Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 562
Filtrar
1.
Biomed Mater ; 19(4)2024 Jun 20.
Artigo em Inglês | MEDLINE | ID: mdl-38857607

RESUMO

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.


Assuntos
Sobrevivência Celular , Matriz Extracelular , Glândula Tireoide , Engenharia Tecidual , Alicerces Teciduais , Engenharia Tecidual/métodos , Humanos , Glândula Tireoide/citologia , Matriz Extracelular/metabolismo , Matriz Extracelular/química , Alicerces Teciduais/química , Colágeno/química , Adesão Celular , Glicosaminoglicanos/metabolismo , Glicosaminoglicanos/química , Linhagem Celular Tumoral , DNA , Módulo de Elasticidade , Proliferação de Células , Neoplasias da Glândula Tireoide/patologia , Matriz Extracelular Descelularizada/química , Laminina/química , Fenômenos Biomecânicos , Diferenciação Celular , Câncer Papilífero da Tireoide/patologia , Fibronectinas/química , Fibronectinas/metabolismo
2.
Int J Biol Macromol ; 271(Pt 1): 132439, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38761907

RESUMO

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.


Assuntos
Macrófagos , Simulação de Acoplamento Molecular , NF-kappa B , Transdução de Sinais , Receptor 4 Toll-Like , Animais , Receptor 4 Toll-Like/metabolismo , Camundongos , Células RAW 264.7 , Transdução de Sinais/efeitos dos fármacos , NF-kappa B/metabolismo , Macrófagos/efeitos dos fármacos , Macrófagos/metabolismo , Macrófagos/imunologia , Fatores Imunológicos/farmacologia , Fatores Imunológicos/química , Glicosaminoglicanos/farmacologia , Glicosaminoglicanos/química , Glicosaminoglicanos/metabolismo , Óxido Nítrico/metabolismo , Fagocitose/efeitos dos fármacos , Espécies Reativas de Oxigênio/metabolismo , Simulação de Dinâmica Molecular , Sobrevivência Celular/efeitos dos fármacos
3.
Int J Biol Macromol ; 253(Pt 4): 126975, 2023 Dec 31.
Artigo em Inglês | MEDLINE | ID: mdl-37739278

RESUMO

Glycosaminoglycans (GAGs) are naturally existing extracellular components with a variety important biological functions. However, their heterogeneous chemical compositions and the challenges in purification have become the main disadvantages for clinical applications. Thus, various synthetic glycopolymers have been designed to mimic the structures and functions of natural GAGs. In the current study, glycopolymers from structurally simple glucose or N-acetylglucosamine monomers were synthesized, which were further subjected to sulfation of different degrees and grafting onto silica nanoparticles, leading to spherical-shaped nano-structures of uniform diameters. With the successively strengthened multivalent effect, the obtained glycopolymer nanoparticles not only showed excellent effects on promotion of cell proliferation by stabilizing growth factors, but also significantly inhibited tumor metastasis by weakening the adhesion between tumor cells and activated platelets. Among the prepared nanoparticles, S3-PGNAc@Si with N-acetylglucosamine segment and the highest sulfation degree exhibited the strongest bioactivities, which were even close to those of heparin. This work presents a novel approach for structural and functional mimicking of natural GAGs from simple and low-cost monosaccharides, holding great potential for a range of biomedical applications.


Assuntos
Glicosaminoglicanos , Nanopartículas , Glicosaminoglicanos/farmacologia , Glicosaminoglicanos/química , Acetilglucosamina , Heparina/farmacologia , Proliferação de Células , Nanopartículas/química
4.
Mar Drugs ; 21(5)2023 Apr 25.
Artigo em Inglês | MEDLINE | ID: mdl-37233458

RESUMO

Sulfated glycans from marine organisms are excellent sources of naturally occurring glycosaminoglycan (GAG) mimetics that demonstrate therapeutic activities, such as antiviral/microbial infection, anticoagulant, anticancer, and anti-inflammation activities. Many viruses use the heparan sulfate (HS) GAG on the surface of host cells as co-receptors for attachment and initiating cell entry. Therefore, virion-HS interactions have been targeted to develop broad-spectrum antiviral therapeutics. Here we report the potential anti-monkeypox virus (MPXV) activities of eight defined marine sulfated glycans, three fucosylated chondroitin sulfates, and three sulfated fucans extracted from the sea cucumber species Isostichopus badionotus, Holothuria floridana, and Pentacta pygmaea, and the sea urchin Lytechinus variegatus, as well as two chemically desulfated derivatives. The inhibitions of these marine sulfated glycans on MPXV A29 and A35 protein-heparin interactions were evaluated using surface plasmon resonance (SPR). These results demonstrated that the viral surface proteins of MPXV A29 and A35 bound to heparin, which is a highly sulfated HS, and sulfated glycans from sea cucumbers showed strong inhibition of MPXV A29 and A35 interactions. The study of molecular interactions between viral proteins and host cell GAGs is important in developing therapeutics for the prevention and treatment of MPXV.


Assuntos
Glicosaminoglicanos , Pepinos-do-Mar , Animais , Glicosaminoglicanos/química , Ressonância de Plasmônio de Superfície , Sulfatos/farmacologia , Sulfatos/química , Heparitina Sulfato/farmacologia , Sulfatos de Condroitina , Heparina/farmacologia , Pepinos-do-Mar/química , Antivirais/farmacologia
5.
Biomolecules ; 13(2)2023 01 28.
Artigo em Inglês | MEDLINE | ID: mdl-36830616

RESUMO

Glycosaminoglycans (GAGs) are a class of linear anionic periodic polysaccharides containing disaccharide repetitive units. These molecules interact with a variety of proteins in the extracellular matrix and so participate in biochemically crucial processes such as cell signalling affecting tissue regeneration as well as the onset of cancer, Alzheimer's or Parkinson's diseases. Due to their flexibility, periodicity and chemical heterogeneity, often termed "sulfation code", GAGs are challenging molecules both for experiments and computation. One of the key questions in the GAG research is the specificity of their intermolecular interactions. In this study, we make a step forward to deciphering the "sulfation code" of chondroitin sulfates-4,6 (CS4, CS6, where the numbers correspond to the position of sulfation in NAcGal residue) and dermatan sulfate (DS), which is different from CSs by the presence of IdoA acid instead of GlcA. We rigorously investigate two sets of these GAGs in dimeric, tetrameric and hexameric forms with molecular dynamics-based descriptors. Our data clearly suggest that CS4, CS6 and DS are substantially different in terms of their structural, conformational and dynamic properties, which contributes to the understanding of how these molecules can be different when they bind proteins, which could have practical implications for the GAG-based drug design strategies in the regenerative medicine.


Assuntos
Dermatan Sulfato , Simulação de Dinâmica Molecular , Dermatan Sulfato/análise , Dermatan Sulfato/química , Dermatan Sulfato/metabolismo , Sulfatos de Condroitina/química , Glicosaminoglicanos/química , Sulfatos
6.
Biomater Adv ; 147: 213319, 2023 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-36758282

RESUMO

Many established bioinks fulfill important requirements regarding fabrication standards and cytocompatibility. Current research focuses on development of functionalized bioinks with an improved support of tissue-specific cell differentiation. Many approaches primarily depend on decellularized extracellular matrices or blood components. In this study, we investigated the combination of a highly viscous alginate-methylcellulose (algMC) bioink with collagen-based artificial extracellular matrix (aECM) as a finely controllable and tailorable system composed of collagen type I (col) with and without chondroitin sulfate (CS) or sulfated hyaluronan (sHA). As an additional stabilizer, the polyphenol tannic acid (TA) was integrated into the inks. The assessment of rheological properties and printability as well as hydrogel microstructure revealed no adverse effect of the integrated components on the inks. Viability, adhesion, and proliferation of bioprinted immortalized human mesenchymal stem cells (hTERT-MSC) was improved indicating enhanced interaction with the designed microenvironment. Furthermore, chondrogenic matrix production (collagen type II and sulfated glycosaminoglycans) by primary human chondrocytes (hChon) was enhanced by aECM. Supplementing the inks with TA was required for these positive effects but caused cytotoxicity as soon as TA concentrations exceeded a certain amount. Thus, combining tailorable aECM with algMC and balanced TA addition proved to be a promising approach for promoting adhesion of immortalized stem cells and differentiation of chondrocytes in bioprinted scaffolds.


Assuntos
Alginatos , Células-Tronco Mesenquimais , Humanos , Células-Tronco Mesenquimais/metabolismo , Glicosaminoglicanos/química , Glicosaminoglicanos/metabolismo , Glicosaminoglicanos/farmacologia , Colágeno Tipo I/metabolismo , Colágeno Tipo I/farmacologia , Diferenciação Celular , Metilcelulose/metabolismo , Metilcelulose/farmacologia , Taninos/metabolismo , Taninos/farmacologia
7.
Methods Mol Biol ; 2619: 153-167, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-36662469

RESUMO

Glycosaminoglycans are long linear periodic anionic polysaccharides consisting of disaccharide units exhibiting different sulfation patterns forming a highly heterogeneous group of molecules. Due to their flexibility, length, high charge, and periodicity, they are challenging for computational approaches. Despite their biological significance in terms of the important role in various diseases (e.g., Alzheimer, cancer, SARS-CoV-2) and proper cell functioning (e.g., proliferation, maturation), there is a lack of effective molecular docking tools designed specifically for glycosaminoglycans due to their challenging physical-chemical nature. In this chapter we present protocols for the Repulsive Scaling Replica Exchange Molecular Dynamics (RS-REMD) methods to dock glycosaminoglycans with both implicit and explicit solvent models implemented. This novel molecular dynamics-based replica exchange technique should help to elevate our current knowledge on the complexes and interactions between glycosaminoglycans and their protein receptors.


Assuntos
COVID-19 , Glicosaminoglicanos , Humanos , Glicosaminoglicanos/química , Simulação de Dinâmica Molecular , Simulação de Acoplamento Molecular , SARS-CoV-2/metabolismo
8.
Protein Sci ; 32(1): e4508, 2023 01.
Artigo em Inglês | MEDLINE | ID: mdl-36369695

RESUMO

Fibrillar collagen-integrin interactions in the extracellular matrix (ECM) regulate a multitude of cellular processes and cell signalling. Collagen I fibrils serve as the molecular scaffolding for connective tissues throughout the human body and are the most abundant protein building blocks in the ECM. The ECM environment is diverse, made up of several ECM proteins, enzymes, and proteoglycans. In particular, glycosaminoglycans (GAGs), anionic polysaccharides that decorate proteoglycans, become depleted in the ECM with natural aging and their mis-regulation has been linked to cancers and other diseases. The impact of GAG depletion in the ECM environment on collagen I protein interactions and on mechanical properties is not well understood. Here, we integrate ELISA protein binding assays with liquid high-resolution atomic force microscopy (AFM) to assess the effects of GAG depletion on the interaction of collagen I fibrils with the integrin α2I domain using separate rat tails. ELISA binding assays demonstrate that α2I preferentially binds to GAG-depleted collagen I fibrils in comparison to native fibrils. By amplitude modulated AFM in air and in solution, we find that GAG-depleted collagen I fibrils retain structural features of the native fibrils, including their characteristic D-banding pattern, a key structural motif. AFM fast force mapping in solution shows that GAG depletion reduces the stiffness of individual fibrils, lowering the indentation modulus by half compared to native fibrils. Together these results shed new light on how GAGs influence collagen I fibril-integrin interactions and may aid in strategies to treat diseases that result from GAG mis-regulation.


Assuntos
Matriz Extracelular , Glicosaminoglicanos , Ratos , Humanos , Animais , Glicosaminoglicanos/análise , Glicosaminoglicanos/química , Glicosaminoglicanos/metabolismo , Matriz Extracelular/química , Proteoglicanas/análise , Proteoglicanas/metabolismo , Microscopia de Força Atômica , Colágeno/química
9.
J R Soc Interface ; 19(193): 20220391, 2022 08.
Artigo em Inglês | MEDLINE | ID: mdl-35919982

RESUMO

Phosphate and sulfate groups are integral to energy metabolism and introduce negative charges into biological macromolecules. One purpose of such modifications is to elicit precise binding/activation of protein partners. The physico-chemical properties of the two groups, while superficially similar, differ in one important respect-the valency of the central (phosphorus or sulfur) atom. This dictates the distinct properties of their respective esters, di-esters and hence their charges, interactions with metal ions and their solubility. These, in turn, determine the contrasting roles for which each group has evolved in biological systems. Biosynthetic links exist between the two modifications; the sulfate donor 3'-phosphoadenosine-5'-phosphosulfate being formed from adenosine triphosphate (ATP) and adenosine phosphosulfate, while the latter is generated from sulfate anions and ATP. Furthermore, phosphorylation, by a xylosyl kinase (Fam20B, glycosaminoglycan xylosylkinase) of the xylose residue of the tetrasaccharide linker region that connects nascent glycosaminoglycan (GAG) chains to their parent proteoglycans, substantially accelerates their biosynthesis. Following observations that GAG chains can enter the cell nucleus, it is hypothesized that sulfated GAGs could influence events in the nucleus, which would complete a feedback loop uniting the complementary anionic modifications of phosphorylation and sulfation through complex, inter-connected signalling networks and warrants further exploration.


Assuntos
Vias Biossintéticas , Glicosaminoglicanos , Trifosfato de Adenosina/metabolismo , Ésteres , Glicosaminoglicanos/química , Fosforilação , Sulfatos/metabolismo
10.
J Comput Chem ; 43(24): 1633-1640, 2022 09 15.
Artigo em Inglês | MEDLINE | ID: mdl-35796487

RESUMO

Glycosaminoglcyans (GAGs), linear anionic periodic polysaccharides, are crucial for many biologically relevant functions in the extracellular matrix. By interacting with proteins GAGs mediate processes such as cancer development, cell proliferation and the onset of neurodegenerative diseases. Despite this eminent importance of GAGs, they still represent a limited focus for the computational community in comparison to other classes of biomolecules. Therefore, there is a lack of modeling tools designed specifically for docking GAGs. One has to rely on existing docking software developed mostly for small drug molecules substantially differing from GAGs in their basic physico-chemical properties. In this study, we present an updated protocol for docking GAGs based on the Repulsive Scaling Replica Exchange Molecular Dynamics (RS-REMD) that includes explicit solvent description. The use of this water model improved docking performance both in terms of its accuracy and speed. This method represents a significant computational progress in GAG-related research.


Assuntos
Glicosaminoglicanos , Simulação de Dinâmica Molecular , Glicosaminoglicanos/química , Proteínas/química , Solventes/química , Água/química
11.
Am J Physiol Cell Physiol ; 322(6): C1187-C1200, 2022 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-35385322

RESUMO

Glycosaminoglycans (GAGs) are an important component of the tumor microenvironment (TME). GAGs can interact with a variety of binding partners and thereby influence cancer progression on multiple levels. GAGs can modulate growth factors and chemokine signaling, invasion, and metastasis formation. Moreover, GAGs are able to change the physical property of the extracellular matrix (ECM). Abnormalities in GAG abundance and structure (e.g., sulfation patterns and molecular weight) are found across various cancer types and show biomarker potential. Targeting GAGs, as well as the usage of GAGs and their mimetics, are promising approaches to interfere with cancer progression. In addition, GAGs can be used as drug and cytokine carriers to induce an antitumor response. In this review, we summarize the role of GAGs in cancer and the potential use of GAGs and GAG derivatives to target cancer.


Assuntos
Glicosaminoglicanos , Neoplasias , Matriz Extracelular/metabolismo , Glicosaminoglicanos/química , Glicosaminoglicanos/metabolismo , Glicosaminoglicanos/uso terapêutico , Humanos , Neoplasias/tratamento farmacológico , Neoplasias/metabolismo , Microambiente Tumoral
12.
Am J Physiol Cell Physiol ; 322(5): C849-C864, 2022 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-35294848

RESUMO

Glycosaminoglycans (GAGs) are long, linear polysaccharides that are ubiquitously expressed on the cell surface and in the extracellular matrix of all animal cells. These complex carbohydrates play important roles in many cellular processes and have been implicated in many disease states, including cancer, inflammation, and genetic disorders. GAGs are among the most complex molecules in biology with enormous information content and extensive structural and functional heterogeneity. GAG biosynthesis is a nontemplate-driven process facilitated by a large group of biosynthetic enzymes that have been extensively characterized over the past few decades. Interestingly, the expression of the enzymes and the consequent structure and function of the polysaccharide chains can vary temporally and spatially during development and under certain pathophysiological conditions, suggesting their assembly is tightly regulated in cells. Due to their many key roles in cell homeostasis and disease, there is much interest in targeting the assembly and function of GAGs as a therapeutic approach. Recent advances in genomics and GAG analytical techniques have pushed the field and generated new perspectives on the regulation of mammalian glycosylation. This review highlights the spatiotemporal diversity of GAGs and the mechanisms guiding their assembly and function in human biology and disease.


Assuntos
Genômica , Glicosaminoglicanos , Animais , Matriz Extracelular/metabolismo , Glicosaminoglicanos/química , Glicosaminoglicanos/metabolismo , Glicosilação , Homeostase , Humanos , Mamíferos/metabolismo
13.
Int J Mol Sci ; 23(3)2022 Jan 27.
Artigo em Inglês | MEDLINE | ID: mdl-35163379

RESUMO

Non-viral gene delivery has become a popular approach in tissue engineering, as it permits the transient delivery of a therapeutic gene, in order to stimulate tissue repair. However, the efficacy of non-viral delivery vectors remains an issue. Our lab has created gene-activated scaffolds by incorporating various non-viral delivery vectors, including the glycosaminoglycan-binding enhanced transduction (GET) peptide into collagen-based scaffolds with proven osteogenic potential. A modification to the GET peptide (FLR) by substitution of arginine residues with histidine (FLH) has been designed to enhance plasmid DNA (pDNA) delivery. In this study, we complexed pDNA with combinations of FLR and FLH peptides, termed GET* nanoparticles. We sought to enhance our gene-activated scaffold platform by incorporating GET* nanoparticles into collagen-nanohydroxyapatite scaffolds with proven osteogenic capacity. GET* N/P 8 was shown to be the most effective formulation for delivery to MSCs in 2D. Furthermore, GET* N/P 8 nanoparticles incorporated into collagen-nanohydroxyapatite (coll-nHA) scaffolds at a 1:1 ratio of collagen:nanohydroxyapatite was shown to be the optimal gene-activated scaffold. pDNA encoding stromal-derived factor 1α (pSDF-1α), an angiogenic chemokine which plays a role in BMP mediated differentiation of MSCs, was then delivered to MSCs using our optimised gene-activated scaffold platform, with the aim of significantly increasing angiogenesis as an important precursor to bone repair. The GET* N/P 8 coll-nHA scaffolds successfully delivered pSDF-1α to MSCs, resulting in a significant, sustained increase in SDF-1α protein production and an enhanced angiogenic effect, a key precursor in the early stages of bone repair.


Assuntos
Peptídeos Penetradores de Células/farmacologia , Quimiocina CXCL12/administração & dosagem , Sistemas de Liberação de Medicamentos , Neovascularização Fisiológica , Engenharia Tecidual , Alicerces Teciduais/química , Ativação Transcricional , Animais , Materiais Biocompatíveis/farmacologia , Quimiocina CXCL12/farmacologia , Colágeno/química , DNA/química , Durapatita/química , Células Progenitoras Endoteliais/metabolismo , Glicosaminoglicanos/química , Nanopartículas , Neovascularização Fisiológica/efeitos dos fármacos , Plasmídeos/química , Ratos Sprague-Dawley , Ativação Transcricional/efeitos dos fármacos , Transfecção
14.
J Mol Graph Model ; 113: 108153, 2022 06.
Artigo em Inglês | MEDLINE | ID: mdl-35183977

RESUMO

Human cysteine cathepsins are lysosomal proteases, which are involved in different biological processes. Their enzymatic activity can be regulated by glycosaminoglycans (GAGs): long linear periodic negatively charged polysaccharides, which dimeric building blocks consist of uronic acid and hexosamine monosaccharide units. In this study, molecular docking simulations of chondroitin 4-sulfate, chondroitin 6-sulfate, heparin, heparan sulfate, dermatan sulfate and hyaluronic acid of various chain lengths were performed with cathepsins B, L, K, S and V and followed by molecular dynamics-based refinement and binding free energy analysis. We concluded that electrostatics might be a driving force for cathepsin-GAG interactions; indeed as in most of characterised systems, the increase of GAG chain length consequently leads to a more pronounced effect on the strength of cathepsin-GAG interactions. Results also suggest that binding of GAGs at different regions on cathepsins surface affect differently their enzymatic activity and could is dependent on cathepsin and GAG type. Present data contribute to systematic description of cathepsin-GAG interactions, which is helpful in understanding the subtle molecular mechanisms of protease regulation behind their biological functions.


Assuntos
Cisteína , Glicosaminoglicanos , Catepsinas/química , Catepsinas/metabolismo , Sulfatos de Condroitina/química , Sulfatos de Condroitina/metabolismo , Glicosaminoglicanos/química , Humanos , Simulação de Acoplamento Molecular
15.
Nat Commun ; 12(1): 7171, 2021 12 09.
Artigo em Inglês | MEDLINE | ID: mdl-34887403

RESUMO

Hedgehog (HH) morphogen signalling, crucial for cell growth and tissue patterning in animals, is initiated by the binding of dually lipidated HH ligands to cell surface receptors. Hedgehog-Interacting Protein (HHIP), the only reported secreted inhibitor of Sonic Hedgehog (SHH) signalling, binds directly to SHH with high nanomolar affinity, sequestering SHH. Here, we report the structure of the HHIP N-terminal domain (HHIP-N) in complex with a glycosaminoglycan (GAG). HHIP-N displays a unique bipartite fold with a GAG-binding domain alongside a Cysteine Rich Domain (CRD). We show that HHIP-N is required to convey full HHIP inhibitory function, likely by interacting with the cholesterol moiety covalently linked to HH ligands, thereby preventing this SHH-attached cholesterol from binding to the HH receptor Patched (PTCH1). We also present the structure of the HHIP C-terminal domain in complex with the GAG heparin. Heparin can bind to both HHIP-N and HHIP-C, thereby inducing clustering at the cell surface and generating a high-avidity platform for SHH sequestration and inhibition. Our data suggest a multimodal mechanism, in which HHIP can bind two specific sites on the SHH morphogen, alongside multiple GAG interactions, to inhibit SHH signalling.


Assuntos
Proteínas de Transporte/química , Proteínas de Transporte/metabolismo , Proteínas Hedgehog/metabolismo , Glicoproteínas de Membrana/química , Glicoproteínas de Membrana/metabolismo , Transdução de Sinais , Proteínas de Transporte/genética , Colesterol/química , Colesterol/metabolismo , Glicosaminoglicanos/química , Glicosaminoglicanos/metabolismo , Proteínas Hedgehog/química , Proteínas Hedgehog/genética , Humanos , Ligantes , Glicoproteínas de Membrana/genética , Ligação Proteica , Domínios Proteicos
16.
Int J Mol Sci ; 22(23)2021 Nov 26.
Artigo em Inglês | MEDLINE | ID: mdl-34884623

RESUMO

The present study analyzes the capacity of collagen (coll)/sulfated glycosaminoglycan (sGAG)-based surface coatings containing bioactive glass nanoparticles (BGN) in promoting the osteogenic differentiation of human mesenchymal stroma cells (hMSC). Physicochemical characteristics of these coatings and their effects on proliferation and osteogenic differentiation of hMSC were investigated. BGN were stably incorporated into the artificial extracellular matrices (aECM). Oscillatory rheology showed predominantly elastic, gel-like properties of the coatings. The complex viscosity increased depending on the GAG component and was further elevated by adding BGN. BGN-containing aECM showed a release of silicon ions as well as an uptake of calcium ions. hMSC were able to proliferate on coll and coll/sGAG coatings, while cellular growth was delayed on aECM containing BGN. However, a stimulating effect of BGN on ALP activity and calcium deposition was shown. Furthermore, a synergistic effect of sGAG and BGN was found for some donors. Our findings demonstrated the promising potential of aECM and BGN combinations in promoting bone regeneration. Still, future work is required to further optimize the BGN/aECM combination for increasing its combined osteogenic effect.


Assuntos
Diferenciação Celular , Matriz Extracelular/química , Vidro/química , Células-Tronco Mesenquimais/citologia , Nanopartículas/administração & dosagem , Osteogênese , Proliferação de Células , Células Cultivadas , Colágeno/química , Glicosaminoglicanos/química , Humanos , Células-Tronco Mesenquimais/efeitos dos fármacos , Nanopartículas/química
17.
Biomolecules ; 11(9)2021 09 12.
Artigo em Inglês | MEDLINE | ID: mdl-34572563

RESUMO

Glycosaminoglycans (GAGs) are linear anionic periodic polysaccharides participating in a number of biologically relevant processes in the extracellular matrix via interactions with their protein targets. Due to their periodicity, conformational flexibility, pseudo-symmetry of the sulfation pattern, and the key role of electrostatics, these molecules are challenging for both experimental and theoretical approaches. In particular, conventional molecular docking applied for GAGs longer than 10-mer experiences severe difficulties. In this work, for the first time, 24- and 48-meric GAGs were docked using all-atomic repulsive-scaling Hamiltonian replica exchange molecular dynamics (RS-REMD), a novel methodology based on replicas with van der Waals radii of interacting molecules being scaled. This approach performed well for proteins complexed with oligomeric GAGs and is independent of their length, which distinguishes it from other molecular docking approaches. We built a model of long GAGs in complex with a proliferation-inducing ligand (APRIL) prebound to its receptors, the B cell maturation antigen and the transmembrane activator and calcium modulator and cyclophilin ligand interactor (TACI). Furthermore, the prediction power of the RS-REMD for this tertiary complex was evaluated. We conclude that the TACI-GAG interaction could be potentially amplified by TACI's binding to APRIL. RS-REMD outperformed Autodock3, the docking program previously proven the best for short GAGs.


Assuntos
Glicosaminoglicanos/química , Simulação de Dinâmica Molecular , Proteína Transmembrana Ativadora e Interagente do CAML/química , Membro 13 da Superfamília de Ligantes de Fatores de Necrose Tumoral/química , Antígeno de Maturação de Linfócitos B/química , Heparina/química , Simulação de Acoplamento Molecular , Ligação Proteica , Termodinâmica
18.
Biol Chem ; 402(11): 1427-1440, 2021 10 26.
Artigo em Inglês | MEDLINE | ID: mdl-34472763

RESUMO

Glycosaminoglycans (GAGs) are essential functional components of the extracellular matrix (ECM). Artificial GAGs like sulfated hyaluronan (sHA) exhibit pro-osteogenic properties and boost healing processes. Hence, they are of high interest for supporting bone regeneration and wound healing. Although sulfated GAGs (sGAGs) appear intracellularly, the knowledge about intracellular effects and putative interaction partners is scarce. Here we used an affinity-purification mass spectrometry-based (AP-MS) approach to identify novel and particularly intracellular sGAG-interacting proteins in human bone marrow stromal cells (hBMSC). Overall, 477 proteins were found interacting with at least one of four distinct sGAGs. Enrichment analysis for protein localization showed that mainly intracellular and cell-associated interacting proteins were identified. The interaction of sGAG with α2-macroglobulin receptor-associated protein (LRPAP1), exportin-1 (XPO1), and serine protease HTRA1 (HTRA1) was confirmed in reverse assays. Consecutive pathway and cluster analysis led to the identification of biological processes, namely processes involving binding and processing of nucleic acids, LRP1-dependent endocytosis, and exosome formation. Respecting the preferentially intracellular localization of sGAG in vesicle-like structures, also the interaction data indicate sGAG-specific modulation of vesicle-based transport processes. By identifying many sGAG-specific interacting proteins, our data provide a resource for upcoming studies aimed at molecular mechanisms and understanding of sGAG cellular effects.


Assuntos
Glicosaminoglicanos/metabolismo , Serina Peptidase 1 de Requerimento de Alta Temperatura A/metabolismo , Carioferinas/metabolismo , Proteína Associada a Proteínas Relacionadas a Receptor de LDL/metabolismo , Receptores Citoplasmáticos e Nucleares/metabolismo , Células Cultivadas , Cromatografia Líquida , Glicosaminoglicanos/química , Serina Peptidase 1 de Requerimento de Alta Temperatura A/química , Serina Peptidase 1 de Requerimento de Alta Temperatura A/isolamento & purificação , Humanos , Carioferinas/química , Carioferinas/isolamento & purificação , Proteína Associada a Proteínas Relacionadas a Receptor de LDL/química , Proteína Associada a Proteínas Relacionadas a Receptor de LDL/isolamento & purificação , Células-Tronco Mesenquimais/química , Células-Tronco Mesenquimais/metabolismo , Receptores Citoplasmáticos e Nucleares/química , Receptores Citoplasmáticos e Nucleares/isolamento & purificação , Espectrometria de Massas em Tandem , Proteína Exportina 1
19.
Biomolecules ; 11(8)2021 08 18.
Artigo em Inglês | MEDLINE | ID: mdl-34439900

RESUMO

Hyaluronic acid (HA), a glycosaminoglycan ubiquitous in the skin, has come into the limelight in recent years for its role in facilitating dermal wound healing. Specifically, HA's length of linearly repeating disaccharides-in other words, its molecular weight (MW)-determines its effects. High molecular weight (HMW)-HA serves an immunosuppressive and anti-inflammatory role, whereas low molecular weight (LMW)-HA contributes to immunostimulation and thus inflammation. During the inflammatory stage of tissue repair, direct and indirect interactions between HA and the innate and adaptive immune systems are of particular interest for their long-lasting impact on wound repair. This review seeks to synthesize the literature on wound healing with a focus on HA's involvement in the immune subsystems.


Assuntos
Ácido Hialurônico/química , Pele/imunologia , Pele/patologia , Cicatrização , Imunidade Adaptativa , Animais , Anti-Inflamatórios , Linfócitos B/imunologia , Linfócitos T CD4-Positivos/imunologia , Células Dendríticas/imunologia , Dissacarídeos/química , Glicosaminoglicanos/química , Humanos , Receptores de Hialuronatos/metabolismo , Sistema Imunitário , Imunidade Inata , Imunossupressores , Inflamação , Linfócitos Intraepiteliais/imunologia , Macrófagos/imunologia , Mastócitos/imunologia , Camundongos , Peso Molecular , Pele/metabolismo , Linfócitos T/imunologia
20.
Biol Chem ; 402(11): 1453-1464, 2021 10 26.
Artigo em Inglês | MEDLINE | ID: mdl-34218538

RESUMO

The delivery of chemotactic signaling molecules via customized biomaterials can effectively guide the migration of cells to improve the regeneration of damaged or diseased tissues. Here, we present a novel biohybrid hydrogel system containing two different sulfated glycosaminoglycans (sGAG)/sGAG derivatives, namely either a mixture of short heparin polymers (Hep-Mal) or structurally defined nona-sulfated tetrahyaluronans (9s-HA4-SH), to precisely control the release of charged signaling molecules. The polymer networks are described in terms of their negative charge, i.e. the anionic sulfate groups on the saccharides, using two parameters, the integral density of negative charge and the local charge distribution (clustering) within the network. The modulation of both parameters was shown to govern the release characteristics of the chemotactic signaling molecule SDF-1 and allows for seamless transitions between burst and sustained release conditions as well as the precise control over the total amount of delivered protein. The obtained hydrogels with well-adjusted release profiles effectively promote MSC migration in vitro and emerge as promising candidates for new treatment modalities in the context of bone repair and wound healing.


Assuntos
Quimiocina CXCL12/metabolismo , Glicosaminoglicanos/metabolismo , Hidrogéis/metabolismo , Quimiocina CXCL12/química , Glicosaminoglicanos/química , Humanos , Hidrogéis/síntese química , Hidrogéis/química , Células-Tronco Mesenquimais/metabolismo , Estrutura Molecular
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA