Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 30
Filter
Add more filters










Publication year range
1.
Cell ; 187(11): 2703-2716.e23, 2024 May 23.
Article in English | MEDLINE | ID: mdl-38657602

ABSTRACT

Antigen presentation defects in tumors are prevalent mechanisms of adaptive immune evasion and resistance to cancer immunotherapy, whereas how tumors evade innate immunity is less clear. Using CRISPR screens, we discovered that IGSF8 expressed on tumors suppresses NK cell function by interacting with human KIR3DL2 and mouse Klra9 receptors on NK cells. IGSF8 is normally expressed in neuronal tissues and is not required for cell survival in vitro or in vivo. It is overexpressed and associated with low antigen presentation, low immune infiltration, and worse clinical outcomes in many tumors. An antibody that blocks IGSF8-NK receptor interaction enhances NK cell killing of malignant cells in vitro and upregulates antigen presentation, NK cell-mediated cytotoxicity, and T cell signaling in vivo. In syngeneic tumor models, anti-IGSF8 alone, or in combination with anti-PD1, inhibits tumor growth. Our results indicate that IGSF8 is an innate immune checkpoint that could be exploited as a therapeutic target.


Subject(s)
Immunity, Innate , Immunotherapy , Killer Cells, Natural , Neoplasms , Animals , Female , Humans , Mice , Antigen Presentation , Cell Line, Tumor , Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , Membrane Proteins/metabolism , Mice, Inbred C57BL , Neoplasms/immunology , Neoplasms/therapy
3.
Mol Cancer Ther ; 21(6): 914-924, 2022 06 01.
Article in English | MEDLINE | ID: mdl-35313332

ABSTRACT

Stimulator of interferon genes (STING) is an innate immune receptor activated by natural or synthetic agonists to elicit antitumoral immune response via type I IFNs and other inflammatory cytokines. Bacillus Calmette-Guerin (BCG) is the standard of care as intravesical therapy for patients with high-risk non-muscle invasive bladder cancer (NMIBC). There are limited options available for patients with NMIBC who developed BCG unresponsiveness. In this study, we characterized in vitro and in vivo antitumor effects of E7766, a macrocyle-bridged STING agonist, via intravesical instillation in two syngeneic orthotopic murine NMIBC tumor models resistant to therapeutic doses of BCG and anti-PD-1 agents. E7766 bound to recombinant STING protein with a Kd value of 40 nmol/L and induced IFNß expression in primary human peripheral blood mononuclear cells harboring any of seven major STING genotypes with EC50 values of 0.15 to 0.79 µmol/L. Intravesical E7766 was efficacious in both NMIBC models with induction of effective immunologic memory in the treated animals. Pharmacologic activation of the STING pathway in the bladder resulted in IFN pathway activation, infiltration of T cells and natural killer (NK) cells, dendritic cell activation, and antigen presentation in bladder epithelium, leading to the antitumor activity and immunity. In addition, measurements of the pharmacodynamic markers, Ifnß1 and CXCL10, in bladder, urine, and plasma, and of STING pathway intactness in cancer cells, supported this mode of action. Taken together, our studies reveal an antitumor immune effect of pharmacologic activation of the STING pathway in bladder epithelium and thus provide a rationale for subsequent clinical studies in patients with NMIBC.


Subject(s)
Phosphatidylinositol 3-Kinases , Urinary Bladder Neoplasms , Animals , BCG Vaccine/pharmacology , Cell Line, Tumor , Cell Proliferation , Humans , Leukocytes, Mononuclear/metabolism , Mice , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction , TOR Serine-Threonine Kinases/metabolism , Urinary Bladder/metabolism , Urinary Bladder/pathology , Urinary Bladder Neoplasms/drug therapy , Urinary Bladder Neoplasms/pathology
4.
ChemMedChem ; 16(11): 1740-1743, 2021 06 07.
Article in English | MEDLINE | ID: mdl-33522135

ABSTRACT

A strategy for creating potent and pan-genotypic stimulator of interferon genes (STING) agonists is described. Locking a bioactive U-shaped conformation of cyclic dinucleotides by introducing a transannular macrocyclic bridge between the nucleic acid bases leads to a topologically novel macrocycle-bridged STING agonist (MBSA). In addition to substantially enhanced potency, the newly designed MBSAs, exemplified by clinical candidate E7766, exhibit broad pan-genotypic activity in all major human STING variants. E7766 is shown to have potent antitumor activity with long lasting immune memory response in a mouse liver metastatic tumor model. Two complementary stereoselective synthetic routes to E7766 are also described.


Subject(s)
Antineoplastic Agents/pharmacology , Interferons/agonists , Macrocyclic Compounds/pharmacology , Animals , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Cell Line, Tumor , Cell Proliferation/drug effects , Drug Screening Assays, Antitumor , Humans , Macrocyclic Compounds/chemical synthesis , Macrocyclic Compounds/chemistry , Mice , Models, Molecular , Molecular Structure , Neoplasms, Experimental/drug therapy , Neoplasms, Experimental/pathology
5.
J Immunother Cancer ; 8(1)2020 06.
Article in English | MEDLINE | ID: mdl-32554609

ABSTRACT

BACKGROUND: E7046 is a highly selective, small-molecule antagonist of the E-type prostanoid receptor 4 (EP4) for prostaglandin E2, an immunosuppressive mediator of the tumor immune microenvironment. This first-in-human phase 1 study assessed the safety, tolerability, pharmacokinetics, pharmacodynamics, maximum tolerated dose (MTD) and recommended phase 2 dose of E7046. METHODS: This first-in-human study enrolled 30 patients with advanced tumors of cancer types associated with high levels of myeloid infiltrates. E7046 was administered orally once-daily in sequential escalating dose cohorts (125, 250, 500, and 750 mg) with ≥6 patients per cohort. Tumor assessments were performed every 6 weeks. Paired tumor biopsies and blood samples, before and on treatment, were collected for pharmacokinetic and pharmacodynamic characterization of the treatment. RESULTS: No dose-limiting toxicities were observed, and the MTD was not reached. E7046 had an elimination half-life (t1/2) of 12 hours, and drug exposure increased dose-dependently from 125 to 500 mg. Target modulation by E7046 was supported by changes in genes downstream of EP4 with concurrent enhanced antitumoral immune responses. A best response of stable disease (per irRECIST) was reported in 23% of patients treated with E7046 (n=30) (125 mg: n=2; 250 mg: n=2; 750 mg: n=3). Over half (4/7) of the patients with stable disease had treatment duration of 18 weeks or more, and three patients (3/15; 20%) achieved metabolic responses. CONCLUSIONS: In this first-in-human study, E7046 administered orally once daily demonstrated manageable tolerability, immunomodulatory effects, and a best response of stable disease (≥18 weeks) in several heavily pretreated patients with advanced malignancies. The 250 and 500 mg doses are proposed for further development in the combination setting. TRIAL REGISTRATION NUMBER: NCT02540291.


Subject(s)
Antineoplastic Agents, Immunological , Benzoates , Neoplasms/drug therapy , Pyrazoles , Receptors, Prostaglandin E, EP4 Subtype/antagonists & inhibitors , Administration, Oral , Adult , Aged , Antineoplastic Agents, Immunological/administration & dosage , Antineoplastic Agents, Immunological/adverse effects , Antineoplastic Agents, Immunological/pharmacokinetics , Benzoates/administration & dosage , Benzoates/adverse effects , Benzoates/pharmacokinetics , Female , Humans , Male , Maximum Tolerated Dose , Middle Aged , Neoplasm Staging , Neoplasms/diagnosis , Neoplasms/pathology , Pyrazoles/administration & dosage , Pyrazoles/adverse effects , Pyrazoles/pharmacokinetics , Response Evaluation Criteria in Solid Tumors , Tumor Microenvironment/drug effects , Tumor Microenvironment/immunology , Young Adult
6.
Cell Chem Biol ; 27(3): 259-268.e5, 2020 03 19.
Article in English | MEDLINE | ID: mdl-32017919

ABSTRACT

Carbamoyl phosphate synthetase 1 (CPS1) catalyzes the first step in the ammonia-detoxifying urea cycle, converting ammonia to carbamoyl phosphate under physiologic conditions. In cancer, CPS1 overexpression supports pyrimidine synthesis to promote tumor growth in some cancer types, while in others CPS1 activity prevents the buildup of toxic levels of intratumoral ammonia to allow for sustained tumor growth. Targeted CPS1 inhibitors may, therefore, provide a therapeutic benefit for cancer patients with tumors overexpressing CPS1. Herein, we describe the discovery of small-molecule CPS1 inhibitors that bind to a previously unknown allosteric pocket to block ATP hydrolysis in the first step of carbamoyl phosphate synthesis. CPS1 inhibitors are active in cellular assays, blocking both urea synthesis and CPS1 support of the pyrimidine biosynthetic pathway, while having no activity against CPS2. These newly discovered CPS1 inhibitors are a first step toward providing researchers with valuable tools for probing CPS1 cancer biology.


Subject(s)
Carbamoyl-Phosphate Synthase (Ammonia)/antagonists & inhibitors , Enzyme Inhibitors/pharmacology , Piperidines/pharmacology , Small Molecule Libraries/pharmacology , Thiazoles/pharmacology , Adenosine Triphosphate/antagonists & inhibitors , Adenosine Triphosphate/metabolism , Allosteric Regulation/drug effects , Carbamoyl-Phosphate Synthase (Ammonia)/genetics , Carbamoyl-Phosphate Synthase (Ammonia)/metabolism , Dose-Response Relationship, Drug , Enzyme Inhibitors/chemistry , Humans , Hydrolysis/drug effects , Models, Molecular , Molecular Structure , Piperidines/chemistry , Small Molecule Libraries/chemistry , Thiazoles/chemistry
7.
Oncoimmunology ; 6(8): e1338239, 2017.
Article in English | MEDLINE | ID: mdl-28920002

ABSTRACT

Reprogramming of immunosuppressive tumor microenvironment (TME) by targeting alternatively activated tumor associated macrophages (M2TAM), myeloid-derived suppressor cells (MDSC), and regulatory T cells (Tregs), represents a promising strategy for developing novel cancer immunotherapy. Prostaglandin E2 (PGE2), an arachidonic acid pathway metabolite and mediator of chronic inflammation, has emerged as a powerful immunosuppressor in the TME through engagement with one or more of its 4 receptors (EP1-EP4). We have developed E7046, an orally bioavailable EP4-specific antagonist and show here that E7046 has specific and potent inhibitory activity on PGE2-mediated pro-tumor myeloid cell differentiation and activation. E7046 treatment reduced the growth or even rejected established tumors in vivo in a manner dependent on both myeloid and CD8+ T cells. Furthermore, co-administration of E7046 and E7777, an IL-2-diphtheria toxin fusion protein that preferentially kills Tregs, synergistically disrupted the myeloid and Treg immunosuppressive networks, resulting in effective and durable anti-tumor immune responses in mouse tumor models. In the TME, E7046 and E7777 markedly increased ratios of CD8+granzymeB+ cytotoxic T cells (CTLs)/live Tregs and of M1-like/M2TAM, and converted a chronic inflammation phenotype into acute inflammation, shown by substantial induction of STAT1/IRF-1 and IFNγ-controlled genes. Notably, E7046 also showed synergistic anti-tumor activity when combined with anti-CTLA-4 antibodies, which have been reported to diminish intratumoral Tregs. Our studies thus reveal a specific myeloid cell differentiation-modifying activity by EP4 blockade and a novel combination of E7046 and E7777 as a means to synergistically mitigate both myeloid and Treg-derived immunosuppression for cancer treatment in preclinical models.

8.
Cancer Biol Ther ; 16(4): 589-601, 2015.
Article in English | MEDLINE | ID: mdl-25729885

ABSTRACT

Natural compound schweinfurthins are of considerable interest for novel therapy development because of their selective anti-proliferative activity against human cancer cells. We previously reported the isolation of highly active schweinfurthins E-H, and in the present study, mechanisms of the potent and selective anti-proliferation were investigated. We found that schweinfurthins preferentially inhibited the proliferation of PTEN deficient cancer cells by indirect inhibition of AKT phosphorylation. Mechanistically, schweinfurthins and their analogs arrested trans-Golgi-network trafficking, an intracellular vesicular trafficking system, resulting in the induction of endoplasmic reticulum stress and the suppression of both lipid raft-mediated PI3K activation and mTOR/RheB complex formation, which collectively led to an effective inhibition of mTOR/AKT signaling. The trans-Golgi-network traffic arresting effect of schweinfurthins was associated with their in vitro binding activity to oxysterol-binding proteins that are known to regulate intracellular vesicular trafficking. Moreover, schweinfurthins were found to be highly toxic toward PTEN-deficient B cell lymphoma cells, and displayed 2 orders of magnitude lower activity toward normal human peripheral blood mononuclear cells and primary fibroblasts in vitro. These results revealed a previously unrecognized role of schweinfurthins in regulating trans-Golgi-network trafficking, and linked mechanistically this cellular effect with mTOR/AKT signaling and with cancer cell survival and growth. Our findings suggest the schweinfurthin class of compounds as a novel approach to modulate oncogenic mTOR/AKT signaling for cancer treatment.


Subject(s)
Cell Proliferation/drug effects , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction/drug effects , Small Molecule Libraries/pharmacology , Stilbenes/pharmacology , TOR Serine-Threonine Kinases/metabolism , trans-Golgi Network/drug effects , Cell Line , Cell Line, Tumor , Cell Survival/drug effects , Fibroblasts/drug effects , Fibroblasts/metabolism , Humans , Leukocytes, Mononuclear/drug effects , Leukocytes, Mononuclear/metabolism , Lymphoma, B-Cell/metabolism , PTEN Phosphohydrolase/metabolism , Phosphatidylinositol 3-Kinases/metabolism
9.
Proc Natl Acad Sci U S A ; 111(22): 8173-8, 2014 Jun 03.
Article in English | MEDLINE | ID: mdl-24835176

ABSTRACT

Identification of carbohydrate sequences that determine affinity to specific chemokines is a critical step for strategies to interfere with chemokine-mediated leukocyte trafficking. Here, we first characterized the development of allergic asthma in Tie2-dependent and inducible Ext1-knockout (Tie2-Ext1(iKO)) mice. We showed that heparan sulfate is essential for leukocyte recruitment in the peribronchial region and bronchoalveolar lavage fluid (BALF), and is crucial for induction of airway hyperresponsiveness. Our glycan microarray showed a unique affinity profile of chemokine CCL20 to substructures of heparin and heparin-like oligo/di/monosaccharides. Among them, we identified a synthetic and not naturally occurring monosaccharide, 2,4-O-di-sulfated iduronic acid (Di-S-IdoA), as a potential inhibitor for CCL20-heparan sulfate interaction. Mice injected with Di-S-IdoA via tail vain or nasal inhalation showed attenuated leukocyte recruitment into inflammatory sites and BALF. These results demonstrate a critical role of chemokine-heparan sulfate interaction in the asthma development and Di-S-IdoA as a potential drug for asthma treatment.


Subject(s)
Asthma/drug therapy , Iduronic Acid/pharmacology , Sulfates/pharmacology , T-Lymphocytes/drug effects , Animals , Asthma/immunology , Bronchoalveolar Lavage Fluid/immunology , Carbohydrate Sequence , Chemokine CCL20/immunology , Chemokine CCL20/metabolism , Chemotaxis/immunology , Disease Models, Animal , Eosinophils/cytology , Eosinophils/drug effects , Eosinophils/immunology , Heparitin Sulfate/immunology , Heparitin Sulfate/metabolism , Iduronic Acid/chemical synthesis , Lung/immunology , Mice , Mice, Knockout , N-Acetylglucosaminyltransferases/genetics , Ovalbumin/immunology , Ovalbumin/pharmacology , Polysaccharides/immunology , Polysaccharides/metabolism , Receptor, TIE-2/genetics , Sulfates/chemical synthesis , T-Lymphocytes/cytology , T-Lymphocytes/immunology
10.
PLoS One ; 8(12): e78191, 2013.
Article in English | MEDLINE | ID: mdl-24312443

ABSTRACT

Approximately 10-15% of individuals infected with Helicobacter pylori will develop ulcer disease (gastric or duodenal ulcer), while most people infected with H. pylori will be asymptomatic. The majority of infected individuals remain asymptomatic partly due to the inhibition of synthesis of cholesteryl α-glucosides in H. pylori cell wall by α1,4-GlcNAc-capped mucin O-glycans, which are expressed in the deeper portion of gastric mucosa. However, it has not been determined how cholesteryl α-glucosyltransferase (αCgT), which forms cholesteryl α-glucosides, functions in the pathogenesis of H. pylori infection. Here, we show that the activity of αCgT from H. pylori clinical isolates is highly correlated with the degree of gastric atrophy. We investigated the role of cholesteryl α-glucosides in various aspects of the immune response. Phagocytosis and activation of dendritic cells were observed at similar degrees in the presence of wild-type H. pylori or variants harboring mutant forms of αCgT showing a range of enzymatic activity. However, cholesteryl α-glucosides were recognized by invariant natural killer T (iNKT) cells, eliciting an immune response in vitro and in vivo. Following inoculation of H. pylori harboring highly active αCgT into iNKT cell-deficient (Jα18(-/-)) or wild-type mice, bacterial recovery significantly increased in Jα18(-/-) compared to wild-type mice. Moreover, cytokine production characteristic of Th1 and Th2 cells dramatically decreased in Jα18(-/-) compared to wild-type mice. These findings demonstrate that cholesteryl α-glucosides play critical roles in H. pylori-mediated gastric inflammation and precancerous atrophic gastritis.


Subject(s)
Gastritis, Atrophic/immunology , Glucosides/immunology , Helicobacter Infections/immunology , Helicobacter pylori/immunology , Helicobacter pylori/pathogenicity , Natural Killer T-Cells/immunology , Adolescent , Adult , Aged , Aged, 80 and over , Animals , Child , Female , Gastritis, Atrophic/genetics , Gastritis, Atrophic/microbiology , Gastritis, Atrophic/pathology , Helicobacter Infections/genetics , Helicobacter Infections/pathology , Helicobacter pylori/genetics , Humans , Male , Mice , Mice, Knockout , Middle Aged , Natural Killer T-Cells/pathology , Phagocytosis/immunology , Th1 Cells/immunology , Th1 Cells/pathology , Th2 Cells/immunology , Th2 Cells/pathology
11.
Mol Biol Cell ; 23(5): 771-80, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22238358

ABSTRACT

Glycans of α-dystroglycan (α-DG), which is expressed at the epithelial cell-basement membrane (BM) interface, play an essential role in epithelium development and tissue organization. Laminin-binding glycans on α-DG expressed on cancer cells suppress tumor progression by attenuating tumor cell migration from the BM. However, mechanisms controlling laminin-binding glycan expression are not known. Here, we used small interfering RNA (siRNA) library screening and identified Fer kinase, a non-receptor-type tyrosine kinase, as a key regulator of laminin-binding glycan expression. Fer overexpression decreased laminin-binding glycan expression, whereas siRNA-mediated down-regulation of Fer kinase increased glycan expression on breast and prostate cancer cell lines. Loss of Fer kinase function via siRNA or mutagenesis increased transcription levels of glycosyltransferases, including protein O-mannosyltransferase 1, ß3-N-acetylglucosaminyltransferase 1, and like-acetylglucosaminyltransferase that are required to synthesize laminin-binding glycans. Consistently, inhibition of Fer expression decreased cell migration in the presence of laminin fragment. Fer kinase regulated STAT3 phosphorylation and consequent activation, whereas knockdown of STAT3 increased laminin-binding glycan expression on cancer cells. These results indicate that the Fer pathway negatively controls expression of genes required to synthesize laminin-binding glycans, thus impairing BM attachment and increasing tumor cell migration.


Subject(s)
Cell Movement , Dystroglycans/metabolism , Neoplasms/pathology , Protein-Tyrosine Kinases/metabolism , Basement Membrane/metabolism , Cell Line, Tumor , Down-Regulation , Gene Knockdown Techniques , Glycosylation , Humans , Lamins/metabolism , Mannosyltransferases/genetics , N-Acetylglucosaminyltransferases/genetics , Neoplasms/genetics , Neoplasms/metabolism , Phosphorylation , Protein-Tyrosine Kinases/genetics , RNA, Small Interfering/genetics , STAT3 Transcription Factor/metabolism , Transcription, Genetic , Tyrosine/metabolism
12.
J Biol Chem ; 286(37): 32824-33, 2011 Sep 16.
Article in English | MEDLINE | ID: mdl-21784847

ABSTRACT

Astrocytic tumor is the most prevalent primary brain tumor. However, the role of cell surface carbohydrates in astrocytic tumor invasion is not known. In a previous study, we showed that polysialic acid facilitates astrocytic tumor invasion and thereby tumor progression. Here, we examined the role of HNK-1 glycan in astrocytic tumor invasion. A Kaplan-Meier analysis of 45 patients revealed that higher HNK-1 expression levels were positively associated with increased survival of patients. To determine the role of HNK-1 glycan, we transfected C6 glioma cells, which lack HNK-1 glycan expression, with ß1,3-glucuronyltransferase-P cDNA, generating HNK-1-positive cells. When these cells were injected into the mouse brain, the resultant tumors were 60% smaller than tumors emerging from injection of the mock-transfected HNK-1-negative C6 cells. HNK-1-positive C6 cells also grew more slowly than mock-transfected C6 cells in anchorage-dependent and anchorage-independent assays. C6-HNK-1 cells migrated well after treatment of anti-ß1 integrin antibody, whereas the same treatment inhibited cell migration of mock-transfected C6 cells. Similarly, α-dystroglycan containing HNK-1 glycan is different from those containing the laminin-binding glycans, supporting the above conclusion that C6-HNK-1 cells migrate independently from ß1-integrin-mediated signaling. Moreover, HNK-1-positive cells exhibited attenuated activation of ERK 1/2 compared with mock-transfected C6 cells, whereas focal adhesion kinase activation was equivalent in both cell types. Overall, these results indicate that HNK-1 glycan functions as a tumor suppressor.


Subject(s)
Antigens, Neoplasm/metabolism , Astrocytoma/metabolism , Brain Neoplasms/metabolism , Cell Movement , Gene Expression Regulation, Neoplastic , Glucans/metabolism , Animals , Antibodies/pharmacology , Astrocytoma/pathology , Brain Neoplasms/pathology , Cell Line, Tumor , Enzyme Activation/drug effects , Female , Focal Adhesion Protein-Tyrosine Kinases/metabolism , Humans , Integrin beta1/metabolism , Male , Mice , Mice, Nude , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Neoplasm Proteins/metabolism , Neoplasm Transplantation
13.
BMC Biochem ; 12: 29, 2011 May 29.
Article in English | MEDLINE | ID: mdl-21619699

ABSTRACT

BACKGROUND: Previously, we have reported the presence of highly sulfated dermatans in solitary ascidians from the orders Phlebobranchia (Phallusia nigra) and Stolidobranchia (Halocynthia pyriformis and Styela plicata). Despite the identical disaccharide backbone, consisting of [→4IdoA(2S)ß-1→3GalNAcß-1→], those polymers differ in the position of sulfation on the N-Acetyl galactosamine, which can occur at carbon 4 or 6. We have shown that position rather than degree of sulfation is important for heparin cofactor II activity. As a consequence, 2,4- and 2,6-sulfated dermatans have high and low heparin cofactor II activities, respectively. In the present study we extended the disaccharide analysis of ascidian dermatan sulfates to additional species of the orders Stolidobranchia (Herdmania pallida, Halocynthia roretzi) and Phlebobranchia (Ciona intestinalis), aiming to investigate how sulfation evolved within Tunicata. In addition, we analysed how heparin cofactor II activity responds to dermatan sulfates containing different proportions of 2,6- or 2,4-disulfated units. RESULTS: Disaccharide analyses indicated a high content of disulfated disaccharide units in the dermatan sulfates from both orders. However, the degree of sulfation decreased from Stolidobranchia to Phlebobranchia. While 76% of the disaccharide units in dermatan sulfates from stolidobranch ascidians are disulfated, 53% of disulfated disaccharides are found in dermatan sulfates from phlebobranch ascidians. Besides this notable difference in the sulfation degree, dermatan sulfates from phlebobranch ascidians contain mainly 2,6-sulfated disaccharides whereas dermatan sulfate from the stolidobranch ascidians contain mostly 2,4-sulfated disaccharides, suggesting that the biosynthesis of dermatan sulfates might be differently regulated during tunicates evolution. Changes in the position of sulfation on N-acetylgalactosamine in the disaccharide [→4IdoA(2-Sulfate)ß-1→3GalNAcß-1→] modulate heparin cofactor II activity of dermatan sulfate polymers. Thus, high and low heparin cofactor II stimulating activity is observed in 2,4-sulfated dermatan sulfates and 2,6-sulfated dermatan sulfates, respectively, confirming the clear correlation between the anticoagulant activities of dermatan sulfates and the presence of 2,4-sulfated units. CONCLUSIONS: Our results indicate that in ascidian dermatan sulfates the position of sulfation on the GalNAc in the disaccharide [→4IdoA(2S)ß-1→3GalNAcß-1→] is directly related to the taxon and that the 6-O sulfation is a novelty apparently restricted to the Phlebobranchia. We also show that the increased content of [→4IdoA(2S)ß-1→3GalNAc(4S)ß-1→] disaccharide units in dermatan sulfates from Stolidobranchia accounts for the increased heparin cofactor II stimulating activity.


Subject(s)
Dermatan Sulfate/chemistry , Dermatan Sulfate/metabolism , Heparin Cofactor II/metabolism , Phylogeny , Urochordata/metabolism , Animals , Antithrombins/chemistry , Antithrombins/metabolism , Carbohydrate Sequence , Chondroitin ABC Lyase/metabolism , Electrophoresis, Agar Gel , Electrophoresis, Polyacrylamide Gel , Evolution, Molecular , Hexuronic Acids/metabolism , Humans , Molecular Sequence Data , Partial Thromboplastin Time , Species Specificity , Urochordata/genetics
14.
Am J Pathol ; 178(1): 423-33, 2011 Jan.
Article in English | MEDLINE | ID: mdl-21224079

ABSTRACT

L-selectin initiates lymphocyte interactions with high endothelial venules (HEVs) of lymphoid organs through binding to ligands with specific glycosylation modifications. 6-Sulfo sLe(x), a sulfated carbohydrate determinant for L-selectin, is carried on core 2 and extended core 1 O-glycans of HEV-expressed glycoproteins. The MECA-79 monoclonal antibody recognizes sulfated extended core 1 O-glycans and partially blocks lymphocyte-HEV interactions in lymphoid organs. Recent evidence has identified the contribution of 6-sulfo sLe(x) carried on N-glycans to lymphocyte homing in mice. Here, we characterize CL40, a novel IgG monoclonal antibody. CL40 equaled or surpassed MECA-79 as a histochemical staining reagent for HEVs and HEV-like vessels in mouse and human. Using synthetic carbohydrates, we found that CL40 bound to 6-sulfo sLe(x) structures, on both core 2 and extended core 1 structures, with an absolute dependency on 6-O-sulfation. Using transfected CHO cells and gene-targeted mice, we observed that CL40 bound its epitope on both N-glycans and O-glycans. Consistent with its broader glycan-binding, CL40 was superior to MECA-79 in blocking lymphocyte-HEV interactions in both wild-type mice and mice deficient in forming O-glycans. This superiority was more marked in human, as CL40 completely blocked lymphocyte binding to tonsillar HEVs, whereas MECA-79 inhibited only 60%. These findings extend the evidence for the importance of N-glycans in lymphocyte homing in mouse and indicate that this dependency also applies to human lymphoid organs.


Subject(s)
Antigens, Surface/immunology , L-Selectin/immunology , Lymph Nodes/immunology , Membrane Proteins/immunology , Palatine Tonsil/immunology , Polysaccharides/immunology , Animals , Antibodies, Monoclonal, Murine-Derived/immunology , CHO Cells , Cricetinae , Cricetulus , Epitopes/immunology , Epitopes/metabolism , Humans , L-Selectin/metabolism , Ligands , Mice , Polysaccharides/metabolism , Protein Processing, Post-Translational , Rats
15.
Immunity ; 33(5): 817-29, 2010 Nov 24.
Article in English | MEDLINE | ID: mdl-21093315

ABSTRACT

Heparan sulfate can bind several adhesion molecules involved in lymphocyte trafficking. However, the in vivo function of endothelial heparan sulfate in lymphocyte homing and stimulation of the immune response has not been elucidated. Here, we generated mutant mice deficient in the enzyme Ext1, which is required for heparan sulfate synthesis, in a Tek-dependent and inducible manner. Chemokine presentation was diminished in the mutant mice, causing the lack of appropriate integrin-mediated adhesion, and resulted in a marked decrease in lymphocyte sticking to high endothelial venules and in recruitment of resident dendritic cells through lymphatic vessels to the lymph nodes. As a consequence, mutant mice displayed a severe impairment in lymphocyte homing and a compromised contact hypersensitivity response. By contrast, lymphocyte rolling was increased because of loss of electrostatic repulsion by heparan sulfate. These results demonstrate critical roles of endothelial heparan sulfate in immune surveillance and immune response generation.


Subject(s)
Chemokines/immunology , Dendritic Cells/immunology , Heparitin Sulfate/immunology , Lymph Nodes/immunology , Lymphocytes/immunology , Animals , Cell Adhesion/immunology , Chemokines/metabolism , Dendritic Cells/metabolism , Endothelium, Vascular/immunology , Heparitin Sulfate/metabolism , Integrins/immunology , Lymph Nodes/metabolism , Lymphocytes/metabolism , Mice , Mice, Inbred C57BL , N-Acetylglucosaminyltransferases/deficiency , Venules/immunology , Venules/metabolism
16.
Methods Enzymol ; 479: 387-96, 2010.
Article in English | MEDLINE | ID: mdl-20816178

ABSTRACT

Interaction of epithelial cells with basement membrane (BM) is mediated by cell-adhesion molecules, which regulate cell proliferation, motility, and differentiation by integrating signals from extracellular matrix and soluble factors. alpha-Dystroglycan (alpha-DG) is one of the most important adhesion molecules in epithelial cell-BM interaction. alpha-DG serves as the cell surface receptor for several major BM proteins, including laminin, perlecan, and agrin. The laminin G-like domain in all these proteins binds to a unique glycan structure, so-called laminin-binding glycan, attached to alpha-DG with high affinity. Formation of the laminin-binding glycan is required for the BM assembly, and loss or deficiency of the glycan causes muscular dystrophy. We studied the role of this alpha-DG-specific glycan modification in tumor development, and identified a tumor suppressor function of the laminin-binding alpha-DG. In this chapter, we describe methods used to isolate the cell populations from human prostate cancer cell line PC3 and characterize their potentials in tumor formation and metastasis in vitro and in vivo.


Subject(s)
Dystroglycans/metabolism , Laminin/metabolism , Tumor Suppressor Proteins/metabolism , Cell Line, Tumor , Flow Cytometry , Humans , Male , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Protein Binding
17.
J Biol Chem ; 285(36): 27673-85, 2010 Sep 03.
Article in English | MEDLINE | ID: mdl-20584902

ABSTRACT

Endogenous pleiotrophin and hepatocyte growth factor (HGF) mediate the neurite outgrowth-promoting activity of chondroitin sulfate (CS)/dermatan sulfate (DS) hybrid chains isolated from embryonic pig brain. CS/DS hybrid chains isolated from shark skin have a different disaccharide composition, but also display these activities. In this study, pleiotrophin- and HGF-binding domains in shark skin CS/DS were investigated. A high affinity CS/DS fraction was isolated using a pleiotrophin-immobilized column. It showed marked neurite outgrowth-promoting activity and strong inhibitory activity against the binding of pleiotrophin to immobilized CS/DS chains from embryonic pig brain. The inhibitory activity was abolished by chondroitinase ABC or B, and partially reduced by chondroitinase AC-I. A pentasulfated hexasaccharide with a novel structure was isolated from the chondroitinase AC-I digest using pleiotrophin affinity and anion exchange chromatographies. It displayed a potent inhibitory effect on the binding of HGF to immobilized shark skin CS/DS chains, suggesting that the pleiotrophin- and HGF-binding domains at least partially overlap in the CS/DS chains involved in the neuritogenic activity. Computational chemistry using molecular modeling and calculations of the electrostatic potential of the hexasaccharide and two pleiotrophin-binding octasaccharides previously isolated from CS/DS hybrid chains of embryonic pig brain identified an electronegative zone potentially involved in the molecular recognition of the oligosaccharides by pleiotrophin. Homology modeling of pleiotrophin based on a related midkine protein structure predicted the binding pocket of pleiotrophin for the oligosaccharides and provided new insights into the molecular mechanism of the interactions between the oligosaccharides and pleiotrophin.


Subject(s)
Carrier Proteins/metabolism , Computer Simulation , Cytokines/metabolism , Hepatocyte Growth Factor/metabolism , Oligosaccharides/chemistry , Oligosaccharides/metabolism , Sulfuric Acids/chemistry , Animals , Brain/cytology , Carbohydrate Conformation , Carbohydrate Sequence , Chondroitin Sulfates/chemistry , Chondroitin Sulfates/metabolism , Chondroitin Sulfates/pharmacology , Chondroitinases and Chondroitin Lyases/metabolism , Dermatan Sulfate/chemistry , Dermatan Sulfate/metabolism , Dermatan Sulfate/pharmacology , Humans , Models, Molecular , Neurites/drug effects , Neurites/metabolism , Protein Binding , Static Electricity
18.
Proc Natl Acad Sci U S A ; 106(29): 12109-14, 2009 Jul 21.
Article in English | MEDLINE | ID: mdl-19587235

ABSTRACT

Alpha-dystroglycan (alpha-DG) represents a highly glycosylated cell surface molecule that is expressed in the epithelial cell-basement membrane (BM) interface and plays an essential role in epithelium development and tissue organization. The alpha-DG-mediated epithelial cell-BM interaction is often impaired in invasive carcinomas, yet roles and underlying mechanisms of such an impaired interaction in tumor progression remain unclear. We report here a suppressor function of laminin-binding glycans on alpha-DG in tumor progression. In aggressive prostate and breast carcinoma cell lines, laminin-binding glycans are dramatically decreased, although the amount of alpha-DG and beta-dystroglycan is maintained. The decrease of laminin-binding glycans and consequent increased cell migration were associated with the decreased expression of beta3-N-acetylglucosaminyltransferase-1 (beta3GnT1). Forced expression of beta3GnT1 in aggressive cancer cells restored the laminin-binding glycans and decreased tumor formation. beta3GnT1 was found to be required for laminin-binding glycan synthesis through formation of a complex with LARGE, thus regulating the function of LARGE. Interaction of the laminin-binding glycans with laminin and other adhesive molecules in BM attenuates tumor cell migratory potential by antagonizing ERK/AKT phosphorylation induced by the components in the ECM. These results identify a previously undescribed role of carbohydrate-dependent cell-BM interaction in tumor suppression and its control by beta3GnT1 and LARGE.


Subject(s)
Dystroglycans/metabolism , Laminin/metabolism , N-Acetylglucosaminyltransferases/metabolism , Tumor Suppressor Proteins/metabolism , Breast Neoplasms/enzymology , Breast Neoplasms/pathology , Cell Movement , Extracellular Matrix/metabolism , Female , Glycosylation , Humans , Integrins/metabolism , Ligands , Male , Models, Biological , Phenotype , Polysaccharides/metabolism , Prostatic Neoplasms/enzymology , Prostatic Neoplasms/pathology , Protein Binding , Signal Transduction
19.
J Biol Chem ; 284(25): 17157-17169, 2009 Jun 19.
Article in English | MEDLINE | ID: mdl-19395705

ABSTRACT

Although there are numerous reports of carbohydrates enriched in cancer cells, very few studies have addressed the functions of carbohydrates present in normal cells that decrease in cancer cells. It has been reported that core3 O-glycans are synthesized in normal gastrointestinal cells but are down-regulated in cancer cells. To determine the roles of core3 O-glycans, we transfected PC3 and LNCaP prostate cancer cells with beta3-N-acetylglucosaminyltransferase-6 (core3 synthase) required to synthesize core3 O-glycans. Both engineered cell lines exhibited reduced migration and invasion through extracellular matrix components compared with mock-transfected cells. Moreover we found that alpha2beta1 integrin acquired core3 O-glycans in cells expressing core3 synthase with decreased maturation of beta1 integrin, leading to decreased levels of the alpha2beta1 integrin complex, decreased activation of focal adhesion kinase, and reduced lamellipodia formation. Upon inoculation into the prostate of nude mice, PC3 cells expressing core3 O-glycans produced much smaller tumors without metastasis to the surrounding lymph nodes in contrast to robust tumor formation and metastasis seen in mock-transfected PC3 cells. Similarly LNCaP cells expressing core3 O-glycans barely produced subcutaneous tumors in contrast to robust tumor formation by mock-transfected LNCaP cells. These findings indicate that addition of core3 O-glycans to beta1 and alpha2 integrin subunits in prostate cancer cells suppresses tumor formation and tumor metastasis.


Subject(s)
Integrin alpha2beta1/metabolism , N-Acetylglucosaminyltransferases/metabolism , Prostatic Neoplasms/metabolism , Animals , Base Sequence , Cell Line, Tumor , DNA Primers/genetics , Down-Regulation , Humans , Lymphatic Metastasis/prevention & control , Male , Mice , Mice, Nude , Neoplasm Transplantation , Polysaccharides/chemistry , Polysaccharides/metabolism , Prostatic Neoplasms/genetics , Prostatic Neoplasms/prevention & control , Prostatic Neoplasms/secondary , Pseudopodia/pathology , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Transfection , Transplantation, Heterologous
SELECTION OF CITATIONS
SEARCH DETAIL
...