Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 7.658
Filtrer
1.
Sci Rep ; 14(1): 14109, 2024 06 19.
Article de Anglais | MEDLINE | ID: mdl-38898080

RÉSUMÉ

Developing a reliable method to predict thrombocytopenia is imperative in drug discovery. Here, we establish an assay using a microphysiological system (MPS) to recapitulate the in-vivo mechanisms of platelet aggregation and adhesion. This assay highlights the role of shear stress on platelet aggregation and their interactions with vascular endothelial cells. Platelet aggregation induced by soluble collagen was detected under agitated, but not static, conditions using a plate shaker and gravity-driven flow using MPS. Notably, aggregates adhered on vascular endothelial cells under gravity-driven flow in the MPS, and this incident increased in a concentration-dependent manner. Upon comparing the soluble collagen-induced aggregation activity in platelet-rich plasma (PRP) and whole blood, remarkable platelet aggregate formation was observed at concentrations of 30 µg/mL and 3 µg/mL in PRP and whole blood, respectively. Moreover, ODN2395, an oligonucleotide, induced platelet aggregation and adhesion to vascular endothelial cells. SYK inhibition, which mediated thrombogenic activity via glycoprotein VI on platelets, ameliorated platelet aggregation in the system, demonstrating that the mechanism of platelet aggregation was induced by soluble collagen and oligonucleotide. Our evaluation system partially recapitulated the aggregation mechanisms in blood vessels and can contribute to the discovery of safe drugs to mitigate the risk of thrombocytopenia.


Sujet(s)
Plaquettes , Agrégation plaquettaire , Thrombopénie , Agrégation plaquettaire/effets des médicaments et des substances chimiques , Humains , Thrombopénie/induit chimiquement , Plaquettes/effets des médicaments et des substances chimiques , Plaquettes/métabolisme , Collagène/métabolisme , Collagène/pharmacologie , Cellules endothéliales/effets des médicaments et des substances chimiques , Cellules endothéliales/métabolisme , Adhésivité plaquettaire/effets des médicaments et des substances chimiques , Syk kinase/métabolisme , Syk kinase/antagonistes et inhibiteurs , Plasma riche en plaquettes/métabolisme , Cellules endothéliales de la veine ombilicale humaine/effets des médicaments et des substances chimiques , Systèmes microphysiologiques
2.
Int J Biol Macromol ; 272(Pt 1): 132848, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38830491

RÉSUMÉ

Collagen-based (COL) hydrogels could be a promising treatment option for injuries to the articular cartilage (AC) becuase of their similarity to AC native extra extracellular matrix. However, the high hydration of COL hydrogels poses challenges for AC's mechanical properties. To address this, we developed a hydrogel platform that incorporating cellulose nanocrystals (CNCs) within COL and followed by plastic compression (PC) procedure to expel the excessive fluid out. This approach significantly improved the mechanical properties of the hydrogels and enhanced the chondrogenic differentiation of mesenchymal stem cells (MSCs). Radially confined PC resulted in higher collagen fibrillar densities together with reducing fibril-fibril distances. Compressed hydrogels containing CNCs exhibited the highest compressive modulus and toughness. MSCs encapsulated in these hydrogels were initially affected by PC, but their viability improved after 7 days. Furthermore, the morphology of the cells and their secretion of glycosaminoglycans (GAGs) were positively influenced by the compressed COL-CNC hydrogel. Our findings shed light on the combined effects of PC and CNCs in improving the physical and mechanical properties of COL and their role in promoting chondrogenesis.


Sujet(s)
Différenciation cellulaire , Cellulose , Chondrogenèse , Collagène , Hydrogels , Cellules souches mésenchymateuses , Nanoparticules , Cellules souches mésenchymateuses/cytologie , Cellules souches mésenchymateuses/effets des médicaments et des substances chimiques , Cellules souches mésenchymateuses/métabolisme , Cellulose/composition chimique , Cellulose/pharmacologie , Chondrogenèse/effets des médicaments et des substances chimiques , Différenciation cellulaire/effets des médicaments et des substances chimiques , Nanoparticules/composition chimique , Collagène/composition chimique , Collagène/pharmacologie , Hydrogels/composition chimique , Hydrogels/pharmacologie , Animaux , Matières plastiques/composition chimique , Matières plastiques/pharmacologie , Survie cellulaire/effets des médicaments et des substances chimiques , Glycosaminoglycanes/métabolisme , Cartilage/cytologie , Cartilage/effets des médicaments et des substances chimiques
3.
Nutrients ; 16(11)2024 May 21.
Article de Anglais | MEDLINE | ID: mdl-38892477

RÉSUMÉ

BACKGROUND: Our objective was to conduct a systematic review of the effects of hydrolyzed collagen supplementation on the proliferation and activation of fibroblasts. METHODS: The search was conducted for journals that published articles in the English language, peer-reviewed, meeting the following criteria: (a) randomized clinical trials, (b) randomized studies in animals or humans, (c) in vitro studies, (d) studies using hydrolyzed collagens or collagen peptides, and (e) studies assessing alterations on fibroblasts as the primary or secondary outcome. We utilized the main journal databases PubMed/Web of Science and ongoing reviews by PROSPERO. For bias risk and methodological quality, we used an adaptation of the Downs and Black checklist. Our review followed the PRISMA checklist, conducted from February 2024 to the first week of March 2024, by two independent researchers (P.A.Q.I. and R.P.V.). RESULTS: Eleven studies were included in this review, where our findings reinforce the notion that hydrolyzed collagens or collagen peptides at concentrations of 50-500 µg/mL are sufficient to stimulate fibroblasts in human and animal tissues without inducing toxicity. Different enzymatic processes may confer distinct biological properties to collagens, allowing for scenarios favoring fibroblast promotion or antioxidant effects. Lastly, collagens with lower molecular weights exhibit greater bioavailability to adjacent tissues. CONCLUSIONS: Hydrolyzed collagens or collagen peptides with molecular sizes ranging from <3 to 3000 KDa promote the stimulation of fibroblasts in human tissues.


Sujet(s)
Collagène , Compléments alimentaires , Fibroblastes , Collagène/pharmacologie , Humains , Fibroblastes/effets des médicaments et des substances chimiques , Animaux , Prolifération cellulaire/effets des médicaments et des substances chimiques , Hydrolyse
4.
Int J Nanomedicine ; 19: 3991-4005, 2024.
Article de Anglais | MEDLINE | ID: mdl-38720939

RÉSUMÉ

Purpose: Surgical site infections pose a significant challenge for medical services. Systemic antibiotics may be insufficient in preventing bacterial biofilm development. With the local administration of antibiotics, it is easier to minimize possible complications, achieve drugs' higher concentration at the injured site, as well as provide their more sustained release. Therefore, the main objective of the proposed herein studies was the fabrication and characterization of innovative hydrogel-based composites for local vancomycin (VAN) therapy. Methods: Presented systems are composed of ionically gelled chitosan particles loaded with vancomycin, embedded into biomimetic collagen/chitosan/hyaluronic acid-based hydrogels crosslinked with genipin and freeze-dried to serve in a flake/disc-like form. VAN-loaded carriers were characterized for their size, stability, and encapsulation efficiency (EE) using dynamic light scattering technique, zeta potential measurements, and UV-Vis spectroscopy, respectively. The synthesized composites were tested in terms of their physicochemical and biological features. Results: Spherical structures with sizes of about 200 nm and encapsulation efficiencies reaching values of approximately 60% were obtained. It was found that the resulting particles exhibit stability over time. The antibacterial activity of the developed materials against Staphylococcus aureus was established. Moreover, in vitro cell culture study revealed that the surfaces of all prepared systems are biocompatible as they supported the proliferation and adhesion of the model MG-63 cells. In addition, we have demonstrated significantly prolonged VAN release while minimizing the initial burst effect for the composites compared to bare nanoparticles and verified their desired physicochemical features during swellability, and degradation experiments. Conclusion: It is expected that the developed herein system will enable direct delivery of the antibiotic at an exposed to infections surgical site, providing drugs sustained release and thus will reduce the risk of systemic toxicity. This strategy would both inhibit biofilm formation and accelerate the healing process.


Sujet(s)
Antibactériens , Chitosane , Hydrogels , Staphylococcus aureus , Vancomycine , Vancomycine/composition chimique , Vancomycine/pharmacologie , Vancomycine/administration et posologie , Vancomycine/pharmacocinétique , Antibactériens/composition chimique , Antibactériens/pharmacologie , Antibactériens/administration et posologie , Hydrogels/composition chimique , Hydrogels/pharmacologie , Staphylococcus aureus/effets des médicaments et des substances chimiques , Humains , Chitosane/composition chimique , Acide hyaluronique/composition chimique , Acide hyaluronique/pharmacologie , Vecteurs de médicaments/composition chimique , Collagène/composition chimique , Collagène/pharmacologie , Taille de particule , Libération de médicament , Infection de plaie opératoire/prévention et contrôle , Infection de plaie opératoire/traitement médicamenteux , Tests de sensibilité microbienne , Biofilms/effets des médicaments et des substances chimiques
5.
ACS Biomater Sci Eng ; 10(6): 3984-3993, 2024 06 10.
Article de Anglais | MEDLINE | ID: mdl-38728538

RÉSUMÉ

Guided bone regeneration (GBR) membranes that reside at the interface between the bone and soft tissues for bone repair attract increasing attention, but currently developed GBR membranes suffer from relatively poor osteogenic and antibacterial effects as well as limited mechanical property and biodegradability. We present here the design and fabrication of a bifunctional Janus GBR membrane based on a shear flow-driven layer by a layer self-assembly approach. The Janus GBR membrane comprises a calcium phosphate-collagen/polyethylene glycol (CaP@COL/PEG) layer and a chitosan/poly(acrylic acid) (CHI/PAA) layer on different sides of a collagen membrane to form a sandwich structure. The membrane exhibits good mechanical stability and tailored biodegradability. It is found that the CaP@COL/PEG layer and CHI/PAA layer contribute to the osteogenic differentiation and antibacterial function, respectively. In comparison with the control group, the Janus GBR membrane displays a 2.52-time and 1.84-time enhancement in respective volume and density of newly generated bone. The greatly improved bone repair ability of the Janus GBR membrane is further confirmed through histological analysis, and it has great potential for practical applications in bone tissue engineering.


Sujet(s)
Antibactériens , Régénération osseuse , Ostéogenèse , Régénération osseuse/effets des médicaments et des substances chimiques , Antibactériens/pharmacologie , Antibactériens/composition chimique , Ostéogenèse/effets des médicaments et des substances chimiques , Animaux , Chitosane/composition chimique , Chitosane/pharmacologie , Phosphates de calcium/composition chimique , Phosphates de calcium/pharmacologie , Membrane artificielle , Collagène/composition chimique , Collagène/pharmacologie , Polyéthylène glycols/composition chimique , Polyéthylène glycols/pharmacologie , Régénération tissulaire guidée/méthodes , Ingénierie tissulaire/méthodes , Différenciation cellulaire/effets des médicaments et des substances chimiques
6.
Mol Biol Rep ; 51(1): 482, 2024 Apr 05.
Article de Anglais | MEDLINE | ID: mdl-38578512

RÉSUMÉ

BACKGROUND: Natural bone grafts are the highly preferred materials for restoring the lost bone, while being constrained of donor availability and risk of disease transmission. As a result, tissue engineering is emerging as an efficacious and competitive technique for bone repair. Bone tissue engineering (TE) scaffolds to support bone regeneration and devoid of aforesaid limitations are being vastly explored and among these the avian eggshell membrane has drawn attention for TE owing to its low immunogenicity, similarity with the extracellular matrix, and easy availability. METHODOLOGY AND RESULTS: In this study, the development of bone ingrowth support system from avian eggshell membrane derived collagen hydrolysates (Col-h) is reported. The hydrolysate, cross-linked with glutaraldehyde, was developed into hydrogels with poly-(vinyl alcohol) (PVA) by freeze-thawing and further characterized with ATR-FTIR, XRD, FESEM. The biodegradability, swelling, mechanical, anti-microbial, and biocompatibility evaluation were performed further for the suitability in bone regeneration. The presence of amide I, amide III, and -OH functional groups at 1639 cm- 1,1264 cm- 1, and 3308 cm- 1 respectively and broad peak between 16°-21° (2θ) in XRD data reinstated the composition and form. CONCLUSIONS: The maximum ratio of Col-h/PVA that produced well defined hydrogels was 50:50. Though all the hydrogel matrices alluded towards their competitive attributes and applicability towards restorative bone repair, the hydrogel with 40:60 ratios showed better mechanical strength and cell proliferation than its counterparts. The prominent E. coli growth inhibition by the hydrogel matrices was also observed, along with excellent biocompatibility with MG-63 osteoblasts. The findings indicate strongly the promising application of avian eggshell-derived Col-h in supporting bone regeneration.


Sujet(s)
Coquille de l'oeuf , Escherichia coli , Animaux , Collagène/pharmacologie , Structures d'échafaudage tissulaires , Ingénierie tissulaire/méthodes , Hydrogels , Régénération osseuse , Amides
7.
Zhongguo Zhong Yao Za Zhi ; 49(3): 789-797, 2024 Feb.
Article de Chinois | MEDLINE | ID: mdl-38621883

RÉSUMÉ

This study aims to investigate the effect and mechanism of Fuyu Decoction(FYD) in the treatment of myocardial fibrosis in the rat model of heart failure(HF). Sixty Wistar rats were randomized into a modeling group(n=50) and a sham group(n=10). A post-myocardial infarction HF model was established by ligating the left anterior descending coronary artery in rats. The successfully modeled rats were assigned into model, low-dose(2.5 g·kg~(-1)) FYD(FYD-L), high-dose(5.0 g·kg~(-1)) FYD(FYD-H), and FYD+Nrf2 inhibitor(ML385, 30 mg·kg~(-1)) groups(n=10). FYD was administrated by gavage and ML385 by intraperitoneal injection. The rats in the sham and model groups were administrated with equal amounts of normal saline by gavage. After 8 weeks of intervention, the cardiac function indicators were measured, and the myocardial tissue morphology and collagen deposition were observed. The positive expression of collagens Ⅰ and Ⅲ, apoptosis, and oxidative stress were examined, and the levels of Fe~(2+) and reactive oxygen species(ROS) were determined. The protein levels of nuclear factor erythroid 2-related factor 2(Nrf2), solute carrier family 7 member 11(SLC7A11), glutathione peroxidase 4(GPX4), and acyl-coenzyme A synthase long chain family member 4(ACSL4) in the myocardial tissue were determined. Compared with sham group, the model group showed decreased left ventricular ejection fraction(LVEF) and left ventricular fractional shortening(LVFS), increased left ventricular end internal dimension in systole(LVIDs), left ventricular internal diameter in diastole(LVIDd), and myocardial collagen deposition, positive expression of collagens Ⅰ and Ⅲ, elevated apoptosis rate and malondialdehyde(MDA), Fe~(2+), and ROS levels, lowered superoxide dismutase(SOD) and glutathione peroxidase(GSH) levels, down-regulated protein levels of Nrf2, SLC7A11, and GPX4, and up-regulated protein level of ACSL4. Compared with the model group, the above indicators were restored by FYD. Moreover, ML385 reversed the protective effect of FYD on myocardial fibrosis in HF rats. In conclusion, FYD can inhibit ferroptosis by activating the Nrf2/GPX4 pathway, thereby ameliorating myocardial fibrosis in HF rats.


Sujet(s)
Ferroptose , Défaillance cardiaque , Rats , Animaux , Rat Sprague-Dawley , Facteur-2 apparenté à NF-E2/génétique , Facteur-2 apparenté à NF-E2/métabolisme , Débit systolique , Espèces réactives de l'oxygène , Fonction ventriculaire gauche , Rat Wistar , Défaillance cardiaque/traitement médicamenteux , Fibrose , Collagène/pharmacologie
8.
J Wound Care ; 33(Sup4a): cxi-cxvii, 2024 Apr 02.
Article de Anglais | MEDLINE | ID: mdl-38588055

RÉSUMÉ

OBJECTIVE: Scar tissue formation, as a normal part of wound healing, initiates in the proliferation phase, continues after the remodelling phase, and may cause an unpleasant appearance or disruption in normal functioning. This study investigated the effects of a topical gel on acute wound healing and reducing scars in a rat model. METHOD: ChitoScar (ChitoTech Company, Iran), a commercial scar-reducing gel based on chitosan, was analysed for antibacterial and antiviral activity through a quantitative suspension test. Its cytotoxic effect was investigated, and then irritation and delayed-type hypersensitivity tests were carried out on rabbits through direct application of the gel. Furthermore, the effect of the chitosan-based gel on wound healing and scar tissue formation was studied in rats with an acute wound in two groups: the treatment group (topical application of the chitosan-based gel); and the control group (without treatment). Histopathological examination was carried out based on the inflammatory cells, collagen fibre, keratinocytes and fibroblasts. RESULTS: Analysis revealed that the chitosan-based gel had no cytotoxicity and caused no erythema, oedema, local or other systemic adverse response. Wound healing occurred earlier in the treatment group, which was a result of a significant increase in re-epithelialisation, angiogenesis, fibroblast population and collagen fibre thickness (p<0.05). In the treatment group, wounds healed completely after 21 days and scars totally disappeared after 28 days, while in the control group, wound healing remained incomplete with distinct scar tissue. CONCLUSION: The results demonstrated the positive effect of the chitosan-based gel on the duration and quality of the wound healing process, as well as minimising the scar tissue formation in this in vivo study.


Sujet(s)
Chitosane , Cicatrice , Rats , Lapins , Animaux , Chitosane/pharmacologie , Chitosane/usage thérapeutique , Cicatrisation de plaie , Peau , Collagène/pharmacologie
9.
Biomed Pharmacother ; 174: 116515, 2024 May.
Article de Anglais | MEDLINE | ID: mdl-38569276

RÉSUMÉ

Mesenchymal stem cell exosome (MSCs-exo) is a class of products secreted by mesenchymal stem cells (MSCs) that contain various biologically active substances. MSCs-exo is a promising alternative to MSCs due to their lower immunogenicity and lack of ethical constraints. Ginsenoside Rh2 (Rh2) is a hydrolyzed component of the primary active substance of ginsenosides. Rh2 has a variety of pharmacological functions, including anti-inflammatory, anti-tumor, and antioxidant. Studies have demonstrated that gut microbiota and metabolites are critical in developing rheumatoid arthritis (RA). In this study, we constructed a collagen-induced arthritis (CIA) model in rats. We used MSCs-exo combined with Rh2 to treat CIA rats. To observe the effect of MSCs-exo combined with Rh2 on joint inflammation, rat feces were collected for 16 rRNA amplicon sequencing and untargeted metabolomics analysis. The results showed that the arthritis index score and joint swelling of CIA rats treated with MSCs-exo in combination with Rh2 were significantly lower than those of the model and MSCs-exo alone groups. MSCs-exo and Rh2 significantly ameliorated the disturbed gut microbiota in CIA rats. The regulation of Candidatus_Saccharibacteria and Clostridium_XlVb regulation may be the most critical. Rh2 enhanced the therapeutic effect of MSCs-exo compared with the MSCs-exo -alone group. Furthermore, significant changes in gut metabolites were observed in the CIA rat group, and these differentially altered metabolites may act as messengers for host-microbiota interactions. These differential metabolites were enriched into relevant critical metabolic pathways, revealing possible pathways for host-microbiota interactions.


Sujet(s)
Arthrite expérimentale , Microbiome gastro-intestinal , Ginsénosides , Cellules souches mésenchymateuses , Animaux , Humains , Mâle , Rats , Arthrite expérimentale/induit chimiquement , Arthrite expérimentale/traitement médicamenteux , Arthrite expérimentale/microbiologie , Arthrite expérimentale/thérapie , Polyarthrite rhumatoïde/induit chimiquement , Polyarthrite rhumatoïde/traitement médicamenteux , Polyarthrite rhumatoïde/microbiologie , Polyarthrite rhumatoïde/thérapie , Exosomes/métabolisme , Microbiome gastro-intestinal/effets des médicaments et des substances chimiques , Ginsénosides/pharmacologie , Cellules souches mésenchymateuses/métabolisme , Cellules souches mésenchymateuses/effets des médicaments et des substances chimiques , Cordon ombilical , Collagène/métabolisme , Collagène/pharmacologie
10.
Sci Rep ; 14(1): 8729, 2024 04 16.
Article de Anglais | MEDLINE | ID: mdl-38622264

RÉSUMÉ

Pirfenidone (PFD), one acceptable medication for treating idiopathic pulmonary fibrosis (IPF), is not well tolerated by patients at full doses. Hence, employing of some approaches such as combination therapy may be applicable for increasing therapeutic efficacy of PFD. Losartan (LOS), an angiotensin II receptor antagonist, could be a suitable candidate for combination therapy because of its stabilizing effect on the pulmonary function of IPF patients. Therefore, this study aimed to investigate the effects of LOS in combination with PFD on bleomycin (BLM)-induced lung fibrosis in rats. BLM-exposed rats were treated with LOS alone or in combination with PFD. The edema, pathological changes, level of transforming growth factor-ß (TGF-ß1), collagen content, and oxidative stress parameters were assessed in the lung tissues. Following BLM exposure, the inflammatory response, collagen levels, and antioxidant markers in rat lung tissues were significantly improved by PFD, and these effects were improved by combination with LOS. The findings of this in vivo study suggest that the combined administration of PFD and LOS may provide more potent protection against IPF than single therapy through boosting its anti-inflammatory, anti-fibrotic, and anti-oxidant effects. These results hold promise in developing a more effective therapeutic strategy for treating of lung fibrosis.


Sujet(s)
Fibrose pulmonaire idiopathique , Losartan , Pyridones , Humains , Rats , Animaux , Losartan/pharmacologie , Losartan/usage thérapeutique , Bléomycine/toxicité , Poumon/anatomopathologie , Fibrose pulmonaire idiopathique/induit chimiquement , Fibrose pulmonaire idiopathique/traitement médicamenteux , Fibrose pulmonaire idiopathique/anatomopathologie , Antioxydants/pharmacologie , Facteur de croissance transformant bêta-1/pharmacologie , Collagène/pharmacologie
11.
Int J Biol Macromol ; 266(Pt 2): 131277, 2024 May.
Article de Anglais | MEDLINE | ID: mdl-38565366

RÉSUMÉ

Bacteria-infected wound healing has attracted widespread attention in biomedical engineering. Wound dressing is a potential strategy for repairing infectious wounds. However, the development of wound dressing with appropriate physiochemical, antibacterial, and hemostatic properties, remains challenging. Hence, there is a motivation to develop new synthetic dressings to improve bacteria-infected wound healing. Here, we fabricate a biocompatible sponge through the covalent crosslinking of collagen (Col), quaternized chitosan (QCS), and graphene oxide (GO). The resulting Col-QCS-GO sponge shows an elastic modulus of 1.93-fold higher than Col sponge due to enhanced crosslinking degree by GO incorporation. Moreover, the fabricated Col-QCS-GO sponge shows favorable porosity (84.30 ± 3.12 %), water absorption / retention (2658.0 ± 113.4 % / 1114.0 ± 65.7 %), and hemostasis capacities (blood loss <50.0 mg). Furthermore, the antibacterial property of the Col-QCS-GO sponge under near-infrared (NIR) irradiation is significantly enhanced (the inhibition rates are 99.9 % for S. aureus and 99.9 % for E. coli) due to the inherent antibacterial properties of QCS and the photothermal antibacterial capabilities of GO. Finally, the Col-QCS-GO+NIR sponge exhibits the lowest percentage of wound area (9.05 ± 1.42 %) at day 14 compared to the control group (31.61 ± 1.76 %). This study provides new insights for developing innovative sponges for bacteria-infected wound healing.


Sujet(s)
Antibactériens , Chitosane , Graphite , Hémostatiques , Cicatrisation de plaie , Animaux , Rats , Antibactériens/pharmacologie , Antibactériens/composition chimique , Bandages , Matériaux biocompatibles/composition chimique , Matériaux biocompatibles/pharmacologie , Chitosane/composition chimique , Chitosane/pharmacologie , Collagène/composition chimique , Collagène/pharmacologie , Escherichia coli/effets des médicaments et des substances chimiques , Graphite/composition chimique , Graphite/pharmacologie , Hémostase/effets des médicaments et des substances chimiques , Hémostatiques/pharmacologie , Hémostatiques/composition chimique , Porosité , Staphylococcus aureus/effets des médicaments et des substances chimiques , Cicatrisation de plaie/effets des médicaments et des substances chimiques
12.
Biomater Adv ; 160: 213847, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38657288

RÉSUMÉ

Three-dimensional (3D) organoid models have been instrumental in understanding molecular mechanisms responsible for many cellular processes and diseases. However, established organic biomaterial scaffolds used for 3D hydrogel cultures, such as Matrigel, are biochemically complex and display significant batch variability, limiting reproducibility in experiments. Recently, there has been significant progress in the development of synthetic hydrogels for in vitro cell culture that are reproducible, mechanically tuneable, and biocompatible. Self-assembling peptide hydrogels (SAPHs) are synthetic biomaterials that can be engineered to be compatible with 3D cell culture. Here we investigate the ability of PeptiGel® SAPHs to model the mammary epithelial cell (MEC) microenvironment in vitro. The positively charged PeptiGel®Alpha4 supported MEC viability, but did not promote formation of polarised acini. Modifying the stiffness of PeptiGel® Alpha4 stimulated changes in MEC viability and changes in protein expression associated with altered MEC function, but did not fully recapitulate the morphologies of MECs grown in Matrigel. To supply the appropriate biochemical signals for MEC organoids, we supplemented PeptiGels® with laminin. Laminin was found to require negatively charged PeptiGel® Alpha7 for functionality, but was then able to provide appropriate signals for correct MEC polarisation and expression of characteristic proteins. Thus, optimisation of SAPH composition and mechanics allows tuning to support tissue-specific organoids.


Sujet(s)
Techniques de cultures cellulaires tridimensionnelles , Collagène , Association médicamenteuse , Cellules épithéliales , Hydrogels , Laminine , Peptides , Protéoglycanes , Laminine/pharmacologie , Laminine/composition chimique , Hydrogels/composition chimique , Hydrogels/pharmacologie , Protéoglycanes/pharmacologie , Protéoglycanes/composition chimique , Collagène/composition chimique , Collagène/pharmacologie , Peptides/pharmacologie , Peptides/composition chimique , Cellules épithéliales/effets des médicaments et des substances chimiques , Cellules épithéliales/cytologie , Humains , Femelle , Techniques de cultures cellulaires tridimensionnelles/méthodes , Survie cellulaire/effets des médicaments et des substances chimiques , Matériaux biocompatibles/composition chimique , Matériaux biocompatibles/pharmacologie , Glandes mammaires humaines/cytologie , Organoïdes/effets des médicaments et des substances chimiques , Organoïdes/cytologie , Techniques de culture cellulaire/méthodes
13.
Int J Biol Macromol ; 270(Pt 1): 131886, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38677696

RÉSUMÉ

Type V collagen is an essential component of the extracellular matrix (ECM), and its remodeling releases specific protein fragments that can specifically inhibit endothelial cell responses such as proliferation, migration, and invasion. In this study, we have successfully constructed two engineered strains of Pichia pastoris capable of producing recombinant collagen through a new genetic engineering approach. Through high-density fermentation, the expression of 1605 protein and 1610 protein could reach 2.72 g/L and 4.36 g/L. With the increase of repetition times, the yield also increased. Bioactivity analysis showed that recombinant collagen could block the angiogenic effect of FGF-2 on endothelial cells by eliminating FGF-2-induced endothelial cell migration and invasion. Collectively, the recombinant proteins we successfully expressed have a wide range of potential for inhibiting angiogenesis in the biomaterials and biomedical fields.


Sujet(s)
Protéines recombinantes , Protéines recombinantes/pharmacologie , Protéines recombinantes/génétique , Humains , Collagène/composition chimique , Collagène/pharmacologie , Mouvement cellulaire/effets des médicaments et des substances chimiques , Séquences répétées d'acides aminés , Séquence d'acides aminés , Cellules endothéliales de la veine ombilicale humaine/effets des médicaments et des substances chimiques , Inhibiteurs de l'angiogenèse/pharmacologie , Inhibiteurs de l'angiogenèse/composition chimique , Facteur de croissance fibroblastique de type 2/génétique , Facteur de croissance fibroblastique de type 2/pharmacologie , Facteur de croissance fibroblastique de type 2/métabolisme , Facteur de croissance fibroblastique de type 2/composition chimique , Expression des gènes , Fermentation , Saccharomycetales/génétique , Saccharomycetales/métabolisme
14.
Biomater Adv ; 159: 213813, 2024 May.
Article de Anglais | MEDLINE | ID: mdl-38428122

RÉSUMÉ

The ability of human tissues to self-repair is limited, which motivates the scientific community to explore new and better therapeutic approaches to tissue regeneration. The present manuscript provides a comparative study between a marine-based composite biomaterial, and another composed of well-established counterparts for bone tissue regeneration. Blue shark skin collagen was combined with bioapatite obtained from blue shark's teeth (mColl:BAp), while bovine collagen was combined with synthetic hydroxyapatite (bColl:Ap) to produce 3D composite scaffolds by freeze-drying. Collagens showed similar profiles, while apatite particles differed in their composition, being the marine bioapatite a fluoride-enriched ceramic. The marine-sourced biomaterials presented higher porosities, improved mechanical properties, and slower degradation rates when compared to synthetic apatite-reinforced bovine collagen. The in vivo performance regarding bone tissue regeneration was evaluated in defects created in femoral condyles in New Zealand rabbits twelve weeks post-surgery. Micro-CT results showed that mColl:BAp implanted condyles had a slower degradation and an higher tissue formation (17.9 ± 6.9 %) when compared with bColl:Ap implanted ones (12.9 ± 7.6 %). The histomorphometry analysis provided supporting evidence, confirming the observed trend by quantifying 13.1 ± 7.9 % of new tissue formation for mColl:BAp composites and 10.4 ± 3.2 % for bColl:Ap composites, suggesting the potential use of marine biomaterials for bone regeneration.


Sujet(s)
Matériaux biocompatibles , Structures d'échafaudage tissulaires , Humains , Animaux , Lapins , Bovins , Matériaux biocompatibles/usage thérapeutique , Apatites , Régénération osseuse , Collagène/pharmacologie
15.
Int J Biol Macromol ; 265(Pt 1): 130843, 2024 Apr.
Article de Anglais | MEDLINE | ID: mdl-38484819

RÉSUMÉ

BACKGROUND: Stem cell exosomes are beneficial in accelerating wound repair. However, the therapeutic function is limited due to its rapid clearance in vivo. To improve the functionality of exosomes in cutaneous wound healing, a novel hydrogel was designed and fabricated by recombinant human collagen I and carboxymethyl chitosan loaded with exosomes derived from human umbilical cord mesenchymal stem cells (hUCMSCs), named as the rhCol I/CMC-Exos hydrogel. METHODS: Exosomes were extracted from hUCMSCs and were characterizated by TEM (Transmission Electron Microscopy), and biomarker detection. The rhCol I hydrogel, rhCol I/carboxymethyl chitosan (rhCol I/CMC) hydrogel and the rhCol I/CMC-Exos hydrogel composites were cross-linked by genipin. These materials were assessed and compared for their physical characteristics, including cross-sectional morphology, porosity, pore distribution, and hydrophilicity. Cell biocompatibility on biomaterials was investigated using scanning electron microscopy and CFDA staining, as well as assessed in vivo through histological examination of major organs in mice. Effects of the hydrogel composite on wound healing were further evaluated by using the full-thickness skin defect mice model. RESULTS: Successful extraction of hUCMSCs-derived exosomes was confirmed by TEM,Western Blotting and flow cytometry. The synthesized rhCol I/CMC-Exos hydrogel composite exhibited cytocompatibility and promoted cell growth in vitro. The rhCol I/CMC-Exos hydrogel showed sustained release of exosomes. In the mice full skin-defects model, the rhCol I/CMC-Exos-treated group showed superior wound healing efficiency, with 15 % faster wound closure compared to controls. Histological examinations revealed thicker dermis formation and more balanced collagen deposition in wounds treated with rhCol I/CMC-Exos hydrogel. Mechanistically, the application of rhCol I/CMC-Exos hydrogel increased fibroblasts proliferation, alleviated inflammation responses as well as promoted angiogenesis, thereby was beneficial in promoting skin wound healing and regeneration. CONCLUSION: Our study, for the first time, introduced recombinant human Collagen I in fabricating a novel hydrogel loaded with hUCMSCs-derived exosomes, which effectively promoted skin wound closure and regeneration, demonstrating a great potential in severe skin wound healing treatment.


Sujet(s)
Chitosane , Exosomes , Cellules souches mésenchymateuses , Humains , Souris , Animaux , Hydrogels/pharmacologie , Cicatrisation de plaie , Chitosane/pharmacologie , Études transversales , Collagène/pharmacologie , Modèles animaux de maladie humaine , Collagène de type I/pharmacologie
16.
Bioorg Chem ; 146: 107286, 2024 May.
Article de Anglais | MEDLINE | ID: mdl-38537336

RÉSUMÉ

Pulmonary fibrosis (PF) poses a significant challenge with limited treatment options and a high mortality rate of approximately 45 %. Qingkailing Granule (QKL), derived from the Angong Niuhuang Pill, shows promise in addressing pulmonary conditions. Using a comprehensive approach, combining network pharmacology analysis with experimental validation, this study explores the therapeutic effects and mechanisms of QKL against PF for the first time. In vivo, QKL reduced collagen deposition and suppressed proinflammatory cytokines in a bleomycin-induced PF mouse model. In vitro studies demonstrated QKL's efficacy in protecting cells from bleomycin-induced injury and reducing collagen accumulation and cell migration in TGF-ß1-induced pulmonary fibrosis cell models. Network pharmacology analysis revealed potential mechanisms, confirmed by western blotting, involving the modulation of PI3K/AKT and SRC/STAT3 signaling pathways. Molecular docking simulations highlighted interactions between QKL's active compounds and key proteins, showing inhibitory effects on epithelial damage and fibrosis. Collectively, these findings underscore the therapeutic potential of QKL in alleviating pulmonary inflammation and fibrosis through the downregulation of PI3K/AKT and SRC/STAT3 signaling pathways, with a pivotal role attributed to its active compounds.


Sujet(s)
Médicaments issus de plantes chinoises , Fibrose pulmonaire , Souris , Animaux , Fibrose pulmonaire/induit chimiquement , Fibrose pulmonaire/traitement médicamenteux , Fibrose pulmonaire/métabolisme , Protéines proto-oncogènes c-akt/métabolisme , Phosphatidylinositol 3-kinases/métabolisme , Simulation de docking moléculaire , Transduction du signal , Collagène/métabolisme , Collagène/pharmacologie , Collagène/usage thérapeutique , Fibrose , Bléomycine/effets indésirables
17.
J Biosci ; 492024.
Article de Anglais | MEDLINE | ID: mdl-38516910

RÉSUMÉ

Snake venom L-amino acid oxidases (LAAOs) are flavoenzymes with diverse physiological and pharmacological effects. These enzymes are found to showcase anticoagulant, antiplatelet, cytotoxicity and other biological effects in bite victims. However, the exact mechanism through which they exhibit several biological properties is not yet fully understood. The current study focussed on the purification of cobra venom LAAO and the functional characterization of purified LAAO. A novel L-amino acid oxidase NNLAAO70 with a molecular weight ~70 kDa was purified from the venom of an Indian spectacled cobra (Naja naja). NNLAAO70 showed high substrate specificity for L-His, L-Leu, and L-Arg during its LAAO activity. It inhibited adenosine di-phosphate (ADP) and collagen-induced platelet aggregation process in a dosedependent manner. About 60% inhibition of collagen-induced and 40% inhibition of ADP-induced platelet aggregation was observed with a 40 µg/ml dose of NNLAAO70. NNLAAO70 exhibited bactericidal activity on Bacillus subtilis, Escherichia coli, Bacillus megaterium, and Pseudomonas fluorescens. NNLAAO70 also showed cytotoxicity on A549 cells in vitro. It showed severe bactericidal activity on P. fluorescens and lysed 55% of cells. NNLAAO70 also exhibited drastic cytotoxicity on A549 cells. At 1 lg/ml dosage, it demonstrated a 60% reduction in A549 viability and induced apoptosis upon 24-h incubation. H2O2 released during oxidative deamination reactions played a major role in NNLAAO70-induced cytotoxicity. NNLAAO70 significantly increased intracellular reactive oxygen species (ROS) levels in A549 cells by six fold when compared to untreated cells. Oxidative stress-mediated cell injury is the primary cause of NNLAAO70-induced apoptosis in A549 cells and prolonged oxidative stress caused DNA fragmentation and activated cellular secondary necrosis.


Sujet(s)
Elapidae , Tumeurs , Animaux , Humains , Naja naja , L-Amino acid oxidase/génétique , L-Amino acid oxidase/pharmacologie , L-Amino acid oxidase/composition chimique , Peroxyde d'hydrogène/pharmacologie , Venins des élapidés/pharmacologie , Apoptose , Nécrose , Collagène/pharmacologie , Poumon
18.
Invest Ophthalmol Vis Sci ; 65(3): 36, 2024 Mar 05.
Article de Anglais | MEDLINE | ID: mdl-38551585

RÉSUMÉ

Purpose: Symptomatic vitreous opacifications, so-called floaters, are difficult to objectively assess majorly limiting the possibility of in vitro studies. Forward light scattering was found previously to be increased in eyes with symptomatic floaters. Using an objective setup to measure forward light scattering, we studied the effects of enzymatically digesting the components of the vitreous body on straylight to develop an in vitro model of vitreous opacifications. Methods: Fifty-seven porcine vitreous bodies were digested using hyaluronidase, collagenase, trypsin, and bromelain, as well as using a combination of hyaluronidase + collagenase and hyaluronidase + bromelain. A modified C-Quant setup was used to objectively assess forward light scattering. Results: Depletion of hyaluronic acid majorly increased vitreous straylight (mean increase 34.4 deg2/sr; P = 0.01), whereas primarily digesting the vitreous gel with collagenase or trypsin did not significantly affect straylight. When collagenase or bromelain is applied in hyaluronic acid depleted vitreous gels, the increase in forward light scattering is reversed partially. Conclusions: The age-related loss of hyaluronic acid primarily drives the increase in vitreous gel straylight induced by conglomerates of collagen. This process can be reversed partially by digesting collagen. This in vitro model allows the objective quantification and statistical comparison of straylight burden caused by vitreous opacities and, thus, can serve as a first testing ground for pharmacological therapies, as demonstrated with bromelain.


Sujet(s)
Bromélaïnes , Lumière , Animaux , Suidae , Hyaluronoglucosaminidase/pharmacologie , Acide hyaluronique/pharmacologie , Trypsine , Vieillissement , Collagène/pharmacologie , Collagenases/pharmacologie , Diffusion de rayonnements
19.
PLoS One ; 19(3): e0295104, 2024.
Article de Anglais | MEDLINE | ID: mdl-38478501

RÉSUMÉ

BACKGROUND: Melatonin (MEL) is an indole amine molecule primarily produced in the pineal gland. Melatonin has been shown in numerous studies to have antifibrotic effects on the kidney, liver, and other organs. However, it is still unclear how melatonin works in bladder fibrosis. We explored how melatonin affects animals with bladder fibrosis and the underlying mechanisms. MATERIALS AND METHODS: MEL was used to treat human bladder smooth muscle cells (HBdSMCs) after they were stimulated with transforming growth factor-ß1 (TGF-ß1) in vitro. Proteomic analysis and bioinformatic analysis of the altered expression of these proteins were subsequently performed on HBdSMCs from the different processing methods. To construct an in vivo bladder fibrosis model, we injected protamine sulfate (PS) and lipopolysaccharide (LPS) twice a week into the rat bladder for six weeks. After two weeks of PS/LPS treatment, the mice in the treatment group were treated with MEL (20 mg/kg/d) for 4 weeks. Finally, we detected the expression of fibrosis markers from different perspectives. The TGF-ß1/Smad pathway and epithelial-mesenchymal transition (EMT) in cell and bladder tissues were also identified. Further proteomic analysis was also performed. RESULTS: In vitro, we found that TGF-ß1 treatment enhanced the expression of the fibrosis markers collagen III and α-SMA in HBdSMCs. E-cadherin expression decreased while the TGF-ß1/Smad pathway was activated. Vimentin and N-cadherin expression was also elevated at the same time. Similar findings were observed in the LPS group. After MEL treatment, the expression of collagen III and α-SMA decreased, the expression of E-cadherin increased, and the expression of vimentin and N-cadherin also decreased. According to our quantitative proteomics analysis, CCN1 and SQLE may be important proteins involved in the development of bladder fibrosis. MEL decreased the expression of these genes, leading to the relief of bladder fibrosis. Bioinformatics analysis revealed that the extracellular space structure related to metabolic pathways, actin filament binding, and stress fibers can serve as a pivotal focus in the management of fibrosis. CONCLUSION: Melatonin attenuates bladder fibrosis by blocking the TGF-ß1/Smad pathway and EMT. CCN1 appears to be a possible therapeutic target for bladder fibrosis.


Sujet(s)
Mélatonine , Facteur de croissance transformant bêta-1 , Rats , Humains , Souris , Animaux , Facteur de croissance transformant bêta-1/métabolisme , Vimentine/métabolisme , Mélatonine/pharmacologie , Mélatonine/usage thérapeutique , Transduction du signal , Vessie urinaire/métabolisme , Lipopolysaccharides/pharmacologie , Protéomique , Fibrose , Transition épithélio-mésenchymateuse , Collagène/pharmacologie , Cadhérines/métabolisme
20.
In Vitro Cell Dev Biol Anim ; 60(3): 287-299, 2024 Mar.
Article de Anglais | MEDLINE | ID: mdl-38485818

RÉSUMÉ

The study aimed to investigate the effect of ginsenoside Rg1 on intervertebral disc degeneration (IVDD) in rats and IL-1ß-induced nucleus pulposus (NP) cells, and explore its underlying mechanism. Forty IVDD rat models were divided into the IVDD group, low-dose (L-Rg1) group (intraperitoneal injection of 20 mg/kg/d ginsenoside Rg1), medium-dose (M-Rg1) group (intraperitoneal injection of 40 mg/kg/d ginsenoside Rg1), and high-dose (H-Rg1) group (intraperitoneal injection of 80 mg/kg/d ginsenoside Rg1). The pathological change was observed by HE and safranin O-fast green staining. The expression of IL-1ß, IL-6, TNF-α, MMP3, aggrecan, and collagen II was detected. The expression of NF-κB p65 in IVD tissues was detected. Rat NP cells were induced by IL-1ß to simulate IVDD environment and divided into the control group, IL-1ß group, and 20, 50, and 100 µmol/L Rg1 groups. The cell proliferation activity, the apoptosis, and the expression of IL-6, TNF-α, MMP3, aggrecan, collagen II, and NF-κB pathway-related protein were detected. In IVDD rats, ginsenoside Rg1 improved the pathology of IVD tissues; suppressed the expression of IL-1ß, IL-6, TNF-α, aggrecan, and collagen II; and inhibited the expression of p-p65/p65 and nuclear translocation of p65, to alleviate the IVDD progression. In the IL-1ß-induced NP cells, ginsenoside Rg1 also improved the cell proliferation and inhibited the apoptosis and the expression of IL-6, TNF-α, aggrecan, collagen II, p-p65/p65, and IκK in a dose-dependent manner. Ginsenoside Rg1 alleviated IVDD in rats and inhibited apoptosis, inflammatory response, and ECM degradation in IL-1ß-induced NP cells. And Rg1 may exert its effect via inhibiting the activation of NF-κB signaling pathway.


Sujet(s)
Ginsénosides , Dégénérescence de disque intervertébral , Disque intervertébral , Nucleus pulposus , Animaux , Rats , Agrécanes/génétique , Apoptose , Collagène/pharmacologie , Inflammation/anatomopathologie , Interleukine-6/métabolisme , Disque intervertébral/métabolisme , Disque intervertébral/anatomopathologie , Dégénérescence de disque intervertébral/traitement médicamenteux , Dégénérescence de disque intervertébral/métabolisme , Dégénérescence de disque intervertébral/anatomopathologie , Matrix metalloproteinase 3/métabolisme , Facteur de transcription NF-kappa B/métabolisme , Transduction du signal , Facteur de nécrose tumorale alpha/métabolisme
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...