Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 88
Filtrar
1.
Adv Funct Mater ; 33(2)2023 Jan 10.
Artigo em Inglês | MEDLINE | ID: mdl-36816838

RESUMO

Peripheral nerve transection has a high prevalence and results in functional loss of affected limbs. The current clinical treatment using suture anastomosis significantly limits nerve recovery due to severe inflammation, secondary damage, and fibrosis. Fibrin glue, a commercial nerve adhesive as an alternative, avoids secondary damage but suffers from poor adhesion strength. To address their limitations, a highly efficacious nerve adhesive based on dual-crosslinking of dopamine-isothiocyanate modified hyaluronic acid and decellularized nerve matrix is reported in this paper. This dual-network nerve adhesive (DNNA) shows controllable gelation behaviors feasible for surgical applications, robust adhesion strength, and promoted axonal outgrowth in vitro. The in vivo therapeutic efficacy is tested using a rat-based sciatic nerve transection model. The DNNA decreases fibrosis and accelerates axon/myelin debris clearance at 10 days post-surgery, compared to suture and commercial fibrin glue treatments. At 10 weeks post-surgery, the strong adhesion and bioactivity allow DNNA to significantly decrease intraneural inflammation and fibrosis, enhance axon connection and remyelination, aid motor and sensory function recovery, as well as improve muscle contraction, compared to suture and fibrin treatments. Overall, this dual-network hydrogel with robust adhesion provides a rapid and highly efficacious nerve transection treatment to facilitate nerve repair and neuromuscular function recovery.

2.
J Transl Med ; 20(1): 209, 2022 05 13.
Artigo em Inglês | MEDLINE | ID: mdl-35562738

RESUMO

BACKGROUND: Colorectal cancer is a commonly diagnosed cancer worldwide. Unfortunately, many patients do not respond to standard chemotherapy treatments and develop disease relapse and metastases. Besides cancer cell specific genetic changes, heterogeneity in the tumor microenvironment contribute to the clinical presentation of the disease and can potentially also influence drug resistance. By using a recently developed patient-derived scaffold method monitoring how a standardized reporter cancer cell line adapts to various microenvironments treated with chemotherapy, we wanted to clarify how individual patient specific microenvironments influence the chemotherapy response in colorectal cancer. METHODS: Surgically resected colorectal cancer specimens from 89 patients were decellularized to produce patient-derived scaffold, which were seeded with HT29 cells, cultured for 3 weeks, and treated with 5-fluorouracil. Gene expression changes of adapted and treated HT29 cells were monitored by qPCR and compared with clinical parameters including disease-free survival. RESULTS: The effects of 5-fluorouracil treatment varied between different patient-derived scaffold, but generally induced a reduced expression of proliferation genes and increased expression of pluripotency and epithelial-to-mesenchymal transition genes. Interestingly, patient-derived scaffold cultures obtained from patients with disease recurrences showed a significantly less pronounced anti-proliferative effect of 5-fluorouracil and more pronounced increase of pluripotency, with MKI67 and POU5F1 being among the most significant genes linked to disease relapse in colorectal cancer. CONCLUSIONS: Colorectal patient-derived scaffold can decode clinically relevant tumor microenvironmental influence of 5-fluorouracil treatment effects opening up for optimized precision medicine in colorectal cancer treatment.


Assuntos
Neoplasias Colorretais , Fluoruracila , Neoplasias Colorretais/tratamento farmacológico , Neoplasias Colorretais/genética , Neoplasias Colorretais/patologia , Fluoruracila/farmacologia , Fluoruracila/uso terapêutico , Células HT29 , Humanos , Recidiva Local de Neoplasia/patologia , Microambiente Tumoral
3.
Cell Tissue Bank ; 21(2): 215-231, 2020 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-32020424

RESUMO

This study aims to evaluate the CD146+ stem cells obtained from the human umbilical cord and their extracellular matrix proteins on in vitro Pseudomonas aeruginosa and Staphylococcus aureus biofilms to understand their possible antimicrobial activity. CD146+ stem cells were determined according to cell surface markers and differentiation capacity. Characterization of the decellularized matrix was done with DAPI, Masson's Trichrome staining and proteome analysis. Cell viability/proliferation of cells in co-cultures was evaluated by WST-1 and crystal-violet staining. The effects of cells and decellularized matrix proteins on biofilms were investigated on a drip flow biofilm reactor and their effects on gene expression were determined by RT-qPCR. We observed that CD146/105+ stem cells could differentiate adipogenically and decellularized matrix showed negative DAPI and positive collagen staining with Masson' s Trichrome. Proteome analysis of the decellularized matrix revealed some matrix components and growth factors. Although the decellularized matrix significantly reduced the cell counts of P. aeruginosa, no significant difference was observed for S. aureus cells in both groups. Supporting data was obtained from the gene expression results of P. aeruginosa with the significant down-regulation of rhlR and lasR. For S. aureus, icaADBC genes were significantly up-regulated when grown on the decellularized matrix.


Assuntos
Biofilmes/crescimento & desenvolvimento , Antígeno CD146/metabolismo , Matriz Extracelular/metabolismo , Pseudomonas aeruginosa/fisiologia , Staphylococcus aureus/fisiologia , Células-Tronco/citologia , Cordão Umbilical/citologia , Biomarcadores/metabolismo , Diferenciação Celular , Proliferação de Células , Forma Celular , Células Cultivadas , Técnicas de Cocultura , Proteínas da Matriz Extracelular/metabolismo , Regulação Bacteriana da Expressão Gênica , Humanos , Proteoma/metabolismo , Pseudomonas aeruginosa/genética , Pseudomonas aeruginosa/ultraestrutura , Staphylococcus aureus/genética , Staphylococcus aureus/ultraestrutura
4.
Cytotherapy ; 20(1): 74-86, 2018 01.
Artigo em Inglês | MEDLINE | ID: mdl-29050915

RESUMO

BACKGROUND AIMS: Retinal progenitor cells (RPCs) are a promising cell therapy treatment for retinal degenerative diseases. However, problems with limited proliferation ability and differentiation preference toward glia rather than neurons restrict the clinical application of these RPCs. The extracellular matrix (ECM) has been recognized to provide an appropriate microenvironment to support stem cell adhesion and direct cell behaviors, such as self-renewal and differentiation. METHODS: In this study, decellularized matrix of adipose-derived mesenchymal stromal cells (DMA) was manufactured using a chemical agent method (0.5% ammonium hydroxide Triton + 20 mmol/L NH4OH) in combination with a biological agent method (DNase solution), and the resulting DMA were evaluated by scanning electron microscopy (SEM) and immunocytochemistry. The effect of DMA on RPC proliferation and differentiation was evaluated by quantitative polymerase chain reaction, Western blot and immunocytochemistry analysis. RESULTS: DMA was successfully fabricated, as demonstrated by SEM and immunocytochemistry. Compared with tissue culture plates, DMA may effectively enhance the proliferation of RPCs by activating Akt and Erk phosphorylation; when the two pathways were blocked, the promoting effect was reversed. Moreover, DMA promoted the differentiation of RPCs toward retinal neurons, especially rhodopsin- and recoverin-positive photoreceptors, which is the most interesting class of cells for retinal degeneration treatment. CONCLUSIONS: These results indicate that DMA has important roles in governing RPC proliferation and differentiation and may contribute to the application of RPCs in treating retinal degenerative diseases.


Assuntos
Tecido Adiposo/citologia , Diferenciação Celular , Matriz Extracelular/metabolismo , Sistema de Sinalização das MAP Quinases , Células-Tronco Mesenquimais/metabolismo , Neurônios/citologia , Proteínas Proto-Oncogênicas c-akt/metabolismo , Retina/citologia , Adolescente , Adulto , Animais , Diferenciação Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Separação Celular , Células Cultivadas , Feminino , Humanos , Camundongos Endogâmicos C57BL , Adulto Jovem
5.
Ultrastruct Pathol ; 42(1): 32-38, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29192810

RESUMO

Recently, interest has been increasing for human decellularized matrices, due to their ability to reduce numerous side effects related to hernia repair. To date, only animal studies investigated the biological interaction post-implant of human decellularized matrices for soft tissue repair. Therefore, the aim of this study was to evaluate the morphological response one year post implant of human decellularized matrix, through morphological analysis of human biopsies. The histological and ultrastructural results revealed a perfect cellular repopulation and neoangiogenesis, with minimal inflammatory response and a well-organized collagen matrix. The results have indicated that this scaffold can be an effective treatment for hernia.


Assuntos
Derme Acelular , Herniorrafia/métodos , Matriz Extracelular/ultraestrutura , Feminino , Humanos , Masculino , Microscopia Eletrônica de Transmissão , Pessoa de Meia-Idade
6.
Oral Dis ; 22(4): 313-23, 2016 May.
Artigo em Inglês | MEDLINE | ID: mdl-26785831

RESUMO

OBJECTIVE: A challenge in engineering tissues is to supply parenchymal cells with suitable scaffolds which ideally reproduce the extracellular matrix (ECM). This study tested the hypothesis of preserving the 'residual connective tissue' remaining after mechanical and enzymatic release of cells from human submandibular gland biopsies (that we named 'natural ExtraCellular Matrix scaffolds', nECMsc) to be used as recycled natural scaffolds. The objective was to test whether nECMsc and native salivary tissue were comparable morphologically, in ECM proteins composition, and in cell seeding efficiency. METHODS: Following cell isolation procedures, nECMsc were kept, either fresh or frozen (sectioned into 12-µm-thick slices), and examined with high-resolution electron microscopy (HRSEM) for its three-dimensional structure, and with picrosirius red staining and immunogold staining for ECM protein composition and distribution, respectively. nECMsc were seeded with human epithelial cells and fibroblasts to assess cell attachment and proliferation in short-term experiments. RESULTS: Under HRSEM, nECMsc had comparable fiber arrangement to original glands. Histochemical and immunogold-labeling examinations revealed the presence of collagen types I, III, and IV. Seeded epithelial cells and fibroblasts attached, proliferated (14-55%), and were alive (86-99%) after 4-8 days of culture. CONCLUSIONS: nECMsc retained native ECM proteins and maintained their distribution. Seeded cells remained viable on nECMsc.


Assuntos
Matriz Extracelular/química , Matriz Extracelular/ultraestrutura , Glândula Submandibular , Engenharia Tecidual/métodos , Alicerces Teciduais , Adulto , Idoso , Adesão Celular , Proliferação de Células , Colágeno Tipo I/análise , Colágeno Tipo III/análise , Colágeno Tipo IV/análise , Células Epiteliais/fisiologia , Fibroblastos/fisiologia , Humanos , Masculino , Pessoa de Meia-Idade , Técnicas de Cultura de Tecidos
7.
Nano Lett ; 14(10): 5792-6, 2014 Oct 08.
Artigo em Inglês | MEDLINE | ID: mdl-25176294

RESUMO

Decellularized matrices are valuable scaffolds for engineering functional cardiac patches for treating myocardial infarction. However, the lack of quick and efficient electrical coupling between adjacent cells may jeopardize the success of the treatment. To address this issue, we have deposited gold nanoparticles on fibrous decellularized omental matrices and investigated their morphology, conductivity, and degradation. We have shown that cardiac cells engineered within the hybrid scaffolds exhibited elongated and aligned morphology, massive striation, and organized connexin 43 electrical coupling proteins. Finally, we have shown that the hybrid patches demonstrated superior function as compared to pristine patches, including a stronger contraction force, lower excitation threshold, and faster calcium transients.


Assuntos
Ouro/química , Nanopartículas Metálicas/química , Miocárdio/citologia , Engenharia Tecidual/métodos , Alicerces Teciduais/química , Animais , Células Cultivadas , Conexina 43/análise , Nanopartículas Metálicas/ultraestrutura , Ratos
8.
Int J Biol Macromol ; 254(Pt 2): 127891, 2024 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-37931866

RESUMO

Tissue engineering (TE) has become a primary research topic for the treatment of diseased or damaged tendon/ligament (T/L) tissue. T/L injuries pose a severe clinical burden worldwide, necessitating the development of effective strategies for T/L repair and tissue regeneration. TE has emerged as a promising strategy for restoring T/L function using decellularized extracellular matrix (dECM)-based scaffolds. dECM scaffolds have gained significant prominence because of their native structure, relatively high bioactivity, low immunogenicity, and ability to function as scaffolds for cell attachment, proliferation, and differentiation, which are difficult to imitate using synthetic materials. Here, we review the recent advances and possible future prospects for the advancement of dECM scaffolds for T/L tissue regeneration. We focus on crucial scaffold properties and functions, as well as various engineering strategies employed for biomaterial design in T/L regeneration. dECM provides both the physical and mechanical microenvironments required by cells to survive and proliferate. Various decellularization methods and sources of allogeneic and xenogeneic dECM in T/L repair and regeneration are critically discussed. Additionally, dECM hydrogels, bio-inks in 3D bioprinting, and nanofibers are briefly explored. Understanding the opportunities and challenges associated with dECM-based scaffold development is crucial for advancing T/L repairs in tissue engineering and regenerative medicine.


Assuntos
Matriz Extracelular , Alicerces Teciduais , Alicerces Teciduais/química , Matriz Extracelular/química , Engenharia Tecidual , Polissacarídeos/análise , Tendões
9.
Biomed Mater ; 19(4)2024 May 07.
Artigo em Inglês | MEDLINE | ID: mdl-38653259

RESUMO

The decellularized matrix has a great potential for tissue remodeling and regeneration; however, decellularization could induce host immune rejection due to incomplete cell removal or detergent residues, thereby posing significant challenges for its clinical application. Therefore, the selection of an appropriate detergent concentration, further optimization of tissue decellularization technique, increased of biosafety in decellularized tissues, and reduction of tissue damage during the decellularization procedures are pivotal issues that need to be investigated. In this study, we tested several conditions and determined that 0.1% Sodium dodecyl sulfate and three decellularization cycles were the optimal conditions for decellularization of pulp tissue. Decellularization efficiency was calculated and the preparation protocol for dental pulp decellularization matrix (DPDM) was further optimized. To characterize the optimized DPDM, the microstructure, odontogenesis-related protein and fiber content were evaluated. Our results showed that the properties of optimized DPDM were superior to those of the non-optimized matrix. We also performed the 4D-Label-free quantitative proteomic analysis of DPDM and demonstrated the preservation of proteins from the natural pulp. This study provides a optimized protocol for the potential application of DPDM in pulp regeneration.


Assuntos
Matriz Extracelular Descelularizada , Polpa Dentária , Proteômica , Engenharia Tecidual , Alicerces Teciduais , Polpa Dentária/citologia , Proteômica/métodos , Animais , Engenharia Tecidual/métodos , Alicerces Teciduais/química , Matriz Extracelular Descelularizada/química , Dodecilsulfato de Sódio/química , Humanos , Odontogênese , Matriz Extracelular/metabolismo , Matriz Extracelular/química
10.
Tissue Eng Part A ; 2024 May 02.
Artigo em Inglês | MEDLINE | ID: mdl-38623816

RESUMO

The arteriovenous loop (AVL) model allows the in vivo engineering of axially vascularized flaps, the so-called AVL flaps. Although AVL flaps can be transplanted microsurgically to cover tissue defects, they lack an epithelial layer on the surface. Therefore, the objective of this study was to engineer axially vascularized AVL flaps with an accompanying epithelial layer for local defect reconstruction. In this study, AVLs were established in 20 male Lewis rats. Minimally invasive injection of keratinocytes onto the surface of the AVL flaps was performed on postoperative day (POD) 21. AVL flaps were explanted from 12 rats on POD 24 or POD 30, then the epithelium formed by the keratinocytes on the surface of the flaps was evaluated using immunofluorescence staining. In six other rats, the AVL flap was locally transposed to cover a critical defect in the rats' leg on POD 30 and explanted for analysis on POD 40. In two control rats, sodium chloride was applied instead of keratinocytes. These control flaps were also transplanted on POD 30 and explanted on POD 40. Our results revealed that 3 days after keratinocyte application, a loose single-layered epithelium was observed histologically on the AVL flaps surface, whereas after 9 days, a multilayered and structured epithelium had grown. The epithelium on the transplanted AVL flaps showed its physiological differentiation when being exposed to an air-liquid interface. Histologically, a layered epithelium identical to the rats' regular skin was formed. In the sodium chloride control group, no epithelium had been grown. This study clearly demonstrates that axially vascularized AVL flaps can be processed in the subcutaneous chamber by minimally invasive injection of keratinocytes. Thus, AVL flaps with an intact epithelial layer were engineered and could be successfully transplanted for local defect coverage in a small animal model.

11.
Biofabrication ; 16(4)2024 Sep 19.
Artigo em Inglês | MEDLINE | ID: mdl-39255833

RESUMO

The transplantation of islet beta cells offers an alternative to heterotopic islet transplantation for treating type 1 diabetes mellitus (T1DM). However, the use of systemic immunosuppressive drugs in islet transplantation poses significant risks to the body. To address this issue, we constructed an encapsulated hybrid scaffold loaded with islet beta cells. This article focuses on the preparation of the encapsulated structure using 3D printing, which incorporates porcine pancreas decellularized extracellular matrix (dECM) to the core scaffold. The improved decellularization method successfully preserved a substantial proportion of protein (such as Collagen I and Laminins) architecture and glycosaminoglycans in the dECM hydrogel, while effectively removing most of the DNA. The inclusion of dECM enhanced the physical and chemical properties of the scaffold, resulting in a porosity of 83.62% ± 1.09% and a tensile stress of 1.85 ± 0.16 MPa. In teams of biological activity, dECM demonstrated enhanced proliferation, differentiation, and expression of transcription factors such as Ki67, PDX1, and NKX6.1, leading to improved insulin secretion function in MIN-6 pancreatic beta cells. In the glucose-stimulated insulin secretion experiment on day 21, the maximum insulin secretion from the encapsulated structure reached 1.96 ± 0.08 mIU ml-1, representing a 44% increase compared to the control group. Furthermore, conventional capsule scaffolds leaverage the compatibility of natural biomaterials with macrophages to mitigate immune rejection. Here, incorporating curcumin into the capsule scaffold significantly reduced the secretion of pro-inflammatory cytokine (IL-1ß, IL-6, TNF-α, IFN-γ) secretion by RAW264.7 macrophages and T cells in T1DM mice. This approach protected pancreatic islet cells against immune cell infiltration mediated by inflammatory factors and prevented insulitis. Overall, the encapsulated scaffold developed in this study shows promise as a natural platform for clinical treatment of T1DM.


Assuntos
Curcumina , Matriz Extracelular Descelularizada , Diabetes Mellitus Tipo 1 , Células Secretoras de Insulina , Alicerces Teciduais , Animais , Diabetes Mellitus Tipo 1/terapia , Células Secretoras de Insulina/efeitos dos fármacos , Células Secretoras de Insulina/metabolismo , Células Secretoras de Insulina/citologia , Alicerces Teciduais/química , Curcumina/farmacologia , Curcumina/química , Camundongos , Matriz Extracelular Descelularizada/química , Matriz Extracelular Descelularizada/farmacologia , Suínos , Transplante das Ilhotas Pancreáticas , Cápsulas/química , Insulina/metabolismo , Diabetes Mellitus Experimental/terapia , Linhagem Celular , Matriz Extracelular/metabolismo , Matriz Extracelular/química
12.
Bioengineering (Basel) ; 11(4)2024 Mar 28.
Artigo em Inglês | MEDLINE | ID: mdl-38671751

RESUMO

Cartilage damage presents a significant clinical challenge due to its intrinsic avascular nature which limits self-repair. Addressing this, our study focuses on an alginate-based bioink, integrating human articular cartilage, for cartilage tissue engineering. This novel bioink was formulated by encapsulating C20A4 human articular chondrocytes in sodium alginate, polyvinyl alcohol, gum arabic, and cartilage extracellular matrix powder sourced from allograft femoral condyle shavings. Using a 3D bioprinter, constructs were biofabricated and cross-linked, followed by culture in standard medium. Evaluations were conducted on cellular viability and gene expression at various stages. Results indicated that the printed constructs maintained a porous structure conducive to cell growth. Cellular viability was 87% post printing, which decreased to 76% after seven days, and significantly recovered to 86% by day 14. There was also a notable upregulation of chondrogenic genes, COL2A1 (p = 0.008) and SOX9 (p = 0.021), suggesting an enhancement in cartilage formation. This study concludes that the innovative bioink shows promise for cartilage regeneration, demonstrating substantial viability and gene expression conducive to repair and suggesting its potential for future therapeutic applications in cartilage repair.

13.
Zhongguo Xiu Fu Chong Jian Wai Ke Za Zhi ; 38(6): 748-754, 2024 Jun 15.
Artigo em Zh | MEDLINE | ID: mdl-38918198

RESUMO

Objective: To investigate the construction of a novel tissue engineered meniscus scaffold based on low temperature deposition three-dimenisonal (3D) printing technology and evaluate its biocompatibility. Methods: The fresh pig meniscus was decellularized by improved physicochemical method to obtain decellularized meniscus matrix homogenate. Gross observation, HE staining, and DAPI staining were used to observe the decellularization effect. Toluidine blue staining, safranin O staining, and sirius red staining were used to evaluate the retention of mucopolysaccharide and collagen. Then, the decellularized meniscus matrix bioink was prepared, and the new tissue engineered meniscus scaffold was prepared by low temperature deposition 3D printing technology. Scanning electron microscopy was used to observe the microstructure. After co-culture with adipose-derived stem cells, the cell compatibility of the scaffolds was observed by cell counting kit 8 (CCK-8), and the cell activity and morphology were observed by dead/live cell staining and cytoskeleton staining. The inflammatory cell infiltration and degradation of the scaffolds were evaluated by subcutaneous experiment in rats. Results: The decellularized meniscus matrix homogenate appeared as a transparent gel. DAPI and histological staining showed that the immunogenic nucleic acids were effectively removed and the active components of mucopolysaccharide and collagen were remained. The new tissue engineered meniscus scaffolds was constructed by low temperature deposition 3D printing technology and it had macroporous-microporous microstructures under scanning electron microscopy. CCK-8 test showed that the scaffolds had good cell compatibility. Dead/live cell staining showed that the scaffold could effectively maintain cell viability (>90%). Cytoskeleton staining showed that the scaffolds were benefit for cell adhesion and spreading. After 1 week of subcutaneous implantation of the scaffolds in rats, there was a mild inflammatory response, but no significant inflammatory response was observed after 3 weeks, and the scaffolds gradually degraded. Conclusion: The novel tissue engineered meniscus scaffold constructed by low temperature deposition 3D printing technology has a graded macroporous-microporous microstructure and good cytocompatibility, which is conducive to cell adhesion and growth, laying the foundation for the in vivo research of tissue engineered meniscus scaffolds in the next step.


Assuntos
Menisco , Impressão Tridimensional , Engenharia Tecidual , Alicerces Teciduais , Animais , Engenharia Tecidual/métodos , Alicerces Teciduais/química , Suínos , Ratos , Menisco/citologia , Materiais Biocompatíveis , Ratos Sprague-Dawley , Células Cultivadas , Meniscos Tibiais/citologia , Microscopia Eletrônica de Varredura
14.
J Biomed Mater Res A ; 112(12): 2273-2288, 2024 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-39015005

RESUMO

The objective of this study was to create injectable photo-crosslinkable biomaterials, using gelatin methacryloyl (GelMA) hydrogel, combined with a decellularized bone matrix (BMdc) and a deproteinized (BMdp) bovine bone matrix. These were intended to serve as bioactive scaffolds for dentin regeneration. The parameters for GelMA hydrogel fabrication were initially selected, followed by the incorporation of BMdc and BMdp at a 1% (w/v) ratio. Nano-hydroxyapatite (nHA) was also included as a control. A physicochemical characterization was conducted, with FTIR analysis indicating that the mineral phase was complexed with GelMA, and BMdc was chemically bonded to the amide groups of gelatin. The porous structure was preserved post-BMdc incorporation, with bone particles incorporated alongside the pores. Conversely, the mineral phase was situated inside the pore opening, affecting the degree of porosity. The mineral phase did not modify the degradability of GelMA, even under conditions of type I collagenase-mediated enzymatic challenge, allowing hydrogel injection and increased mechanical strength. Subsequently, human dental pulp cells (HDPCs) were seeded onto the hydrogels. The cells remained viable and proliferative, irrespective of the GelMA composition. All mineral phases resulted in a significant increase in alkaline phosphatase activity and mineralized matrix deposition. However, GelMA-BMdc exhibited higher cell expression values, significantly surpassing those of all other formulations. In conclusion, our results showed that GelMA-BMdc produced a porous and stable hydrogel, capable of enhancing odontoblastic differentiation and mineral deposition when in contact with HDPCs, thereby showing potential for dentin regeneration.


Assuntos
Polpa Dentária , Dentina , Gelatina , Engenharia Tecidual , Dentina/química , Engenharia Tecidual/métodos , Animais , Bovinos , Gelatina/química , Humanos , Polpa Dentária/citologia , Metacrilatos/química , Reagentes de Ligações Cruzadas/química , Hidrogéis/química , Alicerces Teciduais/química , Osso e Ossos , Células Cultivadas , Porosidade
15.
Front Bioeng Biotechnol ; 12: 1441933, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-39211011

RESUMO

Heart failure is a major health problem in which the heart is unable to pump enough blood to meet the body's needs. It is a progressive disease that becomes more severe over time and can be caused by a variety of factors, including heart attack, cardiomyopathy and heart valve disease. There are various methods to cure this disease, which has many complications and risks. The advancement of knowledge and technology has proposed new methods for many diseases. One of the promising new treatments for heart failure is tissue engineering. Tissue engineering is a field of research that aims to create living tissues and organs to replace damaged or diseased tissue. The goal of tissue engineering in heart failure is to improve cardiac function and reduce the need for heart transplantation. This can be done using the three important principles of cells, biomaterials and signals to improve function or replace heart tissue. The techniques for using cells and biomaterials such as electrospinning, hydrogel synthesis, decellularization, etc. are diverse. Treating heart failure through tissue engineering is still under development and research, but it is hoped that there will be no transplants or invasive surgeries in the near future. In this study, based on the most important research in recent years, we will examine the power of tissue engineering in the treatment of heart failure.

16.
Materials (Basel) ; 16(15)2023 Jul 29.
Artigo em Inglês | MEDLINE | ID: mdl-37570039

RESUMO

Injectable hydrogels possess tremendous merits for use in muscle regeneration; however, they still lack intrinsic biological cues (such as the proliferation and differentiation of myogenic cells), thus considerably restricting their potential for therapeutic use. Herein, we developed a double cross-linked injectable hydrogel composed of methacrylamidated oxidized hyaluronic acid (MOHA) and muscular decellularized matrix (MDM). The chemical composition of the hydrogel was confirmed using 1H NMR and Fourier transform infrared spectroscopy. To achieve cross-linking, the aldehyde groups in MOHA were initially reacted with the amino groups in MDM through a Schiff-based reaction. This relatively weak cross-linking provided the MOHA/MDM hydrogel with satisfactory injectability. Furthermore, the methacrylation of MOHA facilitated a second cross-linking mechanism via UV irradiation, resulting in improved gelation ability, biomechanical properties, and swelling performance. When C2C12 myogenic cells were loaded into the hydrogel, our results showed that the addition of MDM significantly enhanced myoblast proliferation compared to the MOHA hydrogel, as demonstrated by live/dead staining and Cell Counting Kit-8 assay after seven days of in vitro cultivation. In addition, gene expression analysis using quantitative polymerase chain reaction indicated that the MOHA/MDM hydrogel promoted myogenic differentiation of C2C12 cells more effectively than the MOHA hydrogel, as evidenced by elevated expression levels of myogenin, troponin T, and MHC in the MOHA/MDM hydrogel group. Moreover, after four to eight weeks of implantation in a full-thickness abdominal wall-defect model, the MOHA/MDM hydrogel could promote the reconstruction and repair of functional skeletal muscle tissue with enhanced tetanic force and tensile strength. This study provides a new double cross-linked injectable hydrogel for use in muscular tissue engineering.

17.
Acta Biomater ; 156: 37-48, 2023 01 15.
Artigo em Inglês | MEDLINE | ID: mdl-36455855

RESUMO

The sufficient imitation of tissue structures and components represents an effective and promising approach for tissue engineering and regenerative medicine applications. Dental pulp disease is one of the most common oral diseases, although functional pulp regeneration remains challenging. Herein, we propose a strategy that employs hydrogel microspheres incorporated with decellularized dental pulp matrix-derived bioactive factors to simulate a pulp-specific three-dimensional (3D) microenvironment. The dental pulp microenvironment-specific microspheres constructed by this regenerative strategy exhibited favorable plasticity, biocompatibility, and biological performances. Human dental pulp stem cells (hDPSCs) cultured on the constructed microspheres exhibited enhanced pulp-formation ability in vitro. Furthermore, the hDPSCs-microcarriers achieved the regeneration of pulp-like tissue and new dentin in a semi-orthotopic model in vivo. Mechanistically, the decellularized pulp matrix-derived bioactive factors mediated the multi-directional differentiation of hDPSCs to regenerate the pulp tissue by eliciting the secretion of crucial bioactive cues. Our findings demonstrated that a 3D dental pulp-specific microenvironment facilitated by hydrogel microspheres and dental pulp-specific bioactive factors regenerated the pulp-dentin complex and could be served as a promising treatment option for dental pulp disease. STATEMENT OF SIGNIFICANCE: Injectable bioscaffolds are increasingly used for regenerative endodontic treatment. Despite their success related to their ability to load stem cells, bioactive factors, and injectability, conventional bulk bioscaffolds have drawbacks such as ischemic necrosis in the central region. Various studies have shown that ischemic necrosis in the central region can be corrected by injectable hydrogel microspheres. Unfortunately, pristine microspheres or microspheres without dental pulp-specific bioactive factor would oftentimes fail to regulate stem cells fates in dental pulp multi-directional differentiation. Our present study reported the biofabrication of dental pulp-derived decellularized matrix functionalized gelatin microspheres, which contained dental pulp-specific bioactive factors and have the potential application in endodontic regeneration.


Assuntos
Doenças da Polpa Dentária , Hidrogéis , Humanos , Hidrogéis/farmacologia , Polpa Dentária , Microesferas , Regeneração , Diferenciação Celular , Necrose
18.
ACS Appl Mater Interfaces ; 15(2): 2578-2589, 2023 Jan 18.
Artigo em Inglês | MEDLINE | ID: mdl-36598791

RESUMO

Transplantation of exogenous cardiomyocytes (CMs) is a hopeful method to treat myocardial infarction (MI). However, its clinical application still remains challenging due to low retention and survival rates of the transplanted cells. Herein, a stromal cell-derived factor 1 (SDF-1)-loaded injectable hydrogel based on a decellularized porcine extracellular matrix (dECM) is developed to encapsulate and deliver CMs locally to the infarct area of the heart. The soluble porcine cardiac dECM is composed of similar components such as the human cardiac ECM, which could be self-assembled into a nanofibrous hydrogel at physiological temperature to improve the retention of transplanted CMs. Furthermore, the chemokine SDF-1 could recruit endogenous cells to promote angiogenesis, mitigating the ischemic microenvironment and improving the survival of CMs. The results in vitro show that this composite hydrogel exhibits good biocompatibility, anti-apoptosis property, and chemotactic effects for mesenchymal stromal cells and endothelial cells through SDF-1-CXCR4 axis. Moreover, intramyocardial injection of this composite hydrogel to the infarcted area leads to the promotion of angiogenesis and inhibition of fibrosis, reducing the infarction size and improving the cardiac function. The combination of natural biomaterials, exogenous cells, and bioactive factors shows potential for MI treatment in the clinical application.


Assuntos
Quimiocina CXCL12 , Matriz Extracelular Descelularizada , Hidrogéis , Infarto do Miocárdio , Miócitos Cardíacos , Animais , Humanos , Quimiocina CXCL12/química , Quimiocina CXCL12/farmacologia , Matriz Extracelular Descelularizada/química , Matriz Extracelular Descelularizada/farmacologia , Células Endoteliais , Matriz Extracelular , Hidrogéis/farmacologia , Infarto do Miocárdio/terapia , Miócitos Cardíacos/metabolismo , Regeneração , Suínos
19.
J Biomater Appl ; 38(4): 471-483, 2023 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-37670570

RESUMO

Peripheral nerve injury (PNI) is one of the major clinical treatment challenges following an impact on the body. When PNI manifests as nerve gaps, surgical connections and exogenous grafts are required. Recently, electrically conductive polymers (CPs) based nerve guidance conduits have yielded promising results for treating PNI. Polypyrrole (PPy) has become one of the most commonly used CPs in PNI repair due to its advantages of high conductivity and excellent biocompatibility. In this study, we combined different PPy concentrations with a chitosan (CS) temperature-sensitive hydrogel system containing decellularized nerve matrix (DNM) to construct the electrically conductive nerve conduits. We evaluated the physical and biological properties of four groups of nerve conduits. It was found that the PPy concentrations were proportional to the electrical conductivity of the nerve conduits. The mechanical properties of the nerve conduits increased with higher PPy concentrations but decreased when the PPy concentration was as high as 8%. Meanwhile, the co-blending of PPy and DNM gave the nerve conduit suitable degradation properties. Furthermore, in vitro cytotoxicity assay and live/dead assay demonstrated these conduits could support the adhesion and growth of cells. In summary, the electrically conductive nerve conduits with high conductivity, mechanical properties, biodegradation characteristics, and cytocompatibility had potential applications in the field of peripheral nerve regeneration.


Assuntos
Quitosana , Traumatismos dos Nervos Periféricos , Humanos , Polímeros , Hidrogéis , Pirróis , Regeneração Nervosa , Traumatismos dos Nervos Periféricos/terapia
20.
Bioact Mater ; 19: 581-593, 2023 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-35600980

RESUMO

Annulus fibrosus (AF) repair remains a challenge because of its limited self-healing ability. Endogenous repair strategies combining scaffolds and growth factors show great promise in AF repair. Although the unique and beneficial characteristics of decellularized extracellular matrix (ECM) in tissue repair have been demonstrated, the poor mechanical property of ECM hydrogels largely hinders their applications in tissue regeneration. In the present study, we combined polyethylene glycol diacrylate (PEGDA) and decellularized annulus fibrosus matrix (DAFM) to develop an injectable, photocurable hydrogel for AF repair. We found that the addition of PEGDA markedly improved the mechanical strength of DAFM hydrogels while maintaining their porous structure. Transforming growth factor-ß1 (TGF-ß1) was further incorporated into PEGDA/DAFM hydrogels, and it could be continuously released from the hydrogel. The in vitro experiments showed that TGF-ß1 facilitated the migration of AF cells. Furthermore, PEGDA/DAFM/TGF-ß1 hydrogels supported the adhesion, proliferation, and increased ECM production of AF cells. In vivo repair performance of the hydrogels was assessed using a rat AF defect model. The results showed that the implantation of PEGDA/DAFM/TGF-ß1 hydrogels effectively sealed the AF defect, prevented nucleus pulposus atrophy, retained disc height, and partially restored the biomechanical properties of disc. In addition, the implanted hydrogel was infiltrated by cells resembling AF cells and well integrated with adjacent AF tissue. In summary, findings from this study indicate that TGF-ß1-supplemented DAFM hydrogels hold promise for AF repair.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA