Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 94
Filter
Add more filters

Publication year range
1.
Biotechnol Bioeng ; 120(12): 3638-3654, 2023 12.
Article in English | MEDLINE | ID: mdl-37668186

ABSTRACT

Mesenchymal stem cells and macrophages (MQ) are two very important cells involved in the normal wound healing process. It is well understood that topological cues and mechanical factors can lead to different responses in stem cells and MQ by influencing their shape, cytoskeleton proliferation, migration, and differentiation, which play an essential role in the success or failure of biomaterial implantation and more importantly wound healing. On the other hand, the polarization of MQ from proinflammatory (M1) to prohealing (M2) phenotypes has a critical role in the acceleration of wound healing. In this study, the morphology of different MQ subtypes (M0, M1, and M2) was imprinted on a silicon surface (polydimethylsiloxane [PDMS]) to prepare a nano-topography cell-imprinted substrate with the ability to induce anti-inflammatory effects on the mouse adipose-derived stem cells (ADSCs) and RAW264.7 monocyte cell line (MO). The gene expression profiles and flow cytometry of MQ revealed that the cell shape microstructure promoted the MQ phenotypes according to the specific shape of each pattern. The ELISA results were in agreement with the gene expression profiles. The ADSCs on the patterned PDMS exhibited remarkably different shapes from no-patterned PDMS. The MOs grown on M2 morphological patterns showed a significant increase in expression and section of anti-inflammatory cytokine compared with M0 and M1 patterns. The ADSCs homing in niches heavily deformed the cytoskeletal, which is probably why the gene expression and phenotype unexpectedly changed. In conclusion, wound dressings with M2 cell morphology-induced surfaces are suggested as excellent anti-inflammatory and antiscarring dressings.


Subject(s)
Macrophages , Mesenchymal Stem Cells , Mice , Animals , Macrophages/metabolism , Cytokines/metabolism , Wound Healing , Mesenchymal Stem Cells/metabolism , Anti-Inflammatory Agents/pharmacology
2.
Artif Organs ; 47(8): 1267-1284, 2023 Aug.
Article in English | MEDLINE | ID: mdl-36869662

ABSTRACT

BACKGROUND: Enhancing the efficiency of cell-based skin tissue engineering (TE) approaches is possible via designing electrospun scaffolds possessing natural materials like amniotic membrane (AM) with wound healing characteristics. Concentrating on this aim, we fabricated innovative polycaprolactone (PCL)/AM scaffolds through the electrospinning process. METHODS: The manufactured structures were characterized by employing scanning electron microscope (SEM), attenuated total reflection-Fourier transform infrared (ATR-FTIR) spectroscopy, tensile testing, Bradford protein assay, etc. In addition, the mechanical properties of scaffolds were simulated by the multiscale modeling method. RESULTS: As a result of conducting various tests, it was concluded that the uniformity and distribution of fibers decreased with an increase in the amniotic content. Furthermore, PCL-AM scaffolds contained amniotic and PCL characteristic bands. In the case of protein release, greater content of AM led to the release of higher amounts of collagen. Tensile testing revealed that scaffolds' ultimate strength increased when the AM content augmented. The multiscale modeling demonstrated that the scaffold had elastoplastic behavior. In order to assess cellular attachment, viability, and differentiation, human adipose-derived stem cells (ASCs) were seeded on the scaffolds. In this regard, SEM and 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl-2H-tetrazolium bromide (MTT) assays showed significant cellular proliferation and viability on the proposed scaffolds, and these analyses illustrated that higher cell survival and adhesion could be achieved when scaffolds possessed a larger amount of AM. After 21 days of cultivation, particular keratinocyte markers, such as keratin I and involucrin, were identified through utilizing immunofluorescence and real-time polymerase chain reaction (PCR) tests. The markers' expressions were higher in the PCL-AM scaffold with a ratio of 90:10 v v-1 compared with the PCL-epidermal growth factor (EGF) structure. Moreover, the presence of AM in the scaffolds resulted in the keratinogenic differentiation of ASCs even without employing EGF. Consequently, this state-of-the-art experiment suggests that the PCL-AM scaffold can be a promising candidate in skin bioengineering. CONCLUSION: This study showed that mixing AM with PCL, a widely used polymer, in different concentrations can overcome PCL disadvantages such as high hydrophobicity and low cellular compatibility.


Subject(s)
Nanofibers , Tissue Scaffolds , Humans , Tissue Scaffolds/chemistry , Epidermal Growth Factor , Nanofibers/chemistry , Amnion , Wound Healing , Tissue Engineering/methods , Polyesters/chemistry , Cell Proliferation
3.
J Mater Sci Mater Med ; 34(3): 12, 2023 Mar 14.
Article in English | MEDLINE | ID: mdl-36917304

ABSTRACT

Decellularization by chemical approaches has harmful effects on extracellular matrix (ECM) proteins, and damages lots of functional peptides and biomolecules present in the ultrastructure. In this study, we employed a combination of chemical and physical decellularization methods to overcome these disadvantages. The induced osmotic pressure by hypertonic/hypotonic solutions dissociated and removed most of cellular membranes significantly without any detergent or chemical agent. In total, 0.025% trypsin solution was found adequate to remove the remaining debrides, and ultimately 1% Triton X-100 was utilized for final cleansing. In addition, conducting all the decellularization processes at 4 °C yielded an ECM with least damages in the ultrastructure which could be inferred by close mechanical strength and swelling ratio to the native vessel, and high quality and quantity of cell attachment, migration and proliferation which were examined by optical microscopy and scanning electron microscopy (SEM) of the histology samples. Moreover, the obtained biological scaffold (BS) had no cytotoxicity according to the MTT assay, and this scaffold is storable at -20 °C. Employing bioreactor for concurrent cyclic tensile and shear stresses improved the cell migration into pores of the BS and made the cells and the scaffold compact in analogous to native tissue. As opening angle test showed by decellularizing of the blood vessel, the residual stress dropped significantly which revealed the role of cells in the amount of induced stress in the structure. However, intact and healthy ECM explicitly recovered upon recellularization and beat the initial residual stress of the native tissue. The tensile test of the blood vessels in longitudinal and radial directions revealed orthotropic behavior which can be explained by collagen fibers direction in the ECM. Furthermore, by the three regions of the stress-strain curve can be elucidated the roles of cells, elastin and collagen fibers in mechanical behavior of the vascular tissues.


Subject(s)
Extracellular Matrix , Tissue Engineering , Tissue Engineering/methods , Extracellular Matrix/metabolism , Biomimetics , Octoxynol/chemistry , Collagen/chemistry , Tissue Scaffolds/chemistry
4.
J Cell Mol Med ; 26(23): 5929-5942, 2022 12.
Article in English | MEDLINE | ID: mdl-36412036

ABSTRACT

Different growth factors can regulate stem cell differentiation. We used keratinocyte growth factor (KGF) to direct adipose-derived stem cells (ASCs) differentiation into keratinocytes. To enhance KGF bioavailability, we targeted KGF for collagen by fusing it to collagen-binding domain from Vibrio mimicus metalloprotease (vibrioCBD-KGF). KGF and vibrioCBD-KGF were expressed in Escherichia coli and purified to homogeneity. Both proteins displayed comparable activities in stimulating proliferation of HEK-293 and MCF-7 cells. vibrioCBD-KGF demonstrated enhanced collagen-binding affinity in immunofluorescence and ELISA. KGF and vibrioCBD-KGF at different concentrations (2, 10, and 20 ng/ml) were applied for 21 days on ASCs cultured on collagen-coated plates. Keratinocyte differentiation was assessed based on morphological changes, the expression of keratinocyte markers (Keratin-10 and Involucrin), and stem cell markers (Collagen-I and Vimentin) by real-time PCR or immunofluorescence. Our results indicated that the expression of keratinocyte markers was substantially increased at all concentrations of vibrioCBD-KGF, while it was observed for KGF only at 20 ng/ml. Immunofluorescence staining approved this finding. Moreover, down-regulation of Collagen-I, an indicator of differentiation commitment, was more significant in samples treated with vibrioCBD-KGF. The present study showed that vibrioCBD-KGF is more potent in inducing the ASCs differentiation into keratinocytes compared to KGF. Our results have important implications for effective skin regeneration using collagen-based biomaterials.


Subject(s)
Cell Differentiation , Fibroblast Growth Factor 7 , Keratinocytes , Stem Cells , Humans , Collagen , Collagen Type I/genetics , Fibroblast Growth Factor 7/pharmacology , HEK293 Cells , Keratinocytes/cytology , Keratinocytes/drug effects , Stem Cells/cytology , Stem Cells/drug effects
5.
Mol Biol Rep ; 49(6): 4595-4605, 2022 Jun.
Article in English | MEDLINE | ID: mdl-35279778

ABSTRACT

BACKGROUND: The purpose of this research was to investigate the in vitro osteogenic induction of MG-63 cells using topography and collagen protein printed on polydimethyl siloxane (PDMS). METHODS: ALKALINE PHOSPHATASE (ALP) assay, calcium content, alizarin red staining, immunocytochemistry (ICC), and real-time polymerase chain reaction (PCR) were used to evaluate the osteo-differentiation of human adipose stem cells on the MG-63 cell pattern, MG-63 cells/collagen pattern, and collagen pattern. Also, the differentiated cell shape was studied by crystal violet staining and scanning electron microscopy (SEM). RESULTS: Our results showed that calcium content and ALP activity increased significantly on the MG-63 cells /collagen pattern (P < 0.05). The gene expression analysis (ALKALINPHOSPHATASE, COLLAGEN1 and OSTEOCALCIN) and bone marker protein expression (OSTEOCALCIN) confirmed the osteo differentiation of adipose stem cells (ADSCs) seeded on the imprinting substrate. DISCUSSION: Cell and molecular printing enhanced osteogenic development of adipose stem cells, according to our findings.


Subject(s)
Calcium , Tissue Engineering , Adipose Tissue , Calcium/metabolism , Cell Differentiation , Cells, Cultured , Collagen/metabolism , Humans , Osteocalcin/genetics , Osteocalcin/metabolism , Osteogenesis , Tissue Engineering/methods
6.
J Mater Sci Mater Med ; 33(3): 31, 2022 Mar 10.
Article in English | MEDLINE | ID: mdl-35267105

ABSTRACT

Controlled pore size and desirable internal architecture of bone scaffolds play a significant role in bone regeneration efficiency. In addition to choosing appropriate materials, the manufacturing method is another significant factor in fabricating the ideal scaffold. In this study, scaffolds were designed and fabricated by the fused filament fabrication (FFF) technique. Polycaprolactone (PCL) and composites films with various percentages of hydroxyapatite (HA) (up to 20%wt) were used to fabricate filaments. The influence of (HA) addition on the mechanical properties of filaments and scaffolds was investigated. in vitro biological evaluation was examined as well as the apatite formation in simulated body fluid (SBF). The addition of HA particles increased the compressive strength and Young's modulus of filaments and consequently the scaffolds. Compared to PCL, Young's modulus of PCL/HA20% filament and three-dimensional (3D) printed scaffold has increased by 30% and 50%, respectively. Also, Young's modulus for all scaffolds was in the range of 30-70 MPa, which is appropriate to use in spongy bone. Besides, the MTT assay was utilized to evaluate cell viability on the scaffolds. All the samples had qualified cytocompatibility, and it would be anticipated that addition of HA particles raise the biocompatibility in vivo. Alkaline phosphatase (ALP) evaluation shows that the addition of HA caused higher ALP activity in the PCL/HA scaffolds than PCL. Furthermore, calcium deposition in the PCL/HA specimens is higher than control. In conclusion, the addition of HA particles into the PCL matrix, as well as utilizing an inexpensive commercial FFF device, lead to the fabrication of scaffolds with proper mechanical and biological properties for bone tissue engineering applications. Graphical abstract.


Subject(s)
Durapatite , Tissue Engineering , Polyesters , Porosity , Printing, Three-Dimensional , Tissue Engineering/methods , Tissue Scaffolds
7.
Cell Physiol Biochem ; 55(1): 33-60, 2021 Jan 22.
Article in English | MEDLINE | ID: mdl-33474906

ABSTRACT

Many factors including growth factors (GF), scaffold materials, and chemical and physical cues determine the cell behaviors. For many years, growth factors have been considered as the pivotal cell behavior regulators, whereas recent studies emphasize also the key role of physical factors such as mechanical forces, cell shape, surface topographies, and extracellular matrix (ECM) in regulating the cell proliferation, apoptosis, differentiation, etc. through mechanotransduction pathways. In this process, the cell morphology and mechanical properties of the cell's micro/ nano-environments and ECM can be conveyed to the nucleus by regulating transcriptional factors such as Yes-associated protein and transcriptional coactivator with PDZ-binding motif (TAZ). Generally, YAP/TAZ activity is considered as the key factor for the growth of whole organs, however, recent studies have also repeatedly addressed the role of YAP/TAZ in mechanotransduction. In this review, the biological functions of the YAP/TAZ pathway and its contribution to the mechanotransduction and cell behavior regulation in response to the mechanical cues have been summarized. Also, the role of key mechanical checkpoints in the cell including focal adhesions, cytoskeletal tension, Rho small GTPases, and nuclear membrane protein elements involved in the transfer of environmental mechanical cues from the cell surface to the nucleus and their effect in regulating the YAP/TAZ activity are discussed.


Subject(s)
Cell Cycle Proteins/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Mechanotransduction, Cellular , Transcription Factors/metabolism , Cell Shape/physiology , Humans , Transcriptional Coactivator with PDZ-Binding Motif Proteins
8.
J Gene Med ; 22(12): e3288, 2020 12.
Article in English | MEDLINE | ID: mdl-33047833

ABSTRACT

BACKGROUND: Dentin matrix protein 1 (DMP1) is highly expressed in mineralized tooth and bone, playing a critical role in mineralization and phosphate metabolism. One important role for the expression of DMP1 in the nucleus of preosteoblasts is the up-regulation of osteoblast-specific genes such as osteocalcin and alkaline phosphatase1 . The present study aimed to investigate the potential application of human DMP1 promoter as an indicator marker of osteoblastic differentiation. METHODS: In the present study, we developed DMP1 promoter-DsRed-GFP knock-in mesenchymal stem cell (MSCs) via the clustered regularly interspaced short palindromic repeats (CRISPR)/CRISPR-associated protein 9 (Cas9) system that enabled automatic detection of osteoblast differentiation. With the application of a homology-directed knock-in strategy, a 2-kb fragment of DMP1 promoter, which was inserted upstream of the GFP and DsRed reporter cassette, was integrated into the human ROSA locus to generate double fluorescent cells. We further differentiated MSCs under osteogenic media to monitor the fate of MSCs. First, cells were transfected using CRISPR/Cas9 plasmids, which culminated in MSCs with a green fluorescence intensity, then GFP-positive cells were selected using puromycin. Second, the GFP-positive MSCs were differentiated toward osteoblasts, which demonstrated an increased red fluorescence intensity. The osteoblast differentiation of MSCs was also verified by performing alkaline phosphatase and Alizarin Red assays. RESULTS: We have exploited the DMP1 promoter as a predictive marker of MSC differentiation toward osteoblasts. Using the CRISPR/Cas9 technology, we have identified a distinctive change in the fluorescence intensities of GFP knock-in (green) and osteoblast differentiated MSCs 2 . CONCLUSIONS: The data show that DMP1-DsRed-GFP knock-in MSCs through CRISPR/Cas9 technology provide a valuable indicator for osteoblast differentiation. Moreover, The DMP1 promoter might be used as a predictive marker of MSCs differentiated toward osteoblasts.


Subject(s)
CRISPR-Cas Systems , Cell Differentiation , Extracellular Matrix Proteins/antagonists & inhibitors , Gene Knock-In Techniques/methods , Mesenchymal Stem Cells/cytology , Osteoblasts/cytology , Osteogenesis , Phosphoproteins/antagonists & inhibitors , Cell Proliferation , Cells, Cultured , Extracellular Matrix Proteins/genetics , Extracellular Matrix Proteins/metabolism , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Humans , Mesenchymal Stem Cells/metabolism , Osteoblasts/metabolism , Phosphoproteins/genetics , Phosphoproteins/metabolism , Promoter Regions, Genetic
9.
Cell Tissue Res ; 381(2): 255-272, 2020 Aug.
Article in English | MEDLINE | ID: mdl-32405685

ABSTRACT

Mesenchymal stem cells (MSCs) are promising cell candidates for cartilage regeneration. Furthermore, it is important to control the cell-matrix interactions that have a direct influence on cell functions. Providing an appropriate microenvironment for cell differentiation in response to exogenous stimuli is a critical step towards the clinical utilization of MSCs. In this study, hydrogels consisted of different proportions of alginates that were modified using gelatin, collagen type I and arginine-glycine-aspartic acid (RGD) and were evaluated regarding their effects on mesenchymal stem cells. The effect of applying hydrostatic pressure on MSCs encapsulated in collagen-modified alginate with and without chondrogenic medium was evaluated 7, 14 and 21 days after culture, which is a comprehensive evaluation of chondrogenesis in 3D hydrogels with mechanical and chemical stimulants. Alcian blue, safranin O and dimethyl methylene blue (DMMB) staining showed the chondrogenic phenotype of cells seeded in the collagen- and RGD-modified alginate hydrogels with the highest intensity after 21 days of culture. The results of real-time PCR for cartilage-specific extracellular matrix genes indicated the chondrogenic differentiation of MSCs in all hydrogels. Also, the synergic effects of chemical and mechanical stimuli are indicated. The highest expression levels of the studied genes were observed in the cells embedded in collagen-modified alginate by loading after 14 days of exposure to the chondrogenic medium. The effect of using IHP on encapsulated MSCs in modified alginate with collagen type I is equal or even higher than using TGF-beta on encapsulated cells. The results of immunohistochemical assessments also confirmed the real-time PCR data.


Subject(s)
Chondrogenesis , Extracellular Matrix/metabolism , Hydrogels/chemistry , Mechanotransduction, Cellular , Mesenchymal Stem Cells/cytology , Tissue Engineering , Alginates/chemistry , Animals , Cartilage, Articular , Cells, Cultured , Chondrocytes , Collagen Type I/chemistry , Gelatin/chemistry , Male , Peptides/chemistry , Rabbits , Tissue Scaffolds
10.
Biotechnol Appl Biochem ; 66(3): 445-453, 2019 May.
Article in English | MEDLINE | ID: mdl-30817028

ABSTRACT

Cell function regulation is influenced by continuous biochemical and biophysical signal exchange within the body. Substrates with nano/micro-scaled topographies that mimic the physiological niche are widely applied for tissue engineering applications. As the cartilage niche is composed of several stimulating factors, a multifunctional substrate providing topographical features while having the capability of electrical stimulation is presented. Herein, we demonstrate a biocompatible and conductive chondrocyte cell-imprinted substrate using polydimethylsiloxane (PDMS) and carbon nanotubes (CNTs) as conductive fillers. Unlike the conventional silicon wafers or structural photoresist masters used for molding, cell surface topographical replication is challenging as biological cells showed extremely sensitive to chemical solvent residues during molding. The composite showed no significant difference compared with PDMS with regard to cytotoxicity, whereas an enhanced cell adhesion was observed on the conductive composite's surface. Integration of nanomaterials into the cell seeding scaffolds can make tissue regeneration process more efficient.


Subject(s)
Biocompatible Materials/chemistry , Cell Culture Techniques , Chondrocytes/cytology , Dimethylpolysiloxanes/chemistry , Nanotubes, Carbon/chemistry , Animals , Biocompatible Materials/pharmacology , Cell Adhesion/drug effects , Cell Survival/drug effects , Cells, Cultured , Chondrocytes/drug effects , Dimethylpolysiloxanes/pharmacology , Electric Conductivity , Materials Testing , Particle Size , Rabbits , Surface Properties
11.
Artif Organs ; 43(10): E264-E281, 2019 Oct.
Article in English | MEDLINE | ID: mdl-31013365

ABSTRACT

This in vitro study aimed to evaluate the physicochemical and biological activity of the polycaprolactone/chitosan/collagen scaffolds incorporated with 0, 0.5, 3, and 6 wt% of graphene oxide (GO). Using standard tests and MG-63 cells, the characteristics of scaffolds were evaluated, and the behavior of osteoblasts were simulated, respectively. A non-significant decrease in nanofibers diameter was noted in scaffolds with a higher ratio of GO. The hydrophilicity and bioactivity of the scaffold surface, as well as cell attachment and proliferation, increased in correspondence to an increase in GO. The higher ratio of GO also improved the osteogenesis activity. GO increased the degradation rate, but it was negligible and seemed not enough to endanger stability. Modifying the scaffolds with GO did not make a significant change to the antibacterial effect.


Subject(s)
Chitosan/chemistry , Collagen/chemistry , Graphite/chemistry , Polyesters/chemistry , Tissue Scaffolds/chemistry , Biocompatible Materials/chemistry , Cell Line , Humans , Materials Testing , Nanofibers/chemistry , Osteoblasts/cytology , Osteogenesis , Tissue Engineering
12.
Cells Tissues Organs ; 206(6): 317-329, 2018.
Article in English | MEDLINE | ID: mdl-31340210

ABSTRACT

Burn wound treatment is difficult and one of the most challenging problems in the clinic. Researchers have examined the applications of mesenchymal stem cells as a cell-based therapy for skin regeneration. But the role of human bone marrow mesenchymal stem cell conditioned medium (hBM-MSC-CM) in the treatment of burn injury remains unclear. This research aims at detecting whether hBM-MSC-CM can increase the wound healing of deep second-degree burns in male rats. In this study, 32 adult male rats per each time point were randomly divided into four groups: (1) control group, (2) sham group (DMEM), (3) common treatment group (CT), and (4) conditioned media group (CM). A 3 × 3 cm circular burn was created on the back of the rats. On postsurgical days 7, 15, and 28, the wound closure area of each wound was measured and then the skin samples were removed and analyzed using stereological methods. Wound closure area was significantly increased in the CM and CT groups on the 15th and the 28th day after burn injury compared to the control and DMEM groups. The stereological parameters and immunohistochemistry analysis of the wounds revealed significantly improved healing in the CM group compared to the control and other groups. It is concluded that these findings indicate that hBM-MSC-CM promotes skin wound healing by increasing cell proliferation, regulating collagen synthesis and collagen composition, and inducing angiogenesis at the injury site.

13.
Nanotechnology ; 29(28): 285102, 2018 Jul 13.
Article in English | MEDLINE | ID: mdl-29694332

ABSTRACT

Graphene-based nanomaterials contain unique physicochemical properties and have been widely investigated due to a variety of applications particularly in cancer therapy. Furthermore, Ag has been known for its extensive historical background for biomedical applications. Therefore, conjugation of shape-selective Ag nanostructures with graphene may provide new horizons for pharmaceutical applications such as cancer treatments. Here we report on the synthesis of Ag nanoparticles (NPs)/reduced graphene oxide (AgNPs/RGO) conjugate nanomaterials containing various shapes of AgNPs by a novel and simple synthesis route using the deformation of dimethylformamide (DMF) as the reducing and coupling agent. The cytotoxicity and anticancer properties of AgNPs, AgNPs/RGO conjugate nanomaterials, RGO and graphene oxide (GO) were probed against MDA-MB-231 cancer and MCF-10A normal human breast cells in vitro. The AgNPs/RGO nanocomposites exhibited a strong anticancer effect by penetration and apoptosis in cancer cells as well as the lowest influence on the viability of normal cells. It was found that cancer cell viability not only depends on the geometry of Ag nanostructures but also on the interaction between AgNPs and RGO nanoplatelets. It is suggested that AgNPs/RGO conjugate nanomaterials with various shapes of AgNPs is a promising therapeutic platform for cancer therapy.


Subject(s)
Breast Neoplasms/pathology , Graphite/pharmacology , Nanostructures/chemistry , Silver/pharmacology , Cell Death/drug effects , Cell Line, Tumor , Cell Shape/drug effects , Cell Survival/drug effects , Drug Synergism , Endocytosis/drug effects , Female , Humans , Inhibitory Concentration 50 , Nanocomposites/chemistry , Nanocomposites/ultrastructure , Nanostructures/ultrastructure , Particle Size , Spectroscopy, Fourier Transform Infrared , X-Ray Diffraction
14.
J Mater Sci Mater Med ; 30(1): 7, 2018 Dec 29.
Article in English | MEDLINE | ID: mdl-30594964

ABSTRACT

Strategies based on growth factor (GF) delivery have attracted considerable attention in tissue engineering applications. Among different GFs, transforming growth factor beta 1 (TGF-ß1) is considered to be a potent factor for inducing chondrogenesis. In the present study, an expression cassette encoding the TGF-ß1 protein was prepared and transfected into the SP2/0-Ag14 cell line. The confocal microscopy of the transfected cells was performed to confirm the correct transfection process. The expression and in vitro release kinetics of the recombinant TGF-ß1 were assessed by western blot analysis and ELISA, respectively. Moreover, the biological activity of the expressed protein was compared with that of a commercially available product. The chondrogenic effects of the sustained release of the recombinant TGF-ß1 in an in vitro co-culture system were evaluated using a migration assay and real-time PCR. Results of confocal microscopy confirmed the successful transfection of the vector-encoding TGF-ß1 protein into the SP2/0-Ag14 cells. The bioactivity of the produced protein was in the range of the commercial product. The sustained release of the TGF-ß1 protein via SP2/0-Ag14 cells encapsulated in hydrogels encouraged the migration of adipose-derived MSCs. In addition, the expression analysis of chondrogenesis-related genes revealed that the pretreatment of encapsulated Ad-MSCs cells in alginate sulfate hydrogels through their exposure to the sustained release of TGF-ß1 is an efficient approach before transplantation of cells into the body.


Subject(s)
Cell Differentiation/drug effects , Chondrogenesis/physiology , Mesenchymal Stem Cells/drug effects , Transforming Growth Factor beta1/metabolism , Transforming Growth Factor beta1/pharmacology , Alginates/chemistry , Animals , Cell Line , Mesenchymal Stem Cells/physiology , Mice , Transforming Growth Factor beta1/genetics
15.
Lasers Med Sci ; 31(5): 863-73, 2016 Jul.
Article in English | MEDLINE | ID: mdl-27025859

ABSTRACT

Soft tissue seal plays a critical role in long-term success of dental implants, and the effects of implant surface treatments such as laser ablation have been a topic of particular interest in this respect. Considering the existing controversy regarding soft tissue behavior in contact with implant surfaces, this study sought to assess the morphology, proliferation, and gene expression of human gingival fibroblasts (HGFs) on different abutment surfaces. In this in vitro, experimental study, HGFs were cultured on 45 discs (Laser-Lok, titanium, and zirconia). Cell morphology, proliferation rate, and interleukin 10 (IL-10), tumor necrosis factor alpha (TNFα), fibronectin, and integrin gene expressions were assessed by electron microscopy, methyl thiazol tetrazolium (MTT) assay, and real-time polymerase chain reaction (PCR), respectively. Data were analyzed using ANOVA and the Kruskal-Wallis H test. Fibroblast attachment was noted in all the three groups. Spindle-shaped cells with pseudopod-like processes were more frequently seen in the Laser-Lok group. Cell proliferation was significantly higher in the Laser-Lok group compared to those in the other groups (P = 0.0002). Significant differences were found in the expression of IL-10, TNFα, fibronectin, and integrin genes among the groups (P < 0.01). Within the limitations of this study, HGFs on Laser-Lok surfaces had a more mature morphology and greater proliferation and differentiation as compared to those on zirconia and titanium surfaces. This indicates better attachment of these cells to laser-modified surfaces, creating a more efficient soft tissue seal around dental implants.


Subject(s)
Cell Proliferation/radiation effects , Fibroblasts/radiation effects , Gene Expression/radiation effects , Gingiva/cytology , Low-Level Light Therapy/instrumentation , Low-Level Light Therapy/methods , Cells, Cultured , Dental Implants , Fibronectins/radiation effects , Humans , Real-Time Polymerase Chain Reaction , Surface Properties , Titanium , Zirconium
16.
Int J Biol Macromol ; 277(Pt 4): 134412, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39097043

ABSTRACT

Injectable hydrogels are promising for bone tissue engineering due to their minimally invasive application and adaptability to irregular defects. This study presents the development of pluronic grafted silk fibroin (PF-127-g-SF), a temperature-sensitive graft copolymer synthesized from SF and modified PF-127 via a carbodiimide coupling reaction. The PF-127-g-SF copolymer exhibited a higher sol-gel transition temperature (34 °C at 16 % w/v) compared to PF-127 (23 °C), making it suitable for injectable applications. It also showed improved flexibility and strength, with a yielding point increase from <10 % to nearly 30 %. Unlike PF-127 gel, which degrades within 72 h in aqueous media, the PF-127-g-SF copolymer maintained a stable gel structure for over two weeks due to its robust crosslinked hydrogel network. Incorporating hydroxyapatite nanoparticles (n-HA) into the hydrogel reduced pore size and decreased swelling and degradation rates, extending structural stability to four weeks. Increasing n-HA concentration from 0 % to 20 % reduced porosity from 80 % to 66 %. Rheological studies indicated that n-HA enhanced the scaffold's strength and mechanical properties without altering gelation temperature. Cellular studies with MG-63 cells showed that n-HA concentration influenced cell viability and mineralization, highlighting the scaffold's potential in bone tissue engineering.


Subject(s)
Durapatite , Fibroins , Hydrogels , Nanoparticles , Poloxamer , Temperature , Tissue Engineering , Fibroins/chemistry , Tissue Engineering/methods , Durapatite/chemistry , Poloxamer/chemistry , Nanoparticles/chemistry , Hydrogels/chemistry , Hydrogels/chemical synthesis , Hydrogels/pharmacology , Humans , Bone and Bones/drug effects , Tissue Scaffolds/chemistry , Rheology , Injections , Porosity , Biocompatible Materials/chemistry
17.
Stem Cell Rev Rep ; 2024 Jul 27.
Article in English | MEDLINE | ID: mdl-39066936

ABSTRACT

The influence of surface topography on stem cell behavior and differentiation has garnered significant attention in regenerative medicine and tissue engineering. The cell-imprinting method has been introduced as a promising approach to mimic the geometry and topography of cells. The cell-imprinted substrates are designed to replicate the topographies and dimensions of target cells, enabling tailored interactions that promote the differentiation of stem cells towards desired specialized cell types. In fact, by replicating the size and shape of cells, biomimetic substrates provide physical cues that profoundly impact stem cell differentiation. These cues play a pivotal role in directing cell morphology, cytoskeletal organization, and gene expression, ultimately influencing lineage commitment. The biomimetic substrates' ability to emulate the native cellular microenvironment supports the creation of platforms capable of steering stem cell fate with high precision. This review discusses the role of mechanical factors that impact stem cell fate. It also provides an overview of the design and fabrication principles of cell-imprinted substrates. Furthermore, the paper delves into the use of cell-imprinted polydimethylsiloxane (PDMS) substrates to direct adipose-derived stem cells (ADSCs) differentiation into a variety of specialized cells for tissue engineering and regenerative medicine applications. Additionally, the review discusses the limitations of cell-imprinted PDMS substrates and highlights the efforts made to overcome these limitations.

18.
Bioimpacts ; 14(3): 29945, 2024.
Article in English | MEDLINE | ID: mdl-38938752

ABSTRACT

Cell culture-based technologies are widely utilized in various domains such as drug evaluation, toxicity assessment, vaccine and biopharmaceutical development, reproductive technology, and regenerative medicine. It has been demonstrated that pre-adsorption of extracellular matrix (ECM) proteins including collagen, laminin and fibronectin provide more degrees of support for cell adhesion. The purpose of cell imprinting is to imitate the natural topography of cell membranes by gels or polymers to create a reliable environment for the regulation of cell function. The results of recent studies show that cell imprinting is a tool to guide the behavior of cultured cells by controlling their adhesive interactions with surfaces. Therefore, in this review we aim to compare different cell cultures with the imprinting method and discuss different cell imprinting applications in regenerative medicine, personalized medicine, disease modeling, and cell therapy.

19.
J Biomed Mater Res B Appl Biomater ; 112(2): e35346, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38359175

ABSTRACT

Improvement of mechanical properties of injectable tissue engineering scaffolds is a current challenge. The objective of the current study is to produce a highly porous injectable scaffold with improved mechanical properties. For this aim, cellulose nanocrystals-reinforced dual crosslinked porous nanocomposite cryogels were prepared using chemically crosslinked methacrylated gelatin (GelMA) and ionically crosslinked hyaluronic acid (HA) through the cryogelation process. The resulting nanocomposites showed highly porous structures with interconnected porosity (>90%) and mean pore size in the range of 130-296 µm. The prepared nanocomposite containing 3%w/v of GelMA, 20 w/w% of HA, and 1%w/v of CNC showed the highest Young's modulus (10 kPa) and excellent reversibility after 90% compression and could regain its initial shape after injection by a 16-gauge needle in the aqueous media. The in vitro results demonstrated acceptable viability (>90%) and migration of the human chondrocyte cell line (C28/I2), and chondrogenic differentiation of human adipose stem cells. A two-month in vivo assay on a rabbit's ear model confirmed that the regeneration potential of the prepared cryogel is comparable to the natural autologous cartilage graft, suggesting it is a promising alternative for autografts in the treatment of cartilage defects.


Subject(s)
Nanocomposites , Nanoparticles , Animals , Rabbits , Humans , Cryogels/pharmacology , Cryogels/chemistry , Hyaluronic Acid/pharmacology , Hyaluronic Acid/chemistry , Gelatin/pharmacology , Gelatin/chemistry , Cellulose/pharmacology , Cellulose/chemistry , Tissue Scaffolds/chemistry , Cartilage , Tissue Engineering/methods , Nanoparticles/chemistry , Porosity
20.
Int J Biol Macromol ; 277(Pt 1): 133995, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39038571

ABSTRACT

This study investigated the efficacy of using chitosan/alginate nanoparticles loaded with recombinant human bone morphogenetic-2 (rhBMP-2) and SMAD4 encoding plasmid to enhance the chondrogenesis of human bone marrow mesenchymal stem cells (hBM-MSCs) seeded on an extracellular matrix (ECM). The research treatments included the stem cells treated with the biological cocktail (BC), negative control (NC), hBM-MSCs with chondrogenic medium (MCM), hBM-MSCs with naked rhBMP-2 and chondrogenic medium (NB/C), and hBM-MSCs with naked rhBMP-2 and chondrogenic medium plus SMAD4 encoding plasmid transfected with polyethyleneimine (PEI) (NB/C/S/P). The cartilage differentiation was performed with real-time quantitative PCR analysis and alizarin blue staining. The data indicated that the biological cocktail (BC) exhibited significantly higher expression of cartilage-related genes compared to significant differences with MCM and negative control (NC) on chondrogenesis. In the (NB/C/S/P), the expression levels of SOX9 and COLX were lower than those in the BC group. The expression pattern of the ACAN gene was similar to COL2A1 changes suggesting that it holds promising potential for cartilage regeneration.


Subject(s)
Alginates , Bone Morphogenetic Protein 2 , Cartilage, Articular , Chitosan , Chondrogenesis , Extracellular Matrix , Mesenchymal Stem Cells , Nanoparticles , Regeneration , Signal Transduction , Smad4 Protein , Tissue Scaffolds , Mesenchymal Stem Cells/metabolism , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/drug effects , Chitosan/chemistry , Chitosan/pharmacology , Alginates/chemistry , Alginates/pharmacology , Humans , Cartilage, Articular/drug effects , Cartilage, Articular/metabolism , Cartilage, Articular/cytology , Bone Morphogenetic Protein 2/metabolism , Bone Morphogenetic Protein 2/genetics , Nanoparticles/chemistry , Chondrogenesis/drug effects , Tissue Scaffolds/chemistry , Smad4 Protein/metabolism , Smad4 Protein/genetics , Signal Transduction/drug effects , Extracellular Matrix/metabolism , Regeneration/drug effects , Cell Differentiation/drug effects , SOX9 Transcription Factor/metabolism , SOX9 Transcription Factor/genetics , Recombinant Proteins/pharmacology , Transforming Growth Factor beta
SELECTION OF CITATIONS
SEARCH DETAIL