1, HS, or engineered heparan compounds containing 2-O-sulfate groups rescued Sdc1-/- mice from AILI by potentiating Akt signaling and inhibiting GSK-3ß-mediated apoptosis in hepatocytes. In addition, HS showed a significantly prolonged therapeutic efficacy as compared to N-acetylcysteine. CONCLUSION: These results demonstrate that 2-O-sulfated domains in syndecan-1 HS halt disease progression and promote liver repair by enhancing hepatocyte survival in AILI. We propose that syndecan-1 is a critical endogenous factor that controls the balance between prosurvival signaling and apoptosis in hepatocytes in APAP liver disease. (Hepatology 2017;66:1601-1615).


Subject(s)
Acetaminophen/adverse effects , Analgesics, Non-Narcotic/adverse effects , Chemical and Drug Induced Liver Injury/metabolism , Syndecan-1/metabolism , Animals , Apoptosis , Chemical and Drug Induced Liver Injury/etiology , Female , Glycogen Synthase Kinase 3 beta/metabolism , Hepatocytes/drug effects , Male , Mice, Inbred C57BL , Proto-Oncogene Proteins c-akt/metabolism
14.
Blood ; 129(20): 2749-2759, 2017 05 18.
Article in English | MEDLINE | ID: mdl-28381397

ABSTRACT

Antibody secreting cells (ASCs) are critical effector cells and long-lived sentinels for immune memory. ASCs are highly dependent on exogenous soluble factors such as interleukin-6 (IL-6) and APRIL, to prevent their cell death. We have found that the canonical surface marker of ASCs, CD138 (syndecan-1), which is upregulated during ASC maturation, is required in a cell-intrinsic manner to mount an effective long-term humoral immune response following immunization. Surface expression of CD138 increased heparan sulfate levels on ASCs, which are known to bind pro-survival cytokines, leading to increased survival in a cell-intrinsic manner in vivo. In IL-6 and APRIL-deficient hosts, ASCs underwent extensive apoptosis independently of CD138 expression. We propose a model in which CD138 expression on fully mature ASCs provides a selective survival advantage over less mature, newly minted ASCs, by enhancing pro-survival cytokine signaling.


Subject(s)
Cell Differentiation , Plasma Cells/cytology , Plasma Cells/metabolism , Syndecan-1/metabolism , Animals , Antibody Formation/immunology , Apoptosis , Cell Survival , Epitopes/immunology , Germinal Center/immunology , Humans , Immunity, Humoral , Interleukin-6/metabolism , Mice, Inbred C57BL , Signal Transduction/immunology , Tumor Necrosis Factor Ligand Superfamily Member 13/metabolism
15.
J Exp Med ; 214(3): 623-637, 2017 03 06.
Article in English | MEDLINE | ID: mdl-28148688

ABSTRACT

We studied three patients with severe skeletal dysplasia, T cell immunodeficiency, and developmental delay. Whole-exome sequencing revealed homozygous missense mutations affecting exostosin-like 3 (EXTL3), a glycosyltransferase involved in heparan sulfate (HS) biosynthesis. Patient-derived fibroblasts showed abnormal HS composition and altered fibroblast growth factor 2 signaling, which was rescued by overexpression of wild-type EXTL3 cDNA. Interleukin-2-mediated STAT5 phosphorylation in patients' lymphocytes was markedly reduced. Interbreeding of the extl3-mutant zebrafish (box) with Tg(rag2:green fluorescent protein) transgenic zebrafish revealed defective thymopoiesis, which was rescued by injection of wild-type human EXTL3 RNA. Targeted differentiation of patient-derived induced pluripotent stem cells showed a reduced expansion of lymphohematopoietic progenitor cells and defects of thymic epithelial progenitor cell differentiation. These data identify EXTL3 mutations as a novel cause of severe immune deficiency with skeletal dysplasia and developmental delay and underline a crucial role of HS in thymopoiesis and skeletal and brain development.


Subject(s)
Bone Diseases, Developmental/etiology , Developmental Disabilities/etiology , Immunologic Deficiency Syndromes/etiology , Mutation , N-Acetylglucosaminyltransferases/genetics , Animals , Child, Preschool , Female , Heparitin Sulfate/physiology , Humans , Induced Pluripotent Stem Cells/cytology , Infant , Lymphocytes/physiology , Zebrafish
16.
Shock ; 48(3): 340-345, 2017 09.
Article in English | MEDLINE | ID: mdl-28107214

ABSTRACT

Syndecan-1 (Sdc1) is considered a biomarker of injury to the endothelial glycocalyx following hemorrhagic shock, with shedding of Sdc1 deleterious. Resuscitation with fresh frozen plasma (FFP) has been correlated with restitution of pulmonary Sdc1 and reduction of lung injury, but the precise contribution of Sdc1 to FFPs protection in the lung remains unclear. Human lung endothelial cells were used to assess the time and dose-dependent effect of FFP on Sdc1 expression and the effect of Sdc1 silencing on in vitro endothelial cell permeability and actin stress fiber formation. Wild-type and Sdc1 mice were subjected to hemorrhagic shock followed by resuscitation with lactated Ringers (LR) or FFP and compared with shock alone and shams. Lungs were harvested after 3 h for analysis of permeability, histology, and inflammation and for measurement of syndecan- 2 and 4 expression. In vitro, FFP enhanced pulmonary endothelial Sdc1 expression in time- and dose-dependent manners and loss of Sdc1 in pulmonary endothelial cells worsened permeability and stress fiber formation by FFP. Loss of Sdc1 in vivo led to equivalency between LR and FFP in restoring pulmonary injury, inflammation, and permeability after shock. Lastly, Sdc1 mice demonstrated a significant increase in pulmonary syndecan 4 expression after hemorrhagic shock and FFP-based resuscitation. Taken together, our findings support a key role for Sdc1 in modulating pulmonary protection by FFP after hemorrhagic shock. Our results also suggest that other members of the syndecan family may at least be contributing to FFP's effects on the endothelium, an area that warrants further investigation.


Subject(s)
Endothelial Cells/metabolism , Lung/metabolism , Plasma , Shock, Hemorrhagic/metabolism , Syndecan-1/metabolism , Animals , Cells, Cultured , Endothelial Cells/pathology , Lung/pathology , Mice , Mice, Knockout , Resuscitation , Shock, Hemorrhagic/genetics , Shock, Hemorrhagic/pathology , Shock, Hemorrhagic/therapy , Syndecan-1/genetics
17.
Front Biosci (Landmark Ed) ; 21(6): 1260-77, 2016 06 01.
Article in English | MEDLINE | ID: mdl-27100505

ABSTRACT

Glycosaminoglycans (GAGs) are complex linear polysaccharides expressed in intracellular compartments, at the cell surface, and in the extracellular environment where they interact with various molecules to regulate many cellular processes implicated in health and disease. Subversion of GAGs is a pathogenic strategy shared by a wide variety of microbial pathogens, including viruses, bacteria, parasites, and fungi. Pathogens use GAGs at virtually every major portals of entry to promote their attachment and invasion of host cells, movement from one cell to another, and to protect themselves from immune attack. Pathogens co-opt fundamental activities of GAGs to accomplish these tasks. This ingenious strategy to subvert essential activities of GAGs likely prevented host organisms from deleting or inactivating these mechanisms during their evolution. The goal of this review is to provide a mechanistic overview of our current understanding of how microbes subvert GAGs at major steps of pathogenesis, using select GAG-pathogen interactions as representative examples.


Subject(s)
Glycosaminoglycans/metabolism , Infections/metabolism , Animals , Blood-Borne Pathogens , Eye Infections/etiology , Eye Infections/metabolism , Gastrointestinal Diseases/etiology , Gastrointestinal Diseases/metabolism , Host-Pathogen Interactions/physiology , Humans , Infections/etiology , Respiratory Tract Infections/etiology , Respiratory Tract Infections/metabolism , Skin Diseases/etiology , Skin Diseases/metabolism , Urogenital System/metabolism , Urogenital System/microbiology
18.
Am J Respir Cell Mol Biol ; 55(2): 243-51, 2016 08.
Article in English | MEDLINE | ID: mdl-26934670

ABSTRACT

Although neutrophils play critical roles in innate immunity, in excess these cells cause severe tissue damage. Thus, neutrophil activation must be tightly regulated to prevent indiscriminant damage. Previously, we reported that mice lacking matrix metalloproteinase (MMP) 7 are protected from lung injury owing to markedly impaired neutrophil movement from the interstitium into mucosal lumenal spaces. This phenotype resulted from a lack of MMP7 shedding of syndecan-1, a heparan sulfate proteoglycan that carries the neutrophil chemokine CXCL1 as cargo. Here, we assessed if shedding syndecan-1/CXCL1 complexes affects neutrophil activation. Whereas injured monolayers of wild-type alveolar type II cells potently stimulated neutrophil activation, as gauged by release of myeloperoxidase, cells from Mmp7(-/-) or syndecan-1-null (Sdc1(-/-)) mice or human cells with MMP7 knockdown did not. In vivo, we observed reduced myeloperoxidase release relative to neutrophil numbers in bleomycin-injured Mmp7(-/-) and Sdc1(-/-) mice. Furthermore, we determined that soluble syndecan-1 directly stimulated neutrophil activation in the absence of cellular damage. These data indicate that MMP7 shedding of syndecan-1/CXCL1 complexes functions as a checkpoint that restricts neutrophil activation at sites of epithelial injury.


Subject(s)
Chemokine CXCL1/metabolism , Epithelial Cells/metabolism , Matrix Metalloproteinase 7/metabolism , Neutrophil Activation , Syndecan-1/metabolism , Alveolar Epithelial Cells/metabolism , Alveolar Epithelial Cells/pathology , Animals , Female , Male , Mice, Inbred C57BL , Models, Biological , Neutrophils/metabolism
19.
J Cell Biol ; 210(7): 1199-211, 2015 Sep 28.
Article in English | MEDLINE | ID: mdl-26391658

ABSTRACT

Transmembrane heparan sulfate proteoglycans regulate multiple aspects of cell behavior, but the molecular basis of their signaling is unresolved. The major family of transmembrane proteoglycans is the syndecans, present in virtually all nucleated cells, but with mostly unknown functions. Here, we show that syndecans regulate transient receptor potential canonical (TRPCs) channels to control cytosolic calcium equilibria and consequent cell behavior. In fibroblasts, ligand interactions with heparan sulfate of syndecan-4 recruit cytoplasmic protein kinase C to target serine714 of TRPC7 with subsequent control of the cytoskeleton and the myofibroblast phenotype. In epidermal keratinocytes a syndecan-TRPC4 complex controls adhesion, adherens junction composition, and early differentiation in vivo and in vitro. In Caenorhabditis elegans, the TRPC orthologues TRP-1 and -2 genetically complement the loss of syndecan by suppressing neuronal guidance and locomotory defects related to increases in neuronal calcium levels. The widespread and conserved syndecan-TRPC axis therefore fine tunes cytoskeletal organization and cell behavior.


Subject(s)
Caenorhabditis elegans Proteins/metabolism , Caenorhabditis elegans/metabolism , Calcium/metabolism , Cytosol/metabolism , Syndecan-4/metabolism , TRPC Cation Channels/metabolism , Animals , Caenorhabditis elegans/genetics , Caenorhabditis elegans Proteins/genetics , Cell Line , Humans , Mice , Mice, Mutant Strains , Protein Kinase C/genetics , Protein Kinase C/metabolism , Rats , Syndecan-4/genetics , TRPC Cation Channels/genetics
20.
Shock ; 44(5): 452-7, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26263434

ABSTRACT

We have shown in a rodent model of hemorrhagic shock (HS) that fresh frozen plasma (FFP) reduces lung inflammation and injury that are correlated with restitution of syndecan-1. As the gut is believed to contribute to distant organ injury and inflammation after shock, the current study sought to determine if the protective effects of plasma would extend to the gut and to elucidate the contribution of syndecan-1 to this protective effect. We also examined the potential role of TNFα, and a disintegrin and metalloproteinase (ADAM)-17, both intestinal sheddases of syndecan-1. Wild-type (WT) and syndecan-1 (KO) mice were subjected to HS followed by resuscitation with lactated Ringer's (LR) or FFP and compared with shock alone and shams. Small bowel and blood were obtained after 3  h for analysis of mucosal injury and inflammation and TNFα and ADAM-17 protein expression and activity. After HS, gut injury and inflammation were significantly increased compared with shams. Resuscitation with LR decreased both injury and inflammation that were further lessened by FFP. KO mice displayed worsened gut injury and inflammation after HS compared with WT mice, and LR and FFP equivalently inhibited injury and inflammation. Both systemic and intestinal TNFα and ADAM-17 followed similar trends, with increases after HS, reduction by LR, and a further decrease by FFP in WT but not KO mice. In conclusion, FFP decreased gut injury and inflammation after hemorrhagic shock, an effect that was abrogated in syndecan-1 mice. Plasma also decreased TNFα and ADAM-17, representing a potential mechanistic link to its protection via syndecan-1.


Subject(s)
Intestinal Diseases/prevention & control , Plasma , Shock, Hemorrhagic/therapy , Syndecan-1/physiology , ADAM Proteins/metabolism , ADAM17 Protein , Animals , Disease Models, Animal , Enteritis/etiology , Enteritis/metabolism , Enteritis/pathology , Enteritis/prevention & control , Intestinal Diseases/etiology , Intestinal Diseases/metabolism , Intestinal Diseases/pathology , Intestinal Mucosa/metabolism , Intestinal Mucosa/pathology , Intestine, Small/metabolism , Intestine, Small/pathology , Male , Mice, Knockout , Shock, Hemorrhagic/complications , Shock, Hemorrhagic/metabolism , Syndecan-1/deficiency , Syndecan-1/genetics , Tumor Necrosis Factor-alpha/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...