Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 20.683
Filter
1.
Clin Oral Investig ; 28(8): 416, 2024 Jul 06.
Article in English | MEDLINE | ID: mdl-38969964

ABSTRACT

OBJECTIVES: To assess the biocompatibility, bioactivity, and immunomodulatory properties of three new calcium silicate cement-based sealers: Ceraseal (CS), Totalfill BC Sealer (TFbc) and WellRoot ST (WR-ST) on human periodontal ligament stem cells (hPDLSCs). MATERIALS AND METHODS: HPDLSCs were isolated from extracted third molars from healthy patients. Eluates (1:1, 1:2, and 1:4 ratio) and sample discs of CS, TFbc and WR-ST after setting were prepared. A series of assays were performed: cell characterization, cell metabolic activity (MTT assay) cell attachment and morphology (SEM assay), cell migration (wound-healing assay), cytoskeleton organization (phaloidin-based assay); IL-6 and IL-8 release (ELISA); differentiation marker expression (RT-qPCR assay), and cell mineralization (Alizarin Red S staining). HPDLSCs cultured in unconditioned (negative control) or osteogenic (positive control) culture media were used as a comparison. Statistical significance was established at p < 0.05. RESULTS: All the tested sealers exhibited similar results in the cytocompatibility assays (cell metabolic activity, migration, attachment, morphology, and cytoskeleton organization) compared with a negative control group. CS and TFbc exhibited an upregulation of at least one osteo/cementogenic marker compared to the negative and positive control groups. CS and TFbc also showed a significantly higher calcified nodule formation than the negative and positive control groups. Both the marker expression and calcified nodule formation were significantly higher in CS-treated cells than TFbc treated cells. WR-ST exhibited similar results to the control group. CS and TFbc-treated cells exhibited a significant downregulation of IL-6 after 72 h of culture compared to the negative control group (p < 0.05). CONCLUSION: All the tested sealers exhibited an adequate cytocompatibility. CS significantly enhances cell differentiation by upregulating the expression of key genes associated with bone and cementum formation. Additionally, CS was observed to facilitate the mineralization of the extracellular matrix effectively. In contrast, the effects of TFbc and WR-ST on these processes were less pronounced compared to CS. Furthermore, both CS and TFbc exhibited an anti-inflammatory potential, contributing to their potential therapeutic benefits in regenerative endodontics. CLINICAL RELEVANCE: This is the first study to compare the biological properties and immunomodulatory potential of Ceraseal, Totalfill BC Sealer, and WellRoot ST. The results act as supporting evidence for their use in root canal treatment.


Subject(s)
Biocompatible Materials , Calcium Compounds , Materials Testing , Periodontal Ligament , Silicates , Calcium Compounds/pharmacology , Silicates/pharmacology , Humans , Periodontal Ligament/cytology , Periodontal Ligament/drug effects , Biocompatible Materials/pharmacology , In Vitro Techniques , Cells, Cultured , Stem Cells/drug effects , Root Canal Filling Materials/pharmacology , Cell Differentiation/drug effects , Cell Movement/drug effects , Enzyme-Linked Immunosorbent Assay , Cell Adhesion/drug effects , Molar, Third
2.
Cell Biochem Funct ; 42(5): e4090, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38973147

ABSTRACT

Cellular therapy is considered a better option for the treatment of degenerative disorders. Different cell types are being used for tissue regeneration. Despite extensive research in this field, several issues remain to be addressed concerning cell transplantation. One of these issues is the survival and homing of administered cells in the injured tissue, which depends on the ability of these cells to adhere. To enhance cell adherence and survival, Rap1 GTPase was activated in mesenchymal stem cells (MSCs) as well as in cardiomyocytes (CMs) by using 8-pCPT-2'-O-Me-cAMP, and the effect on gene expression dynamics was determined through quantitative reverse transcriptase-polymerase chain reaction analysis. Pharmacological activation of MSCs and CMs resulted in the upregulation of connexin-43 and cell adhesion genes, which increased the cell adhesion ability of MSCs and CMs, and increased the fusion of MSCs with neonatal CMs. Treating stem cells with a pharmacological agent that activates Rap1a before transplantation can enhance their fusion with CMs and increase cellular regeneration.


Subject(s)
Mesenchymal Stem Cells , Myocytes, Cardiac , Mesenchymal Stem Cells/metabolism , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/drug effects , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/cytology , Myocytes, Cardiac/drug effects , Animals , Cell Adhesion/drug effects , Cell Adhesion Molecules/metabolism , Cell Adhesion Molecules/genetics , Cell Fusion , Cells, Cultured , Rats , Animals, Newborn , rap1 GTP-Binding Proteins/metabolism , rap1 GTP-Binding Proteins/genetics
3.
ACS Appl Mater Interfaces ; 16(25): 31922-31935, 2024 Jun 26.
Article in English | MEDLINE | ID: mdl-38874539

ABSTRACT

Poly-l-lysine (PLL) and Matrigel, both classical coating materials for culture substrates in neural stem cell (NSC) research, present distinct interfaces whose effect on NSC behavior at cellular and molecular levels remains ambiguous. Our investigation reveals intriguing disparities: although both PLL and Matrigel interfaces are hydrophilic and feature amine functional groups, Matrigel stands out with lower stiffness and higher roughness. Based on this diversity, Matrigel surpasses PLL, driving NSC adhesion, migration, and proliferation. Intriguingly, PLL promotes NSC differentiation into astrocytes, whereas Matrigel favors neural differentiation and the physiological maturation of neurons. At the molecular level, Matrigel showcases a wider upregulation of genes linked to NSC behavior. Specifically, it enhances ECM-receptor interaction, activates the YAP transcription factor, and heightens glycerophospholipid metabolism, steering NSC proliferation and neural differentiation. Conversely, PLL upregulates genes associated with glial cell differentiation and amino acid metabolism and elevates various amino acid levels, potentially linked to its support for astrocyte differentiation. These distinct transcriptional and metabolic activities jointly shape the divergent NSC behavior on these substrates. This study significantly advances our understanding of substrate regulation on NSC behavior, offering novel insights into optimizing and targeting the application of these surface coating materials in NSC research.


Subject(s)
Cell Differentiation , Cell Proliferation , Collagen , Drug Combinations , Laminin , Neural Stem Cells , Polylysine , Proteoglycans , Polylysine/chemistry , Neural Stem Cells/cytology , Neural Stem Cells/metabolism , Neural Stem Cells/drug effects , Laminin/chemistry , Laminin/pharmacology , Collagen/chemistry , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Proteoglycans/chemistry , Proteoglycans/pharmacology , Animals , Cell Adhesion/drug effects , Cell Movement/drug effects , Mice
4.
Biol Pharm Bull ; 47(6): 1072-1078, 2024.
Article in English | MEDLINE | ID: mdl-38825460

ABSTRACT

In previous studies, my group developed cell-adhesive peptide-polysaccharide complexes as biomaterials for tissue engineering. Having a wide variety of cell-adhesive peptides is important as the biological functions of peptide-polysaccharide complexes are highly dependent on the biological activity of peptides. This paper reviews the biological activities of two types of recently characterized cell-adhesive peptides. The first is peptides rich in basic amino acids originating from octaarginine. We analyzed the relationships between the amino acid composition of basic peptides and cell adhesion, elongation, and proliferation and identified the most suitable peptide for cell culture. The second was arginine-glycine-aspartic acid (RGD)-containing peptides that promote the adhesion of induced pluripotent stem cells (iPSCs). We identified the RGD-surrounding sequences necessary for iPSC adhesion, clarified the underlying mechanism, and improved cell adhesion by modifying the structure-activity relationships. The novel cell-adhesive peptides identified in our previous studies may aid in the development of novel peptide-based biomaterials.


Subject(s)
Biocompatible Materials , Cell Adhesion , Peptides , Cell Adhesion/drug effects , Biocompatible Materials/chemistry , Humans , Peptides/pharmacology , Peptides/chemistry , Animals , Oligopeptides/chemistry , Oligopeptides/pharmacology , Tissue Engineering/methods , Induced Pluripotent Stem Cells/cytology
5.
ACS Appl Mater Interfaces ; 16(25): 31983-31996, 2024 Jun 26.
Article in English | MEDLINE | ID: mdl-38865688

ABSTRACT

Effective osteointegration is of great importance for pedicle screws in spinal fusion surgeries. However, the lack of osteoinductive activity of current screws diminishes their feasibility for osteointegration and fixation, making screw loosening a common complication worldwide. In this study, Ti-6Al-4V pedicle screws with full through-hole design were fabricated via selective laser melting (SLM) 3D printing and then deposited with porous oxide coatings by microarc oxidation (MAO). The porous surface morphology of the oxide coating and the release of bioactive ions could effectively support cell adhesion, migration, vascularization, and osteogenesis in vitro. Furthermore, an in vivo goat model demonstrated the efficacy of modified screws in improving bone maturation and osseointegration, thus providing a promising method for feasible orthopedic internal fixation.


Subject(s)
Ceramics , Goats , Osseointegration , Oxidation-Reduction , Pedicle Screws , Printing, Three-Dimensional , Titanium , Animals , Osseointegration/drug effects , Titanium/chemistry , Titanium/pharmacology , Ceramics/chemistry , Ceramics/pharmacology , Alloys/chemistry , Alloys/pharmacology , Osteogenesis/drug effects , Humans , Porosity , Coated Materials, Biocompatible/chemistry , Coated Materials, Biocompatible/pharmacology , Cell Adhesion/drug effects
6.
Nanoscale ; 16(26): 12510-12522, 2024 Jul 04.
Article in English | MEDLINE | ID: mdl-38874593

ABSTRACT

Titanium-based orthopedic implants are gaining popularity in recent years due to their excellent biocompatibility, superior corrosion resistance and lightweight properties. However, these implants often fail to perform effectively due to poor osseointegration. Nanosurface modification approaches may help to resolve this problem. In this work, TiO2 nanotube (NT) arrays were fabricated on commercially available pure titanium (Ti) surfaces by anodization and annealing. Then, zinc (Zn) and strontium (Sr), important for cell signaling, were doped on the NT surface by hydrothermal treatment. This very simple method of Zn and Sr doping takes less time and energy compared to other complicated techniques. Different surface characterization tools such as scanning electron microscopy (SEM), X-ray photoelectron spectroscopy (XPS), energy-dispersive X-ray spectroscopy (EDS), static water contact angle, X-ray diffraction (XRD) and nanoindentation techniques were used to evaluate the modified surfaces. Then, adipose derived stem cells (ADSCs) were cultured with the surfaces to evaluate cell adhesion, proliferation, and growth on the surfaces. After that, the cells were differentiated towards osteogenic lineage to evaluate alkaline phosphatase (ALP) activity, osteocalcin expression, and calcium phosphate mineralization. Results indicate that NT surfaces doped with Zn and Sr had significantly enhanced ADSC adhesion, proliferation, growth, and osteogenic differentiation compared to an unmodified surface, thus confirming the enhanced performance of these surfaces.


Subject(s)
Cell Proliferation , Nanotubes , Osteogenesis , Strontium , Surface Properties , Titanium , Zinc , Titanium/chemistry , Titanium/pharmacology , Strontium/chemistry , Strontium/pharmacology , Nanotubes/chemistry , Zinc/chemistry , Zinc/pharmacology , Osteogenesis/drug effects , Cell Proliferation/drug effects , Cell Adhesion/drug effects , Cell Differentiation/drug effects , Humans , Alkaline Phosphatase/metabolism , Stem Cells/cytology , Stem Cells/metabolism , Stem Cells/drug effects , Biocompatible Materials/chemistry , Biocompatible Materials/pharmacology , Cells, Cultured
7.
Chem Biol Interact ; 398: 111115, 2024 Aug 01.
Article in English | MEDLINE | ID: mdl-38908811

ABSTRACT

In the present study, the effect of sulfonamide-chalcone 185 (SSC185) was investigated against B16-F10 metastatic melanoma cells aggressive actions, besides migration and adhesion processes, by in silico and in vitro assays. In silico studies were used to characterize the pharmacokinetic profile and possible targets of SSC185, using the pkCSM web server, and docking simulations with AutoDock Tools. Furthermore, the antimetastatic effect of SSC185 was investigated by in vitro experiments using MTT (3-(4,5-Dimethylthiazol-2-yl)-2,5-Diphenyltetrazolium Bromide), colony, scratch, and cell adhesion assays, and atomic force microscopy (AFM). The molecular docking results show better affinity of SSC185 with the metalloproteinases-2 (MMP-2) and α5ß1 integrin. SSC185 effectively restricts the formation of colonies, migration, and adhesion of B16-F10 metastatic melanoma cells. Through the AFM images changes in cells morphology was identified, with a decrease in the filopodia and increase in the average cellular roughness. The results obtained demonstrate the potential of this molecule in inhibit the primordial steps for metastasis, which is responsible for a worse prognosis of late stage cancer, being the main cause of morbidity among cancer patients.


Subject(s)
Cell Adhesion , Cell Movement , Chalcone , Molecular Docking Simulation , Sulfonamides , Cell Movement/drug effects , Cell Adhesion/drug effects , Sulfonamides/pharmacology , Sulfonamides/chemistry , Mice , Animals , Cell Line, Tumor , Chalcone/pharmacology , Chalcone/chemistry , Chalcone/analogs & derivatives , Matrix Metalloproteinase 2/metabolism , Melanoma, Experimental/pathology , Melanoma, Experimental/drug therapy , Melanoma, Experimental/metabolism , Microscopy, Atomic Force , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Chalcones/pharmacology , Chalcones/chemistry , Humans
8.
Int J Biol Macromol ; 273(Pt 1): 132768, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38823733

ABSTRACT

Polylactic acid (PLA), a polymer derived from renewable resources, is gaining increasing attention in the development of biomedical devices due to its cost-effectiveness, low immunogenicity, and biodegradability. However, its inherent hydrophobicity remains a problem, leading to poor cell adhesion features. On this basis, the aim of this work was to develop a method for functionalizing the surface of PLA films with a biopolymer, chitosan (CH), which was proved to be a material with intrinsic cell adhesive properties, but whose mechanical properties are insufficient to be used alone. The combination of the two polymers, PLA as a bulk scaffold and CH as a coating, could be a promising combination to develop a scaffold for cell growth. The modification of PLA films involved several steps: aminolysis followed by bromination to graft amino and then bromide groups, poly(glycidyl methacrylate) (PGMA) grafting by surface-initiated supplemental activator and reducing agent atom transfer radical polymerization (SI-SARA ATRP) and finally the CH grafting. To prove the effective adhesive properties, conjugated and non-conjugated films were tested in vitro as substrates for neuronal cell growth using differentiated neurons from human induced pluripotent stem cells. The results demonstrated enhanced cell growth in the presence of CH.


Subject(s)
Cell Proliferation , Chitosan , Neurons , Polyesters , Tissue Scaffolds , Chitosan/chemistry , Polyesters/chemistry , Humans , Tissue Scaffolds/chemistry , Neurons/cytology , Neurons/drug effects , Cell Proliferation/drug effects , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/drug effects , Polymerization , Cell Adhesion/drug effects , Biocompatible Materials/chemistry
9.
Int J Biol Macromol ; 273(Pt 1): 133064, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38866288

ABSTRACT

Bone tissue regeneration strategies have incorporated the use of natural polymers, such as hydroxyapatite (nHA), chitosan (CH), gelatin (GEL), or alginate (ALG). Additionally, platelet concentrates, such as platelet-rich fibrin (PRF) have been suggested to improve scaffold biocompatibility. This study aimed to develop scaffolds composed of nHA, GEL, and CH, with or without ALG and lyophilized PRF, to evaluate the scaffold's properties, growth factor release, and dental pulp stem cells (DPSC), and osteoblast (OB) derived from DPSC viability. Four scaffold variations were synthesized and lyophilized. Then, degradation, swelling profiles, and morphological analysis were performed. Furthermore, PDGF-BB and FGF-B growth factors release were quantified by ELISA, and cytotoxicity and cell viability were evaluated. The swelling and degradation profiles were similar in all scaffolds, with pore sizes ranging between 100 and 250 µm. FGF-B and PDGF-BB release was evidenced after 24 h of scaffold immersion in cell culture medium. DPSC and OB-DPSC viability was notably increased in PRF-supplemented scaffolds. The nHA-CH-GEL-PRF scaffold demonstrated optimal physical-biological characteristics for stimulating DPSC and OB-DPSC cell viability. These results suggest lyophilized PRF improves scaffold biocompatibility for bone tissue regeneration purposes.


Subject(s)
Alginates , Cell Survival , Chitosan , Dental Pulp , Durapatite , Gelatin , Osteoblasts , Platelet-Rich Fibrin , Stem Cells , Tissue Scaffolds , Humans , Dental Pulp/cytology , Chitosan/chemistry , Chitosan/pharmacology , Gelatin/chemistry , Platelet-Rich Fibrin/chemistry , Platelet-Rich Fibrin/metabolism , Tissue Scaffolds/chemistry , Stem Cells/drug effects , Stem Cells/cytology , Stem Cells/metabolism , Cell Survival/drug effects , Durapatite/chemistry , Durapatite/pharmacology , Alginates/chemistry , Alginates/pharmacology , Osteoblasts/drug effects , Osteoblasts/cytology , Cell Adhesion/drug effects , Tissue Engineering/methods , Cells, Cultured
10.
Int J Biol Macromol ; 273(Pt 2): 133193, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38885859

ABSTRACT

A major problem after tendon injury is adhesion formation to the surrounding tissue leading to a limited range of motion. A viable strategy to reduce adhesion extent is the use of physical barriers that limit the contact between the tendon and the adjacent tissue. The purpose of this study was to fabricate an electrospun bilayered tube of hyaluronic acid/polyethylene oxide (HA/PEO) and biodegradable DegraPol® (DP) to improve the anti-adhesive effect of the implant in a rabbit Achilles tendon full laceration model compared to a pure DP tube. Additionally, the attachment of rabbit tenocytes on pure DP and HA/PEO containing scaffolds was tested and Scanning Electron Microscopy, Fourier-transform Infrared Spectroscopy, Differential Scanning Calorimetry, Water Contact Angle measurements, and testing of mechanical properties were used to characterize the scaffolds. In vivo assessment after three weeks showed that the implant containing a second HA/PEO layer significantly reduced adhesion extent reaching levels comparable to native tendons, compared with a pure DP implant that reduced adhesion formation only by 20 %. Tenocytes were able to attach to and migrate into every scaffold, but cell number was reduced over two weeks. Implants containing HA/PEO showed better mechanical properties than pure DP tubes and with the ability to entirely reduce adhesion extent makes this implant a promising candidate for clinical application in tendon repair.


Subject(s)
Hyaluronic Acid , Polyethylene Glycols , Tissue Scaffolds , Animals , Hyaluronic Acid/chemistry , Hyaluronic Acid/pharmacology , Rabbits , Polyethylene Glycols/chemistry , Tissue Scaffolds/chemistry , Tenocytes/drug effects , Tenocytes/metabolism , Achilles Tendon/drug effects , Tendon Injuries/therapy , Cell Adhesion/drug effects , Tissue Adhesions/prevention & control , Tendons/drug effects , Tissue Engineering/methods , Biocompatible Materials/chemistry , Biocompatible Materials/pharmacology , Polyesters/chemistry , Polyurethanes
11.
Int J Mol Sci ; 25(11)2024 May 29.
Article in English | MEDLINE | ID: mdl-38892114

ABSTRACT

This study presents the effects of treating polystyrene (PS) cell culture plastic with oxidoreductase enzyme laccase and the catechol substrates caffeic acid (CA), L-DOPA, and dopamine on the culturing of normal human epidermal melanocytes (NHEMs) and human embryonal carcinoma cells (NTERA-2). The laccase-substrate treatment improved PS hydrophilicity and roughness, increasing NHEM and NTERA-2 adherence, proliferation, and NHEM melanogenesis to a level comparable with conventional plasma treatment. Cell adherence dynamics and proliferation were evaluated. The NHEM endpoint function was quantified by measuring melanin content. PS surfaces treated with laccase and its substrates demonstrated the forming of polymer-like structures. The surface texture roughness gradient and the peak curvature were higher on PS treated with a combination of laccase and substrates than laccase alone. The number of adherent NHEM and NTERA-2 was significantly higher than on the untreated surface. The proliferation of NHEM and NTERA-2 correspondingly increased on treated surfaces. NHEM melanin content was enhanced 6-10-fold on treated surfaces. In summary, laccase- and laccase-substrate-modified PS possess improved PS surface chemistry/hydrophilicity and altered roughness compared to untreated and plasma-treated surfaces, facilitating cellular adherence, subsequent proliferation, and exertion of the melanotic phenotype. The presented technology is easy to apply and creates a promising custom-made, substrate-based, cell-type-specific platform for both 2D and 3D cell culture.


Subject(s)
Caffeic Acids , Cell Proliferation , Dopamine , Laccase , Melanins , Melanocytes , Polystyrenes , Humans , Laccase/metabolism , Melanocytes/metabolism , Melanocytes/drug effects , Cell Proliferation/drug effects , Polystyrenes/chemistry , Caffeic Acids/pharmacology , Caffeic Acids/chemistry , Dopamine/metabolism , Melanins/metabolism , Cell Adhesion/drug effects , Levodopa/pharmacology , Levodopa/metabolism , Levodopa/chemistry , Surface Properties , Cell Line, Tumor , Embryonal Carcinoma Stem Cells/metabolism , Embryonal Carcinoma Stem Cells/drug effects
12.
ACS Appl Mater Interfaces ; 16(23): 29770-29782, 2024 Jun 12.
Article in English | MEDLINE | ID: mdl-38832565

ABSTRACT

Biomaterial surface engineering and the integration of cell-adhesive ligands are crucial in biological research and biotechnological applications. The interplay between cells and their microenvironment, influenced by chemical and physical cues, impacts cellular behavior. Surface modification of biomaterials profoundly affects cellular responses, especially at the cell-surface interface. This work focuses on enhancing cellular activities through material manipulation, emphasizing silanization for further functionalization with bioactive molecules such as RGD peptides to improve cell adhesion. The grafting of three distinct silanes onto silicon wafers using both spin coating and immersion methods was investigated. This study sheds light on the effects of different alkyl chain lengths and protecting groups on cellular behavior, providing valuable insights into optimizing silane-based self-assembled monolayers (SAMs) before peptide or protein grafting for the first time. Specifically, it challenges the common use of APTES molecules in this context. These findings advance our understanding of surface modification strategies, paving the way for tailoring biomaterial surfaces to modulate the cellular behavior for diverse biotechnological applications.


Subject(s)
Cell Adhesion , Silanes , Silicon , Surface Properties , Cell Adhesion/drug effects , Silicon/chemistry , Silanes/chemistry , Humans , Oligopeptides/chemistry , Oligopeptides/pharmacology , Biocompatible Materials/chemistry , Biocompatible Materials/pharmacology
13.
Int J Nanomedicine ; 19: 5109-5123, 2024.
Article in English | MEDLINE | ID: mdl-38846643

ABSTRACT

Introduction: Lumbar interbody fusion is widely employed for both acute and chronic spinal diseases interventions. However, large incision created during interbody cage implantation may adversely impair spinal tissue and influence postoperative recovery. The aim of this study was to design a shape memory interbody fusion device suitable for small incision implantation. Methods: In this study, we designed and fabricated an intervertebral fusion cage that utilizes near-infrared (NIR) light-responsive shape memory characteristics. This cage was composed of bisphenol A diglycidyl ether, polyether amine D-230, decylamine and iron oxide nanoparticles. A self-hardening calcium phosphate-starch cement (CSC) was injected internally through the injection channel of the cage for healing outcome improvement. Results: The size of the interbody cage is reduced from 22 mm to 8.8 mm to minimize the incision size. Subsequent NIR light irradiation prompted a swift recovery of the cage shape within 5 min at the lesion site. The biocompatibility of the shape memory composite was validated through in vitro MC3T3-E1 cell (osteoblast-like cells) adhesion and proliferation assays and subcutaneous implantation experiments in rats. CSC was injected into the cage, and the relevant results revealed that CSC is uniformly dispersed within the internal space, along with the cage compressive strength increasing from 12 to 20 MPa. Conclusion: The results from this study thus demonstrated that this integrated approach of using a minimally invasive NIR shape memory spinal fusion cage with CSC has potential for lumbar interbody fusion.


Subject(s)
Spinal Fusion , Spinal Fusion/instrumentation , Spinal Fusion/methods , Animals , Mice , Rats , Calcium Phosphates/chemistry , Minimally Invasive Surgical Procedures/instrumentation , Minimally Invasive Surgical Procedures/methods , Lumbar Vertebrae/surgery , Rats, Sprague-Dawley , Male , Compressive Strength , Cell Proliferation/drug effects , Bone Cements/chemistry , Smart Materials/chemistry , Cell Adhesion/drug effects
14.
Sci Rep ; 14(1): 12721, 2024 06 03.
Article in English | MEDLINE | ID: mdl-38830871

ABSTRACT

Surface structure plays a crucial role in determining cell behavior on biomaterials, influencing cell adhesion, proliferation, differentiation, as well as immune cells and macrophage polarization. While grooves and ridges stimulate M2 polarization and pits and bumps promote M1 polarization, these structures do not accurately mimic the real bone surface. Consequently, the impact of mimicking bone surface topography on macrophage polarization remains unknown. Understanding the synergistic sequential roles of M1 and M2 macrophages in osteoimmunomodulation is crucial for effective bone tissue engineering. Thus, exploring the impact of bone surface microstructure mimicking biomaterials on macrophage polarization is critical. In this study, we aimed to sequentially activate M1 and M2 macrophages using Poly-L-Lactic acid (PLA) membranes with bone surface topographical features mimicked through the soft lithography technique. To mimic the bone surface topography, a bovine femur was used as a model surface, and the membranes were further modified with collagen type-I and hydroxyapatite to mimic the bone surface microenvironment. To determine the effect of these biomaterials on macrophage polarization, we conducted experimental analysis that contained estimating cytokine release profiles and characterizing cell morphology. Our results demonstrated the potential of the hydroxyapatite-deposited bone surface-mimicked PLA membranes to trigger sequential and synergistic M1 and M2 macrophage polarizations, suggesting their ability to achieve osteoimmunomodulatory macrophage polarization for bone tissue engineering applications. Although further experimental studies are required to completely investigate the osteoimmunomodulatory effects of these biomaterials, our results provide valuable insights into the potential advantages of biomaterials that mimic the complex microenvironment of bone surfaces.


Subject(s)
Macrophages , Polyesters , Surface Properties , Animals , Macrophages/metabolism , Macrophages/drug effects , Macrophages/immunology , Cattle , Polyesters/chemistry , Mice , Biocompatible Materials/chemistry , Biocompatible Materials/pharmacology , Tissue Engineering/methods , Durapatite/chemistry , Cytokines/metabolism , Bone and Bones/cytology , Cell Differentiation/drug effects , Macrophage Activation/drug effects , Cell Adhesion/drug effects , RAW 264.7 Cells , Cell Polarity/drug effects , Femur , Collagen Type I/metabolism
15.
J Mater Sci Mater Med ; 35(1): 31, 2024 Jun 19.
Article in English | MEDLINE | ID: mdl-38896291

ABSTRACT

Orthopedic and dental implant failure continues to be a significant concern due to localized bacterial infections. Previous studies have attempted to improve implant surfaces by modifying their texture and roughness or coating them with antibiotics to enhance antibacterial properties for implant longevity. However, these approaches have demonstrated limited effectiveness. In this study, we attempted to engineer the titanium (Ti) alloy surface biomimetically at the nanometer scale, inspired by the cicada wing nanostructure using alkaline hydrothermal treatment (AHT) to simultaneously confer antibacterial properties and support the adhesion and proliferation of mammalian cells. The two modified Ti surfaces were developed using a 4 h and 8 h AHT process in 1 N NaOH at 230 °C, followed by a 2-hour post-calcination at 600 °C. We found that the control plates showed a relatively smooth surface, while the treatment groups (4 h & 8 h AHT) displayed nanoflower structures containing randomly distributed nano-spikes. The results demonstrated a statistically significant decrease in the contact angle of the treatment groups, which increased wettability characteristics. The 8 h AHT group exhibited the highest wettability and significant increase in roughness 0.72 ± 0.08 µm (P < 0.05), leading to more osteoblast cell attachment, reduced cytotoxicity effects, and enhanced relative survivability. The alkaline phosphatase activity measured in all different groups indicated that the 8 h AHT group exhibited the highest activity, suggesting that the surface roughness and wettability of the treatment groups may have facilitated cell adhesion and attachment and subsequently increased secretion of extracellular matrix. Overall, the findings indicate that biomimetic nanotextured surfaces created by the AHT process have the potential to be translated as implant coatings to enhance bone regeneration and implant integration.


Subject(s)
Biomimetic Materials , Dental Implants , Osteoblasts , Surface Properties , Titanium , Wettability , Osteoblasts/drug effects , Titanium/chemistry , Animals , Biomimetic Materials/chemistry , Biomimetic Materials/pharmacology , Cell Adhesion/drug effects , Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/pharmacology , Materials Testing , Biomimetics , Humans , Cell Proliferation/drug effects , Alloys/chemistry , Prostheses and Implants , Coated Materials, Biocompatible/chemistry , Coated Materials, Biocompatible/pharmacology , Nanostructures/chemistry , Cell Survival/drug effects , Alkaline Phosphatase/metabolism , Hemiptera , Cell Line
16.
J Appl Biomed ; 22(2): 107-114, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38912866

ABSTRACT

Diffuse large B-cell lymphoma (DLBCL) stands out as the most common type of malignant cancer, representing the majority of cases of non-Hodgkin's lymphoma. Ethyl pyruvate (EP) is a derivative of pyruvic acid and found to have potent anti-tumor properties. Despite its potential benefits, the impact of EP on DLBCL remains ambiguous. Our objective is to elucidate the role of EP in modulating the development of DLBCL. Analysis of cholecystokinin-8 (CCK-8) revealed that treatment with EP significantly diminished the viability of DLBCL cells. Furthermore, EP administration suppressed colony formation and hindered cell adhesion and invasion in DLBCL cells. Examination of cell cycle progression showed that EP treatment induced arrest at the G1 phase and subsequently reduced the S phase population in DLBCL cells. EP treatment consistently exhibited apoptosis-inducing properties in Annexin-V assays, and notably downregulated the expression of Bcl-2 while increasing levels of proapoptotic cleaved caspase 3 and BAX in DLBCL cells. Additionally, EP treatment decreased the overexpression of c-Jun in c-Jun-transfected DLBCL cells. Further, EP demonstrated DNA-damaging effects in TUNEL assays. In vivo, xenograft animal models revealed that EP treatment significantly mitigated DLBCL tumor growth and suppressed DLBCL cell adhesion to bone marrow stromal cells. In summary, these findings suggest that EP mitigates DLBCL progression by inducing apoptosis, inducing cell cycle arrest, and promoting DNA damage.


Subject(s)
Cell Adhesion , Cell Proliferation , Lymphoma, Large B-Cell, Diffuse , Pyruvates , Pyruvates/pharmacology , Pyruvates/therapeutic use , Lymphoma, Large B-Cell, Diffuse/drug therapy , Lymphoma, Large B-Cell, Diffuse/pathology , Humans , Animals , Cell Adhesion/drug effects , Cell Proliferation/drug effects , Cell Line, Tumor , Mice , Apoptosis/drug effects , Proto-Oncogene Proteins c-jun/metabolism , Proto-Oncogene Proteins c-jun/genetics , Xenograft Model Antitumor Assays
17.
Sci Rep ; 14(1): 14110, 2024 06 19.
Article in English | MEDLINE | ID: mdl-38898117

ABSTRACT

Newly synthesized gemini quaternary ammonium salts (QAS) with different counterions (bromide, hydrogen chloride, methylcarbonate, acetate, lactate), chain lengths (C12, C14, C16) and methylene linker (3xCH2) were tested. Dihydrochlorides and dibromides with 12 carbon atoms in hydrophobic chains were characterized by the highest biological activity against planktonic forms of yeast and yeast-like fungi. The tested gemini surfactants also inhibited the production of filaments by C. albicans. Moreover, they reduced the adhesion of C. albicans cells to the surfaces of stainless steel, silicone and glass, and slightly to polystyrene. In particular, the gemini compounds with 16-carbon alkyl chains were most effective against biofilms. It was also found that the tested surfactants were not cytotoxic to yeast cells. Moreover, dimethylcarbonate (2xC12MeCO3G3) did not cause hemolysis of sheep erythrocytes. Dihydrochlorides, dilactate and diacetate showed no mutagenic potential.


Subject(s)
Antifungal Agents , Biofilms , Candida albicans , Quaternary Ammonium Compounds , Biofilms/drug effects , Quaternary Ammonium Compounds/pharmacology , Quaternary Ammonium Compounds/chemistry , Quaternary Ammonium Compounds/chemical synthesis , Antifungal Agents/pharmacology , Antifungal Agents/chemical synthesis , Antifungal Agents/chemistry , Candida albicans/drug effects , Animals , Sheep , Surface-Active Agents/pharmacology , Surface-Active Agents/chemical synthesis , Surface-Active Agents/chemistry , Hemolysis/drug effects , Erythrocytes/drug effects , Microbial Sensitivity Tests , Cell Adhesion/drug effects , Stainless Steel/chemistry
18.
ACS Nano ; 18(24): 15815-15830, 2024 Jun 18.
Article in English | MEDLINE | ID: mdl-38833572

ABSTRACT

Amyloid-like fibrils are garnering keen interest in biotechnology as supramolecular nanofunctional units to be used as biomimetic platforms to control cell behavior. Recent insights into fibril functionality have highlighted their importance in tissue structure, mechanical properties, and improved cell adhesion, emphasizing the need for scalable and high-kinetics fibril synthesis. In this study, we present the instantaneous and bulk formation of amyloid-like nanofibrils from human platelet lysate (PL) using the ionic liquid cholinium tosylate as a fibrillating agent. The instant fibrillation of PL proteins upon supramolecular protein-ionic liquid interactions was confirmed from the protein conformational transition toward cross-ß-sheet-rich structures. These nanofibrils were utilized as building blocks for the formation of thin and flexible free-standing membranes via solvent casting to support cell self-aggregation. These PL-derived fibril membranes reveal a nanotopographically rough surface and high stability over 14 days under cell culture conditions. The culture of mesenchymal stem cells or tumor cells on the top of the membrane demonstrated that cells are able to adhere and self-organize in a three-dimensional (3D) spheroid-like microtissue while tightly folding the fibril membrane. Results suggest that nanofibril membrane incorporation in cell aggregates can improve cell viability and metabolic activity, recreating native tissues' organization. Altogether, these PL-derived nanofibril membranes are suitable bioactive platforms to generate 3D cell-guided microtissues, which can be explored as bottom-up strategies to faithfully emulate native tissues in a fully human microenvironment.


Subject(s)
Blood Platelets , Nanofibers , Humans , Blood Platelets/metabolism , Blood Platelets/chemistry , Nanofibers/chemistry , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/metabolism , Cell Aggregation/drug effects , Cell Adhesion/drug effects , Amyloid/chemistry , Amyloid/metabolism , Membranes, Artificial
19.
J Toxicol Sci ; 49(6): 281-288, 2024.
Article in English | MEDLINE | ID: mdl-38825487

ABSTRACT

Nitric oxide (NO) plays a physiological role in signal transduction and excess or chronic NO has toxic effects as an inflammatory mediator. NO reversibly forms protein S-nitrosylation and exerts toxicological functions related to disease progression. DNA methyltransferases, epigenome-related enzymes, are inhibited in enzymatic activity by S-nitrosylation. Therefore, excess or chronic NO exposure may cause disease by altering gene expression. However, the effects of chronic NO exposure on transcriptome are poorly understood. Here, we performed transcriptome analysis of A549, AGS, HEK293T, and SW48 cells exposed to NO (100 µM) for 48 hr. We showed that the differentially expressed genes were cell-specific. Gene ontology analysis showed that the functional signature of differentially expressed genes related to cell adhesion or migration was upregulated in several cell lines. Gene set enrichment analysis indicated that NO stimulated inflammation-related gene expression in various cell lines. This finding supports previous studies showing that NO is closely involved in inflammatory diseases. Overall, this study elucidates the pathogenesis of NO-associated inflammatory diseases by focusing on changes in gene expression.


Subject(s)
Gene Expression Profiling , Nitric Oxide , Transcriptome , Humans , Nitric Oxide/metabolism , Transcriptome/drug effects , Cell Adhesion/drug effects , Cell Adhesion/genetics , HEK293 Cells , Cell Movement/drug effects , Cell Movement/genetics , Inflammation/genetics , Inflammation/chemically induced , Signal Transduction/drug effects , Signal Transduction/genetics
20.
Sci Rep ; 14(1): 14178, 2024 06 19.
Article in English | MEDLINE | ID: mdl-38898058

ABSTRACT

Increasing evidence supports the hypothesis that cancer progression is under mitochondrial control. Mitochondrial fission plays a pivotal role in the maintenance of cancer cell homeostasis. The inhibition of DRP1, the main regulator of mitochondrial fission, with the mitochondrial division inhibitor (mdivi-1) had been associated with cancer cell sensitivity to chemotherapeutics and decrease proliferation. Here, using breast cancer cells we find that mdivi-1 induces the detachment of the cells, leading to a bulk of floating cells that conserved their viability. Despite a decrease in their proliferative and clonogenic capabilities, these floating cells maintain the capacity to re-adhere upon re-seeding and retain their migratory and invasive potential. Interestingly, the cell detachment induced by mdivi-1 is independent of DRP1 but relies on inhibition of mitochondrial complex I. Furthermore, mdivi-1 induces cell detachment rely on glucose and the pentose phosphate pathway. Our data evidence a novel DRP1-independent effect of mdivi-1 in the attachment of cancer cells. The generation of floating viable cells restricts the use of mdivi-1 as a therapeutic agent and demonstrates that mdivi-1 effect on cancer cells are more complex than anticipated.


Subject(s)
Breast Neoplasms , Dynamins , Extracellular Matrix , Mitochondrial Dynamics , Quinazolinones , Humans , Dynamins/metabolism , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Breast Neoplasms/drug therapy , Female , Extracellular Matrix/metabolism , Extracellular Matrix/drug effects , Cell Line, Tumor , Quinazolinones/pharmacology , Mitochondrial Dynamics/drug effects , Cell Adhesion/drug effects , Cell Movement/drug effects , Cell Survival/drug effects , Cell Proliferation/drug effects , Mitochondria/metabolism , Mitochondria/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...