Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 50
Filter
1.
Sci Rep ; 14(1): 14127, 2024 Jun 19.
Article in English | MEDLINE | ID: mdl-38898063

ABSTRACT

Since conventional PID (Proportional-Integral-Derivative) controllers hardly control the robot to stabilize for constant force grinding under changing environmental conditions, it is necessary to add a compensation term to conventional PID controllers. An optimal parameter finding algorithm based on SAC (Soft-Actor-Critic) is proposed to solve the problem that the compensation term parameters are difficult to obtain, including training state action and normalization preprocessing, reward function design, and targeted deep neural network design. The algorithm is used to find the optimal controller compensation term parameters and applied to the PID controller to complete the compensation through the inverse kinematics of the robot to achieve constant force grinding control. To verify the algorithm's feasibility, a simulation model of a grinding robot with sensible force information is established, and the simulation results show that the controller trained with the algorithm can achieve constant force grinding of the robot. Finally, the robot constant force grinding experimental system platform is built for testing, which verifies the control effect of the optimal parameter finding algorithm on the robot constant force grinding and has specific environmental adaptability.

2.
Rheumatology (Oxford) ; 63(7): e206-e207, 2024 Jul 01.
Article in English | MEDLINE | ID: mdl-38648745
3.
Appl Opt ; 63(6): 1457-1470, 2024 Feb 20.
Article in English | MEDLINE | ID: mdl-38437357

ABSTRACT

Most near-eye displays with one fixed focal plane suffer from the vergence-accommodation conflict and cause visual discomfort to users. In contrast, light field displays can provide natural and comfortable 3D visual sensation to users without the conflict. This paper presents a near-eye light field display consisting of a geometric lightguide and a light field generator, along with a collimator to ensure the light rays propagating in the lightguide are collimated. Unlike most lightguides, which reduce thickness by employing total internal reflection that can easily generate stray light, our lightguide directly propagates light rays without total internal reflection. The partially reflective mirrors of the lightguide expand the exit pupil to achieve an eyebox of 13m m(h o r i z o n t a l)×6.5m m(v e r t i c a l) with an eye relief of 18 mm. The collimator and the light field generator, both having effective focal lengths different in the horizontal and vertical directions, are designed to provide a 40-deg diagonal field of view. The working range of the light field generator, which is 30 cm to infinity, is verified qualitatively and quantitatively by experiments. We optimize the illuminance uniformity and analyze the illuminance variation across the eyebox. Further, we minimize the ghost artifact (referring to the split-up of light fields replicated by the partially reflective mirrors) by orienting the partially reflective mirrors at slightly different angles to enhance the image quality for short-range applications such as medical surgery.

4.
J Nanobiotechnology ; 22(1): 74, 2024 Feb 23.
Article in English | MEDLINE | ID: mdl-38395929

ABSTRACT

Due to its unique structure, articular cartilage has limited abilities to undergo self-repair after injury. Additionally, the repair of articular cartilage after injury has always been a difficult problem in the field of sports medicine. Previous studies have shown that the therapeutic use of mesenchymal stem cells (MSCs) and their extracellular vesicles (EVs) has great potential for promoting cartilage repair. Recent studies have demonstrated that most transplanted stem cells undergo apoptosis in vivo, and the apoptotic EVs (ApoEVs) that are subsequently generated play crucial roles in tissue repair. Additionally, MSCs are known to exist under low-oxygen conditions in the physiological environment, and these hypoxic conditions can alter the functional and secretory properties of MSCs as well as their secretomes. This study aimed to investigate whether ApoEVs that are isolated from adipose-derived MSCs cultured under hypoxic conditions (hypoxic apoptotic EVs [H-ApoEVs]) exert greater effects on cartilage repair than those that are isolated from cells cultured under normoxic conditions. Through in vitro cell proliferation and migration experiments, we demonstrated that H-ApoEVs exerted enhanced effects on stem cell proliferation, stem cell migration, and bone marrow derived macrophages (BMDMs) M2 polarization compared to ApoEVs. Furthermore, we utilized a modified gelatine matrix/3D-printed extracellular matrix (ECM) scaffold complex as a carrier to deliver H-ApoEVs into the joint cavity, thus establishing a cartilage regeneration system. The 3D-printed ECM scaffold provided mechanical support and created a microenvironment that was conducive to cartilage regeneration, and the H-ApoEVs further enhanced the regenerative capacity of endogenous stem cells and the immunomodulatory microenvironment of the joint cavity; thus, this approach significantly promoted cartilage repair. In conclusion, this study confirmed that a ApoEVs delivery system based on a modified gelatine matrix/3D-printed ECM scaffold together with hypoxic preconditioning enhances the functionality of stem cell-derived ApoEVs and represents a promising approach for promoting cartilage regeneration.


Subject(s)
Cartilage, Articular , Extracellular Vesicles , Mesenchymal Stem Cells , Humans , Hydrogels , Tissue Scaffolds/chemistry , Gelatin , Stem Cells , Hypoxia
5.
Cell Prolif ; 57(6): e13605, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38282322

ABSTRACT

Clinicians and researchers have always faced challenges in performing surgery for rotator cuff tears (RCT) due to the intricate nature of the tendon-bone gradient and the limited long-term effectiveness. At the same time, the occurrence of an inflammatory microenvironment further aggravates tissue damage, which has a negative impact on the regeneration process of mesenchymal stem cells (MSCs) and eventually leads to the production of scar tissue. Tetrahedral framework nucleic acids (tFNAs), novel nanomaterials, have shown great potential in biomedicine due to their strong biocompatibility, excellent cellular internalisation ability, and unparalleled programmability. The objective of this research was to examine if tFNAs have a positive effect on regeneration after RCTs. Experiments conducted in a controlled environment demonstrated that tFNAs hindered the assembly of inflammasomes in macrophages, resulting in a decrease in the release of inflammatory factors. Next, tFNAs were shown to exert a protective effect on the osteogenic and chondrogenic differentiation of bone marrow MSCs under inflammatory conditions. The in vitro results also demonstrated the regulatory effect of tFNAs on tendon-related protein expression levels in tenocytes after inflammatory stimulation. Finally, intra-articular injection of tFNAs into a rat RCT model showed that tFNAs improved tendon-to-bone healing, suggesting that tFNAs may be promising tendon-to-bone protective agents for the treatment of RCTs.


Subject(s)
Mesenchymal Stem Cells , Rats, Sprague-Dawley , Rotator Cuff Injuries , Rotator Cuff Injuries/drug therapy , Rotator Cuff Injuries/surgery , Rotator Cuff Injuries/pathology , Animals , Rats , Mesenchymal Stem Cells/metabolism , Mesenchymal Stem Cells/drug effects , Nucleic Acids/pharmacology , Nucleic Acids/metabolism , Cell Differentiation/drug effects , Male , Osteogenesis/drug effects , Tendons/drug effects , Tendons/metabolism , Tendons/pathology , Bone and Bones/drug effects , Bone and Bones/metabolism , Rotator Cuff/surgery , Rotator Cuff/pathology , Chondrogenesis/drug effects , Wound Healing/drug effects
6.
IEEE Trans Med Imaging ; 43(3): 1060-1070, 2024 Mar.
Article in English | MEDLINE | ID: mdl-37874706

ABSTRACT

Semantic segmentation of basal cell carcinoma (BCC) from full-field optical coherence tomography (FF-OCT) images of human skin has received considerable attention in medical imaging. However, it is challenging for dermatopathologists to annotate the training data due to OCT's lack of color specificity. Very often, they are uncertain about the correctness of the annotations they made. In practice, annotations fraught with uncertainty profoundly impact the effectiveness of model training and hence the performance of BCC segmentation. To address this issue, we propose an approach to model training with uncertain annotations. The proposed approach includes a data selection strategy to mitigate the uncertainty of training data, a class expansion to consider sebaceous gland and hair follicle as additional classes to enhance the performance of BCC segmentation, and a self-supervised pre-training procedure to improve the initial weights of the segmentation model parameters. Furthermore, we develop three post-processing techniques to reduce the impact of speckle noise and image discontinuities on BCC segmentation. The mean Dice score of BCC of our model reaches 0.503±0.003, which, to the best of our knowledge, is the best performance to date for semantic segmentation of BCC from FF-OCT images.


Subject(s)
Carcinoma, Basal Cell , Skin Neoplasms , Humans , Skin Neoplasms/diagnostic imaging , Semantics , Uncertainty , Tomography, Optical Coherence/methods , Carcinoma, Basal Cell/diagnostic imaging , Carcinoma, Basal Cell/pathology , Image Processing, Computer-Assisted
7.
Regen Biomater ; 10: rbad085, 2023.
Article in English | MEDLINE | ID: mdl-37814675

ABSTRACT

The field of regenerative medicine faces a notable challenge in terms of the regeneration of articular cartilage. Without proper treatment, it can lead to osteoarthritis. Based on the research findings, human umbilical cord mesenchymal stem cells (hUMSCs) are considered an excellent choice for regenerating cartilage. However, there is still a lack of suitable biomaterials to control their ability to self-renew and differentiate. To address this issue, in this study using tetrahedral framework nucleic acids (tFNAs) as a new method in an in vitro culture setting to manage the behaviour of hUMSCs was proposed. Then, the influence of tFNAs on hUMSC proliferation, migration and chondrogenic differentiation was explored by combining bioinformatics methods. In addition, a variety of molecular biology techniques have been used to investigate deep molecular mechanisms. Relevant results demonstrated that tFNAs can affect the transcriptome and multiple signalling pathways of hUMSCs, among which the PI3K/Akt pathway is significantly activated. Furthermore, tFNAs can regulate the expression levels of multiple proteins (GSK3ß, RhoA and mTOR) downstream of the PI3K-Akt axis to further enhance cell proliferation, migration and hUMSC chondrogenic differentiation. tFNAs provide new insight into enhancing the chondrogenic potential of hUMSCs, which exhibits promising potential for future utilization within the domains of AC regeneration and clinical treatment.

8.
Comput Methods Programs Biomed ; 242: 107824, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37832427

ABSTRACT

Medical image-to-image translation is often difficult and of limited effectiveness due to the differences in image acquisition mechanisms and the diverse structure of biological tissues. This work presents an unpaired image translation model between in-vivo optical coherence tomography (OCT) and ex-vivo Hematoxylin and eosin (H&E) stained images without the need for image stacking, registration, post-processing, and annotation. The model can generate high-quality and highly accurate virtual medical images, and is robust and bidirectional. Our framework introduces random noise to (1) blur redundant features, (2) defend against self-adversarial attacks, (3) stabilize inverse conversion, and (4) mitigate the impact of OCT speckles. We also demonstrate that our model can be pre-trained and then fine-tuned using images from different OCT systems in just a few epochs. Qualitative and quantitative comparisons with traditional image-to-image translation models show the robustness of our proposed signal-to-noise ratio (SNR) cycle-consistency method.


Subject(s)
Algorithms , Image Processing, Computer-Assisted , Signal-To-Noise Ratio , Image Processing, Computer-Assisted/methods , Tomography, Optical Coherence/methods , Cell Nucleus
9.
ACS Appl Mater Interfaces ; 15(19): 22944-22958, 2023 May 17.
Article in English | MEDLINE | ID: mdl-37134259

ABSTRACT

The regeneration and reconstruction of articular cartilage (AC) after a defect are often difficult. The key to the treatment of AC defects lies in regeneration of the defect site and regulation of the inflammatory response. In this investigation, a bioactive multifunctional scaffold was formulated using the aptamer Apt19S as a mediator for mesenchymal stem cell (MSC)-specific recruitment and the enhancement of cellular chondrogenic and inflammatory regulation through the incorporation of Mg2+. Apt19S, which can recruit MSCs in vitro and in vivo, was chemically conjugated to a decellularized cartilage extracellular matrix (ECM)-lysed scaffold. The results from in vitro experiments using the resulting scaffold demonstrated that the inclusion of Mg2+ could stimulate not only the chondrogenic differentiation of synovial MSCs but also the increased polarization of macrophages toward the M2 phenotype. Additionally, Mg2+ inhibited NLRP3 inflammasome activation, thereby decreasing chondrocyte pyroptosis. Subsequently, Mg2+ was incorporated into the bioactive multifunctional scaffold, and the resulting scaffold promoted cartilage regeneration in vivo. In conclusion, this study confirms that the combination of Mg2+ and aptamer-functionalized ECM scaffolds is a promising strategy for AC regeneration based on in situ tissue engineering and early inflammatory regulation.


Subject(s)
Cartilage, Articular , Cartilage, Articular/physiology , Magnesium/pharmacology , Regeneration/physiology , Chondrocytes , Tissue Engineering/methods , Oligonucleotides , Chondrogenesis , Extracellular Matrix/metabolism , Ions/metabolism , Tissue Scaffolds
10.
Front Bioeng Biotechnol ; 11: 1115312, 2023.
Article in English | MEDLINE | ID: mdl-36890920

ABSTRACT

Tendon injuries often result in significant pain and disability and impose severe clinical and financial burdens on our society. Despite considerable achievements in the field of regenerative medicine in the past several decades, effective treatments remain a challenge due to the limited natural healing capacity of tendons caused by poor cell density and vascularization. The development of tissue engineering has provided more promising results in regenerating tendon-like tissues with compositional, structural and functional characteristics comparable to those of native tendon tissues. Tissue engineering is the discipline of regenerative medicine that aims to restore the physiological functions of tissues by using a combination of cells and materials, as well as suitable biochemical and physicochemical factors. In this review, following a discussion of tendon structure, injury and healing, we aim to elucidate the current strategies (biomaterials, scaffold fabrication techniques, cells, biological adjuncts, mechanical loading and bioreactors, and the role of macrophage polarization in tendon regeneration), challenges and future directions in the field of tendon tissue engineering.

11.
Molecules ; 28(4)2023 Feb 12.
Article in English | MEDLINE | ID: mdl-36838740

ABSTRACT

The effects of protein-glutaminase (PG) on calcium sulphate (CaSO4)-induced gels of soy protein isolate (SPI) with different heat treatment levels were investigated. The time-dependent degree of deamidation showed that the mild denaturation of the protein favored the deamidation. The particle size distribution showed that the heat treatment increased the SPI particle size, and the particle size distribution of the SPI shifted to the right or increased the proportion of the large particle size component as the degree of deamidation increased for each sample. Rheological analysis showed that the deamidation substantially pushed up the gel temperature and decreased the value of G'. The gel strength and water-holding capacity showed that the higher the amount of enzyme added, the more significant the decrease in gel strength, while the gel water-holding capacity increased. In summary, the deamidation of PG and heat treatment can affect the gel properties of SPI synergistically.


Subject(s)
Calcium Sulfate , Glutaminase , Glutaminase/metabolism , Soybean Proteins , Gels , Water , Rheology
12.
Biomater Res ; 27(1): 7, 2023 Feb 05.
Article in English | MEDLINE | ID: mdl-36739446

ABSTRACT

BACKGROUND: In recent years, there has been significant research progress on in situ articular cartilage (AC) tissue engineering with endogenous stem cells, which uses biological materials or bioactive factors to improve the regeneration microenvironment and recruit more endogenous stem cells from the joint cavity to the defect area to promote cartilage regeneration. METHOD: In this study, we used ECM alone as a bioink in low-temperature deposition manufacturing (LDM) 3D printing and then successfully fabricated a hierarchical porous ECM scaffold incorporating GDF-5. RESULTS: Comparative in vitro experiments showed that the 7% ECM scaffolds had the best biocompatibility. After the addition of GDF-5 protein, the ECM scaffolds significantly improved bone marrow mesenchymal stem cell (BMSC) migration and chondrogenic differentiation. Most importantly, the in vivo results showed that the ECM/GDF-5 scaffold significantly enhanced in situ cartilage repair. CONCLUSION: In conclusion, this study reports the construction of a new scaffold based on the concept of in situ regeneration, and we believe that our findings will provide a new treatment strategy for AC defect repair.

13.
Molecules ; 27(23)2022 Nov 25.
Article in English | MEDLINE | ID: mdl-36500320

ABSTRACT

This study investigated the effect of ultrasound on gel properties of soy protein isolates (SPIs) at different salt concentrations. The results showed that ultrasound could significantly improve the gel hardness and the water holding capacity (WHC) of the salt-containing gel (p < 0.05). The gel presents a uniform and compact three-dimensional network structure. The combination of 200 mM NaCl with 20 min of ultrasound could significantly increase the gel hardness (four times) and the WHC (p < 0.05) compared with the SPI gel without treatment. With the increase in NaCl concentration, the ζ potential and surface hydrophobicity increased, and the solubility decreased. Ultrasound could improve the protein solubility, compensate for the loss of solubility caused by the addition of NaCl, and further increase the surface hydrophobicity. Ultrasound combined with NaCl allowed proteins to form aggregates of different sizes. In addition, the combined treatment increased the hydrophobic interactions and disulfide bond interactions in the gel. Overall, ultrasound could improve the thermal gel properties of SPI gels with salt addition.


Subject(s)
Hot Temperature , Soybean Proteins , Soybean Proteins/chemistry , Gels/chemistry , Osmolar Concentration , Hydrophobic and Hydrophilic Interactions , Water/chemistry , Sodium Chloride
15.
Biomaterials ; 291: 121888, 2022 12.
Article in English | MEDLINE | ID: mdl-36403324

ABSTRACT

Inferior healing and peritendinous adhesions are the major clinical problems following Achilles tendon injury, leading to impaired motor function and an increased risk of re-rupture. These complications are presumed to be inextricably linked to inflammation and fibroscar formation. Here, microRNA29a is identified as a promising therapeutic target for tendon injury through the cross-regulation of the immune response and matrix remodeling. MiR29a-LNPs were successfully prepared by microfluidic technology. They are then loaded into the core-shell nanofibers to achieve local delivery in the injured tendon, where the shell layer is composed of PELA for anti-adhesion. Our studies reveal that miR29a regulates collagen synthesis and NF-κB activation in tenocytes, and promotes macrophage polarization by inhibiting the inflammasome pathway. In vivo studies of the Achilles tendon-rupture model indicate the best repair in the miR29a group, as evidenced by superior collagen composition and alignment, higher mechanical strength, and better functional recovery. In conclusion, a functionalized anti-adhesive membrane that promotes nascent tendon matrix remodeling and improves the regenerative immune microenvironment is developed for the treatment of tendon injury.


Subject(s)
Nanofibers , Tendon Injuries , Humans , Tendons , Tendon Injuries/therapy , Immunity
16.
Bioact Mater ; 9: 411-427, 2022 Mar.
Article in English | MEDLINE | ID: mdl-34820580

ABSTRACT

Many recent studies have shown that joint-resident mesenchymal stem cells (MSCs) play a vital role in articular cartilage (AC) in situ regeneration. Specifically, synovium-derived MSCs (SMSCs), which have strong chondrogenic differentiation potential, may be the main driver of cartilage repair. However, both the insufficient number of MSCs and the lack of an ideal regenerative microenvironment in the defect area will seriously affect the regeneration of AC. Tetrahedral framework nucleic acids (tFNAs), notable novel nanomaterials, are considered prospective biological regulators in biomedical engineering. Here, we aimed to explore whether tFNAs have positive effects on AC in situ regeneration and to investigate the related mechanism. The results of in vitro experiments showed that the proliferation and migration of SMSCs were significantly enhanced by tFNAs. In addition, tFNAs, which were added to chondrogenic induction medium, were shown to promote the chondrogenic capacity of SMSCs by increasing the phosphorylation of Smad2/3. In animal models, the injection of tFNAs improved the therapeutic outcome of cartilage defects compared with that of the control treatments without tFNAs. In conclusion, this is the first report to demonstrate that tFNAs can promote the chondrogenic differentiation of SMSCs in vitro and enhance AC regeneration in vivo, indicating that tFNAs may become a promising therapeutic for AC regeneration.

17.
Acta Biomater ; 140: 23-42, 2022 03 01.
Article in English | MEDLINE | ID: mdl-34896634

ABSTRACT

The ability of articular cartilage to repair itself is limited because it lacks blood vessels, nerves, and lymph tissue. Once damaged, it can lead to joint swelling and pain, accelerating the progression of osteoarthritis. To date, complete regeneration of hyaline cartilage exhibiting mechanical properties remains an elusive goal, despite the many available technologies. The inflammatory milieu created by cartilage damage is critical for chondrocyte death and hypertrophy, extracellular matrix breakdown, ectopic bone formation, and progression of cartilage injury to osteoarthritis. In the inflammatory microenvironment, mesenchymal stem cells (MSCs) undergo aberrant differentiation, and chondrocytes begin to convert or dedifferentiate into cells with a fibroblast phenotype, thereby resulting in fibrocartilage with poor mechanical qualities. All these factors suggest that inflammatory problems may be a major stumbling block to cartilage repair. To produce a milieu conducive to cartilage repair, multi-dimensional management of the joint inflammatory microenvironment in place and time is required. Therefore, this calls for elucidation of the immune microenvironment of cartilage repair after injury. This review provides a brief overview of: (1) the pathogenesis of cartilage injury; (2) immune cells in cartilage injury and repair; (3) effects of inflammatory cytokines on cartilage repair; (4) clinical strategies for treating cartilage defects; and (5) strategies for targeted immunoregulation in cartilage repair. STATEMENT OF SIGNIFICANCE: Immune response is increasingly considered the key factor affecting cartilage repair. It has both negative and positive regulatory effects on the process of regeneration and repair. Proinflammatory factors are secreted in large numbers, and necrotic cartilage is removed. During the repair period, immune cells can secrete anti-inflammatory factors and chondrogenic cytokines, which can inhibit inflammation and promote cartilage repair. However, inflammatory factors persist, which accelerate the degradation of the cartilage matrix. Furthermore, in an inflammatory microenvironment, MSCs undergo abnormal differentiation, and chondrocytes begin to transform or dedifferentiate into fibroblast-like cells, forming fibrocartilage with poor mechanical properties. Consequently, cartilage regeneration requires multi-dimensional regulation of the joint inflammatory microenvironment in space and time to make it conducive to cartilage regeneration.


Subject(s)
Cartilage Diseases , Cartilage, Articular , Osteoarthritis , Cartilage Diseases/metabolism , Cell Differentiation/genetics , Chondrocytes , Chondrogenesis , Humans , Osteoarthritis/pathology , Tissue Engineering
18.
Biomaterials ; 278: 121131, 2021 11.
Article in English | MEDLINE | ID: mdl-34543785

ABSTRACT

Articular cartilage (AC) injury repair has always been a difficult problem for clinicians and researchers. Recently, a promising therapy based on mesenchymal stem cells (MSCs) has been developed for the regeneration of cartilage defects. As endogenous articular stem cells, synovial MSCs (SMSCs) possess strong chondrogenic differentiation ability and articular specificity. In this study, a cartilage regenerative system was developed based on a chitosan (CS) hydrogel/3D-printed poly(ε-caprolactone) (PCL) hybrid containing SMSCs and recruiting tetrahedral framework nucleic acid (TFNA) injected into the articular cavity. TFNA, which is a promising DNA nanomaterial for improving the regenerative microenvironment, could be taken up into SMSCs and promoted the proliferation and chondrogenic differentiation of SMSCs. CS, as a cationic polysaccharide, can bind to DNA through electrostatic action and recruit free TFNA after articular cavity injection in vivo. The 3D-printed PCL scaffold provided basic mechanical support, and TFNA provided a good microenvironment for the proliferation and chondrogenic differentiation of the delivered SMSCs and promoted cartilage regeneration, thus greatly improving the repair of cartilage defects. In conclusion, this study confirmed that a CS hydrogel/3D-printed PCL hybrid scaffold containing SMSCs could be a promising strategy for cartilage regeneration based on chitosan-directed TFNA recruitment and TFNA-enhanced cell proliferation and chondrogenesis.


Subject(s)
Cartilage, Articular , Chitosan , Mesenchymal Stem Cells , Nucleic Acids , Cell Differentiation , Chondrogenesis , Hydrogels , Polyesters , Printing, Three-Dimensional , Regeneration , Tissue Engineering , Tissue Scaffolds
19.
Front Cell Dev Biol ; 9: 661802, 2021.
Article in English | MEDLINE | ID: mdl-34327197

ABSTRACT

Knee menisci are structurally complex components that preserve appropriate biomechanics of the knee. Meniscal tissue is susceptible to injury and cannot heal spontaneously from most pathologies, especially considering the limited regenerative capacity of the inner avascular region. Conventional clinical treatments span from conservative therapy to meniscus implantation, all with limitations. There have been advances in meniscal tissue engineering and regenerative medicine in terms of potential combinations of polymeric biomaterials, endogenous cells and stimuli, resulting in innovative strategies. Recently, polymeric scaffolds have provided researchers with a powerful instrument to rationally support the requirements for meniscal tissue regeneration, ranging from an ideal architecture to biocompatibility and bioactivity. However, multiple challenges involving the anisotropic structure, sophisticated regenerative process, and challenging healing environment of the meniscus still create barriers to clinical application. Advances in scaffold manufacturing technology, temporal regulation of molecular signaling and investigation of host immunoresponses to scaffolds in tissue engineering provide alternative strategies, and studies have shed light on this field. Accordingly, this review aims to summarize the current polymers used to fabricate meniscal scaffolds and their applications in vivo and in vitro to evaluate their potential utility in meniscal tissue engineering. Recent progress on combinations of two or more types of polymers is described, with a focus on advanced strategies associated with technologies and immune compatibility and tunability. Finally, we discuss the current challenges and future prospects for regenerating injured meniscal tissues.

20.
Front Bioeng Biotechnol ; 9: 662381, 2021.
Article in English | MEDLINE | ID: mdl-33996783

ABSTRACT

Meniscus tissue engineering (MTE) aims to fabricate ideal scaffolds to stimulate the microenvironment for recreating the damaged meniscal tissue. Indeed, favorable mechanical properties, suitable biocompatibility, and inherent chondrogenic capability are crucial in MTE. In this study, we present a composite scaffold by 3D printing a poly(ε-caprolactone) (PCL) scaffold as backbone, followed by injection with the meniscus extracellular matrix (MECM), and modification with kartogenin (KGN)-loaded poly(lactic-co-glycolic) acid (PLGA) microsphere (µS), which serves as a drug delivery system. Therefore, we propose a plan to improve meniscus regeneration via KGN released from the 3D porous PCL/MECM scaffold. The final results showed that the hydrophilicity and bioactivity of the resulting PCL/MECM scaffold were remarkably enhanced. In vitro synovium-derived mesenchymal stem cells (SMSCs) experiments suggested that introducing MECM components helped cell adhesion and proliferation and maintained promising ability to induce cell migration. Moreover, KGN-incorporating PLGA microspheres, which were loaded on scaffolds, showed a prolonged release profile and improved the chondrogenic differentiation of SMSCs during the 14-day culture. Particularly, the PCL/MECM-KGN µS seeded by SMSCs showed the highest secretion of total collagen and aggrecan. More importantly, the synergistic effect of the MECM and sustained release of KGN can endow the PCL/MECM-KGN µS scaffolds with not only excellent cell affinity and cell vitality preservation but also chondrogenic activity. Thus, the PCL/MECM-KGN µS scaffolds are expected to have good application prospects in the field of MTE.

SELECTION OF CITATIONS
SEARCH DETAIL