Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 37
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
País de afiliação
Intervalo de ano de publicação
1.
J Transl Med ; 20(1): 515, 2022 11 08.
Artigo em Inglês | MEDLINE | ID: mdl-36348497

RESUMO

Osteoarthritis (OA) is a chronic degenerative joint disease characterized by the destruction of the articular cartilage, sclerosis of the subchondral bone, and joint dysfunction. Its pathogenesis is attributed to direct damage and mechanical destruction of joint tissues. Mesenchymal stem cells (MSCs), suggested as a potential strategy for the treatment of OA, have shown therapeutic effects on OA. However, the specific fate of MSCs after intraarticular injection, including cell attachment, proliferation, differentiation, and death, is still unclear, and there is no guarantee that stem cells can be retained in the cartilage tissue to enact repair. Direct homing of MSCs is an important determinant of the efficacy of MSC-based cartilage repair. Recent studies have revealed that the unique homing capacity of MSCs and targeted modification can improve their ability to promote tissue regeneration. Here, we comprehensively review the homing effect of stem cells in joints and highlight progress toward the targeted modification of MSCs. In the future, developments of this targeting system that accelerate tissue regeneration will benefit targeted tissue repair.


Assuntos
Cartilagem Articular , Transplante de Células-Tronco Mesenquimais , Células-Tronco Mesenquimais , Osteoartrite , Humanos , Cartilagem Articular/patologia , Osteoartrite/terapia , Osteoartrite/patologia , Diferenciação Celular
2.
J Cell Physiol ; 232(7): 1708-1716, 2017 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-27404036

RESUMO

DNA methylation has emerged as a crucial regulator of chondrocyte dedifferentiation, which severely compromises the outcome of autologous chondrocyte implantation (ACI) treatment for cartilage defects. However, the full-scale DNA methylation profiling in chondrocyte dedifferentiation remains to be determined. Here, we performed a genome-wide DNA methylation profiling of dedifferentiated chondrocytes in monolayer culture and chondrocytes treated with DNA methylation inhibitor 5-azacytidine (5-AzaC). This research revealed that the general methylation level of CpG was increased while the COL-1A1 promoter methylation level was decreased during the chondrocyte dedifferentiation. 5-AzaC could reduce general methylation levels and reverse the chondrocyte dedifferentiation. Surprisingly, the DNA methylation level of COL-1A1 promoter was increased after 5-AzaC treatment. The COL-1A1 expression level was increased while that of SOX-9 was decreased during the chondrocyte dedifferentiation. 5-AzaC treatment up-regulated the SOX-9 expression while down-regulated the COL-1A1 promoter activity and gene expression. Taken together, these results suggested that differential regulation of the DNA methylation level of cartilage-specific genes might contribute to the chondrocyte dedifferentiation. Thus, the epigenetic manipulation of these genes could be a potential strategy to counteract the chondrocyte dedifferentiation accompanying in vitro propagation. J. Cell. Physiol. 232: 1708-1716, 2017. © 2016 Wiley Periodicals, Inc.


Assuntos
Diferenciação Celular/genética , Metilação de DNA/genética , Perfilação da Expressão Gênica , Adulto , Azacitidina/farmacologia , Desdiferenciação Celular/efeitos dos fármacos , Desdiferenciação Celular/genética , Diferenciação Celular/efeitos dos fármacos , Condrócitos/citologia , Condrócitos/efeitos dos fármacos , Condrócitos/metabolismo , Colágeno Tipo I/metabolismo , Ilhas de CpG/genética , Metilação de DNA/efeitos dos fármacos , Regulação para Baixo/efeitos dos fármacos , Regulação para Baixo/genética , Feminino , Ontologia Genética , Humanos , Masculino , Fenótipo , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Fatores de Transcrição SOX9/metabolismo , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/genética , Regulação para Cima/efeitos dos fármacos , Regulação para Cima/genética
3.
Saudi Pharm J ; 25(4): 575-579, 2017 May.
Artigo em Inglês | MEDLINE | ID: mdl-28579894

RESUMO

This study was conducted to investigate the effect of magnetic nanoparticle composite scaffold under a pulsed electromagnetic field on bone marrow mesenchymal stem cells (BMSCs), which was achieved by examining the biological behaviors of cell adhesion, proliferation and differentiation on the surface of the scaffolds. This may provide some experimental evidence for the use of magnetic nanoparticles in medical application. The magnetic nanoparticle composite scaffolds were evaluated and characterized by the following indexes: the cell proliferation was detected by the CCK-8 method, the alkaline phosphatase (ALP) activity was examined by a detection kit, and the expression of type I collagen and osteocalcin gene were evaluated by RT-PCR. The CCK-8 test showed that there was no significant difference in Group A (BMSCs-seeded magnetic scaffolds under the electromagnetic field), B (BMSCs-seeded magnetic scaffolds) and C (BMSCs cultured alone) (P > 0.05). The value for the ALP activity in Group A was higher than the other two groups. In addition, the RT-PCR results showed that the expression of type I collagen gene in Group A was enhanced (P < 0.05), suggesting that the magnetic nanoparticles combined with the pulsed electromagnetic field had a positive effect on the osteogenic differentiation of BMSCs. However, the expression of osteocalcin was not significantly different in three groups (P > 0.05). To conclude, magnetic nanoparticles may induce the osteogenic differentiation with the action of the pulsed electromagnetic field.

4.
Biosens Bioelectron ; 259: 116380, 2024 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-38754193

RESUMO

Exosomes, as novel biomarker for liquid biopsy, exhibit huge important potential value for cancer diagnosis. However, various proteins show different expression levels on exosomal membrane, and the absolute concentration of exosomes in clinical samples is easily influenced by a number of factors. Here, we developed a CRISPR/Cas12a and aptamer-chemiluminescence based analysis (CACBA) for the relative abundance determination of tumor-related protein positive exosomes in plasma for breast cancer diagnosis. The total concentration of exosomes was determined through captured CD63 using a CRISPR/Cas12a-based method with the LoD of 8.97 × 103 particles/µl. Meanwhile, EpCAM and MUC1 positive exosomes were quantitatively detected by aptamer-chemiluminescence (ACL) based method with the LoD of 1.45 × 102 and 3.73 × 102 particles/µl, respectively. It showed that the percentages of EpCAM and MUC1 positive exosomes offered an excellent capability to differentiate breast cancer patients and healthy donors. The high sensitivity, strong specificity, outstanding anti-interference capability, and steady recovery rate of this approach offered higher accuracy and robustness than the commercialized method in clinical trial. In addition with good stability, easy preparation and low cost, this method not only provides a new approach to rapid analysis of exosome proteins, it may be quickly extended to the diagnoses of various cancers.


Assuntos
Aptâmeros de Nucleotídeos , Biomarcadores Tumorais , Técnicas Biossensoriais , Neoplasias da Mama , Sistemas CRISPR-Cas , Molécula de Adesão da Célula Epitelial , Exossomos , Mucina-1 , Humanos , Neoplasias da Mama/diagnóstico , Neoplasias da Mama/sangue , Neoplasias da Mama/genética , Exossomos/química , Exossomos/genética , Feminino , Aptâmeros de Nucleotídeos/química , Técnicas Biossensoriais/métodos , Mucina-1/sangue , Mucina-1/genética , Mucina-1/análise , Biomarcadores Tumorais/sangue , Biomarcadores Tumorais/genética , Molécula de Adesão da Célula Epitelial/genética , Medições Luminescentes/métodos , Tetraspanina 30 , Limite de Detecção
5.
Biomed Pharmacother ; 168: 115715, 2023 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-37857246

RESUMO

Osteoarthritis (OA) is a challenging joint inflammatory disease that often leads to disability. Immunoregulatory Exosomes (Exos) have shown promise in OA and cartilage degeneration treatment. Engineering Exos to deliver therapeutic agents like Kartogenin (KGN) has displayed potential for restoring cartilage regeneration. However, challenges include the uneven distribution of Exos at the injury site and the release of Exos cargo out of chondrocytes. Hydrogel-loaded uMSC-Exo has demonstrated significant therapeutic effects in wound healing and tissue regeneration. Recently, a new version of three-dimensional (3D) bioprinting of hydrogel significantly restored cartilage regeneration in OA joints. Combining immune regulatory Exos with 3D bioprinting hydrogel (3D-BPH-Exos) holds the potential for immunomodulating cartilage tissue and treatment of OA. It can reduce intracellular inflammasome formation and the release of inflammatory agents like IL-1ß, TNF-α, and INF-γ, while also preventing chondrocyte apoptosis by restoring mitochondrial functions and enhancing chondrogenesis in synovial MSCs, osteoprogenitor cells, and osteoclasts. Loading Exos with chondrogenic stimuli agents in the 3D-BPH-Exos approach may offer a faster and safer strategy for cartilage repair while better inhibiting joint inflammation than high doses of anti-inflammatory drugs and cell-based therapies. This review provides a comprehensive overview of hydrogel bioprinting and exosome-based therapy in OA. It emphasizes the potential of 3D-BPH-Exos loaded with chondrogenic stimuli agents for OA treatment, serving as a basis for further research.


Assuntos
Bioimpressão , Exossomos , Células-Tronco Mesenquimais , Osteoartrite , Humanos , Condrogênese , Hidrogéis/farmacologia , Osteoartrite/tratamento farmacológico , Cartilagem , Condrócitos
6.
Oncol Lett ; 26(4): 454, 2023 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-37720673

RESUMO

[This retracts the article DOI: 10.3892/ol.2018.7994.].

7.
Front Bioeng Biotechnol ; 10: 943712, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36466335

RESUMO

Imaging of extracellular vesicles (EVs) will facilitate a better understanding of their biological functions and their potential as therapeutics and drug delivery vehicles. In order to clarify EV-mediated cellular communication in vitro and to track the bio-distribution of EV in vivo, various strategies have been developed to label and image EVs. In this review, we summarized recent advances in the tracking of EVs, demonstrating the methods for labeling and imaging of EVs, in which the labeling methods include direct and indirect labeling and the imaging modalities include fluorescent imaging, bioluminescent imaging, nuclear imaging, and nanoparticle-assisted imaging. These techniques help us better understand the mechanism of uptake, the bio-distribution, and the function of EVs. More importantly, we can evaluate the pharmacokinetic properties of EVs, which will help promote their further clinical application.

8.
Eur J Med Res ; 27(1): 288, 2022 Dec 12.
Artigo em Inglês | MEDLINE | ID: mdl-36503684

RESUMO

Osteoarthritis (OA) is a common chronic disabling disease that affects hundreds of millions of people around the world. The most important pathological feature is the rupture and loss of articular cartilage, and the characteristics of avascular joint tissues lead to limited repair ability. Currently, there is no effective treatment to prevent cartilage degeneration. Studies on the mechanism of cartilage metabolism revealed that hypoxia-inducible factors (HIFs) are key regulatory genes that maintain the balance of cartilage catabolism-matrix anabolism and are considered to be the major OA regulator and promising OA treatment target. Although the exact mechanism of HIFs in OA needs to be further clarified, many drugs that directly or indirectly act on HIF signaling pathways have been confirmed by animal experiments and regarded as promising treatments for OA. Targeting HIFs will provide a promising strategy for the development of new OA drugs. This article reviews the regulation of HIFs on intra-articular cartilage homeostasis and its influence on the progression of osteoarthritis and summarizes the recent advances in OA therapies targeting the HIF system.


Assuntos
Cartilagem Articular , Osteoartrite , Animais , Osteoartrite/tratamento farmacológico , Condrogênese , Hipóxia
9.
Gels ; 8(2)2022 Feb 21.
Artigo em Inglês | MEDLINE | ID: mdl-35200519

RESUMO

Tissue engineering is becoming an effective strategy for repairing cartilage damage. Synthesized nanocomposite hydrogels mimic the structure of natural cartilage extracellular matrices (ECMs), are biocompatible, and exhibit nano-bio effects in response to external stimuli. These inherent characteristics make nanocomposite hydrogels promising scaffold materials for cartilage tissue engineering. This review summarizes the advances made in the field of nanocomposite hydrogels for artificial cartilage. We discuss, in detail, their preparation methods and scope of application. The challenges involved for the application of hydrogel nanocomposites for cartilage repair are also highlighted.

10.
Biomed Pharmacother ; 148: 112782, 2022 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-35248846

RESUMO

Recently, cartilage tissue engineering has become a cornerstone to treat cartilage degeneration and osteoarthritis (OA). Fibronectin1 (FN1) is described as multiple functional glycoproteins that play an essential role in chondrogenic and osteogenic differentiation. Few studies reported the potential of FN1 to enhance tissue engineering and reduce the death of chondrocytes in OA. Further, FN1 possesses multiple binding domains including collagen, integrin, and heparin that can interact with heparan sulfate proteoglycans at the surface of chondrocyte leading to promote cell signaling and differentiation. Recent studies suggested that FN1 can promote chondrocyte differentiation by upregulating TGF-ß/PI3K/Akt pathways. Further, FN1 can inhibit the apoptosis of chondrocytes by preventing the release of metalloproteinases through lowering the expression of p-PI3K/PI3K and p-AKT/AKT pathways. However, the use of FN1 in cartilage repair studies using animal models or clinical trials was rarely reported. Therefore, this article provides new insights into the importance of FN1 in cartilage tissue engineering to encourage more studies concerning FN1 in cartilage repair studies. Further, we provided new suggestions for advanced applications of FN1 to treat OA and cartilage degeneration.


Assuntos
Cartilagem/metabolismo , Condrócitos/metabolismo , Fibronectinas/metabolismo , Engenharia Tecidual/métodos , Animais , Apoptose/fisiologia , Técnicas de Silenciamento de Genes , Humanos , Células-Tronco Mesenquimais/metabolismo , Metaloproteases/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Fator de Crescimento Transformador beta/metabolismo
11.
Int J Bioprint ; 8(2): 517, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35669327

RESUMO

Artificial skins are biomaterials that can replace the lost skin or promote the regeneration of damaged skin. Skin regenerative biomaterials are highly applauded because they can exempt patients with severe burns from the painful procedure of autologous skin transplantation. Notwithstanding decades of research, biocompatible, degradable, and printable biomaterials that can effectively promote skin regeneration as a transplantation replacement in clinical use are still scarce. Here, we report one type of all-protein hydrogel material as the product of the enzymatic crosslinking reaction of gelatin and a recombinant type III collagen (rColIII) protein. Doping the rColIII protein in gelatin reduces the inflammatory response as an implant underneath the skin. The all-protein hydrogel can be bioprinted as scaffolds to support the growth and proliferation of 3T3 fibroblast cells. The hydrogel used as a wound dressing promotes wound healing in a rat model of skin damage, showing a faster and healthier recovery than the controls. The rColIII protein in the hydrogel has been shown to play a critical role in skin regeneration. Altogether, this work manifests the development of all-protein gelatin-rColIII hydrogel and demonstrates its use in wound healing. The gelatin-collagen hydrogel wound dressing thereby may become a promising treatment of severe wounds in the future.

12.
Stem Cells Int ; 2021: 1285087, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34567129

RESUMO

Wound healing and regeneration are a dynamic and complex process that requires a collaborative effort between growth factors, epidermal cells, dermal cells, extracellular matrix, and vessels local to the wound area. Mesenchymal stem cells participate in the recruitment site, mainly by releasing secretory factors and matrix proteins to promote wound healing. Stem cell-derived nanovesicles (CDNs), including microvesicles, exosomes, and exosome mimetics, contain most of the biologically active substances of their parent cells and have similar effects. CDNs can shuttle various proteins, messenger RNAs, and microRNAs to regulate the activity of receptor cells, and they play important roles in skin wound healing. This article reviews recent research progress on CDNs for wound repair. We summarize current knowledge on how CDNs regulate immunity, fibroblast activity, angiogenesis, and scar formation in the wound healing process. This review can help researchers explore new treatment strategies to enhance the therapeutic efficacy of CDNs, which have a promising future as naturally cell-free therapies.

13.
Gels ; 7(3)2021 Sep 16.
Artigo em Inglês | MEDLINE | ID: mdl-34563030

RESUMO

Three-dimensional (3D) bioprinting is an emerging technology based on 3D digital imaging technology and multi-level continuous printing. The precise positioning of biological materials, seed cells, and biological factors, known as "additive biomanufacturing", can provide personalized therapy strategies in regenerative medicine. Over the last two decades, 3D bioprinting hydrogels have significantly advanced the field of cartilage and bone tissue engineering. This article reviews the development of 3D bioprinting and its application in cartilage tissue engineering, followed by a discussion of the current challenges and prospects for 3D bioprinting. This review presents foundational information on the future optimization of the design and manufacturing process of 3D additive biomanufacturing.

14.
Nanoscale ; 13(19): 8740-8750, 2021 May 20.
Artigo em Inglês | MEDLINE | ID: mdl-33969373

RESUMO

With the development of regenerative medicine, tissue repair at the molecular, cellular, tissue, and organ level has seen continuous improvements over traditional techniques. As the core of tissue repair, seed cells are widely used in various fields of regenerative medicine. However, their use is still associated with problems such as decreased cell survival and regeneration capacity after transplantation, immune rejection, and ethical concerns. Therefore, it is difficult to universally and safely apply stem cell banks for regenerative medicine. The paracrine effects of cells, especially secretion of exosomes, play vital roles in cell communication, immune response, angiogenesis, scar formation, tissue repair, and other biological functions. Exosomes are a type of nanoscale extracellular vesicle that contain biologically active molecules such as RNA and proteins; therefore, exosomes can replicate the functions of their parental cells. Meanwhile, exosomes can be used as nanocarriers to deliver active factors or small molecules to promote tissue repair. Preclinical studies of exosomes in tissue engineering and regenerative medicine have been carried in the fields of bone/cartilage repair, nerve regeneration, liver and kidney regeneration, skin repair, vascular tissue regeneration, etc. This review introduces exosomes from the aspects of biogenesis, composition, identification, and isolation, and focuses on the development status of scaffold materials for exosome delivery. In addition, we highlight examples of exosome-laden scaffolds for preclinical applications in tissue repair. We look forward to the broad application prospects of exosome-laden scaffolds.


Assuntos
Exossomos , Células-Tronco Mesenquimais , Rim , Regeneração , Medicina Regenerativa , Engenharia Tecidual , Alicerces Teciduais
15.
Biomater Sci ; 9(7): 2620-2630, 2021 Apr 07.
Artigo em Inglês | MEDLINE | ID: mdl-33595025

RESUMO

Acute injury of the articular cartilage can lead to chronic disabling conditions because of the limited self-repair capability of the cartilage. Implantation of stem cells at the injury site is a viable treatment, but requires a scaffold with a precisely controlled geometry and porosity in the 3D space, high biocompatibility, and the capability of promoting chondrogenic differentiation of the implanted stem cells. Here we report the development of gelatin/hydroxyapatite (HAP) hybrid materials by microextrusion 3D bioprinting and enzymatic cross-linking as the scaffold for human umbilical cord blood-derived mesenchymal stem cells (hUCB-MSCs). The scaffold supports the adhesion, growth, and proliferation of hUCB-MSCs and induces their chondrogenic differentiation in vitro. Doping HAP in the gelatin scaffold increases the fluidity of the hydrogel, improves the gelation kinetics and the rheological properties, and allows better control over 3D printing. Implanting the hUCB-MSC-laden scaffold at the injury site of the articular cartilage effectively repairs the cartilage defects in a pig model. Altogether, this work demonstrates the 3D printing of gelatin-based scaffold materials for hUCB-MSCs to repair cartilage defects as a potential treatment of articular cartilage injury.


Assuntos
Cartilagem Articular , Células-Tronco Mesenquimais , Animais , Diferenciação Celular , Condrogênese , Durapatita , Gelatina , Impressão Tridimensional , Células-Tronco , Suínos , Engenharia Tecidual , Alicerces Teciduais
16.
ACS Omega ; 5(17): 9733-9743, 2020 May 05.
Artigo em Inglês | MEDLINE | ID: mdl-32391460

RESUMO

To provide a novel approach for the clinical treatment of cartilage tissue defects, we prepared a new type of magnetic nanocomposite hydrogel with an optimal raw material ratio using Fe3O4, polyvinyl alcohol (PVA), and type-II collagen (COLII). Briefly, five groups of PVA and collagen hydrogel matrices with different mass ratios were prepared by a combination of repeated thawing cycles and foam-frozen ice crystal separation methods. Microscopic characterization was conducted using electron microscopy, and the biomechanical properties of each group of hydrogels were then tested. The highest performing component hydrogel matrix was selected after which Fe3O4 with different mass ratios was introduced to construct a new Fe3O4/PVA/COLII hydrogel. The prepared composite hydrogels were also microscopically characterized using electron microscopy along with scanning, measurements for porosity and moisture content, and biomechanical, infrared spectrum and degradation performance testing. CCK-8 detection and staining to determine the amount of living and dead cells were also performed. Collectively, these results showed that PVA/COLII,95:5 was the optimal hydrogel matrix. Using this hydrogel matrix, five groups of composite hydrogels with different Fe3O4 mass ratios were then prepared. There was no significant difference in the microscopic characteristics between these different hydrogels. Fe3O4/PVA/COLII,5:95:5 had better physical properties as well as swelling performance and cell compatibility. The PVA/COLII,95:5 hydrogel matrix was determined to be the best, while the new magnetic nanocomposite hydrogel Fe3O4/PVA/COLII,5:95:5 had good, comprehensive properties.

17.
RSC Adv ; 10(1): 541-550, 2019 Dec 20.
Artigo em Inglês | MEDLINE | ID: mdl-35492543

RESUMO

Hydrogel is an important scaffold material in regenerative medicine and cartilage tissue engineering. Hydrogel material combined with pulse electromagnetic fields (PEMFs), PEMFs has the potential to manage the repair of defective articular cartilage. Here, we developed a new type of magnetic hydrogel. The data shows that the magnetic hydrogel had good mechanical properties, and its surface had micropores and unevenness, which was conducive to cell adhesion growth. Infrared spectroscopy analysis showed that the magnetic particles were evenly distributed in the hydrogel, and the addition of constant static magnetic field yielded magnetic water. The hydrogel exhibited good superparamagnetism. The co-culture of the magnetic hydrogel and bone marrow mesenchymal stem cells (BMSCs) showed good biocompatibility. The PEMFs promoted the differentiation of the BMSCs into cartilage, and the index of cartilage differentiation increased obviously. The results of the animal experiments showed that the magnetic hydrogel and BMSCs combined with pulsed electromagnetic field had a strong repair effect. They also showed that the magnetic nano-hydrogel combined with the PEMFs induced chondrogenic differentiation of the BMSCs. The positive experimental results suggested that the combination of magnetic hydrogel and the PEMFs can be used as an effective method for repairing articular cartilage defects in rabbit model.

18.
J Drug Target ; 27(1): 28-32, 2019 01.
Artigo em Inglês | MEDLINE | ID: mdl-29772932

RESUMO

Either osteoarthritis or sports-related injuries can lead to cartilage defects, whereas both chondrocyte self-renewal and conventional treatments face limitations. In cartilage regenerative medicine, growth factors are commonly used to induce chondrogenic differentiation of stem cells. However, application of growth factors is confined by some drawbacks. Emerging small molecules are regarded as an alternative for cartilage regeneration. A recently discovered small-molecule compound, kartogenin (KGN), has been proven to be a chondrogenic and chondroprotective agent and is more effective in inducing cartilage regeneration when compared with growth factors. KGN has been processed and applied in many forms, such as in intra-articular injection, in collaboration with growth factors, in incorporation in drug delivery systems, and in combination with scaffolds. Fortunately, progress has been achieved in KGN applications. The current review discusses the recent advances in KGN for cartilage regeneration and thus presents new concepts in cartilage repair in clinical settings.


Assuntos
Anilidas/administração & dosagem , Cartilagem/efeitos dos fármacos , Condrócitos/citologia , Ácidos Ftálicos/administração & dosagem , Anilidas/farmacologia , Animais , Traumatismos em Atletas/tratamento farmacológico , Traumatismos em Atletas/fisiopatologia , Cartilagem/lesões , Cartilagem/patologia , Diferenciação Celular/efeitos dos fármacos , Condrogênese/efeitos dos fármacos , Sistemas de Liberação de Medicamentos , Humanos , Osteoartrite/tratamento farmacológico , Osteoartrite/fisiopatologia , Ácidos Ftálicos/farmacologia , Regeneração/efeitos dos fármacos
19.
Mater Sci Eng C Mater Biol Appl ; 99: 541-551, 2019 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-30889728

RESUMO

The regeneration of hyaline articular cartilage remains a major challenge due to the limited potential for cartilage to self-repair. Mesenchymal stem cell and hydrogel scaffold-based cartilage tissue engineering is a promising technique for articular cartilage therapy. The purpose of this study was to investigate the use of rabbit synovial fluid mesenchymal stem cells (rbSF-MSCs) encapsulated in an injectable chitosan-based hydrogel to repair full-thickness cartilage defects in femoral patellar grooves in rabbits. The rbSF-MSCs were obtained from rabbit synovial fluid and the surface markers of rbSF-MSCs were coincidental to the identification criteria of MSCs according to flow cytometry. The rbSF-MSCs were able to differentiate into osteogenic, adipogenic and chondrogenic lineages. In the present study, rbSF-MSCs encapsulated in glycol chitosan (GC) and benzaldehyde capped poly (ethylene oxide) (OHC-PEO-CHO) hydrogel were introduced into rabbits to repair articular cartilage defects. The modulus of the hydrogel could be regulated by the concentrations of GC and OHC-PEO-CHO and the hydrogel has a good biocompatibility to rbSF-MSCs. Assessment of in vivo repair indicates using hydrogel/rbSF-MSCs was superior to using the hydrogel scaffold only and the untreated control based on gross appearance and histological grading and evaluation. These preliminary findings suggest using the injectable chitosan-based hydrogel as a scaffold and rbSF-MSCs as seed cells is an alternative for tissue engineering of in vivo treatments for cartilage defects and these rbSF-MSCs allografts may be promising for use in clinical applications.


Assuntos
Cartilagem Articular/patologia , Quitosana/farmacologia , Hidrogéis/farmacologia , Injeções , Transplante de Células-Tronco Mesenquimais , Células-Tronco Mesenquimais/citologia , Líquido Sinovial/citologia , Cicatrização , Animais , Diferenciação Celular/efeitos dos fármacos , Linhagem da Célula/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Modelos Animais de Doenças , Módulo de Elasticidade , Feminino , Células-Tronco Mesenquimais/efeitos dos fármacos , Coelhos , Reologia , Cicatrização/efeitos dos fármacos
20.
ACS Biomater Sci Eng ; 5(5): 2200-2207, 2019 May 13.
Artigo em Inglês | MEDLINE | ID: mdl-33405772

RESUMO

Pulsed electromagnetic field therapy, or pulsed signal therapy, has shown efficacy in treating many illnesses, including knee osteoarthritis. Although the mechanism is not fully understood, magnetic therapy is broadly welcomed because of its safe and noninvasive nature. At the cellular and molecular level, remote control of the cell fate by the magnetic field also has profound applications in both basic science and translational research. Here we demonstrate the use of pulsed electromagnetic field, one of the most benign and noninvasive extracellular cues, as a novel method to control specific chondrogenic differentiation of mesenchymal stem cells (MSCs). Chondrogenesis of transplanted MSCs inside the joint is considered one of the future therapies to rebuild the damaged cartilage. Here we show that pulsed electromagnetic field promotes chondrogenic differentiation of MSCs, and such a promoting effect can be drastically enhanced by the combined use of a magnetic hydrogel as the cell growth matrix. The magnetic hydrogel, synthesized by chemical cross-linking of gelatin and ß-cyclodextrin and by embedding Fe3O4 magnetic nanoparticles in the hydrogel network, supports adhesion, growth, and proliferation of MSCs. Pulsed electromagnetic field boosts chondrogenesis of MSCs grown on the magnetic hydrogel, manifested by enhanced toluidine blue staining; higher expression of collagen II protein; and upregulation of collagen II, aggrecan, and SOX9 genes. Therefore, our work presents a robust method for chondrogenesis of MSCs using magnetic field as the external cue.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA