1
|
Saleh AS, Abdel-Gabbar M, Gabr H, Shams A, Tamur S, Mahdi EA, Ahmed OM. Ameliorative effects of undifferentiated and differentiated BM-MSCs in MIA-induced osteoarthritic Wistar rats: roles of NF-κB and MMPs signaling pathways. Am J Transl Res 2024; 16:2793-2813. [PMID: 39114694 PMCID: PMC11301505 DOI: 10.62347/fghv2647] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2024] [Accepted: 05/15/2024] [Indexed: 08/10/2024]
Abstract
OBJECTIVES Osteoarthritis (OA) is a degenerative joint condition that is persistent. OA affects millions of people throughout the world. Both people and society are heavily economically burdened by osteoarthritis. There is currently no medication that can structurally alter the OA processes or stop the disease from progressing. Stem cells have the potential to revolutionize medicine due to their capacity to differentiate into chondrocytes, capacity to heal tissues and organs including osteoarthritic joints, and immunomodulatory capabilities. Therefore, the goal of the current investigation was to determine how bone marrow-derived mesenchymal stem cells (BM-MSCs) and chondrogenic differentiated mesenchymal stem cells (CD-MSCs) affected the treatment of OA in rats with monosodium iodoacetate (MIA)-induced osteoarthritis. METHODS Male Wistar rats were injected three times with MIA (1 mg)/100 µL isotonic saline to induce osteoarthritis in the ankle joint of the right hind leg. Following the MIA injection, the osteoarthritic rats were given weekly treatments of 1 × 106 BM-MSCs and CD-MSCs into the tail vein for three weeks. RESULTS The obtained results showed that in osteoarthritic rats, BM-MSCs and CD-MSCs dramatically decreased ankle diameter measurements, decreased oxidized glutathione (GSSG) level, and boosted glutathione peroxidase (GPx) and glutathione reductase (GR) activities. Additionally, in rats with MIA-induced OA, BM-MSCs and CD-MSCs dramatically boosted interleukin-10 (IL-10) serum levels while considerably decreasing serum anticitrullinated protein antibodies (ACPA), tumour necrosis factor-α (TNF-α), and interleukin-17 (IL-17) levels as well as ankle transforming growth factor-β1 (TGF-β1) expression. Analysis of histology, immunohistochemistry, and western blots in osteoarthritic joints showed that cartilage breakdown and joint inflammation gradually decreased over time. CONCLUSIONS It is possible to conclude from these results that BM-MSCs and CD-MSCs have anti-arthritic potential in MIA-induced OA, which may be mediated via inhibitory effects on oxidative stress, MMPs and inflammation through suppressing the NF-κB pathway. In osteoarthritis, using CD-MSCs as a treatment is more beneficial therapeutically than using BM-MSCs.
Collapse
Affiliation(s)
- Ablaa S Saleh
- Department of Biochemistry, Faculty of Science, Beni-Suef UniversityBeni-Suef 62521, Egypt
| | - Mohammed Abdel-Gabbar
- Department of Biochemistry, Faculty of Science, Beni-Suef UniversityBeni-Suef 62521, Egypt
| | - Hala Gabr
- Department of Clinical and Chemical Pathology, Faculty of Medicine, Cairo UniversityCairo 11435, Egypt
| | - Anwar Shams
- Department of Pharmacology, College of Medicine, Taif UniversityP.O. Box 11099, Taif 21944, Saudi Arabia
- Research Center for Health Sciences, Deanship of Graduate Studies and Scientific Research, Taif UniversityTaif 26432, Saudi Arabia
- High Altitude Research Center, Taif UniversityP.O. Box 11099, Taif 21944, Saudi Arabia
| | - Shadi Tamur
- Department of Pediatric, College of Medicine, Taif UniversityP.O. Box 11099, Taif 21944, Saudi Arabia
| | - Emad A Mahdi
- Department of Pathology, Faculty of Veterinary Medicine, Beni-Suef UniversityBeni-Suef 62521, Egypt
| | - Osama M Ahmed
- Division of Physiology, Department of Zoology, Faculty of Science, Beni-Suef UniversityBeni-Suef 62521, Egypt
| |
Collapse
|
2
|
Bhogoju S, Khan S, Subramanian A. Continuous Low-Intensity Ultrasound Preserves Chondrogenesis of Mesenchymal Stromal Cells in the Presence of Cytokines by Inhibiting NFκB Activation. Biomolecules 2022; 12:434. [PMID: 35327626 PMCID: PMC8946190 DOI: 10.3390/biom12030434] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2022] [Revised: 02/21/2022] [Accepted: 03/03/2022] [Indexed: 02/04/2023] Open
Abstract
Proinflammatory joint environment, coupled with impeded chondrogenic differentiation of mesenchymal stromal cells (MSCs), led to inferior cartilage repair outcomes. Nuclear translocation of phosphorylated-NFκB downregulates SOX9 and hinders the chondrogenesis of MSCs. Strategies that minimize the deleterious effects of NFκB, while promoting MSC chondrogenesis, are of interest. This study establishes the ability of continuous low-intensity ultrasound (cLIUS) to preserve MSC chondrogenesis in a proinflammatory environment. MSCs were seeded in alginate:collagen hydrogels and cultured for 21 days in an ultrasound-assisted bioreactor (5.0 MHz, 2.5 Vpp; 4 applications/day) in the presence of IL1β and evaluated by qRT-PCR and immunofluorescence. The differential expression of markers associated with the NFκB pathway was assessed upon a single exposure of cLIUS and assayed by Western blotting, qRT-PCR, and immunofluorescence. Mitochondrial potential was evaluated by tetramethylrhodamine methyl ester (TMRM) assay. The chondroinductive potential of cLIUS was noted by the increased expression of SOX9 and COLII. cLIUS extended its chondroprotective effects by stabilizing the NFκB complex in the cytoplasm via engaging the IκBα feedback mechanism, thus preventing its nuclear translocation. cLIUS acted as a mitochondrial protective agent by restoring the mitochondrial potential and the mitochondrial mRNA expression in a proinflammatory environment. Altogether, our results demonstrated the potential of cLIUS for cartilage repair and regeneration under proinflammatory conditions.
Collapse
Affiliation(s)
| | | | - Anuradha Subramanian
- Department of Chemical and Materials Engineering, The University of Alabama in Huntsville, Huntsville, AL 35899, USA; (S.B.); (S.K.)
| |
Collapse
|
3
|
Dos Santos JF, Lazzarin MC, Baptista VIDA, Quintana HT, Ribeiro DA, de Oliveira F. Articular cartilage degeneration and bone adaptation due to lack of dystrophin in mice. J Bone Miner Metab 2022; 40:29-39. [PMID: 34549313 DOI: 10.1007/s00774-021-01270-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Accepted: 09/03/2021] [Indexed: 10/20/2022]
Abstract
INTRODUCTION Duchenne muscular dystrophy is caused by the absence of dystrophin. This study aimed to investigate femoral morphological characteristics of lack of dystrophin in MDX mice, considering that this model, different from DMD patient, is not influenced by corticosteroids administration and limited ambulation. MATERIALS AND METHODS Proximal femur of male 16-week-old Control and MDX mice were submitted to histological, morphometric (volume density of articular cartilage, compact bone, trabecular bone and bone marrow; articular cartilage layers area; articular cartilage cell area), and immunohistochemistry analysis for RUNX-2, RANK-L, MMP-2, MMP-9, Caspase-3 and KI-67. RESULTS MDX showed loss of linearity of articular cartilage with subchondral bone transition and elevation of this subchondral bone to the articular surface when compared with control. In addition, MDX presented morphological difference in the pantographic network of collagen fibers. Volume density of trabecular bone tissue was higher in the MDX than Control, but volume density of articular cartilage was lower in MDX (p < 0.05). The articular cartilage layers and chondrocytes area were significantly smaller in MDX than Control. These results associated to MMPs and osteogenic markers of proximal femur revealed an adaptation process as a consequence of lack of dystrophin. CONCLUSIONS The morphological changes observed in the bone tissue of the MDX may be not only secondary to muscle weakness or chronic use of corticosteroids but also our results indicate connections between decrease of cartilage thickness, collagen network alteration and consequent subchondral changes that may lead to articular cartilage degeneration and bone adaptation mechanism in MDX mice.
Collapse
Affiliation(s)
- José Fontes Dos Santos
- Departamento de Biociências, Universidade Federal de São Paulo (UNIFESP), Rua Silva Jardim, 136 Lab 328, CEP: 11015-020, Santos, SP, 11060-001, Brazil
| | - Mariana Cruz Lazzarin
- Departamento de Biociências, Universidade Federal de São Paulo (UNIFESP), Rua Silva Jardim, 136 Lab 328, CEP: 11015-020, Santos, SP, 11060-001, Brazil
| | - Vivianne Izabelle de Araújo Baptista
- Departamento de Biociências, Universidade Federal de São Paulo (UNIFESP), Rua Silva Jardim, 136 Lab 328, CEP: 11015-020, Santos, SP, 11060-001, Brazil
| | - Hananiah Tardivo Quintana
- Departamento de Biociências, Universidade Federal de São Paulo (UNIFESP), Rua Silva Jardim, 136 Lab 328, CEP: 11015-020, Santos, SP, 11060-001, Brazil
| | - Daniel Araki Ribeiro
- Departamento de Biociências, Universidade Federal de São Paulo (UNIFESP), Rua Silva Jardim, 136 Lab 328, CEP: 11015-020, Santos, SP, 11060-001, Brazil
| | - Flavia de Oliveira
- Departamento de Biociências, Universidade Federal de São Paulo (UNIFESP), Rua Silva Jardim, 136 Lab 328, CEP: 11015-020, Santos, SP, 11060-001, Brazil.
| |
Collapse
|
4
|
Bartolotti I, Roseti L, Petretta M, Grigolo B, Desando G. A Roadmap of In Vitro Models in Osteoarthritis: A Focus on Their Biological Relevance in Regenerative Medicine. J Clin Med 2021; 10:1920. [PMID: 33925222 PMCID: PMC8124812 DOI: 10.3390/jcm10091920] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Revised: 04/20/2021] [Accepted: 04/26/2021] [Indexed: 01/15/2023] Open
Abstract
Osteoarthritis (OA) is a multifaceted musculoskeletal disorder, with a high prevalence worldwide. Articular cartilage and synovial membrane are among the main biological targets in the OA microenvironment. Gaining more knowledge on the accuracy of preclinical in vitro OA models could open innovative avenues in regenerative medicine to bridge major gaps, especially in translation from animals to humans. Our methodological approach entailed searches on Scopus, the Web of Science Core Collection, and EMBASE databases to select the most relevant preclinical in vitro models for studying OA. Predicting the biological response of regenerative strategies requires developing relevant preclinical models able to mimic the OA milieu influencing tissue responses and organ complexity. In this light, standard 2D culture models lack critical properties beyond cell biology, while animal models suffer from several limitations due to species differences. In the literature, most of the in vitro models only recapitulate a tissue compartment, by providing fragmented results. Biotechnological advances may enable scientists to generate new in vitro models that combine easy manipulation and organ complexity. Here, we review the state-of-the-art of preclinical in vitro models in OA and outline how the different preclinical systems (inflammatory/biomechanical/microfluidic models) may be valid tools in regenerative medicine, describing their pros and cons. We then discuss the prospects of specific and combinatorial models to predict biological responses following regenerative approaches focusing on mesenchymal stromal cells (MSCs)-based therapies to reduce animal testing.
Collapse
Affiliation(s)
- Isabella Bartolotti
- Laboratorio RAMSES, IRCCS Istituto Ortopedico Rizzoli, via di Barbiano 1/10, 40136 Bologna, Italy; (I.B.); (M.P.); (B.G.)
| | - Livia Roseti
- Laboratorio RAMSES, IRCCS Istituto Ortopedico Rizzoli, via di Barbiano 1/10, 40136 Bologna, Italy; (I.B.); (M.P.); (B.G.)
| | - Mauro Petretta
- Laboratorio RAMSES, IRCCS Istituto Ortopedico Rizzoli, via di Barbiano 1/10, 40136 Bologna, Italy; (I.B.); (M.P.); (B.G.)
- RegenHu Company, Z.I Du Vivier 22, 1690 Villaz-St-Pierre, Switzerland
| | - Brunella Grigolo
- Laboratorio RAMSES, IRCCS Istituto Ortopedico Rizzoli, via di Barbiano 1/10, 40136 Bologna, Italy; (I.B.); (M.P.); (B.G.)
| | - Giovanna Desando
- Laboratorio RAMSES, IRCCS Istituto Ortopedico Rizzoli, via di Barbiano 1/10, 40136 Bologna, Italy; (I.B.); (M.P.); (B.G.)
| |
Collapse
|
5
|
Ragab GH, Halfaya FM, Ahmed OM, Abou El-Kheir W, Mahdi EA, Ali TM, Almehmadi MM, Hagag U. Platelet-Rich Plasma Ameliorates Monosodium Iodoacetate-Induced Ankle Osteoarthritis in the Rat Model via Suppression of Inflammation and Oxidative Stress. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE : ECAM 2021; 2021:6692432. [PMID: 33531920 PMCID: PMC7837774 DOI: 10.1155/2021/6692432] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/03/2020] [Revised: 12/19/2020] [Accepted: 01/07/2021] [Indexed: 12/26/2022]
Abstract
Until now, there is no treatment that cause complete cure of the chronic inflammatory and degenerative disease, osteoarthritis (OA). Moreover, the underlying mechanisms of OA development and progress are not fully elucidated, and the present pharmacological treatment alternatives are restricted and associated with adverse side effects. Thus, the present study was conducted to evaluate the role of platelet-rich plasma (PRP) in the remedy of OA in the rat model in terms of inflammation, ankle histopathological alterations, and oxidative stress. OA was induced in male Wistar rats by injection of MIA (2 mg)/50 µL isotonic saline in the right ankle joint for two successive days in each rat. After the 2nd MIA injection, the osteoarthritic rats were allocated into two groups such as the MIA group (group 2) and MIA + PRP group (group 3). The MIA + PRP group was treated with PRP (50 µL) by injection into the ankle joint of the right hind limb of each rat at days 14, 21, and 28 after the 2nd injection of MIA. The same equivalent volume of saline, as a substitute of PRP, was injected into the ankle joint of each rat of the normal control group (group 1) and MIA group (group 2) at the same tested periods. Swelling of joint, bodyweight, total leucocytes count (TLC), and morphological as well as histological changes of ankle joints were evaluated. Serum lipid peroxides (LPO), glutathione (GSH), and glutathione S-transferase (GST) levels were examined as biomarkers of oxidative stress. Serum tumor necrosis factor-α (TNF-α), interleukin-17 (IL-17), and interleukin-4 (IL-4) were investigated by ELISA as biomarkers of inflammation. In addition, magnetic resonance imaging (MRI) was carried out to investigate the soft tissues in joints. The obtained results revealed that PRP reduced LPO and increased GSH and GST levels in osteoarthritic rats. Also, PRP significantly diminished serum TNF-α and IL-17 levels, while it increased IL-4 serum levels in rats with MIA-induced OA. Morphological observations, histological analysis, and MRI revealed a gradual diminishing in joint inflammation and destruction of cartilage in PRP-injected osteoarthritic rats. Based on these results, it can be suggested that PRP has antiarthritic potential in MIA-induced OA, which may be mediated via suppression of inflammation and oxidative stress.
Collapse
Affiliation(s)
- G. H. Ragab
- Anesthesiology and Radiology Department, Faculty of Veterinary Medicine, Beni-Suef University, Beni-Suef, Egypt
| | - F. M. Halfaya
- Anesthesiology and Radiology Department, Faculty of Veterinary Medicine, Beni-Suef University, Beni-Suef, Egypt
| | - O. M. Ahmed
- Physiology Division, Zoology Department, Faculty of Science, Beni-Suef University, P.O. Box 62521, Beni-Suef, Egypt
| | - W. Abou El-Kheir
- Department of Immunology, Military Medical Academy, Cairo, Egypt
| | - E. A. Mahdi
- Pathology Department, Faculty of Veterinary Medicine, Beni-Suef University, Beni-Suef, Egypt
| | - T. M. Ali
- Physiology Department, College of Medicine, Taif University, Taif, Saudi Arabia
- Physiology Department, Faculty of Medicine, Beni-Suef University, Beni-Suef, Egypt
| | - M. M. Almehmadi
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Taif University, P.O. Box 11099, Taif 21944, Saudi Arabia
| | - U. Hagag
- Anesthesiology and Radiology Department, Faculty of Veterinary Medicine, Beni-Suef University, Beni-Suef, Egypt
| |
Collapse
|
6
|
Oh J, Son YS, Kim WH, Kwon OK, Kang BJ. Mesenchymal stem cells genetically engineered to express platelet-derived growth factor and heme oxygenase-1 ameliorate osteoarthritis in a canine model. J Orthop Surg Res 2021; 16:43. [PMID: 33430899 PMCID: PMC7802278 DOI: 10.1186/s13018-020-02178-4] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/07/2020] [Accepted: 12/25/2020] [Indexed: 01/14/2023] Open
Abstract
BACKGROUND Mesenchymal stem cells (MSCs) are used for the treatment of osteoarthritis (OA), and MSC genetic engineering is expected to enhance cartilage repair. Here, we aimed to investigate the effect of MSCs overexpressing platelet-derived growth factor (PDGF) or heme oxygenase-1 (HO-1) in chondrocytes and synovial cells with an OA phenotype and assess the in vivo efficacy of intra-articular injections of these MSCs in canine OA models. METHODS Canine adipose-derived MSCs were transfected with canine PDGF (PDGF-MSCs) or HO-1 (HO-1-MSCs) using lentiviral vectors. Canine chondrocytes or synovial cells were stimulated with lipopolysaccharide (LPS) to mimic the inflammatory OA model and then co-cultured with MSCs, PDGF-MSCs, or HO-1-MSCs for 24 h and 72 h. The mRNA levels of pro-inflammatory, extracellular matrix-degradative/synthetic, or pain-related factors were measured after co-culture by real-time PCR. Furthermore, a surgery-induced canine OA model was established and the dogs were randomized into four groups: normal saline (n = 4), MSCs (n = 4), PDGF-MSCs (n = 4), and HO-1-MSCs (n = 4). The OA symptoms, radiographic OA severity, and serum matrix metallopeptidase (MMP)-13 levels were assessed before and 10 weeks after treatment, to evaluate the safety and efficacy of the modified MSCs. RESULTS PDGF or HO-1 overexpression significantly reduced the expression of pro-inflammatory factors, MMP-13, and nerve growth factor elicited by LPS and increased that of aggrecan and collagen type 2 in chondrocytes (P < 0.05). In addition, the expression of aggrecanases was significantly downregulated in synovial cells, whereas that of tissue inhibitor of metalloproteinases was upregulated (P < 0.05). Furthermore, the co-cultured MSCs highly expressed genes that contributed to the maintenance of joint homeostasis (P < 0.05). In vivo studies showed that OA symptoms improved after administration of all MSCs. Also, PDGF-MSCs significantly improved limb function and reduced pain (P < 0.05). The results of the radiographic assessment and serum MMP-13 levels did not vary significantly compared to those of the control. CONCLUSIONS Genetically modifying PDGF and HO-1 in MSCs is an effective strategy for treating OA, suggesting that PDGF-MSCs can be novel therapeutic agents for improving OA symptoms.
Collapse
Affiliation(s)
- Jiwon Oh
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine and Research Institute for Veterinary Science, Seoul National University, Seoul, 08826, South Korea
| | - Yeon Sung Son
- Medical Research Center, College of Medicine, Seoul National University, Seoul, 03080, South Korea
| | - Wan Hee Kim
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine and Research Institute for Veterinary Science, Seoul National University, Seoul, 08826, South Korea
| | - Oh-Kyeong Kwon
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine and Research Institute for Veterinary Science, Seoul National University, Seoul, 08826, South Korea
| | - Byung-Jae Kang
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine and Research Institute for Veterinary Science, Seoul National University, Seoul, 08826, South Korea. .,BK21 PLUS Creative Veterinary Research Center, Seoul National University, Seoul, 08826, South Korea.
| |
Collapse
|
7
|
Xu Y, Wang YQ, Wang AT, Yu CY, Luo Y, Liu RM, Zhao YJ, Xiao JH. Effect of CD44 on differentiation of human amniotic mesenchymal stem cells into chondrocytes via Smad and ERK signaling pathways. Mol Med Rep 2020; 21:2357-2366. [PMID: 32236637 PMCID: PMC7185282 DOI: 10.3892/mmr.2020.11044] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2019] [Accepted: 02/28/2020] [Indexed: 01/22/2023] Open
Abstract
CD44 antigen (CD44) is a transmembrane protein found in cell adhesion molecules and is involved in the regulation of various physiological processes in cells. It was hypothesized that CD44 directly affected the chondrogenic differentiation of human amniotic mesenchymal stem cells (hAMSCs). In the present study, the expression of chondrocyte-associated factors was detected in the absence and presence of the antibody blocker anti-CD44 antibody during the chondrogenic differentiation of hAMSCs. Following inhibition of CD44 expression, the transcriptional levels of chondrocyte-associated genes SRY-box transcription factor 9, aggrecan and collagen type II α 1 chain, as well as the production of chondrocyte markers type II collagen and aggrecan were significantly decreased in hAMSCs. Further investigation indicated that there was no significant change in total ERK1/2 expression following inhibition of CD44 expression; however, phosphorylated (p)-ERK1/2 expression was decreased. The expression of p-Smad2/3 was also upregulated following CD44 inhibition. These data indicated that CD44 may affect the differentiation of hAMSCs into chondrocytes by regulating the Smad2/3 and ERK1/2 signaling pathway.
Collapse
Affiliation(s)
- Yan Xu
- Zunyi Municipal Key Laboratory of Medicinal Biotechnology, Institute of Medicinal Biotechnology, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563003, P.R. China
| | - Yi-Qing Wang
- Zunyi Municipal Key Laboratory of Medicinal Biotechnology, Institute of Medicinal Biotechnology, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563003, P.R. China
| | - Ai-Tong Wang
- Zunyi Municipal Key Laboratory of Medicinal Biotechnology, Institute of Medicinal Biotechnology, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563003, P.R. China
| | - Chang-Yin Yu
- Center for Translational Medicine, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563003, P.R. China
| | - Yi Luo
- Zunyi Municipal Key Laboratory of Medicinal Biotechnology, Institute of Medicinal Biotechnology, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563003, P.R. China
| | - Ru-Ming Liu
- Zunyi Municipal Key Laboratory of Medicinal Biotechnology, Institute of Medicinal Biotechnology, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563003, P.R. China
| | - Yu-Jie Zhao
- Department of Laboratory Medicine, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563003, P.R. China
| | - Jian-Hui Xiao
- Zunyi Municipal Key Laboratory of Medicinal Biotechnology, Institute of Medicinal Biotechnology, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563003, P.R. China
| |
Collapse
|
8
|
Shortt C, Luyt LG, Turley EA, Cowman MK, Kirsch T. A Hyaluronan-binding Peptide (P15-1) Reduces Inflammatory and Catabolic Events in IL-1β-treated Human Articular Chondrocytes. Sci Rep 2020; 10:1441. [PMID: 31996703 PMCID: PMC6989647 DOI: 10.1038/s41598-020-57586-7] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2019] [Accepted: 01/03/2020] [Indexed: 01/01/2023] Open
Abstract
Inflammation plays a critical role in osteoarthritis (OA). It stimulates catabolic events in articular chondrocytes and prevents chondrogenic precursor cells from repairing cartilage lesions, leading to accelerated cartilage degradation. Therefore, the identification of novel factors that reduce catabolic events in chondrocytes and enhances chondrogenic differentiation of precursor cells in an inflammatory environment may provide novel therapeutic strategies for the treatment of OA. The goal of this study was to determine whether a hyaluronan (HA)-binding peptide (P15-1), via interacting with high molecular weight (HMW)HA can enhance the anti-inflammatory properties of HMWHA and decrease catabolic events in interleukin-1beta (IL-1β)-treated human articular chondrocytes. Treatment with P15-1 decreased catabolic events and stimulated anabolic events in articular chondrocytes cultured in an inflammatory environment. P15-1 pre-mixed with HMWHA was more effective in inhibiting catabolic events and stimulating anabolic events than P15-1 or HMWHA alone. Our findings suggest that P15-1 together with HMWHA inhibits catabolic events in articular chondrocytes via the inhibition of p38 mitogen-activated protein kinases (MAPK) and increasing the thickness of the pericellular matrix (PCM) around chondrocytes thereby decreasing catabolic signaling. Finally, conditioned medium from IL-1β and P15-1-treated human articular chondrocytes was less inhibitory for chondrogenic differentiation of precursor cells than conditioned medium from chondrocytes treated with IL-1β alone. In conclusion, P15-1 is proposed to function synergistically with HMWHA to enhance the protective microenvironment for chondrocytes and mesenchymal stem cells during inflammation and regeneration.
Collapse
Affiliation(s)
- Claire Shortt
- Musculoskeletal Research Center, Department of Orthopaedic Surgery, New York University School of Medicine, New York, NY, USA
- FoodMarble Digestive Health, Dublin, 2, Ireland
| | - Leonard G Luyt
- The University of Western Ontario, London, ON, Canada
- London Regional Cancer Program, Lawson Health Research Institute, London, ON, Canada
| | - Eva A Turley
- The University of Western Ontario, London, ON, Canada
- London Regional Cancer Program, Lawson Health Research Institute, London, ON, Canada
| | - Mary K Cowman
- Musculoskeletal Research Center, Department of Orthopaedic Surgery, New York University School of Medicine, New York, NY, USA
- Department of Biomedical Engineering, New York University Tandon School of Engineering, New York, NY, USA
| | - Thorsten Kirsch
- Musculoskeletal Research Center, Department of Orthopaedic Surgery, New York University School of Medicine, New York, NY, USA.
- Department of Biomedical Engineering, New York University Tandon School of Engineering, New York, NY, USA.
| |
Collapse
|
9
|
Cheng BF, Lian JJ, Yang HJ, Wang L, Yu HH, Bi JJ, Gao YX, Chen SJ, Wang M, Feng ZW. Neural cell adhesion molecule regulates chondrocyte hypertrophy in chondrogenic differentiation and experimental osteoarthritis. Stem Cells Transl Med 2019; 9:273-283. [PMID: 31742919 PMCID: PMC6988767 DOI: 10.1002/sctm.19-0190] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2019] [Accepted: 10/25/2019] [Indexed: 12/28/2022] Open
Abstract
Chondrocyte hypertrophy-like change is an important pathological process of osteoarthritis (OA), but the mechanism remains largely unknown. Neural cell adhesion molecule (NCAM) is highly expressed and involved in the chondrocyte differentiation of mesenchymal stem cells (MSCs). In this study, we found that NCAM deficiency accelerates chondrocyte hypertrophy in articular cartilage and growth plate of OA mice. NCAM deficiency leads to hypertrophic chondrocyte differentiation in both murine MSCs and chondrogenic cells, in which extracellular signal-regulated kinase (ERK) signaling plays an important role. Moreover, NCAM expression is downregulated in an interleukin-1β-stimulated OA cellular model and monosodium iodoacetate-induced OA rats. Overexpression of NCAM substantially inhibits hypertrophic differentiation in the OA cellular model. In conclusion, NCAM could inhibit hypertrophic chondrocyte differentiation of MSCs by inhibiting ERK signaling and reduce chondrocyte hypertrophy in experimental OA model, suggesting the potential utility of NCAM as a novel therapeutic target for alleviating chondrocyte hypertrophy of OA.
Collapse
Affiliation(s)
- Bin-Feng Cheng
- School of Life Sciences and Technology, Xinxiang Medical University, Xinxiang, Henan, People's Republic of China
| | - Jun-Jiang Lian
- School of Life Sciences and Technology, Xinxiang Medical University, Xinxiang, Henan, People's Republic of China.,Shanghai Key Laboratory of Signaling and Disease Research, Laboratory of Receptor-based Bio-medicine, Collaborative Innovation Center for Brain Science, School of Life Sciences and Technology, Tongji University, Shanghai, People's Republic of China
| | - Hai-Jie Yang
- School of Life Sciences and Technology, Xinxiang Medical University, Xinxiang, Henan, People's Republic of China
| | - Lei Wang
- School of Life Sciences and Technology, Xinxiang Medical University, Xinxiang, Henan, People's Republic of China
| | - Hao-Heng Yu
- Henan Key Laboratory of Medical Tissue Regeneration, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, Henan, People's Republic of China
| | - Jia-Jia Bi
- School of Life Sciences and Technology, Xinxiang Medical University, Xinxiang, Henan, People's Republic of China
| | - Yao-Xin Gao
- School of Life Sciences and Technology, Xinxiang Medical University, Xinxiang, Henan, People's Republic of China
| | - Su-Juan Chen
- School of Life Sciences and Technology, Xinxiang Medical University, Xinxiang, Henan, People's Republic of China
| | - Mian Wang
- School of Life Sciences and Technology, Xinxiang Medical University, Xinxiang, Henan, People's Republic of China
| | - Zhi-Wei Feng
- School of Life Sciences and Technology, Xinxiang Medical University, Xinxiang, Henan, People's Republic of China
| |
Collapse
|
10
|
Zhao X, Li Y, Lin X, Wang J, Zhao X, Xie J, Sun T, Fu Z. Ozone induces autophagy in rat chondrocytes stimulated with IL-1β through the AMPK/mTOR signaling pathway. J Pain Res 2018; 11:3003-3017. [PMID: 30568481 PMCID: PMC6267635 DOI: 10.2147/jpr.s183594] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Background Ozone injection is generally used for the management of pain in diseases such as osteoarthritis (OA). Recent studies have shown that reduced autophagy in chondrocytes plays an important role in the development of OA. The purpose of this study was to determine whether ozone treats OA by inducing autophagy in OA chondrocytes. Materials and methods In this study, primary chondrocytes were stimulated with IL-1β for 24 hours to simulate an OA chondrocyte model, followed by treatment with ozone (30 µg/ mL) or pretreatment with 3-methyladenine or compound C before ozone treatment. Then, cell viability was detected by a CCK-8 kit, and the AMPK/mTOR signaling pathway and autophagy were detected by Western blotting and immunofluorescence. The mRNA expression levels of IL-6, TNF-α, MMP-13 and TIMP-1 were measured by quantitative real-time PCR. Finally, autophagosomes in chondrocytes were observed by transmission electron microscopy. Results Ozone improved cell viability in chondrocytes stimulated by IL-1β. The decreased level of autophagy in IL-1β-stimulated chondrocytes improved with ozone treatment through activation of the AMPK/mTOR signaling pathway. In addition, the mRNA expression levels of IL-6 and TNF-α were suppressed by ozone treatment in chondrocytes stimulated with IL-1β. Ozone increased the mRNA level of TIMP-1 and decreased the mRNA level of MMP-13 in chondrocytes stimulated with IL-1β. Conclusion These results suggested that ozone improved the decreased level of autophagy in chondrocytes stimulated with IL-1β through activation of the AMPK/mTOR signaling pathway. Moreover, ozone treatment suppressed inflammation and helped maintain metabolic balance in chondrocytes stimulated with IL-1β.
Collapse
Affiliation(s)
- Xu Zhao
- Department of Pain Management, Shandong Provincial Hospital Affiliated to Shandong University, Shandong University, Jinan, Shandong Province 250021, P.R. China,
| | - Yun Li
- Department of Pain Management, Shandong Provincial Hospital Affiliated to Shandong University, Shandong University, Jinan, Shandong Province 250021, P.R. China,
| | - Xiaowen Lin
- Department of Pain Management, Shandong Provincial Hospital Affiliated to Shandong University, Shandong University, Jinan, Shandong Province 250021, P.R. China,
| | - Junnan Wang
- Department of Pain Management, Shandong Provincial Hospital Affiliated to Shandong University, Shandong University, Jinan, Shandong Province 250021, P.R. China,
| | - Xuejun Zhao
- Department of Pain Management, Shandong Provincial Hospital Affiliated to Shandong University, Shandong University, Jinan, Shandong Province 250021, P.R. China,
| | - Juntian Xie
- Department of Pain Management, Shandong Provincial Hospital Affiliated to Shandong University, Shandong University, Jinan, Shandong Province 250021, P.R. China,
| | - Tao Sun
- Department of Pain Management, Shandong Provincial Hospital Affiliated to Shandong University, Shandong University, Jinan, Shandong Province 250021, P.R. China,
| | - Zhijian Fu
- Department of Pain Management, Shandong Provincial Hospital Affiliated to Shandong University, Shandong University, Jinan, Shandong Province 250021, P.R. China,
| |
Collapse
|
11
|
Luteolin inhibits IL-1β-induced inflammation in rat chondrocytes and attenuates osteoarthritis progression in a rat model. Biomed Pharmacother 2018; 109:1586-1592. [PMID: 30551412 DOI: 10.1016/j.biopha.2018.09.161] [Citation(s) in RCA: 147] [Impact Index Per Article: 24.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2018] [Revised: 09/28/2018] [Accepted: 09/28/2018] [Indexed: 01/01/2023] Open
Abstract
Osteoarthritis (OA) is a joint disease characterized by inflammation and cartilage degradation. Accumulating evidence has demonstrated that luteolin, a natural flavonoid, has anti-inflammatory and anticatabolic effects. The present study aimed to assess the protective effect of luteolin on interleukin (IL)-1β-stimulated rat chondrocytes and a monosodium iodoacetate (MIA)-induced model of OA. Rat chondrocytes were pretreated with luteolin (0, 25, 50, and 100 μM for 12 h) prior to stimulation with IL-1β (10 ng/ml for 24 h). Nitric oxide (NO) production was determined using the Griess method. Production of prostaglandin E2 (PGE2), tumor necrosis factor-α (TNF-α), and matrix metalloproteinase-2, -8, and -9 (MMP-2, MMP-8 and MMP-9) was measured by an enzyme-linked immunosorbent assay (ELISA). Protein levels of inducible nitric oxide synthase (iNOS), cyclooxygenase-2 (COX-2), MMP-1, MMP-3, MMP-13, p65, p-p65, IκB, and p-IκB were determined by Western blotting. The OA rats received luteolin (10 mg/kg/day) by gavage in vivo. Morphological and ultrastructural scanning electron microscopy (SEM) observations were performed to assess the severity of OA at 45 days following MIA injection. Collagen II protein expression was determined by immunohistochemistry. In this study, luteolin considerably reduced the IL-1β-induced production of NO, PGE2, TNF-α, MMP-2, MMP-8 and MMP-9 and the expression of COX-2, iNOS, MMP-1, MMP-3 and MMP-13. Luteolin reversed the degradation of collagen II induced by IL-1β. Luteolin also significantly inhibited IL-1β-induced phosphorylation of NF-κB in vitro. Luteolin treatment prevented cartilage destruction and enhanced collagen II expression in OA rats in vivo. Overall, our findings suggest that luteolin may be a useful therapeutic agent for patients with OA.
Collapse
|
12
|
Choi SM, Lee KM, Ryu SB, Park YJ, Hwang YG, Baek D, Choi Y, Park KH, Park KD, Lee JW. Enhanced articular cartilage regeneration with SIRT1-activated MSCs using gelatin-based hydrogel. Cell Death Dis 2018; 9:866. [PMID: 30158625 PMCID: PMC6115405 DOI: 10.1038/s41419-018-0914-1] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2018] [Revised: 06/26/2018] [Accepted: 07/09/2018] [Indexed: 12/27/2022]
Abstract
To investigate the functional effects of resveratrol (RSV) on mesenchymal stem cells (MSCs), we treated MSCs with RSV continuously during ex vivo expansion. MSCs were continuously treated with RSV from passage (P) 0 to P5. A proliferative capacity of RSV-treated MSCs was higher than that of non-treated MSCs and similar with P1-MSCs. Continuous treatment of RSV on MSCs increased the stemness and inhibited the senescence. During chondrogenic differentiation in vitro, RSV-treated MSCs had higher differentiation potential and reduced hypertrophic maturation, which are limitations for hyaline cartilage formation. The histological analysis of micromass demonstrated increased chondrogenic differentiation potential. We further explored the therapeutic effectiveness of this method in a rabbit osteochondral defect model. A rabbit osteochondral defect model was established to investigate the hyaline cartilage regeneration potential of RSV-treated MSCs. Moreover, the cartilage regeneration potential of RSV-treated MSCs was greater than that of untreated MSCs. The expression levels of chondrogenic markers increased and those of hypertrophic markers decreased in RSV-treated MSCs compared with untreated MSCs. Sustained treatment of RSV on MSCs during ex vivo expansion resulted in the maintenance of stemness and enhanced chondrogenic differentiation potential. Consequentially, highly efficient MSCs promoted superior hyaline cartilage regeneration in vivo. This novel treatment method provides a basis for cell-based tissue engineering.
Collapse
Affiliation(s)
- Seong Mi Choi
- Department of Orthopaedic Surgery, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, Seoul, 03722, South Korea
- Brain Korea 21 PLUS Project for Medical Sciences, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, Seoul, 03722, South Korea
| | - Kyoung-Mi Lee
- Department of Orthopaedic Surgery, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, Seoul, 03722, South Korea
- Severance Biomedical Science Institute, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, Seoul, 03722, South Korea
| | - Seung Bae Ryu
- Department of Molecular Science and Technology/Applied Chemistry and Biological Engineering, Ajou University, 206, World cup-ro Yeongtong-gu, Suwon-si, Gyeonggi-do, 16499, South Korea
| | - Yoo Jung Park
- Department of Orthopaedic Surgery, Yonsei University Wonju College of Medicine, 20, Ilsan-ro, Wonju-si, Gangwon-do, 26426, South Korea
| | - Yeok Gu Hwang
- Department of Orthopaedic Surgery, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, Seoul, 03722, South Korea
| | - Dawoon Baek
- Department of Orthopaedic Surgery, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, Seoul, 03722, South Korea
- Brain Korea 21 PLUS Project for Medical Sciences, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, Seoul, 03722, South Korea
| | - Yoorim Choi
- Department of Orthopaedic Surgery, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, Seoul, 03722, South Korea
- Brain Korea 21 PLUS Project for Medical Sciences, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, Seoul, 03722, South Korea
| | - Kwang Hwan Park
- Department of Orthopaedic Surgery, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, Seoul, 03722, South Korea
| | - Ki Dong Park
- Department of Molecular Science and Technology/Applied Chemistry and Biological Engineering, Ajou University, 206, World cup-ro Yeongtong-gu, Suwon-si, Gyeonggi-do, 16499, South Korea
| | - Jin Woo Lee
- Department of Orthopaedic Surgery, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, Seoul, 03722, South Korea.
- Brain Korea 21 PLUS Project for Medical Sciences, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, Seoul, 03722, South Korea.
- Severance Biomedical Science Institute, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, Seoul, 03722, South Korea.
| |
Collapse
|
13
|
van den Akker GG, van Beuningen HM, Vitters EL, Koenders MI, van de Loo FA, van Lent PL, Blaney Davidson EN, van der Kraan PM. Interleukin 1 β-induced SMAD2/3 linker modifications are TAK1 dependent and delay TGFβ signaling in primary human mesenchymal stem cells. Cell Signal 2017; 40:190-199. [PMID: 28943409 DOI: 10.1016/j.cellsig.2017.09.010] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2017] [Revised: 09/07/2017] [Accepted: 09/14/2017] [Indexed: 01/30/2023]
Abstract
BACKGROUND Chondrogenic differentiation of mesenchymal stem cells (MSC) requires transforming growth factor beta (TGFβ) signaling. TGFβ binds to the type I receptor activin-like kinase (ALK)5 and results in C-terminal SMAD2/3 phosphorylation (pSMAD2/3C). In turn pSMAD2/3C translocates to the nucleus and regulates target gene expression. Inflammatory mediators are known to exert an inhibitory effect on MSC differentiation. In this study we investigated the effect of interleukin 1 β (IL1β) on SMAD2/3 signaling dynamics and post-translational modifications. RESULTS Co-stimulation of MSC with TGFβ and IL1β did not affect peak pSMAD2C levels at 1h post-stimulation. Surprisingly, SMAD3 transcriptional activity, as determined by the CAGA12-luciferase reporter construct, was enhanced by co-stimulation of TGFβ and IL1β compared to TGFβ alone. Furthermore, IL1β stimulation induced CAGA12-luciferase activity in a SMAD dependent way. As SMAD function can be modulated independent of canonical TGFβ signaling through the SMAD linker domain, we studied SMAD2 linker phosphorylation at specific threonine and serine residues. SMAD2 linker threonine and serine modifications were observed within 1h following TGFβ, IL1β or TGFβ and IL1β stimulation. Upon co-stimulation linker modified SMAD2 accumulated in the cytoplasm and SMAD2/3 target gene transcription (ID1, JUNB) at 2-4h was inhibited. A detailed time course analysis of IL1β-induced SMAD2 linker modifications revealed a distinct temperospatial pattern compared to TGFβ. Co-stimulation with both factors resulted in a similar kinetic profile as TGFβ alone. Nevertheless, IL1β did subtly alter TGFβ-induced pSMAD2C levels between 8 and 24h post-stimulation, which was reflected by TGFβ target gene expression (PAI1, JUNB). Direct evidence for the importance of SMAD3 linker modifications for the effect of IL1β on TGFβ signaling was obtained by over-expression of SMAD3 or a SMAD3 linker phospho-mutant. Finally, an inhibitor screening was performed to identify kinases involved in SMAD2/3 linker modifications. We identified TAK1 kinase activity as crucial for IL1β-induced SMAD2 linker modifications and CAGA12-luciferase activity. CONCLUSIONS TGFβ and IL1β signaling interact at the SMAD2/3 level in human primary MSC. Down-stream TGFβ target genes were repressed by IL1β independent of C-terminal SMAD2 phosphorylation. We demonstrate that SMAD2/3 linker modifications are required for this interplay and identified TAK1 as a crucial mediator of IL1β-induced TGFβ signal modulation.
Collapse
Affiliation(s)
- Guus G van den Akker
- Radboud University Medical Center, Department of Rheumatology, Experimental Rheumatology, PO Box 9101, 6500 HB Nijmegen, The Netherlands
| | - Henk M van Beuningen
- Radboud University Medical Center, Department of Rheumatology, Experimental Rheumatology, PO Box 9101, 6500 HB Nijmegen, The Netherlands
| | - Elly L Vitters
- Radboud University Medical Center, Department of Rheumatology, Experimental Rheumatology, PO Box 9101, 6500 HB Nijmegen, The Netherlands
| | - Marije I Koenders
- Radboud University Medical Center, Department of Rheumatology, Experimental Rheumatology, PO Box 9101, 6500 HB Nijmegen, The Netherlands
| | - Fons A van de Loo
- Radboud University Medical Center, Department of Rheumatology, Experimental Rheumatology, PO Box 9101, 6500 HB Nijmegen, The Netherlands
| | - Peter L van Lent
- Radboud University Medical Center, Department of Rheumatology, Experimental Rheumatology, PO Box 9101, 6500 HB Nijmegen, The Netherlands
| | - Esmeralda N Blaney Davidson
- Radboud University Medical Center, Department of Rheumatology, Experimental Rheumatology, PO Box 9101, 6500 HB Nijmegen, The Netherlands
| | - Peter M van der Kraan
- Radboud University Medical Center, Department of Rheumatology, Experimental Rheumatology, PO Box 9101, 6500 HB Nijmegen, The Netherlands.
| |
Collapse
|
14
|
Hsu YH, Yang YY, Huwang MH, Weng YH, Jou IM, Wu PT, Lin TY, Wu LW, Chang MS. Anti-IL-20 monoclonal antibody inhibited inflammation and protected against cartilage destruction in murine models of osteoarthritis. PLoS One 2017; 12:e0175802. [PMID: 28426699 PMCID: PMC5398531 DOI: 10.1371/journal.pone.0175802] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2016] [Accepted: 03/06/2017] [Indexed: 12/17/2022] Open
Abstract
Osteoarthritis (OA) is a degenerative joint disease characterized by progressive destruction of articular cartilage. Interleukin (IL)-20 is a proinflammatory cytokine involved in the pathogenesis of rheumatoid arthritis. We investigated the role of IL-20 in OA and evaluated whether anti-IL-20 antibody (7E) treatment attenuates disease severity in murine models of surgery-induced OA. Immunohistochemical staining was used to detect IL-20 and its receptors expression in synovial tissue and cartilage from OA patients, and in OA synovial fibroblasts (OASFs) and chondrocytes (OACCs) from rodents with surgery-induced OA. RTQ-PCR and western blotting were used to determine IL-20-regulated OA-associated gene expression in OASFs and OACCs. OA rats and OA mice were treated with 7E. Arthritis severity was determined based on the degree of cartilage damage and the arthritis severity score. We found that IL-20 and its receptors were expressed in OASFs and OACCs. IL-20 induced TNF-α, IL-1β, MMP-1, and MMP-13 expression by activating ERK-1/2 and JNK signals in OASFs. IL-20 not only upregulated MCP-1, IL-6, MMP-1, and MMP-13 expression, but also downregulated aggrecan, type 2 collagen, TGF-β, and BMP-2 expression in OACCs. Arthritis severity was significantly lower in 7E-treated OA rats, and 7E- or MSC-treated OA mice. Therefore, we concluded that IL-20 was involved in the progression and development of OA through inducing proinflammatory cytokines and OA-associated gene expression in OASFs and OACCs. 7E reduced the severity of arthritis in murine models of surgery-induced OA. Our findings provide evidence that IL-20 is a novel target and that 7E is a potential therapeutic agent for OA.
Collapse
Affiliation(s)
- Yu-Hsiang Hsu
- Institute of Clinical Medicine, College of Medicine, National Cheng Kung University, Tainan, Taiwan.,Research Center of Clinical Medicine, National Cheng Kung University Hospital, Tainan, Taiwan.,Research Center of New Antibody Drug, National Cheng Kung University, Tainan, Taiwan
| | - Ya-Yu Yang
- Department of Biochemistry and Molecular Biology, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Man-Hsiang Huwang
- Department of Biochemistry and Molecular Biology, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Yun-Han Weng
- Research Center of New Antibody Drug, National Cheng Kung University, Tainan, Taiwan.,Department of Biochemistry and Molecular Biology, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - I-Ming Jou
- Department of Orthopedics, National Cheng Kung University Hospital, Tainan, Taiwan
| | - Po-Tin Wu
- Department of Orthopedics, National Cheng Kung University Hospital, Tainan, Taiwan
| | - Tain-Yu Lin
- Institute of Clinical Medicine, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Li-Wha Wu
- Institute of Molecular Medicine, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Ming-Shi Chang
- Research Center of New Antibody Drug, National Cheng Kung University, Tainan, Taiwan.,Department of Biochemistry and Molecular Biology, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| |
Collapse
|
15
|
Hassan Famian M, Montazer Saheb S, Montaseri A. Conditioned Medium of Wharton's Jelly Derived Stem Cells Can Enhance the Cartilage Specific Genes Expression by Chondrocytes in Monolayer and Mass Culture Systems. Adv Pharm Bull 2017; 7:123-130. [PMID: 28507946 PMCID: PMC5426725 DOI: 10.15171/apb.2017.016] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2016] [Revised: 02/28/2017] [Accepted: 03/07/2017] [Indexed: 12/13/2022] Open
Abstract
Purpose: Mesenchymal stem cells (MSCs) have been introduced for cell therapy strategies in osteoarthritis (OA). Despite of their capacity for differentiation into chondrocyte, there are some evidences about their life-threatening problem after transplantation. So, some researchers shifted on the application of stem cells conditioned medium. The goal of this study is to evaluate whether Wharton's jelly derived stem cell conditioned medium (WJSCs-CM) can enhance the gene expression profile by chondrocytes in monolayer and mass culture systems. Methods: Conditioned medium was obtained from WJSCs at fourth passage. Isolated chondrocytes were plated at density of 1×106 for both monolayer and high density culture. Then cells in both groups were divided into control (received medium) and experiment group treated with WJ-CM for 3 and 6 days. Samples were prepared to evaluate gene expression profile of collagen II, aggrecan, cartilage oligomeric matrix protein (COMP) and sox-9 using real-time RT-PCR. Results: After 3 days, Chondrocytes treated with WJSCs-CM expressed significantly higher level of genes compared to the control group in both culture systems. After 6 days, the expression of genes in monolayer cultivated chondrocytes was decreased but that of the mass culture were up-regulated significantly. Conclusion: WJ-SCs-CM can increase the expression of cartilage-specific genes and can be introduced as a promoting factor for cartilage regeneration.
Collapse
Affiliation(s)
- Maryam Hassan Famian
- Department of molecular biology, Ahar Branch, Islamic Azad University, Ahar, Iran
| | | | - Azadeh Montaseri
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
16
|
Potencial regenerativo do tecido cartilaginoso por células‐tronco mesenquimais: atualização, limitações e desafios. Rev Bras Ortop 2017. [DOI: 10.1016/j.rbo.2016.02.007] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
|
17
|
Cruz IBMD, Severo AL, Azzolin VF, Garcia LFM, Kuhn A, Lech O. Regenerative potential of the cartilaginous tissue in mesenchymal stem cells: update, limitations, and challenges. Rev Bras Ortop 2017; 52:2-10. [PMID: 28194374 PMCID: PMC5290078 DOI: 10.1016/j.rboe.2016.11.005] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2016] [Accepted: 02/15/2016] [Indexed: 12/26/2022] Open
Abstract
Advances in the studies with adult mesenchymal stem cells (MSCs) have turned tissue regenerative therapy into a promising tool in many areas of medicine. In orthopedics, one of the main challenges has been the regeneration of cartilage tissue, mainly in diarthroses. In the induction of the MSCs, in addition to cytodifferentiation, the microenvironmental context of the tissue to be regenerated and an appropriate spatial arrangement are extremely important factors. Furthermore, it is known that MSC differentiation is fundamentally determined by mechanisms such as cell proliferation (mitosis), biochemical-molecular interactions, movement, cell adhesion, and apoptosis. Although the use of MSCs for cartilage regeneration remains at a research level, there are important questions to be resolved in order to make this therapy efficient and safe. It is known, for instance, that the expansion of chondrocytes in cultivation, needed to increase the number of cells, could end up producing fibrocartilage instead of hyaline cartilage. However, the latest results are promising. In 2014, the first stage I/II clinical trial to evaluate the efficacy and safety of the intra-articular injection of MSCs in femorotibial cartilage regeneration was published, indicating a decrease in injured areas. One issue to be explored is how many modifications in the articulate inflammatory environment could induce differentiation of MSCs already allocated in that region. Such issue arose from studies that suggested that the suppression of the inflammation may increase the efficiency of tissue regeneration. Considering the complexity of the events related to the chondrogenesis and cartilage repair, it can be concluded that the road ahead is still long, and that further studies are needed.
Collapse
Affiliation(s)
- Ivana Beatrice Mânica da Cruz
- Universidade Federal de Santa Maria (UFSM), Centro de Ciências da Saúde, Santa Maria, RS, Brazil
- Universidade Federal de Santa Maria (UFSM), Laboratório de Biogenômica, Santa Maria, RS, Brazil
| | - Antônio Lourenço Severo
- Universidade Federal da Fronteira Sul (UFFS), Hospital São Vicente de Paulo, Instituto de Ortopedia e Traumatologia, Passo Fundo, RS, Brazil
| | - Verônica Farina Azzolin
- Universidade Federal de Santa Maria (UFSM), Laboratório de Biogenômica, Santa Maria, RS, Brazil
| | | | - André Kuhn
- Universidade Federal da Fronteira Sul (UFFS), Hospital São Vicente de Paulo, Instituto de Ortopedia e Traumatologia, Passo Fundo, RS, Brazil
| | - Osvandré Lech
- Universidade Federal da Fronteira Sul (UFFS), Hospital São Vicente de Paulo, Instituto de Ortopedia e Traumatologia, Passo Fundo, RS, Brazil
| |
Collapse
|
18
|
Lian JJ, Cheng BF, Gao YX, Xue H, Wang L, Wang M, Yang HJ, Feng ZW. Protective effect of kaempferol, a flavonoid widely present in varieties of edible plants, on IL-1β-induced inflammatory response via inhibiting MAPK, Akt, and NF-κB signalling in SW982 cells. J Funct Foods 2016. [DOI: 10.1016/j.jff.2016.09.003] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
|
19
|
Manoto SL, Maepa MJ, Motaung SK. Medical ozone therapy as a potential treatment modality for regeneration of damaged articular cartilage in osteoarthritis. Saudi J Biol Sci 2016; 25:672-679. [PMID: 29736142 PMCID: PMC5935866 DOI: 10.1016/j.sjbs.2016.02.002] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2015] [Revised: 01/19/2016] [Accepted: 02/01/2016] [Indexed: 12/19/2022] Open
Abstract
Osteoarthritis (OA) is the most common degenerative joint disease and a growing health problem affecting more than half of the population over the age of 65. It is characterized by inflammation in the cartilage and synovium, resulting in the loss of joint structure and progressive damage to the cartilage. Many pro-inflammatory mediators are elevated in OA, including reactive oxygen species (ROS) such as nitric oxide (NO) and hydrogen peroxide (H2O2). Damaged articular cartilage remains a challenge to treat due to the limited self-healing capacity of the tissue and unsuccessful biological interventions. This highlights the need for better therapeutic strategies to heal damaged articular cartilage. Ozone (O3) therapy has been shown to have positive results in the treatment of OA; however the use of O3 therapy as a therapeutic agent is controversial. There is a perception that O3 is always toxic, whereas evidence indicates that when it is applied following a specified method, O3 can be effective in the treatment of degenerative diseases. The mechanism of action of O3 therapy in OA is not fully understood and this review summarizes the use of O3 therapy in the treatment of damaged articular cartilage in OA.
Collapse
Affiliation(s)
- Sello Lebohang Manoto
- Faculty of Science, Department of Biomedical Sciences, Tshwane University of Technology, Pretoria, South Africa
| | - Makwese Johaness Maepa
- Faculty of Science, Department of Biomedical Sciences, Tshwane University of Technology, Pretoria, South Africa
| | - Shirley Keolebogile Motaung
- Faculty of Science, Department of Biomedical Sciences, Tshwane University of Technology, Pretoria, South Africa
| |
Collapse
|