Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 303
Filter
1.
Cell Biochem Funct ; 42(4): e4038, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38736214

ABSTRACT

The generation of insulin-producing cells (IPCs) is an attractive approach for replacing damaged ß cells in diabetic patients. In the present work, we introduced a hybrid platform of decellularized amniotic membrane (dAM) and fibrin encapsulation for differentiating adipose tissue-derived stem cells (ASCs) into IPCs. ASCs were isolated from healthy donors and characterized. Human AM was decellularized, and its morphology, DNA, collagen, glycosaminoglycan (GAG) contents, and biocompatibility were evaluated. ASCs were subjected to four IPC differentiation methods, and the most efficient method was selected for the experiment. ASCs were seeded onto dAM, alone or encapsulated in fibrin gel with various thrombin concentrations, and differentiated into IPCs according to a method applying serum-free media containing 2-mercaptoethanol, nicotinamide, and exendin-4. PDX-1, GLUT-2 and insulin expression were evaluated in differentiated cells using real-time PCR. Structural integrity and collagen and GAG contents of AM were preserved after decellularization, while DNA content was minimized. Cultivating ASCs on dAM augmented their attachment, proliferation, and viability and enhanced the expression of PDX-1, GLUT-2, and insulin in differentiated cells. Encapsulating ASCs in fibrin gel containing 2 mg/ml fibrinogen and 10 units/ml thrombin increased their differentiation into IPCs. dAM and fibrin gel synergistically enhanced the differentiation of ASCs into IPCs, which could be considered an appropriate strategy for replacing damaged ß cells.


Subject(s)
Adipose Tissue , Cell Differentiation , Fibrin , Insulin , Stem Cells , Humans , Cell Differentiation/drug effects , Fibrin/chemistry , Fibrin/metabolism , Adipose Tissue/cytology , Adipose Tissue/metabolism , Stem Cells/metabolism , Stem Cells/cytology , Insulin/metabolism , Cells, Cultured , Insulin-Secreting Cells/metabolism , Insulin-Secreting Cells/cytology , Decellularized Extracellular Matrix/chemistry , Decellularized Extracellular Matrix/metabolism , Decellularized Extracellular Matrix/pharmacology , Amnion/cytology , Amnion/metabolism , Amnion/chemistry
2.
J Biomater Appl ; 39(1): 24-39, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38616137

ABSTRACT

The reparative properties of amniotic membrane allografts are well-suited for a broad spectrum of specialties. Further enhancement of their utility can be achieved by designing to the needs of each application through the development of novel processing techniques and tissue configurations. As such, this study evaluated the material characteristics and biological properties of two PURION® processed amniotic membrane products, a lyophilized human amnion, intermediate layer, and chorion membrane (LHACM) and a dehydrated human amnion, chorion membrane (DHACM). LHACM is thicker; therefore, its handling properties are ideal for deep, soft tissue deficits; whereas DHACM is more similar to a film-like overlay and may be used for shallow defects or surgical on-lays. Characterization of the similarities and differences between LHACM and DHACM was conducted through a series of in vitro and in vivo studies relevant to the healing cascade. Compositional analysis was performed through histological staining along with assessment of barrier membrane properties through equilibrium dialysis. In vitro cellular response was assessed in fibroblasts and endothelial cells using cell proliferation, migration, and metabolic assays. The in vivo cellular response was assessed in an athymic nude mouse subcutaneous implantation model. The results indicated the PURION® process preserved the native membrane structure, nonviable cells and collagen distributed in the individual layers of both products. Although, LHACM is thicker than DHACM, a similar composition of growth factors, cytokines, chemokines and proteases is retained and consequently elicit comparable in vitro and in vivo cellular responses. In culture, both treatments behaved as potent mitogens, chemoattractants and stimulants, which translated to the promotion of cellular infiltration, neocollagen deposition and angiogenesis in a murine model. PURION® processed LHACM and DHACM differ in physical properties but possess similar in vitro and in vivo activities highlighting the impact of processing method on the versatility of clinical use of amniotic membrane allografts.


Subject(s)
Allografts , Amnion , Chorion , Mice, Nude , Chorion/cytology , Amnion/chemistry , Animals , Humans , Mice , Wound Healing , Cell Proliferation , Biocompatible Materials/chemistry , Biocompatible Materials/metabolism , Female , Fibroblasts/cytology , Fibroblasts/metabolism , Materials Testing , Cell Movement
3.
Biotechnol Lett ; 46(3): 469-481, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38368285

ABSTRACT

PURPOSE: Based on the clinical need for grafts for vascular tissue regeneration, our group developed a customizable scaffold derived from the human amniotic membrane. Our approach consists of rolling the decellularized amniotic membrane around a mandrel to form a multilayered tubular scaffold with tunable diameter and wall thickness. Herein, we aimed to investigate if silica nanoparticles (SiNP) could enhance the adhesion of the amnion layers within these rolled grafts. METHODS: To test this, we assessed the structural integrity and mechanical properties of SiNP-treated scaffolds. Mechanical tests were repeated after six months to evaluate adhesion stability in aqueous environments. RESULTS: Our results showed that the rolled SiNP-treated scaffolds maintained their tubular shape upon hydration, while non-treated scaffolds collapsed. By scanning electron microscopy, SiNP-treated scaffolds presented more densely packed layers than untreated controls. Mechanical analysis showed that SiNP treatment increased the scaffold's tensile strength up to tenfold in relation to non-treated controls and changed the mechanism of failure from interfacial slipping to single-point fracture. The nanoparticles reinforced the scaffolds both at the interface between two distinct layers and within each layer of the extracellular matrix. Finally, SiNP-treated scaffolds significantly increased the suture pullout force in comparison to untreated controls. CONCLUSION: Our study demonstrated that SiNP prevents the unraveling of a multilayered extracellular matrix graft while improving the scaffolds' overall mechanical properties. In addition to the generation of a robust biomaterial for vascular tissue regeneration, this novel layering technology is a promising strategy for a number of bioengineering applications.


Subject(s)
Extracellular Matrix , Nanoparticles , Silicon Dioxide , Tissue Scaffolds , Silicon Dioxide/chemistry , Tissue Scaffolds/chemistry , Nanoparticles/chemistry , Humans , Extracellular Matrix/chemistry , Tissue Engineering/methods , Amnion/chemistry , Regeneration/drug effects , Tensile Strength
4.
J Biomed Mater Res A ; 112(7): 1041-1056, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38380793

ABSTRACT

Extracellular matrix-based bio-scaffolds are useful for tissue engineering as they retain the unique structural, mechanical, and physiological microenvironment of the tissue thus facilitating cellular attachment and matrix activities. However, considering its potential, a comprehensive understanding of the protein profile remains elusive. Herein, we evaluate the impact of decellularization on the human amniotic membrane (hAM) based on its proteome profile, physicochemical features, as well as the attachment, viability, and proliferation of umbilical cord-derived mesenchymal stem cells (hUC-MSC). Proteome profiles of decellularized hAM (D-hAM) were compared with hAM, and gene ontology (GO) enrichment analysis was performed. Proteomic data revealed that D-hAM retained a total of 249 proteins, predominantly comprised of extracellular matrix proteins including collagens (collagen I, collagen IV, collagen VI, collagen VII, and collagen XII), proteoglycans (biglycan, decorin, lumican, mimecan, and versican), glycoproteins (dermatopontin, fibrinogen, fibrillin, laminin, and vitronectin), and growth factors including transforming growth factor beta (TGF-ß) and fibroblast growth factor (FGF) while eliminated most of the intracellular proteins. Scanning electron microscopy was used to analyze the epithelial and basal surfaces of D-hAM. The D-hAM displayed variability in fibril morphology and porosity as compared with hAM, showing loosely packed collagen fibers and prominent large pore areas on the basal side of D-hAM. Both sides of D-hAM supported the growth and proliferation of hUC-MSC. Comparative investigations, however, demonstrated that the basal side of D-hAM displayed higher hUC-MSC proliferation than the epithelial side. These findings highlight the importance of understanding the micro-environmental differences between the two sides of D-hAM while optimizing cell-based therapeutic applications.


Subject(s)
Amnion , Mesenchymal Stem Cells , Proteome , Umbilical Cord , Humans , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/metabolism , Amnion/cytology , Amnion/chemistry , Amnion/metabolism , Umbilical Cord/cytology , Proteome/analysis , Cell Proliferation , Decellularized Extracellular Matrix/chemistry , Biocompatible Materials/chemistry
5.
Cell Tissue Bank ; 25(1): 305-323, 2024 Mar.
Article in English | MEDLINE | ID: mdl-37840108

ABSTRACT

Amniotic membrane (AM), the innermost layer of the placenta, is an exceptionally effective biomaterial with divers applications in clinical medicine. It possesses various biological functions, including scar reduction, anti-inflammatory properties, support for epithelialization, as well as anti-microbial, anti-fibrotic and angio-modulatory effects. Furthermore, its abundant availability, cost-effectiveness, and ethical acceptability make it a compelling biomaterial in the field of medicine. Given the potential unavailability of fresh tissue when needed, the preservation of AM is crucial to ensure a readily accessible and continuous supply for clinical use. However, preserving the properties of AM presents a significant challenge. Therefore, the establishment of standardized protocols for the collection and preservation of AM is vital to ensure optimal tissue quality and enhance patient safety. Various preservation methods, such as cryopreservation, lyophilization, and air-drying, have been employed over the years. However, identifying a preservation method that effectively safeguards AM properties remains an ongoing endeavor. This article aims to review and discuss different sterilization and preservation procedures for AM, as well as their impacts on its histological, physical, and biochemical characteristics.


Subject(s)
Amnion , Cryopreservation , Pregnancy , Female , Humans , Amnion/chemistry , Cryopreservation/methods , Freeze Drying/methods , Placenta , Biocompatible Materials/pharmacology
6.
J Biomater Appl ; 37(8): 1341-1354, 2023 03.
Article in English | MEDLINE | ID: mdl-36331116

ABSTRACT

The developing fetus is wrapped by a human amniotic membrane or amnion. Amnion is a promising human tissue allograft in clinical application because of its chemical composition, collagen-based, and mechanical properties of the extracellular matrix. In addition, amnion contains cells and growth factors; therefore, meets the essential parameters of tissue engineering. No donor morbidity, easy processing and storage, fewer ethical issue, anti-inflammatory, antioxidant, antibacterial, and non-immunogenic properties are other advantages of amnion usage. For these reasons, amnion can resolve some bottlenecks in the regenerative medicine issues such as tissue engineering and cell therapy. Over the last decades, biomedical applications of amnion have evolved from a simple sheet for skin or cornea repair to high-technology applications such as amnion nanocomposite, powder, or hydrogel for the regeneration of cartilage, muscle, tendon, and heart. Furthermore, amnion has anticancer as well as drug/cell delivery capacity. This review highlights various ancient and new applications of amnion in research and clinical applications, from regenerative medicine to cancer therapy, focusing on articles published during the last decade that also revealed information regarding amnion-based products. Challenges and future perspectives of the amnion in regenerative medicine are also discussed.


Subject(s)
Amnion , Regenerative Medicine , Humans , Amnion/chemistry , Tissue Engineering , Skin
7.
J Endod ; 48(10): 1285-1293.e2, 2022 Oct.
Article in English | MEDLINE | ID: mdl-35850299

ABSTRACT

INTRODUCTION: During regenerative endodontic procedures, the microenvironment of the canal is formed by the degree of disinfection and release of ions from the applied materials onto the top surface. This study aimed to characterize the effects of amnion-chorion membrane and collagen membrane on pulp-dentin regeneration compared to calcium silicate cements (CSCs), focusing on cell migration, mineralization potential, anti-inflammation, and angiogenesis. METHODS: Two CSCs and 2 membranes were used: ProRoot MTA (Dentsply, Tulsa, OK, USA), RetroMTA (BioMTA, Seoul, Korea), Collagen Membrane (Genoss, Suwon, Korea), and BioXclude (amnion-chorion membrane; Snoasis Medical, Colorado, USA). Transwell and scratch assays were used to evaluate cell migration and wound healing. Mineralization potential was evaluated using alkaline phosphatase activity, Alizarin red S staining, and quantitative real-time polymerase chain reaction for the expression of marker genes. Quantitative real-time polymerase chain reaction was used to measure the levels of angiogenic genes and inflammatory mediators. An endothelial tube formation assay was used to assess angiogenesis. RESULTS: The membranes showed superior migration and wound healing compared with CSCs. Except for RetroMTA, ProRoot MTA and the 2 membranes showed high alkaline phosphatase activity and mineralization nodule formation and upregulated mRNA expression of markers for mineralization. Membranes upregulated the mRNA of angiogenesis genes and increased the capillary tube formation of endothelial cells compared to CSCs. Furthermore, the membrane matrix decreased the expression of inflammatory genes. CONCLUSIONS: Collagen membrane and Amnion-chorion membrane showed prominent cell migration, angiogenesis, and healing effects against inflammation, as well as comparable mineralization potential compared to CSCs, recommending the use of membrane as a matrix.


Subject(s)
Regenerative Endodontics , Alkaline Phosphatase/metabolism , Amnion/chemistry , Amnion/metabolism , Calcium Compounds/pharmacology , Chorion , Collagen/pharmacology , Dental Pulp , Endothelial Cells/metabolism , Inflammation Mediators , RNA, Messenger , Silicates/pharmacology
8.
Mol Biol Rep ; 49(8): 7731-7741, 2022 Aug.
Article in English | MEDLINE | ID: mdl-35716291

ABSTRACT

BACKGROUND: The human Amniotic Membrane (hAM) has been studied as a potential therapeutic option in cancer, namely in hepatocellular carcinoma. Previously, our research group evaluated the effect of human Amniotic Membrane Protein Extracts (hAMPE) in cancer therapy, demonstrating that hAMPE inhibit the metabolic activity of human hepatocellular carcinoma cell lines: Hep3B2.1-7, HepG2 and Huh7. Therefore, and considering the close relationship between metabolic activity and oxidative stress, the aim of this study was to evaluate the effect of hAMPE treatment in glucose metabolism and its role in oxidative stress of hepatocellular carcinoma. METHODS AND RESULTS: Glucose uptake and lactate production was assessed by 1 H-NMR, and the expression of several mediators of the glycolytic pathway was evaluated by Western blot or fluorescence. Total antioxidant capacity (TAC) and biomarkers of oxidative stress effects in proteins were detected. Our results showed that hAMPE treatment increased glucose consumption on Hep3B2.1-7, HepG2, and Huh7 through the increase of GLUT1 in Hep3B2.1-7 and Huh7, and GLUT3 in HepG2 cells. It was observed an increased expression of 6-phosphofrutokinase (PFK-1L) in all cell lines though glucose was not converted to lactate on HepG2 and Huh7 cells, suggesting that hAMPE treatment may counteract the Warburg effect observed in carcinogenesis. In Hep3B2.1-7, hAMPE treatment induced an increase in expression of lactate dehydrogenase (LDH) and monocarboxylate transporter isoform 4 (MCT4). We further detected that hAMPE enhances the TAC of culture media after 2 and 8 h. This was followed by a degree of protection against proteins nitration and carbonylation. CONCLUSIONS: Overall, this work highlights the potential usefulness of hAMPE as anticancer therapy through the modulation of the glycolytic and oxidative profile in human hepatocellular carcinoma.


Subject(s)
Carcinoma, Hepatocellular , Liver Neoplasms , Amnion/chemistry , Amnion/metabolism , Biomarkers/metabolism , Carcinoma, Hepatocellular/metabolism , Cell Line, Tumor , Glucose/metabolism , Glycolysis , Humans , Lactic Acid/metabolism , Liver Neoplasms/metabolism , Oxidative Stress
9.
J Mater Sci Mater Med ; 33(3): 32, 2022 Mar 10.
Article in English | MEDLINE | ID: mdl-35267104

ABSTRACT

Amniotic membrane (AM) is a biological tissue that surrounds the fetus in the mother's womb. It has pluripotent cells, immune modulators, collagen, cytokines with anti-fibrotic and anti-inflammatory effect, matrix proteins, and growth factors. In spite of the biological characteristics, some results have been released in preventing the adhesion on traumatized surfaces. Application of the AM as a scaffold is limited due to its low biomechanical resistance and rapid biodegradation. Therefore, for using the AM during surgery, its modification by different methods such as cross-linking of the membrane collagen is necessary, because the cross-linking is an effective way to reduce the rate of biodegradation of the biological materials. In addition, their cross-linking is likely an efficient way to increase the tensile properties of the material, so that they can be easily handled or sutured. In this regard, various methods related to cross-linking of the AM subsuming the composite materials, physical cross-linking, and chemical cross-linking with the glutraldehyde, carbodiimide, genipin, aluminum sulfate, etc. are reviewed along with its advantages and disadvantages in the current work.


Subject(s)
Amnion , Carbodiimides , Amnion/chemistry , Biocompatible Materials/chemistry , Carbodiimides/chemistry , Collagen/chemistry , Cross-Linking Reagents/chemistry , Tissue Engineering/methods , Tissue Scaffolds/chemistry
10.
Proc Natl Acad Sci U S A ; 119(4)2022 01 25.
Article in English | MEDLINE | ID: mdl-35046053

ABSTRACT

Current treatment strategies for osteoarthritis (OA) predominantly address symptoms with limited disease-modifying potential. There is a growing interest in the use of adipose-derived stem cells (ADSCs) for OA treatment and developing biomimetic injectable hydrogels as cell delivery systems. Biomimetic injectable hydrogels can simulate the native tissue microenvironment by providing appropriate biological and chemical cues for tissue regeneration. A biomimetic injectable hydrogel using amnion membrane (AM) was developed which can self-assemble in situ and retain the stem cells at the target site. In the present study, we evaluated the efficacy of intraarticular injections of AM hydrogels with and without ADSCs in reducing inflammation and cartilage degeneration in a collagenase-induced OA rat model. A week after the induction of OA, rats were treated with control (phosphate-buffered saline), ADSCs, AM gel, and AM-ADSCs. Inflammation and cartilage regeneration was evaluated by joint swelling, analysis of serum by cytokine profiling and Raman spectroscopy, gross appearance, and histology. Both AM and ADSC possess antiinflammatory and chondroprotective properties to target the sites of inflammation in an osteoarthritic joint, thereby reducing the inflammation-mediated damage to the articular cartilage. The present study demonstrated the potential of AM hydrogel to foster cartilage tissue regeneration, a comparable regenerative effect of AM hydrogel and ADSCs, and the synergistic antiinflammatory and chondroprotective effects of AM and ADSC to regenerate cartilage tissue in a rat OA model.


Subject(s)
Adipose Tissue/cytology , Amnion , Hydrogels , Osteoarthritis/therapy , Stem Cell Transplantation , Stem Cells/metabolism , Amnion/chemistry , Animals , Cell Differentiation , Cells, Cultured , Chromatography, Liquid , Cytokines/metabolism , Hydrogels/chemistry , Immunohistochemistry , Injections, Intra-Articular , Mass Spectrometry , Osteoarthritis/etiology , Osteoarthritis/pathology , Rats , Spectrum Analysis, Raman , Stem Cell Transplantation/methods , Stem Cells/cytology , Treatment Outcome
11.
Tissue Cell ; 74: 101693, 2022 Feb.
Article in English | MEDLINE | ID: mdl-34856451

ABSTRACT

As a promising approach in tissue engineering, decellularization has become one of the mostly-studied research areas in tissue engineering thanks to its potential to bring about several advantages over synthetic materials since it can provide a 3-dimensional ECM structure with matching biomechanical properties of the target tissue. Amniotic membranes are the tissues that nurture the embryos during labor. Similarly, these materials have also been proposed for tissue regeneration in several applications. The main drawback in using amniotic membranes is the limited thickness of these materials since most tissues require a 3D matrix for an enhance regeneration. In order to prevent this limitation, here we report a facile fabrication methodology for multilayered amniotic membrane-based tissue constructs. The amniotic membranes of Wistar albino rats were first decellularized with the physical and chemical methods and utilized as scaffolds. Secondly, the prepared decellularized membranes were sutured to form a multilayered 3D structure. Within the study, 7 groups including control (PBS), were prepared based on physical and chemical decellularization methods. UV exposure and freezing techniques were used as a physical decellularization methods while hypertonic medium and SDS (sodium dodecyl sulfate) protocols were used as chemical decellularization methods. The combinations of both protocols were also used. In groups, A was the control and group B was applied just UV. In group C was applied UV and freezing. In addition to UV and freezing, in group D was applied hypertonic solution while group E was applied SDS (0.03 %). In group F was applied UV, freezing, hypertonic solution and SDS (0.03 %). In group G was applied UV, hypertonic solution, SDS (0.03 %) and freezing, respectively. Based on the histological and quantitative analyses, F and G groups were found as the most efficient decellularization protocols in rat amniotic membranes. Then, group F and G decellularized amniotic membranes were used to form scaffolds and thus-formed matrices were further characterized in vitro cell culture studies and mechanical tests. Cytotoxicity analyses performed using MTT showed a good cell viability in F and G groups scaffolds. The percentage viability rate was higher in G group (81.3 %) compared to F (75.33 %) and also cell viability in G group was found more meaningful according to p value which was obtained 0.007. Cellular adhesions after in vitro cell culture and morphology of scaffolds were evaluated by scanning electron microscopy (SEM). It was observed that the cells cultivated in equal amounts of tissue scaffolds were higher in the F compared to that observed in group G. The mechanical testing with 40 N force revealed 0.77 mm displacement in group F while it was 0.75 mm in group G. Moreover, according to force-controlled test, 2.9 mm displacement of F group and 1.2 mm displacement of G group was measured. As a result, this study shows that the multilayered decellularized amniotic membrane scaffolds support cell survival and adhesion and can form a flexible biomaterial with desired handling properties.


Subject(s)
Amnion/chemistry , Tissue Engineering , Tissue Scaffolds/chemistry , Animals , Cell Line , Female , Mice , Rats , Rats, Wistar
12.
Carbohydr Polym ; 276: 118752, 2022 Jan 15.
Article in English | MEDLINE | ID: mdl-34823781

ABSTRACT

Hydrogels have long been used for encapsulating stem cell-derived conditioned mediums to achieve skin regeneration after wounding. However, inappropriate mechanical strength, low adhesion and low elasticity limit their clinical application. To address these challenges, we engineered a hyaluronic acid-based hydrogel grafted with methacrylic anhydride and N-(2-aminoethyl)-4-[4-(hydroxymethyl)-2-methoxy-5-nitrophenoxy]-butanamide (NB) groups to encapsulate a lyophilized amnion-derived conditioned medium (AM-CM). This hydrogel can photopolymerize in situ within 3 s by photo-initiated free-radical crosslinking between methacrylate moieties. Meanwhile, the formed o-nitrosobenzaldehyde groups by photo-irradiation could covalently bond with the amino groups of tissue surface, which allowed strong tissue adhesion. Furthermore, the hydrogel possessed excellent mechanical properties, high elasticity, favorable biocompatibility and prolonged AM-CM release. Our further vitro and in vivo studies showed that the hydrogel significantly accelerated diabetic wound healing by regulating macrophage polarization and promoting angiogenesis. The engineered hydrogel with AM-CM release has high potential to treat chronic wounds in clinics.


Subject(s)
Amnion/chemistry , Diabetes Mellitus/drug therapy , Hyaluronic Acid/pharmacology , Hydrogels/pharmacology , Wound Healing/drug effects , Adhesives , Anhydrides/chemistry , Animals , Cell Proliferation/drug effects , Culture Media, Conditioned , Diabetes Mellitus/metabolism , Human Umbilical Vein Endothelial Cells , Humans , Hyaluronic Acid/chemistry , Hydrogels/chemistry , Macrophages/drug effects , Male , Mice , Mice, Inbred C57BL , Rheology/methods , Skin/drug effects , Stem Cells/metabolism , Tissue Adhesions/metabolism
13.
Sci Rep ; 11(1): 22508, 2021 11 18.
Article in English | MEDLINE | ID: mdl-34795361

ABSTRACT

Decellularized and de-epithelialized placenta membranes have widely been used as scaffolds and grafts in tissue engineering and regenerative medicine. Exceptional pro-angiogenic and biomechanical properties and low immunogenicity have made the amniochorionic membrane a unique substrate which provides an enriched niche for cellular growth. Herein, an optimized combination of enzymatic solutions (based on streptokinase) with mechanical scrapping is used to remove the amniotic epithelium and chorion trophoblastic layer, which resulted in exposing the basement membranes of both sides without their separation and subsequent damages to the in-between spongy layer. Biomechanical and biodegradability properties, endothelial proliferation capacity, and in vivo pro-angiogenic capabilities of the substrate were also evaluated. Histological staining, immunohistochemistry (IHC) staining for collagen IV, and scanning electron microscope demonstrated that the underlying amniotic and chorionic basement membranes remained intact while the epithelial and trophoblastic layers were entirely removed without considerable damage to basement membranes. The biomechanical evaluation showed that the scaffold is suturable. Proliferation assay, real-time polymerase chain reaction for endothelial adhesion molecules, and IHC demonstrated that both side basement membranes could support the growth of endothelial cells without altering endothelial characteristics. The dorsal skinfold chamber animal model indicated that both side basement membranes could promote angiogenesis. This bi-sided substrate with two exposed surfaces for cultivating various cells would have potential applications in the skin, cardiac, vascularized composite allografts, and microvascular tissue engineering.


Subject(s)
Basement Membrane/metabolism , Cell Culture Techniques/methods , Endothelial Cells/metabolism , Placenta/metabolism , Trophoblasts/metabolism , Amnion/chemistry , Animals , Antigens, CD/biosynthesis , Biomechanical Phenomena , Cadherins/biosynthesis , Cell Proliferation , Female , Human Umbilical Vein Endothelial Cells , Humans , Immunohistochemistry , Male , Microcirculation , Neovascularization, Pathologic , Platelet Endothelial Cell Adhesion Molecule-1/biosynthesis , Pregnancy , Rats , Regenerative Medicine/methods , Time Factors , Tissue Engineering/methods , Tissue Scaffolds/chemistry , Vascular Endothelial Growth Factor A
14.
Biomed Mater ; 16(6)2021 09 23.
Article in English | MEDLINE | ID: mdl-34492639

ABSTRACT

Due to its excellent biocompatibility and anti-inflammatory activity, amniotic membrane (AM) has attracted much attention from scholars. However, its clinical application in vascular reconstruction was limited for poor processability, rapid biodegradation, and insufficient hemocompatibility. A naturally extracted substance with good cytocompatibility, phytic acid (PA), which can quickly form strong and stable hydrogen bonds on the tissue surface, was used to crosslink decellularized AM (DAM) to prepare a novel vascular replacement material. The results showed that PA-fixed AM had excellent mechanical strength and resistance to enzymatic degradation as well as appropriate surface hydrophilicity. Among all samples, 2% PA-fixed specimen showed excellent human umbilical vein endothelial cells (HUVECs)-cytocompatibility and hemocompatibility. It could also stimulate the secretion of vascular endothelial growth factor and endothelin-1 from seeded HUVECs, indicating that PA might promote neovascularization after implantation of PA-fixed specimens. Also, 2% PA-fixed specimen could inhibit the secretion of tumor necrosis factor-αfrom co-cultured macrophages, thus might reduce the inflammatory response after sample implantation. Finally, the results ofex vivoblood test andin vivoexperiments confirmed our deduction that PA might promote neovascularization after implantation. All the results indicated that prepared PA-fixed DAM could be considered as a promising small-diameter vascular replacement material.


Subject(s)
Amnion , Anti-Inflammatory Agents , Decellularized Extracellular Matrix , Phytic Acid , Amnion/chemistry , Amnion/cytology , Animals , Anti-Inflammatory Agents/chemistry , Anti-Inflammatory Agents/pharmacology , Blood Vessels/metabolism , Cell Survival/drug effects , Cells, Cultured , Decellularized Extracellular Matrix/chemistry , Decellularized Extracellular Matrix/pharmacology , Decellularized Extracellular Matrix/toxicity , Female , Human Umbilical Vein Endothelial Cells , Humans , Materials Testing , Phytic Acid/chemistry , Phytic Acid/pharmacology , Rabbits , Rats , Vascular Endothelial Growth Factor A/metabolism
15.
Sci Rep ; 11(1): 13754, 2021 07 02.
Article in English | MEDLINE | ID: mdl-34215804

ABSTRACT

Diseases and injuries that compromise the ocular surface cause considerable patient distress and have long term consequences for their quality of life. Treatment modalities that can address the delicate balance of tissue regeneration, inflammation and maintenance of corneal transparency are therefore needed. We have recently formulated two novel eye drops from placental tissues: cord blood platelet lysate (CBED) and amniotic membrane extract eye drops (AMED), which can be used to treat severe ocular disorders. Here we characterise these two preparations by measuring: (a) growth factors (GF) and cytokines composition, (b) promotion of human corneal epithelial cell (HCEC) growth and (c) effects on immune cells in a lymphocyte culture assay. Finally, their bioavailability was assayed in an ex vivo porcine corneal model. We show that both preparations contain GF and cytokines that were able to promote the in vitro growth of HCEC and support repair in an in vitro scratch test. When assessed in a lymphocyte culture, both favoured immune suppression reducing the cellular expression of NKG2D and CD107a as well as the production of interferon gamma (IFN-γ) in natural killer, NKT and T cells. Regarding bioavailability, CBED active molecules were found mainly in the pre-corneal fraction with some penetration into the corneal fraction, in an ex vivo model. In summary, both placental-derived allogeneic preparations, CBED and AMED, display regenerative and immunomodulatory capabilities. These results will help define mechanisms of action and the best indications and doses of each product for use in a particular patient and support the development of off-the-shelf therapies for ocular surface pathologies in which wound healing defects and inflammatory events are contributing factors.


Subject(s)
Cornea/drug effects , Corneal Diseases/drug therapy , Ophthalmic Solutions/pharmacology , Regeneration/drug effects , Amnion/chemistry , Animals , Blood Platelets/immunology , Cell Proliferation/drug effects , Cornea/growth & development , Corneal Diseases/pathology , Epithelial Cells/drug effects , Female , Fetal Blood/chemistry , Fetal Blood/immunology , Humans , Intercellular Signaling Peptides and Proteins/genetics , Intercellular Signaling Peptides and Proteins/pharmacology , Lymphocytes/drug effects , Lymphocytes/immunology , Ophthalmic Solutions/chemistry , Placenta/chemistry , Placenta/immunology , Pregnancy , Quality of Life , Swine , Wound Healing
16.
Genes (Basel) ; 12(5)2021 05 11.
Article in English | MEDLINE | ID: mdl-34064633

ABSTRACT

Junctional epidermolysis bullosa (JEB) is a clinically and genetically heterogeneous skin fragility disorder frequently caused by mutations in genes encoding the epithelial laminin isoform, laminin-332. JEB patients also present mucosal involvement, including painful corneal lesions. Recurrent corneal abrasions may lead to corneal opacities and visual impairment. Current treatments are merely supportive. We report a novel JEB phenotype distinguished by the complete resolution of skin fragility in infancy and persistent ocular involvement with unremitting and painful corneal abrasions. Biallelic LAMB3 mutations c.3052-5C>G and c.3492_3493delCG were identified as the molecular basis for this phenotype, with one mutation being a hypomorphic splice variant that allows residual wild-type laminin-332 production. The reduced laminin-332 level was associated with impaired keratinocyte adhesion. Then, we also investigated the therapeutic power of a human amniotic membrane (AM) eyedrop preparation for corneal lesions. AM were isolated from placenta donors, according to a procedure preserving the AM biological characteristics as a tissue, and confirmed to contain laminin-332. We found that AM eyedrop preparation could restore keratinocyte adhesion in an in vitro assay. Of note, AM eyedrop administration to the patient resulted in long-lasting remission of her ocular manifestations. Our findings suggest that AM eyedrops could represent an effective, non-invasive, simple-to-handle treatment for corneal lesions in patients with JEB and possibly other EB forms.


Subject(s)
Corneal Dystrophies, Hereditary/genetics , Epidermolysis Bullosa, Junctional/genetics , Epithelium, Corneal/pathology , Ophthalmic Solutions/therapeutic use , Phenotype , Amnion/chemistry , Cell Adhesion , Cell Adhesion Molecules/genetics , Cells, Cultured , Child, Preschool , Corneal Dystrophies, Hereditary/drug therapy , Corneal Dystrophies, Hereditary/pathology , Epidermolysis Bullosa, Junctional/drug therapy , Epidermolysis Bullosa, Junctional/pathology , Female , Humans , Keratinocytes/drug effects , Keratinocytes/pathology , Keratinocytes/physiology , Mutation , Ophthalmic Solutions/chemistry , Ophthalmic Solutions/pharmacology , Skin/pathology , Kalinin
17.
Biomed Res Int ; 2021: 5597758, 2021.
Article in English | MEDLINE | ID: mdl-34124249

ABSTRACT

BACKGROUND: Investigating the viability and proliferative rates of fibroblast cells on human amniotic membrane (HAM) as a scaffold will be an important subject for further research. The aim of this study was to assess the fibroblast viability seeded on acellular HAM, since foreskin neonatal allogenic fibroblasts seeded on HAM accelerate the wound healing process. METHODS: Fibroblasts were retrieved from the foreskin of a genetically healthy male infant, and we exploited AM of healthy term neonates to prepare the amniotic scaffold for fibroblast transfer. After cell culture, preparation of acellular HAM, and seeding of cells on HAM based on the protocol, different methods including 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT), 4',6-Diamidino-2-phenylindole dihydrochloride (DAPI), and propidium iodide (PI) staining were employed for assessment of fibroblast viability on HAM. RESULTS: Based on the results obtained from the DAPI and PI staining, the percentage of viable cells in the former staining was clearly higher than that of the dead cells in the latter one. The results of DAPI and PI staining were in accordance with the findings of MTT assay, confirming that fibroblasts were viable and even proliferate on HAM. CONCLUSION: Our findings showed the viability of fibroblasts seeded on the acellular HAM using MTT assay, DAPI, and PI staining; however, this study had some limitations. It would be an interesting subject for future research to compare the viability and proliferation rate of fibroblasts seeded on both cellular and acellular HAM.


Subject(s)
Amnion/chemistry , Cell Culture Techniques , Fibroblasts/metabolism , Tissue Scaffolds/chemistry , Cell Survival , Fibroblasts/cytology , Humans , Male
18.
Biol Reprod ; 105(2): 439-448, 2021 08 03.
Article in English | MEDLINE | ID: mdl-34057176

ABSTRACT

Serotonin or 5-hydroxytryptamine (5-HT) is a biogenic amine involved in regulating several functions, including development. However, its impact on human embryo development has been poorly studied. The present work investigated the expression and distribution of the main components of the serotoninergic system in human amniotic tissue and human amniotic epithelial cells (hAEC) in vitro, as an alternative model of early human embryo development. Amniotic membranes from full-term healthy pregnancies were used. Human amnion tissue or hAEC isolated from the amnion was processed for reverse transcription-polymerase chain reaction and immunofluorescence analyses of the main components of the serotoninergic system. We found the expression of tryptophan hydroxylase type 1 (TPH1), type 2 (TPH2), serotonin transporter (SERT), monoamine oxidase-A (MAOA), as well as HTR1D and HTR7 receptors at mRNA level in amnion tissue as well in hAEC. Interestingly, we found the presence of 5-HT in the nucleus of the cells in amnion tissue, whereas it was located in the cytoplasm of isolated hAEC. We detected TPH1, TPH2, and HTR1D receptor in both the nucleus and cytoplasm. SERT, MAOA, and HTR7 receptor were only observed in the cytoplasm. The results presented herein show, for the first time, the presence of the serotoninergic system in human amnion in vivo and in vitro.


Subject(s)
Amnion/metabolism , Epithelial Cells/metabolism , Serotonin/metabolism , Amnion/chemistry , Humans
19.
J Biomater Appl ; 35(6): 592-601, 2021 01.
Article in English | MEDLINE | ID: mdl-32873118

ABSTRACT

Amniotic membrane (AM) has been shown to enhance corneal wound healing due to the abundance of growth factors, cytokines, and extracellular matrix (ECM) proteins inherent to the tissue. As such, AM has garnered widespread clinical utility as a biological dressing for a number of ophthalmic and soft tissue applications. The preparation, sterilization, and storage procedures used to manufacture AM grafts are extremely important for the conservation of inherent biological components within the membrane. Current processing techniques use harsh chemicals and sterilization agents that can compromise the fundamental wound healing properties of AM. Furthermore, commercially available cryopreserved AM products require specific storage conditions (e.g., ultra-low freezers) thereby limiting their clinical availability in austere environments. Supercritical carbon dioxide (SCCO2) technology allows for the sterilization of biological tissues without the resulting degradation of integral ECM proteins and other factors often seen with current tissue sterilization processes. With this study we demonstrate that lyophilized AM, sterilized using SCCO2, maintains similar biochemical properties and biocompatibility as that of commercially available AM products requiring specialized cold storage conditions.


Subject(s)
Allografts/chemistry , Amnion/chemistry , Biocompatible Materials/chemistry , Carbon Dioxide/chemistry , Freeze Drying/methods , Allografts/metabolism , Amnion/metabolism , Animals , Bandages , Biocompatible Materials/metabolism , Collodion/chemistry , Cornea/metabolism , Cytokines/metabolism , Extracellular Matrix/metabolism , Humans , Intercellular Signaling Peptides and Proteins/metabolism , Male , Rabbits , Sterilization , Wound Healing/drug effects
20.
Int J Biol Macromol ; 169: 492-499, 2021 Feb 01.
Article in English | MEDLINE | ID: mdl-33358948

ABSTRACT

Corneal transplantation, by which the damaged cornea is replaced by a new one, suffers from limited access to HLA-compatible-donors and high maintenance and surgical costs. Therefore, artificial corneas are considered as alternative tools with promising prospects. In our previous study, a two-part-polymeric artificial cornea was composed of enhanced hydrophilic surface electrospun poly(ε-caprolactone) nanofibrous scaffold that is thermally connected to a polyvinyl alcohol-based hydrogel disk was prepared. Characterization tests revealed the prepared artificial cornea had similar biocompatible and structural characteristics regarding the natural cornea. In current study, human amniotic membrane extract containing growth factors, cytokines, anti-inflammatory factors, and anti-angiogenic factors was prepared, nano-encapsulated in chitosan-dextran nanoparticles, and physically decorated on the poly(ε-caprolactone)-polyvinyl-alcohol artificial cornea. Physicochemical and biological characterizations revealed the nano-decorated artificial cornea has more biocompatibility than the unmodified one. Our study demonstrated the bioactive macromolecules loaded on chitosan-dextran nanoparticles enhanced the anti-angiogenic property of artificial cornea through the sustained release of anti-angiogenic factors such as thrombospondin-1, endostatin, and heparin sulfate proteoglycan. Real-time-PCR and flow-cytometry assessments elucidated the vascularization was inhibited through a decrease in the expression of cluster of differentiation 31 and von-Willebrand-Factor. Our study proposed the use of biocompatible artificial cornea could be a promising strategy in corneal transplantation.


Subject(s)
Amnion/chemistry , Chitosan/chemistry , Dextrans/chemistry , Amnion/drug effects , Amnion/metabolism , Biocompatible Materials/chemistry , Cornea/drug effects , Corneal Transplantation/methods , Humans , Materials Testing , Nanoparticles/chemistry , Polymers/chemistry , Polyvinyl Alcohol/chemistry , Tissue Engineering/methods , Tissue Scaffolds/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL
...