Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
Add more filters










Publication year range
1.
ACS Appl Bio Mater ; 2024 Apr 10.
Article in English | MEDLINE | ID: mdl-38598256

ABSTRACT

Traditional tissue engineering methods face challenges, such as fabrication, implantation of irregularly shaped scaffolds, and limited accessibility for immediate healthcare providers. In situ bioprinting, an alternate strategy, involves direct deposition of biomaterials, cells, and bioactive factors at the site, facilitating on-site fabrication of intricate tissue, which can offer a patient-specific personalized approach and align with the principles of precision medicine. It can be applied using a handled device and robotic arms to various tissues, including skin, bone, cartilage, muscle, and composite tissues. Bioinks, the critical components of bioprinting that support cell viability and tissue development, play a crucial role in the success of in situ bioprinting. This review discusses in situ bioprinting techniques, the materials used for bioinks, and their critical properties for successful applications. Finally, we discuss the challenges and future trends in accelerating in situ printing to translate this technology in a clinical settings for personalized regenerative medicine.

2.
Dent Mater ; 40(1): 151-157, 2024 Jan.
Article in English | MEDLINE | ID: mdl-37945385

ABSTRACT

OBJECTIVE: Regenerative dentistry (RD) is an innovative strategy for treating necrotic teeth and regenerating damaged dental tissue. Biocompatible materials are pivotal for the advancement of RD, and the rising interest in environmental sustainability drives exploration of sustainable materials for dentistry. Bacterial nanocellulose (BNC) has emerged as a promising eco-friendly option and this study aims to assess BNC's suitability as scaffolds for regenerative dentistry applications. METHODS: Different in vitro methods have been utilized to characterize the properties of BNC scaffolds in regenerative dentistry, such as scanning electron microscopy (SEM) to analyse surface property and porosity, as well as examining their absorption behaviour using phosphate-buffered saline and bovine serum. Dental pulp stem cell (DPSCs) attachment, viability, and proliferation were evaluated using SEM, live and dead, and tetrazolium reduction assays. The odontogenic potential of the scaffold was evaluated using Alizarin Red staining and qPCR (14 and 21 days). RESULTS: Scanning electron microscopy (SEM) images and ethanol displacement method demonstrated the porous architecture of the BNC scaffold with an average porosity of 70.02 ± 4.74% and 50.26 ± 1.43% respectively. The scaffold absorbed 2846.54 ± 258.95 of BSA and 1648.63 ± 50.37% PBS after immersion in solution for 1 h, following pseudo first and second order kinetics. The biocompatibility assay indicated that cell density increased with time and that the scaffold was appropriate for cell adhesion and migration. Moreover, the BNC led to significantly higher mineralization and odontogenic expression compared to the control (BNC in conditioned media). SIGNIFICANCE: BNC showed fast adsorption of bovine serum, allowed DPSC attachment, migration, and odontogenic differentiation. This suggests its suitability as a biocompatible scaffold for triggering in situ mineralized tissue regeneration for regenerative dental applications.


Subject(s)
Biocompatible Materials , Tissue Scaffolds , Biocompatible Materials/pharmacology , Cell Differentiation , Odontogenesis , Bacteria , Dentistry , Dental Pulp , Tissue Engineering
3.
SLAS Technol ; 28(3): 152-164, 2023 06.
Article in English | MEDLINE | ID: mdl-37019216

ABSTRACT

Cancer treatment development is a complex process, with tumor heterogeneity and inter-patient variations limiting the success of therapeutic intervention. Traditional two-dimensional cell culture has been used to study cancer metabolism, but it fails to capture physiologically relevant cell-cell and cell-environment interactions required to mimic tumor-specific architecture. Over the past three decades, research efforts in the field of 3D cancer model fabrication using tissue engineering have addressed this unmet need. The self-organized and scaffold-based model has shown potential to study the cancer microenvironment and eventually bridge the gap between 2D cell culture and animal models. Recently, three-dimensional (3D) bioprinting has emerged as an exciting and novel biofabrication strategy aimed at developing a 3D compartmentalized hierarchical organization with the precise positioning of biomolecules, including living cells. In this review, we discuss the advancements in 3D culture techniques for the fabrication of cancer models, as well as their benefits and limitations. We also highlight future directions associated with technological advances, detailed applicative research, patient compliance, and regulatory challenges to achieve a successful bed-to-bench transition.


Subject(s)
Neoplasms , Tissue Engineering , Animals , Tissue Engineering/methods , Tumor Microenvironment , Neoplasms/therapy , Cell Culture Techniques/methods , Printing, Three-Dimensional
4.
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
5.
J Funct Biomater ; 15(1)2023 Dec 19.
Article in English | MEDLINE | ID: mdl-38276474

ABSTRACT

The liquid extract method is commonly used to evaluate the cytotoxicity and bioactivity of materials. Although ISO has recommended guidelines for test methods, variations in elution period, and shape of samples can influence the biological outcomes. The aim of this study was to investigate the influence of material form and elution period of Biodentine on dental pulp stem cells (DPSCs)' proliferation and mineralization. Biodentine (0.2 g) discs or powder were immersed in culture media (10 mL) for 1, 3 or 7 days (D1, D3 and D7). The eluents were filtered and used to treat DPSC. The calcium release profile and pH were determined. Cell proliferation was evaluated by MTS for 3 days, and mineralization and differentiation were assessed by alizarin red S staining (Ca2+/ng of DNA) and qRT-PCR (MEPE, DSPP, DMP-1, RUNX2, COL-I and OCN) for 14 days. Statistical analysis was performed with a one or two-way ANOVA and post hoc Tukey's test (pH, calcium release and proliferation) or Mann-Whitney test (α = 0.05). pH and calcium ion release of powdered eluents were significantly higher than disc eluents. Powdered eluent promoted extensive cell death, while the disc form was cytocompatible. All disc eluents significantly increased the gene expression and mineralization after 14 days compared to the untreated control. D7 induced less mineralization and differentiation compared to D1 and D3. Thus, the materials' form and elution time are critical aspects to be considered when evaluating the bioactivity of materials, since this binomial can affect positively and negatively the biological outcomes.

6.
Dent Mater ; 38(9): 1507-1517, 2022 09.
Article in English | MEDLINE | ID: mdl-35882570

ABSTRACT

OBJECTIVES: This work sought to formulate photocrosslinkable chlorhexidine (CHX)-laden methacrylated gelatin (CHX/GelMA) hydrogels with broad spectrum of action against endodontic pathogens as a clinically viable cell-friendly disinfection therapy prior to regenerative endodontics procedures. METHODS: CHX/GelMA hydrogel formulations were successfully synthesized using CHX concentrations between 0.12 % and 5 % w/v. Hydrogel microstructure was evaluated by scanning electron microscopy (SEM). Swelling and enzymatic degradation were assessed to determine microenvironmental effects. Compression test was performed to investigate the influence of CHX incorporation on the hydrogels' biomechanics. The antimicrobial and anti-biofilm potential of the formulated hydrogels were assessed using agar diffusion assays and a microcosms biofilm model, respectively. The cytocompatibility was evaluated by exposing stem cells from human exfoliated deciduous teeth (SHEDs) to hydrogel extracts (i.e., leachable byproducts obtained from overtime hydrogel incubation in phosphate buffer saline). The data were analyzed using One- and Two-way ANOVA and Tukey's test (α = 0.05). RESULTS: CHX/GelMA hydrogels were effectively prepared. NMR spectroscopy confirmed the incorporation of CHX into GelMA. The addition of CHX did not change the micromorphology (pore size) nor the swelling profile (p > 0.05). CHX incorporation reduced the degradation rate of the hydrogels (p < 0.001); whereas, it contributed to increased compressive modulus (p < 0.05). Regarding the antimicrobial properties, the incorporation of CHX showed a statistically significant decrease in the number of bacteria colonies at 0.12 % and 0.5 % concentration (p < 0.001) and completely inhibited the growth of biofilm at concentration levels 1 %, 2 %, and 5 %. Meanwhile, the addition of CHX, regardless of the concentration, did not lead to cell toxicity, as cell viability values were above 70 %. SIGNIFICANCE: The addition of CHX into GelMA showed significant antimicrobial action against the pathogens tested, even at low concentrations, with the potential to be used as a cell-friendly injectable drug delivery system for root canal disinfection prior to regenerative endodontics.


Subject(s)
Gelatin , Regenerative Endodontics , Cell Survival , Chlorhexidine/pharmacology , Gelatin/chemistry , Gelatin/pharmacology , Humans , Hydrogels/chemistry , Hydrogels/pharmacology , Tissue Engineering/methods
7.
Biomaterials ; 287: 121639, 2022 Aug.
Article in English | MEDLINE | ID: mdl-35779481

ABSTRACT

Tissue/organ shortage is a major medical challenge due to donor scarcity and patient immune rejections. Furthermore, it is difficult to predict or mimic the human disease condition in animal models during preclinical studies because disease phenotype differs between humans and animals. Three-dimensional bioprinting (3DBP) is evolving into an unparalleled multidisciplinary technology for engineering three-dimensional (3D) biological tissue with complex architecture and composition. The technology has emerged as a key driver by precise deposition and assembly of biomaterials with patient's/donor cells. This advancement has aided in the successful fabrication of in vitro models, preclinical implants, and tissue/organs-like structures. Here, we critically reviewed the current state of 3D-bioprinting strategies for regenerative therapy in eight organ systems, including nervous, cardiovascular, skeletal, integumentary, endocrine and exocrine, gastrointestinal, respiratory, and urinary systems. We also focus on the application of 3D bioprinting to fabricated in vitro models to study cancer, infection, drug testing, and safety assessment. The concept of in situ 3D bioprinting is discussed, which is the direct printing of tissues at the injury or defect site for reparative and regenerative therapy. Finally, issues such as scalability, immune response, and regulatory approval are discussed, as well as recently developed tools and technologies such as four-dimensional and convergence bioprinting. In addition, information about clinical trials using 3D printing has been included.

8.
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.

9.
Int J Mol Sci ; 23(2)2022 Jan 16.
Article in English | MEDLINE | ID: mdl-35055155

ABSTRACT

This study aimed at engineering cytocompatible and injectable antibiotic-laden fibrous microparticles gelatin methacryloyl (GelMA) hydrogels for endodontic infection ablation. Clindamycin (CLIN) or metronidazole (MET) was added to a polymer solution and electrospun into fibrous mats, which were processed via cryomilling to obtain CLIN- or MET-laden fibrous microparticles. Then, GelMA was modified with CLIN- or MET-laden microparticles or by using equal amounts of each set of fibrous microparticles. Morphological characterization of electrospun fibers and cryomilled particles was performed via scanning electron microscopy (SEM). The experimental hydrogels were further examined for swelling, degradation, and toxicity to dental stem cells, as well as antimicrobial action against endodontic pathogens (agar diffusion) and biofilm inhibition, evaluated both quantitatively (CFU/mL) and qualitatively via confocal laser scanning microscopy (CLSM) and SEM. Data were analyzed using ANOVA and Tukey's test (α = 0.05). The modification of GelMA with antibiotic-laden fibrous microparticles increased the hydrogel swelling ratio and degradation rate. Cell viability was slightly reduced, although without any significant toxicity (cell viability > 50%). All hydrogels containing antibiotic-laden fibrous microparticles displayed antibiofilm effects, with the dentin substrate showing nearly complete elimination of viable bacteria. Altogether, our findings suggest that the engineered injectable antibiotic-laden fibrous microparticles hydrogels hold clinical prospects for endodontic infection ablation.


Subject(s)
Anti-Bacterial Agents/pharmacology , Clindamycin/pharmacology , Dental Pulp Diseases/microbiology , Gelatin/chemistry , Methacrylates/chemistry , Metronidazole/pharmacology , Stem Cells/cytology , Anti-Bacterial Agents/chemistry , Biofilms/drug effects , Cells, Cultured , Clindamycin/chemistry , Dental Pulp Diseases/drug therapy , Humans , Hydrogels , Injections , Metronidazole/chemistry , Microscopy, Confocal , Microscopy, Electron, Scanning , Microspheres , Particle Size , Stem Cells/drug effects
10.
Dent Mater ; 38(3): 529-539, 2022 03.
Article in English | MEDLINE | ID: mdl-35074166

ABSTRACT

OBJECTIVE: Alveolar bone defects can be highly variable in their morphology and, as the defect size increases, they become more challenging to treat with currently available therapeutics and biomaterials. This investigation sought to devise a protocol for fabricating customized clinical scale and patient-specific, bioceramic scaffolds for reconstruction of large alveolar bone defects. METHODS: Two types of calcium phosphate (CaP)-based bioceramic scaffolds (alginate/ß-TCP and hydroxyapatite/α-TCP, hereafter referred to as hybrid CaP and Osteoink™, respectively) were designed, 3D printed, and their biocompatibility with alveolar bone marrow stem cells and mechanical properties were determined. Following scaffold optimization, a workflow was developed to use cone beam computed tomographic (CBCT) imaging to design and 3D print, defect-specific bioceramic scaffolds for clinical-scale bone defects. RESULTS: Osteoink™ scaffolds had the highest compressive strength when compared to hybrid CaP with different infill orientation. In cell culture medium, hybrid CaP degradation resulted in decreased pH (6.3) and toxicity to stem cells; however, OsteoInk™ scaffolds maintained a stable pH (7.2) in culture and passed the ISO standard for cytotoxicity. Finally, a clinically feasible laboratory workflow was developed and evaluated using CBCT imaging to engineer customized and defect-specific CaP scaffolds using OsteoInk™. It was determined that printed scaffolds had a high degree of accuracy to fit the respective clinical defects for which they were designed (0.27 mm morphological deviation of printed scaffolds from digital design). SIGNIFICANCE: From patient to patient, large alveolar bone defects are difficult to treat due to high variability in their complex morphologies and architecture. Our findings shows that Osteoink™ is a biocompatible material for 3D printing of clinically acceptable, patient-specific scaffolds with precision-fit for use in alveolar bone reconstructive procedures. Collectively, emerging digital technologies including CBCT imaging, 3D surgical planning, and (bio)printing can be integrated to address this unmet clinical challenge.


Subject(s)
Printing, Three-Dimensional , Tissue Scaffolds , Biocompatible Materials/chemistry , Bone Regeneration , Calcium Phosphates/chemistry , Durapatite , Humans , Tissue Engineering , Tissue Scaffolds/chemistry
11.
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
12.
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
13.
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
14.
Biomacromolecules ; 21(9): 3945-3956, 2020 09 14.
Article in English | MEDLINE | ID: mdl-32786527

ABSTRACT

Oral bacterial infection represents the leading cause of the gradual destruction of tooth and periodontal structures anchoring the teeth. Lately, injectable hydrogels have gained increased attention as a promising minimally invasive platform for localized delivery of personalized therapeutics. Here, an injectable and photocrosslinkable gelatin methacryloyl (GelMA) hydrogel is successfully engineered with ciprofloxacin (CIP)-eluting short nanofibers for oral infection ablation. For this purpose, CIP or its ß-cyclodextrin (ß-CD)-inclusion complex (CIP/ß-CD-IC) has been incorporated into polymeric electrospun fibers, which were subsequently cut into short nanofibers, and then embedded in GelMA to obtain an injectable hybrid antimicrobial hydrogel. Thanks to the solubility enhancement of CIP by ß-CD-IC and the tunable degradation profile of GelMA, the hydrogels promote localized, sustained, and yet effective cell-friendly antibiotic doses, as measured by a series of bacterial assays that demonstrated efficacy in attenuating the growth of Gram-positive Enterococcus faecalis. Altogether, we foresee significant potential in translating this innovative hybrid hydrogel as an injectable platform technology that may have broad applications in oral infection ablation, such as periodontal disease and pulpal pathology.


Subject(s)
Anti-Infective Agents , Nanofibers , Anti-Bacterial Agents/pharmacology , Gelatin , Hydrogels
15.
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
16.
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
17.
Colloids Surf B Biointerfaces ; 191: 111011, 2020 Jul.
Article in English | MEDLINE | ID: mdl-32334136

ABSTRACT

Beta-cyclodextrin (ß-CD) is an oligosaccharide commonly used to improve the aqueous solubility of lipophilic drugs (e.g., dexamethasone, DEX). Here we present the development of a drug delivery system to provide sustained release of DEX by ß-CD-inclusion complex (IC) to amplify the mineralization capacity of stem cells from human-extracted deciduous teeth (SHEDs) as a potential direct pulp capping strategy. First, IC of DEX (DEX-CD-IC) was synthesized with ß-CD. To confirm DEX-CD-IC complex formation, X-ray diffraction (XRD) and Fourier transform infrared spectroscopy (FTIR) analyses were performed. XRD data indicated that IC formation was achieved due to formation of a new crystalline structure, whereas FTIR revealed the presence of the IC from the shifting of the peaks of each component in DEX-CD-IC. Then, electrospun poly(lactic-co-glycolic acid, PLGA) fibers (PLGA/DEX-CD-IC) were processed by varying the concentration of DEX-CD-IC (5%, 10 %, and 15 %). The release of DEX from fibers was determined by ultraperformance liquid chromatography for 28 days. Thanks to the solubility enhancement of DEX by IC, electrospun PLGA/DEX-CD-IC fibers released DEX in a more sustained fashion compared to PLGA/DEX fibers. No deleterious effect was found in terms of SHEDs' proliferation when cultured with or on electrospun fibers, regardless of the IC presence. Importantly, a more pronounced odontogenic differentiation was stimulated by electrospun fibers loaded with the lowest DEX-CD-IC concentration (5%), as a result of the sustained DEX release. In sum, PLGA/DEX-CD-IC fibers have great potential in vital dental pulp therapy, owing to its sustained DEX release, cytocompatibility, and odontogenic differentiation capacity.


Subject(s)
Cyclodextrins/pharmacology , Dental Pulp/drug effects , Dexamethasone/pharmacology , Nanofibers/chemistry , Polymers/pharmacology , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Cells, Cultured , Child , Cyclodextrins/chemistry , Dental Pulp/pathology , Dexamethasone/chemistry , Drug Liberation , Humans , Particle Size , Polymers/chemistry , Surface Properties
18.
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
19.
Article in English | MEDLINE | ID: mdl-33575013

ABSTRACT

Background. This study aimed to investigate the endodontic debridement efficacy of different sodium hypochlorite (NaOCl) irrigation regimens with and without ultrasonic agitation, followed by ethylenediaminetetraacetic acid (EDTA) via scanning electron microscopy (SEM) after using a rotary instrumentation system. Methods. Mandibular premolars (n=50) were randomly divided into five experimental groups (n=10) for root canal instrumentation with ProTaper Universal rotary system up to F3. The root canal system was treated with intracanal-heated NaOCl (100°C) or preheated NaOCl (55°C), followed by ultrasonic agitation and EDTA treatment. Samples irrigated with conventional needle irrigation (CNI) using normal saline solution were used as controls. Debridement efficacy was analyzed by SEM. A five-point scale was used to estimate the presence/absence of debris for each canal segment (coronal, middle, and apical). The results were analyzed using one-way ANOVA and post hoc Tukey tests (P<0.05). Results. The experimental groups exhibited less debris compared to CNI with saline (P<0.05). The amount of debris decreased significantly for the group with NaOCl intracanal heating compared to extraoral heating. Ultrasonic agitation further enhanced the root canal debridement efficacy of NaOCl. Conclusion. In summary, intracanal heating of NaOCl with and without ultrasonic agitation followed by EDTA appears to be a promising method to flush debris from the root canal system.

20.
J Endod ; 45(7): 882-889, 2019 Jul.
Article in English | MEDLINE | ID: mdl-31133343

ABSTRACT

INTRODUCTION: This study aimed to compare the cytocompatibility and angiogenic potential of 2 antibiotics (clindamycin [CLIN] and minocycline [MINO]) at distinct concentrations on dental pulp stem cells (DPSCs) and human umbilical vein endothelial cells (HUVECs). METHODS: DPSCs and HUVECs were exposed to cell culture media modified with CLIN or MINO at concentrations ranging from 30 µg/mL-1000 µg/mL. Cell toxicity and proliferation were investigated using the lactate dehydrogenase and tetrazolium reduction assays, respectively. A capillarylike tube formation in vitro assay was conducted to determine the angiogenic potential associated with each antibiotic. Additionally, selected morphometric angiogenesis parameters were determined using dedicated software (WimTube; Onimagin Technologies SCA, Córdoba, Spain). All statistical analyses were performed using 1-way analysis of variance and the Tukey post hoc test (α= .05). RESULTS: The collected data showed that compared with the control (cell culture media, alpha-minimum essential medium Eagle) increasing the antibiotic concentration significantly decreased cell viability and proliferation of both DPSCs and HUVECs. In terms of angiogenic potential, when tested at 30 µg/mL and 50 µg/mL, CLIN significantly amplified tube formation when compared with MINO with angiogenesis parameters (ie, tube length and tube number) similar to the effect promoted by exogenous vascular endothelial growth factor (50 ng/mL). CONCLUSIONS: CLIN was less cytotoxic when compared with MINO at higher concentrations. Of note, CLIN did not hinder the proangiogenic activity induced by vascular endothelial growth factor to the same extent as MINO, suggesting that the replacement of MINO by CLIN might translate into positive implications in the overall regenerative outcome.


Subject(s)
Anti-Bacterial Agents , Clindamycin , Minocycline , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/toxicity , Cell Proliferation , Cells, Cultured , Clindamycin/pharmacology , Clindamycin/toxicity , Dental Pulp/drug effects , Human Umbilical Vein Endothelial Cells , Humans , Minocycline/pharmacology , Minocycline/toxicity , Neovascularization, Physiologic , Spain , Vascular Endothelial Growth Factor A
SELECTION OF CITATIONS
SEARCH DETAIL
...