Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 344
Filter
1.
Nat Commun ; 15(1): 3543, 2024 May 10.
Article in English | MEDLINE | ID: mdl-38730244

ABSTRACT

ß-N-Acetylgalactosamine-containing glycans play essential roles in several biological processes, including cell adhesion, signal transduction, and immune responses. ß-N-Acetylgalactosaminidases hydrolyze ß-N-acetylgalactosamine linkages of various glycoconjugates. However, their biological significance remains ambiguous, primarily because only one type of enzyme, exo-ß-N-acetylgalactosaminidases that specifically act on ß-N-acetylgalactosamine residues, has been documented to date. In this study, we identify four groups distributed among all three domains of life and characterize eight ß-N-acetylgalactosaminidases and ß-N-acetylhexosaminidase through sequence-based screening of deep-sea metagenomes and subsequent searching of public protein databases. Despite low sequence similarity, the crystal structures of these enzymes demonstrate that all enzymes share a prototype structure and have diversified their substrate specificities (oligosaccharide-releasing, oligosaccharide/monosaccharide-releasing, and monosaccharide-releasing) through the accumulation of mutations and insertional amino acid sequences. The diverse ß-N-acetylgalactosaminidases reported in this study could facilitate the comprehension of their structures and functions and present evolutionary pathways for expanding their substrate specificity.


Subject(s)
Acetylgalactosamine , Glycoside Hydrolases , Metagenome , Metagenome/genetics , Substrate Specificity , Acetylgalactosamine/metabolism , Acetylgalactosamine/chemistry , Glycoside Hydrolases/metabolism , Glycoside Hydrolases/genetics , Glycoside Hydrolases/chemistry , beta-N-Acetylhexosaminidases/metabolism , beta-N-Acetylhexosaminidases/genetics , beta-N-Acetylhexosaminidases/chemistry , Phylogeny , Crystallography, X-Ray , Amino Acid Sequence , Animals
2.
Mar Drugs ; 22(4)2024 Apr 19.
Article in English | MEDLINE | ID: mdl-38667801

ABSTRACT

Fucosylated chondroitin sulfate is a unique glycosaminoglycan isolated from sea cucumbers, with excellent anticoagulant activity. The fucosyl branch in FCS is generally located at the 3-OH of D-glucuronic acid but, recently, a novel structure with α-L-fucose linked to the 6-OH of N-acetyl-galactosamine has been found. Here, using functionalized monosaccharide building blocks, we prepared novel FCS tetrasaccharides with fucosyl branches both at the 6-OH of GalNAc and 3-OH of GlcA. In the synthesis, the protective group strategy of selective O-sulfation, as well as stereoselective glycosylation, was established, which enabled the efficient synthesis of the specific tetrasaccharide compounds. This research enriches knowledge on the structural types of FCS oligosaccharides and facilitates the exploration of the structure-activity relationship in the future.


Subject(s)
Chondroitin Sulfates , Oligosaccharides , Sea Cucumbers , Chondroitin Sulfates/chemistry , Chondroitin Sulfates/chemical synthesis , Chondroitin Sulfates/pharmacology , Animals , Oligosaccharides/chemical synthesis , Oligosaccharides/chemistry , Sea Cucumbers/chemistry , Glycosylation , Fucose/chemistry , Anticoagulants/pharmacology , Anticoagulants/chemistry , Anticoagulants/chemical synthesis , Structure-Activity Relationship , Acetylgalactosamine/chemistry , Acetylgalactosamine/analogs & derivatives
3.
Int J Biol Macromol ; 266(Pt 2): 131283, 2024 May.
Article in English | MEDLINE | ID: mdl-38561119

ABSTRACT

Glycosaminoglycan (GAG) lyases are important tools for investigating the structure of GAGs and preparing low-molecular-weight GAGs. The PL35 family, a recently established polysaccharide lyase family, should be further investigated. In this study, we discovered a new GAG lyase, CHa1, which belongs to the PL35 family. When expressed heterologously in Escherichia coli (BL21), CHa1 exhibited high expression levels and solubility. The optimal activity was observed in Tris-HCl buffer (pH 7.0) or sodium phosphate buffer (pH 8.0) at 30 °C. The specific activities towards HA, CSA, CSC, CSD, CSE, and HS were 3.81, 13.03, 36.47, 18.46, 6.46, and 0.50 U/mg protein, respectively. CHa1 digests substrate chains randomly that acting as an endolytic lyase and shows a significant preference for GlcA-containing structures, prefers larger oligosaccharides (≥UDP8) and can generate a series of oligosaccharides composed mainly of the A unit when digesting CSA. These oligosaccharides include ΔC-A, ΔC-A-A, ΔC-A-A-A, ΔC-A-A-A-A, and ΔC-A-A-A-A-A. The residues Tyr257 and His421 play crucial roles in the catalytic process, and Ser211, Asn212, Asn213, Trp214, Gln216, Lys360, Arg460 and Gln462 may participate in the binding process of CHa1. This study on CHa1 contributes to our understanding of the PL35 family and provides valuable tools for investigating the structure of GAGs.


Subject(s)
Polysaccharide-Lyases , Polysaccharide-Lyases/chemistry , Polysaccharide-Lyases/metabolism , Polysaccharide-Lyases/genetics , Substrate Specificity , Acetylgalactosamine/chemistry , Acetylgalactosamine/metabolism , Escherichia coli/genetics , Glycosaminoglycans/metabolism , Glycosaminoglycans/chemistry , Amino Acid Sequence , Oligosaccharides/chemistry , Oligosaccharides/metabolism
4.
Eur J Med Chem ; 269: 116278, 2024 Apr 05.
Article in English | MEDLINE | ID: mdl-38479165

ABSTRACT

Asialoglycoprotein receptor (ASGPR) specifically recognizes glycans terminated with ß-d-galactose or N-acetylgalactosamine. Its exclusive expression in mammalian hepatocytes renders it an ideal hepatic-targeted biomarker. To date, ASGPR-targeted ligands have been actively developed for drug delivery and hepatic imaging. This review provides a comprehensive summary of the progress achieved to-date in the field of developing ASGPR-targeted nuclear medicine imaging (NMI) radiotracers, highlighting the recent advancements over the last decade in terms of structure, radionuclides and labeling strategies. The biodistribution patterns, imaging characteristics, challenges and future prospective are discussed.


Subject(s)
Nuclear Medicine , Animals , Asialoglycoprotein Receptor/chemistry , Asialoglycoprotein Receptor/metabolism , Hepatocytes/metabolism , Liver/diagnostic imaging , Liver/metabolism , Mammals/metabolism , Tissue Distribution , Acetylgalactosamine/chemistry , Acetylgalactosamine/metabolism
5.
Int J Biol Macromol ; 263(Pt 2): 130347, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38401583

ABSTRACT

Polypeptide N-acetylgalactosamine transferase 9 (GALNT9) catalyzes the initial step of mucin-type O-glycosylation via linking N-acetylgalactosamine (GalNAc) to serine/threonine in a protein. To unravel the association of GALNT9 with Parkinson's disease (PD), a progressive neurodegenerative disorder, GALNT9 levels were evaluated in the patients with PD and mice treated with 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine, and statistically analyzed based on the GEO datasets of GSE114918 and GSE216281. Glycoproteins with exposing GalNAc were purified using lectin affinity chromatography and identified by LC-MS/MS. The influence of GALNT9 on cells was evaluated via introducing a GALNT9-specific siRNA into SH-SY5Y cells. Consequently, GALNT9 deficiency was found to occur under PD conditions. GALNT9 silencing contributed to a causative factor in PD pathogenesis via reducing the levels of intracellular dopamine, tyrosine hydroxylase and soluble α-synuclein, and promoting α-synuclein aggregates. MS identification revealed 14 glycoproteins. 5 glycoproteins, including ACO2, ATP5B, CKB, CKMT1A, ALDOC, were associated with energy metabolism. GALNT9 silencing resulted in mitochondrial dysfunctions via increasing ROS accumulation, mitochondrial membrane depolarization, mPTPs opening, Ca2+ releasing and activation of the CytC-related apoptotic pathway. The dysfunctional mitochondria then triggered mitophagy, possibly intermediated by adenine nucleotide translocase 1. Our study suggests that GALNT9 is potentially developed into an auxiliary diagnostic index and therapeutic target of PD.


Subject(s)
Mitochondrial Diseases , N-Acetylgalactosaminyltransferases , Neuroblastoma , Parkinson Disease , Humans , Mice , Animals , Parkinson Disease/metabolism , alpha-Synuclein/chemistry , Acetylgalactosamine/chemistry , Transferases , Chromatography, Liquid , Tandem Mass Spectrometry , Peptides , Glycoproteins , N-Acetylgalactosaminyltransferases/genetics , N-Acetylgalactosaminyltransferases/metabolism , Creatine Kinase
6.
Mol Ther ; 32(3): 637-645, 2024 Mar 06.
Article in English | MEDLINE | ID: mdl-38204163

ABSTRACT

N-Acetylgalactosamine (GalNAc)-conjugated small interfering RNA (siRNA) therapies have received approval for treating both orphan and prevalent diseases. To improve in vivo efficacy and streamline the chemical synthesis process for efficient and cost-effective manufacturing, we conducted this study to identify better designs of GalNAc-siRNA conjugates for therapeutic development. Here, we present data on redesigned GalNAc-based ligands conjugated with siRNAs against angiopoietin-like 3 (ANGPTL3) and lipoprotein (a) (Lp(a)), two target molecules with the potential to address large unmet medical needs in atherosclerotic cardiovascular diseases. By attaching a novel pyran-derived scaffold to serial monovalent GalNAc units before solid-phase oligonucleotide synthesis, we achieved increased GalNAc-siRNA production efficiency with fewer synthesis steps compared to the standard triantennary GalNAc construct L96. The improved GalNAc-siRNA conjugates demonstrated equivalent or superior in vivo efficacy compared to triantennary GalNAc-conjugated siRNAs.


Subject(s)
Cardiovascular Diseases , Hepatocytes , Humans , RNA, Small Interfering/genetics , RNA, Small Interfering/chemistry , Cost-Benefit Analysis , RNA, Double-Stranded , Acetylgalactosamine/chemistry , Angiopoietin-Like Protein 3
7.
Clin Pharmacokinet ; 62(12): 1661-1672, 2023 12.
Article in English | MEDLINE | ID: mdl-37824025

ABSTRACT

Small-interfering ribonucleic acids (siRNAs) with N-acetylgalactosamine (GalNAc) conjugation for improved liver uptake represent an emerging class of drugs that modulate liver-expressed therapeutic targets. The pharmacokinetics of GalNAc-siRNAs are characterized by a rapid distribution from plasma to tissue (hours) and a long terminal plasma half-life, analyzed in the form of the antisense strand, driven by redistribution from tissue (weeks). Understanding how clinical pharmacokinetics relate to the dose and type of siRNA chemical stabilizing method used is critical, e.g., to design studies, to investigate safety windows, and to predict the pharmacokinetics of new preclinical assets. To this end, we collected and analyzed pharmacokinetic data from the literature regarding nine GalNAc-siRNAs. Based on this analysis, we showed that the clinical plasma pharmacokinetics of GalNAc-siRNAs are approximately dose proportional and similar between chemical stabilizing methods. This holds for both the area under the concentration-time curve (AUC) and the maximum plasma concentration (Cmax). Corresponding rat and monkey pharmacokinetic data for a subset of the nine GalNAc-siRNAs show dose-proportional Cmax, supra-dose-proportional AUC, and similar pharmacokinetics between chemical stabilizing methods​. Together, the animal and human pharmacokinetic data indicate that plasma clearance divided by bioavailability follows allometric principles and scales between species with an exponent of 0.75. Finally, the clinical plasma concentration-time profiles can be empirically described by standard one-compartment kinetics with first-order absorption up to 24 h after subcutaneous dosing, and by three-compartment kinetics with first-order absorption in general. To describe the system more mechanistically, we report a corrected and unambiguously defined version of a previously published physiologically based pharmacokinetic model.


Subject(s)
Acetylgalactosamine , Liver , Humans , Rats , Animals , Acetylgalactosamine/chemistry , Acetylgalactosamine/metabolism , Liver/metabolism , RNA, Small Interfering/chemistry , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Biological Availability
8.
J Med Chem ; 66(4): 2506-2523, 2023 02 23.
Article in English | MEDLINE | ID: mdl-36757090

ABSTRACT

Conjugation of synthetic triantennary N-acetyl-d-galactosamine (GalNAc) to small interfering RNA (siRNA) mediates binding to the asialoglycoprotein receptor (ASGPR) on the surface of hepatocytes, facilitating liver-specific uptake and siRNA-mediated gene silencing. The natural ß-glycosidic bond of the GalNAc ligand is rapidly cleaved by glycosidases in vivo. Novel GalNAc ligands with S-, and C-glycosides with both α- and ß-anomeric linkages, N-glycosides with ß-anomeric linkage, and the O-glycoside with α-anomeric linkage were synthesized and conjugated to siRNA either on-column during siRNA synthesis or through a high-throughput, post-synthetic method. Unlike natural GalNAc, modified ligands were resistant to glycosidase activity. The siRNAs conjugated to newly designed ligands had similar affinities for ASGPR and similar silencing activity in mice as the parent GalNAc-siRNA conjugate. These data suggest that other factors, such as protein-nucleic acid interactions and loading of the antisense strand into the RNA-induced silencing complex (RISC), are more critical to the duration of action than the stereochemistry and stability of the anomeric linkage between the GalNAc moiety of the ligand conjugated to the sense strand of the siRNA.


Subject(s)
Asialoglycoprotein Receptor , Galactosamine , RNA, Small Interfering , RNA-Induced Silencing Complex , Animals , Mice , Acetylgalactosamine/chemistry , Asialoglycoprotein Receptor/metabolism , Glycoside Hydrolases/metabolism , Glycosides/metabolism , Hepatocytes/metabolism , Ligands , RNA, Small Interfering/metabolism , RNA-Induced Silencing Complex/metabolism
9.
Molecules ; 27(12)2022 Jun 20.
Article in English | MEDLINE | ID: mdl-35745067

ABSTRACT

The development of oligonucleotide conjugates for in vivo targeting is one of the most exciting areas for oligonucleotide therapeutics. A major breakthrough in this field was the development of multifunctional GalNAc-oligonucleotides with high affinity to asialoglycoprotein receptors (ASGPR) that directed therapeutic oligonucleotides to hepatocytes. In the present study, we explore the use of G-rich sequences functionalized with one unit of GalNAc at the 3'-end for the formation of tetrameric GalNAc nanostructures upon formation of a parallel G-quadruplex. These compounds are expected to facilitate the synthetic protocols by providing the multifunctionality needed for the binding to ASGPR. To this end, several G-rich oligonucleotides carrying a TGGGGGGT sequence at the 3'-end functionalized with one molecule of N-acetylgalactosamine (GalNAc) were synthesized together with appropriate control sequences. The formation of a self-assembled parallel G-quadruplex was confirmed through various biophysical techniques such as circular dichroism, nuclear magnetic resonance, polyacrylamide electrophoresis and denaturation curves. Binding experiments to ASGPR show that the size and the relative position of the therapeutic cargo are critical for the binding of these nanostructures. The biological properties of the resulting parallel G-quadruplex were evaluated demonstrating the absence of the toxicity in cell lines. The internalization preferences of GalNAc-quadruplexes to hepatic cells were also demonstrated as well as the enhancement of the luciferase inhibition using the luciferase assay in HepG2 cell lines versus HeLa cells. All together, we demonstrate that tetramerization of G-rich oligonucleotide is a novel and simple route to obtain the beneficial effects of multivalent N-acetylgalactosamine functionalization.


Subject(s)
Acetylgalactosamine , G-Quadruplexes , Acetylgalactosamine/chemistry , Asialoglycoprotein Receptor/metabolism , HeLa Cells , Hepatocytes , Humans , Oligonucleotides/metabolism
10.
J Biol Chem ; 298(3): 101720, 2022 03.
Article in English | MEDLINE | ID: mdl-35151686

ABSTRACT

Glycosylphosphatidylinositol (GPI) is a posttranslational glycolipid modification of proteins that anchors proteins in lipid rafts on the cell surface. Although some GPI-anchored proteins (GPI-APs), including the prion protein PrPC, have a glycan side chain composed of N-acetylgalactosamine (GalNAc)-galactose-sialic acid on the core structure of GPI glycolipid, in vivo functions of this GPI-GalNAc side chain are largely unresolved. Here, we investigated the physiological and pathological roles of the GPI-GalNAc side chain in vivo by knocking out its initiation enzyme, PGAP4, in mice. We show that Pgap4 mRNA is highly expressed in the brain, particularly in neurons, and mass spectrometry analysis confirmed the loss of the GalNAc side chain in PrPC GPI in PGAP4-KO mouse brains. Furthermore, PGAP4-KO mice exhibited various phenotypes, including an elevated blood alkaline phosphatase level, impaired bone formation, decreased locomotor activity, and impaired memory, despite normal expression levels and lipid raft association of various GPI-APs. Thus, we conclude that the GPI-GalNAc side chain is required for in vivo functions of GPI-APs in mammals, especially in bone and the brain. Moreover, PGAP4-KO mice were more vulnerable to prion diseases and died earlier after intracerebral inoculation of the pathogenic prion strains than wildtype mice, highlighting the protective roles of the GalNAc side chain against prion diseases.


Subject(s)
Acetylgalactosamine , Glycosylphosphatidylinositols , Prion Diseases , Prions , Acetylgalactosamine/chemistry , Acetylgalactosamine/metabolism , Animals , Brain/metabolism , Glycosylphosphatidylinositols/chemistry , Glycosylphosphatidylinositols/metabolism , Mice , Osteogenesis , Prion Diseases/metabolism , Prions/metabolism , Structure-Activity Relationship
11.
Nature ; 598(7880): 332-337, 2021 10.
Article in English | MEDLINE | ID: mdl-34616040

ABSTRACT

Humans have co-evolved with a dense community of microbial symbionts that inhabit the lower intestine. In the colon, secreted mucus creates a barrier that separates these microorganisms from the intestinal epithelium1. Some gut bacteria are able to utilize mucin glycoproteins, the main mucus component, as a nutrient source. However, it remains unclear which bacterial enzymes initiate degradation of the complex O-glycans found in mucins. In the distal colon, these glycans are heavily sulfated, but specific sulfatases that are active on colonic mucins have not been identified. Here we show that sulfatases are essential to the utilization of distal colonic mucin O-glycans by the human gut symbiont Bacteroides thetaiotaomicron. We characterized the activity of 12 different sulfatases produced by this species, showing that they are collectively active on all known sulfate linkages in O-glycans. Crystal structures of three enzymes provide mechanistic insight into the molecular basis of substrate specificity. Unexpectedly, we found that a single sulfatase is essential for utilization of sulfated O-glycans in vitro and also has a major role in vivo. Our results provide insight into the mechanisms of mucin degradation by a prominent group of gut bacteria, an important process for both normal microbial gut colonization2 and diseases such as inflammatory bowel disease3.


Subject(s)
Bacteroides/enzymology , Colon/metabolism , Colon/microbiology , Gastrointestinal Microbiome , Mucins/metabolism , Sulfatases/metabolism , Acetylgalactosamine/chemistry , Acetylgalactosamine/metabolism , Animals , Colon/chemistry , Crystallography, X-Ray , Female , Galactose/metabolism , Humans , Male , Mice , Models, Molecular , Substrate Specificity , Sulfatases/chemistry
12.
Arterioscler Thromb Vasc Biol ; 41(12): 2855-2865, 2021 12.
Article in English | MEDLINE | ID: mdl-34645280

ABSTRACT

While the promise of oligonucleotide therapeutics, such as (chemically modified) ASO (antisense oligonucleotides) and short interfering RNAs, is undisputed from their introduction onwards, their unfavorable pharmacokinetics and intrinsic capacity to mobilize innate immune responses, were limiting widespread clinical use. However, these major setbacks have been tackled by breakthroughs in chemistry, stability and delivery. When aiming an intervention hepatic targets, such as lipid and sugar metabolism, coagulation, not to mention cancer and virus infection, introduction of N-acetylgalactosamine aided targeting technology has advanced the field profoundly and by now a dozen of N-acetylgalactosamine therapeutics for these indications have been approved for clinical use or have progressed to clinical trial stage 2 to 3 testing. This technology, in combination with major advances in oligonucleotide stability allows safe and durable intervention in targets that were previously deemed undruggable, such as Lp(a) and PCSK9 (proprotein convertase subtilisin/kexin type 9), at high efficacy and specificity, often with as little as 2 doses per year. Their successful use even the most visionary would not have predicted 2 decades ago. Here, we will review the evolution of N-acetylgalactosamine technology. We shall outline their fundamental design principles and merits, and their application for the delivery of oligonucleotide therapeutics to the liver. Finally, we will discuss the perspectives of N-acetylgalactosamine technology and propose directions for future research in receptor targeted delivery of these gene medicines.


Subject(s)
Acetylgalactosamine/chemistry , Cardiovascular Diseases/drug therapy , Drug Delivery Systems , Genetic Therapy/methods , Liver/drug effects , Oligonucleotides/administration & dosage , Hepatocytes/drug effects , Hepatocytes/metabolism , Humans , Liver/metabolism , RNAi Therapeutics
13.
Anal Bioanal Chem ; 413(29): 7229-7240, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34327564

ABSTRACT

Negative ion collision-induced dissociation (CID) of underivatized N-glycans has proved to be a simple, yet powerful method for their structural determination. Recently, we have identified a series of such structures with GalNAc rather than the more common galactose capping the antennae of hybrid and complex glycans. As part of a series of publications describing the negative ion fragmentation of different types of N-glycan, this paper describes their CID spectra and estimated nitrogen cross sections recorded by travelling wave ion mobility mass spectrometry (TWIMS). Most of the glycans were derived from the recombinant glycoproteins gp120 and gp41 from the human immunodeficiency virus (HIV), recombinantly derived from human embryonic kidney (HEK 293T) cells. Twenty-six GalNAc-capped hybrid and complex N-glycans were identified by a combination of TWIMS, negative ion CID, and exoglycosidase digestions. They were present as the neutral glycans and their sulfated and α2→3-linked sialylated analogues. Overall, negative ion fragmentation of glycans generates fingerprints that reveal their structural identity.


Subject(s)
Glycoproteins/chemistry , Ion Mobility Spectrometry/methods , Polysaccharides/analysis , Polysaccharides/chemistry , Acetylgalactosamine/chemistry , Glycoproteins/genetics , HEK293 Cells , HIV Envelope Protein gp120/chemistry , HIV Envelope Protein gp120/genetics , HIV Envelope Protein gp41/chemistry , HIV Envelope Protein gp41/genetics , Humans , Nitrogen/chemistry , Protein Multimerization , Recombinant Proteins/genetics , Spectrometry, Mass, Electrospray Ionization
14.
Eur J Med Chem ; 222: 113605, 2021 Oct 15.
Article in English | MEDLINE | ID: mdl-34126457

ABSTRACT

In this study, we designed and developed a novel asialoglycoprotein receptor (ASGPR)-targeted PEGylated paclitaxel (PTX) nanoliposome for hepatocellular carcinoma (HCC). N-acetylgalactosamine with α configuration (Tn) was synthesized and used as the active targeting ligand. Notably, Tn modified nanoliposomes loaded with PTX (Tn-Lipo-PTX) showed a narrow distribution (PDI = 0.18-0.20) with 74 ± 0.36 nm of average sizes. Tn-Lipo-PTX has a high encapsulation efficiency of more than 93.0% and 13% of drug loading (DL). Compared with no targeted Con-Lipo-PTX, Tn-Lipo-PTX showed lower and sustained release characteristic in PBS in vitro. Tn targeting ASGPR was confirmed by HepG-2 cells uptake experiment by fluorescence microscopy analysis. Tn-Lipo-PTX accumulated in HepG-2 cells and this process was inhibited by adding Tn ligand, supporting receptor-mediated endocytosis mechanism. MTT assays was implemented in four cell lines. Tn-Lipo-PTX exhibited superior inhibition against ASGPR on over-expressing HepG-2 (IC50 = 1.93 nM). The cell cycle experiments showed that Tn-Lipo-PTX could efficiently increase the percentage of cells arrest in the G2/M phase. Through western blotting analysis, the ß-tubulin and cyclin B1 expression in the Tn-Lipo-PTX group were significantly higher compared with other groups and the CDK1 was down-regulated compared with PTX group, which indicated that targeting liposome delivery system could not only change periodic proteins expression, but also improve the killing effect of PTX on hepatocarcinoma cell. Tn-installed PEGylated nanoliposomes have a great potential for targeted cancer chemotherapy.


Subject(s)
Acetylgalactosamine/chemistry , Antineoplastic Agents, Phytogenic/pharmacology , Carcinoma, Hepatocellular/drug therapy , Drug Delivery Systems , Liver Neoplasms/drug therapy , Nanoparticles/chemistry , Paclitaxel/pharmacology , Antineoplastic Agents, Phytogenic/chemistry , Carcinoma, Hepatocellular/pathology , Cell Cycle/drug effects , Cell Proliferation/drug effects , Cell Survival/drug effects , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Hep G2 Cells , Humans , Liposomes/chemistry , Liver Neoplasms/pathology , Molecular Structure , Paclitaxel/chemistry , Structure-Activity Relationship
15.
Mol Ther ; 29(10): 2910-2919, 2021 10 06.
Article in English | MEDLINE | ID: mdl-34091052

ABSTRACT

N-Acetylgalactosamine (GalNAc) conjugated short interfering RNAs (siRNAs) are a leading RNA interference (RNAi) platform allowing targeted inhibition of disease-causing genes in hepatocytes. More than a decade of development has recently resulted in the first approvals for this class of drugs. While substantial effort has been made to improve nucleic acid modification patterns for better payload stability and efficacy, relatively little attention has been given to the GalNAc targeting ligand. In addition, the lack of an intrinsic endosomal release mechanism has limited potency. Here, we report a stepwise analysis of the structure activity relationships (SAR) of the components comprising these targeting ligands. We show that there is relatively little difference in biological performance between bi-, tri-, and tetravalent ligand structures while identifying other features that affect their biological activity more significantly. Further, we demonstrate that subcutaneous co-administration of a GalNAc-functionalized, pH responsive endosomal release agent markedly improved the activity and duration of effect for siRNA conjugates, without compromising tolerability, in non-human primates. These findings could address a significant bottleneck for future siRNA ligand conjugate development.


Subject(s)
Acetylgalactosamine/chemistry , Asialoglycoprotein Receptor/metabolism , RNA, Small Interfering/administration & dosage , Animals , Female , Hep G2 Cells , Humans , Injections, Subcutaneous , Ligands , Liposomes , Male , Mice , Nanoparticles , Primates , RNA, Small Interfering/chemistry , Structure-Activity Relationship
16.
J Med Chem ; 64(10): 6838-6855, 2021 05 27.
Article in English | MEDLINE | ID: mdl-33950677

ABSTRACT

A morpholine-based nucleotide analog was developed as a building block for hepatic siRNA targeting and stabilization. Attachment of an asialoglycoprotein-binding GalNAc ligand at the morpholine nitrogen was realized with different linkers. The obtained morpholino GalNAc scaffolds were coupled to the sense strand of a transthyretin-targeting siRNA and tested for their knockdown potency in vitro and in vivo. A clear structure-activity relationship was developed with regard to the linker type and length as well as the attachment site of the morpholino GalNAc moieties at the siRNA sense strand. Further, simple alkylation of the morpholine nitrogen led to a nucleotide analog, which increased siRNA stability, when used as a double 3'-overhang at the sense strand sequence. Combination of the best morpholino GalNAc building blocks as targeting nucleotides with an optimized stabilizing alkyl-substituted morpholine as 3'-overhangs resulted in siRNAs without any phosphorothioate stabilization in the sense strand and clearly improved the duration of action in vivo.


Subject(s)
Morpholines/chemistry , Nucleotides/chemistry , RNA, Small Interfering/metabolism , Acetylgalactosamine/chemistry , Animals , Cells, Cultured , Female , Hepatocytes/cytology , Hepatocytes/metabolism , Ligands , Mice , Mice, Inbred C57BL , Nucleotides/chemical synthesis , Nucleotides/metabolism , Prealbumin/antagonists & inhibitors , Prealbumin/genetics , Prealbumin/metabolism , RNA Interference , RNA Stability , RNA, Small Interfering/chemistry
17.
Int J Biol Macromol ; 181: 928-936, 2021 Jun 30.
Article in English | MEDLINE | ID: mdl-33878355

ABSTRACT

AGL, a 15-kDa lectin from Amaranthus gangeticus seeds was isolated using ion-exchange and gel filtration chromatography. AGL contained 8.55% of neutral sugar and became specifically inhibited by N-acetyl-D-galactosamine. Hemagglutination activity of the lectin was maximum over the pH range of 4.0-6.0 and temperatures of 30-60 °C though it lost the activity when treated with urea and EDTA. With an LC50 value of 250 µg/ml, AGL showed mild toxicity against Artemia nauplii. It inhibited the growth of pathogenic bacteria like Shigella boydii, Shigella dysenteriae and Staphylococcus aureus when treated for 8 and 16 h, respectively, but lost the antibacterial activity during a 24 h treatment. AGL could not inhibit the growth of Escherichia coli and mitogenic growth (7.0-9.0%) was observed instead. AGL inhibited 37.14%, 65.71% and 82.85% of biofilm formation of Escherichia coli at the concentrations of 250, 500 and 1000 µg/ml, respectively. Marked inhibition of the proliferation of Ehrlich ascites carcinoma cells was determined when treated with various doses of AGL. AGL inhibited 65.89% and 81.25% of the in vivo growth of EAC cells in mice at the doses of 2.0 and 4.0 mg/kg/day, respectively. Significant alteration of the expression of apoptosis related genes Fas, NF-kB and MAPK were observed.


Subject(s)
Amaranthus/chemistry , Biofilms/drug effects , Carcinoma, Ehrlich Tumor/drug therapy , Lectins/pharmacology , Acetylgalactosamine/antagonists & inhibitors , Acetylgalactosamine/chemistry , Animals , Apoptosis , Ascites/drug therapy , Ascites/genetics , Ascites/pathology , Biofilms/growth & development , Carcinoma, Ehrlich Tumor/genetics , Carcinoma, Ehrlich Tumor/pathology , Cell Line, Tumor , Cell Proliferation , Humans , Lectins/chemistry , Mice , NF-kappa B/genetics , Plant Lectins/chemistry , Seeds/chemistry
18.
Cancer Sci ; 112(6): 2481-2492, 2021 Jun.
Article in English | MEDLINE | ID: mdl-33792132

ABSTRACT

Hepatocellular carcinoma (HCC) is one of the most lethal cancers in humans. The inhibition of peptidyl-prolyl cis/trans isomerase (Pin1) gene expression may have great potential in the treatment of HCC. N-Acetylgalactosamine (GalNAc) was used to target the liver. Cholesterol-modified antimicrobial peptide DP7 (DP7-C) acts as a carrier, the GalNAc-siRNA/DP7-C complex increases the uptake of GalNAc-siRNA and the escape of endosomes in hepatocytes. In addition, DP7-C nanoparticles and hydrogel-assisted GalNAc-Pin1 siRNA delivery can effectively enhance the stability and prolong the silencing effects of Pin1 siRNA. In an orthotopic liver cancer model, the GalNAc-Pin1 siRNA/DP7-C/hydrogel complex can potentially regulate Pin1 expression in hepatocellular carcinoma cells and effectively inhibit tumor progression. Our study proves that Pin1 siRNA is an efficient method for the treatment of HCC and provides a sustainable and effective drug delivery system for the suppression of liver cancer.


Subject(s)
Acetylgalactosamine/chemistry , Carcinoma, Hepatocellular/drug therapy , Liver Neoplasms/drug therapy , NIMA-Interacting Peptidylprolyl Isomerase/antagonists & inhibitors , RNA, Small Interfering/administration & dosage , Animals , Carcinoma, Hepatocellular/genetics , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Delayed-Action Preparations , Drug Compounding , Female , Gene Expression Regulation, Neoplastic/drug effects , Humans , Hydrogels/chemistry , Injections, Subcutaneous , Liver Neoplasms/genetics , Mice , NIMA-Interacting Peptidylprolyl Isomerase/genetics , Pore Forming Cytotoxic Proteins/chemistry , RNA, Small Interfering/chemistry , RNA, Small Interfering/pharmacology , Thermodynamics , Xenograft Model Antitumor Assays
19.
Bioconjug Chem ; 32(4): 763-781, 2021 04 21.
Article in English | MEDLINE | ID: mdl-33691403

ABSTRACT

Herein, we describe the design, synthesis, and biological evaluation of novel betulin and N-acetyl-d-galactosamine (GalNAc) glycoconjugates and suggest them as targeted agents against hepatocellular carcinoma. We prepared six conjugates derived via the C-3 and C-28 positions of betulin with one or two saccharide ligands. These molecules demonstrate high affinity to the asialoglycoprotein receptor (ASGPR) of hepatocytes assessed by in silico modeling and surface plasmon resonance tests. Cytotoxicity studies in vitro revealed a bivalent conjugate with moderate activity, selectivity of action, and cytostatic properties against hepatocellular carcinoma cells HepG2. An additional investigation confirmed the specific engagement with HepG2 cells by the enhanced generation of reactive oxygen species. Stability tests demonstrated its lability to acidic media and to intracellular enzymes. Therefore, the selected bivalent conjugate represents a new potential agent targeted against hepatocellular carcinoma. Further extensive studies of the cellular uptake in vitro and the real-time microdistribution in the murine liver in vivo for fluorescent dye-labeled analogue showed its selective internalization into hepatocytes due to the presence of GalNAc ligand in comparison with reference compounds. The betulin and GalNAc glycoconjugates can therefore be considered as a new strategy for developing therapeutic agents based on natural triterpenoids.


Subject(s)
Acetylgalactosamine/chemistry , Antineoplastic Agents/pharmacology , Asialoglycoprotein Receptor/drug effects , Carcinoma, Hepatocellular/pathology , Liver Neoplasms/pathology , Triterpenes/chemistry , Antineoplastic Agents/chemistry , Cell Line, Tumor , Drug Delivery Systems , Drug Discovery , Drug Screening Assays, Antitumor , Fluorescent Dyes/chemistry , Humans , Hydrophobic and Hydrophilic Interactions , Molecular Docking Simulation , Surface Plasmon Resonance
20.
J Biol Chem ; 296: 100299, 2021.
Article in English | MEDLINE | ID: mdl-33460651

ABSTRACT

The human Gb3/CD77 synthase, encoded by the A4GALT gene, is an unusually promiscuous glycosyltransferase. It synthesizes the Galα1→4Gal linkage on two different glycosphingolipids (GSLs), producing globotriaosylceramide (Gb3, CD77, Pk) and the P1 antigen. Gb3 is the major receptor for Shiga toxins (Stxs) produced by enterohemorrhagic Escherichia coli. A single amino acid substitution (p.Q211E) ramps up the enzyme's promiscuity, rendering it able to attach Gal both to another Gal residue and to GalNAc, giving rise to NOR1 and NOR2 GSLs. Human Gb3/CD77 synthase was long believed to transfer Gal only to GSL acceptors, therefore its GSL products were, by default, considered the only human Stx receptors. Here, using soluble, recombinant human Gb3/CD77 synthase and p.Q211E mutein, we demonstrate that both enzymes can synthesize the P1 glycotope (terminal Galα1→4Galß1→4GlcNAc-R) on a complex type N-glycan and a synthetic N-glycoprotein (saposin D). Moreover, by transfection of CHO-Lec2 cells with vectors encoding human Gb3/CD77 synthase and its p.Q211E mutein, we demonstrate that both enzymes produce P1 glycotopes on N-glycoproteins, with the mutein exhibiting elevated activity. These P1-terminated N-glycoproteins are recognized by Stx1 but not Stx2 B subunits. Finally, cytotoxicity assays show that Stx1 can use P1 N-glycoproteins produced in CHO-Lec2 cells as functional receptors. We conclude that Stx1 can recognize and use P1 N-glycoproteins in addition to its canonical GSL receptors to enter and kill the cells, while Stx2 can use GSLs only. Collectively, these results may have important implications for our understanding of the Shiga toxin pathology.


Subject(s)
Galactosyltransferases/chemistry , Globosides/chemistry , Shiga Toxin 1/chemistry , Trihexosylceramides/chemistry , Acetylgalactosamine/chemistry , Acetylgalactosamine/metabolism , Acetylglucosamine/chemistry , Acetylglucosamine/metabolism , Animals , Binding Sites , CHO Cells , Carbohydrate Sequence , Cricetulus , Enterohemorrhagic Escherichia coli/chemistry , Enterohemorrhagic Escherichia coli/pathogenicity , Galactose/chemistry , Galactose/metabolism , Galactosyltransferases/genetics , Galactosyltransferases/metabolism , Gene Expression , Globosides/biosynthesis , Globosides/metabolism , Glucose/chemistry , Glucose/metabolism , Humans , Models, Molecular , Mutation , Protein Binding , Protein Conformation, alpha-Helical , Protein Conformation, beta-Strand , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Shiga Toxin 1/metabolism , Shiga Toxin 2/chemistry , Shiga Toxin 2/metabolism , Trihexosylceramides/biosynthesis
SELECTION OF CITATIONS
SEARCH DETAIL
...