ArticlePublisher preview availableLiterature Review

The useful agent to have an ideal biological scaffold

Authors:
To read the full-text of this research, you can request a copy directly from the authors.

Abstract and Figures

Tissue engineering which is applied in regenerative medicine has three basic components: cells, scaffolds and growth factors. This multidisciplinary field can regulate cell behaviors in different conditions using scaffolds and growth factors. Scaffolds perform this regulation with their structural, mechanical, functional and bioinductive properties and growth factors by attaching to and activating their receptors in cells. There are various types of biological extracellular matrix (ECM) and polymeric scaffolds in tissue engineering. Recently, many researchers have turned to using biological ECM rather than polymeric scaffolds because of its safety and growth factors. Therefore, selection the right scaffold with the best properties tailored to clinical use is an ideal way to regulate cell behaviors in order to repair or improve damaged tissue functions in regenerative medicine. In this review we first divided properties of biological scaffold into intrinsic and extrinsic elements and then explain the components of each element. Finally, the types of scaffold storage methods and their advantages and disadvantages are examined.
This content is subject to copyright. Terms and conditions apply.
FULL LENGTH REVIEW
The useful agent to have an ideal biological scaffold
Raziyeh Kheirjou .Jafar Soleimani Rad .Ahad Ferdowsi Khosroshahi .
Leila Roshangar
Received: 9 April 2020 / Accepted: 3 November 2020 / Published online: 22 November 2020
ÓSpringer Nature B.V. 2020
Abstract Tissue engineering which is applied in
regenerative medicine has three basic components:
cells, scaffolds and growth factors. This multidisci-
plinary field can regulate cell behaviors in different
conditions using scaffolds and growth factors. Scaf-
folds perform this regulation with their structural,
mechanical, functional and bioinductive properties
and growth factors by attaching to and activating their
receptors in cells. There are various types of biological
extracellular matrix (ECM) and polymeric scaffolds in
tissue engineering. Recently, many researchers have
turned to using biological ECM rather than polymeric
scaffolds because of its safety and growth factors.
Therefore, selection the right scaffold with the best
properties tailored to clinical use is an ideal way to
regulate cell behaviors in order to repair or improve
damaged tissue functions in regenerative medicine. In
this review we first divided properties of biological
scaffold into intrinsic and extrinsic elements and then
explain the components of each element. Finally, the
types of scaffold storage methods and their advantages
and disadvantages are examined.
Keywords Tissue engineering Decellularization
Biological scaffold Extracellular matrix Tissue
banking Storage
Introduction
Tissue engineering has appeared in the 1980s. This
multidisciplinary field is applied in regenerative
medicine to help various damaged tissues and organs,
and it is based on using of cells, scaffolds, and
bioactive factors. Scaffolds not only provide a sup-
portive template for cell attachment, but they also
create a biomechanical and physical environment. So
the scaffolds play an active role in the regulation of
cell behaviors (Qiu 2012).
Because of the toxic and inflammatory capacity of
synthetic polymers, which lead to reducing extracel-
lular matrix (ECM) remodeling and growth capacity,
the xeno-or allogeneic tissues are substituted to
biodegradable synthetic scaffolds (Thompson 1992).
The cells of xeno-or allogeneic tissues as biological
scaffolds, are removed, and their ECM remains as
3-dimensioal (3D) structure (Badylak et al. 2009).
These natural ECMs decrease immune and inflamma-
tory response in grafting through decellularization,
and serve as inductive means through their structural
and functional proteins and endogenous growth fac-
tors (Assmann 2013; Badylak et al. 2012).
R. Kheirjou A. F. Khosroshahi
Department of Anatomical Sciences, Faculty of Medicine,
Tabriz University of Medical Sciences, Tabriz, Iran
J. S. Rad L. Roshangar (&)
Stem Cell Research Center, Tabriz University of Medical
Sciences, 33363879 Tabriz, Iran
e-mail: lroshangar@yahoo.com
123
Cell Tissue Bank (2021) 22:225–239
https://doi.org/10.1007/s10561-020-09881-w(0123456789().,-volV)(0123456789().,-volV)
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
... 7 This therapy is based on the tissue engineering triad: stem cells, growth factors and matrices, 8,9 requiring the association of these three components for the repair process of these structures to occur. 10,11 One of the most important matrix structures is the scaffold, which can be defined as structural support that will fill and support the regenerated tissue. These biomaterials act mainly on dimensional and biomechanical stability, to the point that the regenerated tissue itself can assume this function. ...
... 15,16,17,18 Selecting the scaffold composition according to the desired clinical situation is one way to regulate the cells behavior to repair or improve the functions of damaged tissue in regenerative medicine. 11 Within the variety of scaffolds and membrane matrix compositions, Chitosan and Xanthan gum polymeric complexes have shown promising results when applied in advanced cell therapy. 19,20 When chitosan and xanthan are combined to form the chitosan-xanthan complex, there are an improvement on the mechanical properties and on the stability to pH variations, without compromising the physicochemical and biological properties of chitosan and xanthan when not associated. ...
Article
The aim of this paper was to synthesize and characterize polymeric scaffolds of Chitosan/Xanthan/Hydroxyapatite-Graphene Oxide nanocomposite associated with mesenchymal stem cells for regenerative dentistry application. The chitosan-xanthan gum (CX) complex was associated with Hydroxyapatite-Graphene Oxide (HA-GO) nanocomposite with different Graphene Oxides (GO) concentration (0.5 wt%; 1.0 wt%; 1.5 wt%). The scaffolds characterizations were performed by X-ray diffraction (XRD), Fourier Transform Infrared Spectroscopy (FTIR), Raman spectroscopy, thermogravimetric analysis (TGA), scanning electron microscopy (SEM) and contact angle. The mechanical properties were assessed by compressive strength. The in vitro bioactivity and the in vitro cytotoxicity test (MTT test) were analyzed as well. The data was submitted to the Normality and Homogeneity tests. In vitro Indirect Cytotoxicity assay data was statistically analyzed by ANOVA two-way, followed by Tukey’s test (α = 0.05). Compressive strength and contact angle data were statistically analyzed by one-way ANOVA, followed by Tukey’s test (α = 0.05). XRD showed the presence of Hydroxyapatite (HA) peaks in the structures CXHA, CXHAGO 0.5%,1.0% and 1.5%. FT-IR showed amino and carboxylic bands characteristic of CX. Raman spectroscopy analysis evidenced a high quality of the GO. In the TGA it was observed the mass loss associated with the CX degradation by depolymerization. SEM analysis showed pores in the scaffolds, in addition to HA incorporated and adhered to the polymer. Contact angle test showed that scaffolds have a hydrophilic characteristic, with the CX group the highest contact angle and CXHA the lowest ( p < 0.05). 1.0 wt% GO significantly increased the compressive strength compared to other compositions. In the bioactivity test, the apatite crystals precipitation on the scaffold surface was observed. MTT test showed high cell viability in CXHAGO 1.0% and CXHAGO 1.5% scaffold. CXHAGO scaffolds are promising for regenerative dentistry application because they have morphological characteristics, mechanical and biological properties favorable for the regeneration process.
... Tissue engineering is considered an important therapeutic tool in regenerative medicine; one of the crucial factors for tissue engineering is the scaffolds, which provide a platform for cells' adhesion while allowing their growth [1,2]. Scaffolds are usually made from different materials, including ceramics and polymers. ...
Article
Full-text available
It has been reported that chitosan scaffolds, due to their physicochemical properties, stimulate cell proliferation in different tissues of the human body. This study aimed to determine the physicochemical, mechanical, and biological properties of chitosan scaffolds crosslinked with ammonium hydroxide, with different pH values, to better understand cell behavior depending on the pH of the biomaterial. Scaffolds were either neutralized with sodium hydroxide solution, washed with distilled water until reaching a neutral pH, or kept at alkaline pH. Physicochemical characterization included scanning electron microscopy (SEM), elemental composition (EDX), Fourier-transform infrared (FTIR) spectroscopy, Raman spectroscopy, thermogravimetric analysis (TGA), and mechanical testing. In vitro cytotoxicity was assessed via dental-pulp stem cells’ (DPSCs’) biocompatibility. The results revealed that the neutralized scaffolds exhibited better cell proliferation and morphology. It was concluded that the chitosan scaffolds’ high pH (due to residual ammonium hydroxide) decreases DPSCs’ cell viability.
... 5 Since cellular and nuclear contents are proinflammatory, removal of cellular content would decrease recipient graft rejection. 1,2,12 The preservation of ECM components allows for retention of specific tissue composition, biologic activity, and mechanical integrity. 1 Acellular, tissue-specific ECM bio-scaffolds can generate appropriate spatiotemporal and biochemical signaling to promote tissue regeneration and host endogenous stem and progenitor cell differentiation. ...
Article
Full-text available
Vascularized composite allotransplantation is becoming the emerging standard for reconstructive surgery treatment for patients with limb trauma and facial injuries involving soft tissue loss. Due to the complex immunogenicity of composite grafts, patients who undergo vascularized composite allotransplantation are reliant on lifelong immunosuppressive therapy. Decellularization of donor grafts to create an extracellular matrix bio-scaffold provides an immunomodulatory graft that preserves the structural and bioactive function of the extracellular matrix. Retention of extracellular matrix proteins, growth factors, and signaling cascades allow for cell adhesion, migration, proliferation, and tissue regeneration. Perfusion decellularization of detergents through the graft vasculature allows for increased regent access to all tissue layers, and removal of cellular debris through the venous system. Grafts can subsequently be repopulated with appropriate cells through the vasculature to facilitate tissue regeneration. The present work reviews methods of decellularization, process parameters, evaluation of adequate cellular and nuclear removal, successful applications of perfusion decellularization for use in vascularized composite allotransplantation, and current limitations.
Article
Poly (L-lactic acid) microparticles (PLLAms) loaded with growth factors were prepared through the modified (ethanol as the modifier) solution enhanced dispersion by supercritical CO2 (SEDS) for controlling release rate while maintaining bioactivity. Trypsin was used to explore operational parameters and their influence on release. Results showed that the optimized flow rate of polymer solution was 1.0 mL/min. Release rate of trypsin-PLLAms was regulated by molecular weight of PLLA and Polyethylene glycol (PEG) ratio. Bone morphogenetic protein (BMP-2), fibroblast growth factor-2 (FGF-2), and vascular endothelial growth factor (VEGF) was loaded into PLLAms. Encapsulation efficiency was 65.9%, 63.9%, and 64.3%, cumulative release rate was 63.2%, 74.8% and 84.1% for 7 days, respectively. BMP-2, VEGF and FGF-2 released from PLLAms significantly promoted cell proliferation and differentiation. Therefore, the modified SEDS effectively maintained drug bioactivity, which could be used to fabricate carriers for controlled bioactive drugs release.
Article
Rupture of tendons and ligaments (T/L) is a major clinical challenge due to T/L possess anisotropic mechanical properties and hierarchical structures. Here, to imitate these characteristics, an approach is presented by fabricating hybrid nanofibrous composites. First, hybrid fiber‐reinforced yarns are fabricated via successively electrospinning poly(L‑lactide‑co‑ε‑caprolactone) (PLCL) and gelatin (Ge) nanofibers onto polyethylene terephthalate (PET) fibers to improve biodurability and biocompatibility. Then, by comparing different manufacturing methods, the knitted structure succeeds in simulating anisotropic mechanical properties, even being stronger than natural ligaments, and possessing comfort compliance superior to clinically used ligament advanced reinforcement system (LARS) ligament. Moreover, after inoculation with tendon‐derived stem cells and transplantation in vivo, hybrid nanofibrous composites are integrated with native tendons to guide surrounding tissue ingrowth due to the highly interconnected and porous structure. The knitted hybrid nanofibrous composites are also ligamentized and remodeled in vivo to promote tendon regeneration. Specifically, after the use of optimized anisotropic hybrid nanofibrous composites to repair tendon, the deposition of tendon‐associated extracellular matrix proteins is more significant. Thus, this study indicates a strategy of manufacturing anisotropic hybrid nanofibrous composites with superior mechanical properties and good histocompatibility for clinical reconstruction.
Article
Full-text available
Injury from the severe burn is exacerbated by a persistent inflammatory response. This response is mediated by cytokines and chemokines, which are released from various immune cells, including mast cells. In this study, the ability of the acellular ovine small intestine submucosa (AOSIS) to load and release of Mineral Pitch (MP) was first investigated, and it was found that the preparation of the scaffold by a modified method enables it to load and release water-soluble drugs. Then, 32 male Wistar rats were divided into four groups, a third-degree burn was created, and except for the control group, the others were treated with: AOSIS, WJ-MSCs seeded AOSIS, or AOSIS loaded with WJ-MSCs and MP. Wound sampling on the 5th day after treatment showed that the number of intact and degranulated mast cells in the treatment groups was associated with a decrease compared to the control group. In the last group, this decrease was the largest (and statically significant (p < 0.05)). Also, by measuring the level of inflammatory factors in blood serum, it was found that in the treatment groups compared to the control group, IL-10 was associated with an increase, and TNF-α was associated with a decrease. The changes in inflammatory factors were more significant (p < 0.05) in the last group. So, our results indicate that AOSIS loaded with WJ-MSCs and MP could be used as an innovative tissue-engineered device to control inflammatory condition during burn wound healing.
Article
Full-text available
Amniotic membrane has been widely applied as a biological graft in both medical and veterinary practice. In ophthalmology, epidermal growth factor (EGF) in human amniotic membrane (HAM) promotes corneal epithelial cell proliferation and migration, thus it facilitates corneal wound healing. In dogs, with limited cryopreserved HAM availability, different cold glycerol preserving protocols have been developed for the storage canine amniotic membrane (CAM). This study aimed to study protein expression of EGF in CAM preserved with different concentrations of glycerol and storage temperatures, using enzyme-linked immunosorbent assay. CAM preserved in 50% glycerol and 99.5% glycerol and kept at 4 and − 20 °C for 7–30 days were compared. We found that preserving membrane with 50% glycerol at − 20 °C has significantly higher EGF protein expression compared with that at 4 °C (p < 0.05). There was a trend that the storage in 50% glycerol achieved higher EGF protein expression than 99.5% glycerol at both 4 °C and − 20 °C. In conclusion, 50% glycerol at − 20 °C was the best condition to preserve CAM in our study. Therefore, there is likely an alternative method to maintain level of EGF protein expression in preserved CAM.
Article
Full-text available
The purpose of the current study was to compare the effects of drying and fresh-freezing on human amniotic membrane (HAM) and amnion/chorion membrane (HACM) in terms of histological and structural characteristics and cytokine levels. HAM and HACM samples, obtained from six placentae, were investigated. HAM and HACM were dried, electron beam-irradiated (dehydration group; d-HAM/d-HACM), or fresh-frozen (freezing group; f-HAM/f-HACM). Luminex assay was used to assay the levels of 15 cytokines. The ultrastructural characteristics of HAM and HACM were evaluated using light and transmission electron microscopies. Total cytokine contents did not show the statistical difference between dehydration and fresh-freezing process. Significantly higher levels of total cytokines were observed in HACM than in HAM. Epidermal growth factor (EGF) level was significantly higher in d-HAM than in the other samples. The levels of most of the other growth factors were higher in HACM than in HAM, but there was no statistical difference between the dehydration process and the fresh-freezing process. The levels of the cytokines, other than the growth factors, were higher in HACM than in HAM, and higher concentrations of cytokines were observed in the freezing group than in the dehydration group. Histological examination revealed that the dehydration group had thinner tissues than the freezing group, but the structural stability, including the basement membrane, did not differ between the two groups. Microscopic structures such as microvilli and nuclei were well-preserved in the freezing group, based on the results of the transmission electron microscopy. Our dehydration process maintained the histological structure of HAM/HACM and a variety of growth factors and cytokines were identified. Especially, the HAM, processed with the dehydration method, had a higher EGF level than that processed with the fresh-freezing method. Therefore, dehydration method can be used to effectively promote wound repair.
Article
Full-text available
Currently available bioprosthetic heart valves have been successfully used clinically; however, they have several limitations. Alternatively, tissue-engineering techniques can be used. However, there are limited data concerning the impact of storage conditions of scaffolds on their biomechanics and morphology. The aim of this study was to determine the effect of different storage conditions on the biomechanics and morphology of pulmonary valve dedicated for the acellular scaffold preparation to achieve optimal conditions to obtain stable heart valve prostheses. Scaffold can then be used for the construction of tissue-engineered heart valve, for this reason evaluation of these parameters can determine the success of the clinical application this type of bioprosthesis. Pulmonary heart valves were collected from adult porcines. Materials were divided into five groups depending on the storage conditions. Biomechanical tests were performed, both the static tensile test, and examination of viscoelastic properties. Extracellular matrix morphology was evaluated using transmission electron microscopy and immunohistochemistry. Tissue stored at 4 °C exhibited a higher modulus of elasticity than the control (native) and fresh acellular, which indicated the stiffening of the tissue and changes of the viscoelastic properties. Such changes were not observed in the radial direction. Percent strain was not significantly different in the study groups. The storage conditions affected the acellularization efficiency and tissue morphology. To the best of our knowledge, this study is the first that attributes the mechanical properties of pulmonary valve tissue to the biomechanical changes in the collagen network due to different storage conditions. Storage conditions of scaffolds for tissue-engineered heart valves may have a significant impact on the haemodynamic and clinical effects of the used bioprostheses.
Article
Full-text available
Background: Recent data suggest an increased risk for infection when acellular dermal matrix is used in breast reconstruction. This may be because some acellular dermal matrices are actually not terminally sterilized but are instead "aseptically processed." This study evaluates aseptic and sterile matrices for evidence of bacterial contamination and whether or not terminal sterilization affects matrix collagen architecture and stem cell ingrowth. Methods: Five separate samples of 14 different matrices were analyzed by fluorescent in situ hybridization using a bacterial DNA probe to detect bacterial DNA on the matrices. Separate samples were incubated for bacteria, acid-fast bacilli, and fungi for 2 to 6 weeks to detect living organisms. The impact of terminal sterilization on the collagen network and stem cell ingrowth on the matrices was then assessed. Results: Traces of bacterial DNA were encountered on all matrices, with more bacteria in the aseptic group compared with the sterile group (3.4 versus 1.6; p = 0.003). The number of positive cultures was the same between groups (3.8 percent). Electron microscopy demonstrated decreased collagen organization in the sterile group. Stem cell seeding on the matrices displayed a wide variation of cellular ingrowth between matrices, with no difference between aseptic and sterile groups (p = 0.2). Conclusions: Although there was more evidence of prior bacterial contamination on aseptically processed matrices compared with sterile matrices; clinical cultures did not differ between groups. Terminal sterilization does not appear to affect stem cell ingrowth but may come at the cost of damaging the collagen network. Clinical question/level of evidence: Therapeutic, V.
Article
Full-text available
Oesophageal tissue engineering is a therapeutic alternative when oesophageal replacement is required. Decellularised scaffolds are ideal as they are derived from tissue-specific extracellular matrix and are non-immunogenic. However, appropriate preservation may significantly affect scaffold behaviour. Here we aim to prove that an effective method for short- and long-term preservation can be applied to tissue engineered products allowing their translation to clinical application. Rabbit oesophagi were decellularised using the detergent-enzymatic treatment (DET), a combination of deionised water, sodium deoxycholate and DNase-I. Samples were stored in phosphate-buffered saline solution at 4°C (4°C) or slow cooled in medium with 10% Me2SO at -1°C/min followed by storage in liquid nitrogen (SCM). Structural and functional analyses were performed prior to and after 2 and 4 weeks and 3 and 6 months of storage under each condition. Efficient decellularisation was achieved after 2 cycles of DET as determined with histology and DNA quantification, with preservation of the ECM. Only the SCM method, commonly used for cell storage, maintained the architecture and biomechanical properties of the scaffold up to 6 months. On the contrary, 4°C method was effective for short-term storage but led to a progressive distortion and degradation of the tissue architecture at the following time points. Efficient storage allows a timely use of decellularised oesophagi, essential for clinical translation. Here we describe that slow cooling with cryoprotectant solution in liquid nitrogen vapour leads to reliable long-term storage of decellularised oesophageal scaffolds for tissue engineering purposes.
Article
Full-text available
To improve the safe use of allograft bone, decellularization techniques may be utilized to produce acellular scaffolds. Such scaffolds should retain their innate biological and biomechanical capacity and support mesenchymal stem cell (MSC) osteogenic differentiation. However, as allograft bone is derived from a wide age-range, this study aimed to determine whether donor age impacts on the ability an osteoinductive, acellular scaffold produced from human bone to promote the osteogenic differentiation of bone marrow MSCs (BM-MSC). BM-MSCs from young and old donors were seeded on acellular bone cubes from young and old donors undergoing osteoarthritis related hip surgery. All combinations resulted in increased osteogenic gene expression, and alkaline phosphatase (ALP) enzyme activity, however BM-MSCs cultured on old donor bone displayed the largest increases. BM-MSCs cultured in old donor bone conditioned media also displayed higher osteogenic gene expression and ALP activity than those exposed to young donor bone conditioned media. ELISA and Luminex analysis of conditioned media demonstrated similar levels of bioactive factors between age groups; however, IGF binding protein 1 (IGFBP1) concentration was significantly higher in young donor samples. Additionally, structural analysis of old donor bone indicated an increased porosity compared to young donor bone. These results demonstrate the ability of a decellularized scaffold produced from young and old donors to support osteogenic differentiation of cells from young and old donors. Significantly, the older donor bone produced greater osteogenic differentiation which may be related to reduced IGFBP1 bioavailability and increased porosity, potentially explaining the excellent clinical results seen with the use of allograft from aged donors.
Article
When implanted as a biomaterial for tissue replacement, selected submucosal layers of porcine small intestine induce site‐specific tissue remodeling. Small intestinal submucosa (SIS), as isolated, is primarily an acellular extracellular matrix material. In an attempt to discover the components of small intestinal submucosa which are able to induce this tissue remodeling, the material was extracted and extracts were tested for the ability to stimulate Swiss 3T3 fibroblasts to synthesize DNA and proliferate. Each of the four different extracts of small intestinal submucosa had measurable cell‐stimulating activity when analyzed in both a whole cell proliferation assay (alamarBlue dye reduction) and a DNA synthesis assay ([³H]‐thymidine incorporation). Proteins extracted from SIS with 2 M urea induced activity profiles in the two assays which were very similar to the activity profiles of basic fibroblast growth factor (FGF‐2) in the assays. As well, the changes in cell morphology in response to the extracted proteins mimicked the changes induced by FGF‐2. Neutralization experiments with specific antibodies to this growth factor confirmed the presence of FGF‐2 and indicated that it was responsible for 60% of the fibroblast‐stimulating activity of the urea extract of small intestinal submucosa. Western blot analysis with a monoclonal antibody specific for FGF‐2 detected a reactive doublet at approximately 19 kDa and further confirmed the presence of FGF‐2. Cell stimulating activity of proteins extracted from SIS with 4 M guanidine was neutralized by an antibody specific for transforming growth factor β (TGFβ). Changes in the morphology of the fibroblasts exposed to this extract were nearly identical to changes induced by TGFβ. Although no reactive protein band was detected at 25 kDa in nonreduced western blot analysis, several bands were reactive at higher molecular weight. The identity of this TGFβ‐related component of small intestinal submucosa is unknown. Identification of FGF‐2 and TGFβ‐related activities in SIS, two growth factors known to significantly affect critical processes of tissue development and differentiation, provides the opportunity to further elucidate the mechanisms by which this extracellular matrix biomaterial modulates wound healing and tissue remodeling. J. Cell. Biochem. 67:478–491, 1997. © 1997 Wiley‐Liss, Inc.
Article
Engineering of functional vascularized liver tissues hold great promise in addressing donor organ shortage for transplantation. Whole organ decellularization is a cell removal method that retains the native vascular structures of the organ such that it can be anastomosed with the recipient circulation after recellularization with healthy cells. However, a main hurdle to successful implantation of bioengineered organ is the inability to efficiently re-endothelialize the vasculature with a functional endothelium, resulting in blood clotting which is the primary cause of failure in early transplant studies. Here, we present an efficient approach for enhancing re-endothelialization of decellularized rat liver scaffolds by conjugating the REDV cell-binding domain to improve attachment of endothelial cells (EC) on vascular wall surfaces. In order to facilitate expression and purification of the peptide, REDV was fused with elastin-like peptide (ELP) that confers thermally triggered aggregation behavior to the fusion protein. After validating the adhesive properties of the REDV-ELP peptide, we covalently coupled REDV-ELP to the blood vasculature of decellularized rat livers and seeded EC using perfusion of the portal vein. We showed that REDV-ELP increased cell attachment, spreading and proliferation of EC within the construct resulting in uniform endothelial lining of the scaffold vasculature. We further observed that REDV-ELP conjugation dramatically reduced platelet adhesion and activation. Altogether, our results demonstrate that this method allowed functional re-endothelialization of liver scaffold and show great potential toward the generation of functional bioengineered liver for long-term transplantation. Statement of significance: There is a critical need for novel organ replacement therapies as the grafts for transplantation fall short of demand. Recent advances in tissue engineering, through the use of decellularized scaffolds, have opened the possibility that engineered grafts could be used as substitutes for donor livers. However, successful implantation has been challenged by the inability to create a functional vasculature. Our research study reports a new strategy to increase efficiency of endothelialization by increasing the affinity of the vascular matrix for endothelial cells. We functionalized decellularized liver scaffold using elastin-like peptides grafted with REDV cell binding domain. We showed that REDV-ELP conjugation improve endothelial cell attachment and proliferation within the scaffold, demonstrating the feasibility of re-endothelializing a whole liver vasculature using our technique.
Article
The regional injection of connective tissue growth factor (CTGF) for diabetic wound healing requires multiple components and results in a substantial loss of its biological activity. Acellular dermal matrix (ADM) scaffolds are optimal candidates for delivering these factors to local ischemic environments. In this study, we explored whether CTGF loaded on ADM scaffolds can enhance fibronectin (FN) expression to accelerate diabetic wound healing via the Protein kinase C (PKC) signaling pathway. The performance of CTGF and CTGF+PKC inhibitor, which were loaded on ADM scaffolds to treat dorsal skin wounds in streptozotocin (STZ)-induced diabetic mice, were evaluated with naked ADM as a control. Wound closure showed that ADM scaffolds loaded with CTGF induced greater diabetic wound healing in the early stage of the wound in diabetic mice. Moreover, ADM scaffolds loaded with CTGF obviously increased the expression of FN both at the mRNA and protein levels, while the expression of FN was significantly reduced in the inhibitor group. Furthermore, the ADM+CTGF group, which produce FN, obviously promoted α-SMA and TGF-ß expression and enhanced neovasculature and collagen synthesis at the wound sites. ADM scaffolds loaded with CTGF+PKC inhibitor delayed diabetic wound healing, indicating that FN expression was mediated by the PKC signaling pathway. Our findings offer new perspectives for the treatment of diabetic wound healing and suggest a rationale for the clinical evaluation of CTGF use in diabetic wound healing.
Article
Extracellular matrix-based scaffolds derived from mammalian tissues have been used in tissue engineering applications. Among all the tissues, decellularized small intestine submucosal layer (SIS) has been recently investigated for its exceptional characteristics and biocompatibilities. These investigations have been mainly focused on the decellularized porcine SIS; however, there has not been any report on ovine SIS (OSIS) layer. In this study, OSIS was decellularized and its physical, chemical, and morphological properties were evaluated. Decellularization was carried out using chemical reagents and various physical conditions. The effects of different conditions were evaluated on histological and biomechanical properties, quality of residual DNA, GAPDH gene expression, and biocompatibility. Results revealed satisfactory decellularization of OSIS which could be due to its thin thickness. Mechanical properties, structural form, and glycosaminoglycan contents were preserved in all the decellularized groups. In SDS-treated groups, further cells and DNA residues were removed compared to the groups treated with Triton X-100 only. No toxicity was observed in all treatments, and viability, expansion, and cell proliferation were supported. In conclusion, our results suggest that OSIS decellularized scaffold could be considered as an appropriate biological scaffold for tissue engineering applications. © 2017 Wiley Periodicals, Inc. J Biomed Mater Res Part B: Appl Biomater, 2017.