Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 2.011
Filter
1.
Proc Inst Mech Eng H ; 238(7): 741-754, 2024 Jul.
Article in English | MEDLINE | ID: mdl-39109566

ABSTRACT

The objectives of this study were to compare the chondrogenic potential of cells derived from different layers of Mandibular condyle cartilage and to gain further understanding of the impact of chondrogenic cues when embedded into a novel hydrogel scaffold (PGH, a polymer blend of poly (ethylene glycol), gelatin, and heparin) compared to a gelatin hydrogel scaffold (GEL). Cartilage layer cells (CLCs) and fibroblastic superficial layer cells (SLCs) were harvested from the mandibular condyle of boer goats obtained from a local abattoir. After expansion, cells were seeded into PGH and GEL hydrogels and cultured in chondrogenic media for 3 weeks. Scaffolds were harvested at 0, 1, and 3 week(s) and processed for gross appearance, histochemical, biochemical, and mechanical assays. In terms of chondrogenesis, major differences were observed between scaffold materials, but not cell types. Glycosaminoglycan (GAG) staining showed GEL scaffolds deposited GAG during the 3 week period, which was also confirmed with the biochemical testing. Moreover, GEL scaffolds had significantly higher compressive modulus and peak stress than PGH scaffolds at all time points with the largest difference seen in week 3. It can be concluded that GEL outperformed PGH in chondrogenesis. It can also be concluded that materials play a more important role in the process of chondrogenesis than the tested cell populations. Fibroblastic SLCs were shown to have similar chondrogenic potential as CLCs cells, suggesting a rich pool of progenitor cells in the superficial fibroblastic layer capable of undergoing chondrogenesis given appropriate physical and chemical cues.


Subject(s)
Cartilage , Chondrogenesis , Gelatin , Goats , Hydrogels , Temporomandibular Joint , Hydrogels/chemistry , Gelatin/chemistry , Animals , Cartilage/cytology , Temporomandibular Joint/cytology , Tissue Scaffolds/chemistry , Polymerization , Mandibular Condyle/cytology , Mandibular Condyle/physiology , Photochemical Processes , Glycosaminoglycans/chemistry , Glycosaminoglycans/metabolism
2.
ACS Sens ; 9(7): 3794-3804, 2024 Jul 26.
Article in English | MEDLINE | ID: mdl-38976969

ABSTRACT

Articular cartilage is a complex tissue, and early detection of osteoarthritis (OA) is crucial for effective treatment. However, current imaging modalities lack molecular specificity and primarily detect late-stage changes. In this study, we propose the use of spatially offset Raman spectroscopy (SORS) for noninvasive, depth-dependent, and molecular-specific diagnostics of articular cartilage. We demonstrate the potential of SORS to penetrate deep layers of cartilage, providing a comprehensive understanding of disease progression. Our SORS measurements were characterized and validated through mechanical and histological techniques, revealing strong correlations between spectroscopic measurements and both Young's modulus and depth of cartilage damage. By longitudinally monitoring enzymatically degraded condyles, we further developed a depth-dependent damage-tracking method. Our analysis revealed distinct components related to sample depth and glycosaminoglycan (GAG) changes, offering a comprehensive picture of cartilage health. Collectively, these findings highlight the potential of SORS as a valuable tool for enhancing OA management and improving patient outcomes.


Subject(s)
Cartilage, Articular , Osteoarthritis , Spectrum Analysis, Raman , Spectrum Analysis, Raman/methods , Cartilage, Articular/diagnostic imaging , Cartilage, Articular/pathology , Osteoarthritis/diagnosis , Animals , Glycosaminoglycans/analysis , Glycosaminoglycans/chemistry , Humans , Cattle
3.
ACS Appl Mater Interfaces ; 16(31): 40455-40468, 2024 Aug 07.
Article in English | MEDLINE | ID: mdl-39072446

ABSTRACT

Bone morphogenetic proteins (BMPs) are important targets to incorporate in biomaterial scaffolds to orchestrate tissue repair. Glycosaminoglycans (GAGs) such as heparin allow the capture of BMPs and their retention at the surface of biomaterials at safe concentrations. Although heparin has strong affinities for BMP2 and BMP4, two important types of growth factors regulating bone and tissue repair, it remains difficult to embed stably at the surface of a broad range of biomaterials and degrades rapidly in vitro and in vivo. In this report, biomimetic poly(sulfopropyl methacrylate) (PSPMA) brushes are proposed as sulfated GAG mimetic interfaces for the stable capture of BMPs. The growth of PSPMA brushes via a surface-initiated activator regenerated by electron transfer polymerization is investigated via ellipsometry, prior to characterization of swelling and surface chemistry via X-ray photoelectron spectroscopy and Fourier transform infrared. The capacity of PSPMA brushes to bind BMP2 and BMP4 is then characterized via surface plasmon resonance. BMP2 is found to anchor particularly stably and at high density at the surface of PSPMA brushes, and a strong impact of the brush architecture on binding capacity is observed. These results are further confirmed using a quartz crystal microbalance with dissipation monitoring, providing some insights into the mode of adsorption of BMPs at the surface of PSPMA brushes. Primary adsorption of BMP2, with relatively little infiltration, is observed on thick dense brushes, implying that this growth factor should be accessible for further binding of corresponding cell membrane receptors. Finally, to demonstrate the impact of PSPMA brushes for BMP2 capture, dermal fibroblasts were then cultured at the surface of functionalized PSPMA brushes. The presence of BMP2 and the architecture of the brush are found to have a significant impact on matrix deposition at the corresponding interfaces. Therefore, PSPMA brushes emerge as attractive coatings for scaffold engineering and stable capture of BMP2 for regenerative medicine applications.


Subject(s)
Bone Morphogenetic Protein 2 , Bone Morphogenetic Protein 4 , Bone Morphogenetic Protein 2/chemistry , Bone Morphogenetic Protein 2/metabolism , Bone Morphogenetic Protein 4/chemistry , Bone Morphogenetic Protein 4/metabolism , Humans , Sulfonic Acids/chemistry , Methacrylates/chemistry , Surface Properties , Biocompatible Materials/chemistry , Biocompatible Materials/pharmacology , Glycosaminoglycans/chemistry , Glycosaminoglycans/metabolism
4.
Carbohydr Res ; 543: 109201, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39013335

ABSTRACT

Cysteine cathepsins constitute the largest cathepsin family, with 11 proteases in human that are present primarily within acidic endosomal and lysosomal compartments. They are involved in the turnover of intracellular and extracellular proteins. They are synthesized as inactive procathepsins that are converted to mature active forms. Cathepsins play important roles in physiological and pathological processes and, therefore, receive increasing attention as potential therapeutic targets. Their maturation and activity can be regulated by glycosaminoglycans (GAGs), long linear negatively charged polysaccharides composed of recurring dimeric units. In this review, we summarize recent computational progress in the field of (pro)cathepsin-GAG complexes analyses.


Subject(s)
Cathepsins , Glycosaminoglycans , Humans , Glycosaminoglycans/chemistry , Glycosaminoglycans/metabolism , Cathepsins/metabolism , Cathepsins/chemistry , Computer Simulation , Cysteine/chemistry , Cysteine/metabolism
5.
J Biol Chem ; 300(7): 107466, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38876302

ABSTRACT

Glycosaminoglycan (GAG) lyases are often strictly substrate specific, and it is especially difficult to simultaneously degrade GAGs with different types of glycosidic bonds. Herein, we found a new class of GAG lyases (GAGases) from different bacteria. These GAGases belong to polysaccharide lyase 35 family and share quite low homology with the identified GAG lyases. The most surprising thing is that GAGases can not only degrade three types of GAGs: hyaluronan, chondroitin sulfate, and heparan sulfate but also even one of them can also degrade alginate. Further investigation of structural preferences revealed that GAGases selectively act on GAG domains composed of non/6-O-/N-sulfated hexosamines and d-glucoronic acids as well as on alginate domains composed of d-mannuronic acids. In addition, GAG lyases were once speculated to have evolved from alginate lyases, but no transitional enzymes have been found. The discovery of GAGases not only broadens the category of GAG lyases, provides new enzymatic tools for the structural and functional studies of GAGs with specific structures, but also provides candidates for the evolution of GAG lyases.


Subject(s)
Glycosaminoglycans , Polysaccharide-Lyases , Substrate Specificity , Glycosaminoglycans/metabolism , Glycosaminoglycans/chemistry , Polysaccharide-Lyases/metabolism , Polysaccharide-Lyases/chemistry , Bacterial Proteins/chemistry , Bacterial Proteins/metabolism , Bacterial Proteins/genetics , Chondroitin Sulfates/metabolism , Chondroitin Sulfates/chemistry
6.
Biotechnol Adv ; 74: 108394, 2024 Sep.
Article in English | MEDLINE | ID: mdl-38857660

ABSTRACT

Glycosaminoglycans (GAGs) are a family of structurally complex heteropolysaccharides that play pivotal roles in biological functions, including the regulation of cell proliferation, enzyme inhibition, and activation of growth factor receptors. Therefore, the synthesis of GAGs is a hot research topic in drug development. The enzymatic synthesis of GAGs has received widespread attention due to their eco-friendly nature, high regioselectivity, and stereoselectivity. The enhancement of the enzymatic synthesis process is the key to its industrial applications. In this review, we overviewed the construction of more efficient in vitro biomimetic synthesis systems of glycosaminoglycans and presented the different strategies to improve enzyme catalysis, including the combination of chemical and enzymatic methods, solid-phase synthesis, and protein engineering to solve the problems of enzyme stability, separation and purification of the product, preparation of structurally defined sugar chains, etc., and discussed the challenges and opportunities in large-scale green synthesis of GAGs.


Subject(s)
Glycosaminoglycans , Green Chemistry Technology , Glycosaminoglycans/chemistry , Green Chemistry Technology/methods , Biocatalysis , Protein Engineering/methods , Enzymes/chemistry , Enzymes/metabolism , Catalysis
7.
Biomed Mater ; 19(4)2024 Jun 20.
Article in English | MEDLINE | ID: mdl-38857607

ABSTRACT

Hypothyroidism is caused by insufficient stimulation or disruption of the thyroid. However, the drawbacks of thyroid transplantation have led to the search for new treatments. Decellularization allows tissue transplants to maintain their biomimetic structures while preserving cell adhesion, proliferation, and differentiation. This study aimed to decellularize human thyroid tissues using a structure-preserving optimization strategy and present preliminary data on recellularization. Nine methods were used for physical and chemical decellularization. Quantitative and immunohistochemical analyses were performed to investigate the DNA and extracellular matrix components of the tissues. Biomechanical properties were determined by compression test, and cell viability was examined after seeding MDA-T32 papillary thyroid cancer (PTC) cells onto the decellularized tissues. Decellularized tissues exhibited a notable decrease (<50 ng mg-1DNA, except for Groups 2 and 7) compared to the native thyroid tissue. Nonetheless, collagen and glycosaminoglycans were shown to be conserved in all decellularized tissues. Laminin and fibronectin were preserved at comparatively higher levels, and Young's modulus was elevated when decellularization included SDS. It was observed that the strain value in Group 1 (1.63 ± 0.14 MPa) was significantly greater than that in the decellularized tissues between Groups 2-9, ranging from 0.13 ± 0.03-0.72 ± 0.29 MPa. Finally, viability assessment demonstrated that PTC cells within the recellularized tissue groups successfully attached to the 3D scaffolds and sustained metabolic activity throughout the incubation period. We successfully established a decellularization optimization for human thyroid tissues, which has potential applications in tissue engineering and transplantation research. Our next goal is to conduct recellularization using the methods utilized in Group 1 and transplant the primary thyroid follicular cell-seeded tissues into anin vivoanimal model, particularly due to their remarkable 3D structural preservation and cell adhesion-promoting properties.


Subject(s)
Cell Survival , Extracellular Matrix , Thyroid Gland , Tissue Engineering , Tissue Scaffolds , Tissue Engineering/methods , Humans , Thyroid Gland/cytology , Extracellular Matrix/metabolism , Extracellular Matrix/chemistry , Tissue Scaffolds/chemistry , Collagen/chemistry , Cell Adhesion , Glycosaminoglycans/metabolism , Glycosaminoglycans/chemistry , Cell Line, Tumor , DNA , Elastic Modulus , Cell Proliferation , Thyroid Neoplasms/pathology , Decellularized Extracellular Matrix/chemistry , Laminin/chemistry , Biomechanical Phenomena , Cell Differentiation , Thyroid Cancer, Papillary/pathology , Fibronectins/chemistry , Fibronectins/metabolism
8.
Nat Commun ; 15(1): 4912, 2024 Jun 08.
Article in English | MEDLINE | ID: mdl-38851738

ABSTRACT

Bacterial adhesion is a fundamental process which enables colonisation of niche environments and is key for infection. However, in Legionella pneumophila, the causative agent of Legionnaires' disease, these processes are not well understood. The Legionella collagen-like protein (Lcl) is an extracellular peripheral membrane protein that recognises sulphated glycosaminoglycans on the surface of eukaryotic cells, but also stimulates bacterial aggregation in response to divalent cations. Here we report the crystal structure of the Lcl C-terminal domain (Lcl-CTD) and present a model for intact Lcl. Our data reveal that Lcl-CTD forms an unusual trimer arrangement with a positively charged external surface and negatively charged solvent exposed internal cavity. Through molecular dynamics simulations, we show how the glycosaminoglycan chondroitin-4-sulphate associates with the Lcl-CTD surface via distinct binding modes. Our findings show that Lcl homologs are present across both the Pseudomonadota and Fibrobacterota-Chlorobiota-Bacteroidota phyla and suggest that Lcl may represent a versatile carbohydrate-binding mechanism.


Subject(s)
Bacterial Proteins , Collagen , Glycosaminoglycans , Legionella pneumophila , Molecular Dynamics Simulation , Protein Binding , Glycosaminoglycans/metabolism , Glycosaminoglycans/chemistry , Bacterial Proteins/metabolism , Bacterial Proteins/chemistry , Legionella pneumophila/metabolism , Collagen/metabolism , Collagen/chemistry , Crystallography, X-Ray , Chondroitin Sulfates/metabolism , Chondroitin Sulfates/chemistry , Bacterial Adhesion , Protein Domains , Legionnaires' Disease/microbiology , Legionnaires' Disease/metabolism , Humans , Amino Acid Sequence
9.
Langmuir ; 40(27): 14007-14015, 2024 Jul 09.
Article in English | MEDLINE | ID: mdl-38916446

ABSTRACT

Allograft transplantation is an important method for tendon reconstruction after injury, and its clinical success highly relies on the storage and transportation of the grafts. Cryopreservation is a promising strategy for tendon storage. In this study, we report a novel cryopreservation agent (CPA) formulation with a high biocompatibility for tendon cryopreservation. Mainly composed of natural zwitterionic betaine and the biocompatible polymer poly(vinylpyrrolidone) (PVP), it exhibited ideal abilities to depress the freezing point and inhibit ice growth and recrystallization. Notably, after cryopreservation via plunge-freezing for 1 month, Young's modulus (144 MPa, 98% of fresh tendons) and ultimate stress (46.7 MPa, 99% of fresh tendons) remained stable, and the cross-linking of collagen microfibers, protein structures, and glycosaminoglycan (GAG) contents changed slightly. These results indicate that the formulation (5 wt % betaine and 5 wt % PVP in phosphate-buffered saline, PBS solution) effectively maintains the biomechanical properties and tissue structure. This work offers a novel cryopreservation method for tendons and may also provide insights into the long-term preservation of various other tissues.


Subject(s)
Betaine , Cryopreservation , Tendons , Cryopreservation/methods , Tendons/drug effects , Betaine/chemistry , Animals , Freezing , Cryoprotective Agents/chemistry , Cryoprotective Agents/pharmacology , Povidone/chemistry , Collagen/chemistry , Glycosaminoglycans/chemistry , Biocompatible Materials/chemistry , Biocompatible Materials/pharmacology
10.
Carbohydr Polym ; 341: 122294, 2024 Oct 01.
Article in English | MEDLINE | ID: mdl-38876708

ABSTRACT

The role of glycosaminoglycans (GAGs) in modulating bone morphogenetic protein (BMP) signaling represents a recent and underexplored area. Conflicting reports suggest a dual effect: some indicate a positive influence, while others demonstrate a negative impact. This duality suggests that the localization of GAGs (either at the cell surface or within the extracellular matrix) or the specific type of GAG may dictate their signaling role. The precise sulfation patterns of heparan sulfate (HS) responsible for BMP2 binding remain elusive. BMP2 exhibits a preference for binding to HS over other GAGs. Using well-characterized biomaterials mimicking the extracellular matrix, our research reveals that HS promotes BMP2 signaling in the extracellular space, contrary to chondroitin sulfate (CS), which enhances BMP2 bioactivity at the cell surface. Further observations indicate that a central IdoA (2S)-GlcNS (6S) tri-sulfated motif within HS hexasaccharides enhances binding. Nevertheless, BMP2 exhibits a degree of adaptability to various HS sulfation types and sequences. Molecular dynamic simulations attribute this adaptability to the BMP2 N-terminal end flexibility. Our findings illustrate the complex interplay between GAGs and BMP signaling, highlighting the importance of localization and specific sulfation patterns. This understanding has implications for the development of biomaterials with tailored properties for therapeutic applications targeting BMP signaling pathways.


Subject(s)
Bone Morphogenetic Protein 2 , Glycosaminoglycans , Heparitin Sulfate , Signal Transduction , Bone Morphogenetic Protein 2/metabolism , Heparitin Sulfate/metabolism , Heparitin Sulfate/chemistry , Humans , Glycosaminoglycans/metabolism , Glycosaminoglycans/chemistry , Chondroitin Sulfates/chemistry , Chondroitin Sulfates/metabolism , Molecular Dynamics Simulation , Animals , Protein Binding
11.
Acta Biomater ; 182: 42-53, 2024 07 01.
Article in English | MEDLINE | ID: mdl-38729549

ABSTRACT

Magnetic resonance elastography (MRE) and diffusion-weighted imaging (DWI) are complementary imaging techniques that detect disease based on viscoelasticity and water mobility, respectively. However, the relationship between viscoelasticity and water diffusion is still poorly understood, hindering the clinical translation of combined DWI-MRE markers. We used DWI-MRE to study 129 biomaterial samples including native and cross-linked collagen, glycosaminoglycans (GAGs) with different sulfation levels, and decellularized specimens of pancreas and liver, all with different proportions of solid tissue, or solid fractions. We developed a theoretical framework of the relationship between mechanical loss and tissue-water mobility based on two parameters, solid and fluid viscosity. These parameters revealed distinct DWI-MRE property clusters characterizing weak, moderate, and strong water-network interactions. Sparse networks interacting weakly with water, such as collagen or diluted decellularized tissue, resulted in marginal changes in water diffusion over increasing solid viscosity. In contrast, dense networks with larger solid fractions exhibited both free and hindered water diffusion depending on the polarity of the solid components. For example, polar and highly sulfated GAGs as well as native soft tissues hindered water diffusion despite relatively low solid viscosity. Our results suggest that two fundamental properties of tissue networks, solid fraction and network polarity, critically influence solid and fluid viscosity in biological tissues. Since clinical DWI and MRE are sensitive to these viscosity parameters, the framework we present here can be used to detect tissue remodeling and architectural changes in the setting of diagnostic imaging. STATEMENT OF SIGNIFICANCE: The viscoelastic properties of biological tissues provide a wealth of information on the vital state of cells and host matrix. Combined measurement of viscoelasticity and water diffusion by medical imaging is sensitive to tissue microarchitecture. However, the relationship between viscoelasticity and water diffusion is still poorly understood, hindering full exploitation of these properties as a combined clinical biomarker. Therefore, we analyzed the parameter space accessible by diffusion-weighted imaging (DWI) and magnetic resonance elastography (MRE) and developed a theoretical framework for the relationship between water mobility and mechanical parameters in biomaterials. Our theory of solid material properties related to particle motion can be translated to clinical radiology using clinically established MRE and DWI.


Subject(s)
Elasticity , Water , Viscosity , Water/chemistry , Diffusion , Animals , Elasticity Imaging Techniques/methods , Humans , Diffusion Magnetic Resonance Imaging/methods , Collagen/chemistry , Glycosaminoglycans/metabolism , Glycosaminoglycans/chemistry , Liver/diagnostic imaging
12.
N Biotechnol ; 82: 75-84, 2024 Sep 25.
Article in English | MEDLINE | ID: mdl-38750817

ABSTRACT

Natural biopolymers become increasingly attractive as bio-based alternatives to petrol-based rheological modifiers, especially in personal care applications. However, many polysaccharides exhibit undesired properties in cosmetic applications such as limited viscosifying characteristics, unpleasant sensory properties, or incompatibility with certain formulation compounds. Here, a comprehensive rheological analysis of non-decorated acetan-like heteroexopolysaccharides derived from two Kozakia baliensis strains was performed in selected surfactant formulations. The results were compared to native xanthan gum and a genetically engineered xanthan variant, Xan∆gumFGL, which lacks any acetyl- and pyruvyl moieties and whose rheological properties are unaffected by saline environments. All four polysaccharides displayed a highly similar rheological performance in the non-ionic surfactant lauryl glucoside, while the rheological properties differed in amphoteric and anionic surfactants cocamidopropyl betaine and sodium laureth sulfate due to minor changes in side chain composition. Polysaccharide precipitation was observed in the presence of the cationic surfactant. Nevertheless, the native heteroexopolysaccharide derived from K. baliensis LMG 27018 shows significant potential as a salt-independent rheological modifier compared to the genetically engineered Xan∆gumFGL variant. In addition, blends of heteroexopolysaccharides from K. baliensis and several galactomannans displayed synergistic effects which were comparable to native xanthan gum-galactomannan blends. This study shows that heteroexopolysaccharides of K. baliensis are capable of further extending the portfolio of bio-based rheological modifiers.


Subject(s)
Galactose , Mannans , Polysaccharides, Bacterial , Rheology , Surface-Active Agents , Mannans/chemistry , Galactose/analogs & derivatives , Galactose/chemistry , Surface-Active Agents/chemistry , Polysaccharides, Bacterial/chemistry , Glycosaminoglycans/chemistry , Glycosaminoglycans/metabolism
13.
Mar Drugs ; 22(5)2024 Apr 26.
Article in English | MEDLINE | ID: mdl-38786589

ABSTRACT

Glycosaminoglycans (GAGs) are valuable bioactive polysaccharides with promising biomedical and pharmaceutical applications. In this study, we analyzed GAGs using HPLC-MS/MS from the bone (B), muscle (M), skin (S), and viscera (V) of Scophthalmus maximus (SM), Paralichthysi (P), Limanda ferruginea (LF), Cleisthenes herzensteini (G), Platichthys bicoloratus (PB), Pleuronichthys cornutus (PC), and Cleisthenes herzensteini (CH). Unsaturated disaccharide products were obtained by enzymatic hydrolysis of the GAGs and subjected to compositional analysis of chondroitin sulfate (CS), heparin sulfate (HS), and hyaluronic acid (HA), including the sulfation degree of CS and HS, as well as the content of each GAG. The contents of GAGs in the tissues and the sulfation degree differed significantly among the fish. The bone of S. maximus contained more than 12 µg of CS per mg of dry tissue. Although the fish typically contained high levels of CSA (CS-4S), some fish bone tissue exhibited elevated levels of CSC (CS-6S). The HS content was found to range from 10-150 ug/g, primarily distributed in viscera, with a predominant non-sulfated structure (HS-0S). The structure of HA is well-defined without sulfation modification. These analytical results are independent of biological classification. We provide a high-throughput rapid detection method for tissue samples using HPLC-MS/MS to rapidly screen ideal sources of GAG. On this basis, four kinds of CS were prepared and purified from flounder bone, and their molecular weight was determined to be 23-28 kDa by HPGPC-MALLS, and the disaccharide component unit was dominated by CS-6S, which is a potential substitute for CSC derived from shark cartilage.


Subject(s)
Chondroitin Sulfates , Flounder , Glycosaminoglycans , Tandem Mass Spectrometry , Animals , Chondroitin Sulfates/chemistry , Chondroitin Sulfates/isolation & purification , Glycosaminoglycans/isolation & purification , Glycosaminoglycans/chemistry , Chromatography, High Pressure Liquid , Bone and Bones/chemistry , Skin/chemistry , Skin/metabolism , Hyaluronic Acid/chemistry , Hyaluronic Acid/isolation & purification , Muscles/chemistry
14.
Int J Biol Macromol ; 271(Pt 1): 132439, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38761907

ABSTRACT

This study explored the immunomodulatory impact and potential mechanisms on macrophages RAW264.7 using a purified macromolecular sulfate glycosaminoglycan (SBSG) from the swim bladder, whose structure was similar to chondroitin sulfate A. The results showed that SBSG at 0.25-1 mg/mL increased the viability and phagocytosis of RAW264.7 cells. Meanwhile, SBSG promoted the secretion of tumor necrosis factor α (TNF-α), interleukin 10 (IL-10), and nitric oxide (NO), as well as the production of reactive oxygen species (ROS). According to the RT-PCR and Western blot data, SBSG activated TLR4-nuclear factor kappa B (NF-κB) signaling pathways, which decreased the relative mRNA and protein levels of Toll-like receptor 4 (TLR4), IκB kinase ß (IKKß), NF-κB p65, and p-NF-κB p65. The molecular docking and molecular dynamic simulation findings revealed that the main binding force between TLR4 and SBSG was conventional hydrogen bond interaction, resulting in more stable ligand receptor complexes. In summary, SBSG exhibits significant immunomodulatory potential, similar to chondroitin sulfate C. The underlying molecular mechanism involved the binding of SBSG through hydrogen bonding to TLR4 receptors, triggering the NF-κB signaling pathway to downregulate the expression of related genes and proteins. This, in turn, regulated the secretion of various cytokines that were mediated by macrophages to exert the immunity of the body.


Subject(s)
Macrophages , Molecular Docking Simulation , NF-kappa B , Signal Transduction , Toll-Like Receptor 4 , Animals , Mice , Cell Survival/drug effects , Glycosaminoglycans/pharmacology , Glycosaminoglycans/chemistry , Glycosaminoglycans/metabolism , Immunologic Factors/pharmacology , Immunologic Factors/chemistry , Macrophages/drug effects , Macrophages/metabolism , Macrophages/immunology , Molecular Dynamics Simulation , NF-kappa B/metabolism , Nitric Oxide/metabolism , Phagocytosis/drug effects , RAW 264.7 Cells , Reactive Oxygen Species/metabolism , Signal Transduction/drug effects , Toll-Like Receptor 4/metabolism
15.
Chemistry ; 30(35): e202400783, 2024 Jun 20.
Article in English | MEDLINE | ID: mdl-38629399

ABSTRACT

Glycosaminoglycans (GAGs) are linear and acidic polysaccharides. They are ubiquitous molecules, which are involved in a wide range of biological processes. Despite being structurally simple at first glance, with a repeating backbone of alternating hexuronic acid and hexosamine dimers, GAGs display a highly complex structure, which predominantly results from their heterogeneous sulfation patterns. The commonly applied method for compositional analysis of all GAGs is "disaccharide analysis." In this process, GAGs are enzymatically depolymerized into disaccharides, derivatized with a fluorescent label, and then analysed through liquid chromatography. The limiting factor in the high throughput analysis of GAG disaccharides is the time-consuming liquid chromatography. To address this limitation, we here utilized trapped ion mobility-mass spectrometry (TIM-MS) for the separation of isomeric GAG disaccharides, which reduces the measurement time from hours to a few minutes. A full set of disaccharides comprises twelve structures, with eight possessing isomers. Most disaccharides cannot be differentiated by TIM-MS in underivatized form. Therefore, we developed chemical modifications to reduce sample complexity and enhance differentiability. Quantification is performed using stable isotope labelled standards, which are easily available due to the nature of the performed modifications.


Subject(s)
Disaccharides , Glycosaminoglycans , Disaccharides/chemistry , Disaccharides/analysis , Glycosaminoglycans/chemistry , Glycosaminoglycans/analysis , Ion Mobility Spectrometry/methods , Mass Spectrometry/methods , Isomerism , Chromatography, Liquid/methods
16.
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
17.
FEBS J ; 291(15): 3331-3366, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38500384

ABSTRACT

Mammalian glycosaminoglycans (GAGs), except hyaluronan (HA), are sulfated polysaccharides that are covalently attached to core proteins to form proteoglycans (PGs). This article summarizes key biological findings for the most widespread GAGs, namely HA, chondroitin sulfate/dermatan sulfate (CS/DS), keratan sulfate (KS), and heparan sulfate (HS). It focuses on the major processes that remain to be deciphered to get a comprehensive view of the mechanisms mediating GAG biological functions. They include the regulation of GAG biosynthesis and postsynthetic modifications in heparin (HP) and HS, the composition, heterogeneity, and function of the tetrasaccharide linkage region and its role in disease, the functional characterization of the new PGs recently identified by glycoproteomics, the selectivity of interactions mediated by GAG chains, the display of GAG chains and PGs at the cell surface and their impact on the availability and activity of soluble ligands, and on their move through the glycocalyx layer to reach their receptors, the human GAG profile in health and disease, the roles of GAGs and particular PGs (syndecans, decorin, and biglycan) involved in cancer, inflammation, and fibrosis, the possible use of GAGs and PGs as disease biomarkers, and the design of inhibitors targeting GAG biosynthetic enzymes and GAG-protein interactions to develop novel therapeutic approaches.


Subject(s)
Glycosaminoglycans , Humans , Glycosaminoglycans/metabolism , Glycosaminoglycans/chemistry , Animals , Heparitin Sulfate/metabolism , Heparitin Sulfate/chemistry , Proteoglycans/metabolism , Dermatan Sulfate/metabolism , Dermatan Sulfate/chemistry , Neoplasms/metabolism , Neoplasms/drug therapy , Neoplasms/pathology , Hyaluronic Acid/metabolism , Hyaluronic Acid/chemistry , Keratan Sulfate/metabolism , Keratan Sulfate/chemistry , Chondroitin Sulfates/metabolism , Chondroitin Sulfates/chemistry
18.
Biotechnol Appl Biochem ; 71(3): 651-660, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38449083

ABSTRACT

Glycosaminoglycans (GAGs) serve as a biomarker for mucopolysaccharidoses disease. In this study, a novel fluorometric method was developed to measure total GAGs in urine. Graphene oxide (GO) and rhodamine B (RhB), a cationic fluorescent dye, were employed in the development of the method. RhB attaches to the GO surface via electrostatic attraction, leading to the quenching of its fluorescence upon the establishment of the RhB-GO complex. However, the presence of GAGs prompts a resurgence of intense fluorescence. The linear range of the method is between 5.00 and 70.00 mg/L. The total GAG levels of urine samples analyzed using the method agree with the results of the biochemistry analysis laboratory (65.85 and 79.18 mg/L; 73.30 ± 1.76 and 72.21 ± 2.21). The method is simple, accurate, and sensitive and may be used for both first-step diagnosis of the mucopolysaccharidoses and detection of individual GAGs for studies of GAG-related research and other biological applications.


Subject(s)
Glycosaminoglycans , Graphite , Graphite/chemistry , Glycosaminoglycans/urine , Glycosaminoglycans/chemistry , Humans , Spectrometry, Fluorescence , Rhodamines/chemistry , Fluorescent Dyes/chemistry , Fluorescence , Mucopolysaccharidoses/urine , Mucopolysaccharidoses/diagnosis
19.
J Chem Inf Model ; 64(5): 1691-1703, 2024 03 11.
Article in English | MEDLINE | ID: mdl-38410841

ABSTRACT

Glycosaminoglycans (GAGs) made of repeating disaccharide units intricately engage with proteins, playing a crucial role in the spatial organization of the extracellular matrix (ECM) and the transduction of biological signals in cells to modulate a number of biochemical processes. Exploring protein-GAG interactions reveals several challenges for their analysis, namely, the highly charged and periodic nature of GAGs, their multipose binding, and the abundance of the interfacial water molecules in the protein-GAG complexes. Most of the studies on protein-GAG interactions are conducted using the TIP3P water model, and there are no data on the effect of various water models on the results obtained in molecular dynamics (MD) simulations of protein-GAG complexes. Hence, it is essential to perform a systematic analysis of different water models in MD simulations for these systems. In this work, we aim to evaluate the properties of the protein-GAG complexes in MD simulations using different explicit: TIP3P, SPC/E, TIP4P, TIP4PEw, OPC, and TIP5P and implicit: IGB = 1, 2, 5, 7, and 8 water models to find out which of them are best suited to study the dynamics of protein-GAG complexes. The FF14SB and GLYCAM06 force fields were used for the proteins and GAGs, respectively. The interactions of several GAG types, such as heparin, chondroitin sulfate, and hyaluronic acid with basic fibroblast growth factor, cathepsin K, and CD44 receptor, respectively, are investigated. The observed variations in different descriptors used to study the binding in these complexes emphasize the relevance of the choice of water models for the MD simulation of these complexes.


Subject(s)
Glycosaminoglycans , Molecular Dynamics Simulation , Glycosaminoglycans/chemistry , Water/chemistry , Benchmarking , Heparin/chemistry , Proteins/chemistry
20.
Int J Biol Macromol ; 263(Pt 1): 130128, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38350587

ABSTRACT

Bone morphogenetic protein-2 (BMP-2) is a critical growth factor of bone extracellular matrix (ECM), pivotal for osteogenesis. Glycosaminoglycans (GAGs), another vital ECM biomolecules, interact with growth factors, affecting signal transduction. Our study primarily focused on hyaluronic acid (HA), a prevalent GAG, and its sulfated derivative (SHA). We explored their impact on BMP-2's conformation, aggregation, and mechanistic pathways of aggregation using diverse optical and rheological methods. In the presence of HA and SHA, the secondary structure of BMP-2 underwent a structured transformation, characterized by a substantial increase in beta sheet content, and a detrimental alteration, manifesting as a shift towards unstructured content, respectively. Although both HA and SHA induced BMP-2 aggregation, their mechanisms differed. SHA led to rapid amorphous aggregates, while HA promoted amyloid fibrils with a lag phase and sigmoidal kinetics. Aggregate size and shape varied; HA produced larger structures, SHA smaller. Each aggregation type followed distinct pathways influenced by viscosity and excluded volume. Higher viscosity, low diffusivity of protein and higher excluded volume In the presence of HA promotes fibrillation having size in micrometer range. Low viscosity, high diffusivity of protein and lesser excluded volume leads to amorphous aggregate of size in nanometer range.


Subject(s)
Glycosaminoglycans , Hyaluronic Acid , Hyaluronic Acid/chemistry , Glycosaminoglycans/chemistry , Extracellular Matrix/metabolism , Bone Morphogenetic Protein 2/metabolism , Chemical Phenomena , Osteogenesis
SELECTION OF CITATIONS
SEARCH DETAIL