Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.450
Filter
1.
Biomaterials ; 312: 122741, 2025 Jan.
Article in English | MEDLINE | ID: mdl-39121727

ABSTRACT

Last twenties, tissue engineering has rapidly advanced to address the shortage of organ donors. Decellularization techniques have been developed to mitigate immune rejection and alloresponse in transplantation. However, a clear definition of effective decellularization remains elusive. This study compares various decellularization protocols using the human fascia lata model. Morphological, structural and cytotoxicity/viability analyses indicated that all the five tested protocols were equivalent and met Crapo's criteria for successful decellularization. Interestingly, only the in vivo immunization test on rats revealed differences. Only one protocol exhibited Human Leucocyte Antigen (HLA) content below 1% residual threshold, the only criterion preventing rat immunization with an absence of rat anti-human IgG switch after one month (N=4 donors for each of the 7 groups, added by negative and positive controls, n=28). By respecting a refined set of criteria, i.e. lack of visible nuclear material, <50ng DNA/mg dry weight of extracellular matrix, and <1% residual HLA content, the potential for adverse host reactions can be drastically reduced. In conclusion, this study emphasizes the importance of considering not only nuclear components but also major histocompatibility complex in decellularization protocols and proposes new guidelines to promote safer clinical development and use of bioengineered scaffolds.


Subject(s)
Fascia Lata , HLA Antigens , Tissue Engineering , Humans , Animals , Tissue Engineering/methods , HLA Antigens/immunology , Rats , Tissue Scaffolds/chemistry , Biocompatible Materials/chemistry , Male , Decellularized Extracellular Matrix/chemistry , Extracellular Matrix/chemistry , Extracellular Matrix/metabolism
2.
Bioact Mater ; 43: 114-128, 2025 Jan.
Article in English | MEDLINE | ID: mdl-39376928

ABSTRACT

Decellularized tissues offer significant potential as biological materials for tissue regeneration given their ability to preserve the complex compositions and architecture of the native extracellular matrix (ECM). However, the evaluation and derivation of decellularized matrices from human bone tissue remains largely unexplored. We examined how the physiochemical and biological properties of ECM hydrogels derived from human bone ECM could be controlled by manipulating bone powder size (45-250 µm, 250-1000 µm, and 1000-2000 µm) and ECM composition through modulation of enzyme digestion time (3-5-7 days). A reduction in material bone powder size and an increase in ECM digestion time produced enhanced protein concentrations in the ECM hydrogels, accompanied by the presence of a diverse array of proteins and improved gelation strength. Human bone marrow-derived stromal cells (HBMSCs) cultured on ECM hydrogels from 45 to 250 µm bone powder, over 7 days, demonstrated enhanced osteogenic differentiation compared to hydrogels derived from larger bone powders and collagen gels confirming the potential of the hydrogels as biologically active materials for bone regeneration. Digestion time and bone powder size modulation enabled the generation of hydrogels with enhanced release of ECM proteins and appropriate gelation and rheological properties, offering new opportunities for application in bone repair.

3.
Int J Biol Macromol ; 281(Pt 2): 136340, 2024 Oct 05.
Article in English | MEDLINE | ID: mdl-39374728

ABSTRACT

Novel biomaterials are necessary to fabricate biomimetic scaffolds for bone tissue engineering. In the present experiment, we aimed to fabricate and evaluate the osteogenic properties of nanohydroxyapatite/chitosan/decellularized placenta (nHA.Cs.dPL) composite scaffolds. The human placenta was decellularized (dPL), characterized, and digested in pepsin to form the hydrogel. nHA.Cs.dPL scaffolds were fabricated using salt leaching/freeze drying and evaluated for their morphology, chemical composition, swelling, porosity, degradation, mechanical strength, and biocompatibility. Saos-2 cells were seeded on scaffolds, and their osteogenic properties were investigated by evaluating alkaline phosphatase (ALP), osteocalcin (OCN), collagen type 1 (COL I) expression, and calcium deposition under osteogenic differentiation. The dPL was prepared with minimized DNA content and a well-preserved porous structure. Scaffolds were highly porous with interconnected pores and exhibited appropriate swelling and degradation rates supporting saos-2 cell attachment and proliferation. dPL improved scaffold physicochemical features and increased cell proliferation, ALP, OCN, COL I expression, and calcium deposition under osteogenic differentiation induction. nHA.Cs.dPL composite scaffolds provide a 3D microenvironment with superior physicochemical features that support saos-2 cell adhesion, proliferation, and osteogenic differentiation.

4.
Sci Rep ; 14(1): 23192, 2024 10 05.
Article in English | MEDLINE | ID: mdl-39369012

ABSTRACT

Liver disease cases are rapidly expanding worldwide, and transplantation remains the only effective cure for end-stage disease. There is an increasing demand for developing potential drug treatments, and regenerative therapies using in-vitro culture platforms. Human decellularized extracellular matrix (dECM) is an appealing alternative to conventional animal tissues as it contains human-specific proteins and can serve as scaffolding materials. Herein we exploit this with human donor tissue from discarded liver which was not suitable for transplant using a synergistic approach to combining biological and topographical cues in electrospun materials as an in-vitro culture platform. To realise this, we developed a methodology for incorporating human liver dECM into electrospun polycaprolactone (PCL) fibres with surface nanotopographies (230-580 nm). The hybrid scaffolds were fabricated using varying concentrations of dECM; their morphology, mechanical properties, hydrophilicity and stability were analysed. The scaffolds were validated using HepG2 and primary mouse hepatocytes, with subsequent results indicating that the modified scaffolds-maintained cell growth and influenced cell attachment, proliferation and hepatic-related gene expression. This work demonstrates a novel approach to harvesting the potential from decellularized human tissues in the form of innovative in-vitro culture platforms for liver.


Subject(s)
Hepatocytes , Liver , Tissue Engineering , Tissue Scaffolds , Humans , Tissue Scaffolds/chemistry , Tissue Engineering/methods , Animals , Liver/metabolism , Mice , Hepatocytes/cytology , Hep G2 Cells , Extracellular Matrix/metabolism , Polyesters/chemistry , Decellularized Extracellular Matrix/chemistry , Cell Proliferation , Cellular Microenvironment , Cell Adhesion
5.
J Biomed Mater Res B Appl Biomater ; 112(10): e35489, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39377466

ABSTRACT

Creating acellular vascularized constructs from animal and plant tissue is one of the well-known strategies for scaffold assembly. Decellularization takes an important position among these strategies. The most common method is chemical decellularization. This approach employs high concentrations of detergents, primarily Triton X-100, sodium dodecyl sulfate (SDS), and sodium hypochlorite (SH). In this work, novel techniques for decellularizing spinach were developed using detergents frequently utilized in laboratories. Spinach leaves were decellularized using Tween-20, SDS, and SH at low concentrations to generate an acellular plant matrix for tissue engineering. We measured the quantities of DNA and protein, as well as the decellularization using hematoxylin and eosin (H&E) staining. The biocompatibility and capacity of the biostructures to stimulate fibroblast wound healing were assessed using MTT and the Scratch assay. The antibacterial activity of the scaffolds was also tested against a gram-positive bacterium, Staphilococcus aureus, which is a common pathogen associated with wound healing. The best shape, evident vascularization, and good biocompatibility were seen in the Tween-20 decellularized samples at 1% concentration at 21°C and 37°C through the enhancement of cell proliferation and wound healing. In terms of antibacterial activity, all scaffold samples had a significant effect on Staphilococcus aureus, where the number of bacterial colonies in all six scaffold groups became zero after 4 h of treatment. The scaffolds also showed a 100% kill rate on Staphilococcus aureus, which could avoid wound infection during the repair process, and that can be suggested as a scaffold for tissue engineering applications and an important constituent for pharmacological activities.


Subject(s)
Anti-Bacterial Agents , Spinacia oleracea , Staphylococcus aureus , Wound Healing , Wound Healing/drug effects , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/chemistry , Spinacia oleracea/chemistry , Animals , Staphylococcus aureus/drug effects , Staphylococcus aureus/growth & development , Mice , Decellularized Extracellular Matrix/chemistry , Decellularized Extracellular Matrix/pharmacology , Fibroblasts , Tissue Scaffolds/chemistry , Humans , Materials Testing
6.
Biomed Mater Eng ; 2024 Sep 26.
Article in English | MEDLINE | ID: mdl-39331089

ABSTRACT

BACKGROUND: Restoration of the abdominal wall defects due to herniation or other complications represents a challenging task of the reconstructive surgery. Synthetic grafts or crosslinked animal-derived grafts, are utilized, followed by significant adverse reactions. OBJECTIVE: This study aimed to evaluate primarily the production of a decellularized abdominal wall scaffold and secondly its biocompatibility upon transplantation in an animal model. METHODS: Full-thickness abdominal wall samples were harvested from Wistar Rats and then decellularized utilizing a three-cycle process. To evaluate the decellularization efficacy, histological, biochemical and biomechanical analyses were performed. The biocompatibility assessment involved the implantation of the produced scaffolds to Sprague Dawley rats. The grafts remained for a total period of 4 weeks, followed by immunohistochemistry for the detection of CD11b+, CD4+ and CD8+ cells. RESULTS: Histological, biochemical and biomechanical results, indicated the production of compatible acellular full-thickness abdominal wall samples. After 4 weeks of implantation, a minor presence of immunity cells was observed. CONCLUSION: The data of this study indicated the successful production of a full-thickness abdominal wall scaffold. Biologically derived full-thickness abdominal wall scaffolds may have greater potential in restoration of the abdominal wall defects, bringing them one step closer to their clinical utility.

7.
Int J Mol Sci ; 25(18)2024 Sep 20.
Article in English | MEDLINE | ID: mdl-39337615

ABSTRACT

Several lung diseases can cause structural damage, making lung transplantation the only therapeutic option for advanced disease stages. However, the transplantation success rate remains limited. Lung bioengineering using the natural extracellular matrix (ECM) of decellularized lungs is a potential alternative. The use of undifferentiated cells to seed the ECM is practical; however, sterilizing the organ for recellularization is challenging. Photobiomodulation therapy (PBMT) may offer a solution, in which the wavelength is crucial for tissue penetration. This study aimed to explore the potential of optimizing lung recellularization with mesenchymal stem cells using PBMT (660 nm) after sterilization with PBMT (880 nm). The lungs from C57BL/6 mice were decellularized using 1% SDS and sterilized using PBMT (880 nm, 100 mW, 30 s). Recellularization was performed in two groups: (1) recellularized lung and (2) recellularized lung + 660 nm PBMT (660 nm, 100 mW, 30 s). Both were seeded with mesenchymal stem cells from human tooth pulp (DPSc) and incubated for 24 h at 37 °C and 5% CO2 in bioreactor-like conditions with continuous positive airway pressure (CPAP) at 20 cmH2O and 90% O2. The culture medium was analyzed after 24 h. H&E, immunostaining, SEM, and ELISA assays were performed. Viable biological scaffolds were produced, which were free of cell DNA and preserved the glycosaminoglycans; collagens I, III, and IV; fibronectin; laminin; elastin; and the lung structure (SEM). The IL-6 and IL-8 levels were stable during the 24 h culture, but the IFN-γ levels showed significant differences in the recellularized lung and recellularized lung + 660 nm PBMT groups. Greater immunological modulation was observed in the recellularized groups regarding pro-inflammatory cytokines (IL-6, IFN-γ, and IL-8). These findings suggest that PBMT plays a role in cytokine regulation and antimicrobial activity, thus offering promise for enhanced therapeutic strategies in lung bioengineering.


Subject(s)
Cytokines , Low-Level Light Therapy , Lung , Mesenchymal Stem Cells , Mice, Inbred C57BL , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/metabolism , Animals , Mice , Lung/metabolism , Low-Level Light Therapy/methods , Humans , Cytokines/metabolism , Mesenchymal Stem Cell Transplantation/methods , Sterilization/methods , Extracellular Matrix/metabolism , Tissue Engineering/methods
8.
Mater Today Bio ; 28: 101228, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39296356

ABSTRACT

Full osteochondral regeneration remains a major clinical challenge. Among other experimental cartilage regenerative approaches, decellularized cartilage (DCC) is considered a promising material for generating potentially implantable scaffolds useful as cartilage repair strategy. In this work, we focus on screening and comparing different decellularization methods, aiming to generate DCC potentially useful in biomedical context, and therefore, with biological activity and functional properties in terms of induction of differentiation and regeneration. Data indicates that enzymatic and detergents-based decellularization methods differentially affect ECM components, and that it has consequences in further biological behavior. SDS-treated DCC powder is not useful to be further processed in 2D or 3D structures, because these structures tend to rapidly solubilize, or disaggregate, in physiologic media conditions. Conversely, Trypsin-treated DCC powders can be processed to mechanically stable 2D films and 3D solid-foam scaffolds, presumably due to partial digestion of collagens during decellularization, which would ease crosslinking at DCC during solubilization and processing. In vitro cell culture studies indicate that these structures are biocompatible and induce and potentiate chondrogenic differentiation. In vivo implantation of DCC derived 3D porous scaffolds in rabbit osteochondral defects induce subchondral bone regeneration and fibrocartilage tissue formation after implantation. Therefore, this work defines an optimal cartilage tissue decellularization protocol able to generate DCC powders processable to biocompatible and bioactive 2D and 3D structures. These structures are useful for in vitro cartilage research and in vivo subchondral bone regeneration, while hyaline cartilage regeneration with DCC alone as implantable material remains elusive.

9.
Front Pharmacol ; 15: 1435359, 2024.
Article in English | MEDLINE | ID: mdl-39286627

ABSTRACT

The excessive deposition and cross-linking of core matrisome components typically result in abnormal remodeling of the extracellular matrix (ECM), leading to increased liver stiffness and worsening liver fibrosis. Exploring the biochemical properties of the ECM scaffold can deepen our understanding of the pathological mechanisms driving liver fibrosis and potentially facilitate the identification of therapeutic targets. While traditional sodium dodecyl sulfate (SDS)-based liver decellularization followed by proteomics can uncover the matrisome components within the ECM scaffold, it lacks the ability to reveal physicochemical characteristics like solubility. In our present study, using adult mouse liver as an example, we introduced a novel two-step workflow that combines our previously enhanced SDS (ESDS) decellularization with the conventional SDS method, enabling the identification of matrisome members with mild and/or high solubilities. Through this approach, we visualized the atlas of the mildly and highly insoluble matrisome contents in the adult mouse liver, as well as the regulatory network of highly insoluble matrisome that largely governs liver stiffness. Given the strong correlation between increased matrisome insolubility and heightened ECM stiffness, we believe that this methodology holds promise for future research focused on liver stiffness.

10.
Stem Cell Rev Rep ; 2024 Sep 11.
Article in English | MEDLINE | ID: mdl-39259445

ABSTRACT

BACKGROUND: Derivation of hepatocytes from stem cells has been established through various protocols involving growth factor (GF) and small molecule (SM) agents, among others. However, mesenchymal stem cell-based derivation of hepatocytes still remains expensive due to the use of a cocktail of growth factors, and a long duration of differentiation is needed, thus limiting its potential clinical application. METHODS: In this study, we developed a chemically defined differentiation strategy that is exclusively based on SM and takes 14 days, while the GF-based protocol requires 23-28 days. RESULTS: We optimized a stage-specific differentiation protocol for the differentiation of rat bone marrow-derived mesenchymal stem cells (MSCs) into functional hepatocyte-like cells (dHeps) that involved four stages, i.e., definitive endoderm (DE), hepatic competence (HC), hepatic specification (HS) and hepatic differentiation and growth. We further generated hepatic tissue using human decellularized liver extracellular matrix and compared it with hepatic tissue derived from the growth factor-based protocol at the transcriptional level. dHep, upon transplantation in a rat model of acute liver injury (ALI), was capable of ameliorating liver injury in rats and improving liver function and tissue damage compared to those in the ALI model. CONCLUSIONS: In summary, this is the first study in which hepatocytes and hepatic tissue were derived from MSCs utilizing a stage-specific strategy by exclusively using SM as a differentiation factor.

11.
Regen Biomater ; 11: rbae098, 2024.
Article in English | MEDLINE | ID: mdl-39224131

ABSTRACT

Coronary artery bypass grafting is acknowledged as a major clinical approach for treatment of severe coronary artery atherosclerotic heart disease. This procedure typically requires autologous small-diameter vascular grafts. However, the limited availability of the donor vessels and associated trauma during tissue harvest underscore the necessity for artificial arterial alternatives. Herein, decellularized bovine intercostal arteries were successfully fabricated with lengths ranging from 15 to 30 cm, which also closely match the inner diameters of human coronary arteries. These decellularized arterial grafts exhibited great promise following poly(2-methacryloyloxyethyl phosphorylcholine) (PMPC) grafting from the inner surface. Such surface modification endowed the decellularized arteries with superior mechanical strength, enhanced anticoagulant properties and improved biocompatibility, compared to the decellularized bovine intercostal arteries alone, or even those decellularized grafts modified with both heparin and vascular endothelial growth factor. After replacement of the carotid arteries in rabbits, all surface-modified vascular grafts have shown good patency within 30 days post-implantation. Notably, strong signal was observed after α-SMA immunofluorescence staining on the PMPC-grafted vessels, indicating significant potential for regenerating the vascular smooth muscle layer and thereby restoring full structures of the artery. Consequently, the decellularized bovine intercostal arteries surface modified by PMPC can emerge as a potent candidate for small-diameter artificial blood vessels, and have shown great promise to serve as viable substitutes of arterial autografts.

12.
Tissue Eng Part A ; 2024 Sep 25.
Article in English | MEDLINE | ID: mdl-39276095

ABSTRACT

A bioengineered liver has the potential to save patients with end-stage liver disease, and a three-dimensional decellularized scaffold is a promising approach for practical use. The main challenge in bioengineered liver transplantation is thrombogenicity during blood perfusion. We aimed to apply a novel antithrombotic polymer to revascularize liver scaffolds and evaluate the thrombogenicity and biosafety of the polymer-treated scaffolds. A biomimetic polymer, 2-metacryloyloxyethyl phosphorylcholine (MPC) was prepared for modification of the extracellular matrix in liver scaffolds. The polymer was injected into the rat liver scaffolds' portal vein and could extensively react to the vessel walls. In an ex vivo blood perfusion experiment, we demonstrated significantly less platelet deposition in the polymer-treated scaffolds than nontreated or re-endothelialized scaffolds with human umbilical vein endothelial cells. In the heterotopic transplantation model, liver volume was better maintained in the polymer-treated groups, and platelet deposition was suppressed in these groups. Additionally, the polymer-treated liver scaffolds maintained the metabolic function of the recellularized rat primary hepatocytes during perfusion culture. The MPC polymer treatment efficiently suppressed thrombus formation during blood perfusion in liver scaffolds and maintained the function of recellularized hepatocytes. Revascularizing liver scaffolds using this polymer is a promising approach for bioengineered liver transplantation.

13.
Mol Cell Biochem ; 2024 Sep 10.
Article in English | MEDLINE | ID: mdl-39251464

ABSTRACT

Scaffolds used in tissue engineering can be obtained from synthetic or natural materials, always focusing the effort on mimicking the extracellular matrix of human native tissue. In this study, a decellularization process is used to obtain an acellular, biocompatible non-cytotoxic human pericardium graft as a bio-substitute. An enzymatic and hypertonic method was used to decellularize the pericardium. Histological analyses were performed to determine the absence of cells and ensure the integrity of the extracellular matrix (ECM). In order to measure the effect of the decellularization process on the tissue's biological and mechanical properties, residual genetic content and ECM biomolecules (collagen, elastin, and glycosaminoglycan) were quantified and the tissue's tensile strength was tested. Preservation of the biomolecules, a residual genetic content below 50 ng/mg dry tissue, and maintenance of the histological structure provided evidence for the efficacy of the decellularization process, while preserving the ECM. Moreover, the acellular tissue retains its mechanical properties, as shown by the biomechanical tests. Our group has shown that the acellular pericardial matrix obtained through the super-fast decellularization protocol developed recently retains the desired biomechanical and structural properties, suggesting that it is suitable for a broad range of clinical indications.

14.
MethodsX ; 13: 102919, 2024 Dec.
Article in English | MEDLINE | ID: mdl-39280757

ABSTRACT

In recent years, minimally invasive biopsy techniques have been widely used to generate small tissue samples that require processing in clinical pathology. However, small paraffin-embedded tissues are prone to loss due to their small size. To prevent the loss of small tissues, researchers have employed nonbiological embedding materials for preembedding, but this approach can lead to cumbersome experimental procedures and increase the chances of tissue loss. This study aimed to develop a convenient decellularized embedding material derived from biological membrane tissues to effectively protect small tissues from loss during paraffin embedding. This study decellularized three types of fresh animal-derived membrane tissues and selected the small intestine as the most suitable decellularized raw material through attempts at softening, comparing physical properties, and using tissue as the starting material. Subsequently, small tissues from various tissue sources were embedded, followed by H&E staining, Masson staining, immunofluorescence staining, and immunohistochemical staining. The decellularized material derived from biomembrane tissues (DMBT) developed in this study can reduce the loss of small tissues without the need for preembedding, thereby shortening the embedding process. This provides a new pathological embedding tool for future laboratory and clinical research and work.•The fat layer of the pig's small intestine is scraped off, and chemical reagents are used to defat and decellularize it.•Chemical reagents are used to soften and make the pig's small intestine transparent, and the decellularized pig's small intestine is dried.•DMBT is used for embedding and staining the biological tissue.

15.
FASEB J ; 38(16): e70004, 2024 Aug 31.
Article in English | MEDLINE | ID: mdl-39190010

ABSTRACT

Previous studies showed that the bladder extracellular matrix (B-ECM) could increase the differentiation efficiency of mesenchymal cells into smooth muscle cells (SMC). This study investigates the potential of human amniotic membrane-derived hydrogel (HAM-hydrogel) as an alternative to xenogeneic B-ECM for the myogenic differentiation of the rabbit adipose tissue-derived MSC (AD-MSC). Decellularized human amniotic membrane (HAM) and sheep urinary bladder (SUB) were utilized to create pre-gel solutions for hydrogel formation. Rabbit AD-MSCs were cultured on SUB-hydrogel or HAM-hydrogel-coated plates supplemented with differentiation media containing myogenic growth factors (PDGF-BB and TGF-ß1). An uncoated plate served as the control. After 2 weeks, real-time qPCR, immunocytochemistry, flow cytometry, and western blot were employed to assess the expression of SMC-specific markers (MHC and α-SMA) at both protein and mRNA levels. Our decellularization protocol efficiently removed cell nuclei from the bladder and amniotic tissues, preserving key ECM components (collagen, mucopolysaccharides, and elastin) within the hydrogels. Compared to the control, the hydrogel-coated groups exhibited significantly upregulated expression of SMC markers (p ≤ .05). These findings suggest HAM-hydrogel as a promising xenogeneic-free alternative for bladder tissue engineering, potentially overcoming limitations associated with ethical concerns and contamination risks of xenogeneic materials.


Subject(s)
Amnion , Cell Differentiation , Hydrogels , Mesenchymal Stem Cells , Myocytes, Smooth Muscle , Animals , Amnion/cytology , Amnion/metabolism , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/metabolism , Myocytes, Smooth Muscle/cytology , Myocytes, Smooth Muscle/metabolism , Rabbits , Humans , Hydrogels/chemistry , Urinary Bladder/cytology , Urinary Bladder/metabolism , Extracellular Matrix/metabolism , Sheep , Cells, Cultured , Tissue Engineering/methods
16.
Curr Issues Mol Biol ; 46(8): 9179-9214, 2024 Aug 22.
Article in English | MEDLINE | ID: mdl-39194760

ABSTRACT

The recellularization of tissues after decellularization is a relatively new technology in the field of tissue engineering (TE). Decellularization involves removing cells from a tissue or organ, leaving only the extracellular matrix (ECM). This can then be recellularized with new cells to create functional tissues or organs. The first significant mention of recellularization in decellularized tissues can be traced to research conducted in the early 2000s. One of the landmark studies in this field was published in 2008 by Ott, where researchers demonstrated the recellularization of a decellularized rat heart with cardiac cells, resulting in a functional organ capable of contraction. Since then, other important studies have been published. These studies paved the way for the widespread application of recellularization in TE, demonstrating the potential of decellularized ECM to serve as a scaffold for regenerating functional tissues. Thus, although the concept of recellularization was initially explored in previous decades, these studies from the 2000s marked a major turning point in the development and practical application of the technology for the recellularization of decellularized tissues. The article reviews the historical advances and limitations in organ recellularization in TE over the last two decades.

17.
Regen Med ; 19(7-8): 421-437, 2024.
Article in English | MEDLINE | ID: mdl-39101556

ABSTRACT

The paper highlights how significant characteristics of liver can be modeled in tissue-engineered constructs using unconventional scaffolds. Hepatic lobular organization and metabolic zonation can be mimicked with decellularized plant structures with vasculature resembling a native-hepatic lobule vascular arrangement or silk blend scaffolds meticulously designed for guided cellular arrangement as hepatic patches or metabolic activities. The functionality of hepatocytes can be enhanced and maintained for long periods in naturally fibrous structures paving way for bioartificial liver development. The phase I enzymatic activity in hepatic models can be raised exploiting the microfibrillar structure of paper to allow cellular stacking creating hypoxic conditions to induce in vivo-like xenobiotic metabolism. Lastly, the paper introduces amalgamation of carbon-based nanomaterials into existing scaffolds in liver tissue engineering.


Unconventional scaffolds have the potential to meet the current challenges in liver tissue engineering- loss of hepatic morphology and functions over long-term culture, absence of native-like cell-cell and cell-matrix interactions, organization of hepatocytes into lobular structures exhibiting metabolic variations-which hinder pharmaceutical analysis, regenerative therapies and artificial organ development. Paper with cellulose microfibril network develops cellular aggregates with hypoxic conditions that influence enzymes of xenobiotic metabolism proving to be a better scaffold for hepatotoxicity testing compared with conventional monolayers in tissue culture plates. Decellularized plant stems provide already-built vasculature to be exploited for the development of intricate vessel networks that exist in hepatic lobules aiding in regenerative medicine for hepatic pathologies. Fibrous plant structures are excellent materials for the immobilization of hepatocytes and improve albumin secretion enabling their use in bioartificial liver development. Biomimicry of metabolic zonation in hepatic lobules can be achieved with perfusion culture using silk blend scaffolds with varying proportions of the liver matrix that orchestrate cellular function. The mechanical properties of silk allow the fabrication of structures that resemble liver anatomy to generate native-like hepatic lobules. Nanomaterials have immense potential as a component of composite material development for scaffolds to achieve improved predictive ability in pharmacokinetics. Most of these unconventional scaffolds have the added advantage of being readily available, accessible, affordable and sustainable for liver tissue engineering applications. Conclusively, the shift of attention away from conventional scaffolds poses a promising future in the field of tissue engineering.


Subject(s)
Liver , Nanostructures , Silk , Tissue Engineering , Tissue Scaffolds , Tissue Engineering/methods , Tissue Scaffolds/chemistry , Nanostructures/chemistry , Humans , Liver/cytology , Liver/metabolism , Silk/chemistry , Animals , Paper , Plants/metabolism , Hepatocytes/cytology , Hepatocytes/metabolism
18.
J Biomater Sci Polym Ed ; : 1-23, 2024 Aug 27.
Article in English | MEDLINE | ID: mdl-39190661

ABSTRACT

Decellularized tissue hydrogels, especially that mimic the native tissue, have a high potential for tissue engineering, three-dimensional (3D) cell culture, bioprinting, and therapeutic agent encapsulation due to their excellent biocompatibility and ability to facilitate the growth of cells. It is important to note that the decellularization process significantly affects the structural integrity and properties of the extracellular matrix, which in turn shapes the characteristics of the resulting hydrogels at the macromolecular level. Therefore, our study aims to identify an effective chemical decellularization method for sheep lung tissue, using a mixing/agitation technique with a range of detergents, including commonly [Sodium dodecyl sulfate (SDS), Triton X-100, and 3-((3-cholamidopropyl) dimethylammonio)-1-propanesulfonate] (CHAPS), and rarely used (sodium cholate hydrate, NP-40, and 3-[N,N-Dimethyl(3-myristoylaminopropyl)ammonio]propanesulfonate) (ASB-14). After the effectiveness of the used detergents on decellularization was determined by histological and biochemical methods, lung derived decellularized extracellular matrix was converted into hydrogel. We investigated the interactions between lung cells and decellularized extracellular matrix using proliferation assay, scanning electron microscopy, and immunofluorescence microscopy methods on BEAS-2B cells in air-liquid interface. Notably, this study emphasizes the effectiveness of ASB-14 in the decellularization process, showcasing its crucial role in removing cellular components while preserving vital extracellular matrix biological macromolecules, including glycosaminoglycans, collagen, and elastin. The resulting hydrogels demonstrated favorable mechanical properties and are compatible with both cell-cell and cell-extracellular matrix interactions.

19.
Colloids Surf B Biointerfaces ; 244: 114175, 2024 Dec.
Article in English | MEDLINE | ID: mdl-39216442

ABSTRACT

Decellularized extracellular matrix (dECM) hydrogels are engineered constructs that are widely-used in the field of regenerative medicine. However, the development of ECM-based hydrogels for bone tissue engineering requires enhancement in its osteogenic properties. For this purpose, we initially employed bone-derived dECM hydrogel (dECM-Hy) in combination with calcium phosphate cement (CPC) paste to improve the biological and structural properties of the dECM hydrogel. A decellularization protocol for bovine bone was developed to prepare dECM-Hy, and the mechanically-tuned dECM/CPC-Hy was built based on both rheological and mechanical characteristics. The dECM/CPC-Hy displayed a double swelling ratio and compressive strength. An interconnected structure with distinct hydroxyapatite crystals was evident in dECM/CPC-Hy. The expression levels of Alp, Runx2 and Ocn genes were upregulated in dECM/CPC-Hy compared to the dECM-Hy. A 14-day follow-up of the rats receiving subcutaneous implanted dECM-Hy, dECM/CPC-Hy and mesenchymal stem cells (MSCs)-embedded (dECM/CPC/MSCs-Hy) showed no toxicity, inflammatory factor expression or pathological changes. Radiography and computed tomography (CT) of the calvarial defects revealed new bone formation and elevated number of osteoblasts-osteocytes and osteons in dECM/CPC-Hy and dECM/CPC/MSCs-Hy compared to the control groups. These findings indicate that the dECM/CPC-Hy has substantial potential for bone tissue engineering.


Subject(s)
Bone Cements , Bone Regeneration , Calcium Phosphates , Mesenchymal Stem Cells , Animals , Calcium Phosphates/chemistry , Bone Regeneration/drug effects , Cattle , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/metabolism , Rats , Bone Cements/chemistry , Bone Cements/pharmacology , Decellularized Extracellular Matrix/chemistry , Decellularized Extracellular Matrix/pharmacology , Hydrogels/chemistry , Hydrogels/pharmacology , Osteogenesis/drug effects , Rats, Sprague-Dawley , Tissue Engineering , Core Binding Factor Alpha 1 Subunit/metabolism , Core Binding Factor Alpha 1 Subunit/genetics , Extracellular Matrix/chemistry , Extracellular Matrix/metabolism
20.
Int Dent J ; 2024 Aug 06.
Article in English | MEDLINE | ID: mdl-39112112

ABSTRACT

OBJECTIVES: This study describes a robust and versatile method for decellularization of rat submandibular glands (SMGs). METHODS: Briefly, rat SMGs were harvested and subjected to perfusion cycles using an anionic detergent. Native and decellularized SMG tissues were subjected to histological analysis using hematoxylin and eosin (H&E) stain and immunohistochemical staining using Hoescht reagent. Further, complementary DNA was synthesized using the native and decellularized SMG tissues and subjected to quantitative reverse transcription polymerase chain reaction (RT-PCR) using rat-specific genes (i.e., α-amylase [Amyl], aquaporin 5 [Aqp5], mucin 19 [Muc19] and glyceraldehyde-3-phosphate dehydrogenase [GAPDH]). The total DNA within native and decellularized SMG tissues were also quantified. RESULTS: H&E staining of SMG tissues revealed preserved ECM content. Decellularized SMG scaffolds lacked cellular material but retained collagen bundles similar to native SMGs. Hoechst reagent immunostaining showed cell nuclei and DNA present in native SMG but not in decellularized SMG scaffolds. Quantitative RT-PCR analysis showed specific amplification products of salivary gland-specific genes (Amyl, Muc19 and Aqp5) and GAPDH in the native SMG tissues. However, no amplification product was observed in the cDNAs from the decellularized SMG scaffolds, confirming the absence of DNA. Quantification of the DNA content showed that the decellularized SMG scaffolds had significantly lower DNA content than native SMG tissue. CONCLUSIONS: Results from this study demonstrated that the decellularization protocol was effective in removing cellular material while preserving the extracellular matrix components and structural integrity of the native SMG tissue.

SELECTION OF CITATIONS
SEARCH DETAIL