1
|
Wang M, Yang J, Wu Y, Li H, Zhong Y, Luo Y, Xie R. Curcumin-activated Wnt5a pathway mediates Ca 2+ channel opening to affect myoblast differentiation and skeletal muscle regeneration. J Cachexia Sarcopenia Muscle 2024; 15:1834-1849. [PMID: 38982896 PMCID: PMC11446719 DOI: 10.1002/jcsm.13535] [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: 08/21/2023] [Revised: 05/17/2024] [Accepted: 06/03/2024] [Indexed: 07/11/2024] Open
Abstract
BACKGROUND Skeletal muscle injury is one of the most common sports injuries; if not properly treated or not effective rehabilitation treatment after injury, it can be transformed into chronic cumulative injury. Curcumin, an herbal ingredient, has been found to promote skeletal muscle injury repair and regeneration. The Wnt5a pathway is related to the expression of myogenic regulatory factors, and Ca2+ promotes the differentiation and fusion process of myoblasts. This study explored the effect and mechanism of curcumin on myoblast differentiation during the repair and regeneration of injured skeletal muscle and its relationship with the Wnt5a pathway and Ca2+ channel. METHODS Myogenic differentiation of C2C12 cells was induced with 2% horse serum, and a mouse (male, 10 weeks old) model of acute skeletal muscle injury was established using cardiotoxin (20 μL). In addition, we constructed a Wnt5a knockdown C2C12 cell model and a Wnt5a knockout mouse model. Besides, curcumin was added to the cell culture solution (80 mg/L) and fed to the mice (50 mg/kg). Fluorescence microscopy was used to determine the concentration of Ca2+. Western blot and RT-qPCR were used to detect the protein and mRNA levels of Wnt5a, CaN, NFAT2, MyoD, Myf5, Pax7, and Myogenin. The expression levels of MyoD, Myf5, Myogenin, MHC, Desmin, and NFAT2 were detected using immunofluorescence techniques. In addition, MyoD expression was observed using immunohistochemistry, and morphological changes in mouse muscle tissue were observed using HE staining. RESULTS During myoblast differentiation and muscle regeneration, Wnt5a expression was upregulated (P < 0.001) and the Wnt5a signalling pathway was activated. Wnt5a overexpression promoted the expression of MyoD, Myf5, Myogenin, MHC, and Desmin (P < 0.05), and conversely, knockdown of Wnt5a inhibited their expression (P < 0.001). The Wnt5a pathway mediated the opening of Ca2+ channels, regulated the expression levels of CaN, NFAT2, MyoD, Myf5, Myogenin, MHC, and Desmin (P < 0.01) and promoted the differentiation of C2C12 myoblasts and the repair and regeneration of injured skeletal muscle. The expression of Wnt5a, CaN, NFAT2, MyoD, Myogenin, Myf5, and MHC in C2C12 myoblast was significantly increased after curcumin intervention (P < 0.05); however, their expression decreased significantly after knocking down Wnt5a on the basis of curcumin intervention (P < 0.05). Similarly, in Wnt5a knockout mice, the promotion of muscle regeneration by curcumin was significantly attenuated. CONCLUSIONS Curcumin can activate the Wnt5a signalling pathway and mediate the opening of Ca2+ channels to accelerate the myogenic differentiation of C2C12 cells and the repair and regeneration of injured skeletal muscle.
Collapse
Affiliation(s)
- Mao‐yuan Wang
- Department of Rehabilitation MedicineFirst Affiliated Hospital of Gannan Medical UniversityGanzhouChina
- Ganzhou Key Laboratory of Rehabilitation MedicineGanzhouChina
| | - Jia‐ming Yang
- Department of Rehabilitation MedicineFirst Affiliated Hospital of Gannan Medical UniversityGanzhouChina
| | - Yi Wu
- Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases, Ministry of EducationGannan Medical UniversityGanzhouChina
| | - Hai Li
- Department of Rehabilitation MedicineFirst Affiliated Hospital of Gannan Medical UniversityGanzhouChina
| | - Yan‐biao Zhong
- Department of Rehabilitation MedicineFirst Affiliated Hospital of Gannan Medical UniversityGanzhouChina
- Ganzhou Intelligent Rehabilitation Technology Innovation CenterGanzhouChina
| | - Yun Luo
- Department of Rehabilitation MedicineFirst Affiliated Hospital of Gannan Medical UniversityGanzhouChina
| | - Rui‐lian Xie
- Department of OncologyFirst Affiliated Hospital of Gannan Medical UniversityGanzhouChina
| |
Collapse
|
2
|
Ding Y, Chen ZQ, Pan WF, Chen HJ, Wu M, Lyu YQ, Xie H, Huang YC, Chen ZZ, Chen F. The association and underlying mechanism of the digit ratio (2D:4D) in hypospadias. Asian J Androl 2024; 26:356-365. [PMID: 38563741 PMCID: PMC11280205 DOI: 10.4103/aja202377] [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: 07/29/2023] [Accepted: 01/14/2024] [Indexed: 04/04/2024] Open
Abstract
The second-to-fourth digit (2D:4D) ratio is thought to be associated with prenatal androgen exposure. However, the relationship between the 2D:4D ratio and hypospadias is poorly understood, and its molecular mechanism is not clear. In this study, by analyzing the hand digit length of 142 boys with hypospadias (23 distal, 68 middle, and 51 proximal) and 196 controls enrolled in Shanghai Children's Hospital (Shanghai, China) from December 2020 to December 2021, we found that the 2D:4D ratio was significantly increased in boys with hypospadias ( P < 0.001) and it was positively correlated with the severity of the hypospadias. This was further verified by the comparison of control mice and prenatal low testosterone mice model obtained by knocking out the risk gene (dynein axonemal heavy chain 8 [ DNAH8 ]) associated with hypospadias. Furthermore, the discrepancy was mainly caused by a shift in 4D. Proteomic characterization of a mouse model validated that low testosterone levels during pregnancy can impair the growth and development of 4D. Comprehensive mechanistic explorations revealed that during the androgen-sensitive window, the downregulation of the androgen receptor (AR) caused by low testosterone levels, as well as the suppressed expression of chondrocyte proliferation-related genes such as Wnt family member 5a ( Wnt5a ), Wnt5b , Smad family member 2 ( Smad2 ), and Smad3 ; mitochondrial function-related genes in cartilage such as AMP-activated protein kinase ( AMPK ) and nuclear respiratory factor 1 ( Nrf-1 ); and vascular development-related genes such as myosin light chain ( MLC ), notch receptor 3 ( Notch3 ), and sphingosine kinase 1 ( Sphk1 ), are responsible for the limitation of 4D growth, which results in a higher 2D:4D ratio in boys with hypospadias via decreased endochondral ossification. This study indicates that the ratio of 2D:4D is a risk marker of hypospadias and provides a potential molecular mechanism.
Collapse
Affiliation(s)
- Yu Ding
- Department of Urology, Shanghai Children’s Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200062, China
| | - Zu-Quan Chen
- Department of Urology, Shanghai Children’s Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200062, China
| | - Wen-Feng Pan
- Department of Urology, Shanghai Children’s Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200062, China
| | - Hao-Jie Chen
- Department of Urology, Shanghai Children’s Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200062, China
| | - Min Wu
- Department of Urology, Shanghai Children’s Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200062, China
| | - Yi-Qing Lyu
- Department of Urology, Shanghai Children’s Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200062, China
| | - Hua Xie
- Department of Urology, Shanghai Children’s Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200062, China
| | - Yi-Chen Huang
- Department of Urology, Shanghai Children’s Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200062, China
| | - Zhong-Zhong Chen
- Department of Urology, Shanghai Children’s Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200062, China
- Urogenital Development Research Center, Shanghai Children’s Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200062, China
| | - Fang Chen
- Department of Urology, Shanghai Children’s Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200062, China
- Clinical Research Center for Hypospadias, Pediatric College, Shanghai Jiao Tong University School of Medicine, Shanghai 200062, China
| |
Collapse
|
3
|
Zhou Z, Wang J, Jiang C, Xu K, Xu T, Yu X, Fang J, Yang Y, Dai X. Advances in Hydrogels for Meniscus Tissue Engineering: A Focus on Biomaterials, Crosslinking, Therapeutic Additives. Gels 2024; 10:114. [PMID: 38391445 PMCID: PMC10887778 DOI: 10.3390/gels10020114] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2024] [Revised: 01/23/2024] [Accepted: 01/26/2024] [Indexed: 02/24/2024] Open
Abstract
Meniscus tissue engineering (MTE) has emerged as a promising strategy for meniscus repair and regeneration. As versatile platforms, hydrogels have gained significant attention in this field, as they possess tunable properties that allow them to mimic native extracellular matrices and provide a suitable microenvironment. Additionally, hydrogels can be minimally invasively injected and can be adjusted to match the shape of the implant site. They can conveniently and effectively deliver bioactive additives and demonstrate good compatibility with other functional materials. These inherent qualities have made hydrogel a promising candidate for therapeutic approaches in meniscus repair and regeneration. This article provides a comprehensive review of the advancements made in the research on hydrogel application for meniscus tissue engineering. Firstly, the biomaterials and crosslinking strategies used in the formation of hydrogels are summarized and analyzed. Subsequently, the role of therapeutic additives, including cells, growth factors, and other active products, in facilitating meniscus repair and regeneration is thoroughly discussed. Furthermore, we summarize the key issues for designing hydrogels used in MTE. Finally, we conclude with the current challenges encountered by hydrogel applications and suggest potential solutions for addressing these challenges in the field of MTE. We hope this review provides a resource for researchers and practitioners interested in this field, thereby facilitating the exploration of new design possibilities.
Collapse
Affiliation(s)
- Zhuxing Zhou
- Department of Orthopedic Surgery, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310000, China
- Orthopedics Research Institute of Zhejiang University, Hangzhou 310000, China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou 310000, China
- Clinical Research Center of Motor System Disease of Zhejiang Province, Hangzhou 310000, China
| | - Jiajie Wang
- Department of Orthopedic Surgery, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310000, China
- Orthopedics Research Institute of Zhejiang University, Hangzhou 310000, China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou 310000, China
- Clinical Research Center of Motor System Disease of Zhejiang Province, Hangzhou 310000, China
| | - Chaoqian Jiang
- School of Materials and Engineering, Zhengzhou University, Zhengzhou 450001, China
| | - Kaiwang Xu
- Department of Orthopedic Surgery, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310000, China
- Orthopedics Research Institute of Zhejiang University, Hangzhou 310000, China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou 310000, China
- Clinical Research Center of Motor System Disease of Zhejiang Province, Hangzhou 310000, China
| | - Tengjing Xu
- Department of Orthopedic Surgery, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310000, China
- Orthopedics Research Institute of Zhejiang University, Hangzhou 310000, China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou 310000, China
- Clinical Research Center of Motor System Disease of Zhejiang Province, Hangzhou 310000, China
| | - Xinning Yu
- Department of Orthopedic Surgery, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310000, China
- Orthopedics Research Institute of Zhejiang University, Hangzhou 310000, China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou 310000, China
- Clinical Research Center of Motor System Disease of Zhejiang Province, Hangzhou 310000, China
| | - Jinghua Fang
- Department of Orthopedic Surgery, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310000, China
- Orthopedics Research Institute of Zhejiang University, Hangzhou 310000, China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou 310000, China
- Clinical Research Center of Motor System Disease of Zhejiang Province, Hangzhou 310000, China
| | - Yanyu Yang
- School of Materials and Engineering, Zhengzhou University, Zhengzhou 450001, China
| | - Xuesong Dai
- Department of Orthopedic Surgery, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310000, China
- Orthopedics Research Institute of Zhejiang University, Hangzhou 310000, China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou 310000, China
- Clinical Research Center of Motor System Disease of Zhejiang Province, Hangzhou 310000, China
| |
Collapse
|
4
|
Duarte-Olivenza C, Hurle JM, Montero JA, Lorda-Diez CI. Modeling the Differentiation of Embryonic Limb Chondroprogenitors by Cell Death and Cell Senescence in High Density Micromass Cultures and Their Regulation by FGF Signaling. Cells 2022; 12:cells12010175. [PMID: 36611968 PMCID: PMC9818968 DOI: 10.3390/cells12010175] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2022] [Revised: 12/20/2022] [Accepted: 12/28/2022] [Indexed: 01/04/2023] Open
Abstract
Considering the importance of programmed cell death in the formation of the skeleton during embryonic development, the aim of the present study was to analyze whether regulated cell degeneration also accompanies the differentiation of embryonic limb skeletal progenitors in high-density tridimensional cultures (micromass cultures). Our results show that the formation of primary cartilage nodules in the micromass culture assay involves a patterned process of cell death and cell senescence, complementary to the pattern of chondrogenesis. As occurs in vivo, the degenerative events were preceded by DNA damage detectable by γH2AX immunolabeling and proceeded via apoptosis and cell senescence. Combined treatments of the cultures with growth factors active during limb skeletogenesis, including FGF, BMP, and WNT revealed that FGF signaling modulates the response of progenitors to signaling pathways implicated in cell death. Transcriptional changes induced by FGF treatments suggested that this function is mediated by the positive regulation of the genetic machinery responsible for apoptosis and cell senescence together with hypomethylation of the Sox9 gene promoter. We propose that FGF signaling exerts a primordial function in the embryonic limb conferring chondroprogenitors with their biological properties.
Collapse
Affiliation(s)
| | | | - Juan A. Montero
- Correspondence: (J.A.M.); (C.I.L.-D.); Fax: +34-942201923 (J.A.M. and C.I.L.-D.)
| | - Carlos I. Lorda-Diez
- Correspondence: (J.A.M.); (C.I.L.-D.); Fax: +34-942201923 (J.A.M. and C.I.L.-D.)
| |
Collapse
|
5
|
Li Z, Zhang Y, Li Y, Xing S, Li S, Lyu J, Ban Z. PROSER2 is a poor prognostic biomarker for patients with osteosarcoma and promotes proliferation, migration and invasion of osteosarcoma cells. Exp Ther Med 2022; 24:750. [PMID: 36561964 PMCID: PMC9748638 DOI: 10.3892/etm.2022.11686] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Accepted: 09/14/2022] [Indexed: 11/10/2022] Open
Abstract
Proline- and serine-rich 2 (PROSER2) is encoded by the 47th open reading frame on human chromosome 10. Bioinformatic analysis has shown PROSER2 was significantly correlated with prognostic outcome of osteosarcoma patients. Its role in the progression and metastasis of human osteosarcoma has not been elucidated until now. Bioinformatics analysis was performed on 101 patients with osteosarcoma from The Cancer Genome Atlas database. High levels of PROSER2 were associated with a poor prognosis in patients with osteosarcoma. PROSER2 expression was significantly upregulated in clinical specimens from patients with osteosarcoma and osteosarcoma cell lines. MTT assay was performed to test the cell viability and Transwell assay was used to test the migration and invasion of MG63 cells. PROSER2 knockdown inhibited the viability, migration and invasion of MG63 cells. Gene Set Enrichment Analysis and Gene Ontology/Kyoto Encyclopedia of Genes and Genomes analysis showed that the differentially expressed genes were primarily involved in 'calcium signaling pathway' and 'Wnt signaling' in patients with osteosarcoma and high PROSER2 expression. Western blotting analysis revealed that PROSER2 regulated migration and invasion of osteosarcoma via the Wnt/nuclear factor of activated T-cells (NFAT)c1 signaling pathway. In conclusion, PROSER2 promoted the proliferation, migration and invasion of osteosarcoma cells via the Wnt/Ca2+/NFATc1 signaling pathway by increasing nuclear localization of NFATc1.
Collapse
Affiliation(s)
- Zhengjiang Li
- Department of Orthopedics, Chengdu Fifth People's Hospital, Chengdu, Sichuan 611130, P.R. China
| | - Yan Zhang
- Department of Orthopedics, Chengdu Fifth People's Hospital, Chengdu, Sichuan 611130, P.R. China
| | - Yongkui Li
- Department of Orthopedics, Chengdu Fifth People's Hospital, Chengdu, Sichuan 611130, P.R. China
| | - Shuxing Xing
- Department of Orthopedics, Chengdu Fifth People's Hospital, Chengdu, Sichuan 611130, P.R. China
| | - Shunqiang Li
- Department of Orthopedics, Chengdu Fifth People's Hospital, Chengdu, Sichuan 611130, P.R. China
| | - Jing Lyu
- Department of Orthopedics, Chengdu Fifth People's Hospital, Chengdu, Sichuan 611130, P.R. China
| | - Zhaonan Ban
- Department of Orthopedics, Chengdu Fifth People's Hospital, Chengdu, Sichuan 611130, P.R. China,Correspondence to: Dr Zhaonan Ban, Department of Orthopedics, Chengdu Fifth People's Hospital, 33 Mashi Street, Wenjiang, Chengdu, Sichuan 611130, P.R. China
| |
Collapse
|
6
|
Cao H, Zhou Q, Liu C, Zhang Y, Xie M, Qiao W, Dong N. Substrate stiffness regulates differentiation of induced pluripotent stem cells into heart valve endothelial cells. Acta Biomater 2022; 143:115-126. [PMID: 35235867 DOI: 10.1016/j.actbio.2022.02.032] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Revised: 02/08/2022] [Accepted: 02/21/2022] [Indexed: 12/15/2022]
Abstract
Substrate stiffness has been indicated as a primary determinant for stem cell fate, being capable of influencing motility, proliferation, and differentiation. Although the effects of stiffness on cardiac differentiation of human-induced pluripotent stem cells (h-iPSCs) have been reported, whether stiffness of polydimethylsiloxane-based substrates could enhance differentiation of h-iPSCs toward heart valve endothelial cells lineage (VECs) or not remains unknown. Herein, we modulated the substrate stiffness to evaluate its effect on the differentiation of h-iPSCs into valve endothelial-like cells (h-iVECs) in vitro and determine the suitable stiffness. The results revealed that VECs-related genes (PECAM1, CDH5, NFATC1, etc.) were significantly increased in h-iVECs obtained from the three substrates compared with h-iPSCs. Gene expression levels and differentiation efficiency were higher in the medium group than in the stiff and soft groups. An increase in substrate stiffness to 2.8 GPa decreased the efficiency of h-iPSCs differentiation into h-iVECs and downregulated VECs specific genes. Through mRNA sequencing, we determined the key genetic markers involved in stiffness guiding the differentiation of cardiac progenitor cells into h-iVECs. Unsupervised hierarchical clustering showed that medium stiffness were more suitable for the differentiation of h-iPSCs into h-iVECs in vitro. Moreover, this process is regulated by the WNT/Calcineurin signaling pathway. Overall, this study demonstrates how stiffness can be used to enhance the h-iVECs differentiation of iPSCs and emphasizes the importance of using substrate stiffness to accomplish a more specific and mature differentiation of h-iVECs for future therapeutic and tissue engineering valve applications. STATEMENT OF SIGNIFICANCE: Several studies have examined the stiffness-induced cell fate from pluripotent stem cells during the stage of mesoderm cell differentiation. This is the first research that rigorously examines the effect of substrate stiffness on human valve endothelial-like cells differentiation from cardiac progenitor cells. We found that the medium stiffness can increase the differentiation efficiency of h-iVECs from 40% to about 60%, and this process was regulated by the WNT/CaN signaling pathway through the activation of WNT5a. Substrate stiffness not only increases the differentiation efficiency of h-iVECs, but also improves its cellular functions such as low-density lipoprotein uptake and NO release. This study emphasizes the importance of using substrate stiffness to accomplish a more specific and mature differentiation of h-iVECs.
Collapse
|
7
|
De Palma A, Nalesso G. WNT Signalling in Osteoarthritis and Its Pharmacological Targeting. Handb Exp Pharmacol 2021; 269:337-356. [PMID: 34510305 DOI: 10.1007/164_2021_525] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Osteoarthritis (OA) is a highly disabling musculoskeletal condition affecting millions of people worldwide. OA is characterised by progressive destruction and irreversible morphological changes of joint tissues and architecture. At molecular level, de-regulation of several pathways contributes to the disruption of tissue homeostasis in the joint. Overactivation of the WNT/β-catenin signalling pathway has been associated with degenerative processes in OA. However, the multiple layers of complexity in the modulation of the signalling and the still insufficient knowledge of the specific molecular drivers of pathogenetic mechanisms have made difficult the pharmacological targeting of this pathway for therapeutic purposes. This review aims to provide an overview of the WNT/β-catenin signalling in OA with a particular focus on its role in the articular cartilage. Pathway components whose targeting showed therapeutic potential will be highlighted and described. A specific section will be dedicated to Lorecivivint, the first inhibitor of the β-catenin-dependent pathway currently in phase III clinical trial as OA-modifying agent.
Collapse
Affiliation(s)
- Anna De Palma
- Department of Veterinary Pre-Clinical Sciences, School of Veterinary Medicine, University of Surrey, Guildford, UK
| | - Giovanna Nalesso
- Department of Veterinary Pre-Clinical Sciences, School of Veterinary Medicine, University of Surrey, Guildford, UK.
| |
Collapse
|