Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 31.396
Filter
1.
J Nanobiotechnology ; 22(1): 445, 2024 Jul 28.
Article in English | MEDLINE | ID: mdl-39069607

ABSTRACT

BACKGROUND: The incidence of osteochondral defects caused by trauma, arthritis or tumours is increasing annually, but progress has not been made in terms of treatment methods. Due to the heterogeneous structure and biological characteristics of cartilage and subchondral bone, the integration of osteochondral repair is still a challenge. RESULTS: In the present study, a novel bilayer hydrogel scaffold was designed based on anatomical characteristics to imitate superficial cartilage and subchondral bone. The scaffold showed favourable biocompatibility, and the addition of an antioxidant nanozyme (LiMn2O4) promoted reactive oxygen species (ROS) scavenging by upregulating antioxidant proteins. The cartilage layer effectively protects against chondrocyte degradation in the inflammatory microenvironment. Subchondral bionic hydrogel scaffolds promote osteogenic differentiation of rat bone marrow mesenchymal stem cells (BMSCs) by regulating the AMPK pathway in vitro. Finally, an in vivo rat preclinical osteochondral defect model confirmed that the bilayer hydrogel scaffold efficiently promoted cartilage and subchondral bone regeneration. CONCLUSIONS: In general, our biomimetic hydrogel scaffold with the ability to regulate the inflammatory microenvironment can effectively repair osteochondral defects. This strategy provides a promising method for regenerating tissues with heterogeneous structures and biological characteristics.


Subject(s)
Bone Regeneration , Hydrogels , Mesenchymal Stem Cells , Osteogenesis , Rats, Sprague-Dawley , Tissue Scaffolds , Animals , Hydrogels/chemistry , Hydrogels/pharmacology , Tissue Scaffolds/chemistry , Rats , Mesenchymal Stem Cells/drug effects , Bone Regeneration/drug effects , Osteogenesis/drug effects , Chondrocytes/drug effects , Male , Cell Differentiation/drug effects , Inflammation , Tissue Engineering/methods , Reactive Oxygen Species/metabolism , Chondrogenesis/drug effects , Cartilage/drug effects , Cartilage, Articular/drug effects , Cells, Cultured
2.
Biomolecules ; 14(7)2024 Jul 18.
Article in English | MEDLINE | ID: mdl-39062575

ABSTRACT

Congenital heart disease (CHD) is the most common birth defect, requiring invasive surgery often before a child's first birthday. Current materials used during CHD surgery lack the ability to grow, remodel, and regenerate. To solve those limitations, 3D bioprinting is an emerging tool with the capability to create tailored constructs based on patients' own imaging data with the ability to grow and remodel once implanted in children with CHD. It has the potential to integrate multiple bioinks with several cell types and biomolecules within 3D-bioprinted constructs that exhibit good structural fidelity, stability, and mechanical integrity. This review gives an overview of CHD and recent advancements in 3D bioprinting technologies with potential use in the treatment of CHD. Moreover, the selection of appropriate biomaterials based on their chemical, physical, and biological properties that are further manipulated to suit their application are also discussed. An introduction to bioink formulations composed of various biomaterials with emphasis on multiple cell types and biomolecules is briefly overviewed. Vasculogenesis and angiogenesis of prefabricated 3D-bioprinted structures and novel 4D printing technology are also summarized. Finally, we discuss several restrictions and our perspective on future directions in 3D bioprinting technologies in the treatment of CHD.


Subject(s)
Biocompatible Materials , Bioprinting , Heart Defects, Congenital , Hydrogels , Printing, Three-Dimensional , Tissue Engineering , Humans , Heart Defects, Congenital/therapy , Bioprinting/methods , Hydrogels/chemistry , Tissue Engineering/methods , Biocompatible Materials/chemistry , Biocompatible Materials/therapeutic use , Tissue Scaffolds/chemistry , Animals
3.
Gastroenterol Clin North Am ; 53(3): 461-472, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39068007

ABSTRACT

Intestinal failure manifests as an impaired capacity of the intestine to sufficiently absorb vital nutrients and electrolytes essential for growth and well-being in pediatric and adult populations. Although parenteral nutrition remains the mainstay therapeutic approach, the pursuit of a definitive and curative strategy, such as regenerative medicine, is imperative. Substantial advancements in the field of engineered intestinal tissues present a promising avenue for addressing intestinal failure; nevertheless, extensive research is still necessary for effective translation from experimental benchwork to clinical bedside applications.


Subject(s)
Intestines , Tissue Engineering , Humans , Tissue Engineering/methods , Intestines/transplantation , Intestinal Failure/therapy , Bioengineering/methods , Regenerative Medicine/methods , Tissue Scaffolds
4.
Skin Res Technol ; 30(7): e13860, 2024 Jul.
Article in English | MEDLINE | ID: mdl-39073182

ABSTRACT

BACKGROUND: The perfect repair of damaged skin has always been a constant goal for scientists; however, the repair and reconstruction of skin is still a major problem and challenge in injury and burns medicine. Human amniotic membrane (hAM), with its good mechanical properties and anti-inflammatory, antioxidant and antimicrobial benefits, containing growth factors that promote wound healing, has evolved over the last few decades from simple skin sheets to high-tech dressings, such as being made into nanocomposites, hydrogels, powders, and electrostatically spun scaffolds. This paper aims to explore the historical development, applications, trends, and research hotspots of hAM in wound healing. METHODS: We examined 2660 publications indexed in the Web of Science Core Collection (WoSCC) from January 1, 1975 to July 12, 2023. Utilizing bibliometric methods, we employed VOSviewer, CiteSpace, and R-bibliometrix to characterize general information, identify development trends, and highlight research hotspots. Subsequently, we identified a collection of high-quality English articles focusing on the roles of human amniotic epithelial stem cells (hAESCs), human amniotic mesenchymal stem cells (hAMSCs), and amniotic membrane (AM) scaffolds in regenerative medicine and tissue engineering. RESULTS: Bibliometric analysis identified Udice-French Research Universities as the most productive affiliation and Tseng S.C.G. as the most prolific author. Keyword analysis, historical direct quotations network, and thematic analysis helped us review the historical and major themes in this field. Our examination included the knowledge structure, global status, trends, and research hotspots regarding the application of hAM in wound healing. Our findings indicate that contemporary research emphasizes the preparation and application of products derived from hAM. Notably, both hAM and the cells isolated from it - hADSCs and hAESCs are prominent and promising areas of research in regenerative medicine and tissue engineering. CONCLUSION: This research delivers a comprehensive understanding of the knowledge frameworks, global dynamics, emerging patterns, and primary research foci in the realm of hAM applications for wound healing. The field is rapidly evolving, and our findings offer valuable insights for researchers. Future research outcomes are anticipated to be applied in clinical practice, enhancing methods for disease prevention, diagnosis, and treatment.


Subject(s)
Amnion , Wound Healing , Humans , Tissue Engineering/methods , Biological Dressings , Tissue Scaffolds , Epithelial Cells/physiology
5.
J Mater Sci Mater Med ; 35(1): 45, 2024 Jul 29.
Article in English | MEDLINE | ID: mdl-39073649

ABSTRACT

Electroconductive polymers are the materials of interest for the fabrication of electro-conductive tissues. Metal ions through the redox systems offer polymers with electrical conductivity. In this study, we processed a gelatin methacrylate (GelMA) network with gold nanoparticles (GNPs) through a redox system with parahydroxybenzaldehyde (PHB) or curcumin to enhance its electrical conductivity. Induction of the redox system with both PHB and curcumin into the GelMA, introduced some new functional groups into the polymeric network, as it has been confirmed by H-NMR and FTIR. These new bonds resulted in higher electro-conductivity when GNPs were added to the polymer. Higher electroactivity was achieved by PHB compared to the curcumin-induced redox system, and the addition of GNPs without redox system induction showed the lowest electroactivity. MTT was used to evaluate the biocompatibility of the resultant polymers, and the PHB-treated hydrogels showed higher proliferative effects on the cells. The findings of this study suggest that the introduction of a redox system by PHB in the GelMA network along with GNPs can contribute to the electrochemical properties of the material. This electroactivity can be advantageous for tissue engineering of electro-conductive tissues like cardiac and nervous tissues.


Subject(s)
Benzaldehydes , Biocompatible Materials , Curcumin , Electric Conductivity , Gelatin , Gold , Hydrogels , Metal Nanoparticles , Methacrylates , Tissue Engineering , Gelatin/chemistry , Gold/chemistry , Tissue Engineering/methods , Metal Nanoparticles/chemistry , Hydrogels/chemistry , Benzaldehydes/chemistry , Curcumin/chemistry , Curcumin/pharmacology , Methacrylates/chemistry , Biocompatible Materials/chemistry , Prohibitins , Spectroscopy, Fourier Transform Infrared , Materials Testing , Animals , Humans , Cell Proliferation/drug effects , Oxidation-Reduction , Tissue Scaffolds/chemistry
6.
Int J Mol Sci ; 25(14)2024 Jul 17.
Article in English | MEDLINE | ID: mdl-39063052

ABSTRACT

Nowadays, as a result of the frequent occurrence of accidental injuries and traumas such as bone damage, the number of people causing bone injuries or fractures is increasing around the world. The design and fabrication of ideal bone tissue engineering (BTE) materials have become a research hotspot in the scientific community, and thus provide a novel path for the treatment of bone diseases. Among the materials used to construct scaffolds in BTE, including metals, bioceramics, bioglasses, biomacromolecules, synthetic organic polymers, etc., natural biopolymers have more advantages against them because they can interact with cells well, causing natural polymers to be widely studied and applied in the field of BTE. In particular, alginate has the advantages of excellent biocompatibility, good biodegradability, non-immunogenicity, non-toxicity, wide sources, low price, and easy gelation, enabling itself to be widely used as a biomaterial. However, pure alginate hydrogel as a BTE scaffold material still has many shortcomings, such as insufficient mechanical properties, easy disintegration of materials in physiological environments, and lack of cell-specific recognition sites, which severely limits its clinical application in BTE. In order to overcome the defects of single alginate hydrogels, researchers prepared alginate composite hydrogels by adding one or more materials to the alginate matrix in a certain proportion to improve their bioapplicability. For this reason, this review will introduce in detail the methods for constructing alginate composite hydrogels, including alginate/polymer composite hydrogels, alginate/bioprotein or polypeptide composite hydrogels, alginate/bioceramic composite hydrogels, alginate/bioceramic composite hydrogels, and alginate/nanoclay composite hydrogels, as well as their biological application trends in BTE scaffold materials, and look forward to their future research direction. These alginate composite hydrogel scaffolds exhibit both unexceptionable mechanical and biochemical properties, which exhibit their high application value in bone tissue repair and regeneration, thus providing a theoretical basis for the development and sustainable application of alginate-based functional biomedical materials.


Subject(s)
Alginates , Biocompatible Materials , Bone and Bones , Hydrogels , Tissue Engineering , Tissue Scaffolds , Alginates/chemistry , Tissue Engineering/methods , Hydrogels/chemistry , Humans , Tissue Scaffolds/chemistry , Biocompatible Materials/chemistry , Animals , Bone Regeneration/drug effects
7.
Molecules ; 29(14)2024 Jul 10.
Article in English | MEDLINE | ID: mdl-39064841

ABSTRACT

Bone tissue engineering is a promising alternative to repair wounds caused by cellular or physical accidents that humans face daily. In this sense, the search for new graphene oxide (GO) nanofillers related to their degree of oxidation is born as an alternative bioactive component in forming new scaffolds. In the present study, three different GOs were synthesized with varying degrees of oxidation and studied chemically and tissue-wise. The oxidation degree was determined through infrared (FTIR), X-ray diffraction (XRD), X-ray photoelectron (XPS), and Raman spectroscopy (RS). The morphology of the samples was analyzed using scanning electron microscopy (SEM). The oxygen content was deeply described using the deconvolution of RS and XPS techniques. The latter represents the oxidation degree for each of the samples and the formation of new bonds promoted by the graphitization of the material. In the RS, two characteristic bands were observed according to the degree of oxidation and the degree of graphitization of the material represented in bands D and G with different relative intensities, suggesting that the samples have different crystallite sizes. This size was described using the Tuinstra-Koenig model, ranging between 18.7 and 25.1 nm. Finally, the bone neoformation observed in the cranial defects of critical size indicates that the F1 and F2 samples, besides being compatible and resorbable, acted as a bridge for bone healing through regeneration. This promoted healing by restoring bone and tissue structure without triggering a strong immune response.


Subject(s)
Bone Regeneration , Graphite , Tissue Engineering , Tissue Scaffolds , Graphite/chemistry , Bone Regeneration/drug effects , Tissue Engineering/methods , Animals , Tissue Scaffolds/chemistry , Nanostructures/chemistry , Bone and Bones/drug effects , Spectrum Analysis, Raman , Oxidation-Reduction , X-Ray Diffraction , Biocompatible Materials/chemistry , Biocompatible Materials/pharmacology , Rats , Spectroscopy, Fourier Transform Infrared
8.
Mar Drugs ; 22(7)2024 Jun 26.
Article in English | MEDLINE | ID: mdl-39057404

ABSTRACT

Recently, the need to develop a robust three-dimensional (3D) cell culture system that serves as a valuable in vitro tumor model has been emphasized. This system should closely mimic the tumor growth behaviors observed in vivo and replicate the key elements and characteristics of human tumors for the effective discovery and development of anti-tumor therapeutics. Therefore, in this study, we developed an effective 3D in vitro model of human prostate cancer (PC) using a marine collagen-based biomimetic 3D scaffold. The model displayed distinctive molecular profiles and cellular properties compared with those of the 2D PC cell culture. This was evidenced by (1) increased cell proliferation, migration, invasion, colony formation, and chemoresistance; (2) upregulated expression of crucial multidrug-resistance- and cancer-stemness-related genes; (3) heightened expression of key molecules associated with malignant progressions, such as epithelial-mesenchymal transition transcription factors, Notch, matrix metalloproteinases, and pluripotency biomarkers; (4) robust enrichment of prostate cancer stem cells (CSCs); and (5) enhanced expression of integrins. These results suggest that our 3D in vitro PC model has the potential to serve as a research platform for studying PC and prostate CSC biology, as well as for screening novel therapies targeting PC and prostate CSCs.


Subject(s)
Antineoplastic Agents , Cell Proliferation , Collagen , Neoplastic Stem Cells , Prostatic Neoplasms , Humans , Male , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/pathology , Antineoplastic Agents/pharmacology , Cell Proliferation/drug effects , Cell Line, Tumor , Neoplastic Stem Cells/drug effects , Cell Culture Techniques, Three Dimensional/methods , Animals , Cell Movement/drug effects , Tissue Scaffolds , Epithelial-Mesenchymal Transition/drug effects , Aquatic Organisms , Drug Discovery/methods
9.
Colloids Surf B Biointerfaces ; 241: 114064, 2024 Sep.
Article in English | MEDLINE | ID: mdl-38954937

ABSTRACT

Bile duct injury presents a significant clinical challenge following hepatobiliary surgery, necessitating advancements in the repair of damaged bile ducts is a persistent issue in biliary surgery. 3D printed tubular scaffolds have emerged as a promising approach for the repair of ductal tissues, yet the development of scaffolds that balance exceptional mechanical properties with biocompatibility remains an ongoing challenge. This study introduces a novel, bio-fabricated bilayer bile duct scaffold using a 3D printing technique. The scaffold comprises an inner layer of polyethylene glycol diacrylate (PEGDA) to provide high mechanical strength, and an outer layer of biocompatible, methacryloylated recombinant collagen type III (rColMA) loaded with basic fibroblast growth factor (bFGF)-encapsulated liposomes (bFGF@Lip). This design enables the controlled release of bFGF, creating an optimal environment for the growth and differentiation of bone marrow mesenchymal stem cells (BMSCs) into cholangiocyte-like cells. These cells are instrumental in the regeneration of bile duct tissues, evidenced by the pronounced expression of cholangiocyte differentiation markers CK19 and CFTR. The PEGDA//rColMA/bFGF@Lip bilayer bile duct scaffold can well simulate the bile duct structure, and the outer rColMA/bFGF@Lip hydrogel can well promote the growth and differentiation of BMSCs into bile duct epithelial cells. In vivo experiments showed that the scaffold did not cause cholestasis in the body. This new in vitro pre-differentiated active 3D printed scaffold provides new ideas for the study of bile duct tissue replacement.


Subject(s)
Bile Ducts , Cell Differentiation , Hydrogels , Mesenchymal Stem Cells , Polyethylene Glycols , Printing, Three-Dimensional , Polyethylene Glycols/chemistry , Hydrogels/chemistry , Hydrogels/pharmacology , Mesenchymal Stem Cells/drug effects , Mesenchymal Stem Cells/cytology , Animals , Cell Differentiation/drug effects , Recombinant Proteins/pharmacology , Collagen/chemistry , Tissue Scaffolds/chemistry , Mice , Fibroblast Growth Factor 2/pharmacology , Cells, Cultured , Humans , Male
10.
J Control Release ; 372: 846-861, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38955252

ABSTRACT

Bone defect is one of the urgent problems to be solved in clinics, and it is very important to construct efficient scaffold materials to facilitate bone tissue regeneration. Hydrogels, characterized by their unique three-dimensional network structure, serve as excellent biological scaffold materials. Their internal pores are capable of loading osteogenic drugs to expedite bone formation. The rate and quality of new bone formation are intimately linked with immune regulation and vascular remodeling. The strategic sequential release of drugs to balance inflammation and regulate vascular remodeling is crucial for initiating the osteogenic process. Through the design of hydrogel microstructures, it is possible to achieve sequential drug release and the drug action time can be prolonged, thereby catering to the multi-systemic collaborative regulation needs of osteosynthesis. The drug release rate within the hydrogel is governed by swelling control systems, physical control systems, chemical control systems, and environmental control systems. Utilizing these control systems to design hydrogel materials capable of multi-drug delivery optimizes the construction of the bone microenvironment. Consequently, this facilitates the spatiotemporal controlled released of drugs, promoting bone tissue regeneration. This paper reviews the principles of the controlled release system of various sustained-release hydrogels and the advancements in research on hydrogel multi-drug delivery systems for bone tissue regeneration.


Subject(s)
Bone Regeneration , Delayed-Action Preparations , Hydrogels , Hydrogels/chemistry , Bone Regeneration/drug effects , Humans , Animals , Drug Liberation , Drug Delivery Systems , Osteogenesis/drug effects , Tissue Scaffolds/chemistry
11.
Sci Rep ; 14(1): 15556, 2024 07 05.
Article in English | MEDLINE | ID: mdl-38969656

ABSTRACT

Previously, we reported successful cellular expansion of a murine colorectal carcinoma cell line (CT-26) using a three-dimensional (3D) engineered extracellular matrix (EECM) fibrillar scaffold structure. CCL-247 were grown over a limited time period of 8 days on 3D EECM or tissue culture polystyrene (TCPS). Cells were then assayed for growth, electroporation efficiency and Vigil manufacturing release criteria. Using EECM scaffolds, we report an expansion of CCL-247 (HCT116), a colorectal carcinoma cell line, from a starting concentration of 2.45 × 105 cells to 1.9 × 106 cells per scaffold. Following expansion, 3D EECM-derived cells were assessed based on clinical release criteria of the Vigil manufacturing process utilized for Phase IIb trial operation with the FDA. 3D EECM-derived cells passed all Vigil manufacturing release criteria including cytokine expression. Here, we demonstrate successful Vigil product manufacture achieving the specifications necessary for the clinical trial product release of Vigil treatment. Our results confirm that 3D EECM can be utilized for the expansion of human cancer cell CCL-247, justifying further clinical development involving human tissue sample manufacturing including core needle biopsy and minimal ascites samples.


Subject(s)
Extracellular Matrix , Immunotherapy , Tissue Scaffolds , Humans , Tissue Scaffolds/chemistry , Immunotherapy/methods , Tissue Engineering/methods , HCT116 Cells , Colorectal Neoplasms/pathology , Animals , Mice , Cell Proliferation , Cell Line, Tumor , Cell Culture Techniques, Three Dimensional/methods
12.
Sci Rep ; 14(1): 15695, 2024 07 08.
Article in English | MEDLINE | ID: mdl-38977824

ABSTRACT

Hydrogels are extensively explored as biomaterials for tissue scaffolds, and their controlled fabrication has been the subject of wide investigation. However, the tedious mechanical property adjusting process through formula control hindered their application for diverse tissue scaffolds. To overcome this limitation, we proposed a two-step process to realize simple adjustment of mechanical modulus over a broad range, by combining digital light processing (DLP) and post-processing steps. UV-curable hydrogels (polyacrylamide-alginate) are 3D printed via DLP, with the ability to create complex 3D patterns. Subsequent post-processing with Fe3+ ions bath induces secondary crosslinking of hydrogel scaffolds, tuning the modulus as required through soaking in solutions with different Fe3+ concentrations. This innovative two-step process offers high-precision (10 µm) and broad modulus adjusting capability (15.8-345 kPa), covering a broad range of tissues in the human body. As a practical demonstration, hydrogel scaffolds with tissue-mimicking patterns were printed for cultivating cardiac tissue and vascular scaffolds, which can effectively support tissue growth and induce tissue morphologies.


Subject(s)
Hydrogels , Printing, Three-Dimensional , Tissue Engineering , Tissue Scaffolds , Tissue Scaffolds/chemistry , Hydrogels/chemistry , Tissue Engineering/methods , Humans , Alginates/chemistry , Biocompatible Materials/chemistry , Acrylic Resins/chemistry , Elastic Modulus , Light
13.
J Mech Behav Biomed Mater ; 157: 106646, 2024 Sep.
Article in English | MEDLINE | ID: mdl-38981181

ABSTRACT

Graphene oxide (GO) exhibits excellent mechanical strength and modulus. However, its effectiveness in mechanically reinforcing polymer materials is limited due to issues with interfacial bonding and dispersion arising from differences in the physicochemical properties between GO and polymers. Surface modification using coupling agents is an effective method to improve the bonding problem between polymer and GO, but there may be biocompatibility issues when used in the biomedical field. In this study, the biomolecule L-lysine, was applied to improve the interfacial bonding and dispersion of GO in polylactic acid (PLA) without compromising biocompatibility. The PLA/L-lysine-modified GO (PLA/L-GO) bone scaffold with triply periodic minimal surface (TPMS) structure was prepared using fused deposition modeling (FDM). The FTIR results revealed successful grafting of L-lysine onto GO through the reaction between their -COOH and -NH2 groups. The macroscopic and microscopic morphology characterization indicated that the PLA/L-GO scaffolds exhibited an characteristics of dynamic diameter changes, with good interlayer bonding. It was noteworthy that the L-lysine modification promoted the dispersion of GO and the interfacial bonding with the PLA matrix, as characterized by SEM. As a result, the PLA/0.1L-GO scaffold exhibited higher compressive strength (13.2 MPa) and elastic modulus (226.8 MPa) than PLA/0.1GO. Moreover, PLA/L-GO composite scaffold exhibited superior biomineralization capacity and cell response compared to PLA/GO. In summary, L-lysine not only improved the dispersion and interfacial bonding of GO with PLA, enhancing the mechanical properties, but also improved the biological properties. This study suggests that biomolecules like L-lysine may replace traditional modifiers as an innovative bio-modifier to improve the performance of polymer/inorganic composite biomaterials.


Subject(s)
Graphite , Lysine , Materials Testing , Mechanical Phenomena , Polyesters , Printing, Three-Dimensional , Tissue Scaffolds , Polyesters/chemistry , Tissue Scaffolds/chemistry , Porosity , Graphite/chemistry , Lysine/chemistry , Biocompatible Materials/chemistry , Biocompatible Materials/pharmacology , Animals
14.
Tissue Cell ; 89: 102461, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38991272

ABSTRACT

The current study was constructed to fabricate polyamide based nanofibrous scaffolds (NS) and to define the most promising one for the generation of cardiomyocytes from adipose tissue derived mesenchymal stem cells (ADMSCs). This purpose was extended to assess the potentiality of the generated cardiomyocytes in relieving myocardial infarction (MI) in rats. Production and characterization of NSs were carried out. ADMSCs were cultured on NS and induced to differentiate into cardiomyocytes by specific growth factors. Molecular analysis for myocyte-specific enhancer factor 2 C (MEF2C) and alpha sarcomeric actin (α-SCA) expression was done to confirm the differentiation of ADMSCs into cardiomyocytes for further transplantation into MI induced rats. Implantation of cells in MI afflicted rats boosted heart rate, ST height and PR interval and lessened P duration, RR, QTc and QRS intervals. Also, this type of medication minified serum lactate dehydrogenase (LDH) and creatine kinase-MB (CK-MB) enzymes activity as well as serum and cardiac troponin T (Tn-T) levels and upraised serum and cardiac α-SCA and cardiac connexin 43 (CX 43) levels. Microscopic feature of cardiac tissue sections of rats in the treated groups revealed great renovation in the cardiac microarchitecture. Conclusively, this attempt gains insight into a realistic strategy for recovery of MI through systemic employment of in vitro generated cardiomyocytes.


Subject(s)
Cell Differentiation , Mesenchymal Stem Cells , Myocardial Infarction , Myocytes, Cardiac , Nanofibers , Tissue Scaffolds , Animals , Myocardial Infarction/therapy , Myocardial Infarction/pathology , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/cytology , Tissue Scaffolds/chemistry , Nanofibers/chemistry , Rats , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/metabolism , Cells, Cultured , Male
15.
Tissue Cell ; 89: 102462, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39002289

ABSTRACT

Successful treatment of diabetic wounds requires multifactorial approaches. Herein we investigated the effects of a bioengineered three-dimensional dermal derived matrix-scaffold (DMS) in combination with hyperbaric oxygen (HBO) in repairing of wound model in diabetic rats. Thirty days after induction of diabetes, a circular wound was created and treatments were performed for 21 days. Animals were randomly allocated into the untreated group, DMS group, HBO group, and DMS+HBO group. On days 7, 14, and 21, tissue samples were obtained for stereological, molecular, and tensiometrical assessments. Our results showed that the wound closure rate, volume of new dermis and epidermis, numerical density fibroblasts and blood vessels, collagen density, and biomechanical characterize were significantly higher in the treatment groups than in the untreated group, and these changes were more obvious in the DMS+HBO ones. Moreover, the expression of TGF-ß, bFGF, miRNA-21, miRNA-146a, and VEGF genes were meaningfully upregulated in treatment groups compared to the untreated group and were greater in the DMS+HBO group. This is while expression of TNF-α and IL-1ß, as well as the numerical density of neutrophil and macrophage decreased more considerably in the DMS+HBO group than in the other groups. Overall, using both DMS engraftment and HBO treatment has more effects on diabetic wound healing.


Subject(s)
Diabetes Mellitus, Experimental , Hyperbaric Oxygenation , Tissue Scaffolds , Wound Healing , Animals , Diabetes Mellitus, Experimental/therapy , Diabetes Mellitus, Experimental/pathology , Rats , Tissue Scaffolds/chemistry , Male , Rats, Sprague-Dawley
16.
J Mech Behav Biomed Mater ; 157: 106661, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39018918

ABSTRACT

Addressing infected bone defects remains a significant challenge in orthopedics, requiring effective infection control and bone defect repair. A promising therapeutic approach involves the development of dual-functional engineered biomaterials with drug delivery systems that combine antibacterial properties with osteogenesis promotion. The Hydroxyapatite composite scaffolds offer a one-stage treatment, eliminating the need for multiple surgeries and thereby streamlining the process and reducing treatment time. This review delves into the impaired bone repair mechanisms within pathogen-infected and inflamed microenvironments, providing a theoretical foundation for treating infectious bone defects. Additionally, it explores composite scaffolds made of antibacterial and osteogenic materials, along with advanced drug delivery systems that possess both antibacterial and bone-regenerative properties. By offering a comprehensive understanding of the microenvironment of infectious bone defects and innovative design strategies for dual-function scaffolds, this review presents significant advancements in treatment methods for infectious bone defects. Continued research and clinical validation are essential to refine these innovations, ensuring biocompatibility and safety, achieving controlled release and stability, and developing scalable manufacturing processes for widespread clinical application.


Subject(s)
Bone Regeneration , Drug Delivery Systems , Durapatite , Tissue Scaffolds , Bone Regeneration/drug effects , Durapatite/chemistry , Tissue Scaffolds/chemistry , Humans , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/chemistry , Animals , Biocompatible Materials/chemistry , Biocompatible Materials/pharmacology
17.
Theranostics ; 14(9): 3739-3759, 2024.
Article in English | MEDLINE | ID: mdl-38948054

ABSTRACT

Background: The repair of osteoporotic bone defects remains challenging due to excessive reactive oxygen species (ROS), persistent inflammation, and an imbalance between osteogenesis and osteoclastogenesis. Methods: Here, an injectable H2-releasing hydrogel (magnesium@polyethylene glycol-poly(lactic-co-glycolic acid), Mg@PEG-PLGA) was developed to remodel the challenging bone environment and accelerate the repair of osteoporotic bone defects. Results: This Mg@PEG-PLGA gel shows excellent injectability, shape adaptability, and phase-transition ability, can fill irregular bone defect areas via minimally invasive injection, and can transform into a porous scaffold in situ to provide mechanical support. With the appropriate release of H2 and magnesium ions, the 2Mg@PEG-PLGA gel (loaded with 2 mg of Mg) displayed significant immunomodulatory effects through reducing intracellular ROS, guiding macrophage polarization toward the M2 phenotype, and inhibiting the IκB/NF-κB signaling pathway. Moreover, in vitro experiments showed that the 2Mg@PEG-PLGA gel inhibited osteoclastogenesis while promoting osteogenesis. Most notably, in animal experiments, the 2Mg@PEG-PLGA gel significantly promoted the repair of osteoporotic bone defects in vivo by scavenging ROS and inhibiting inflammation and osteoclastogenesis. Conclusions: Overall, our study provides critical insight into the design and development of H2-releasing magnesium-based hydrogels as potential implants for repairing osteoporotic bone defects.


Subject(s)
Bone Regeneration , Hydrogels , Hydrogen , Magnesium , Osteogenesis , Osteoporosis , Polyethylene Glycols , Reactive Oxygen Species , Animals , Magnesium/chemistry , Magnesium/administration & dosage , Reactive Oxygen Species/metabolism , Mice , Polyethylene Glycols/chemistry , Hydrogels/chemistry , Osteoporosis/drug therapy , Osteogenesis/drug effects , Hydrogen/pharmacology , Hydrogen/administration & dosage , Hydrogen/chemistry , RAW 264.7 Cells , Bone Regeneration/drug effects , Immunomodulation/drug effects , Tissue Scaffolds/chemistry , Macrophages/drug effects , Macrophages/metabolism , Polyesters
18.
Adv Protein Chem Struct Biol ; 141: 331-360, 2024.
Article in English | MEDLINE | ID: mdl-38960479

ABSTRACT

We recently identified TMEM230 as a master regulator of the endomembrane system of cells. TMEM230 expression is necessary for promoting motor protein dependent intracellular trafficking of metalloproteins for cellular energy production in mitochondria. TMEM230 is also required for transport and secretion of metalloproteinases for autophagy and phagosome dependent clearance of misfolded proteins, defective RNAs and damaged cells, activities that decline with aging. This suggests that aberrant levels of TMEM230 may contribute to aging and regain of proper levels may have therapeutic applications. The components of the endomembrane system include the Golgi complex, other membrane bound organelles, and secreted vesicles and factors. Secreted cellular components modulate immune response and tissue regeneration in aging. Upregulation of intracellular packaging, trafficking and secretion of endosome components while necessary for tissue homeostasis and normal wound healing, also promote secretion of pro-inflammatory and pro-senescence factors. We recently determined that TMEM230 is co-regulated with trafficked cargo of the endomembrane system, including lysosome factors such as RNASET2. Normal tissue regeneration (in aging), repair (following injury) and aberrant destructive tissue remodeling (in cancer or autoimmunity) likely are regulated by TMEM230 activities of the endomembrane system, mitochondria and autophagosomes. The role of TMEM230 in aging is supported by its ability to regulate the pro-inflammatory secretome and senescence-associated secretory phenotype in tissue cells of patients with advanced age and chronic disease. Identifying secreted factors regulated by TMEM230 in young patients and patients of advanced age will facilitate identification of aging associated targets that aberrantly promote, inhibit or reverse aging. Ex situ culture of patient derived cells for identifying secreted factors in tissue regeneration and aging provides opportunities in developing therapeutic and personalized medicine strategies. Identification and validation of human secreted factors in tissue regeneration requires long-term stabile scaffold culture conditions that are different from those previously reported for cell lines used as cell models for aging. We describe a 3 dimensional (3D) platform utilizing non-biogenic and non-labile poly ε-caprolactone scaffolds that supports maintenance of long-term continuous cultures of human stem cells, in vitro generated 3D organoids and patient derived tissue. Combined with animal component free culture media, non-biogenic scaffolds are suitable for proteomic and glycobiological analyses to identify human factors in aging. Applications of electrospun nanofiber technologies in 3D cell culture allow for ex situ screening and the development of patient personalized therapeutic strategies and predicting their effectiveness in mitigating or promoting aging.


Subject(s)
Aging , Organoids , Humans , Organoids/metabolism , Aging/metabolism , Membrane Proteins/metabolism , Cellular Senescence , Female , Tissue Scaffolds/chemistry , Mammary Glands, Human/metabolism , Mammary Glands, Human/cytology
19.
Sci Rep ; 14(1): 15196, 2024 07 02.
Article in English | MEDLINE | ID: mdl-38956215

ABSTRACT

Despite recent advancements in peripheral nerve regeneration, the creation of nerve conduits with chemical and physical cues to enhance glial cell function and support axonal growth remains challenging. This study aimed to assess the impact of electrical stimulation (ES) using a conductive nerve conduit on sciatic nerve regeneration in a rat model with transection injury. The study involved the fabrication of conductive nerve conduits using silk fibroin and Au nanoparticles (AuNPs). Collagen hydrogel loaded with green fluorescent protein (GFP)-positive adipose-derived mesenchymal stem cells (ADSCs) served as the filling for the conduit. Both conductive and non-conductive conduits were applied with and without ES in rat models. Locomotor recovery was assessed using walking track analysis. Histological evaluations were performed using H&E, luxol fast blue staining and immunohistochemistry. Moreover, TEM analysis was conducted to distinguish various ultrastructural aspects of sciatic tissue. In the ES + conductive conduit group, higher S100 (p < 0.0001) and neurofilament (p < 0.001) expression was seen after 6 weeks. Ultrastructural evaluations showed that conductive scaffolds with ES minimized Wallerian degeneration. Furthermore, the conductive conduit with ES group demonstrated significantly increased myelin sheet thickness and decreased G. ratio compared to the autograft. Immunofluorescent images confirmed the presence of GFP-positive ADSCs by the 6th week. Locomotor recovery assessments revealed improved function in the conductive conduit with ES group compared to the control group and groups without ES. These results show that a Silk/AuNPs conduit filled with ADSC-seeded collagen hydrogel can function as a nerve conduit, aiding in the restoration of substantial gaps in the sciatic nerve with ES. Histological and locomotor evaluations indicated that ES had a greater impact on functional recovery compared to using a conductive conduit alone, although the use of conductive conduits did enhance the effects of ES.


Subject(s)
Nerve Regeneration , Sciatic Nerve , Tissue Scaffolds , Animals , Sciatic Nerve/physiology , Rats , Tissue Scaffolds/chemistry , Gold/chemistry , Rats, Sprague-Dawley , Silk/chemistry , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/metabolism , Electric Stimulation/methods , Fibroins/chemistry , Metal Nanoparticles/chemistry , Male , Recovery of Function , Guided Tissue Regeneration/methods , Hydrogels/chemistry
20.
J Mater Sci Mater Med ; 35(1): 38, 2024 Jul 03.
Article in English | MEDLINE | ID: mdl-38958834

ABSTRACT

Fabrication of porous tissue-engineering scaffolds from bioactive glasses (BAG) is complicated by the tendency of BAG compositions to crystallize in thermal treatments during scaffold manufacture. Here, experimental biocompatible glass S59 (SiO2 59.7 wt%, Na2O 25.5 wt%, CaO 11.0 wt%, P2O5 2.5 wt%, B2O3 1.3 wt%), known to be resistant to crystallization, was used in sintering of glass granules (300-500 µm) into porous scaffolds. The dissolution behavior of the scaffolds was then studied in vivo in rabbit femurs and under continuous flow conditions in vitro (14 days in vitro/56 days in vivo). The scaffolds were osteoconductive in vivo, as bone could grow into the scaffold structure. Still, the scaffolds could not induce sufficiently rapid bone ingrowth to replace the strength lost due to dissolution. The scaffolds lost their structure and strength as the scaffold necks dissolved. In vitro, S59 dissolved congruently throughout the 14-day experiments, resulting in only a slight reaction layer formation. Manufacturing BAG scaffolds from S59 that retain their amorphous structure was thus possible. The relatively rapid and stable dissolution of the scaffold implies that the glass S59 may have the potential to be used in composite implants providing initial strength and stable, predictable release of ions over longer exposure times.


Subject(s)
Biocompatible Materials , Glass , Materials Testing , Tissue Engineering , Tissue Scaffolds , Animals , Rabbits , Tissue Scaffolds/chemistry , Glass/chemistry , Biocompatible Materials/chemistry , Porosity , Tissue Engineering/methods , Femur , Solubility , Bone Substitutes/chemistry , Bone Regeneration
SELECTION OF CITATIONS
SEARCH DETAIL