Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
Add more filters










Publication year range
1.
Bioact Mater ; 37: 424-438, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38689661

ABSTRACT

Bone nonunion poses an urgent clinical challenge that needs to be addressed. Recent studies have revealed that the metabolic microenvironment plays a vital role in fracture healing. Macrophages and bone marrow-derived mesenchymal stromal cells (BMSCs) are important targets for therapeutic interventions in bone fractures. Itaconate is a TCA cycle metabolite that has emerged as a potent macrophage immunomodulator that limits the inflammatory response. During osteogenic differentiation, BMSCs tend to undergo aerobic glycolysis and metabolize glucose to lactate. Copper ion (Cu2+) is an essential trace element that participates in glucose metabolism and may stimulate glycolysis in BMSCs and promote osteogenesis. In this study, we develop a 4-octyl itaconate (4-OI)@Cu@Gel nanocomposite hydrogel that can effectively deliver and release 4-OI and Cu2+ to modulate the metabolic microenvironment and improve the functions of cells involved in the fracture healing process. The findings reveal that burst release of 4-OI reduces the inflammatory response, promotes M2 macrophage polarization, and alleviates oxidative stress, while sustained release of Cu2+ stimulates BMSC glycolysis and osteogenic differentiation and enhances endothelial cell angiogenesis. Consequently, the 4-OI@Cu@Gel system achieves rapid fracture healing in mice. Thus, this study proposes a promising regenerative strategy to expedite bone fracture healing through metabolic reprogramming of macrophages and BMSCs.

2.
Front Bioeng Biotechnol ; 11: 1308184, 2023.
Article in English | MEDLINE | ID: mdl-38026853

ABSTRACT

The regeneration of skin tissue is often impeded by bacterial infection seriously. At the same time, reactive oxygen species (ROS) are often overexpressed in infected skin wounds, causing persistent inflammation that further hinders the skin repair process. All of these make the treatment of infected wounds is still a great challenge in clinic. In this study, we fabricate Cu(II)@MXene photothermal complex based on electrostatic self-assembly between Cu2+ and MXene, which are then introduced into a hyaluronic acid (HA) hydrogel to form an antibacterial dressing. The rapid adhesion, self-healing, and injectability of the dressing allows the hydrogel to be easily applied to different wound shapes and to provide long-term wound protection. More importantly, this easily prepared Cu(II)@MXene complex can act as a photothermal antibacterial barrier, ROS scavenger and angiogenesis promoter simultaneously to accelerate the healing rate of infected wounds. Our in vivo experiments strongly proved that the inflammatory condition, collagen deposition, vessel formation, and the final wound closure area were all improved by the application of Cu(II)@MXene photothermal hydrogel dressing.

3.
ACS Nano ; 17(17): 17199-17216, 2023 09 12.
Article in English | MEDLINE | ID: mdl-37624642

ABSTRACT

The clinical applications of currently used photosensitizers are limited by high costs, inconvenient preparation, suboptimal biodegradability, and a lack of biological activity. Humic acids (HAs) show photothermal activity and can be used as a photosensitizer for photothermal therapy. In the presence of various functional groups, HAs are endowed with anti-inflammatory and antioxidant activities. The solubility of HAs is dependent on the pH value, which is soluble in neutral to alkaline conditions and undergoes a conformational change to a coiled and compact structure in acidic conditions. Additionally, Cu2+ is an emerging therapeutic agent for cutaneous wounds and can be chelated by HAs to form complexes. In this study, we explore the ability of HAs to modulate the inflammatory response, particularly macrophage polarization, and the potential underlying mechanism. We fabricate a near-infrared (NIR)/pH dual-responsive Cu-HAs nanoparticle (NP)-based poly(vinyl alcohol) (PVA) hydrogel film loaded with SEW2871 (SEW), a macrophage recruitment agent, to treat bacteria-infected cutaneous wounds. The results show that HAs could promote M2 macrophage polarization in a dose-dependent manner. The Cu-HAs NPs successfully eradicated bacterial infection through NIR-induced local hyperthermia. This PVA@Cu-HAs NPs@SEW hydrogel film improves tissue regeneration by promoting M2 macrophage polarization, alleviating oxidative stress, enhancing angiogenesis, and facilitating collagen deposition. These findings highlight the therapeutic potential of PVA@Cu-HAs NPs@SEW hydrogel film for the treatment of bacterially infected cutaneous wound healing.


Subject(s)
Copper , Humic Substances , Hydrogels/pharmacology , Bacteria , Hydrogen-Ion Concentration
4.
Biomater Res ; 27(1): 76, 2023 Aug 04.
Article in English | MEDLINE | ID: mdl-37542353

ABSTRACT

Various joint pathologies such as osteochondritis dissecans, osteonecrosis, rheumatic disease, and trauma, may result in severe damage of articular cartilage and other joint structures, ranging from focal defects to osteoarthritis (OA). The osteochondral unit is one of the critical actors in this pathophysiological process. New approaches and applications in tissue engineering and regenerative medicine continue to drive the development of OA treatment. Hydrogel scaffolds, a component of tissue engineering, play an indispensable role in osteochondral regeneration. In this review, tissue engineering strategies regarding osteochondral regeneration were highlighted and summarized. The application of hydrogels for osteochondral regeneration within the last five years was evaluated with an emphasis on functionalized physical and chemical properties of hydrogel scaffolds, functionalized delivery hydrogel scaffolds as well as functionalized intelligent response hydrogel scaffolds. Lastly, to serve as guidance for future efforts in the creation of bioinspired hydrogel scaffolds, a succinct summary and new views for specific mechanisms, applications, and existing limitations of the newly designed functionalized hydrogel scaffolds were offered.

5.
Bioact Mater ; 30: 29-45, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37521275

ABSTRACT

The diabetic wounds remain to be unsettled clinically, with chronic wounds characterized by drug-resistant bacterial infections, compromised angiogenesis and oxidative damage to the microenvironment. To ameliorate oxidative stress and applying antioxidant treatment in the wound site, we explore the function of folliculin-interacting protein 1 (FNIP1), a mitochondrial gatekeeper protein works to alter mitochondrial morphology, reduce oxidative phosphorylation and protect cells from unwarranted ROS accumulation. And our in vitro experiments showed the effects of FNIP1 in ameliorating oxidative stress and rescued impaired angiogenesis of HUVECs in high glucose environment. To realize the drug delivery and local regulation of FNIP1 in diabetic wound sites, a novel designed glucose-responsive HA-PBA-FA/EN106 hydrogel is introduced for improving diabetic wound healing. Due to the dynamic phenylboronate ester structure with a phenylboronic acid group between hyaluronic acid (HA) and phenylboronic acid (PBA), the hydrogel is able to realize a glucose-responsive release of drugs. Fulvic acid (FA) is added in the hydrogel, which not only severs as crosslinking agent but also provides antibacterial and anti-inflammatory abilities. Moreover, the release of FEM1b-FNIP1 axis inhibitor EN106 ameliorated oxidative stress and stimulated angiogenesis through FEM1b-FNIP1 axis regulation. These in vivo and in vitro results demonstrated that accelerated diabetic wounds repair with the use of the HA-PBA-FA/EN106 hydrogel, which may provide a promising strategy for chronic diabetic wound repair.

6.
Biomater Sci ; 11(18): 6035-6059, 2023 Sep 12.
Article in English | MEDLINE | ID: mdl-37522328

ABSTRACT

Immune homeostasis is delicately mediated by the dynamic balance between effector immune cells and regulatory immune cells. Local deviations from immune homeostasis in the microenvironment of bone fractures, caused by an increased ratio of effector to regulatory cues, can lead to excessive inflammatory conditions and hinder bone regeneration. Therefore, achieving effective and localized immunomodulation of bone fractures is crucial for successful bone regeneration. Recent research has focused on developing localized and specific immunomodulatory strategies using local hydrogel-based delivery systems. In this review, we aim to emphasize the significant role of immune homeostasis in bone regeneration, explore local hydrogel-based delivery systems, discuss emerging trends in immunomodulation for enhancing bone regeneration, and address the limitations of current delivery strategies along with the challenges of clinical translation.


Subject(s)
Fractures, Bone , Hydrogels , Humans , Bone Regeneration , Immunomodulation
7.
Stem Cells Int ; 2023: 7638842, 2023.
Article in English | MEDLINE | ID: mdl-37274021

ABSTRACT

Bone mesenchymal stem cells (BMSCs) play an important role in maintaining the dynamic balance of bone metabolism. Recent studies have reported that a decrease in the osteogenic function of MSCs is strongly associated with osteoporosis. Melatonin is a neuroendocrine hormone produced in the pineal gland and is essential in the physiological regulation. This study is aimed at exploring the effect of melatonin on MSCs osteoblastic differentiation and elucidate the underlying mechanisms. We isolated BMSCs from rat bone marrow and demonstrated that melatonin improved osteogenic differentiation of BMSCs by the alizarin red staining and ALP staining. We then showed that melatonin enhanced osteogenic gene expression in BMSCs, including ALP, Col 1, OCN, OPN, and RUNX2. We further revealed that melatonin inhibited the inflammatory response of BMSCs by suppressing the NF-κB signaling pathways. In light of this, we found that the NF-κB pathway-specific activator TNF-α activated the NF-κB pathway, inhibited osteogenic differentiation, and induced inflammatory response in BMSCs. Melatonin was found to reverse the inhibitory effect of TNF-α on osteogenic differentiation and inflammation in BMSCs. Taken together, these findings indicated that melatonin may have therapeutic potential to be used for the treatment of osteoporosis.

8.
Adv Healthc Mater ; 12(25): e2300779, 2023 10.
Article in English | MEDLINE | ID: mdl-37051860

ABSTRACT

Diabetic wounds are characterized by drug-resistant bacterial infections, biofilm formation, impaired angiogenesis and perfusion, and oxidative damage to the microenvironment. Given their complex nature, diabetic wounds remain a major challenge in clinical practice. Reactive oxygen species (ROS), which have been shown to trigger hyperinflammation and excessive cellular apoptosis, play a pivotal role in the pathogenesis of diabetic wounds. ROS-scavenging nanosystems have recently emerged as smart and multifunctional nanomedicines with broad synergistic applicability. The documented anti-inflammatory and pro-angiogenic ability of ROS-scavenging treatments predestines these nanosystems as promising options for the treatment of diabetic wounds. Yet, in this context, the therapeutic applicability and efficacy of ROS-scavenging nanosystems remain to be elucidated. Herein, the role of ROS in diabetic wounds is deciphered, and the properties and strengths of nanosystems with ROS-scavenging capacity for the treatment of diabetic wounds are summarized. In addition, the current challenges of such nanosystems and their potential future directions are discussed through a clinical-translational lens.


Subject(s)
Diabetes Mellitus , Oxidative Stress , Reactive Oxygen Species , Apoptosis , Cell Aggregation , Nanomedicine
9.
Adv Mater ; 35(19): e2212300, 2023 May.
Article in English | MEDLINE | ID: mdl-36811203

ABSTRACT

Diabetic wound (DW) therapy is currently a big challenge in medicine and strategies to enhance neurogenesis and angiogenesis have appeared to be a promising direction. However, the current treatments have failed to coordinate neurogenesis and angiogenesis simultaneously, leading to an increased disability rate caused by DWs. Herein, a whole-course-repair system is introduced by a hydrogel to concurrently achieve a mutually supportive cycle of neurogenesis-angiogenesis under a favorable immune-microenvironment. This hydrogel can first be one-step packaged in a syringe for later in situ local injections to cover wounds long-termly for accelerated wound healing via the synergistic effect of magnesium ions (Mg2+ ) and engineered small extracellular vesicles (sEVs). The self-healing and bio-adhesive properties of the hydrogel make it an ideal physical barrier for DWs. At the inflammation stage, the formulation can recruit bone marrow-derived mesenchymal stem cells to the wound sites and stimulate them toward neurogenic differentiation, while providing a favorable immune microenvironment via macrophage reprogramming. At the proliferation stage of wound repair, robust angiogenesis occurs by the synergistic effect of the newly differentiated neural cells and the released Mg2+ , allowing a regenerative neurogenesis-angiogenesis cycle to take place at the wound site. This whole-course-repair system provides a novel platform for combined DW therapy.


Subject(s)
Diabetes Mellitus , Wound Healing , Humans , Hydrogels/pharmacology , Macrophages , Neurogenesis
10.
Mil Med Res ; 9(1): 65, 2022 11 19.
Article in English | MEDLINE | ID: mdl-36401295

ABSTRACT

Bone, cartilage, and soft tissue regeneration is a complex spatiotemporal process recruiting a variety of cell types, whose activity and interplay must be precisely mediated for effective healing post-injury. Although extensive strides have been made in the understanding of the immune microenvironment processes governing bone, cartilage, and soft tissue regeneration, effective clinical translation of these mechanisms remains a challenge. Regulation of the immune microenvironment is increasingly becoming a favorable target for bone, cartilage, and soft tissue regeneration; therefore, an in-depth understanding of the communication between immune cells and functional tissue cells would be valuable. Herein, we review the regulatory role of the immune microenvironment in the promotion and maintenance of stem cell states in the context of bone, cartilage, and soft tissue repair and regeneration. We discuss the roles of various immune cell subsets in bone, cartilage, and soft tissue repair and regeneration processes and introduce novel strategies, for example, biomaterial-targeting of immune cell activity, aimed at regulating healing. Understanding the mechanisms of the crosstalk between the immune microenvironment and regeneration pathways may shed light on new therapeutic opportunities for enhancing bone, cartilage, and soft tissue regeneration through regulation of the immune microenvironment.


Subject(s)
Bone and Bones , Cartilage , Humans , Wound Healing
11.
Exp Mol Med ; 54(7): 961-972, 2022 07.
Article in English | MEDLINE | ID: mdl-35831436

ABSTRACT

MicroRNAs (miRNAs) broadly regulate normal biological functions of bone and the progression of fracture healing and osteoporosis. Recently, it has been reported that miR-1224-5p in fracture plasma is a potential therapy for osteogenesis. To investigate the roles of miR-1224-5p and the Rap1 signaling pathway in fracture healing and osteoporosis development and progression, we used BMMs, BMSCs, and skull osteoblast precursor cells for in vitro osteogenesis and osteoclastogenesis studies. Osteoblastogenesis and osteoclastogenesis were detected by ALP, ARS, and TRAP staining and bone slice resorption pit assays. The miR-1224-5p target gene was assessed by siRNA-mediated target gene knockdown and luciferase reporter assays. To explore the Rap1 pathway, we performed high-throughput sequencing, western blotting, RT-PCR, chromatin immunoprecipitation assays and immunohistochemical staining. In vivo, bone healing was judged by the cortical femoral defect, cranial bone defect and femoral fracture models. Progression of osteoporosis was evaluated by an ovariectomy model and an aged osteoporosis model. We discovered that the expression of miR-1224-5p was positively correlated with fracture healing progression. Moreover, in vitro, overexpression of miR-1224-5p slowed Rankl-induced osteoclast differentiation and promoted osteoblast differentiation via the Rap1-signaling pathway by targeting ADCY2. In addition, in vivo overexpression of miR-1224-5p significantly promoted fracture healing and ameliorated the progression of osteoporosis caused by estrogen deficiency or aging. Furthermore, knockdown of miRNA-1224-5p inhibited bone regeneration in mice and accelerated the progression of osteoporosis in elderly mice. Taken together, these results identify miR-1224-5p as a key bone osteogenic regulator, which may be a potential therapeutic target for osteoporosis and fracture nonunion.


Subject(s)
Bone Resorption , MicroRNAs , Osteoporosis , Adenylyl Cyclases , Animals , Bone Resorption/metabolism , Cell Differentiation/genetics , Female , Mice , MicroRNAs/genetics , MicroRNAs/metabolism , Osteoblasts/metabolism , Osteoclasts/metabolism , Osteogenesis/genetics , Osteoporosis/genetics , Signal Transduction , rap1 GTP-Binding Proteins
12.
Front Cell Dev Biol ; 10: 826023, 2022.
Article in English | MEDLINE | ID: mdl-35445030

ABSTRACT

With the worldwide aging population, the prevalence of osteoporosis is on the rise, particularly the number of postmenopausal women with the condition. However, the various adverse side effects associated with the currently available treatment options underscore the need to develop novel therapies. In this study, we investigated the use of AQX-1125, a novel clinical-stage activator of inositol phosphatase-1 (SHIP1), in ovariectomized (OVX) mice, identifying a protective role. We then found that the effect was likely due to increased osteogenesis and mineralization and decreased osteoclastogenesis caused by AQX-1125 in a time- and dose-dependent manner. The effect against OVX-induced bone loss was identified to be SHIP1-dependent as pretreatment of BMSCs and BMMs with SHIP1 RNAi could greatly diminish the osteoprotective effects. Furthermore, SHIP1 RNAi administration in vivo induced significant bone loss and decreased bone mass. Mechanistically, AQX-1125 upregulated the expression level and activity of SHIP1, followed upregulating the phosphorylation levels of PI3K and Akt to promote osteoblast-related gene expressions, including Alp, cbfa1, Col1a1, and osteocalcin (OCN). NF-κB signaling was also inhibited through suppression of the phosphorylation of IκBα and P65 induced by RANKL, resulting in diminished osteoclastogenesis. Taken together, our results demonstrate that AQX-1125 may be a promising candidate for preventing and treating bone loss.

13.
Front Cell Dev Biol ; 10: 824812, 2022.
Article in English | MEDLINE | ID: mdl-35281084

ABSTRACT

Although bone is an organ that displays potential for self-healing after damage, bone regeneration does not occur properly in some cases, and it is still a challenge to treat large bone defects. The development of bone tissue engineering provides a new approach to the treatment of bone defects. Among various cell types, mesenchymal stem cells (MSCs) represent one of the most promising seed cells in bone tissue engineering due to their functions of osteogenic differentiation, immunomodulation, and secretion of cytokines. Regulation of osteogenic differentiation of MSCs has become an area of extensive research over the past few years. This review provides an overview of recent research progress on enhancement strategies for MSC osteogenesis, including improvement in methods of cell origin selection, culture conditions, biophysical stimulation, crosstalk with macrophages and endothelial cells, and scaffolds. This is favorable for further understanding MSC osteogenesis and the development of MSC-based bone tissue engineering.

14.
J Nanobiotechnology ; 20(1): 25, 2022 Jan 06.
Article in English | MEDLINE | ID: mdl-34991615

ABSTRACT

BACKGROUND: The regeneration and repair of articular cartilage remains a major challenge for clinicians and scientists due to the poor intrinsic healing of this tissue. Since cartilage injuries are often clinically irregular, tissue-engineered scaffolds that can be easily molded to fill cartilage defects of any shape that fit tightly into the host cartilage are needed. METHOD: In this study, bone marrow mesenchymal stem cell (BMSC) affinity peptide sequence PFSSTKT (PFS)-modified chondrocyte extracellular matrix (ECM) particles combined with GelMA hydrogel were constructed. RESULTS: In vitro experiments showed that the pore size and porosity of the solid-supported composite scaffolds were appropriate and that the scaffolds provided a three-dimensional microenvironment supporting cell adhesion, proliferation and chondrogenic differentiation. In vitro experiments also showed that GelMA/ECM-PFS could regulate the migration of rabbit BMSCs. Two weeks after implantation in vivo, the GelMA/ECM-PFS functional scaffold system promoted the recruitment of endogenous mesenchymal stem cells from the defect site. GelMA/ECM-PFS achieved successful hyaline cartilage repair in rabbits in vivo, while the control treatment mostly resulted in fibrous tissue repair. CONCLUSION: This combination of endogenous cell recruitment and chondrogenesis is an ideal strategy for repairing irregular cartilage defects.


Subject(s)
Chondrogenesis/drug effects , Decellularized Extracellular Matrix , Hydrogels , Oligopeptides , Tissue Scaffolds/chemistry , Animals , Cartilage, Articular/cytology , Decellularized Extracellular Matrix/chemistry , Decellularized Extracellular Matrix/pharmacology , Hydrogels/chemistry , Hydrogels/pharmacology , Male , Mesenchymal Stem Cells/drug effects , Oligopeptides/chemistry , Oligopeptides/pharmacology , Rabbits , Tissue Engineering/methods
15.
Front Bioeng Biotechnol ; 9: 664592, 2021.
Article in English | MEDLINE | ID: mdl-34017827

ABSTRACT

Biomaterials play a core role in cartilage repair and regeneration. The success or failure of an implanted biomaterial is largely dependent on host response following implantation. Host response has been considered to be influenced by numerous factors, such as immune components of materials, cytokines and inflammatory agents induced by implants. Both synthetic and native materials involve immune components, which are also termed as immunogenicity. Generally, the innate and adaptive immune system will be activated and various cytokines and inflammatory agents will be consequently released after biomaterials implantation, and further triggers host response to biomaterials. This will guide the constructive remolding process of damaged tissue. Therefore, biomaterial immunogenicity should be given more attention. Further understanding the specific biological mechanisms of host response to biomaterials and the effects of the host-biomaterial interaction may be beneficial to promote cartilage repair and regeneration. In this review, we summarized the characteristics of the host response to implants and the immunomodulatory properties of varied biomaterial. We hope this review will provide scientists with inspiration in cartilage regeneration by controlling immune components of biomaterials and modulating the immune system.

16.
ACS Appl Mater Interfaces ; 13(20): 23369-23383, 2021 May 26.
Article in English | MEDLINE | ID: mdl-33979130

ABSTRACT

Articular cartilage (AC) lesions are fairly common but remain an obstacle for clinicians and researchers due to their poor self-healing capacity. Recently, a promising therapy based on the recruitment of autologous mesenchymal stem cells (MSCs) has been developed for the regeneration of full-thickness cartilage defects in the knee joint. In this study, a 3D-bioprinted difunctional scaffold was developed based on aptamer HM69-mediated MSC-specific recruitment and growth factor-enhanced cell chondrogenesis. The aptamer, which can specifically recognize and recruit MSCs, was first chemically conjugated to the decellularized cartilage extracellular matrix and then mixed with gelatin methacrylate to form a photocrosslinkable bioink ready for 3D bioprinting. Together with the growth factor that promoted cell chondrogenic differentiation, the biodegradable polymer poly(ε-caprolactone) was further chosen to impart mechanical strength to the 3D bioprinted constructs. The difunctional scaffold specifically recruited MSCs, provided a favorable microenvironment for cell adhesion and proliferation, promoted chondrogenesis, and thus greatly improved cartilage repair in rabbit full-thickness defects. In conclusion, this study demonstrated that 3D bioprinting of difunctional scaffolds could be a promising strategy for in situ AC regeneration based on aptamer-directed cell recruitment and growth-factor-enhanced cell chondrogenesis.


Subject(s)
Aptamers, Nucleotide/pharmacology , Bioprinting , Cartilage, Articular , Chondrogenesis , Tissue Engineering/methods , Animals , Cartilage, Articular/cytology , Cartilage, Articular/metabolism , Cells, Cultured , Chondrocytes/cytology , Chondrocytes/drug effects , Chondrogenesis/drug effects , Chondrogenesis/physiology , Intercellular Signaling Peptides and Proteins/pharmacology , Male , Printing, Three-Dimensional , Rabbits , Rats , Tissue Scaffolds/chemistry
17.
J Tissue Eng Regen Med ; 15(9): 776-787, 2021 09.
Article in English | MEDLINE | ID: mdl-34044473

ABSTRACT

Xenogeneic porcine fibrin sealant (PFS), derived from porcine blood, was used as a scaffold for cartilage tissue engineering. PFS has a porous microstructure, biocompatibility and degradation, and it provides a perfect extracellular matrix environment for the adhesion and proliferation of chondrocytes. Recently, PFS in combination with autologous chondrocytes (ACs) were used to study the microstructure of PFS scaffolds and promotion effect on the proliferation and migration of ACs. In this study, we investigated the effects of PFS in combination with ACs on the healing of cartilage defects in rabbits. A full-thickness cartilage defect was made in the femoral trochlear in rabbits, subsequently, three surgical procedures were used to repair the defect, namely: the defect was treated with microfracture (MF group); the defect was filled with PFS alone (PFS group) or in combination with ACs (PFS + ACs group); the unrepaired cartilage defects served as the control group (CD group). Three and 6 months after the operation, the reparative effect was evaluated using medical imaging, gross scoring, pathological staining, biomechanical testing and biochemical examination. The PFS group showed a limited effect on defect repair, this result was significantly worse than the MF group. The best reparative effect was observed in the PFS + ACs group. These results hinted that PFS in combination with autologous chondrocytes has broad prospects for clinical applications in cartilage tissue engineering.


Subject(s)
Cartilage, Articular/pathology , Chondrocytes/transplantation , Fibrin Tissue Adhesive/pharmacology , Regeneration , Animals , Biomechanical Phenomena/drug effects , Bone Regeneration/drug effects , Cartilage, Articular/diagnostic imaging , Cartilage, Articular/drug effects , Cell Movement/drug effects , Collagen Type II/metabolism , Disease Models, Animal , Magnetic Resonance Imaging , Rabbits , Regeneration/drug effects , Swine , Wound Healing/drug effects , X-Ray Microtomography
18.
Stem Cells Int ; 2021: 8830834, 2021.
Article in English | MEDLINE | ID: mdl-33824665

ABSTRACT

Articular cartilage is susceptible to damage, but its self-repair is hindered by its avascular nature. Traditional treatment methods are not able to achieve satisfactory repair effects, and the development of tissue engineering techniques has shed new light on cartilage regeneration. Mesenchymal stem cells (MSCs) are one of the most commonly used seed cells in cartilage tissue engineering. However, MSCs tend to lose their multipotency, and the composition and structure of cartilage-like tissues formed by MSCs are far from those of native cartilage. Thus, there is an urgent need to develop strategies that promote MSC chondrogenic differentiation to give rise to durable and phenotypically correct regenerated cartilage. This review provides an overview of recent advances in enhancement strategies for MSC chondrogenic differentiation, including optimization of bioactive factors, culture conditions, cell type selection, coculture, gene editing, scaffolds, and physical stimulation. This review will aid the further understanding of the MSC chondrogenic differentiation process and enable improvement of MSC-based cartilage tissue engineering.

19.
Acta Biomater ; 127: 131-145, 2021 06.
Article in English | MEDLINE | ID: mdl-33812074

ABSTRACT

Cartilage regeneration is a complex physiological process. Synovial macrophages play a critical immunomodulatory role in the acute inflammatory response surrounding joint injury. Due to the contrasting differences and heterogeneity of macrophage, the phenotype of macrophages are the key determinants of the healing response after cartilage injury. Biomaterials derived from extracellular matrix have been used for the repair and reconstruction of a variety of tissues by modulating the host macrophage response. However, the immunomodulatory effect of decellularized cartilage extracellular matrix (ECM) on macrophages has not been elucidated. It is necessary to clarify the immunomodulatory properties of decellularized cartilage matrix (DCM) to guide the design of cartilage regeneration materials. Here, we prepared porcine articular cartilage derived DCM and determined the response of mouse bone marrow-derived macrophages (BMDMs) to the pepsin-solubilized DCM (PDCM) in vitro. Macrophages activated by the PDCM could promote bone marrow-derived mesenchymal stem cells (BMSCs) invasion, migration, proliferation, and chondrogenic differentiation. Then, we verified that early optimization of the immunomodulatory effects of the cell-free DCM scaffold using IL-4 in vivo could achieve good cartilage regeneration in a rat knee osteochondral defect model. Therefore, this decellularized cartilage ECM scaffold combined with accurate and active immunomodulatory strategies provides a new approach for the development of cartilage regeneration materials. STATEMENT OF SIGNIFICANCE: This work reports a decellularized cartilage extracellular matrix (DCM) scaffold combined with an accurate and active immunomodulatory strategy to improve cartilage regeneration. Our findings demonstrated that the pepsin-solubilized DCM (PDCM) activated bone marrow-derived macrophages to polarize to a constructive macrophage phenotype. These polarized macrophages promoted bone marrow-derived mesenchymal stem cell invasion, migration, proliferation, and chondrogenic differentiation. DCM scaffolds combined with early-stage intra-articular injection of IL-4 created a wound-healing microenvironment and improved cartilage regeneration in a rat knee osteochondral defect model.


Subject(s)
Cartilage, Articular , Interleukin-4 , Animals , Chondrogenesis , Extracellular Matrix , Macrophages , Mice , Rats , Swine , Tissue Engineering , Tissue Scaffolds
20.
Cell Prolif ; 54(5): e13017, 2021 May.
Article in English | MEDLINE | ID: mdl-33704842

ABSTRACT

OBJECTIVES: CD49f is expressed on a variety of stem cells and has certain effects on their cytological functions, such as proliferation and differentiation potential. However, whether CD49f is expressed on the surface of adipose tissue-derived mesenchymal stem cells (ADSCs) and its effect on ADSCs has not been clarified. MATERIALS AND METHODS: The effects of in vitro culture passage and inflammatory factor treatment on CD49f expression and the adhesion ability of ADSCs from mice and rats were investigated. CD49f+ cells were selected from rat ADSCs (rADSCs) by magnetic-activated cell sorting (MACS), and the cellular functions of CD49f+ ADSCs and unsorted ADSCs, including their clonogenic, proliferation, adipogenic and osteogenic differentiation, migration and anti-apoptotic capacities, were compared. RESULTS: CD49f expression and the adhesion ability of ADSCs decreased with increasing in vitro culture passage number. TNF-α and IFN-γ treatment decreased CD49f expression but increased the adhesion ability of ADSCs. After CD49f was blocked with an anti-CD49f antibody, the adhesion ability of ADSCs was decreased. No significant difference in clonogenic activity was observed between unsorted ADSCs and CD49f+ ADSCs. CD49f+ ADSCs had greater proliferation, adipogenic and osteogenic differentiation, migration and anti-apoptotic capacities than unsorted ADSCs. CONCLUSION: In the current study, the expression of CD49f on ADSCs was identified for the first time. The expression of CD49f on ADSCs was influenced by in vitro culture passage number and inflammatory factor treatment. Compared with unsorted ADSCs, CD49f + ADSCs exhibited superior cellular functions, thus may have great application value in mesenchymal stem cell (MSC)-based therapies.


Subject(s)
Biomarkers/metabolism , Integrin alpha6/metabolism , Mesenchymal Stem Cells/metabolism , Adipogenesis , Adipose Tissue/cytology , Animals , Cell Differentiation , Cell Movement/drug effects , Cell Proliferation , Cells, Cultured , Gene Expression/drug effects , Integrin alpha6/genetics , Interleukin-1beta/pharmacology , Mesenchymal Stem Cells/cytology , Mice , Mice, Inbred C57BL , Osteogenesis , Rats , Rats, Sprague-Dawley , Tumor Necrosis Factor-alpha/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL