Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
PeerJ ; 11: e15636, 2023.
Article in English | MEDLINE | ID: mdl-37465155

ABSTRACT

Understanding how environmental drivers influence shark and ray spatial and temporal patterns can provide crucial knowledge for their evidence-based protection and long-term monitoring. However, information on which drivers of variation are most important for elasmobranch communities on soft sediments is limited. Using baited remote underwater stereo-video systems (stereo-BRUVs), we investigated how seasonal and environmental variables affected the elasmobranchs of the iSimangaliso Wetland Park marine protected area (MPA) in South Africa (SA). In total, 11 species were identified from 48 sites between 12 m and 33 m water depth in a sandy habitat. While species richness was similar across seasons, the total abundance of elasmobranchs recorded was higher in winter than summer. The species assemblage composition varied significantly between seasons, with the Human's whaler shark Carcharhinus humani prevalent in summer and the Critically Endangered whitespotted wedgefish Rhynchobatus djiddensis more abundant during winter. Most species were sighted throughout the entire depth range, but rays were more common in shallower waters (< 25 m depth), while C. humani and R. djiddensis were more common in the deeper depth zone of this study. This research provides baseline information about this previously unexplored sandy habitat for elasmobranchs in a site of regional and global significance. Records of species of conservation concern in the sampling area highlight the importance of protecting sand environments within an MPA.


Subject(s)
Sharks , Wetlands , Animals , Ecosystem , Sand , Seasons , South Africa
2.
J Mater Chem B ; 11(17): 3823-3835, 2023 05 03.
Article in English | MEDLINE | ID: mdl-36946228

ABSTRACT

Regenerative endodontics represents a paradigm shift in dental pulp therapy for necrotic young permanent teeth. However, there are still challenges associated with attaining maximum root canal disinfection while supporting angiogenesis and preserving resident stem cells viability and differentiation capacity. Here, we developed a hydrogel system by incorporating antibiotic-eluting fiber-based microparticles in gelatin methacryloyl (GelMA) hydrogel to gather antimicrobial and angiogenic properties while prompting minimum cell toxicity. Minocycline (MINO) or clindamycin (CLIN) was introduced into a polymer solution and electrospun into fibers, which were further cryomilled to attain MINO- or CLIN-eluting fibrous microparticles. To obtain hydrogels with multi-therapeutic effects, MINO- or CLIN-eluting microparticles were suspended in GelMA at distinct concentrations. The engineered hydrogels demonstrated antibiotic-dependent swelling and degradability while inhibiting bacterial growth with minimum toxicity in dental-derived stem cells. Notably, compared to MINO, CLIN hydrogels enhanced the formation of capillary-like networks of endothelial cells in vitro and the presence of widespread vascularization with functioning blood vessels in vivo. Our data shed new light onto the clinical potential of antibiotic-eluting gelatin methacryloyl hydrogel as an injectable scaffold with multi-therapeutic effects to promote antimicrobial disinfection and angiogenesis for regenerative endodontics.


Subject(s)
Anti-Infective Agents , Regenerative Endodontics , Endothelial Cells , Disinfection , Hydrogels/pharmacology , Anti-Bacterial Agents/pharmacology , Clindamycin , Minocycline
3.
Bioact Mater ; 19: 268-281, 2023 Jan.
Article in English | MEDLINE | ID: mdl-35574052

ABSTRACT

Periodontitis is a chronic inflammatory condition that often causes serious damage to tooth-supporting tissues. The limited successful outcomes of clinically available approaches underscore the need for therapeutics that cannot only provide structural guidance to cells but can also modulate the local immune response. Here, three-dimensional melt electrowritten (i.e., poly(ε-caprolactone)) scaffolds with tissue-specific attributes were engineered to guide differentiation of human-derived periodontal ligament stem cells (hPDLSCs) and mediate macrophage polarization. The investigated tissue-specific scaffold attributes comprised fiber morphology (aligned vs. random) and highly-ordered architectures with distinct strand spacings (small 250 µm and large 500 µm). Macrophages exhibited an elongated morphology in aligned and highly-ordered scaffolds, while maintaining their round-shape on randomly-oriented fibrous scaffolds. Expressions of periostin and IL-10 were more pronounced on the aligned and highly-ordered scaffolds. While hPDLSCs on the scaffolds with 500 µm strand spacing show higher expression of osteogenic marker (Runx2) over 21 days, cells on randomly-oriented fibrous scaffolds showed upregulation of M1 markers. In an orthotopic mandibular fenestration defect model, findings revealed that the tissue-specific scaffolds (i.e., aligned fibers for periodontal ligament and highly-ordered 500 µm strand spacing fluorinated calcium phosphate [F/CaP]-coated fibers for bone) could enhance the mimicking of regeneration of natural periodontal tissues.

4.
Dent Mater ; 38(11): 1749-1762, 2022 11.
Article in English | MEDLINE | ID: mdl-36180310

ABSTRACT

OBJECTIVES: Electrospun scaffolds are a versatile biomaterial platform to mimic fibrillar structure of native tissues extracellular matrix, and facilitate the incorporation of biomolecules for regenerative therapies. Self-assembling peptide P11-4 has emerged as a promising strategy to induce mineralization; however, P11-4 application has been mostly addressed for early caries lesions repair on dental enamel. Here, to investigate P11-4's efficacy on bone regeneration, polymeric electrospun scaffolds were developed, and then distinct concentrations of P11-4 were physically adsorbed on the scaffolds. METHODS: P11-4-laden and pristine (P11-4-free) electrospun scaffolds were immersed in simulated body fluid and mineral precipitation identified by SEM. Functional groups and crystalline phases were analyzed by FTIR and XRD, respectively. Cytocompatibility, mineralization, and gene expression assays were conducted using stem cells from human exfoliated deciduous teeth. To investigate P11-4-laden scaffolds potential to induce in vivo mineralization, an established rat calvaria critical-size defect model was used. RESULTS: We successfully synthesized nanofibrous (∼ 500 nm fiber diameter) scaffolds and observed that functionalization with P11-4 did not affect the fibers' diameter. SEM images indicated mineral precipitation, while FTIR and XRD confirmed apatite-like formation and crystallization for P11-4-laden scaffolds. In addition, P11-4-laden scaffolds were cytocompatible, highly stimulated cell-mediated mineral deposition, and upregulated the expression of mineralization-related genes compared to pristine scaffolds. P11-4-laden scaffolds led to enhanced in vivo bone regeneration after 8 weeks compared to pristine PCL. SIGNIFICANCE: Electrospun scaffolds functionalized with P11-4 are a promising strategy for inducing mineralized tissues regeneration in the craniomaxillofacial complex.


Subject(s)
Nanofibers , Tissue Scaffolds , Animals , Apatites , Biocompatible Materials , Bone Regeneration , Humans , Nanofibers/chemistry , Peptides , Polyesters/chemistry , Rats , Tissue Engineering/methods , Tissue Scaffolds/chemistry
5.
Int Mater Rev ; 67(4): 347-384, 2022.
Article in English | MEDLINE | ID: mdl-35754978

ABSTRACT

From a materials perspective, the pillars for the development of clinically translatable scaffold-based strategies for craniomaxillofacial (CMF) bone and periodontal regeneration have included electrospinning and 3D printing (biofabrication) technologies. Here, we offer a detailed analysis of the latest innovations in 3D (bio)printing strategies for CMF bone and periodontal regeneration and provide future directions envisioning the development of advanced 3D architectures for successful clinical translation. First, the principles of electrospinning applied to the generation of biodegradable scaffolds are discussed. Next, we present on extrusion-based 3D printing technologies with a focus on creating scaffolds with improved regenerative capacity. In addition, we offer a critical appraisal on 3D (bio)printing and multitechnology convergence to enable the reconstruction of CMF bones and periodontal tissues. As a future outlook, we highlight future directions associated with the utilization of complementary biomaterials and (bio)fabrication technologies for effective translation of personalized and functional scaffolds into the clinics.

6.
ACS Appl Mater Interfaces ; 13(42): 49642-49657, 2021 Oct 27.
Article in English | MEDLINE | ID: mdl-34637255

ABSTRACT

Periodontitis compromises the integrity and function of tooth-supporting structures. Although therapeutic approaches have been offered, predictable regeneration of periodontal tissues remains intangible, particularly in anatomically complex defects. In this work, personalized and defect-specific antibiotic-laden polymeric scaffolds containing metronidazole (MET), tetracycline (TCH), or their combination (MET/TCH) were created via electrospinning. An initial screening of the synthesized fibers comprising chemo-morphological analyses, cytocompatibility assessment, and antimicrobial validation against periodontopathogens was accomplished to determine the cell-friendly and anti-infective nature of the scaffolds. According to the cytocompatibility and antimicrobial data, the 1:3 MET/TCH formulation was used to obtain three-dimensional defect-specific scaffolds to treat periodontally compromised three-wall osseous defects in rats. Inflammatory cell response and new bone formation were assessed by histology. Micro-computerized tomography was performed to assess bone loss in the furcation area at 2 and 6 weeks post implantation. Chemo-morphological and cell compatibility analyses confirmed the synthesis of cytocompatible antibiotic-laden fibers with antimicrobial action. Importantly, the 1:3 MET/TCH defect-specific scaffolds led to increased new bone formation, lower bone loss, and reduced inflammatory response when compared to antibiotic-free scaffolds. Altogether, our results suggest that the fabrication of defect-specific antibiotic-laden scaffolds holds great potential toward the development of personalized (i.e., patient-specific medication) scaffolds to ablate infection while affording regenerative properties.


Subject(s)
Anti-Bacterial Agents/pharmacology , Metronidazole/pharmacology , Periodontitis/drug therapy , Tetracycline/pharmacology , Tissue Scaffolds/chemistry , Anti-Bacterial Agents/chemistry , Bone Regeneration/drug effects , Fusobacterium nucleatum/drug effects , Materials Testing , Metronidazole/chemistry , Microbial Sensitivity Tests , Particle Size , Periodontitis/microbiology , Periodontitis/pathology , Porphyromonas gingivalis/drug effects , Prevotella intermedia/drug effects , Tetracycline/chemistry
7.
Adv Healthc Mater ; 10(21): e2101152, 2021 11.
Article in English | MEDLINE | ID: mdl-34342173

ABSTRACT

Periodontitis is a chronic inflammatory, bacteria-triggered disorder affecting nearly half of American adults. Although some level of tissue regeneration is realized, its low success in complex cases demands superior strategies to amplify regenerative capacity. Herein, highly ordered scaffolds are engineered via Melt ElectroWriting (MEW), and the effects of strand spacing, as well as the presence of a nanostructured fluorinated calcium phosphate (F/CaP) coating on the adhesion/proliferation, and osteogenic differentiation of human-derived periodontal ligament stem cells, are investigated. Upon initial cell-scaffold interaction screening aimed at defining the most suitable design, MEW poly(ε-caprolactone) scaffolds with 500 µm strand spacing are chosen. Following an alkali treatment, scaffolds are immersed in a pre-established solution to allow for coating formation. The presence of a nanostructured F/CaP coating leads to a marked upregulation of osteogenic genes and attenuated bacterial growth. In vivo findings confirm that the F/CaP-coated scaffolds are biocompatible and lead to periodontal regeneration when implanted in a rat mandibular periodontal fenestration defect model. In aggregate, it is considered that this work can contribute to the development of personalized scaffolds capable of enabling tissue-specific differentiation of progenitor cells, and thus guide simultaneous and coordinated regeneration of soft and hard periodontal tissues, while providing antimicrobial protection.


Subject(s)
Osteogenesis , Tissue Scaffolds , Animals , Periodontium , Polyesters , Rats , Tissue Engineering , Wound Healing
8.
J Mech Behav Biomed Mater ; 116: 104293, 2021 04.
Article in English | MEDLINE | ID: mdl-33588247

ABSTRACT

The aim of this investigation was to engineer metformin (MF)-loaded mesoporous silica nanospheres (MSNs)-laden gelatin methacryloyl (GelMA) photocrosslinkable hydrogels and test their effects on the mechanical properties, swelling ratio, drug release, cytocompatibility, and osteogenic differentiation of stem cells from human exfoliated deciduous teeth (SHEDs). As-received and carboxylated MSNs (MSNs-COOH) were characterized by scanning and transmission electron microscopies (SEM and TEM), as well as Fourier-transform infrared spectroscopy (FTIR) prior to hydrogel modification. MF-MSNs-COOH were obtained by loading MF into MSNs at a 1:1 mass ratio. Upon MSNs-COOH laden-hydrogels fabrication, the mechanical properties, swelling ratio and MF release were evaluated. SHEDs were seeded on the hydrogels and cytocompatibility was examined. The effects of the MF-MSNs-COOH/GelMA on the osteogenic differentiation of SHEDs were measured by ALP activity, Alizarin Red assay, and Real-time PCR. Statistics were performed using one-way ANOVA (α = 0.05). Morphological (SEM and TEM) analyses of pristine and carboxylated MSNs revealed a mean particle size of 200 nm and 218 nm, respectively. Importantly, an intrinsic nanoporous structure was noticed. Incorporation of MSNs-COOH at 1.5 mg/mL in GelMA led to the highest compressive modulus and swelling ratio. The addition of MSNs-COOH (up to 3 mg/mL) in GelMA did not impact cell viability. The presence of MF in MSNs-COOH/GelMA significantly promoted cell proliferation. Significant upregulation of osteogenic-related genes (except OCN) were seen for modified (MSNs-COOH and MF-MSNs-COOH) hydrogels when compared to GelMA. Altogether, the engineered MF-MSNs-COOH/GelMA shows great promise in craniomaxillofacial applications as an injectable, cell-free and bioactive therapeutics for bone regeneration.


Subject(s)
Metformin , Nanospheres , Biocompatible Materials , Gelatin , Humans , Hydrogels , Metformin/pharmacology , Osteogenesis , Tissue Engineering
9.
ACS Appl Bio Mater ; 4(9): 6993-7006, 2021 09 20.
Article in English | MEDLINE | ID: mdl-35006932

ABSTRACT

Engineering multifunctional hydrogel systems capable of amplifying the regenerative capacity of endogenous progenitor cells via localized presentation of therapeutics under tissue inflammation is central to the translation of effective strategies for hard tissue regeneration. Here, we loaded dexamethasone (DEX), a pleotropic drug with anti-inflammatory and mineralizing abilities, into aluminosilicate clay nanotubes (halloysite clay nanotubes (HNTs)) to engineer an injectable multifunctional drug delivery system based on photo-cross-linkable gelatin methacryloyl (GelMA) hydrogel. In detail, a series of hydrogels based on GelMA formulations containing distinct amounts of DEX-loaded nanotubes was analyzed for physicochemical and mechanical properties and kinetics of DEX release as well as compatibility with mesenchymal stem cells from human exfoliated deciduous teeth (SHEDs). The anti-inflammatory response and mineralization potential of the engineered hydrogels were determined in vitro and in vivo. DEX conjugation with HNTs was confirmed by FTIR analysis. The incorporation of DEX-loaded nanotubes enhanced the mechanical strength of GelMA with no effect on its degradation and swelling ratio. Scanning electron microscopy (SEM) images demonstrated the porous architecture of GelMA, which was not significantly altered by DEX-loaded nanotubes' (HNTs/DEX) incorporation. All GelMA formulations showed cytocompatibility with SHEDs (p < 0.05) regardless of the presence of HNTs or HNTs/DEX. However, the highest osteogenic cell differentiation was noticed with the addition of HNT/DEX 10% in GelMA formulations (p < 0.01). The controlled release of DEX over 7 days restored the expression of alkaline phosphatase and mineralization (p < 0.0001) in lipopolysaccharide (LPS)-stimulated SHEDs in vitro. Importantly, in vivo data revealed that DEX-loaded nanotube-modified GelMA (5.0% HNT/DEX 10%) led to enhanced bone formation after 6 weeks (p < 0.0001) compared to DEX-free formulations with a minimum localized inflammatory response after 7 days. Altogether, our findings show that the engineered DEX-loaded nanotube-modified hydrogel may possess great potential to trigger in situ mineralized tissue regeneration under inflammatory conditions.


Subject(s)
Hydrogels , Tissue Engineering , Clay/chemistry , Drug Delivery Systems , Gelatin , Humans , Hydrogels/pharmacology , Methacrylates , Tissue Engineering/methods
10.
Acta Biomater ; 113: 164-176, 2020 09 01.
Article in English | MEDLINE | ID: mdl-32540497

ABSTRACT

One of the most damaging pathologies that affects the health of both soft and hard tissues around the tooth is periodontitis. Clinically, periodontal tissue destruction has been managed by an integrated approach involving elimination of injured tissues followed by regenerative strategies with bone substitutes and/or barrier membranes. Regrettably, a barrier membrane with predictable mechanical integrity and multifunctional therapeutic features has yet to be established. Herein, we report a fiber-reinforced hydrogel with unprecedented tunability in terms of mechanical competence and therapeutic features by integration of highly porous poly(ε-caprolactone) fibrous mesh(es) with well-controlled 3D architecture into bioactive amorphous magnesium phosphate-laden gelatin methacryloyl hydrogels. The presence of amorphous magnesium phosphate and PCL mesh in the hydrogel can control the mechanical properties and improve the osteogenic ability, opening a tremendous opportunity in guided bone regeneration (GBR). Results demonstrate that the presence of PCL meshes fabricated via melt electrowriting can delay hydrogel degradation preventing soft tissue invasion and providing the mechanical barrier to allow time for slower migrating progenitor cells to participate in bone regeneration due to their ability to differentiate into bone-forming cells. Altogether, our approach offers a platform technology for the development of the next-generation of GBR membranes with tunable mechanical and therapeutic properties to amplify bone regeneration in compromised sites. STATEMENT OF SIGNIFICANCE: In this study, we developed a fiber-reinforced hydrogel platform with unprecedented tunability in terms of mechanical competence and therapeutic features for guided bone regeneration. We successfully integrated highly porous poly(ε-caprolactone) [PCL] mesh(es) into amorphous magnesium phosphate-laden hydrogels. The stiffness of the engineered hydrogel was significantly enhanced, and this reinforcing effect could be modulated by altering the number of PCL meshes and tailoring the AMP concentration. Furthermore, the fiber-reinforced hydrogel showed favorable cellular responses, significantly higher rates of mineralization, upregulation of osteogenic-related genes and bone formation. In sum, these fiber-reinforced membranes in combination with therapeutic agent(s) embedded in the hydrogel offer a robust, highly tunable platform to amplify bone regeneration not only in periodontal defects, but also in other craniomaxillofacial sites.


Subject(s)
Bone Regeneration , Hydrogels , Animals , Gelatin , Hydrogels/pharmacology , Male , Osteogenesis , Polyesters , Rats , Stem Cells
11.
Dent Mater ; 36(7): 865-883, 2020 07.
Article in English | MEDLINE | ID: mdl-32451208

ABSTRACT

OBJECTIVE: The aim of this study was to develop bioactive and osseointegrable polyetheretherketone (PEEK)-based composite filaments melt-blended with novel amorphous magnesium phosphate (AMP) particles for 3D printing of dental and orthopedic implants. MATERIALS AND METHODS: A series of materials and biological analyses of AMP-PEEK were performed. Thermal stability, thermogravimetric and differential scanning calorimetry curves of as-synthesized AMP were measured. Complex viscosity, elastic modulus and viscous modulus were determined using a rotational rheometer. In vitro bioactivity was analyzed using SBF immersion method. SEM, EDS and XRD were used to study the apatite-forming ability of the AMP-PEEK filaments. Mouse pre-osteoblasts (MC3T3-E1) were cultured and analyzed for cell viability, proliferation and gene expression. For in vivo analyses, bare PEEK was used as the control and 15AMP-PEEK was chosen based on its in vitro cell-related results. After 4 or 12 weeks, animals were euthanized, and the femurs were collected for micro-computed tomography (µ-CT) and histology. RESULTS: The collected findings confirmed the homogeneous dispersion of AMP particles within the PEEK matrix with no phase degradation. Rheological studies demonstrated that AMP-PEEK composites are good candidates for 3D printing by exhibiting high zero-shear and low infinite-shear viscosities. In vitro results revealed enhanced bioactivity and superior pre-osteoblast cell function in the case of AMP-PEEK composites as compared to bare PEEK. In vivo analyses further corroborated the enhanced osseointegration capacity for AMP-PEEK implants. SIGNIFICANCE: Collectively, the present investigation demonstrated that AMP-PEEK composite filaments can serve as feedstock for 3D printing of orthopedic and dental implants due to enhanced bioactivity and osseointegration capacity.


Subject(s)
Dental Implants , Animals , Benzophenones , Ketones , Magnesium Compounds , Mice , Phosphates , Polyethylene Glycols , Polymers , Printing, Three-Dimensional , X-Ray Microtomography
12.
ACS Appl Mater Interfaces ; 12(21): 23752-23763, 2020 May 27.
Article in English | MEDLINE | ID: mdl-32352748

ABSTRACT

Bioprinting, a promising field in regenerative medicine, holds great potential to create three-dimensional, defect-specific vascularized bones with tremendous opportunities to address unmet craniomaxillofacial reconstructive challenges. A cytocompatible bioink is a critical prerequisite to successfully regenerate functional bone tissue. Synthetic self-assembling peptides have a nanofibrous structure resembling the native extracellular matrix (ECM), making them an excellent bioink component. Amorphous magnesium phosphates (AMPs) have shown greater levels of resorption while maintaining high biocompatibility, osteoinductivity, and low inflammatory response, as compared to their calcium phosphate counterparts. Here, we have established a novel bioink formulation (ECM/AMP) that combines an ECM-based hydrogel containing 2% octapeptide FEFEFKFK and 98% water with AMP particles to realize high cell function with desirable bioprintability. We analyzed the osteogenic differentiation of dental pulp stem cells (DPSCs) encapsulated in the bioink, as well as in vivo bone regeneration, to define the potential of the formulated bioink as a growth factor-free bone-forming strategy. Cell-laden AMP-modified bioprinted constructs showed an improved cell morphology but similar cell viability (∼90%) compared to their AMP-free counterpart. In functional assays, the cell-laden bioprinted constructs modified with AMP exhibited a high level of mineralization and osteogenic gene expression without the use of growth factors, thus suggesting that the presence of AMP-triggered DPSCs' osteogenic differentiation. Cell-free ECM-based bioprinted constructs were implanted in vivo. In comparison with the ECM group, bone volume per total volume for ECM/1.0AMP was approximately 1.7- and 1.4-fold higher at 4 and 8 weeks, respectively. Further, a significant increase in the bone density was observed in ECM/1.0AMP from 4 to 8 weeks. These results demonstrate that the presence of AMP in the bioink significantly increased bone formation, thus showing promise for in situ bioprinting strategies. We foresee significant potential in translating this innovative bioink toward the regeneration of patient-specific bone tissue for regenerative dentistry.


Subject(s)
Bone Regeneration/drug effects , Extracellular Matrix/chemistry , Hydrogels/chemistry , Ink , Magnesium Compounds/chemistry , Phosphates/chemistry , Skull/metabolism , Animals , Bioprinting/methods , Cell Differentiation/drug effects , Male , Osteogenesis/drug effects , Printing, Three-Dimensional , Proof of Concept Study , Rats, Inbred F344 , Stem Cells/metabolism , Tissue Engineering/methods
13.
ACS Appl Mater Interfaces ; 12(14): 16006-16017, 2020 Apr 08.
Article in English | MEDLINE | ID: mdl-32180395

ABSTRACT

A photocrosslinkable gelatin methacryloyl (GelMA) hydrogel has been widely examined in regenerative engineering because of its good cell-tissue affinity and degradability in the presence of matrix metalloproteinases. A halloysite aluminosilicate nanotube (HNT) is a known reservoir for the loading and sustained delivery of therapeutics. Here, we formulate injectable chlorhexidine (CHX)-loaded nanotube-modified GelMA hydrogel that is cytocompatible and biodegradable and provides sustained release of CHX for infection ablation while displaying good biocompatibility. The effects of HNTs and CHX on hydrogel degradability and mechanical properties, as well as on the kinetics of CHX release, and on the antimicrobial efficacy against oral pathogens were systematically assessed. Cytocompatibility in stem cells from human exfoliated deciduous teeth and inflammatory response in vivo using a subcutaneous rat model were determined. Our hydrogel system, that is, (CHX)-loaded nanotube-modified GelMA showed minimum localized inflammatory responses, supporting its ability for drug delivery applications. Moreover, we showed that the incorporation of CHX-loaded nanotubes reduces the mechanical properties, increases the swelling ratio, and diminishes the degradation rate of the hydrogels. Importantly, the presence of CHX-loaded nanotubes inhibits bacterial growth with minimal cell toxicity. Our findings provide a new strategy to modify GelMA hydrogel with chlorhexidine-loaded nanotubes for clinical use as an injectable drug delivery strategy for dental infection ablation.


Subject(s)
Aluminum Silicates/pharmacology , Biodegradable Plastics/pharmacology , Infection Control, Dental/methods , Nanotubes/chemistry , Aluminum Silicates/chemistry , Animals , Biocompatible Materials/chemistry , Biocompatible Materials/pharmacology , Biodegradable Plastics/chemistry , Chlorhexidine/chemistry , Clay/chemistry , Gelatin/chemistry , Gelatin/pharmacology , Humans , Hydrogels/chemistry , Hydrogels/pharmacology , Kinetics , Rats , Regenerative Medicine , Stem Cells/drug effects , Tissue Engineering/methods
14.
J Endod ; 45(11): 1371-1377, 2019 Nov.
Article in English | MEDLINE | ID: mdl-31542283

ABSTRACT

INTRODUCTION: Curcumin incorporation into polymeric fibers was tested for its antimicrobial properties and potential use in root canal disinfection. METHODS: Curcumin-modified fibers were processed via electrospinning and tested against a 7-day old established Actinomyces naeslundii biofilm. The medicaments tested were as follows: curcumin-modified fibers at 2.5 and 5.0 mg/mL, curcumin-based irrigant at 2.5 and 5.0 mg/mL, saline solution (negative control), and the following positive controls: 2% chlorhexidine, 1% sodium hypochlorite, and triple antibiotic paste (TAP, 1 mg/mL). All medicaments, except for the positive controls, were allocated according to the light exposure protocol (ie, photoactivation with a light-emitting diode every 30 seconds for 4 minutes or without photoactivation). After treatment, the medicaments were removed, and 1 mL saline solution was added; the biofilm was scraped from the well and used to prepare a 1:2000 dilution. Spiral plating was performed using anaerobic blood agar plates. After 24 hours, colony-forming units (colony-forming units/mL, n = 11/group) were counted to determine the antimicrobial effects. RESULTS: Data exhibited significant antimicrobial effects on the positive control groups followed by the curcumin irrigants and, lastly, the photoactivated curcumin-modified fibers. There was a significant reduction of viable bacteria in curcumin-based irrigants, which was greater than the TAP-treated group. Curcumin-free fibers, saline, and the nonphotoactivated curcumin-modified fibers did not display antimicrobial activity. CONCLUSIONS: Curcumin seems to be a potential alternative to TAP when controlling infection, but it requires a minimal concentration (2.5 mg/mL) to be effective. Photoactivation of curcumin-based medicaments seems to be essential to obtain greater antibiofilm activity.


Subject(s)
Curcumin , Disinfectants , Root Canal Therapy , Actinomyces/drug effects , Biofilms , Curcumin/pharmacokinetics , Curcumin/pharmacology , Dental Pulp Cavity , Disinfectants/pharmacokinetics , Disinfectants/pharmacology , Disinfection , Drug Liberation , Enterococcus faecalis , Root Canal Irrigants , Sodium Hypochlorite
15.
Int J Dent ; 2018: 4089274, 2018.
Article in English | MEDLINE | ID: mdl-30416524

ABSTRACT

Surface treatment alone does not determine the final microtopography of a dental implant, which can be influenced by implant design and the surgical procedure. This study investigated the effect of surgical placement of dental implants with same surface treatments on surface roughness. Three implants (SIN) of each group with different macrogeometries (Strong, Stylus, and Tryon) were analyzed using laser interferometry and scanning electron microscopy to evaluate surface topography. All threaded regions of the implants, namely, top, flank, and valley, were analyzed individually. Relevant surface parameters (S a, S sk, S ku, S tr, and S dq) were calculated for the different regions on each implant before (B) (n = 9) and after (A) (n = 9) placement into porcine rib bones. The behavior and proliferation of a preosteoblastic cell line MC3T3-E1 on titanium surface, cell viability, and osteopontin secretion were evaluated after 24 h, 48 h, and 96 h, also before (n = 18) and after (n = 18) implant placement into porcine ribs bone. As results, the valleys of all implants had an increase in S a values after implant placement. By contrast, the tops of the Stylus A implant and the flanks of the Tryon A implant showed a significant decrease in mean height of the irregularities (S a), 0.16 µm and 1.25 µm, respectively. The Stylus implant presented significantly (p < 0.05) higher asymmetry values on the distribution curve for irregularity heights (S ku) in all regions after insertion into bone (6.99 for tops, 9.54 for flanks, and 17.64 for valleys), indicating a greater preponderance of peaks over valleys. An increase in roughness gradients (S dq) was observed for all macrogeometries after insertion into bone. The cell culture results showed no significant difference (p > 0.05) for all macrogeometries after bone placement. In conclusion, a subtle change in implant surface roughness was detected after insertion into bone for all the macrogeometries, without significantly affecting the cellular parameters studied.

SELECTION OF CITATIONS
SEARCH DETAIL
...