Displaying publications 41 - 60 of 77 in total

Abstract:
Sort:
  1. Dhand C, Balakrishnan Y, Ong ST, Dwivedi N, Venugopal JR, Harini S, et al.
    Int J Nanomedicine, 2018;13:4473-4492.
    PMID: 30122921 DOI: 10.2147/IJN.S159770
    Introduction: In search for cross-linkers with multifunctional characteristics, the present work investigated the utility of quaternary ammonium organosilane (QOS) as a potential cross-linker for electrospun collagen nanofibers. We hypothesized that the quaternary ammonium ions improve the electrospinnability by reducing the surface tension and confer antimicrobial properties, while the formation of siloxane after alkaline hydrolysis could cross-link collagen and stimulate cell proliferation.

    Materials and methods: QOS collagen nanofibers were electrospun by incorporating various concentrations of QOS (0.1%-10% w/w) and were cross-linked in situ after exposure to ammonium carbonate. The QOS cross-linked scaffolds were characterized and their biological properties were evaluated in terms of their biocompatibility, cellular adhesion and metabolic activity for primary human dermal fibroblasts and human fetal osteoblasts.

    Results and discussion: The study revealed that 1) QOS cross-linking increased the flexibility of otherwise rigid collagen nanofibers and improved the thermal stability; 2) QOS cross-linked mats displayed potent antibacterial activity and 3) the biocompatibility of the composite mats depended on the amount of QOS present in dope solution - at low QOS concentrations (0.1% w/w), the mats promoted mammalian cell proliferation and growth, whereas at higher QOS concentrations, cytotoxic effect was observed.

    Conclusion: This study demonstrates that QOS cross-linked mats possess anti-infective properties and confer niches for cellular growth and proliferation, thus offering a useful approach, which is important for hard and soft tissue engineering and regenerative medicine.

    Matched MeSH terms: Tissue Scaffolds/chemistry*
  2. Ezhilarasu H, Sadiq A, Ratheesh G, Sridhar S, Ramakrishna S, Ab Rahim MH, et al.
    Nanomedicine (Lond), 2019 01;14(2):201-214.
    PMID: 30526272 DOI: 10.2217/nnm-2018-0271
    AIM: Atherosclerosis is a common cardiovascular disease causing medical problems globally leading to coronary artery bypass surgery. The present study is to fabricate core/shell nanofibers to encapsulate VEGF for the differentiation of mesenchymal stem cells (MSCs) into smooth muscle cells to develop vascular grafts.

    MATERIALS & METHODS: The fabricated core/shell nanofibers contained polycaprolactone/gelatin as the shell, and silk fibroin/VEGF as the core materials.

    RESULTS: The results observed that the core/shell nanofibers interact to differentiate MSCs into smooth muscle cells by the expression of vascular smooth muscle cell (VSMC) contractile proteins α-actinin, myosin and F-actin.

    CONCLUSION: The functionalized polycaprolactone/gelatin/silk fibroin/VEGF (250 ng) core/shell nanofibers were fabricated for the controlled release of VEGF in a persistent manner for the differentiation of MSCs into smooth muscle cells for vascular tissue engineering.

    Matched MeSH terms: Tissue Scaffolds/chemistry
  3. Latfi ASA, Pramanik S, Poon CT, Gumel AM, Lai KW, Annuar MSM, et al.
    J Biomater Appl, 2019 01;33(6):854-865.
    PMID: 30458659 DOI: 10.1177/0885328218812490
    Natural biopolymers have many attractive medical applications; however, complications due to fibrosis caused a reduction in diffusion and dispersal of nutrients and waste products. Consequently, severe immunocompatibility problems and poor mechanical and degradation properties in synthetic polymers ensue. Hence, the present study investigates a novel hydrogel material synthesized from caprolactone, ethylene glycol, ethylenediamine, polyethylene glycol, ammonium persulfate, and tetramethylethylenediamine via chemo-enzymatic route. Spectroscopic analyses indicated the formation of polyurea and polyhydroxyurethane as the primary building block of the hydrogel starting material. Biocompatibility studies showed positive observation in biosafety test using direct contact cytotoxicity assay in addition to active cellular growth on the hydrogel scaffold based on fluorescence observation. The synthesized hydrogel also exhibited (self)fluorescence properties under specific wavelength excitation. Hence, synthesized hydrogel could be a potential candidate for medical imaging as well as tissue engineering applications as a tissue expander, coating material, biosensor, and drug delivery system.
    Matched MeSH terms: Tissue Scaffolds/chemistry
  4. Zulkifli FH, Hussain FSJ, Harun WSW, Yusoff MM
    Int J Biol Macromol, 2019 Feb 01;122:562-571.
    PMID: 30365990 DOI: 10.1016/j.ijbiomac.2018.10.156
    This study is focusing to develop a porous biocompatible scaffold using hydroxyethyl cellulose (HEC) and poly (vinyl alcohol) (PVA) with improved cellular adhesion profiles and stability. The combination of HEC and PVA were synthesized using freeze-drying technique and characterized using SEM, ATR-FTIR, TGA, DSC, and UTM. Pore size of HEC/PVA (2-40 μm) scaffolds showed diameter in a range of both pure HEC (2-20 μm) and PVA (14-70 μm). All scaffolds revealed high porosity above 85%. The water uptake of HEC was controlled by PVA cooperation in the polymer matrix. After 7 days, all blended scaffolds showed low degradation rate with the increased of PVA composition. The FTIR and TGA results explicit possible chemical interactions and mass loss of blended scaffolds, respectively. The Tg values of DSC curved in range of HEC and PVA represented the miscibility of HEC/PVA blend polymers. Higher Young's modulus was obtained with the increasing of HEC value. Cell-scaffolds interaction demonstrated that human fibroblast (hFB) cells adhered to polymer matrices with better cell proliferation observed after 7 days of cultivation. These results suggested that biocompatible of HEC/PVA scaffolds fabricated by freeze-drying method might be suitable for skin tissue engineering applications.
    Matched MeSH terms: Tissue Scaffolds/chemistry*
  5. Gnaneshwar PV, Sudakaran SV, Abisegapriyan S, Sherine J, Ramakrishna S, Rahim MHA, et al.
    Mater Sci Eng C Mater Biol Appl, 2019 Mar;96:337-346.
    PMID: 30606541 DOI: 10.1016/j.msec.2018.11.033
    Far-flung evolution in tissue engineering enabled the development of bioactive and biodegradable materials to generate biocomposite nanofibrous scaffolds for bone repair and replacement therapies. Polymeric bioactive nanofibers are to biomimic the native extracellular matrix (ECM), delivering tremendous regenerative potentials for drug delivery and tissue engineering applications. It's been known from few decades that Zinc oxide (ZnO) nanoparticles are enhancing bone growth and providing proliferation of osteoblasts when incorporated with hydroxyapatite (HAp). We attempted to investigate the interaction between the human foetal osteoblasts (hFOB) with ZnO doped HAp incorporated biocomposite poly(L-lactic acid)-co-poly(ε-caprolactone) and silk fibroin (PLACL/SF) nanofibrous scaffolds for osteoblasts mineralization in bone tissue regeneration. The present study, we doped ZnO with HAp (ZnO(HAp) using the sol-gel ethanol condensation technique. The properties of PLACL/SF/ZnO(HAp) biocomposite nanofibrous scaffolds enhanced with doped and blended ZnO/HAp were characterized using Scanning electron microscopy (SEM), Fourier transform infrared spectroscopy (FTIR), Contact angle and Tensile studies to determine the morphology, functionality, wettability and stability. The in vitro study results showed that the addition of ZnO and HAp enhances the secretion of bone mineral matrix (98%) with smaller fiber diameter (139.4 ± 27 nm) due to the presence of silk fibroin showing potential tensile properties (322.4%), and increased the proliferation of osteoblasts for bone tissue regeneration.
    Matched MeSH terms: Tissue Scaffolds/chemistry*
  6. Anita Lett J, Sundareswari M, Ravichandran K, Latha B, Sagadevan S
    Mater Sci Eng C Mater Biol Appl, 2019 Mar;96:487-495.
    PMID: 30606558 DOI: 10.1016/j.msec.2018.11.082
    The practice of bone implants is the standard procedure for the treatment of skeletal fissures, or to substitute and re-establish lost bone. A perfect scaffold ought to be made of biomaterials that duplicate the structure and properties of natural bone. However, the production of living tissue constructs that are architecturally, functionally and mechanically comparable to natural bone is the major challenge in the treatment and regeneration of bone tissue in orthopaedics and in dentistry. In this work, we have employed a polymeric replication method to fabricate hydroxyapatite (HAP) scaffolds using gum tragacanth (GT) as a natural binder. GT is a natural gum collected from the dried sap of several species of Middle Eastern legumes of the genus Astragalus, possessing antibacterial and wound healing properties. The synthesized porous HAP scaffolds were analyzed structurally and characterized for their phase purity and mechanical properties. The biocompatibility of the porous HAP scaffold was confirmed by seeding the scaffold with Vero cells, and its bioactivity assessed by immersing the scaffold in simulated body fluid (SBF). Our characterization data showed that the biocompatible porous HAP scaffolds were composed of highly interconnecting pores with compressive strength ranging from 0.036 MPa to 2.954 MPa, comparable to that of spongy bone. These can be prepared in a controlled manner by using an appropriate binder concentration and sintering temperature. These HAP scaffolds have properties consistent with normal bone and should be further developed for potential application in bone implants.
    Matched MeSH terms: Tissue Scaffolds/chemistry*
  7. Pourshahrestani S, Zeimaran E, Kadri NA, Gargiulo N, Jindal HM, Hasikin K, et al.
    Mater Sci Eng C Mater Biol Appl, 2019 May;98:1022-1033.
    PMID: 30812986 DOI: 10.1016/j.msec.2019.01.022
    A novel series of silver-doped mesoporous bioactive glass/poly(1,8-octanediol citrate) (AgMBG/POC) elastomeric biocomposite scaffolds were successfully constructed by a salt-leaching technique for the first time and the effect of inclusion of different AgMBG contents (5, 10, and 20 wt%) on physicochemical and biological properties of pure POC elastomer was evaluated. Results indicated that AgMBG particles were uniformly dispersed in the POC matrix and increasing the AgMBG concentration into POC matrix up to 20 wt% enhanced thermal behaviour, mechanical properties and water uptake ability of the composite scaffolds compared to those from POC. The 20%AgMBG/POC additionally showed higher degradation rate in Tris(hydroxymethyl)-aminomethane-HCl (Tris-HCl) compared with pure POC and lost about 26% of its initial weight after soaking for 28 days. The AgMBG phase incorporation also significantly endowed the resulting composite scaffolds with efficient antibacterial properties against Escherichia coli and Staphylococcus aureus bacteria while preserving their favorable biocompatibility with soft tissue cells (i.e., human dermal fibroblast cells). Taken together, our results suggest that the synergistic effect of both AgMBG and POC make these newly designed AgMBG/POC composite scaffold an attractive candidate for soft tissue engineering applications.
    Matched MeSH terms: Tissue Scaffolds/chemistry
  8. Mh Busra F, Rajab NF, Tabata Y, Saim AB, B H Idrus R, Chowdhury SR
    J Tissue Eng Regen Med, 2019 05;13(5):874-891.
    PMID: 30811090 DOI: 10.1002/term.2842
    The full-thickness skin wound is a common skin complication affecting millions of people worldwide. Delayed treatment of this condition causes the loss of skin function and integrity that could lead to the development of chronic wounds or even death. This study was aimed to develop a rapid wound treatment modality using ovine tendon collagen type I (OTC-I) bio-scaffold with or without noncultured skin cells. Genipin (GNP) and carbodiimide (EDC) were used to cross-link OTC-I scaffold to improve the mechanical strength of the bio-scaffold. The physicochemical, biomechanical, biodegradation, biocompatibility, and immunogenicity properties of OTC-I scaffolds were investigated. The efficacy of this treatment approach was evaluated in an in vivo skin wound model. The results demonstrated that GNP cross-linked OTC-I scaffold (OTC-I_GNP) had better physicochemical and mechanical properties compared with EDC cross-linked OTC-I scaffold (OTC-I_EDC) and noncross-link OTC-I scaffold (OTC-I_NC). OTC-I_GNP and OTC-I_NC demonstrated no toxic effect on cells as it promoted higher cell attachment and proliferation of both primary human epidermal keratinocytes and human dermal fibroblasts compared with OTC-I_EDC. Both OTC-I_GNP and OTC-I_NC exhibited spontaneous formation of bilayer structure in vitro. Immunogenic evaluation of OTC-I scaffolds, in vitro and in vivo, revealed no sign of immune response. Finally, implantation of OTC-I_NC and OTC-I_GNP scaffolds with noncultured skin cells demonstrated enhanced healing with superior skin maturity and microstructure features, resembling native skin in contrast to other treatment (without noncultured skin cells) and control group. The findings of this study, therefore, suggested that both OTC-I scaffolds with noncultured skin cells could be promising for the rapid treatment of full-thickness skin wound.
    Matched MeSH terms: Tissue Scaffolds/chemistry*
  9. Md Saad AP, Prakoso AT, Sulong MA, Basri H, Wahjuningrum DA, Syahrom A
    Biomech Model Mechanobiol, 2019 Jun;18(3):797-811.
    PMID: 30607641 DOI: 10.1007/s10237-018-01115-z
    This study employs a computational approach to analyse the impact of morphological changes on the structural properties of biodegradable porous Mg subjected to a dynamic immersion test for its application as a bone scaffold. Porous Mg was immersed in a dynamic immersion test for 24, 48, and 72 h. Twelve specimens were prepared and scanned using micro-CT and then reconstructed into a 3D model for finite element analysis. The structural properties from the numerical simulation were then compared to the experimental values. Correlations between morphological parameters, structural properties, and fracture type were then made. The relative losses were observed to be in agreement with relative mass loss done experimentally. The degradation rates determined using exact (degraded) surface area at particular immersion times were on average 20% higher than the degradation rates obtained using original surface area. The dynamic degradation has significantly impacted the morphological changes of porous Mg in volume fraction, surface area, and trabecular separation, which in turn affects its structural properties and increases the immersion time.
    Matched MeSH terms: Tissue Scaffolds/chemistry*
  10. Kouhi M, Jayarama Reddy V, Ramakrishna S
    Appl Biochem Biotechnol, 2019 Jun;188(2):357-368.
    PMID: 30456599 DOI: 10.1007/s12010-018-2922-0
    Bioceramic nanoparticles with high specific surface area often tend to agglomerate in the polymer matrix, which results in undesirable mechanical properties of the composites and poor cell spreading and attachment. In the present work, bredigite (BR) nanoparticles were modified with an organosilane coupling agent, 3-glycidoxypropyltrimethoxysilane (GPTMS), to enhance its dispersibility in the polymer matrix. The polyhydroxybutyrate-co-hydroxyvaletare (PHBV) nanofibrous scaffolds containing either bredigite or GPTMS-modified bredigite (G-BR) nanoparticles were fabricated using electrospinning technique and characterized using scanning electron microscopy, transmission electron microscopy, and tensile strength. Results demonstrated that modification of bredigite was effective in enhancing nanoparticle dispersion in the PHBV matrix. PHBV/G-BR scaffold showed improved mechanical properties compared to PHBV and PHBV/BR, especially at the higher concentration of nanoparticles. In vitro bioactivity assay performed in the simulated body fluid (SBF) indicated that composite PHBV scaffolds were able to induce the formation of apatite deposits after incubation in SBF. From the results of in vitro biological assay, it is concluded that the synergetic effect of BR and GPTMS provided an enhanced hFob cells attachment and proliferation. The developed PHBV/G-BR nanofibrous scaffolds may be considered for application in bone tissue engineering.
    Matched MeSH terms: Tissue Scaffolds/chemistry*
  11. Rizwan M, Yahya R, Hassan A, Yar M, Abd Halim AA, Rageh Al-Maleki A, et al.
    J Mater Sci Mater Med, 2019 Jun 11;30(6):72.
    PMID: 31187295 DOI: 10.1007/s10856-019-6273-3
    The success of wound healing depends upon the proper growth of vascular system in time in the damaged tissues. Poor blood supply to wounded tissues or tissue engineered grafts leads to the failure of wound healing or rejection of grafts. In present paper, we report the synthesis of novel organosoluble and pro-angiogenic chitosan derivative (CSD) by the reaction of chitosan with 1,3-dimethylbarbituric acid and triethylorthoformate (TEOF). The synthesized material was characterized by FTIR and 13C-NMR to confirm the incorporated functional groups and new covalent connectivities. Biodegradability of the synthesized chitosan derivative was tested in the presence of lysozyme and was found to be comparable with CS. The cytotoxicity and apoptosis effect of new derivative was determined against gastric adenocarcinoma (AGS) cells and was found to be non-toxic. The CSD was found to be soluble in majority of organic solvents. It was blended with polycaprolactone (PCL) to form composite scaffolds. From an ex ovo CAM assay, it was noted that CSD stimulated the angiogenesis.
    Matched MeSH terms: Tissue Scaffolds/chemistry*
  12. Dayaghi E, Bakhsheshi-Rad HR, Hamzah E, Akhavan-Farid A, Ismail AF, Aziz M, et al.
    Mater Sci Eng C Mater Biol Appl, 2019 Sep;102:53-65.
    PMID: 31147024 DOI: 10.1016/j.msec.2019.04.010
    Recently, porous magnesium and its alloys are receiving great consideration as biocompatible and biodegradable scaffolds for bone tissue engineering application. However, they presented poor antibacterial performance and corrosion resistance which limited their clinical applications. In this study, Mg-Zn (MZ) scaffold containing different concentrations of tetracycline (MZ-xTC, x = 1, 5 and 10%) were fabricated by space holder technique to meet the desirable antibacterial activity and corrosion resistance properties. The MZ-TC contains total porosity of 63-65% with pore sizes in the range of 600-800 μm in order to accommodate bone cells. The MZ scaffold presented higher compressive strength and corrosion resistance compared to pure Mg scaffold. However, tetracycline incorporation has less significant effect on the mechanical and corrosion properties of the scaffolds. Moreover, MZ-xTC scaffolds drug release profiles show an initial immediate release which is followed by more stable release patterns. The bioactivity test reveals that the MZ-xTC scaffolds are capable of developing the formation of HA layers in simulated body fluid (SBF). Next, Staphylococcus aureus and Escherichia coli bacteria were utilized to assess the antimicrobial activity of the MZ-xTC scaffolds. The findings indicate that those scaffolds that incorporate a high level concentration of tetracycline are tougher against bacterial organization than MZ scaffolds. However, the MTT assay demonstrates that the MZ scaffolds containing 1 to 5% tetracycline are more effective to sustain cell viability, whereas MZ-10TC shows some toxicity. The alkaline phosphatase (ALP) activity of the MZ-(1-5)TC was considerably higher than that of MZ-10TC on the 3 and 7 days, implying higher osteoblastic differentiation. All the findings suggest that the MZ-xTC scaffolds containing 1 to 5% tetracycline is a promising candidate for bone tissue healing due to excellent antibacterial activity and biocompatibility.
    Matched MeSH terms: Tissue Scaffolds/chemistry*
  13. Ramli K, Gasim AI, Ahmad AA, Htwe O, Mohamed Haflah NH, Law ZK, et al.
    Tissue Eng Part A, 2019 10;25(19-20):1438-1455.
    PMID: 30848172 DOI: 10.1089/ten.TEA.2018.0279
    We investigated the efficacy of a muscle-stuffed vein (MSV) seeded with neural-transdifferentiated human mesenchymal stem cells as an alternative nerve conduit to repair a 15-mm sciatic nerve defect in athymic rats. Other rats received MSV conduit alone, commercial polyglycolic acid conduit (Neurotube®), reverse autograft, or were left untreated. Motor and sensory functions as well as nerve conductivity were evaluated for 12 weeks, after which the grafts were harvested for histological analyses. All rats in the treatment groups demonstrated a progressive increase in the mean Sciatic Functional Index (motor function) and nerve conduction amplitude (electrophysiological function) and showed positive withdrawal reflex (sensory function) by the 10th week of postimplantation. Autotomy, which is associated with neuropathic pain, was severe in rats treated with conduit without cells; there was mild or no autotomy in the rats of other groups. Histologically, harvested grafts from all except the untreated groups exhibited axonal regeneration with the presence of mature myelinated axons. In conclusion, treatment with MSV conduit is comparable to that of other treatment groups in supporting functional recovery following sciatic nerve injury; and the addition of cells in the conduit alleviates neuropathic pain. Impact Statement It is shown that pretreated muscle-stuffed vein conduit is comparable to that of commercial nerve conduit and autograft in supporting functional recovery following peripheral nerve injury. The addition of neural-differentiated mesenchymal stem cells in the conduit is shown to alleviate neuropathic pain.
    Matched MeSH terms: Tissue Scaffolds/chemistry*
  14. Lim WL, Liau LL, Ng MH, Chowdhury SR, Law JX
    Tissue Eng Regen Med, 2019 Dec;16(6):549-571.
    PMID: 31824819 DOI: 10.1007/s13770-019-00196-w
    BACKGROUND: Tendon and ligament injuries accounted for 30% of all musculoskeletal consultations with 4 million new incidences worldwide each year and thus imposed a significant burden to the society and the economy. Damaged tendon and ligament can severely affect the normal body movement and might lead to many complications if not treated promptly and adequately. Current conventional treatment through surgical repair and tissue graft are ineffective with a high rate of recurrence.

    METHODS: In this review, we first discussed the anatomy, physiology and pathophysiology of tendon and ligament injuries and its current treatment. Secondly, we explored the current role of tendon and ligament tissue engineering, describing its recent advances. After that, we also described stem cell and cell secreted product approaches in tendon and ligament injuries. Lastly, we examined the role of the bioreactor and mechanical loading in in vitro maturation of engineered tendon and ligament.

    RESULTS: Tissue engineering offers various alternative ways of treatment from biological tissue constructs to stem cell therapy and cell secreted products. Bioreactor with mechanical stimulation is instrumental in preparing mature engineered tendon and ligament substitutes in vitro.

    CONCLUSIONS: Tissue engineering showed great promise in replacing the damaged tendon and ligament. However, more study is needed to develop ideal engineered tendon and ligament.

    Matched MeSH terms: Tissue Scaffolds/chemistry
  15. Hazwani A, Sha'Ban M, Azhim A
    Organogenesis, 2019;15(4):120-136.
    PMID: 31495272 DOI: 10.1080/15476278.2019.1656997
    Extracellular matrix (ECM) based bioscaffolds prepared by decellularization has increasingly emerged in tissue engineering application because it has structural, biochemical, and biomechanical cues that have dramatic effects upon cell behaviors. Therefore, we developed a closed sonication decellularization system to prepare ideal bioscaffolds with minimal adverse effects on the ECM. The decellularization was achieved at 170 kHz of ultrasound frequency in 0.1% and 2% Sodium Dodecyl Sulphate (SDS) solution for 10 hours. The immersion treatment as control was performed to compare the decellularization efficiency with our system. Cell removal and ECM structure were determined by histological staining and biochemical assay. Biomechanical properties were investigated by the indentation testing to test the stiffness, a residual force and compression of bioscaffolds. Additionally, in vivo implantation was performed in rat to investigate host tissue response. Compared to native tissues, histological staining and biochemical assay confirm the absence of cellularity with preservation of ECM structure. Moreover, sonication treatment has not affected the stiffness [N/mm] and a residual force [N] of the aortic scaffolds except for compression [%] which 2% SDS significantly decreased compared to native tissues showing higher SDS has a detrimental effect on ECM structure. Finally, minimal inflammatory response was observed after 1 and 5 weeks of implantation. This study reported that the novelty of our developed closed sonication system to prepare ideal bioscaffolds for tissue engineering applications.
    Matched MeSH terms: Tissue Scaffolds/chemistry*
  16. Busra MFM, Lokanathan Y
    Curr Pharm Biotechnol, 2019;20(12):992-1003.
    PMID: 31364511 DOI: 10.2174/1389201020666190731121016
    Tissue engineering focuses on developing biological substitutes to restore, maintain or improve tissue functions. The three main components of its application are scaffold, cell and growthstimulating signals. Scaffolds composed of biomaterials mainly function as the structural support for ex vivo cells to attach and proliferate. They also provide physical, mechanical and biochemical cues for the differentiation of cells before transferring to the in vivo site. Collagen has been long used in various clinical applications, including drug delivery. The wide usage of collagen in the clinical field can be attributed to its abundance in nature, biocompatibility, low antigenicity and biodegradability. In addition, the high tensile strength and fibril-forming ability of collagen enable its fabrication into various forms, such as sheet/membrane, sponge, hydrogel, beads, nanofibre and nanoparticle, and as a coating material. The wide option of fabrication technology together with the excellent biological and physicochemical characteristics of collagen has stimulated the use of collagen scaffolds in various tissue engineering applications. This review describes the fabrication methods used to produce various forms of scaffolds used in tissue engineering applications.
    Matched MeSH terms: Tissue Scaffolds/chemistry*
  17. Jiang H, Mani MP, Jaganathan SK
    Int J Nanomedicine, 2019;14:8149-8159.
    PMID: 31632024 DOI: 10.2147/IJN.S214646
    Introduction: Recently several new approaches were emerging in bone tissue engineering to develop a substitute for remodelling the damaged tissue. In order to resemble the native extracellular matrix (ECM) of the human tissue, the bone scaffolds must possess necessary requirements like large surface area, interconnected pores and sufficient mechanical strength.

    Materials and methods: A novel bone scaffold has been developed using polyurethane (PE) added with wintergreen (WG) and titanium dioxide (TiO2). The developed nanocomposites were characterized through field emission scanning electron microscopy (FESEM), Fourier transform and infrared spectroscopy (FTIR), X-ray diffraction (XRD), contact angle measurement, thermogravimetric analysis (TGA), atomic force microscopy (AFM) and tensile testing. Furthermore, anticoagulant assays, cell viability analysis and calcium deposition were used to investigate the biological properties of the prepared hybrid nanocomposites.

    Results: FESEM depicted the reduced fibre diameter for the electrospun PE/WG and PE/WG/TiO2 than the pristine PE. The addition of WG and TiO2 resulted in the alteration in peak intensity of PE as revealed in the FTIR. Wettability measurements showed the PE/WG showed decreased wettability and the PE/WG/TiO2 exhibited improved wettability than the pristine PE. TGA measurements showed the improved thermal behaviour for the PE with the addition of WG and TiO2. Surface analysis indicated that the composite has a smoother surface rather than the pristine PE. Further, the incorporation of WG and TiO2 improved the anticoagulant nature of the pristine PE. In vitro cytotoxicity assay has been performed using fibroblast cells which revealed that the electrospun composites showed good cell attachment and proliferation after 5 days. Moreover, the bone apatite formation study revealed the enhanced deposition of calcium content in the fabricated composites than the pristine PE.

    Conclusion: Fabricated nanocomposites rendered improved physico-chemical properties, biocompatibility and calcium deposition which are conducive for bone tissue engineering.

    Matched MeSH terms: Tissue Scaffolds/chemistry*
  18. Yusof F, Sha'ban M, Azhim A
    Int J Nanomedicine, 2019;14:5491-5502.
    PMID: 31410000 DOI: 10.2147/IJN.S207270
    PURPOSE: Meniscus is a fibrocartilagenous tissue that cannot effectively heal due to its complex structure and presence of avascular zone. Thus, tissue engineering and regenerative medicine offer an alternative for the regeneration of meniscus tissues using bioscaffolds as a replacement for the damaged one. The aim of this study was to prepare an ideal meniscus bioscaffold with minimal adverse effect on extracellular matrix components (ECMs) using a sonication treatment system.

    METHODS: The decellularization was achieved using a developed closed sonication treatment system for 10 hrs, and continued with a washing process for 5 days. For the control, a simple immersion treatment was set as a benchmark to compare the decellularization efficiency. Histological and biochemical assays were conducted to investigate the cell removal and retention of the vital extracellular matrix. Surface ultrastructure of the prepared scaffolds was evaluated using scanning electron microscope at 5,000× magnification viewed from cross and longitudinal sections. In addition, the biomechanical properties were investigated through ball indentation testing to study the stiffness, residual forces and compression characteristics. Statistical significance between the samples was determined with p-value =0.05.

    RESULTS: Histological and biochemical assays confirmed the elimination of antigenic cellular components with the retention of the vital extracellular matrix within the sonicated scaffolds. However, there was a significant removal of sulfated glycosaminoglycans. The surface histoarchitecture portrayed the preserved collagen fibril orientation and arrangement. However, there were minor disruptions on the structure, with few empty micropores formed which represented cell lacunae. The biomechanical properties of bioscaffolds showed the retention of viscoelastic behavior of the scaffolds which mimic native tissues. After immersion treatment, those scaffolds had poor results compared to the sonicated scaffolds due to the inefficiency of the treatment.

    CONCLUSION: In conclusion, this study reported that the closed sonication treatment system had high capabilities to prepare ideal bioscaffolds with excellent removal of cellular components, and retained extracellular matrix and biomechanical properties.

    Matched MeSH terms: Tissue Scaffolds/chemistry*
  19. Geetha Bai R, Muthoosamy K, Manickam S, Hilal-Alnaqbi A
    Int J Nanomedicine, 2019;14:5753-5783.
    PMID: 31413573 DOI: 10.2147/IJN.S192779
    Tissue engineering embraces the potential of recreating and replacing defective body parts by advancements in the medical field. Being a biocompatible nanomaterial with outstanding physical, chemical, optical, and biological properties, graphene-based materials were successfully employed in creating the perfect scaffold for a range of organs, starting from the skin through to the brain. Investigations on 2D and 3D tissue culture scaffolds incorporated with graphene or its derivatives have revealed the capability of this carbon material in mimicking in vivo environment. The porous morphology, great surface area, selective permeability of gases, excellent mechanical strength, good thermal and electrical conductivity, good optical properties, and biodegradability enable graphene materials to be the best component for scaffold engineering. Along with the apt microenvironment, this material was found to be efficient in differentiating stem cells into specific cell types. Furthermore, the scope of graphene nanomaterials in liver tissue engineering as a promising biomaterial is also discussed. This review critically looks into the unlimited potential of graphene-based nanomaterials in future tissue engineering and regenerative therapy.
    Matched MeSH terms: Tissue Scaffolds/chemistry*
  20. Alias MA, Buenzli PR
    Int J Numer Method Biomed Eng, 2020 01;36(1):e3279.
    PMID: 31724309 DOI: 10.1002/cnm.3279
    Most biological tissues grow by the synthesis of new material close to the tissue's interface, where spatial interactions can exert strong geometric influences on the local rate of growth. These geometric influences may be mechanistic or cell behavioural in nature. The control of geometry on tissue growth has been evidenced in many in vivo and in vitro experiments, including bone remodelling, wound healing, and tissue engineering scaffolds. In this paper, we propose a generalisation of a mathematical model that captures the mechanistic influence of curvature on the joint evolution of cell density and tissue shape during tissue growth. This generalisation allows us to simulate abrupt topological changes such as tissue fragmentation and tissue fusion, as well as three dimensional cases, through a level-set-based method. The level-set method developed introduces another Eulerian field than the level-set function. This additional field represents the surface density of tissue-synthesising cells, anticipated at future locations of the interface. Numerical tests performed with this level-set-based method show that numerical conservation of cells is a good indicator of simulation accuracy, particularly when cusps develop in the tissue's interface. We apply this new model to several situations of curvature-controlled tissue evolutions that include fragmentation and fusion.
    Matched MeSH terms: Tissue Scaffolds/chemistry
Filters
Contact Us

Please provide feedback to Administrator (afdal@afpm.org.my)

External Links