1
|
Gao Y, Feng J, Chen Q, Wang Z, Yang Z. Milk fat globule-epidermal growth factor 8 exerts anti-osteoarthritis effects by inhibiting apoptosis and inducing autophagy in mouse chondrocytes. Biochem Biophys Res Commun 2024; 741:151022. [PMID: 39608050 DOI: 10.1016/j.bbrc.2024.151022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2024] [Revised: 11/13/2024] [Accepted: 11/18/2024] [Indexed: 11/30/2024]
Abstract
Osteoarthritis (OA) is a chronic degenerative joint disease, characterized by cartilage injury. Milk fat globule-epidermal growth factor 8 (MFG-E8) exhibited anti-inflammatory effects, with undefined mechanism in OA. Eighteen C57BL/6 J mice were randomized into Sham and destabilization of medial meniscus (DMM) groups, with DMM surgery for OA model establishment. Subsequently, DMM mice received rmMFG-E8 (50 ng/g) as rmMFG-E8 group (n = 6). HE staining and Safranin O/Fast green staining for cartilage tissue pathological damage, TUNEL staining for apoptosis, ELISA for pro-inflammatory factors, and immunohistochemistry were performed. Following extraction of primary mouse chondrocytes, cells were randomized into Control, OA (10 ng/mL IL-1β), OA + rmMFG-E8 (500 ng/mL), and OA + rmMFG-E8+3-MA (autophagy inhibitor, 5 mM) groups. Cell viability by CCK8, pro-inflammatory factors by qRT-PCR, and apoptosis by flow cytometry were detected. In vivo and in vitro, transmission electron microscopy for autophagy and Western blot for autophagy- and apoptosis-related expression were conducted. In vivo, DMM group showed severe cartilage tissue damage, higher matrix metalloproteinase 13 (MMP13), Cleaved caspase-3, Cleaved PARP, Bax, TNF-α, IL-1β, IL-6 levels, and lower Bcl-2, MFG-E8, Collagen II, LC3II/LC3I, Beclin1, and ATG5 expression. Further rmMFG-E8 intervention improved mobility and pathological damage in DMM mice, with lower MMP13 expression. In vitro, rmMFG-E8 in OA group reduced TNF-α, IL-1β, IL-6, Cleaved caspase-3, Cleaved PARP, Bax, and P62 levels, and enhanced cell viability, Bcl-2, LC3II/LC3I, Beclin1, and ATG5 expression. Further 3-MA treatment up-regulated apoptosis and decreased cell viability and autophagy. Therefore, MFG-E8 exerts anti-OA effects by inhibiting apoptosis and inducing autophagy, offering a new potential target for OA treatment.
Collapse
Affiliation(s)
- Yang Gao
- Department of Orthopedics, Affiliated Xiaoshan Hospital, Hangzhou Normal University, Hangzhou, Zhejiang, 311200, China
| | - Jian Feng
- Department of Orthopedics, Affiliated Xiaoshan Hospital, Hangzhou Normal University, Hangzhou, Zhejiang, 311200, China
| | - Qiang Chen
- Department of Orthopedics, Affiliated Xiaoshan Hospital, Hangzhou Normal University, Hangzhou, Zhejiang, 311200, China
| | - Zhe Wang
- Department of Orthopedics, Affiliated Xiaoshan Hospital, Hangzhou Normal University, Hangzhou, Zhejiang, 311200, China
| | - Zhihua Yang
- Department of Orthopedics, Affiliated Xiaoshan Hospital, Hangzhou Normal University, Hangzhou, Zhejiang, 311200, China.
| |
Collapse
|
2
|
Shen Y, Jiang R, Huang Y, Wang Y, Zhan S, Tang X, Yi P. Identification of hub genes through integrated single-cell and microarray transcriptome analysis in osteoarthritic meniscus. J Orthop Surg Res 2024; 19:682. [PMID: 39438957 PMCID: PMC11515729 DOI: 10.1186/s13018-024-05175-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/14/2024] [Accepted: 10/13/2024] [Indexed: 10/25/2024] Open
Abstract
BACKGROUND Osteoarthritis (OA) is marked by the progressive degradation of joint cartilage and subchondral bone. The precise molecular mechanisms driving meniscus deterioration in OA, especially at the single-cell level, remain poorly understood. METHOD We analyzed two datasets from the GEO database, GSE220243 and GSE98918, focusing on meniscus tissue sequencing data from OA and non-OA patients. The standard Seurat procedure was employed to process single-cell data and identify differentially expressed genes (DEGs). Immune cell infiltration was assessed using the Microenvironment Cell Populations (MCP) counter and CIBERSORT algorithms. For the microarray data, DEGs were identified with the limma package, and Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) analyses were performed using ClusterProfiler. The overlapping DEGs from both datasets were imported into Cytoscape to generate protein-protein interaction (PPI) networks and identify hub genes. Transcription factor (TF) and miRNA interaction networks were analyzed using NetworkAnalyst, and gene-related predictive drugs were enriched through the DSigDB platform. RESULT After quality control, 34,763 cells from the OA patients and 34,145 cells from the healthy controls were analyzed. UMAP identified and SingleR annotated 14 cell clusters. The 10 largest cell clusters were selected for further analysis. The OA group exhibited a notable increase in macrophages and a reduction in cytotoxic lymphocytes and endothelial cells in the meniscus. In GSE98918, 220 DEGs were identified, and the MCODE plug-in in Cytoscape pinpointed a key module containing 12 candidate genes. The MCC methodfiltered the top 20 DEGs in each GSE220243 cluster. Overlapping DEGs from GSE220243 and GSE98918 identified COL1A1, COL3A1, COL5A2, COL6A3, LOX, and VEGFA as significantly decreased in OA, with COL3A1, COL5A2, LOX, and VEGFA upregulated in meniscal chondrocytes. The interaction network highlighted 3 key miRNAs and 13 shared TFs. Ten gene-related predictive drug molecules were identified. CONCLUSION This research highlights crucial genes in the OA meniscus and uncovers their differing regulatory patterns between chondrocytes and non-chondrocytes. These findings enhance our understanding of the molecular mechanisms driving OA pathogenesis and aid in identifying potential drug targets.
Collapse
Affiliation(s)
- Yanzhu Shen
- China-Japan Friendship Hospital (Institute of Clinical Medical Sciences), Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100730 , China
- Department of Orthopaedics, China-Japan Friendship Hospital, Beijing, 100029, China
| | - Ruichen Jiang
- Graduate School, Beijing University of Chinese Medicine, Beijing, 100029, China
- Department of Orthopaedics, China-Japan Friendship Hospital, Beijing, 100029, China
| | - Yanjun Huang
- China-Japan Friendship Hospital (Institute of Clinical Medical Sciences), Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100730 , China
- Department of Orthopaedics, China-Japan Friendship Hospital, Beijing, 100029, China
| | - Yuming Wang
- Department of Orthopaedics, China-Japan Friendship Hospital, Beijing, 100029, China
| | - Sizheng Zhan
- Department of Orthopaedics, China-Japan Friendship Hospital, Beijing, 100029, China
| | - Xiangsheng Tang
- China-Japan Friendship Hospital (Institute of Clinical Medical Sciences), Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100730 , China
- Department of Orthopaedics, China-Japan Friendship Hospital, Beijing, 100029, China
| | - Ping Yi
- China-Japan Friendship Hospital (Institute of Clinical Medical Sciences), Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100730 , China.
- Department of Orthopaedics, China-Japan Friendship Hospital, Beijing, 100029, China.
| |
Collapse
|
3
|
Chen W, Lu Y, Zhang Y, Wu J, McVicar A, Chen Y, Zhu S, Zhu G, Lu Y, Zhang J, McConnell M, Li YP. Cbfβ regulates Wnt/β-catenin, Hippo/Yap, and Tgfβ signaling pathways in articular cartilage homeostasis and protects from ACLT surgery-induced osteoarthritis. eLife 2024; 13:e95640. [PMID: 38805545 PMCID: PMC11132684 DOI: 10.7554/elife.95640] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2024] [Accepted: 04/30/2024] [Indexed: 05/30/2024] Open
Abstract
As the most common degenerative joint disease, osteoarthritis (OA) contributes significantly to pain and disability during aging. Several genes of interest involved in articular cartilage damage in OA have been identified. However, the direct causes of OA are poorly understood. Evaluating the public human RNA-seq dataset showed that CBFB (subunit of a heterodimeric Cbfβ/Runx1, Runx2, or Runx3 complex) expression is decreased in the cartilage of patients with OA. Here, we found that the chondrocyte-specific deletion of Cbfb in tamoxifen-induced Cbfbf/f;Col2a1-CreERT mice caused a spontaneous OA phenotype, worn articular cartilage, increased inflammation, and osteophytes. RNA-sequencing analysis showed that Cbfβ deficiency in articular cartilage resulted in reduced cartilage regeneration, increased canonical Wnt signaling and inflammatory response, and decreased Hippo/Yap signaling and Tgfβ signaling. Immunostaining and western blot validated these RNA-seq analysis results. ACLT surgery-induced OA decreased Cbfβ and Yap expression and increased active β-catenin expression in articular cartilage, while local AAV-mediated Cbfb overexpression promoted Yap expression and diminished active β-catenin expression in OA lesions. Remarkably, AAV-mediated Cbfb overexpression in knee joints of mice with OA showed the significant protective effect of Cbfβ on articular cartilage in the ACLT OA mouse model. Overall, this study, using loss-of-function and gain-of-function approaches, uncovered that low expression of Cbfβ may be the cause of OA. Moreover, Local admission of Cbfb may rescue and protect OA through decreasing Wnt/β-catenin signaling, and increasing Hippo/Yap signaling and Tgfβ/Smad2/3 signaling in OA articular cartilage, indicating that local Cbfb overexpression could be an effective strategy for treatment of OA.
Collapse
Affiliation(s)
- Wei Chen
- Division in Cellular and Molecular Medicine, Department of Pathology and Laboratory Medicine, Tulane University School of Medicine, Tulane UniversityNew OrleansUnited States
- Department of Pathology, School of Medicine, University of Alabama at BirminghamBirminghamUnited States
| | - Yun Lu
- Department of Pathology, School of Medicine, University of Alabama at BirminghamBirminghamUnited States
| | - Yan Zhang
- Department of Pathology, School of Medicine, University of Alabama at BirminghamBirminghamUnited States
| | - Jinjin Wu
- Department of Pathology, School of Medicine, University of Alabama at BirminghamBirminghamUnited States
| | - Abigail McVicar
- Division in Cellular and Molecular Medicine, Department of Pathology and Laboratory Medicine, Tulane University School of Medicine, Tulane UniversityNew OrleansUnited States
| | - Yilin Chen
- Division in Cellular and Molecular Medicine, Department of Pathology and Laboratory Medicine, Tulane University School of Medicine, Tulane UniversityNew OrleansUnited States
| | - Siyu Zhu
- Division in Cellular and Molecular Medicine, Department of Pathology and Laboratory Medicine, Tulane University School of Medicine, Tulane UniversityNew OrleansUnited States
| | - Guochun Zhu
- Department of Pathology, School of Medicine, University of Alabama at BirminghamBirminghamUnited States
| | - You Lu
- Division in Cellular and Molecular Medicine, Department of Pathology and Laboratory Medicine, Tulane University School of Medicine, Tulane UniversityNew OrleansUnited States
| | - Jiayang Zhang
- Division in Cellular and Molecular Medicine, Department of Pathology and Laboratory Medicine, Tulane University School of Medicine, Tulane UniversityNew OrleansUnited States
| | - Matthew McConnell
- Division in Cellular and Molecular Medicine, Department of Pathology and Laboratory Medicine, Tulane University School of Medicine, Tulane UniversityNew OrleansUnited States
| | - Yi-Ping Li
- Division in Cellular and Molecular Medicine, Department of Pathology and Laboratory Medicine, Tulane University School of Medicine, Tulane UniversityNew OrleansUnited States
- Department of Pathology, School of Medicine, University of Alabama at BirminghamBirminghamUnited States
| |
Collapse
|
4
|
Chen W, Lu Y, Zhang Y, Wu J, McVicar A, Chen Y, Zhu S, Zhu G, Lu Y, Zhang J, McConnell M, Li YP. Cbfβ regulates Wnt/β-catenin, Hippo/Yap, and TGFβ signaling pathways in articular cartilage homeostasis and protects from ACLT surgery-induced osteoarthritis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.01.15.575763. [PMID: 38293189 PMCID: PMC10827176 DOI: 10.1101/2024.01.15.575763] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/01/2024]
Abstract
As the most common degenerative joint disease, osteoarthritis (OA) contributes significantly to pain and disability during aging. Several genes of interest involved in articular cartilage damage in OA have been identified. However, the direct causes of OA are poorly understood. Evaluating the public human RNA-seq dataset showed that Cbfβ, (subunit of a heterodimeric Cbfβ/Runx1,Runx2, or Runx3 complex) expression is decreased in the cartilage of patients with OA. Here, we found that the chondrocyte-specific deletion of Cbfβ in tamoxifen-induced Cbfβf/fCol2α1-CreERT mice caused a spontaneous OA phenotype, worn articular cartilage, increased inflammation, and osteophytes. RNA-sequencing analysis showed that Cbfβ deficiency in articular cartilage resulted in reduced cartilage regeneration, increased canonical Wnt signaling and inflammatory response, and decreased Hippo/YAP signaling and TGF-β signaling. Immunostaining and western blot validated these RNA-seq analysis results. ACLT surgery-induced OA decreased Cbfβ and Yap expression and increased active β-catenin expression in articular cartilage, while local AAV-mediated Cbfβ overexpression promoted Yap expression and diminished active β-catenin expression in OA lesions. Remarkably, AAV-mediated Cbfβ overexpression in knee joints of mice with OA showed the significant protective effect of Cbfβ on articular cartilage in the ACLT OA mouse model. Overall, this study, using loss-of-function and gain-of-function approaches, uncovered that low expression of Cbfβ may be the cause of OA. Moreover, Local admission of Cbfβ may rescue and protect OA through decreasing Wnt/β-catenin signaling, and increasing Hippo/Yap signaling and TGFβ/Smad2/3 signaling in OA articular cartilage, indicating that local Cbfβ overexpression could be an effective strategy for treatment of OA.
Collapse
Affiliation(s)
- Wei Chen
- Division in Cellular and Molecular Medicine, Department of Pathology and Laboratory Medicine, Tulane University School of Medicine, Tulane University, New Orleans, Louisiana, USA
| | - Yun Lu
- Department of Pathology, School of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Yan Zhang
- Department of Pathology, School of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Jinjin Wu
- Department of Pathology, School of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Abigail McVicar
- Division in Cellular and Molecular Medicine, Department of Pathology and Laboratory Medicine, Tulane University School of Medicine, Tulane University, New Orleans, Louisiana, USA
| | - Yilin Chen
- Division in Cellular and Molecular Medicine, Department of Pathology and Laboratory Medicine, Tulane University School of Medicine, Tulane University, New Orleans, Louisiana, USA
| | - Siyu Zhu
- Division in Cellular and Molecular Medicine, Department of Pathology and Laboratory Medicine, Tulane University School of Medicine, Tulane University, New Orleans, Louisiana, USA
| | - Guochun Zhu
- Department of Pathology, School of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - You Lu
- Division in Cellular and Molecular Medicine, Department of Pathology and Laboratory Medicine, Tulane University School of Medicine, Tulane University, New Orleans, Louisiana, USA
| | - Jiayang Zhang
- Division in Cellular and Molecular Medicine, Department of Pathology and Laboratory Medicine, Tulane University School of Medicine, Tulane University, New Orleans, Louisiana, USA
| | - Matthew McConnell
- Division in Cellular and Molecular Medicine, Department of Pathology and Laboratory Medicine, Tulane University School of Medicine, Tulane University, New Orleans, Louisiana, USA
| | - Yi-Ping Li
- Division in Cellular and Molecular Medicine, Department of Pathology and Laboratory Medicine, Tulane University School of Medicine, Tulane University, New Orleans, Louisiana, USA
| |
Collapse
|
5
|
Steinwerth P, Bertrand J, Sandt V, Marchal S, Sahana J, Bollmann M, Schulz H, Kopp S, Grimm D, Wehland M. Structural and Molecular Changes of Human Chondrocytes Exposed to the Rotating Wall Vessel Bioreactor. Biomolecules 2023; 14:25. [PMID: 38254625 PMCID: PMC10813504 DOI: 10.3390/biom14010025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Revised: 12/21/2023] [Accepted: 12/22/2023] [Indexed: 01/24/2024] Open
Abstract
Over the last 30 years, the prevalence of osteoarthritis (OA), a disease characterized by a loss of articular cartilage, has more than doubled worldwide. Patients suffer from pain and progressive loss of joint function. Cartilage is an avascular tissue mostly consisting of extracellular matrix with embedded chondrocytes. As such, it does not regenerate naturally, which makes an early onset of OA prevention and treatment a necessity to sustain the patients' quality of life. In recent years, tissue engineering strategies for the regeneration of cartilage lesions have gained more and more momentum. In this study, we aimed to investigate the scaffold-free 3D cartilage tissue formation under simulated microgravity in the NASA-developed rotating wall vessel (RWV) bioreactor. For this purpose, we cultured both primary human chondrocytes as well as cells from the immortalized line C28/I2 for up to 14 days on the RWV and analyzed tissue morphology, development of apoptosis, and expression of cartilage-specific proteins and genes by histological staining, TUNEL-assays, immunohistochemical detection of collagen species, and quantitative real-time PCR, respectively. We observed spheroid formation in both cell types starting on day 3. After 14 days, constructs from C28/I2 cells had diameters of up to 5 mm, while primary chondrocyte spheroids were slightly smaller with 3 mm. Further inspection of the 14-day-old C28/I2 spheroids revealed a characteristic cartilage morphology with collagen-type 1, -type 2, and -type 10 positivity. Interestingly, these tissues were less susceptible to RWV-induced differential gene expression than those formed from primary chondrocytes, which showed significant changes in the regulation of IL6, ACTB, TUBB, VIM, COL1A1, COL10A1, MMP1, MMP3, MMP13, ITGB1, LAMA1, RUNX3, SOX9, and CASP3 gene expression. These diverging findings might reflect the differences between primary and immortalized cells. Taken together, this study shows that simulated microgravity using the RWV bioreactor is suitable to engineer dense 3D cartilage-like tissue without addition of scaffolds or any other artificial materials. Both primary articular cells and the stable chondrocyte cell line C28/I2 formed 3D neocartilage when exposed for 14 days to an RWV.
Collapse
Affiliation(s)
- Paul Steinwerth
- Department of Microgravity and Translational Regenerative Medicine, University Clinic for Plastic, Aesthetic and Hand Surgery, Otto von Guericke University, 39106 Magdeburg, Germany; (P.S.); (V.S.); (S.M.); (H.S.); (M.W.)
| | - Jessica Bertrand
- Department of Orthopaedic Surgery, Otto-von-Guericke-University Magdeburg, 39120 Magdeburg, Germany; (J.B.); (M.B.)
- Research Group “Magdeburger Arbeitsgemeinschaft für Forschung unter Raumfahrt-und Schwerelosigkeitsbedingungen” (MARS), Otto von Guericke University, 39106 Magdeburg, Germany;
| | - Viviann Sandt
- Department of Microgravity and Translational Regenerative Medicine, University Clinic for Plastic, Aesthetic and Hand Surgery, Otto von Guericke University, 39106 Magdeburg, Germany; (P.S.); (V.S.); (S.M.); (H.S.); (M.W.)
| | - Shannon Marchal
- Department of Microgravity and Translational Regenerative Medicine, University Clinic for Plastic, Aesthetic and Hand Surgery, Otto von Guericke University, 39106 Magdeburg, Germany; (P.S.); (V.S.); (S.M.); (H.S.); (M.W.)
| | - Jayashree Sahana
- Department of Biomedicine, Aarhus University, Ole Worms Allé 4, 8000 Aarhus, Denmark;
| | - Miriam Bollmann
- Department of Orthopaedic Surgery, Otto-von-Guericke-University Magdeburg, 39120 Magdeburg, Germany; (J.B.); (M.B.)
| | - Herbert Schulz
- Department of Microgravity and Translational Regenerative Medicine, University Clinic for Plastic, Aesthetic and Hand Surgery, Otto von Guericke University, 39106 Magdeburg, Germany; (P.S.); (V.S.); (S.M.); (H.S.); (M.W.)
- Research Group “Magdeburger Arbeitsgemeinschaft für Forschung unter Raumfahrt-und Schwerelosigkeitsbedingungen” (MARS), Otto von Guericke University, 39106 Magdeburg, Germany;
| | - Sascha Kopp
- Research Group “Magdeburger Arbeitsgemeinschaft für Forschung unter Raumfahrt-und Schwerelosigkeitsbedingungen” (MARS), Otto von Guericke University, 39106 Magdeburg, Germany;
- Core Facility Tissue Engineering, Otto von Guericke University, 39106 Magdeburg, Germany
| | - Daniela Grimm
- Department of Microgravity and Translational Regenerative Medicine, University Clinic for Plastic, Aesthetic and Hand Surgery, Otto von Guericke University, 39106 Magdeburg, Germany; (P.S.); (V.S.); (S.M.); (H.S.); (M.W.)
- Research Group “Magdeburger Arbeitsgemeinschaft für Forschung unter Raumfahrt-und Schwerelosigkeitsbedingungen” (MARS), Otto von Guericke University, 39106 Magdeburg, Germany;
- Department of Biomedicine, Aarhus University, Ole Worms Allé 4, 8000 Aarhus, Denmark;
| | - Markus Wehland
- Department of Microgravity and Translational Regenerative Medicine, University Clinic for Plastic, Aesthetic and Hand Surgery, Otto von Guericke University, 39106 Magdeburg, Germany; (P.S.); (V.S.); (S.M.); (H.S.); (M.W.)
- Research Group “Magdeburger Arbeitsgemeinschaft für Forschung unter Raumfahrt-und Schwerelosigkeitsbedingungen” (MARS), Otto von Guericke University, 39106 Magdeburg, Germany;
| |
Collapse
|