Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters








Publication year range
1.
Int J Biol Macromol ; 279(Pt 1): 135187, 2024 Nov.
Article in English | MEDLINE | ID: mdl-39216568

ABSTRACT

Periodontal disease (PD) prevention and treatment products typically demonstrate excellent antibacterial activity, but recent studies have raised concerns about their toxicity on oral tissues. Therefore, finding a biocompatible alternative that retains antimicrobial properties is imperative. In this study, a chemically modified hyaluronic acid (HA) hydrogel containing mangostanin (MGTN) was developed. Native HA was chemically modified, incorporating amino and aldehyde groups in different batches of HA, allowing spontaneous crosslinking and gelation when combined at room temperature. MGTN at different concentrations was incorporated before gelation. The structure, swelling characteristics MGTN release, rheological parameters, and in vitro degradation performance of the loaded hydrogel were first evaluated in the study. Then, antimicrobial properties were tested on Porphyromonas gingivalis and its biocompatibility in 3D-engineered human gingiva. HA hydrogel was very stable and showed a sustained release for MGTN for at least 7 days. MGTN-loaded HA hydrogel showed equivalent antimicrobial activity compared to a commercial gel of HA containing 0.2 % chlorhexidine (CHX). In contrast, while MGTN HA hydrogel was biocompatible, CHX gel showed high cytotoxicity, causing cell death and tissue damage. Modified HA hydrogel allows controlled release of MGTN, resulting in a highly biocompatible hydrogel with antibacterial properties. This hydrogel is a suitable alternative therapy to prevent and treat PD.


Subject(s)
Biocompatible Materials , Chlorhexidine , Hyaluronic Acid , Hydrogels , Hyaluronic Acid/chemistry , Hyaluronic Acid/pharmacology , Chlorhexidine/chemistry , Chlorhexidine/pharmacology , Hydrogels/chemistry , Hydrogels/pharmacology , Humans , Biocompatible Materials/chemistry , Biocompatible Materials/pharmacology , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/chemistry , Gingiva/drug effects , Gingiva/cytology , Porphyromonas gingivalis/drug effects , Rheology , Drug Liberation
2.
ACS Omega ; 9(28): 30534-30543, 2024 Jul 16.
Article in English | MEDLINE | ID: mdl-39035935

ABSTRACT

Improved soft tissue integration (STI) around dental implants is key for implant success. The formation of an early and long-lasting transmucosal seal around the implant abutment might help to prevent peri-implantitis, one of the major causes of late implant failure. In natural teeth, collagen fibers are firmly inserted and fixed in the cementum of the tooth and emerge perpendicular to the gingival tissue. In contrast, around dental implants, collagen fibers run predominantly parallel to the implant surface, allowing bacterial migration into the peri-implant interface that might lead to peri-implantitis. Previous studies have shown that nanostructured Ti surfaces improve gingival cell response in monolayer cell cultures. Here, we aimed at evaluating the implant-tissue interface using a 3D gingival tissue equivalent (GTE). First, we evaluated the GTE response to a nanostructured (NN) and machined Ti surface after the stimulation with Porphyromonas gingivalis lipopolysaccharide (LPS), to simulate peri-implantitis conditions. Thus, GTE viability, through MTT assay, the release of metalloproteinase-1 (MMP1) and its inhibitor (TIMP1) through ELISA, and the gene expression of extracellular matrix turnover genes by real-time RT-PCR were analyzed. Second, GTE-implant interaction was characterized by serial block face scanning electron microscopy, and collagen-1 orientation at the tissue-implant interface was analyzed by immunofluorescence. While a similar GTE response to LPS stimulation was found for both implant surfaces, a higher proportion of collagen oriented perpendicular to the implant was observed on the NN implant surface. Thus, our results indicate that the nanostructuration of titanium dental implant abutments could allow the correct orientation of collagen fibers and greater soft tissue sealing, while keeping biocompatibility levels and LPS response comparable.

3.
ACS Pharmacol Transl Sci ; 7(5): 1507-1517, 2024 May 10.
Article in English | MEDLINE | ID: mdl-38751630

ABSTRACT

Skin and oral tissue infections pose significant health challenges worldwide, necessitating the exploration of new antiseptic agents that are both effective and biocompatible. This study evaluated the antibacterial efficacy and biocompatibility of mangostanin (MGTN), a xanthone derived from Garcinia mangostana L., against commercial antiseptics across various bacterial strains (Porphyromonas gingivalis, Streptococcus mutans, Staphylococcus aureus, Staphylococcus epidermidis, Streptococcus pyogenes, and Cutibacterium acnes) and in vitro models of skin and oral tissues. MGTN demonstrated significant antimicrobial activity against all tested pathogens concurrently exhibiting negligible cytotoxic effects on human gingival fibroblasts as well as on three-dimensional (3D) models of human epidermis and oral epithelium. Furthermore, using pooled human saliva, MGTN effectively inhibited plaque biofilm formation, suggesting its potential as a natural, biocompatible antiseptic for skin and oral health applications. These findings position MGTN as a promising candidate for further development into antiseptic formulations, offering a natural alternative to current synthetic options.

4.
Int J Mol Sci ; 24(13)2023 Jun 22.
Article in English | MEDLINE | ID: mdl-37445663

ABSTRACT

Periodontal therapies use immune mediators, but their side effects can increase with dosage. Micro-immunotherapy (MI) is a promising alternative that employs immune regulators at low and ultralow doses to minimize adverse effects. In this study, the effects of 5 capsules and the entire 10-capsule sequence of the sequential MI medicine (MIM-seq) were tested in two in vitro models of periodontitis. Firstly, human gingival fibroblasts (hGFs) exposed to interleukin (IL)-1ß to induce inflammation were treated with five different capsules of MIM-seq for 3 days or with MIM-seq for 24 days. Subsequently, MIM-seq was analyzed in a 3D model of human tissue equivalent of gingiva (GTE) under the same inflammatory stimulus. Simultaneously, a non-IL-1ß-treated control and a vehicle were included. The effects of the treatments on cytotoxicity, collagen deposition, and the secreted levels of IL-1α, IL-6, prostaglandin E2 (PGE2), matrix metalloproteinase-1 (MMP-1), and tissue inhibitor of metalloproteinases-1 (TIMP-1) were evaluated. None of the tested items were cytotoxic. The complete sequence of MIM-seq decreased PGE2 release and restored collagen deposition levels induced by IL-1ß treatment in hGFs exposed to IL-1ß. MIM-seq treatment restored collagen production levels in both models. These promising preclinical findings suggest that MIM-seq should be further investigated for periodontitis treatment.


Subject(s)
Gingiva , Periodontitis , Humans , Dinoprostone/pharmacology , Capsules , Periodontitis/therapy , Collagen/pharmacology , Immunotherapy , Fibroblasts , Cells, Cultured
5.
J Vis Exp ; (192)2023 02 17.
Article in English | MEDLINE | ID: mdl-36876950

ABSTRACT

Periodontitis (PD) is a highly prevalent, chronic immune-inflammatory disease of the periodontium, that results in a loss of gingival soft tissue, periodontal ligament, cementum, and alveolar bone. In this study, a simple method of PD induction in rats is described. We provide detailed instructions for placement of the ligature model around the first maxillary molars (M1) and a combination of injections of lipopolysaccharide (LPS), derived from Porphyromonas gingivalis at the mesio-palatal side of the M1. The induction of periodontitis was maintained for 14 days, promoting the accumulation of bacteria biofilm and inflammation. To validate the animal model, IL-1ß, a key inflammatory mediator, was determined by an immunoassay in the gingival crevicular fluid (GCF), and alveolar bone loss was calculated using cone beam computed tomography (CBCT). This technique was effective in promoting gingiva recession, alveolar bone loss, and an increase in IL-1ß levels in the GCF at the end of the experimental procedure after 14 days. This method was effective in inducing PD, thus being able to be used in studies on disease progression mechanisms and future possible treatments.


Subject(s)
Alveolar Bone Loss , Periodontitis , Animals , Rats , Lipopolysaccharides , Inflammation , Biofilms
6.
Gels ; 8(10)2022 Oct 05.
Article in English | MEDLINE | ID: mdl-36286131

ABSTRACT

Gelatin methacryloyl (GelMA) hydrogels have been widely used for different biomedical applications due to their tunable physical characteristics and appropriate biological properties. In addition, GelMA could be modified with the addition of functional groups providing inherent antibacterial capabilities. Here, GelMA-based hydrogels were developed through the combination of a GelMA unmodified and modified polymer with quaternary ammonium groups (GelMAQ). The GelMAQ was synthesized from GelMA with a low degree of substitution of methacrylamide groups (DSMA) and grafted with glycidyltrimethylammonium chloride in the free amine groups of the lysine moieties present in the original gelatin. GelMAs with high DSMA and GelMAQ were combined 50/50% or 25/75% (w/w), respectively, and compared to controls GelMA and GelMA with added chlorhexidine (CHX) at 0.2%. The different hydrogels were characterized using 1H-NMR spectroscopy and swelling behavior and tested in (1) Porphyromonas gingivalis to evaluate their antibacterial properties and (2) human gingival fibroblast to evaluate their cell biocompatibility and regenerative properties. GelMA/GelMAQ 25/75% showed good antibacterial properties but also excellent biocompatibility and regenerative properties toward human fibroblasts in the wound healing assay. Taken together, these results suggest that the modification of GelMA with quaternary groups could facilitate periodontal tissue regeneration, with good biocompatibility and added antibacterial properties.

7.
Int J Mol Sci ; 23(14)2022 Jul 11.
Article in English | MEDLINE | ID: mdl-35887008

ABSTRACT

Gingival regeneration aims at restoring the architecture and functionality of oral damaged tissue. Different biomaterials or biological materials have been tested for tissue repair, such as platelet concentrates such as PL. In this article, the use of extracellular vesicles (EVs) derived from platelet lysate (PL) and their combination with hyaluronic acid biomaterials (HA) in an in vitro wound healing assay is investigated. EVs were isolated by size exclusion chromatography from PL. In addition, HA gels were formulated with PL or EVs. EVs or HA combined with EVs (HA-EVs) were tested in vitro in gingival fibroblasts and keratinocytes for biocompatibility (LDH activity and metabolic activity) and by an in vitro wound-healing assay and gene expression analysis. EVs and EVs-HA treatments were biocompatible in gingival fibroblasts and keratinocytes and showed an increase in wound healing in vitro compared to control. Moreover, changes in gene expression related to extracellular matrix remodeling were observed after the treatment with EVs. EVs can be combined with HA biomaterials, showing good biocompatibility and preserving their activity and functionality. Therefore, platelet-derived EVs could emerge as a new application for periodontal regeneration in combination with biomaterials in order to enhance their clinical use.


Subject(s)
Extracellular Vesicles , Gingiva , Biocompatible Materials/metabolism , Extracellular Vesicles/metabolism , Fibroblasts , Keratinocytes
8.
Pharmaceutics ; 13(9)2021 Sep 18.
Article in English | MEDLINE | ID: mdl-34575578

ABSTRACT

In the last years, several studies testing commercial periodontal gels that contain chlorhexidine (CHX) or other antibacterial agents, have raised concerns regarding their cytotoxicity in periodontal tissues. We aimed at comparing the biocompatibility but also the efficacy as regards to the antibacterial and wound healing ability of different commercial periodontal gels. In vitro human gingival fibroblasts (GF) and a 3D model of human tissue equivalents of gingiva (GTE) were used under inflammatory conditions to evaluate wound closure, cytotoxicity and gene expression. Antibacterial effects were also investigated on Porphyromonas gingivalis growth, viability and gingipain activity. In GF and in the bacterial study, we found cytotoxic effects on GF and a high inhibition on bacterial growth rate in gels containing CHX, asiaticoside, enoxolone, cetylpyridinium chloride, propolis and eugenol. Of the two gels that were non-cytotoxic, Syntoss Biogel (containing chondrontin sulfate) and Emdogain (EMD, containing amelogenin and propylene glycol alginate), EMD showed the best wound closure, with no effect on P. gingivalis growth but decreased gingipain activity. On the other hand, Syntoss Biogel reduced viability and gingipain activity of P. gingivalis, but lack wound healing capacity. In the 3D GTE, Syntoss Biogel and EMD showed a good biocompatibility. Among all the tested gels, formulations containing CHX, asiaticoside, enoxolone, cetylpyridinium chloride, propolis and eugenol showed high antibacterial effect but also showed high cytotoxicity in eukaryotic cells. EMD was the one with the best biocompatibility and wound healing ability at the conditions tested.

9.
J Periodontol ; 92(10): 1448-1459, 2021 10.
Article in English | MEDLINE | ID: mdl-33393105

ABSTRACT

BACKGROUND: We aimed to evaluate the effect of low doses (LD) bone morphogenetic protein-2 (BMP2) and BMP4 micro-immunotherapy (MI) in two in vitro models of periodontal wound healing/regeneration. METHODS: We first evaluated the effect of LD of BMP2 and BMP4 MI on a 2D cell culture using human gingival fibroblasts (hGF) under inflammatory conditions induced by IL1ß. Biocompatibility, inflammatory response (Prostaglandin E2 (PGE2) release), collagen deposition and release of extracellular matrix (ECM) organization-related enzymes (matrix metalloproteinase-1 (MMP1) and metalloproteinase inhibitor 1 (TIMP1)) were evaluated after short (3 days) and long-term (24 days) treatment with BMP2 or BMP4 MI. Then, given the results obtained in the 2D cell culture, LD BMP4 MI treatment was evaluated in a 3D cell culture model of human tissue equivalent of gingiva (GTE) under the same inflammatory stimulus, evaluating the biocompatibility, inflammatory response and effect on MMP1 and TIMP1 release. RESULTS: LD BMP4 was able to decrease the release of the inflammatory mediator PGE2 and completely re-establish the impaired collagen metabolism induced by IL1ß treatment. In the 3D model, LD BMP4 treatment improved tissue viability compared with the vehicle, with similar levels to 3D tissues without inflammation. No significant effects were observed on PGE2 levels nor MMP1/TIMP1 ratio after LD BMP4 treatment, although a tendency to decrease PGE2 levels was observed after 3 days. CONCLUSIONS: LD BMP4 MI treatment shows anti-inflammatory and regenerative properties on hGF, and improved viability of 3D gingiva under inflammatory conditions. LD BMP4 MI treatment could be used on primary prevention or maintenance care of periodontitis.


Subject(s)
Dinoprostone , Gingiva , Bone Morphogenetic Protein 4 , Cells, Cultured , Collagen , Fibroblasts , Humans , Immunotherapy , Tissue Survival
11.
Sci Rep ; 9(1): 3575, 2019 03 05.
Article in English | MEDLINE | ID: mdl-30837659

ABSTRACT

In the current scenario of high antibiotic resistance, the search for therapeutic options against Pseudomonas aeruginosa must be approached from different perspectives: cell-wall biology as source of bacterial weak points and our immune system as source of weapons. Our recent study suggests that once the permeability barrier has been overcome, the activity of our cell-wall-targeting immune proteins is notably enhanced, more in mutants with impaired peptidoglycan recycling. The present work aims at analyzing the activity of these proteins [lysozyme and Peptidoglycan-Recognition-Proteins (PGLYRPs)], alone or with a permeabilizer (subinhibitory colistin) in clinical strains, along with other features related to the cell-wall. We compared the most relevant and complementary scenarios: acute (bacteremia) and chronic infections [early/late isolates from lungs of cystic fibrosis (CF) patients]. Although a low activity of lysozyme/PGLYRPs per se (except punctual highly susceptible strains) was found, the colistin addition significantly increased their activity regardless of the strains' colistin resistance levels. Our results show increased susceptibility in late CF isolates, suggesting that CF adaptation renders P. aeruginosa more vulnerable to proteins targeting the cell-wall. Thus, our work suggests that attacking some P. aeruginosa cell-wall biology-related elements to increase the activity of our innate weapons could be a promising therapeutic strategy.


Subject(s)
Cell Wall/metabolism , Cytokines/metabolism , Pseudomonas aeruginosa/physiology , Bacteremia/immunology , Bacteremia/metabolism , Cystic Fibrosis/immunology , Cystic Fibrosis/metabolism , Humans , Immunity, Innate , Muramidase/metabolism , beta-Defensins/metabolism
12.
PLoS One ; 12(7): e0181932, 2017.
Article in English | MEDLINE | ID: mdl-28742861

ABSTRACT

Antimicrobial resistance is a continuously increasing threat that severely compromises our antibiotic arsenal and causes thousands of deaths due to hospital-acquired infections by pathogens such as Pseudomonas aeruginosa, situation further aggravated by the limited development of new antibiotics. Thus, alternative strategies such as those targeting bacterial resistance mechanisms, virulence or potentiating the activity of our immune system resources are urgently needed. We have recently shown that mutations simultaneously causing the peptidoglycan recycling blockage and the ß-lactamase AmpC overexpression impair the virulence of P.aeruginosa. These findings suggested that peptidoglycan metabolism might be a good target not only for fighting antibiotic resistance, but also for the attenuation of virulence and/or potentiation of our innate immune weapons. Here we analyzed the activity of the innate immune elements peptidoglycan recognition proteins (PGRPs) and lysozyme against P. aeruginosa. We show that while lysozyme and PGRPs have a very modest basal effect over P. aeruginosa, their bactericidal activity is dramatically increased in the presence of subinhibitory concentrations of the permeabilizing agent colistin. We also show that the P. aeruginosa lysozyme inhibitors seem to play a very residual protective role even in permeabilizing conditions. In contrast, we demonstrate that, once the permeability barrier is overpassed, the activity of lysozyme and PGRPs is dramatically enhanced when inhibiting key peptidoglycan recycling components (such as the 3 AmpDs, AmpG or NagZ), indicating a decisive protective role for cell-wall recycling and that direct peptidoglycan-binding supports, at least partially, the activity of these enzymes. Finally, we show that recycling blockade when occurring simultaneously with AmpC overexpression determines a further decrease in the resistance against PGRP2 and lysozyme, linked to quantitative changes in the cell-wall. Thus, our results help to delineate new strategies against P. aeruginosa infections, simultaneously targeting ß-lactam resistance, cell-wall metabolism and virulence, ultimately enhancing the activity of our innate immune weapons.


Subject(s)
Carrier Proteins/immunology , Cell Membrane Permeability , Immunity, Innate/immunology , Peptidoglycan/metabolism , Pseudomonas aeruginosa/immunology , Anti-Bacterial Agents/pharmacology , Carrier Proteins/metabolism , Colistin/pharmacology , Gene Knockdown Techniques , Microbial Sensitivity Tests , Muramidase/immunology , Muramidase/metabolism , Nod Signaling Adaptor Proteins/metabolism , Pseudomonas aeruginosa/drug effects , Pseudomonas aeruginosa/metabolism , Pseudomonas aeruginosa/pathogenicity
SELECTION OF CITATIONS
SEARCH DETAIL