Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 30
Filter
Add more filters










Publication year range
1.
BMC Biotechnol ; 24(1): 23, 2024 Apr 26.
Article in English | MEDLINE | ID: mdl-38671404

ABSTRACT

Volumetric loss is one of the challenging issues in muscle tissue structure that causes functio laesa. Tissue engineering of muscle tissue using suitable hydrogels is an alternative to restoring the physiological properties of the injured area. Here, myogenic properties of type I collagen (0.5%) and keratin (0.5%) were investigated in a mouse model of biceps femoris injury. Using FTIR, gelation time, and rheological analysis, the physicochemical properties of the collagen (Col)/Keratin scaffold were analyzed. Mouse C2C12 myoblast-laden Col/Keratin hydrogels were injected into the injury site and histological examination plus western blotting were performed to measure myogenic potential after 15 days. FTIR indicated an appropriate interaction between keratin and collagen. The blend of Col/Keratin delayed gelation time when compared to the collagen alone group. Rheological analysis revealed decreased stiffening in blended Col/Keratin hydrogel which is favorable for the extrudability of the hydrogel. Transplantation of C2C12 myoblast-laden Col/Keratin hydrogel to injured muscle tissues led to the formation of newly generated myofibers compared to cell-free hydrogel and collagen groups (p < 0.05). In the C2C12 myoblast-laden Col/Keratin group, a low number of CD31+ cells with minimum inflammatory cells was evident. Western blotting indicated the promotion of MyoD in mice that received cell-laden Col/Keratin hydrogel compared to the other groups (p < 0.05). Despite the increase of the myosin cell-laden Col/Keratin hydrogel group, no significant differences were obtained related to other groups (p > 0.05). The blend of Col/Keratin loaded with myoblasts provides a suitable myogenic platform for the alleviation of injured muscle tissue.


Subject(s)
Keratins , Muscle Development , Muscle, Skeletal , Animals , Mice , Muscle, Skeletal/injuries , Muscle, Skeletal/metabolism , Keratins/metabolism , Cell Line , Hydrogels/chemistry , Neovascularization, Physiologic/drug effects , Tissue Engineering/methods , Disease Models, Animal , Collagen/metabolism , Myoblasts/metabolism , Myoblasts/cytology , Male , Tissue Scaffolds/chemistry , Angiogenesis
2.
Int J Hematol Oncol Stem Cell Res ; 17(4): 245-256, 2023 Oct 01.
Article in English | MEDLINE | ID: mdl-38076776

ABSTRACT

Background: Platelets play a key role in the treatment of thrombocytopenia. Nowadays, platelets (PLTs) are only obtained through blood donation. However, due to the limitations in their preparation and storage, they are produced in laboratories, especially through bioreactors that convert megakaryocytes from stem cells into large-scale injectable PLTs. Materials and Methods: In this study, the CD34 cells isolated from cord blood were differentiated into megakaryocytes. A 6-chamber bioreactor with a two-layer collagen scaffold, several ECM factors, and human cryoprecipitate were used to simulate the structure of the bone marrow. After the addition of megakaryocytes to the scaffold, PLTs were produced due to the flow pressure and the interaction between the scaffold structure and the ECM factors. Results: CD41 + cells were expanded 100 times as much as CD34 + cord blood stem cells. The mean PLT release from one megakaryocyte in the pure collagen scaffold was 17.42 PLTs. Once fibrin, fibronectin, hyaluronic acid, and cryoprecipitates were added to collagen, the mean PLT release was 21.4, 22.4, 23.9, and 27.37, respectively. With the simultaneous addition of three factors to collagen (CFFH) and then four factors (CFFHC), the number of PLTs reached 30.52 and then 34. Conclusion: Functional PLTs can be produced on a large scale with a multi-chamber bioreactor using a combination of ECM and cryoprecipitate with collagen scaffolding.

3.
Int J Biol Macromol ; 253(Pt 5): 127209, 2023 Dec 31.
Article in English | MEDLINE | ID: mdl-37804896

ABSTRACT

Osteogenic properties of phenolated alginate (1.2 %) hydrogel containing collagen (0.5 %)/nano-hydroxyapatite (1 %) were studied on human mesenchymal stem cells in vitro. The phenolation rate and physical properties of the hydrogel were assessed using nuclear magnetic resonance (NMR), Fourier-transform infrared spectroscopy (FTIR), Scanning electron microscope (SEM), swelling ratio, gelation time, mechanical assay, and degradation rate. The viability of encapsulated cells was monitored on days 7, 14, and 21 using an MTT assay. Osteoblast differentiation was studied using western blotting, and real-time PCR. Using PCR array analysis, the role of the Wnt signaling pathway was also investigated. Data showed that the combination of alginate/collagen/nanohydroxyapatite yielded proper mechanical features. The addition of nanohydroxyapatite, and collagen reduced degradation, swelling rate coincided with increased stiffness. Elasticity and pore size were also diminished. NMR and FTIR revealed suitable incorporation of collagen and nanohydroxyapatite in the structure of alginate. Real-time PCR analysis and western blotting indicated the expression of osteoblast-related genes such as Runx2 and osteocalcin. PCR array revealed the induction of numerous genes related to Wnt signaling pathways during the maturation of human stem cells toward osteoblast-like cells. In vivo data indicated that transplantation of phenolated alginate/collagen/nanohydroxyapatite hydrogel led to enhanced de novo bone formation in rats with critical-sized calvarial defects. Phenolated alginate hydrogel can promote the osteogenic capacity of human amniotic membrane mesenchymal stem cells in the presence of nanohydroxyapatite and collagen via engaging the Wnt signaling pathway.


Subject(s)
Mesenchymal Stem Cells , Osteogenesis , Humans , Rats , Animals , Hydrogels/chemistry , Wnt Signaling Pathway , Alginates/chemistry , Collagen/metabolism , Cell Differentiation , Cells, Cultured , Tissue Scaffolds/chemistry
4.
J Biol Eng ; 17(1): 38, 2023 Jun 05.
Article in English | MEDLINE | ID: mdl-37277832

ABSTRACT

To evaluate shear stress influence on ex vivo expansion of hematopoietic cell lineages for clinical application, in this study, human pro-monocytic cell (namely U937 cell line) was selected as a hematopoietic stem cell (HSC) model and cultured in suspension mode at two different agitation rates (50, 100 rpm) in the stirred bioreactor. At the agitation rate of 50 rpm, the cells achieved higher expansion folds (27.4 fold) with minimal morphological changes as well as apoptotic cell death, while at 100 rpm the expansion fold decreased after 5-day of culture in suspension culture in comparison with static culture and reached 24.5 fold at the end of the culture. The results of glucose consumption and lactate production were also in agreement with the data of fold expansion and indicated the preference of culture in the stirred bioreactor when agitated at 50 rpm. This study indicated the stirred bioreactor system with an agitation rate of 50 rpm and surface aeration may be used as a potential dynamic culture system for clinical applications of hematopoietic cell lineage. The current experiments shed data related to the effect of shear stress on human U937 cells, as a hematopoietic cell model, to set a protocol for expansion of HSCs for biomedical applications.

5.
J Cardiovasc Thorac Res ; 14(3): 172-179, 2022.
Article in English | MEDLINE | ID: mdl-36398045

ABSTRACT

Introduction: Here, we monitored the cytocompatibility of scaffolds consisting of poly (glycerol sebacate) (PGS) coated with collagen (Col) for endothelial cell activity after 72 hours. Methods: Human endothelial cells were allocated into Control, PGS, and PGS+Col groups. Scaffolds were characterized using FTIR and HNMR spectroscopy. Contact angel analysis and SEM were used to study wettability, surface morphology, and cell attachment. Cell survival was assessed using LDH leakage assay. Levels of Tie-1, Tie-2, VE-Cadherin, and VEGFR-2 were measured using western blotting and real-time PCR. Results: FTIR and HNMR analyses revealed the proper blending in PGS+Col group. SEM imaging exhibited a flat surface in the PGS group while thin Col fibers were detected in PGS+Col surface. The addition of Col to the PGS reduced the contract angle values from 97.3˚ to 81.1˚. Compared to PGS substrate alone, in PGS+Col group, cells appropriately attached to the surface. PGS and PGS+Col did not alter the leakage of LDH to the supernatant compared to control cells, showing the cytocopatiblity of PGS-based scaffolds. SOD and NO levels were increased significantly in PGS (p<0.05) and PGS+Col groups (p<0.001), respectively. We found that PGS+Col decreased Tie-1 content in endothelial cells whereas protein levels of Tie-2 and VE-Cadherin and expression of VEGFR-2 remained unchanged compared to PGS and control groups. Conclusion: Simultaneous application of Col and PGS can stimulate normal endothleial cell morphology without the alteration of tyrosine kinases receptors and cadherin.

6.
J Nanobiotechnology ; 20(1): 310, 2022 Jun 28.
Article in English | MEDLINE | ID: mdl-35765003

ABSTRACT

BACKGROUND: Hydrogels based on organic/inorganic composites have been at the center of attention for the fabrication of engineered bone constructs. The establishment of a straightforward 3D microenvironment is critical to maintaining cell-to-cell interaction and cellular function, leading to appropriate regeneration. Ionic cross-linkers, Ca2+, Ba2+, and Sr2+, were used for the fabrication of Alginate-Nanohydroxyapatite-Collagen (Alg-nHA-Col) microspheres, and osteogenic properties of human osteoblasts were examined in in vitro and in vivo conditions after 21 days. RESULTS: Physicochemical properties of hydrogels illustrated that microspheres cross-linked with Sr2+ had reduced swelling, enhanced stability, and mechanical strength, as compared to the other groups. Human MG-63 osteoblasts inside Sr2+ cross-linked microspheres exhibited enhanced viability and osteogenic capacity indicated by mineralization and the increase of relevant proteins related to bone formation. PCR (Polymerase Chain Reaction) array analysis of the Wnt (Wingless-related integration site) signaling pathway revealed that Sr2+ cross-linked microspheres appropriately induced various signaling transduction pathways in human osteoblasts leading to osteogenic activity and dynamic growth. Transplantation of Sr2+ cross-linked microspheres with rat osteoblasts into cranium with critical size defect in the rat model accelerated bone formation analyzed with micro-CT and histological examination. CONCLUSION: Sr2+ cross-linked Alg-nHA-Col hydrogel can promote functionality and dynamic growth of osteoblasts.


Subject(s)
Osteogenesis , Strontium , Alginates/pharmacology , Animals , Collagen , Durapatite , Hydrogels/pharmacology , Rats , Strontium/pharmacology
7.
Stem Cell Res Ther ; 13(1): 143, 2022 04 04.
Article in English | MEDLINE | ID: mdl-35379318

ABSTRACT

BACKGROUND: The bone tissue engineering (BTE) approach has been introduced as an alternative to conventional treatments for large non-healing bone defects. Magnetism promotes stem cells' adherence to biocompatible scaffolds toward osteoblast differentiation. Furthermore, osteogenic differentiation media are expensive and any changes in its composition affect stem cells differentiation. Moreover, media growth factors possess a short half-life resulting in the rapid loss of their functions in vivo. With the above in mind, we fabricated a multilayered nanocomposite scaffold containing the wild type of Type I collagen (Col I) with endogenous magnetic property to promote osteogenesis in rat ADSCs with the minimum requirement of osteogenic differentiation medium. METHODS: Fe3O4 NPs were synthesized by co-precipitation method and characterized using SEM, VSM, and FTIR. Then, a PCL/Col I nanocomposite scaffold entrapping Fe3O4 NPs was fabricated by electrospinning and characterized using SEM, TEM, AFM, VSM, Contact Angle, tensile stretching, and FTIR. ADSCs were isolated from rat adipose tissue and identified by flow cytometry. ADSCs were loaded onto PCL/Col I and PCL/Col I/Fe3O4-scaffolds for 1-3 weeks with/without osteogenic media conditions. The cell viability, cell adhesion, and osteogenic differentiation were evaluated using MTT assay, SEM, DAPI staining, ALP/ARS staining, RT-PCR, and western blotting, respectively. RESULTS: SEM, VSM, and FTIR results indicated that Fe3O4 was synthesized in nano-sized (15-30 nm) particles with spherical-shaped morphology and superparamagnetic properties with approved chemical structure as FTIR revealed. According to SEM images, the fabricated magnetic scaffolds consisted of nanofiber (500-700 nm). TEM images have shown the Fe3O4 NPs entrapped in the scaffold's fiber without bead formation. FTIR spectra analysis confirmed the maintenance of the natural structure of Col I, PCL, and Fe3O4 upon electrospinning. AFM data have shown that MNPs incorporation introduced stripe-like topography to nanofibers, while the depth of the grooves has decreased from 800 to 500 nm. Flow cytometry confirmed the phenotype of ADSCs according to their surface markers (i.e., CD29 and CD105). Additionally, Fe3O4 NP improved nanocomposite scaffold strength, wettability, porosity, biocompatibility and also facilitates the ALP activity, calcium-mineralization. Finally, magnetic nanocomposite scaffolds upregulated osteogenic-related genes or proteins' expression (e.g., Col I, Runx2, OCN, ON, BMP2) in seeded ADSCs with/without osteo-differentiation media conditions. CONCLUSIONS: Together, these results indicate that Fe3O4 NPs within the natural structure of Col I increase osteogenic differentiation in osteogenic cues-free media conditions. This effect could be translated in vivo toward bone defects healing. These findings support the use of natural ECM materials alongside magnetic particles as composite scaffolds to achieve their full therapeutic potential in BTE treatments.


Subject(s)
Nanocomposites , Osteogenesis , Animals , Cells, Cultured , Magnetic Phenomena , Osteogenesis/genetics , Rats , Tissue Scaffolds/chemistry
8.
AAPS PharmSciTech ; 23(3): 80, 2022 Mar 01.
Article in English | MEDLINE | ID: mdl-35233687

ABSTRACT

Generally, supersaturation of weakly basic drug solution in the gastrointestinal tract can be followed by precipitation, and this can compromise the bioavailability of drugs. The purpose of this study was to evaluate the effect of Eudragit® S100 on the pH-induced supersaturation of cinnarizine and to examine the preserving mechanism of cinnarizine supersaturation by Eudragit®. Variables, including pH of media, ionic strength, and degree of supersaturation, were studied to investigate the effects of these parameters on cinnarizine supersaturation in the presence and absence of Eudragit®. The size of the Eudragit® aggregate in solution using dynamic light scattering was determined. The effect of Eudragit® on the transport of cinnarizine through the Caco-2 membrane was also investigated. The particle size study of Eudragit® aggregates showed that the size of these aggregates become large when the pH was lowered. Supersaturation experiments also demonstrated that Eudragit® preserved higher cinnarizine supersaturation with increasing ionic strength of the solution. The phase separation behavior of cinnarizine solution as a function of the degree of the supersaturation could be readily explained by considering the drug amorphous solubility. In vitro permeation studies revealed that the rate of cinnarizine permeation across Caco-2 cells increased in the presence of Eudragit®. According to the obtained results, the aggregation status of Eudragit® and nonspecific hydrophobic cinnarizine-Eudragit® interactions seemed to be essential in determining the effect of Eudragit® on cinnarizine supersaturation.


Subject(s)
Cinnarizine , Caco-2 Cells , Cinnarizine/chemistry , Humans , Polymethacrylic Acids/chemistry , Solubility
9.
Carbohydr Polym ; 277: 118807, 2022 Feb 01.
Article in English | MEDLINE | ID: mdl-34893227

ABSTRACT

The addition of nano-hydroxyapatite (nHA) and collagen (Col) to the alginate (Alg) microcapsule hydrogel reduced swelling and degradation ratios while the compressive strength increased compared to Alg, Alg-Col, and Alg-nHA groups. MTT assay and Calcein-AM staining revealed an enhanced MG-63 osteoblasts viability in the Alg-nHA-Col hydrogel compared to the other groups. SEM showed the attachment of MG-63 osteoblasts inside Alg-Col hydrogels. Non-significant differences were found in antioxidant capacity of cells inside the Alg-nHA-Col hydrogel compared to the Alg group. Hematoxylin-Eosin staining showed the distribution of MG-63 osteoblasts inside microspheres. Calcium deposits, alkaline phosphatase (ALP) activity with the increase of intracellular calcium were found in Alg-nHA-Col group. Western blotting showed that levels of osteocalcin, ColA2, Sox-9, and ColA1 also significantly increased compared to the Alg, Alg-Col, Alg-nHA groups. The present study demonstrated that the addition of mineral nHA and protein (Col) into the Alg improves osteogenic potential and provides a 3D platform for modular bone tissue engineering.


Subject(s)
Alginates/pharmacology , Biocompatible Materials/pharmacology , Collagen/pharmacology , Durapatite/pharmacology , Nanoparticles/chemistry , Osteogenesis/drug effects , Alginates/chemistry , Biocompatible Materials/chemistry , Bone Regeneration/drug effects , Cells, Cultured , Collagen/chemistry , Durapatite/chemistry , Humans , Microspheres , Tissue Engineering , Tissue Scaffolds/chemistry
10.
BMC Res Notes ; 14(1): 384, 2021 Sep 28.
Article in English | MEDLINE | ID: mdl-34583753

ABSTRACT

OBJECTIVE: We aimed to detect the effect of a couple of parameters including Alg, H2O2, and HRP concentrations on the gelation time of Alg-based hydrogels using an enzymatic cross-linked procedure. RESULTS: NMR, UV-Vis, and ATR-FTIR analyses confirmed the conjugation of Ph to the Alg backbone. Data showed gelation time was delayed with the increase and reduction of H2O2 and HRP, respectively. We noted that hydrogel consisted of 1.2% (w/v) Alg, 5 U HRP, and 100 mM H2O2 yielded an appropriate gelation time with appropriate mechanical properties. The addition of 0.5% (v/v) Col developed hydrogel increased the gelation time. The data showed that Alg, HRP, and H2O2 with the ratio of 1:0.54:0.54 had proper physicochemical features for cartilage engineering.


Subject(s)
Cartilage, Articular , Hydrogels , Alginates , Horseradish Peroxidase , Hydrogen Peroxide , Tissue Engineering
11.
J Biol Eng ; 15(1): 14, 2021 Apr 17.
Article in English | MEDLINE | ID: mdl-33865460

ABSTRACT

A small scale stirred bioreactor was designed and the effect of different agitation rates (30, 60 and 100 rpm) was investigated on HepG2 cells cultured in alginate-chitosan (AC) core-shell microcapsule in terms of the cell proliferation and liver-specific function. The microencapsulated hepatic cells could proliferate well when they were cultured for 10 days at 30 rpm while the cell-laden microcapsules showed no cell proliferation at 100 rpm in the bioreactor system. Albumin production rate, as an important liver function, increased also 1.8- and 1.5- fold under stirring rate of 30 rpm compared to the static culture and 60 rpm of agitation, respectively. Moreover, In comparison with the static culture, about 1.5-fold increment in urea production was observed at 30 rpm. Similarly, the highest expressions of albumin and P450 genes were found at 30 rpm stirring rate, which were 4.9- and 19.2-fold of the static culture. Addition of collagen to the microcapsule core composition (ACol/C) could improve the cell proliferation and functionality at 60 rpm in comparison with the cell-laden microcapsules without collagen. The study demonstrated the hepatic cell-laden ACol/C microcapsule hydrogel cultured in the small scale stirred bioreactor at low mixing rate has a great potential for mass production of the hepatic cells while maintaining liver-specific functions.

12.
Biotechnol Lett ; 43(5): 959-966, 2021 May.
Article in English | MEDLINE | ID: mdl-33554302

ABSTRACT

OBJECTIVE: The novel engineered bioprocess, which was designed and modeled to provide the clinically relevant cell numbers for different therapies in our previous work (Kaleybar et al. Food Bioprod Process 122:254-268, https://doi.org/10.1016/j.fbp.2020.04.012 , 2020), was evaluated by using U937 as hematopoietic model cells. RESULTS: The culture system showed a 30-fold expansion of U937 cells in one-step during a 10-day culture period. The cell growth profile, the substrate and oxygen consumptions, and byproduct formations were all in agreement with the model predications during 7 days. The cell proliferation decrease after 7 days was attributed to optional oxygen limiting condition in the last days of culture. The bioreactor culture system revealed also a slight enhancement of lactate dehydrogenase (LDH) production as compared to the 2D conventional culture system, indicating the low impact of shear stress on cellular damage in the dynamic system. CONCLUSIONS: The results demonstrated that the conceptual bioprocess for suspended stem cell production has a great potential in practice although additional experiments are required to improve the system.


Subject(s)
Batch Cell Culture Techniques/methods , Hematopoietic Stem Cells/cytology , Batch Cell Culture Techniques/instrumentation , Bioreactors , Cell Proliferation , Cell Survival , Culture Media/chemistry , Culture Media/metabolism , Hematopoietic Stem Cells/metabolism , Humans , Models, Biological , Oxygen/analysis , U937 Cells
13.
Stem Cell Rev Rep ; 17(4): 1294-1311, 2021 08.
Article in English | MEDLINE | ID: mdl-33547591

ABSTRACT

The homeostasis of osteochondral tissue is tightly controlled by articular cartilage chondrocytes and underlying subchondral bone osteoblasts via different internal and external clues. As a correlate, the osteochondral region is frequently exposed to physical forces and mechanical pressure. On this basis, distinct sets of substrates and physicochemical properties of the surrounding matrix affect the regeneration capacity of chondrocytes and osteoblasts. Stem cells are touted as an alternative cell source for the alleviation of osteochondral diseases. These cells appropriately respond to the physicochemical properties of different biomaterials. This review aimed to address some of the essential factors which participate in the chondrogenic and osteogenic capacity of stem cells. Elements consisted of biomechanical forces, electrical fields, and biochemical and physical properties of the extracellular matrix are the major determinant of stem cell differentiation capacity. It is suggested that an additional certain mechanism related to signal-transduction pathways could also mediate the chondro-osteogenic differentiation of stem cells. The discovery of these clues can enable us to modulate the regeneration capacity of stem cells in osteochondral injuries and lead to the improvement of more operative approaches using tissue engineering modalities.


Subject(s)
Chondrogenesis , Osteogenesis , Stem Cells , Tissue Engineering , Humans , Regeneration
14.
BMC Biotechnol ; 21(1): 6, 2021 01 11.
Article in English | MEDLINE | ID: mdl-33430842

ABSTRACT

BACKGROUND: Microcapsule is considered as a promising 3D microenvironment for Bone Tissue Engineering (BTE) applications. Microencapsulation of cells in an appropriate scaffold not only protected the cells against excess stress but also promoted cell proliferation and differentiation. Through the current study, we aimed to microcapsulate the human Dental Pulp Stem Cells (hDPSCs) and evaluated the proliferation and osteogenic differentiation of those cells by using MTT assay, qRT-PCR, Alkaline phosphatase, and Alizarine Red S. RESULTS: The SEM results revealed that Alg/Gel microcapsules containing nHA showed a rough and more compact surface morphology in comparison with the Alg/Gel microcapsules. Moreover, the microencapsulation by Alg/Gel/nHA could improve cell proliferation and induce osteogenic differentiation. The cells cultured in the Alg/Gel and Alg/Gel/nHA microcapsules showed 1.4-fold and 1.7-fold activity of BMP-2 gene expression more in comparison with the control group after 21 days. The mentioned amounts for the BMP-2 gene were 2.5-fold and 4-fold more expression for the Alg/Gel and Alg/Gel/nHA microcapsules after 28 days. The nHA, addition to hDPSCs-laden Alg/Gel microcapsule, could up-regulate the bone-related gene expressions of osteocalcin, osteonectin, and RUNX-2 during the 21 and 28 days through the culturing period, too. Calcium deposition and ALP activities of the cells were observed in accordance with the proliferation results as well as the gene expression analysis. CONCLUSION: The present study demonstrated that microencapsulation of the hDPSCs inside the Alg/Gel/nHA hydrogel could be a potential approach for regenerative dentistry in the near future.


Subject(s)
Alginates/pharmacology , Capsules/pharmacology , Cell Differentiation/drug effects , Dental Pulp/metabolism , Durapatite/pharmacology , Gelatin/pharmacology , Osteogenesis/drug effects , Stem Cells/metabolism , Alginates/chemistry , Alkaline Phosphatase/metabolism , Calcium , Cell Differentiation/physiology , Cell Proliferation/drug effects , Durapatite/chemistry , Gelatin/chemistry , Gene Expression , Humans , Hydrogels , Nanostructures/chemistry , Osteocalcin/genetics , Osteocalcin/metabolism , Osteogenesis/physiology , Tissue Engineering
15.
J Biomater Appl ; 36(1): 179-190, 2021 07.
Article in English | MEDLINE | ID: mdl-33302758

ABSTRACT

Pectin has recently attracted increasing attention for biomedical and pharmaceutical applications. Due to the lack of adhesion molecules in polysaccharides, phenolic hydroxyl conjugated gelatin was added to enzymatically-gellable peroxidase-modified pectin derivative and compared with phenolic hydroxyl -pectin/collagen. Both pectin and gelatin were modified by tyramine hydrochloride in the presence of EDC/NHS. The phenolic hydroxyl -pectin/phenolic hydroxyl -gelatin, phenolic hydroxyl-pectin/collagen, and phenolic hydroxyl -pectin hydrogels were prepared using horseradish peroxidase and hydrogen peroxide. The hydrogels were characterized by gelation time analysis. Morphology, enzymatic biodegradation, mechanical and swelling properties as well as water vapor transmission rate were also evaluated. Fibroblasts were cultured for 7 days, and the survival rate was evaluated using conventional MTT assay. Hydrogels composed of Ph-pectin/Ph-gelatin showed decreased biodegradation rate, and WVTR and further improved mechanical performance in comparison with other groups. Both phenolic hydroxyl -pectin/collagen and phenolic hydroxyl -pectin/phenolic hydroxyl -gelatin hydrogels exhibited porous structures. The hydrogels composed of collagen promoted cell survival rate 1.4 and 3.5 times compared to phenolic hydroxyl -gelatin and phenolic hydroxyl -pectin based hydrogels at the end of 7 days, respectively (p < 0.001). The study demonstrated the potential of enzymatically-gellable pectin-based hydrogels as cost-effective frameworks for use in tissue engineering applications.


Subject(s)
Collagen/chemistry , Fibroblasts , Gelatin/chemistry , Hydrogels/chemistry , Pectins/chemistry , Peroxidase/chemistry , Cell Survival , Horseradish Peroxidase , Peroxidase/metabolism , Peroxidases , Succinimides , Tissue Engineering
16.
Microvasc Res ; 133: 104073, 2021 01.
Article in English | MEDLINE | ID: mdl-32949575

ABSTRACT

In this study, the angiogenic capacity of human endothelial cells was studied after being plated on the surface of polyurethane-poly caprolactone (PU/PCL) scaffolds for 72 h. In this study, cells were designated into five different groups, including PU, PU/PCL (2:1), PU/PCL (1:1); PU/PCL (1:2); and PCL. Data revealed that the PU/PCL (2:1) composition had a higher modulus and breakpoint in comparison with the other groups (p < 0.05). Compared to the other groups, the PU/PCL scaffold with a molar ratio of 2:1 had lower the contact angle θ and higher tensile stress (p < 0.05). The mean size of the PU nanofibers was reduced after the addition of PCL (p < 0.05). Based on our data, the culture of endothelial cells on the surface of PU/PCL (2:1) did not cause nitrosative stress and cytotoxic effects under static conditions compared to cells plated on a conventional plastic surface (p > 0.05). Based on data from the static condition, we fabricated a tubular PU/PCL (2:1) construct for six-day dynamic cell culture inside loop air-lift bioreactors. Scanning electron microscopy showed the attachment of endothelial cells to the luminal surface of the PU/PCL scaffold. Cells were flattened and aligned under the culture medium flow. Immunofluorescence imaging showed the attachment of cells to the luminal surface indicated by blue nuclei on the luminal surface. These data demonstrated that the application of PU/PCL substrate could stimulate endothelial cells activity under static and dynamic conditions.


Subject(s)
Human Umbilical Vein Endothelial Cells/physiology , Nanofibers , Polyesters/chemistry , Polyurethanes/chemistry , Tissue Scaffolds , Bioreactors , Cell Adhesion , Cell Culture Techniques , Cell Proliferation , Cell Survival , Cells, Cultured , Elastic Modulus , Human Umbilical Vein Endothelial Cells/ultrastructure , Humans , Tensile Strength , Time Factors
17.
J Biomed Mater Res B Appl Biomater ; 108(7): 2950-2960, 2020 10.
Article in English | MEDLINE | ID: mdl-32351038

ABSTRACT

To develop an efficient injectable alginate-based hydrogel for soft tissue engineering applications, phenol moiety (Ph) was introduced into alginate (Alg-Ph), and the influence of gelatin as cell adhesive molecule was evaluated on the peroxidase-mediated alginate hydrogel properties and cultured chondrocytic cell behavior. Addition of gelatin (1.5% w/v) to Alg-Ph (1.5% w/v) hydrogels (Alg-Ph/gelatin) regulated characteristics of the enzymatically gellable alginate hydrogel with increasing gelation time to 5.1 min (76%). Swelling ratio and degradation rates of the Alg-Ph/gelatin hydrogel also increased 60 and 100%, respectively, while the mechanical strength value was 35% less than the Alg-Ph hydrogel. Scanning electron microscopy images showed that the addition of gelatin could also increase uniformity of pore sizes inside the Alg-Ph/gelatin hydrogels. The chondrocyte cells maintained their original phenotype and revealed statistically more metabolic activities in the Alg-Ph/gelatin hydrogel. Hydrogels subscutaneously implanted in rats could also be identified readily without complete absorption and signs of toxicity or any untoward reactions after 1 month. Viable chondrocyte cells inside globular aggregates were seen as red colored areas in the cell-laden hydrogels. The study demonstrates that enzymatically gellable alginate/gelatin hydrogel has fair potential as a natural-based injectable hydrogel for soft tissue engineering applications.


Subject(s)
Alginates/chemistry , Chondrocytes/metabolism , Gelatin/chemistry , Hydrogels/chemistry , Peroxidase/chemistry , Tissue Scaffolds/chemistry , Animals , Cell Adhesion , Cell Survival , Male , Rats , Rats, Wistar , Tissue Engineering
18.
Int J Biol Macromol ; 156: 1270-1278, 2020 Aug 01.
Article in English | MEDLINE | ID: mdl-31760032

ABSTRACT

To provide comparable hepatic tissue microenvironment and induce functional behavior for hepatocytes, galctosylated-chitosan (GC) as well as collagen (Col) was added to alginate microcapsule coated with extra layer of chitosan. Four different hydrogel groups of alginate/chitosan (AC); alginate-galactosylated chitosan/chitosan (AGC/C); alginate-collagen/chitosan (ACol/C); and alginate-galactosylated chitosan-collagen/chitosan (AGCCol/C) were prepared and characterized for physical properties such as porosity, swelling, degradation rate, and stiffness. Introduction of GC as well as Col to alginate regulated significantly the physical properties of the resultant hydrogels. GC addition decreased dramatically swelling, degradation, pore size and mechanical properties of the resultant hydrogel. However, the influence of GC on the physical properties in the presence of Col (AGCCol/A) was in a reverse manner, as compared to the AGC/C hydrogel. The AGCCol/C microenvironment also promoted proliferation of microencapsulated HepG2 cells, as a model of hepatocyte, compared to the control-matched groups. Biochemical analysis after 10 days revealed a superior effect of AGCCol/C on the secretion of albumin and urea compared to other groups (P < 0.05). These features were coincided with the mRNA up-regulation of P450 and albumin in the AGCCol/C groups compared to the AGC/C and ACol/C groups (P < 0.05). The study demonstrated that enrichment of alginate-based hydrogels with Col and GC could be touted as an appropriate 3D platform for modular hepatic tissue engineering.


Subject(s)
Alginates/chemistry , Chitosan/chemistry , Chitosan/pharmacology , Collagen/chemistry , Galactose/chemistry , Hepatocytes/drug effects , Hydrogels/chemistry , Capsules , Cell Survival/drug effects , Hep G2 Cells , Hepatocytes/cytology , Humans , Mechanical Phenomena
19.
Biochem Biophys Res Commun ; 518(4): 625-631, 2019 10 22.
Article in English | MEDLINE | ID: mdl-31447120

ABSTRACT

The influence of collagen as an effective substitute for gelatin was investigated on properties of chitosan/gelatin hydrogels for fibroblasts growth and attachment for wound dressing applications. We synthesized hydrogels based on chitosan associated with collagen and gelatin biopolymers (in the ratio of 1:5 and 1:1, respectively). The hydrogels properties such as morphology, swelling ratio, mechanical characteristics, water vapor loss, water vapor transmission rate (WVTR), and biodegradation were analyzed. 1 × 105 human fibroblasts were seeded per ml of hydrogels and maintained for 7 days. Cell viability was assessed by using MTT. The presence of collagen caused reduced swelling ratio, and biodegradation rate compared to chitosan/gelatin hydrogels (p < 0.05). The introduction of collagen into chitosan hydrogels improved the mechanical strength compared to gelatin. Hydrogels with collagen possessed an optimum WVTR compared to the chitosan group and hydrogels with gelatin (p < 0.05). Analyzing the morphology of hydrogels revealed that the addition of collagen leads to a homogenous and interconnected structure. Collagen impregnation promoted cell survival and attachment compared with chitosan hydrogels after 7 days (p < 0.05). Collectively, these results demonstrated the potential of the chitosan/collagen hydrogels for wound dressing applications.


Subject(s)
Chitosan/chemistry , Collagen/chemistry , Fibroblasts/cytology , Gelatin/chemistry , Hydrogels/chemistry , Animals , Bandages , Biocompatible Materials/chemistry , Biocompatible Materials/pharmacology , Cattle , Cell Line , Cell Proliferation/drug effects , Cell Survival/drug effects , Chitosan/pharmacology , Collagen/pharmacology , Fibroblasts/drug effects , Gelatin/pharmacology , Humans , Hydrogels/pharmacology , Steam/analysis , Wound Healing/drug effects
20.
J Cell Biochem ; 120(9): 15069-15082, 2019 09.
Article in English | MEDLINE | ID: mdl-31020682

ABSTRACT

Modular bone tissue engineering is touted as an alternative approach to replace the damaged bone tissue. Hydrogel microcapsules could promote therapeutic properties by providing 3D condition and an increased cell-to-cell interaction. We investigated the osteogenic properties of alginate-nano-silica hydrogels enriched with collagen and gelatin on human osteoblast-like MG-63 cells. For encapsulation, cells were divided into three groups; control (alginate+ nano-silica), collagen (alginate + collagen + nano-silica), and gelatin (alginate + gelatin + nano-silica) and expanded for 28 days. Cell survival was determined by trypan blue staining and 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl tetrazolium bromide (MTT) assay. To confirm the osteogenic potential, we measured the alkaline phosphatase activity. Alizarin red S staining was used to reveal the existence of hydroxyapatite and transcription BMP-2, osteocalcin and osteonectin evaluated by the real-time polymerase chain reaction. Collagen substrate caused a reduced swelling ratio compared with the control and gelatin groups (P < 0.05). Compared with other groups, collagen had potential to improve mechanical strength and generate porous membrane structure. The addition of collagen (4-fold) and gelatin (1.5-fold) increased cell proliferation rate compared with the control (P < 0.05). Biochemical analysis and Alizarin red S staining showed that collagen-induced osteogenesis by induction of alkaline phosphatase and matrix mineralization. The expression of osteocalcin and BMP-2 was increased in cells from the collagen group. As a result, the combination of natural polymers collagen and gelatin with alginate + nano-silica can increase the osteogenic potential of human osteoblasts.


Subject(s)
Alginates/pharmacology , Collagen/pharmacology , Microspheres , Osteoblasts/metabolism , Osteogenesis/drug effects , Silicon Dioxide/pharmacology , Alkaline Phosphatase/metabolism , Animals , Biocompatible Materials/pharmacology , Bone Morphogenetic Protein 2/genetics , Bone Morphogenetic Protein 2/metabolism , Calcium/metabolism , Cattle , Cell Line , Cell Survival/drug effects , Gelatin/pharmacology , Humans , Hydrogels/chemistry , Mechanical Phenomena , Osteoblasts/drug effects , Osteoblasts/enzymology , Osteocalcin/genetics , Osteocalcin/metabolism , Osteogenesis/genetics , Osteonectin/genetics , Osteonectin/metabolism , Tissue Scaffolds/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL
...