Displaying publications 1 - 20 of 255 in total

Abstract:
Sort:
  1. Danagody B, Bose N, Rajappan K, Iqbal A, Ramanujam GM, Anilkumar AK
    ACS Biomater Sci Eng, 2024 Jan 08;10(1):468-481.
    PMID: 38078836 DOI: 10.1021/acsbiomaterials.3c00892
    Developing biomaterial scaffolds using tissue engineering with physical and chemical surface modification processes can improve the bioactivity and biocompatibility of the materials. The appropriate substrate and site for cell attachment are crucial in cell behavior and biological activities. Therefore, the study aims to develop a conventional electrospun nanofibrous biomaterial using reproducible surface topography, which offers beneficial effects on the cell activities of bone cells. The bioactive MgO/gC3N4 was incorporated on PAN/PEG and fabricated into a nanofibrous membrane using electrospinning. The nanocomposite uniformly distributed on the PAN/PEG nanofiber helps to increase the number of induced pores and reduce the hydrophobicity of PAN. The physiochemical characterization of prepared nanoparticles and nanofibers was carried out using FTIR, X-ray diffraction (XRD), thermogravimetry analysis (TGA), X-ray photoelectron spectroscopy (XPS), and water contact angle measurements. SEM and TEM analyses examined the nanofibrous morphology and the structure of MgO/gC3N4. In vitro studies such as on ALP activity demonstrated the membrane's ability to regenerate new bone and healing capacity. Furthermore, alizarin red staining showed the increasing ability of the cell-cell interaction and calcium content for tissue regeneration. The cytotoxicity of the prepared membrane was about 97.09% of live THP-1 cells on the surface of the MgO/gC3N4@PAN/PEG membrane evaluated using MTT dye staining. The soil burial degradation analysis exhibited that the maximum degradation occurs on the 45th day because of microbial activity. In vitro PBS degradation was observed on the 15th day after the bulk hydrolysis mechanism. Hence, on the basis of the study outcomes, we affirm that the MgO/gC3N4@PAN/PEG nanofibrous membrane can act as a potential bone regenerative substrate.
    Matched MeSH terms: Bone Regeneration
  2. Abdelrasoul M, El-Fattah AA, Kotry G, Ramadan O, Essawy M, Kamaldin J, et al.
    Oral Dis, 2023 Nov;29(8):3583-3598.
    PMID: 35839150 DOI: 10.1111/odi.14314
    BACKGROUND: Periodontal regenerative therapy using bone-substituting materials has gained favorable clinical significance in enhancing osseous regeneration. These materials should be biocompatible, osteogenic, malleable, and biodegradable. This study assessed the periodontal regenerative capacity of a novel biodegradable bioactive hydrogel template of organic-inorganic composite loaded with melatonin.

    MATERIALS AND METHODS: A melatonin-loaded alginate-chitosan/beta-tricalcium phosphate composite hydrogel was successfully prepared and characterized. Thirty-six critical-sized bilateral class II furcation defects were created in six Mongrel dogs, and were randomly divided and allocated to three cohorts; sham, unloaded composite, and melatonin-loaded. Periodontal regenerative capacity was evaluated via histologic and histomorphometric analysis.

    RESULTS: Melatonin-treated group showed accelerated bone formation and advanced maturity, with a significant twofold increase in newly formed inter-radicular bone compared with the unloaded composite. The short-term regenerative efficacy was evident 4 weeks postoperatively as a significant increase in cementum length concurrent with reduction of entrapped epithelium. After 8 weeks, the scaffold produced a quality of newly synthesized bone similar to normal compact bone, with potent periodontal ligament attachment.

    CONCLUSIONS: Melatonin-loaded hydrogel template accelerated formation and enhanced quality of newly formed bone, allowing complete periodontal regeneration. Furthermore, the scaffold prevented overgrowth and entrapment of epithelial cells in furcation defects.

    Matched MeSH terms: Bone Regeneration; Guided Tissue Regeneration, Periodontal
  3. Tuygunov N, Zakaria MN, Yahya NA, Abdul Aziz A, Cahyanto A
    J Mech Behav Biomed Mater, 2023 Oct;146:106099.
    PMID: 37660446 DOI: 10.1016/j.jmbbm.2023.106099
    Bone regeneration is a rapidly growing field that seeks to develop new biomaterials to regenerate bone defects. Conventional bone graft materials have limitations, such as limited availability, complication, and rejection. Glass ionomer cement (GIC) is a biomaterial with the potential for bone regeneration due to its bone-contact biocompatibility, ease of use, and cost-effectiveness. GIC is a two-component material that adheres to the bone and releases ions that promote bone growth and mineralization. A systematic literature search was conducted using PubMed-MEDLINE, Scopus, and Web of Science databases and registered in the PROSPERO database to determine the evidence regarding the efficacy and bone-contact biocompatibility of GIC as bone cement. Out of 3715 initial results, thirteen studies were included in the qualitative synthesis. Two tools were employed in evaluating the Risk of Bias (RoB): the QUIN tool for assessing in vitro studies and SYRCLE for in vivo. The results indicate that GIC has demonstrated the ability to adhere to bone and promote bone growth. Establishing a chemical bond occurs at the interface between the GIC and the mineral phase of bone. This interaction allows the GIC to exhibit osteoconductive properties and promote the growth of bone tissue. GIC's bone-contact biocompatibility, ease of preparation, and cost-effectiveness make it a promising alternative to conventional bone grafts. However, further research is required to fully evaluate the potential application of GIC in bone regeneration. The findings hold implications for advancing material development in identifying the optimal composition and fabrication of GIC as a bone repair material.
    Matched MeSH terms: Bone Regeneration
  4. Shahemi NH, Mahat MM, Asri NAN, Amir MA, Ab Rahim S, Kasri MA
    ACS Biomater Sci Eng, 2023 Jul 10;9(7):4045-4085.
    PMID: 37364251 DOI: 10.1021/acsbiomaterials.3c00194
    Spinal cord injury (SCI) causes severe motor or sensory damage that leads to long-term disabilities due to disruption of electrical conduction in neuronal pathways. Despite current clinical therapies being used to limit the propagation of cell or tissue damage, the need for neuroregenerative therapies remains. Conductive hydrogels have been considered a promising neuroregenerative therapy due to their ability to provide a pro-regenerative microenvironment and flexible structure, which conforms to a complex SCI lesion. Furthermore, their conductivity can be utilized for noninvasive electrical signaling in dictating neuronal cell behavior. However, the ability of hydrogels to guide directional axon growth to reach the distal end for complete nerve reconnection remains a critical challenge. In this Review, we highlight recent advances in conductive hydrogels, including the incorporation of conductive materials, fabrication techniques, and cross-linking interactions. We also discuss important characteristics for designing conductive hydrogels for directional growth and regenerative therapy. We propose insights into electrical conductivity properties in a hydrogel that could be implemented as guidance for directional cell growth for SCI applications. Specifically, we highlight the practical implications of recent findings in the field, including the potential for conductive hydrogels to be used in clinical applications. We conclude that conductive hydrogels are a promising neuroregenerative therapy for SCI and that further research is needed to optimize their design and application.
    Matched MeSH terms: Nerve Regeneration
  5. Heng BC, Bai Y, Li X, Meng Y, Lu Y, Zhang X, et al.
    Animal Model Exp Med, 2023 Apr;6(2):120-130.
    PMID: 36856186 DOI: 10.1002/ame2.12300
    Understanding the bioelectrical properties of bone tissue is key to developing new treatment strategies for bone diseases and injuries, as well as improving the design and fabrication of scaffold implants for bone tissue engineering. The bioelectrical properties of bone tissue can be attributed to the interaction of its various cell lineages (osteocyte, osteoblast and osteoclast) with the surrounding extracellular matrix, in the presence of various biomechanical stimuli arising from routine physical activities; and is best described as a combination and overlap of dielectric, piezoelectric, pyroelectric and ferroelectric properties, together with streaming potential and electro-osmosis. There is close interdependence and interaction of the various electroactive and electrosensitive components of bone tissue, including cell membrane potential, voltage-gated ion channels, intracellular signaling pathways, and cell surface receptors, together with various matrix components such as collagen, hydroxyapatite, proteoglycans and glycosaminoglycans. It is the remarkably complex web of interactive cross-talk between the organic and non-organic components of bone that define its electrophysiological properties, which in turn exerts a profound influence on its metabolism, homeostasis and regeneration in health and disease. This has spurred increasing interest in application of electroactive scaffolds in bone tissue engineering, to recapitulate the natural electrophysiological microenvironment of healthy bone tissue to facilitate bone defect repair.
    Matched MeSH terms: Bone Regeneration*
  6. Al Qabbani A, Rani KGA, AlKawas S, Sheikh Abdul Hamid S, Yap Abdullah A, Samsudin AR, et al.
    PLoS One, 2023;18(12):e0294291.
    PMID: 38127838 DOI: 10.1371/journal.pone.0294291
    The aim of this study was to compare the ability of demineralized (DMB) and decellularized (DCC) bovine bone granules to support bone regeneration in rat calvaria critical-size defects. DMB and DCC were prepared using a previously published method. The granule size used ranged between 500 and 750 μm. A total of forty-eight Sprague-Dawley rats were divided into two groups (n = 24). A pair of 5 mm diameter defects were created on the calvaria of the rats in the right and left parietal bone in both groups. Group A animals received DMB granules and Group B received DCC granules in the right parietal defect side while the left parietal untreated defect acted as sham surgery for both groups. Four animals per group were euthanized in a CO2 chamber at day 7, 14 and 21 post-surgery and the calvaria implantation site biopsy harvested was subjected to osteogenic gene expression analysis. Another four animals per group were euthanized at days 15, 30 and 60 post surgery and the calvaria implantation site biopsy harvested was subjected to histological, immunohistochemistry, RAMAN spectroscopy and Micro-CT analysis at the mentioned time points. Statistical analysis was conducted using t-tests and ANOVA. Histomorphometry showed significantly higher new bone formation in the DCC sites (p<0.05) compared to DMB. Both DMB and DCC implantation sites showed distinct staining for osteocalcin and osteopontin proteins compared to their respective sham sites. By day 21 after implantation, DCC sites demonstrated significantly elevated mRNA levels of osteonectin (p<0.001), osteopontin (p<0.001), osteocalcin (p<0.0001), ALP (p<0.01), and BMP-2 (p<0.001) compared to DMB. However, VEGF expression showed no significant differences at this time point between the two groups. Micro-CT analysis also showed enhanced defect closure and higher bone density in DCC implanted sites while RAMAN spectra demonstrated increased abundance of collagen and bone minerals, especially, PO43- ions than DMB. In conclusion, both DMB and DCC granules demonstrated favorable osteogenic potential in critical-sized defects, with DCC exhibited superior osteoconductive, osteoinductive and osteogenesis properties.
    Matched MeSH terms: Bone Regeneration
  7. Vijayanathan Y, Hamzah NM, Lim SM, Lim FT, Tan MP, Majeed ABA, et al.
    Brain Res Bull, 2022 Nov;190:218-233.
    PMID: 36228872 DOI: 10.1016/j.brainresbull.2022.10.001
    In order to understand the biological processes underlying dopaminergic neurons (DpN) regeneration in a 6-hydroxydopamine(6-OHDA)-induced adult zebrafish-based Parkinson's disease model, this study investigated the specific phases of neuroregeneration in a time-based manner. Bromodeoxyuridine (BrdU) was administered 24 h before the harvest of brain tissues at day three, five, seven, nine, 12 and 14 postlesion. Potential migration of proliferative cells was tracked over 14 days postlesion through double-pulse tracking [BrdU and 5-ethynyl-2'-deoxyuridine (EdU)] of cells and immunohistostaining of astrocytes [glial fibrillary acidic protein (GFAP)]. Gene expression of foxa2 and nurr1 (nr4a2a) at day three, nine, 14, 18, 22 and 30 postlesion was quantified using qPCR. Protein expression of foxa2 at day three, seven, 14 and 22 postlesion was validated using the western blot technique. Double labelling [EdU and tyrosine hydroxylase (TH)] of proliferative cells was performed to ascertain their fate after the neuroregeneration processes. It was found that whilst cell proliferation remained unchanged in the area of substantial DpN loss, the ventral diencephalon (vDn), there was a transient increase of cell proliferation in the olfactory bulb (OB) and telencephalon (Tel) seven days postlesion. BrdU-immunoreactive (ir)/ EdU-ir cells and activated astrocytes were later found to be significantly increased in the vDn and its nearby area (Tel) 14 days postlesion. There was a significant but transient downregulation of foxa2 at day three and nine postlesion, and nr4a2a at day three, nine and 14 postlesion. The expression of both genes remained unchanged in the OB and Tel. There was a transient downregulation of foxa2 protein expression at day three and seven postlesion. The significant increase of EdU-ir/ TH-ir cells in the vDn 30 days postlesion indicates maturation of proliferative cells (formed between day five-seven postlesion) into DpN. The present findings warrant future investigation of critical factors that govern the distinctive phases of DpN regeneration.
    Matched MeSH terms: Nerve Regeneration/physiology
  8. Luchman NA, Megat Abdul Wahab R, Zainal Ariffin SH, Nasruddin NS, Lau SF, Yazid F
    PeerJ, 2022;10:e13356.
    PMID: 35529494 DOI: 10.7717/peerj.13356
    BACKGROUND: The selection of appropriate scaffold plays an important role in ensuring the success of bone regeneration. The use of scaffolds with different materials and their effect on the osteogenic performance of cells is not well studied and this can affect the selection of suitable scaffolds for transplantation. Hence, this study aimed to investigate the comparative ability of two different synthetic scaffolds, mainly hydroxyapatite (HA) and polycaprolactone (PCL) scaffolds in promoting in vitro and in vivo bone regeneration.

    METHOD: In vitro cell viability, morphology, and alkaline phosphatase (ALP) activity of MC3T3-E1 cells on HA and PCL scaffolds were determined in comparison to the accepted model outlined for two-dimensional systems. An in vivo study involving the transplantation of MC3T3-E1 cells with scaffolds into an artificial bone defect of 4 mm length and 1.5 mm depth in the rat's left maxilla was conducted. Three-dimensional analysis using micro-computed tomography (micro-CT), hematoxylin and eosin (H&E), and immunohistochemistry analyses evaluation were performed after six weeks of transplantation.

    RESULTS: MC3T3-E1 cells on the HA scaffold showed the highest cell viability. The cell viability on both scaffolds decreased after 14 days of culture, which reflects the dominant occurrence of osteoblast differentiation. An early sign of osteoblast differentiation can be detected on the PCL scaffold. However, cells on the HA scaffold showed more prominent results with intense mineralized nodules and significantly (p 

    Matched MeSH terms: Bone Regeneration
  9. Lee SY, Thow SY, Abdullah S, Ng MH, Mohamed Haflah NH
    Int J Nanomedicine, 2022;17:6723-6758.
    PMID: 36600878 DOI: 10.2147/IJN.S362144
    Peripheral nerve injury (PNI) is a worldwide problem which hugely affects the quality of patients' life. Nerve conduits are now the alternative for treatment of PNI to mimic the gold standard, autologous nerve graft. In that case, with the advantages of electrospun micro- or nano-fibers nerve conduit, the peripheral nerve growth can be escalated, in a better way. In this systematic review, we focused on 39 preclinical studies of electrospun nerve conduit, which include the in vitro and in vivo evaluation from animal peripheral nerve defect models, to provide an update on the progress of the development of electrospun nerve conduit over the last 5 years (2016-2021). The physical characteristics, biocompatibility, functional and morphological outcomes of nerve conduits from different studies would be compared, to give a better strategy for treatment of PNI.
    Matched MeSH terms: Nerve Regeneration/physiology
  10. Ngah NA, Dias GJ, Tong DC, Mohd Noor SNF, Ratnayake J, Cooper PR, et al.
    Molecules, 2021 Nov 25;26(23).
    PMID: 34885714 DOI: 10.3390/molecules26237131
    BACKGROUND: Platelet-rich fibrin (PRF) has gained popularity in craniofacial surgery, as it provides an excellent reservoir of autologous growth factors (GFs) that are essential for bone regeneration. However, the low elastic modulus, short-term clinical application, poor storage potential and limitations in emergency therapy use restrict its more widespread clinical application. This study fabricates lyophilised PRF (Ly-PRF), evaluates its physical and biological properties, and explores its application for craniofacial tissue engineering purposes.

    MATERIAL AND METHODS: A lyophilisation method was applied, and the outcome was evaluated and compared with traditionally prepared PRF. We investigated how lyophilisation affected PRF's physical characteristics and biological properties by determining: (1) the physical and morphological architecture of Ly-PRF using SEM, and (2) the kinetic release of PDGF-AB using ELISA.

    RESULTS: Ly-PRF exhibited a dense and homogeneous interconnected 3D fibrin network. Moreover, clusters of morphologically consistent cells of platelets and leukocytes were apparent within Ly-PRF, along with evidence of PDGF-AB release in accordance with previously reports.

    CONCLUSIONS: The protocol established in this study for Ly-PRF preparation demonstrated versatility, and provides a biomaterial with growth factor release for potential use as a craniofacial bioscaffold.

    Matched MeSH terms: Bone Regeneration/drug effects
  11. Yadav A, Huang TC, Chen SH, Ramasamy TS, Hsueh YY, Lin SP, et al.
    J Neuroinflammation, 2021 Oct 16;18(1):238.
    PMID: 34656124 DOI: 10.1186/s12974-021-02273-1
    BACKGROUND: Epigenetic regulation by histone deacetylases (HDACs) in Schwann cells (SCs) after injury facilitates them to undergo de- and redifferentiation processes necessary to support various stages of nerve repair. Although de-differentiation activates the synthesis and secretion of inflammatory cytokines by SCs to initiate an immune response during nerve repair, changes in either the timing or duration of prolonged inflammation mediated by SCs can affect later processes associated with repair and regeneration. Limited studies have investigated the regulatory processes through which HDACs in SCs control inflammatory cytokines to provide a favorable environment for peripheral nerve regeneration.

    METHODS: We employed the HDAC inhibitor (HDACi) sodium phenylbutyrate (PBA) to address this question in an in vitro RT4 SC inflammation model and an in vivo sciatic nerve transection injury model to examine the effects of HDAC inhibition on the expression of pro-inflammatory cytokines. Furthermore, we assessed the outcomes of suppression of extended inflammation on the regenerative potential of nerves by assessing axonal regeneration, remyelination, and reinnervation.

    RESULTS: Significant reductions in lipopolysaccharide (LPS)-induced pro-inflammatory cytokine (tumor necrosis factor-α [TNFα]) expression and secretion were observed in vitro following PBA treatment. PBA treatment also affected the transient changes in nuclear factor κB (NFκB)-p65 phosphorylation and translocation in response to LPS induction in RT4 SCs. Similarly, PBA mediated long-term suppressive effects on HDAC3 expression and activity. PBA administration resulted in marked inhibition of pro-inflammatory cytokine secretion at the site of transection injury when compared with that in the hydrogel control group at 6-week post-injury. A conducive microenvironment for axonal regrowth and remyelination was generated by increasing expression levels of protein gene product 9.5 (PGP9.5) and myelin basic protein (MBP) in regenerating nerve tissues. PBA administration increased the relative gastrocnemius muscle weight percentage and maintained the intactness of muscle bundles when compared with those in the hydrogel control group.

    CONCLUSIONS: Suppressing the lengthened state of inflammation using PBA treatment favors axonal regrowth and remyelination following nerve transection injury. PBA treatment also regulates pro-inflammatory cytokine expression by inhibiting the transcriptional activation of NFκB-p65 and HDAC3 in SCs in vitro.

    Matched MeSH terms: Nerve Regeneration/drug effects; Nerve Regeneration/physiology*
  12. Ballouze R, Marahat MH, Mohamad S, Saidin NA, Kasim SR, Ooi JP
    J Biomed Mater Res B Appl Biomater, 2021 Oct;109(10):1426-1435.
    PMID: 33484103 DOI: 10.1002/jbm.b.34802
    Autologous bone grafting remains the gold standard for almost all bone void-filling orthopedic surgery. However, autologous bone grafting has several limitations, thus scientists are trying to identify an ideal synthetic material as an alternative bone graft substitute. Magnesium-doped biphasic calcium phosphate (Mg-BCP) has recently been in the spotlight and is considered to be a potential bone substitute. The Mg-BCP is a mixture of two bioceramics, that is, hydroxyapatite (HA) and β-tricalcium phosphate (β-TCP), doped with Mg2+ , and can be synthesized through chemical wet-precipitation, sol-gel, single diffusion gel, and solid state reactions. Regardless of the synthesis routes, it is found that the Mg2+ preferentially accommodates in β-TCP lattice instead of the HA lattice. The addition of Mg2+ to BCP leads to desirable physicochemical properties and is found to enhance the apatite-forming ability as compared to pristine BCP. In vitro results suggest that the Mg-BCP is bioactive and not toxic to cells. Implantation of Mg-BCP in in vivo models further affirmed its biocompatibility and efficacy as a bone substitute. However, like the other bioceramics, the optimum physicochemical properties of the Mg-BCP scaffold have yet to be determined. Further investigations are required regarding Mg-BCP applications in bone tissue engineering.
    Matched MeSH terms: Bone Regeneration
  13. Ching KY, Andriotis O, Sengers B, Stolz M
    J Biomater Appl, 2021 09;36(3):503-516.
    PMID: 33730922 DOI: 10.1177/08853282211002015
    Towards optimizing the growth of extracellular matrix to produce repair cartilage for healing articular cartilage (AC) defects in joints, scaffold-based tissue engineering approaches have recently become a focus of clinical research. Scaffold-based approaches by electrospinning aim to support the differentiation of chondrocytes by providing an ultrastructure similar to the fibrillar meshwork in native cartilage. In a first step, we demonstrate how the blending of chitosan with poly(ethylene oxide) (PEO) allows concentrated chitosan solution to become electrospinnable. The chitosan-based scaffolds share the chemical structure and characteristics of glycosaminoglycans, which are important structural components of the cartilage extracellular matrix. Electrospinning produced nanofibrils of ∼100 nm thickness that are closely mimicking the size of collagen fibrils in human AC. The polymer scaffolds were stabilized in physiological conditions and their stiffness was tuned by introducing the biocompatible natural crosslinker genipin. We produced scaffolds that were crosslinked with 1.0% genipin to obtain values of stiffness that were in between the stiffness of the superficial zone human AC of 600 ± 150 kPa and deep zone AC of 1854 ± 483 kPa, whereas the stiffness of 1.5% genipin crosslinked scaffold was similar to the stiffness of deep zone AC. The scaffolds were degradable, which was indicated by changes in the fibril structure and a decrease in the scaffold stiffness after seven months. Histological and immunohistochemical analysis after three weeks of culture with human articular chondrocytes (HACs) showed a cell viability of over 90% on the scaffolds and new extracellular matrix deposited on the scaffolds.
    Matched MeSH terms: Regeneration
  14. Yow YY, Goh TK, Nyiew KY, Lim LW, Phang SM, Lim SH, et al.
    Cells, 2021 08 25;10(9).
    PMID: 34571842 DOI: 10.3390/cells10092194
    Despite the progressive advances, current standards of treatments for peripheral nerve injury do not guarantee complete recovery. Thus, alternative therapeutic interventions should be considered. Complementary and alternative medicines (CAMs) are widely explored for their therapeutic value, but their potential use in peripheral nerve regeneration is underappreciated. The present systematic review, designed according to guidelines of Preferred Reporting Items for Systematic Review and Meta-Analysis Protocols, aims to present and discuss the current literature on the neuroregenerative potential of CAMs, focusing on plants or herbs, mushrooms, decoctions, and their respective natural products. The available literature on CAMs associated with peripheral nerve regeneration published up to 2020 were retrieved from PubMed, Scopus, and Web of Science. According to current literature, the neuroregenerative potential of Achyranthes bidentata, Astragalus membranaceus, Curcuma longa, Panax ginseng, and Hericium erinaceus are the most widely studied. Various CAMs enhanced proliferation and migration of Schwann cells in vitro, primarily through activation of MAPK pathway and FGF-2 signaling, respectively. Animal studies demonstrated the ability of CAMs to promote peripheral nerve regeneration and functional recovery, which are partially associated with modulations of neurotrophic factors, pro-inflammatory cytokines, and anti-apoptotic signaling. This systematic review provides evidence for the potential use of CAMs in the management of peripheral nerve injury.
    Matched MeSH terms: Nerve Regeneration/drug effects*
  15. Subramaniam T, Fauzi MB, Lokanathan Y, Law JX
    Int J Mol Sci, 2021 Jun 17;22(12).
    PMID: 34204292 DOI: 10.3390/ijms22126486
    Skin injury is quite common, and the wound healing is a complex process involving many types of cells, the extracellular matrix, and soluble mediators. Cell differentiation, migration, and proliferation are essential in restoring the integrity of the injured tissue. Despite the advances in science and technology, we have yet to find the ideal dressing that can support the healing of cutaneous wounds effectively, particularly for difficult-to-heal chronic wounds such as diabetic foot ulcers, bed sores, and venous ulcers. Hence, there is a need to identify and incorporate new ideas and methods to design a more effective dressing that not only can expedite wound healing but also can reduce scarring. Calcium has been identified to influence the wound healing process. This review explores the functions and roles of calcium in skin regeneration and reconstruction during would healing. Furthermore, this review also investigates the possibility of incorporating calcium into scaffolds and examines how it modulates cutaneous wound healing. In summary, the preliminary findings are promising. However, some challenges remain to be addressed before calcium can be used for cutaneous wound healing in clinical settings.
    Matched MeSH terms: Regeneration
  16. Bajuri MY, Selvanathan N, Dzeidee Schaff FN, Abdul Suki MH, Ng AMH
    Tissue Eng Regen Med, 2021 06;18(3):377-385.
    PMID: 34043210 DOI: 10.1007/s13770-021-00343-2
    BACKGROUND: Managing massive bone defects, a great challenge to orthopaedics reconstructive surgery. The problem arise is the supply of suitable bone is limited with many complications. Tissue-engineered hydroxyapatite bone (TEHB) scaffold impregnated with osteoprogenitor cells developed as an alternative to promote bone regeneration.

    METHODS: This animal protocol has been approved by Universiti Kebangsaan Malaysia Animal Ethical Committee. The TEHB scaffold prepared from hydroxyapatite using gel casting method. A total of six adolescent female sheep were chosen for this study. Later, all the sheep were euthanized in a proper manner and the bone harvested for biomechanical study. Bone marrow was collected from iliac crest of the sheep and bone marrow stem cells (BMSCs) isolated and cultured. BMSCs then cultured in osteogenic medium for osteoprogenitor cells development and the plasma collected was seeded with osteoprogenitor cells mixed with calcium chloride. Bone defect of 3 cm length of tibia bone created from each sheep leg and implanted with autologous and TEHB scaffold in 2 different groups of sheep. Wound site was monitored weekly until the wound completely healed and conventional X-ray performed at week 1 and 24. Shear test was conducted to determine the shear force on the autologous bone and TEHB scaffold after implantation for 24 weeks.

    RESULTS: All of the sheep survived without any complications during the study period and radiograph showed new bone formation. Later, the bone harvested was for biomechanical study. The highest shear force for the autologous group was 13 MPa and the lowest was 5 MPa while for the scaffold group, the highest was 10 MPa and the lowest was 3 MPa. Although, proximal and distal interface of autologous bone graft shows higher shear strength compared to the TEHB scaffold but there is no significant difference in both groups, p value > 0.05. Histologically in both proximal and distal interface in both arms shows bone healing and woven bone formation.

    CONCLUSION: TEHB scaffold impregnated with osteoprogenitor cells has the potential to be developed as a bone substitute in view of its strength and capability to promote bone regeneration.

    Matched MeSH terms: Bone Regeneration
  17. Amuthan A, Devi V, Shreedhara CS, Rao V, Jasphin S, Kumar N
    J Tradit Complement Med, 2021 May;11(3):279-286.
    PMID: 34012874 DOI: 10.1016/j.jtcme.2020.08.004
    Background: Traditional Siddha Medicine advises using metal-based formulations to treat cancers. In the case of any toxicities during the therapy, Siddha physicians use Vernonia cinerea (VC) whole plant kashayam (crude aqueous extract-CAE) to reverse the toxic effects.

    Aim: To evaluate the nephroprotective activity of CAE and its fractions in cisplatin-induced nephrotoxicity and to assess whether they compromise the anticancer efficacy of cisplatin.

    Materials and methods: Cisplatin-induced renal damage was induced in Ehrlich Ascites Carcinoma (EAC) bearing mice during mild phase of tumor growth. CAE and its butanol (BF) and aqueous (AF) fractions were administered orally from the 5th day for five days. Nephroprotective potential (serum urea, creatinine, renal histology) and effect of VC on cisplatin anticancer efficacy (tumor volume, viable tumor cells, percentage increase in life span (% ILS)) were calculated.

    Result: CAE and its fractions significantly reversed the cisplatin-induced renal damage. CAE and BF treated animals showed regeneration of 50%-75% of proximal tubular cells. Compared to EAC control mice, the % ILS of the cisplatin-treated group was 244% and it was further extended to 379% after CAE administration. The % ILS in the CAE treated group was 1.6 times higher than the cisplatin alone treated group. GC-MS study showed the presence of astaxanthin and betulin.

    Conclusion: CAE of VC reverses cisplatin-induced kidney damage as well as regenerates proximal tubular epithelial cells, without compromising the anticancer effect of cisplatin. When CAE was further fractionated, the nephroprotective activity was retained, but the beneficial anticancer effect of cisplatin was compromised.

    Matched MeSH terms: Regeneration
  18. Cui YC, Qiu YS, Wu Q, Bu G, Peli A, Teh SW, et al.
    Exp Biol Med (Maywood), 2021 May;246(10):1177-1183.
    PMID: 33535809 DOI: 10.1177/1535370220985468
    Osteoblasts play an important role in bone regeneration and repair. The hypoxia condition in bone occurs when bone undergoes fracture, and this will trigger a series of biochemical and mechanical changes to enable bone repair. Hence, it is interesting to observe the metabolites and metabolism changes when osteoblasts are exposed to hypoxic condition. This study has looked into the response of human osteoblast hFOB 1.19 under normoxic and hypoxic conditions by observing the cell growth and utilization of metabolites via Phenotype MicroArrays™ under these two different oxygen concentrations. The cell growth of hFOB 1.19 under hypoxic condition showed better growth compared to hFOB 1.19 under normal condition. In this study, osteoblast used glycolysis as the main pathway to produce energy as hFOB 1.19 in both hypoxic and normoxic conditions showed cell growth in well containing dextrin, glycogen, maltotriose, D-maltose, D-glucose-6-phospate, D-glucose, D-mannose, D-Turanose, D-fructose-6-phosphate, D-galactose, uridine, adenosine, inosine and α-keto-glutaric acid. In hypoxia, the cells have utilized additional metabolites such as α-D-glucose-1-phosphate and D-fructose, indicating possible activation of glycogen synthesis and glycogenolysis to metabolize α-D-glucose-1-phosphate. Meanwhile, during normoxia, D-L-α-glycerol phosphate was used, and this implies that the osteoblast may use glycerol-3-phosphate shuttle and oxidative phosphorylation to metabolize glycerol-3-phosphate.
    Matched MeSH terms: Bone Regeneration
  19. Nizam NUM, Hanafiah MM, Mahmoudi E, Halim AA, Mohammad AW
    Sci Rep, 2021 Apr 21;11(1):8623.
    PMID: 33883637 DOI: 10.1038/s41598-021-88084-z
    In this study, two biomass-based adsorbents were used as new precursors for optimizing synthesis conditions of a cost-effective powdered activated carbon (PAC). The PAC removed dyes from an aqueous solution using carbonization and activation by KOH, NaOH, and H2SO4. The optimum synthesis, activation temperature, time and impregnation ratio, removal rate, and uptake capacity were determined. The optimum PAC was analyzed and characterized using Fourier-transform infrared spectroscopy (FTIR), x-ray diffraction (XRD), a field emission scanning electron microscope (FESEM), Zeta potential, and Raman spectroscopy. Morphological studies showed single-layered planes with highly porous surfaces, especially PAC activated by NaOH and H2SO4. The results showed that the experimental data were well-fitted with a pseudo-second-order model. Based on Langmuir isotherm, the maximum adsorption capacity for removing methylene blue (MB) was 769.23 mg g-1 and 458.43 mg g-1 for congo red (CR). Based on the isotherm models, more than one mechanism was involved in the adsorption process, monolayer for the anionic dye and multilayer for the cationic dye. Elovich and intraparticle diffusion kinetic models showed that rubber seed shells (RSS) has higher α values with a greater tendency to adsorb dyes compared to rubber seed (RS). A thermodynamic study showed that both dyes' adsorption process was spontaneous and exothermic due to the negative values of the enthalpy (ΔH) and Gibbs free energy (ΔG). The change in removal efficiency of adsorbent for regeneration study was observed in the seventh cycles, with a 3% decline in the CR and 2% decline in MB removal performance. This study showed that the presence of functional groups and active sites on the produced adsorbent (hydroxyl, alkoxy, carboxyl, and π - π) contributed to its considerable affinity for adsorption in dye removal. Therefore, the optimum PAC can serve as efficient and cost-effective adsorbents to remove dyes from industrial wastewater.
    Matched MeSH terms: Regeneration
  20. Nour S, Imani R, Chaudhry GR, Sharifi AM
    J Biomed Mater Res A, 2021 04;109(4):453-478.
    PMID: 32985051 DOI: 10.1002/jbm.a.37105
    Skin injuries and in particular, chronic wounds, are one of the major prevalent medical problems, worldwide. Due to the pivotal role of angiogenesis in tissue regeneration, impaired angiogenesis can cause several complications during the wound healing process and skin regeneration. Therefore, induction or promotion of angiogenesis can be considered as a promising approach to accelerate wound healing. This article presents a comprehensive overview of current and emerging angiogenesis induction methods applied in several studies for skin regeneration, which are classified into the cell, growth factor, scaffold, and biological/chemical compound-based strategies. In addition, the advantages and disadvantages of these angiogenic strategies along with related research examples are discussed in order to demonstrate their potential in the treatment of wounds.
    Matched MeSH terms: Regeneration/drug effects
Filters
Contact Us

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

External Links