Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
Int Immunopharmacol ; 131: 111888, 2024 Apr 20.
Article in English | MEDLINE | ID: mdl-38522139

ABSTRACT

OBJECTIVES: Osteoarthritis (OA) is a whole-joint disease in which the role of the infrapatellar fat pad (IFP) in its pathogenesis is unclear. Our study explored the cellular heterogeneity of IFP to understand OA and identify therapeutic targets. METHODS: Single-cell and single-nuclei RNA sequencing were used to analyze 10 IFP samples, comprising 5 from OA patients and 5 from healthy controls. Analyses included differential gene expression, enrichment, pseudotime trajectory, and cellular communication, along with comparative studies with visceral and subcutaneous fats. Key subcluster and pathways were validated using multiplex immunohistochemistry. RESULTS: The scRNA-seq performed on the IFPs of the OA and control group profiled the gene expressions of over 49,674 cells belonging to 11 major cell types. We discovered that adipose stem and progenitor cells (ASPCs), contributing to the formation of both adipocytes and synovial-lining fibroblasts (SLF). Interstitial inflammatory fibroblasts (iiFBs) were a subcluster of ASPCs that exhibit notable pro-inflammatory and proliferative characteristics. We identified four adipocyte subtypes, with one subtype showing a reduced lipid synthesis ability. Furthermore, iiFBs modulated the activities of macrophages and T cells in the IFP. Compared to subcutaneous and visceral adipose tissues, iiFBs represented a distinctive subpopulation of ASPCs in IFP that regulated cartilage proliferation through the MK pathway. CONCLUSION: This study presents a comprehensive single-cell transcriptomic atlas of IFP, uncovering its complex cellular landscape and potential impact on OA progression. Our findings highlight the role of iiFBs in OA, especially through MK pathway, opening new avenues for understanding OA pathogenesis and developing novel targeted therapies.


Subject(s)
Osteoarthritis, Knee , Humans , Osteoarthritis, Knee/pathology , Adipose Tissue/pathology , Knee Joint/pathology , Gene Expression Profiling , Fibroblasts/metabolism
2.
Int Immunopharmacol ; 120: 110289, 2023 Jul.
Article in English | MEDLINE | ID: mdl-37182456

ABSTRACT

The chronic articular disease osteoarthritis (OA) is characterized by osteophyte generation, subchondral bone remodeling, and cartilage deterioration. Low levels of H2S catalyzed by cystathionine-γ-lyase (CSE) encoded by Cthhas neuroprotective, cardioprotective, anti-apoptotic, and anti-inflammatory effects thus, Cth is being developed as a potential therapy for the management of the pathogenesis and symptoms of osteoarthritis. Single-cell RNA sequencing (scRNA-seq) and immunohistochemistry of human cartilage revealed that the expression of CTH was decreased in OA patients. We found that Cthoverexpression decrease IL-1ß-induced overactivation of the NF-κB signaling pathway. In vivo, Cthoverexpression relieved pain response and cartilage damage in the anterior cruciate ligament transection (ACLT) rat model. In vitro, CSE alleviated chondrocytes catabolism, inflammation, apoptosis, and senescence, and suppressed the NF-κB pathway. We postulate that CSE has therapeutic effects in suppressing inflammation and degeneration in OA and should be further investigated clinically.


Subject(s)
Cartilage, Articular , Osteoarthritis , Humans , Rats , Animals , NF-kappa B/metabolism , Cystathionine/metabolism , Cystathionine/pharmacology , Cystathionine/therapeutic use , Cystathionine gamma-Lyase/metabolism , Osteoarthritis/metabolism , Inflammation/metabolism , Pain/pathology , Chondrocytes , Cartilage, Articular/pathology , Disease Models, Animal
3.
Int J Mol Sci ; 24(6)2023 Mar 10.
Article in English | MEDLINE | ID: mdl-36982442

ABSTRACT

Osteoarthritis (OA), the most common chronic inflammatory joint disease, is characterized by progressive cartilage degeneration, subchondral bone sclerosis, synovitis, and osteophyte formation. Metformin, a hypoglycemic agent used in the treatment of type 2 diabetes, has been evidenced to have anti-inflammatory properties to treat OA. It hampers the M1 polarization of synovial sublining macrophages, which promotes synovitis and exacerbates OA, thus lessening cartilage loss. In this study, metformin prevented the pro-inflammatory cytokines secreted by M1 macrophages, suppressed the inflammatory response of chondrocytes cultured with conditional medium (CM) from M1 macrophages, and mitigated the migration of M1 macrophages induced by interleukin-1ß (IL-1ß)-treated chondrocytes in vitro. In the meantime, metformin reduced the invasion of M1 macrophages in synovial regions brought about by the destabilization of medial meniscus (DMM) surgery in mice, and alleviated cartilage degeneration. Mechanistically, metformin regulated PI3K/AKT and downstream pathways in M1 macrophages. Overall, we demonstrated the therapeutic potential of metformin targeting synovial M1 macrophages in OA.


Subject(s)
Diabetes Mellitus, Type 2 , Metformin , Osteoarthritis , Synovitis , Mice , Animals , Metformin/pharmacology , Metformin/therapeutic use , Metformin/metabolism , Diabetes Mellitus, Type 2/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Osteoarthritis/drug therapy , Osteoarthritis/metabolism , Macrophages/metabolism , Chondrocytes/metabolism , Synovitis/drug therapy , Synovitis/metabolism
4.
Carbohydr Polym ; 304: 120492, 2023 Mar 15.
Article in English | MEDLINE | ID: mdl-36641179

ABSTRACT

Regeneration of injured articular cartilage is limited by low early-stage recruitment of stem cells and insufficient chondrogenic differentiation. Hydrogels are widely used to repair cartilage because they have excellent mechanical and biological properties. In this study, a dual drug-loaded thermosensitive hydroxypropyl chitin hydrogel (HPCH) system was prepared to release stromal-derived factor-1α-like polypeptides (SDFP) and kartogenin (KGN) for stem-cell recruitment and chondrogenic differentiation. The hydrogel had a network structure that promoted cell growth and nutrient exchange. Moreover, it was temperature sensitive and suitable for filling irregular defects. The system showed good biocompatibility in vitro and promoted stem-cell recruitment and chondrogenic differentiation. Furthermore, it reduced chondrocyte catabolism under inflammatory conditions. Animal experiments demonstrated that the dual-drug hydrogel systems can promote the regeneration of articular cartilage in rats. This study confirmed that an HPCH system loaded with KGN and SDFP could effectively repair articular cartilage defects and represents a viable treatment strategy.


Subject(s)
Cartilage, Articular , Hydrogels , Rats , Animals , Hydrogels/pharmacology , Hydrogels/chemistry , Chemokine CXCL12/chemistry , Regeneration , Cell Differentiation , Chondrogenesis
5.
Aging (Albany NY) ; 13(8): 11646-11664, 2021 04 20.
Article in English | MEDLINE | ID: mdl-33879632

ABSTRACT

OBJECTIVE: To investigate the heterogeneous responses of in vitro expanded chondrocytes, which were cultured in an interleukin (IL)-1ß -induced inflammatory environment. METHOD: Human articular chondrocytes were expanded, in vitro, for 13 days and treated with IL-1ß for 0, 24, and 48 h. Cells were collected and subjected to single-cell RNA sequencing. Multiple bioinformatics tools were used to determine the signatures that define chondrocyte physiology. RESULTS: Two major cell clusters with distinct expression patterns were identified at the initial phase and were with heterogeneous variation that coincides with inflammation progress. They transformed into two terminal cell clusters one of which exhibited OA-phenotype and proinflammatory characteristics through two paths, "response-to-inflammation" and "atypical response-to-inflammation", respectively. The involved cell clusters exhibited intrinsic relationship with cell types within native cartilage from OA patients. Genes controlling cell transformation to OA-phenotype were relating to the tumor necrosis factor (TNF) signaling pathway via NFKB, up-regulated KRAS signaling and the IL2/STAT5 signaling pathway and pathways relating to apoptosis and reactive oxygen species. CONCLUSION: The in vitro expanded chondrocytes under IL-1ß-induced inflammatory progression behave heterogeneously. One of the initial cell clusters could transform into a proinflammatory subpopulation through a termed response-to-inflammation path, which may serve as the core target to alleviate OA progression.


Subject(s)
Chondrocytes/pathology , Gene Expression Regulation/immunology , Osteoarthritis/immunology , Signal Transduction/genetics , Cartilage, Articular/cytology , Cells, Cultured , Child , Chondrocytes/immunology , Computational Biology , Culture Media/metabolism , Humans , Interleukin-1beta/metabolism , Osteoarthritis/genetics , Osteoarthritis/pathology , Primary Cell Culture , RNA-Seq , Signal Transduction/immunology , Single-Cell Analysis
6.
Theranostics ; 10(6): 2872-2887, 2020.
Article in English | MEDLINE | ID: mdl-32194841

ABSTRACT

Repairing cartilage defects using thermosensitive hydrogels is an attractive treatment strategy, but the poor mechanical properties and limited understanding of the interactions between hydrogels and cells limit their application. Methods: In this study, a thermosensitive hydroxypropyl chitin hydrogel (HPCH) was functionalized with methacrylate groups to synthesize photocrosslinkable glycidyl methacrylate-modified HPCH (GM-HPCH). GM-HPCH could form a gel in situ through a thermosensitive sol-gel transition and its mechanical properties can be improved by UV irradiation. Cell viability, cell adhesion and anti-apoptosis activity of GM-HPCH were evaluated. Transforming growth factor-ß1 (TGFß1) was introduced into the GM-HPCH hydrogel to fabricate the composite hydrogel. The macrophage immunomodulation, MSC recruitment and chondrogenesis of the composite hydrogel were evaluated. Results: With high biocompatibility, GM-HPCH could protect chondrocytes from apoptosis. Both the in vitro and in vivo experiments showed that GM-HPCH + TGFß1 shifted the recruited macrophages from M1 to M2 and promoted chondrogenic gene expression. Additionally, the composite hydrogel could promote the migration of marrow stromal cells (MSCs) in the Transwell test and increase migrated gene expression. The fluorescent tracking of MSCs confirmed MSC homing in the rat chondral defect with the help of GM-HPCH. The macroscopic evaluation and histological results at 6 weeks and 12 weeks postsurgery showed that GM-HPCH + TGFß1 can achieve superior cartilage healing. Conclusions: The GM-HPCH + TGFß1 hydrogel effectively promoted cartilage repair via immunomodulating macrophages, recruiting MSCs and promoting chondrogenesis; thus it is a promising injectable hydrogel for cartilage regeneration.


Subject(s)
Cartilage Diseases/therapy , Chitin/pharmacology , Chondrogenesis/drug effects , Hydrogels , Transforming Growth Factor beta1/pharmacology , Animals , Cell Adhesion/drug effects , Cell Line, Tumor , Cell Survival/drug effects , Cell- and Tissue-Based Therapy , Chitin/analogs & derivatives , Human Umbilical Vein Endothelial Cells , Humans , Hydrogels/pharmacology , Hydrogels/therapeutic use , Male , Mesenchymal Stem Cells , Mice , RAW 264.7 Cells , Rats , Rats, Sprague-Dawley , Tissue Engineering , Transforming Growth Factor beta1/therapeutic use
7.
Theranostics ; 10(2): 725-740, 2020.
Article in English | MEDLINE | ID: mdl-31903147

ABSTRACT

Chitin-derived hydrogels are commonly used in bone regeneration because of their high cell compatibility; however, their poor mechanical properties and little knowledge of the interaction between the materials and host cells have limited their practical application. Methods: To evaluate osteoinductivity and enhance the mechanical properties of a newly synthesized thermosensitive hydroxypropyl chitin hydrogel (HPCH), a mesenchymal stem cell (MSC)-encapsulated HPCH was infused into a three-dimensional-printed poly (ε-caprolactone) (PCL)/ nano-hydroxyapatite (nHA) scaffold to form a hybrid scaffold. The mechanical properties and cell compatibility of the scaffold were tested. The interaction between macrophages and scaffold for angiogenesis and osteogenesis were explored in vitro and in vivo. Results: The hybrid scaffold showed improved mechanical properties and high cell viability. When MSCs were encapsulated in HPCH, osteo-differentiation was promoted properly via endochondral ossification. The co-culture experiments showed that the hybrid scaffold facilitated growth factor secretion from macrophages, thus promoting vascularization and osteoinduction. The Transwell culture proved that MSCs modulated the inflammatory response of HPCH. Additionally, subcutaneous implantation of MSC-encapsulated HPCH confirmed M2 activation. In situ evaluation of calvarial defects confirmed that the repair was optimal in the MSC-loaded HPCH + PCL/nHA group. Conclusions: PCL/nHA + HPCH hybrid scaffolds effectively promoted vascularization and osteoinduction via osteogenesis promotion and immunomodulation, which suggests promising applications for bone regeneration.


Subject(s)
Bone Regeneration , Hydrogels/chemistry , Immunomodulation , Mesenchymal Stem Cells/cytology , Neovascularization, Physiologic , Osteogenesis , Tissue Scaffolds/chemistry , Animals , Biocompatible Materials/chemistry , Biocompatible Materials/pharmacology , Cell Adhesion , Cell Proliferation , Cell Survival , Cells, Cultured , Chitin/chemistry , Disease Models, Animal , Durapatite/chemistry , Male , Mesenchymal Stem Cells/metabolism , Polyesters/chemistry , Rats , Rats, Sprague-Dawley , Temperature , Tissue Engineering/methods
SELECTION OF CITATIONS
SEARCH DETAIL
...