Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 263
1.
J Med Chem ; 63(22): 13913-13950, 2020 11 25.
Article En | MEDLINE | ID: mdl-33155811

A series of tetrahydroisoquinoline-based benzodiazepine dimers were synthesized and tested for in vitro cytotoxicity against a panel of cancer cell lines. Structure-activity relationship investigation of various spacers guided by molecular modeling studies helped to identify compounds with picomolar activity. Payload 17 was conjugated to anti-mesothelin and anti-fucosylated monosialotetrahexosylganglioside (FucGM1) antibodies using lysosome-cleavable valine-citrulline dipeptide linkers via heterogeneous lysine conjugation and bacterial transglutaminase-mediated site-specific conjugation. In vitro, these antibody drug conjugates (ADCs) exhibited significant cytotoxic and target-mediated selectivity on human cancer cell lines. The pharmacokinetics and efficacy of these ADCs were further evaluated in gastric and lung cancer xenograft models in mice. Consistent pharmacokinetic profiles, high target specificity, and robust antitumor activity were observed in these models after a single dose of the ADC-46 (0.02 µmol/kg).


Antibodies, Monoclonal/chemistry , Antineoplastic Agents/pharmacology , Benzodiazepines/chemistry , Drug Design , Immunoconjugates/pharmacology , Small Cell Lung Carcinoma/drug therapy , Stomach Neoplasms/drug therapy , Tetrahydroisoquinolines/chemistry , Animals , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/metabolism , Antineoplastic Agents/chemistry , Apoptosis , Benzodiazepines/metabolism , Cell Proliferation , Female , G(M1) Ganglioside/analogs & derivatives , G(M1) Ganglioside/immunology , GPI-Linked Proteins/immunology , Humans , Immunoconjugates/chemistry , Lung Neoplasms/drug therapy , Lung Neoplasms/pathology , Mesothelin , Mice , Mice, SCID , Small Cell Lung Carcinoma/pathology , Stomach Neoplasms/pathology , Structure-Activity Relationship , Tetrahydroisoquinolines/metabolism , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
2.
Nanoscale ; 12(5): 2946-2960, 2020 Feb 07.
Article En | MEDLINE | ID: mdl-31994576

Effective treatment of glioma and other central nervous system (CNS) diseases is hindered by the presence of the blood-brain barrier (BBB). A novel nano-delivery vehicle system composed of PLGA-lysoGM1/DOX micelles was developed to cross the BBB for CNS treatment. We have shown that doxorubicin (DOX) as a model drug encapsulated in PLGA-lysoGM1 micelles can achieve up to 3.8% loading efficiency and 61.6% encapsulation efficiency by the orthogonal test design. Our in vitro experiments demonstrated that PLGA-lysoGM1/DOX micelles had a slow and sustainable drug release under physiological conditions and exhibited a high cellular uptake through the macropinocytosis and the autophagy/lysosomal pathways. In vivo experimental studies in zebrafish and mice confirmed that PLGA-lysoGM1/DOX micelles could cross the BBB and be specifically accumulated in the brain. Moreover, an excellent anti-glioma effect was observed in intracranial glioma-bearing rats. Therefore, PLGA-lysoGM1/DOX micelles not only effectively can cross the BBB, but our results also suggest that they have great potential for anti-glioma therapy and other central nervous system diseases.


Blood-Brain Barrier/metabolism , Brain Neoplasms/drug therapy , Doxorubicin , Drug Delivery Systems , G(M1) Ganglioside/analogs & derivatives , Glioma/drug therapy , Polylactic Acid-Polyglycolic Acid Copolymer , Animals , Blood-Brain Barrier/pathology , Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Doxorubicin/pharmacokinetics , Doxorubicin/pharmacology , G(M1) Ganglioside/pharmacokinetics , G(M1) Ganglioside/pharmacology , Glioma/metabolism , Glioma/pathology , Humans , Male , Mice , Micelles , Polylactic Acid-Polyglycolic Acid Copolymer/pharmacokinetics , Polylactic Acid-Polyglycolic Acid Copolymer/pharmacology , Zebrafish
3.
FEBS J ; 287(12): 2612-2630, 2020 06.
Article En | MEDLINE | ID: mdl-31769916

A 15-kDa lectin, termed SeviL, was isolated from Mytilisepta virgata (purplish bifurcate mussel). SeviL forms a noncovalent dimer that binds strongly to ganglio-series GM1b oligosaccharide (Neu5Acɑ2-3Galß1-3GalNAcß1-4Galß1-4Glc) and its precursor, asialo-GM1 (Galß1-3GalNAcß1-4Galß1-4Glc). SeviL also interacts weakly with the glycan moiety of SSEA-4 hexaose (Neu5Acα2-3Galß1-3GalNAcß1-3Galα1-4Galß1-4Glc). A partial protein sequence of the lectin was determined by mass spectrometry, and the complete sequence was identified from transcriptomic analysis. SeviL, consisting of 129 amino acids, was classified as an R(icin B)-type lectin, based on the presence of the QxW motif characteristic of this fold. SeviL mRNA is highly expressed in gills and, in particular, mantle rim tissues. Orthologue sequences were identified in other species of the family Mytilidae, including Mytilus galloprovincialis, from which lectin MytiLec-1 was isolated and characterized in our previous studies. Thus, mytilid species contain lectins belonging to at least two distinct families (R-type lectins and mytilectins) that have a common ß-trefoil fold structure but differing glycan-binding specificities. SeviL displayed notable cytotoxic (apoptotic) effects against various cultured cell lines (human breast, ovarian, and colonic cancer; dog kidney) that possess asialo-GM1 oligosaccharide at the cell surface. This cytotoxic effect was inhibited by the presence of anti-asialo-GM1 oligosaccharide antibodies. With HeLa ovarian cancer cells, SeviL showed dose- and time-dependent activation of kinase MKK3/6, p38 MAPK, and caspase-3/9. The transduction pathways activated by SeviL via the glycosphingolipid oligosaccharide were triggered apoptosis. DATABASE: Nucleotide sequence data have been deposited in the GenBank database under accession numbers MK434191, MK434192, MK434193, MK434194, MK434195, MK434196, MK434197, MK434198, MK434199, MK434200, and MK434201.


G(M1) Ganglioside/analogs & derivatives , Lectins/chemistry , Lectins/metabolism , Mitogen-Activated Protein Kinases/metabolism , Mytilidae/chemistry , Oligosaccharides/metabolism , Animals , Binding Sites , G(M1) Ganglioside/chemistry , G(M1) Ganglioside/metabolism , HeLa Cells , Humans , Lectins/isolation & purification , Mitogen-Activated Protein Kinases/chemistry , Mytilidae/metabolism , Oligosaccharides/chemistry , Species Specificity
4.
Sci Rep ; 9(1): 12243, 2019 08 22.
Article En | MEDLINE | ID: mdl-31439922

Cholera is a life-threatening diarrhoeal disease caused by the human pathogen Vibrio cholerae. Infection occurs after ingestion of the bacteria, which colonize the human small intestine and secrete their major virulence factor - the cholera toxin (CT). The GM1 ganglioside is considered the primary receptor of the CT, but recent studies suggest that also fucosylated receptors such as histo-blood group antigens are important for cellular uptake and toxicity. Recently, a special focus has been on the histo-blood group antigen Lewisx (Lex), however, where and how the CT binds to Lex remains unclear. Here we report the high-resolution crystal structure (1.5 Å) of the receptor-binding B-subunits of the CT bound to the Lex trisaccharide, and complementary quantitative binding data for CT holotoxins. Lex, and also L-fucose alone, bind to the secondary binding site of the toxin, distinct from the GM1 binding site. In contrast, fucosyl-GM1 mainly binds to the primary binding site due to high-affinity interactions of its GM1 core. Lex is the first histo-blood group antigen of non-secretor phenotype structurally investigated in complex with CT. Together with the quantitative binding data, this allows unique insight into why individuals with non-secretor phenotype are more prone to severe cholera than so-called 'secretors'.


Blood Group Antigens/metabolism , Cholera Toxin/chemistry , Cholera/metabolism , G(M1) Ganglioside/analogs & derivatives , Vibrio cholerae/metabolism , Binding Sites , Blood Group Antigens/chemistry , Cholera/microbiology , Cholera Toxin/metabolism , G(M1) Ganglioside/chemistry , G(M1) Ganglioside/metabolism , Glycosylation , Humans , Protein Binding , Vibrio cholerae/chemistry , Vibrio cholerae/genetics
5.
Nano Lett ; 19(3): 1875-1882, 2019 03 13.
Article En | MEDLINE | ID: mdl-30719917

Viruses, such as influenza A, typically bind to the plasma membrane of their host by engaging multiple membrane receptors in parallel, thereby forming so-called multivalent interactions that are created by the collective action of multiple weak ligand-receptor bonds. The overall interaction strength can be modulated by changing the number of engaged receptors. This feature is used by viruses to achieve a sufficiently firm attachment to the host's plasma membrane but also allows progeny viruses to leave the plasma membrane after completing the virus replication cycle. Design of strategies to prevent infection, for example, by disturbing these attachment and detachment processes upon application of antivirals, requires quantification of the underlying multivalent interaction in absence and presence of antivirals. This is still an unresolved problem, as there is currently no approach available that allows for determining the valency (i.e., of the number of receptors bound to a particular virus) on the level of single viruses under equilibrium conditions. Herein, we track the motion of single influenza A/X31 viruses (IAVs; interacting with the ganglioside GD1a incorporated in a supported lipid bilayer) using total internal reflection fluorescence microscopy and show that IAV residence time distributions can be deconvoluted from valency effects by taking the IAV mobility into account. The so-derived off-rate distributions, expressed in dependence of an average, apparent valency, show the expected decrease in off-rate with increasing valency but also show an unexpected peak structure, which can be linked to a competition in the opposing functionalities of the two influenza A virus spike proteins, hemagglutinin (HA), and neuraminidase (NA). By application of the antiviral zanamivir that inhibits the activity of NA, we provide direct evidence, how the HA/NA balance modulates this virus-receptor interaction, allowing us to assess the inhibition concentration of zanamivir based on its effect on the multivalent interaction.


Hemagglutinins/chemistry , Influenza, Human/virology , Neuraminidase/chemistry , Receptors, Virus/chemistry , Cell Membrane/chemistry , G(M1) Ganglioside/analogs & derivatives , G(M1) Ganglioside/chemistry , Hemagglutinins/genetics , Humans , Influenza A virus/chemistry , Influenza A virus/genetics , Influenza A virus/pathogenicity , Influenza, Human/genetics , Lipid Bilayers/chemistry , Neuraminidase/antagonists & inhibitors , Neuraminidase/genetics , Receptors, Virus/genetics , Zanamivir
6.
Curr Eye Res ; 44(6): 664-670, 2019 06.
Article En | MEDLINE | ID: mdl-30688114

Purpose: Most complex gangliosides in vertebrates are formed from ganglioside GM3. GM3 deficiency in humans can result in epilepsy and visual impairment. To investigate whether a deficiency of GM3 is involved in visual function, ST3GAL5-/- mice with mutations in the ST3GAL5 gene-coded GM3 synthase were employed. Materials and Methods: Sixty mice were employed in this study. The glycosphingolipids of mice retinas were analyzed through high performance thin layer chromatography. The morphology of the optic nerves and retinas were evaluated by hematoxylin and eosin staining and immunohistochemical analysis using an anti-glial fibrillary acidic protein (GFAP) antibody. An electroretinogram (ERG) was applied on the eyes of 4, 9, 12, and 14-month-old mice. Also, visual evoked potential (VEP) was applied on 13-month-old mice. Results: The GM3 in the retinas was detected in ST3GAL5+/+ mice but not ST3GAL5-/- mice. Also, GM1b and GD1α expressions and lactosylceramide accumulation were found in the ST3GAL5-/- mouse retinas. There was no significant difference in GFAP expression in the retinas or optic discs between ST3GAL5+/+ and ST3GAL5-/- mice. Furthermore, the outcome of ERG and VEP analysis showed no disparity between the two strains in 13 and 14-month-old mice. Conclusion: In the eye, neither histopathological abnormalities nor abnormal functions of the retina were found in GM3-deficient mice. Differing from the situation in patients with GM3 deficiency, the lack of GM3 in mice did not lead to optic nerve atrophy.


Retina/enzymology , Sialyltransferases/deficiency , Visual Acuity/physiology , Animals , Antigens, CD/metabolism , Drug Combinations , Electroretinography , Evoked Potentials, Visual/physiology , G(M1) Ganglioside/analogs & derivatives , G(M1) Ganglioside/metabolism , Glial Fibrillary Acidic Protein/metabolism , Lactosylceramides/metabolism , Mice , Mice, Inbred C57BL , Mydriatics/pharmacology , Phenylephrine/pharmacology , Protein Kinase C-alpha/metabolism , Pupil/drug effects , Tropicamide/pharmacology
7.
Glycobiology ; 29(3): 242-259, 2019 03 01.
Article En | MEDLINE | ID: mdl-30535277

The hepatitis B virus (HBV)-induced chronic liver diseases are serious health threats worldwide. There is evidence to display the alterations of salivary N-linked glycans related to the development of HBV-infected liver diseases. Here, we further investigated the alterations of fucosylated N/O-glycans recognized by LTL in saliva from 120 subjects (30 healthy volunteers (HV), 30 patients with hepatitis B (HB), 30 patients with hepatic cirrhosis (HC), and 30 patients with hepatocellular carcinoma (HCC)) using salivary microarrys and MALDI-TOF/TOF-MS. The results showed that the expression level of fucosylated glycans recognized by LTL was significantly increased in HCC compared with other subjects (P < 0.0001). Besides, the fucosylated glycoproteins were isolated from pooled saliva of HV, HB, HC, and HCC by LTL-magnetic particle conjugates. Then, N/O- glycans were released from the isolated glycoproteins with PNGase F and NaClO, and were identified by MALDI-TOF-MS, respectively. Totally, there were 21/20, 25/18, 29/19, and 28/24 N/O-glycan peaks that were identified and annotated with proposed structures in saliva of HV, HB, HC, and HCC. Among the total, there were 8 N-glycan peaks (e.g., m/z 1905.634, 2158.777 and 2905.036) and 15 O-glycan peaks (e.g., 1177.407, 1308.444 and 1322.444) that only presented in patients with HBV-induced liver diseases. One N-glycan peak (m/z 2205.766) was unique in HC, and 9 O-glycan peaks (e.g., m/z 1157.420, 1163.417 and 1193.402) were unique in HCC. This study could facilitate the discovery of biomarkers for HC and HCC based on precise alterations of fucosylated N/O-glycans in saliva.


Biomarkers, Tumor/genetics , Hepatitis B virus/genetics , Polysaccharides/genetics , Protein Array Analysis , Biomarkers, Tumor/chemistry , Biomarkers, Tumor/isolation & purification , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/virology , Female , Fibrosis/genetics , Fibrosis/virology , G(M1) Ganglioside/analogs & derivatives , G(M1) Ganglioside/chemistry , G(M1) Ganglioside/genetics , Hepatitis B virus/isolation & purification , Hepatitis B virus/pathogenicity , Hepatitis, Chronic/genetics , Hepatitis, Chronic/virology , Humans , Lectins/chemistry , Liver Neoplasms/genetics , Liver Neoplasms/virology , Male , Polysaccharides/chemistry , Polysaccharides/isolation & purification , Saliva/chemistry , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization
8.
Clin Cancer Res ; 24(20): 5178-5189, 2018 10 15.
Article En | MEDLINE | ID: mdl-30021910

Purpose: The ganglioside fucosyl-GM1 (FucGM1) is a tumor-associated antigen expressed in a large percentage of human small cell lung cancer (SCLC) tumors, but absent in most normal adult tissues, making it a promising target in immuno-oncology. This study was undertaken to evaluate the preclinical efficacy of BMS-986012, a novel, nonfucosylated, fully human IgG1 antibody that binds specifically to FucGM1.Experimental Design: The antitumor activity of BMS-986012 was evaluated in in vitro assays using SCLC cells and in mouse xenograft and syngeneic tumor models, with and without chemotherapeutic agents and checkpoint inhibitors.Results: BMS-986012 showed a high binding affinity for FcγRIIIa (CD16), which resulted in enhanced antibody-dependent cellular cytotoxicity (ADCC) against FucGM1-expressing tumor cell lines. BMS-986012-mediated tumor cell killing was also observed in complement-dependent cytotoxicity (CDC) and antibody-dependent cellular phagocytosis (ADCP) assays. In several mouse SCLC models, BMS-986012 demonstrated efficacy and was well tolerated. In the DMS79 xenograft model, tumor regression was achieved with BMS-986012 doses of 0.3 mg/kg and greater; antitumor activity was enhanced when BMS-986012 was combined with standard-of-care cisplatin or etoposide. In a syngeneic model, tumors derived from a genetically engineered model of SCLC were treated with BMS-986012 or anti-FucGM1 with a mouse IgG2a Fc and their responses evaluated; when BMS-986012 was combined with anti-PD-1 or anti-CD137 antibody, therapeutic responses significantly improved.Conclusions: Single-agent BMS-986012 demonstrated robust antitumor activity, with the addition of chemotherapeutic or immunomodulatory agents further inhibiting SCLC growth in the same models. These preclinical data supported evaluation of BMS-986012 in a phase I clinical trial of patients with relapsed, refractory SCLC. Clin Cancer Res; 24(20); 5178-89. ©2018 AACR.


Antineoplastic Agents, Immunological/pharmacology , G(M1) Ganglioside/analogs & derivatives , Animals , Antibody-Dependent Cell Cytotoxicity/immunology , Antigens, Neoplasm/immunology , Carcinoma, Small Cell/drug therapy , Carcinoma, Small Cell/metabolism , Carcinoma, Small Cell/pathology , Cell Line, Tumor , Disease Models, Animal , Dose-Response Relationship, Drug , Drug Evaluation, Preclinical , G(M1) Ganglioside/antagonists & inhibitors , G(M1) Ganglioside/immunology , G(M1) Ganglioside/metabolism , Humans , Immunohistochemistry , Immunomodulation/drug effects , Lung Neoplasms/drug therapy , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Mice , Protein Binding , Receptors, IgG/metabolism , Tumor Necrosis Factor Receptor Superfamily, Member 9/antagonists & inhibitors , Xenograft Model Antitumor Assays
9.
Rapid Commun Mass Spectrom ; 32(17): 1481-1490, 2018 Sep 15.
Article En | MEDLINE | ID: mdl-29876976

RATIONALE: Certain lung cancer patients express elevated Fucosyl Monosialoganglioside (Fuc-GM1) in circulation compared to control groups. Several sensitive methods involving characterization of Fuc-GM1 have been reported. However, a highly specific and sensitive method for quantifying multiple potential Fuc-GM1 biomarkers present in various biological matrices has not been reported to date. METHODS: Individual Fuc-GM1 analogs in a commercially obtained standard mixture were characterized using HPLC/UV/MS and high-resolution mass spectrometry (HRMS). Proprietary antibodies, mAb1 and mAb2, were used to selectively capture and pre-concentrate the soluble and drug-bound forms of Fuc-GM1 molecules present in human serum and whole blood, eliminating the background matrix components. Immunocapture extraction (ICE) followed by HPLC/MS/MS was used to quantify specific Fuc-GM1 analogs in biological matrices. RESULTS: The concentration of individual Fuc-GM1 analogs in the standard mixture was estimated to be 7-34%, using HPLC/UV/MS. Using the standard mixture spiked into the biological matrices (100 µL), the lower limit of quantification (LLOQ) of each analog was 0.2-0.4 ng/mL with a dynamic range of up to 200 ng/mL. The applicability of the ICE-HPLC/MS/MS method was demonstrated by detecting endogenous Fuc-GM1 analogs present in rat blood and in several lung cancer cell lines. CONCLUSIONS: This highly specific and sensitive HPLC/MS/MS method for quantifying individual potential Fuc-GM1 biomarkers in serum and whole blood can play a critical role in patient stratification strategies and during drug treatment. This method can be employed for monitoring both free (soluble) form and antibody drug-bound Fuc-GM1.


Chromatography, High Pressure Liquid/methods , G(M1) Ganglioside/analogs & derivatives , Lung Neoplasms/blood , Tandem Mass Spectrometry/methods , Animals , Antibodies, Monoclonal/analysis , Biomarkers/blood , Biomarkers/chemistry , G(M1) Ganglioside/blood , G(M1) Ganglioside/chemistry , G(M1) Ganglioside/isolation & purification , Humans , Rats
10.
Org Biomol Chem ; 16(22): 4076-4080, 2018 06 06.
Article En | MEDLINE | ID: mdl-29789847

A highly efficient streamlined chemoenzymatic strategy for total synthesis of four prioritized ganglioside cancer antigens GD2, GD3, fucosyl GM1, and GM3 from commercially available lactose and phytosphingosine is demonstrated. Lactosyl sphingosine (LacßSph) was chemically synthesized (on a 13 g scale), subjected to sequential one-pot multienzyme (OPME) glycosylation reactions with facile C18-cartridge purification, followed by improved acylation conditions to form target gangliosides, including fucosyl GM1 which has never been synthesized before.


Antigens, Neoplasm/chemistry , G(M1) Ganglioside/analogs & derivatives , G(M3) Ganglioside/chemical synthesis , G(M1) Ganglioside/chemical synthesis , Glycosylation , Lactose/chemistry , Sphingosine/analogs & derivatives , Sphingosine/chemistry
11.
Methods Enzymol ; 597: 239-263, 2017.
Article En | MEDLINE | ID: mdl-28935104

Gangliosides, glycosphingolipids containing one or more sialic acids in the glycan chain, are involved in various important biological processes in cell plasma membranes (PMs). However, the behaviors and functions of gangliosides are poorly understood, primarily because of the lack of fluorescent analogs that are equivalent to native gangliosides that can be used as chemical and physical probes. In this study, we developed entirely chemical methods to synthesize fluorescent gangliosides (GM3, GM2, GM1, and GD1b) in which the glycan components are site-specifically labeled with various fluorescent dyes. The functional evaluations of the synthesized fluorescent gangliosides demonstrated the great influence of fluorescent dye on the physical properties of gangliosides in PMs and revealed the fluorescent ganglioside analogs which show similar behaviors to the native gangliosides.


Biochemistry/methods , G(M1) Ganglioside/chemistry , G(M2) Ganglioside/chemistry , G(M3) Ganglioside/chemistry , Gangliosides/chemistry , Animals , Cell Membrane/chemistry , Cell Membrane/metabolism , Fluorescent Dyes/chemical synthesis , Fluorescent Dyes/chemistry , G(M1) Ganglioside/analogs & derivatives , G(M1) Ganglioside/chemical synthesis , G(M2) Ganglioside/analogs & derivatives , G(M2) Ganglioside/chemical synthesis , G(M3) Ganglioside/analogs & derivatives , G(M3) Ganglioside/chemical synthesis , Gangliosides/chemical synthesis , Glycosphingolipids/chemical synthesis , Glycosphingolipids/chemistry , Membrane Microdomains , Sialic Acids/chemistry
12.
Protein Expr Purif ; 138: 1-6, 2017 Oct.
Article En | MEDLINE | ID: mdl-28673733

The cholera toxin B subunit (CTB) is a nontoxic portion of the cholera toxin that retains mucosal adjuvant properties. Expression of CTB in Escherichia coli is difficult as CTB aggregates and accumulates as insoluble inclusion bodies. To remedy this problem, the periplasmic chaperone, SKP, was investigated as possible co-expression partner to increase the solubility of recombinant CTB (rCTB) in E. coli. The result showed co-expression of SKP enhanced the soluble expression of rCTB in E. coli. Moreover, soluble rCTB was successfully expressed and secreted into the periplasmic space through the direction of the LTB leader signal. rCTB in periplasm was purified using an immobilized d-galactose resin; GM1-ELISA experiments showed that rCTB retains strong GM1 ganglioside-binding activity. Intranasal administration of ovalbumin (OVA) with rCTB significantly induced both mucosal and humoral immune responses specific to OVA. These data indicate that co-expression of the molecular chaperone SKP with CTB increased the solubility of rCTB while maintaining its function.


Adjuvants, Immunologic/genetics , Cholera Toxin/genetics , Escherichia coli/genetics , Immunity, Mucosal/drug effects , S-Phase Kinase-Associated Proteins/genetics , Adjuvants, Immunologic/administration & dosage , Administration, Intranasal , Animals , Cholera Toxin/administration & dosage , Cholera Toxin/immunology , Cloning, Molecular , Escherichia coli/metabolism , G(M1) Ganglioside/analogs & derivatives , G(M1) Ganglioside/chemistry , G(M1) Ganglioside/metabolism , Gene Expression , Immunization , Mice , Mice, Inbred C57BL , Ovalbumin/administration & dosage , Ovalbumin/immunology , Periplasm/chemistry , Periplasm/metabolism , Plasmids/chemistry , Plasmids/metabolism , Protein Binding , Recombinant Proteins/administration & dosage , Recombinant Proteins/genetics , Recombinant Proteins/immunology , S-Phase Kinase-Associated Proteins/metabolism , Solubility
13.
Biochim Biophys Acta Gen Subj ; 1861(10): 2494-2506, 2017 Oct.
Article En | MEDLINE | ID: mdl-28734966

Gangliosides are involved in a variety of biological roles and are a component of lipid rafts found in cell plasma membranes (PMs). Gangliosides are especially abundant in neuronal PMs and are essential to their physiological functions. However, the dynamic behaviors of gangliosides have not been investigated in living cells due to a lack of fluorescent probes that behave like their parental molecules. We have recently developed, using an entirely chemical method, four new ganglioside probes (GM1, GM2, GM3, and GD1b) that act similarly to their parental molecules in terms of raft partitioning and binding affinity. Using single fluorescent-molecule imaging, we have found that ganglioside probes dynamically enter and leave rafts featuring CD59, a GPI-anchored protein. This occurs both before and after stimulation. The residency time of our ganglioside probes in rafts with CD59 oligomers was 48ms, after stimulation. The residency times in CD59 homodimer and monomer rafts were 40ms and 12ms, respectively. In this review, we introduce an entirely chemical-based ganglioside analog synthesis method and describe its application in single-molecule imaging and for the study of the dynamic behavior of gangliosides in cell PMs. Finally, we discuss how raft domains are formed, both before and after receptor engagement. This article is part of a Special Issue entitled Neuro-glycoscience, edited by Kenji Kadomatsu and Hiroshi Kitagawa.


G(M1) Ganglioside/chemical synthesis , G(M2) Ganglioside/chemical synthesis , G(M3) Ganglioside/chemical synthesis , Gangliosides/chemical synthesis , Membrane Microdomains/metabolism , Molecular Probes/chemical synthesis , CD59 Antigens/chemistry , CD59 Antigens/metabolism , Carbohydrate Conformation , Carbohydrate Sequence , Cell Line , Epithelial Cells/cytology , Epithelial Cells/metabolism , Fluorescent Dyes/chemical synthesis , Fluorescent Dyes/metabolism , G(M1) Ganglioside/analogs & derivatives , G(M1) Ganglioside/metabolism , G(M2) Ganglioside/analogs & derivatives , G(M2) Ganglioside/metabolism , G(M3) Ganglioside/analogs & derivatives , G(M3) Ganglioside/metabolism , Gangliosides/metabolism , Humans , Hydrophobic and Hydrophilic Interactions , Membrane Microdomains/ultrastructure , Molecular Probes/metabolism , Single Molecule Imaging
14.
Int J Mol Sci ; 17(8)2016 Aug 11.
Article En | MEDLINE | ID: mdl-27529215

The ST6GALNAC5 gene that encodes an α2,6-sialyltransferase involved in the biosynthesis of α-series gangliosides, was previously identified as one of the genes that mediate breast cancer metastasis to the brain. We have shown that the expression of ST6GALNAC5 in MDA-MB-231 breast cancer cells resulted in the expression of GD1α ganglioside at the cell surface. By using a human blood-brain barrier in vitro model recently developed, consisting in CD34⁺ derived endothelial cells co-cultivated with pericytes, we show that ST6GALNAC5 expression decreased the interactions between the breast cancer cells and the human blood-brain barrier.


Blood-Brain Barrier/metabolism , Breast Neoplasms/metabolism , Sialyltransferases/metabolism , Animals , Antigens, CD34/metabolism , Breast Neoplasms/genetics , Cell Line, Tumor , Female , G(M1) Ganglioside/analogs & derivatives , G(M1) Ganglioside/metabolism , Humans , Mice , Pericytes/metabolism , Sialyltransferases/genetics
15.
ACS Chem Neurosci ; 7(8): 1107-11, 2016 08 17.
Article En | MEDLINE | ID: mdl-27276519

Ganglioside Hp-s1 is isolated from the ovary of sea urchin Diadema setosum. It exhibited better neuritogenic activity than GM1 in pheochromocytoma 12 cells. To explore the roles of glucosyl moiety of Hp-s1 in contributing to the neurogenic activity, we developed feasible procedures for synthesis of Hp-s1 analogues (2a-2f). The glucosyl moiety of Hp-s1 was replaced with α-glucose, α-galactose, ß-galactose, α-mannose, and ß-mannose, and their biological activities on SH-SY5Y cells and natural killer T (NKT) cells were evaluated. We found that the orientation of C-2 hydroxyl group at glucosyl moiety of Hp-s1 plays an important role to induce neurite outgrowth of SH-SY5Y cells. Surprisingly, compound 2d could activate NKT cells to produce interleukin 2, although it did not show great activity on neurite outgrowth of SH-SY5Y cells. In general, the Hp-s1 might be considered as a lead compound for the development of novel drugs aimed at modulating the activity of neuronal cells.


Cell Differentiation/drug effects , G(M1) Ganglioside , Neurogenesis/drug effects , Animals , Carbohydrate Conformation , Cell Line, Tumor , Cytokines/metabolism , G(M1) Ganglioside/analogs & derivatives , G(M1) Ganglioside/chemistry , G(M1) Ganglioside/pharmacology , Glycosylation/drug effects , Humans , Killer Cells, Natural/drug effects , Killer Cells, Natural/metabolism , Magnetic Resonance Spectroscopy , Mass Spectrometry , Mice , Models, Chemical , Molecular Structure , Neurites/drug effects , Neuroblastoma
16.
PLoS One ; 11(4): e0153265, 2016.
Article En | MEDLINE | ID: mdl-27070150

Protein-glycan recognition is often mediated by multivalent binding. These multivalent bindings can be further complicated by cooperative interactions between glycans and individual glycan binding subunits. Here we have demonstrated a nanocube-based lipid bilayer array capable of quantitatively elucidating binding dissociation constants, maximum binding capacity, and binding cooperativity in a high-throughput format. Taking cholera toxin B subunit (CTB) as a model cooperativity system, we studied both GM1 and GM1-like gangliosides binding to CTB. We confirmed the previously observed CTB-GM1 positive cooperativity. Surprisingly, we demonstrated fucosyl-GM1 has approximately 7 times higher CTB binding capacity than GM1. In order to explain this phenomenon, we hypothesized that the reduced binding cooperativity of fucosyl-GM1 caused the increased binding capacity. This was unintuitive, as GM1 exhibited higher binding avidity (16 times lower dissociation constant). We confirmed the hypothesis using a theoretical stepwise binding model of CTB. Moreover, by taking a mixture of fucosyl-GM1 and GM2, we observed the mild binding avidity fucosyl-GM1 activated GM2 receptors enhancing the binding capacity of the lipid bilayer surface. This was unexpected as GM2 receptors have negligible binding avidity in pure GM2 bilayers. These unexpected discoveries demonstrate the importance of binding cooperativity in multivalent binding mechanisms. Thus, quantitative analysis of multivalent protein-glycan interactions in heterogeneous glycan systems is of critical importance. Our user-friendly, robust, and high-throughput nanocube-based lipid bilayer array offers an attractive method for dissecting these complex mechanisms.


Cholera Toxin/metabolism , G(M1) Ganglioside/analogs & derivatives , Lipid Bilayers/metabolism , Cholera Toxin/chemistry , Cryoelectron Microscopy , G(M1) Ganglioside/chemistry , G(M1) Ganglioside/metabolism , G(M2) Ganglioside/chemistry , G(M2) Ganglioside/metabolism , In Vitro Techniques , Kinetics , Lipid Bilayers/chemistry , Metal Nanoparticles/chemistry , Metal Nanoparticles/ultrastructure , Models, Biological , Nanotechnology , Protein Binding , Silicon Dioxide/chemistry , Silver/chemistry , Surface Plasmon Resonance
17.
Glycobiology ; 26(9): 984-998, 2016 09.
Article En | MEDLINE | ID: mdl-27102283

Some gangliosides, sialic acid-containing glycosphingolipids, have been considered as tumor-associated antigens. GD1α or a GD1α synthase gene ST6GalNAc5 was reported to be involved in the metastasis of murine lymphomas or human breast cancers, respectively. But expression patterns of 0-series gangliosides GD1α and its precursor GM1b in human cancers have not yet been investigated mainly due to lack of specific antibodies. We established specific monoclonal antibodies (mAbs) reactive with GD1α or GM1b using gangliosides from brain tissues of GM3 synthase (St3gal5)-deficient mice as immunogens. We used GM2/GD2 synthase (B4galnt1)-deficient mice to immunize by liposomes embedded with GD1α or acidic glycolipid fractions from brain of St3gal5-deficient mice. Specificities of established mAbs as analyzed by enzyme-linked immunosorbent assay and thin-layer chromatography-immunostaining were very high among various gangliosides. Increased expression of GD1α and reduced GM1b in the St6galnac5 cDNA-transfected RAW117 cell line also substantiated the specificities of two mAbs. Then, we analyzed expression of GD1α and GM1b, and of relevant glycosyltransferase genes in various human cancer cell lines using generated anti-GD1α mAb 122 or anti-GM1b mAb MR155A-7. A few human cancer cell lines showed significant expression of these gangliosides with reasonable expression of relevant glycosyltransferase genes.


G(M1) Ganglioside/analogs & derivatives , N-Acetylgalactosaminyltransferases/genetics , Sialyltransferases/genetics , Animals , Antibodies, Monoclonal/genetics , Antibodies, Monoclonal/immunology , Antigens, Neoplasm/genetics , Cell Line, Tumor , G(M1) Ganglioside/biosynthesis , G(M1) Ganglioside/genetics , G(M1) Ganglioside/metabolism , Gangliosides/genetics , Gangliosides/metabolism , Gene Expression Regulation, Enzymologic , Glycosphingolipids/genetics , Glycosphingolipids/immunology , Glycosphingolipids/metabolism , Humans , Mice , Mice, Knockout , N-Acetylgalactosaminyltransferases/metabolism , Neoplasm Metastasis , Sialyltransferases/metabolism
18.
Biochim Biophys Acta ; 1861(7): 606-16, 2016 Jul.
Article En | MEDLINE | ID: mdl-27102612

Cholesterol oxidation products frequently have a high biological activity. In the present study, we have used microelectrode recording of end plate currents and FM-based optical detection of synaptic vesicle exo-endocytosis to investigate the effects of two structurally similar oxysterols, olesoxime (cholest-4-en-3-one, oxime) and 5ɑ-cholestan-3-one (5ɑCh3), on neurotransmission at the frog neuromuscular junction. Olesoxime is an exogenous, potentially neuroprotective, substance and 5ɑCh3 is an intermediate product in cholesterol metabolism, which is elevated in the case of cerebrotendinous xanthomatosis. We found that olesoxime slightly increased evoked neurotransmitter release in response to a single stimulus and significantly reduced synaptic depression during high frequency activity. The last effect was due to an increase in both the number of synaptic vesicles involved in exo-endocytosis and the rate of synaptic vesicle recycling. In contrast, 5ɑCh3 reduced evoked neurotransmitter release during the low- and high frequency synaptic activities. The depressant action of 5ɑCh3 was associated with a reduction in the number of synaptic vesicles participating in exo- and endocytosis during high frequency stimulation, without a change in rate of the synaptic vesicle recycling. Of note, olesoxime increased the staining of synaptic membranes with the B-subunit of cholera toxin and the formation of fluorescent ganglioside GM1 clusters, and decreased the fluorescence of 22-NBD-cholesterol, while 5ɑCh3 had the opposite effects, suggesting that the two oxysterols have different effects on lipid raft stability. Taken together, these data show that these two structurally similar oxysterols induce marked different changes in neuromuscular transmission which are related with the alteration in synaptic vesicle cycle.


Cholestanes/pharmacology , Cholestenones/pharmacology , Excitatory Postsynaptic Potentials/drug effects , Muscle, Skeletal/drug effects , Neuromuscular Junction/drug effects , Synaptic Transmission/drug effects , Animals , Cholera Toxin/chemistry , Endocytosis/drug effects , Excitatory Postsynaptic Potentials/physiology , Exocytosis/drug effects , G(M1) Ganglioside/analogs & derivatives , G(M1) Ganglioside/chemistry , Membrane Microdomains/drug effects , Microelectrodes , Muscle, Skeletal/innervation , Muscle, Skeletal/physiology , Neuromuscular Junction/physiology , Rana ridibunda , Staining and Labeling , Synaptic Vesicles/drug effects , Tissue Culture Techniques
19.
J Reprod Dev ; 62(3): 249-55, 2016 Jun 17.
Article En | MEDLINE | ID: mdl-26860251

Gangliosides are key lipid molecules required for the regulation of cellular processes such as proliferation, differentiation, and cell signaling, including signaling of epidermal growth factor receptor (EGFR). Epidermal growth factor (EGF) has long been considered a potential regulator of meiotic and cytoplasmic maturation in mammalian oocytes. However, there is no report on the direct effect of ganglioside GD1a in porcine oocyte maturation. In this study, we first investigated a functional link between GD1a and meiotic maturation during in vitro maturation (IVM) of porcine embryos. Moreover, we confirmed the effect of exogenous GD1a treatment on blastocyst development, quality, and fertilization rate in early embryonic development. First, we observed that the protein level of ST3GAL2, a GD1a synthesizing enzyme, significantly increased (P < 0.01) in cumulus-oocyte-complexes (COCs) during IVM progress. The proportion of arrested germinal vesicles (GV) increased in oocytes treated with EGF+GD1a (41.6 ± 1.5%) at the IVM I stage. Upon completion of meiotic maturation, the proportion of metaphase II (M II) was significantly higher (P < 0.05) in the EGF+GD1a (89.9 ± 3.6%) treated group. After IVF, the percentage of penetrated oocytes was significantly higher (P < 0.05) in the EGF+GD1a (89.1 ± 2.3%) treated group than in the control group. Furthermore, exogenous GD1a treatment improved the developmental competence and quality of blastocysts during preimplantation embryo development stage. These results suggest that ganglioside GD1a may play an important role in IVM mechanisms of porcine maturation capacity. Furthermore, our findings will be helpful for better promoting the embryo development and blastocyst quality in pigs.


Blastocyst/cytology , G(M1) Ganglioside/analogs & derivatives , Oocytes/cytology , Animals , Apoptosis , Cell Nucleus/metabolism , Cells, Cultured , Cleavage Stage, Ovum , Cumulus Cells/cytology , Embryonic Development , Epidermal Growth Factor/metabolism , ErbB Receptors/metabolism , Female , Fertilization , G(M1) Ganglioside/physiology , Meiosis , Metaphase , Ovary/metabolism , Sialyltransferases/metabolism , Swine , beta-Galactoside alpha-2,3-Sialyltransferase
20.
Glycobiology ; 26(5): 532-9, 2016 May.
Article En | MEDLINE | ID: mdl-26715202

Mammalian cell surfaces are decorated with a variety of glycan chains that orchestrate development and defense and are exploited by pathogens for cellular attachment and entry. While glycosidic linkages are, in principle, flexible, the conformational space that a given glycan can sample is subject to spatial and electrostatic restrictions imposed by its overall chemical structure. Here, we show how the glycan moiety of the GM1 ganglioside, a branched, monosialylated pentasaccharide that serves as a ligand for various proteins, undergoes differential conformational selection in its interactions with different lectins. Using STD NMR and X-ray crystallography, we found that the innate immune regulator complement Factor H (FH) binds a previously not reported GM1 conformation that is not compatible with the GM1-binding sites of other structurally characterized GM1-binding lectins such as the Simian Virus 40 (SV40) capsid. Molecular dynamics simulations of the free glycan in explicit solvent on the 10 µs timescale reveal that the FH-bound conformation nevertheless corresponds to a minimum in the Gibbs free energy plot. In contrast to the GM1 conformation recognized by SV40, the FH-bound GM1 conformation is associated with poor NOE restraints, explaining how it escaped(1)H-(1)H NOE-restrained modeling in the past and highlighting the necessity for ensemble representations of glycan structures.


Capsid/chemistry , Complement Factor H/chemistry , G(M1) Ganglioside/analogs & derivatives , Molecular Dynamics Simulation , Simian virus 40/chemistry , Capsid/metabolism , Complement Factor H/metabolism , G(M1) Ganglioside/chemistry , G(M1) Ganglioside/metabolism , Humans , Simian virus 40/metabolism
...