1
|
Jin Y, Mao F, Wang X, Zhang J, Gao Y, Fan Y. The sonic hedgehog signaling inhibitor cyclopamine improves pulmonary arterial hypertension via regulating the bone morphogenetic protein receptor 2 pathway. Sci Rep 2025; 15:12512. [PMID: 40216933 PMCID: PMC11992095 DOI: 10.1038/s41598-025-97627-7] [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: 08/08/2024] [Accepted: 04/07/2025] [Indexed: 04/14/2025] Open
Abstract
Pulmonary arterial hypertension (PAH) is a severe and progressive disease with hallmarks of pulmonary vascular remodeling and bone morphogenetic protein receptor 2 (BMPR2) mutation. Recent studies indicate Sonic hedgehog (SHH) signaling is involved in the proliferation of human pulmonary arterial smooth muscle cells (hPASMCs) but the role of the SHH signaling inhibitor cyclopamine in monocrotaline (MCT)-induced PAH has not been investigated. We hypothesized SHH promotes pulmonary vascular remodeling and that inhibition of SHH signaling by cyclopamine could attenuate pulmonary hypertension via the bone morphogenetic protein (BMP) pathway. SHH and BMPR2 proteins were measured in pulmonary arteries isolated from MCT-induced PAH rats and in hPASMCs. The therapeutic effects of cyclopamine were tested in PAH rats and in BMPR2 knockdown hPASMCs. SHH protein levels were increased in PAH rats and exogenous recombinant SHH protein promoted proliferation of hPASMCs via BMPR2 and osteopontin. Furthermore, cyclopamine attenuated hemodynamics and vascular remodeling via the BMP pathway in PAH rats. Finally, cyclopamine enhanced apoptosis and reduced proliferation in hPASMCs with impaired BMPR2. The findings of this study provide evidence that SHH has a role in pulmonary vascular remodeling via BMP4/BMPR2/ID1, and its inhibition by cyclopamine could be a potential therapeutic target in PAH.
Collapse
Affiliation(s)
- Youpeng Jin
- Department of Pediatrics, Shandong Provincial Hospital Affiliated to Shandong First Medical University, 324 Jingwuweiqi Road, Jinan, 250014, Shandong, PR China
| | - Fei Mao
- Department of Pediatrics, Shandong Provincial Hospital Affiliated to Shandong First Medical University, 324 Jingwuweiqi Road, Jinan, 250014, Shandong, PR China
- Department of Neurology, The First Affiliated Hospital of Shandong First Medical University, Jinan, 250014, PR China
| | - Xuehui Wang
- Department of Pediatrics, Shandong Provincial Hospital Affiliated to Shandong First Medical University, 324 Jingwuweiqi Road, Jinan, 250014, Shandong, PR China
| | - Jie Zhang
- Department of Pediatrics, Shandong Provincial Hospital Affiliated to Shandong First Medical University, 324 Jingwuweiqi Road, Jinan, 250014, Shandong, PR China
| | - Yanting Gao
- Department of Pediatrics, Shandong Provincial Hospital Affiliated to Shandong First Medical University, 324 Jingwuweiqi Road, Jinan, 250014, Shandong, PR China
| | - Youfei Fan
- Department of Pediatrics, Shandong Provincial Hospital Affiliated to Shandong First Medical University, 324 Jingwuweiqi Road, Jinan, 250014, Shandong, PR China.
| |
Collapse
|
2
|
Zhang Y, Shen Y, Kou D, Yu T. Identification and experimental verification of biomarkers related to butyrate metabolism in osteoarthritis. Sci Rep 2025; 15:11884. [PMID: 40195426 PMCID: PMC11977226 DOI: 10.1038/s41598-025-97346-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] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Accepted: 04/03/2025] [Indexed: 04/09/2025] Open
Abstract
Butyrate plays a crucial role in osteoarthritis (OA) development. However, the relationship between butyrate metabolism-related genes (BMRGs) and OA remains unclear. This study investigates the potential correlation between BMRGs and OA using OA-related datasets (GSE55235, GSE12021 and GSE143514). Differential expression analysis identified 38 differentially expressed butyrate metabolism-related genes (DE-BMRGs) from the overlap of 782 OA-related differentially expressed genes (DEGs) and 385 BMRGs in GSE55235. Enrichment analysis indicated that these DE-BMRGs were tightly associated with cell proliferation, differentiation, and apoptosis, which are key processes in OA pathogenesis. Six candidate biomarkers (IL1B, IGF1, CXCL8, PTGS2, SERPINE1, MMP9) were identified through two machine-learning algorithms. IL1B, CXCL8, and PTGS2 were upregulated in controls, exhibiting consistent patterns across validation datasets. Gene set enrichment analysis (GSEA) revealed that dysregulated expression of these biomarkers lead to abnormal cell proliferation and differentiation, contributing to OA progression. Furthermore, significant differences in immune cell infiltration-particularly activated and resting mast cells-along with correlations to immune regulatory factors (CD86, CXCL12, TNFSF9, IL6), highlighted potential therapeutic targets. Quantitative RT-PCR further confirmed elevated expression of IL1B, CXCL8 and PTGS2 in control group. This study identifies IL1B, CXCL8 and PTGS2 as OA-related biomarkers linked to butyrate metabolism, offering a theoretical foundation and potential therapeutic strategies.
Collapse
Affiliation(s)
- Yi Zhang
- Department of Orthopedics, Affiliated Hospital of Qingdao University, 16 Jiangsu Road, Shinan District, QingDao, 266003, China
- Traumatic Orthopedics Institute of Shandong, Affiliated Hospital of Qingdao University, Qingdao, China
| | - Youliang Shen
- Department of Joint Surgery, Affiliated Hospital of Qingdao University, QingDao, China
| | - Dewei Kou
- Department of Pain Management, Affiliated Hospital of Qingdao University, QingDao, China
| | - Tengbo Yu
- Department of Orthopedics, Affiliated Hospital of Qingdao University, 16 Jiangsu Road, Shinan District, QingDao, 266003, China.
- Department of Orthopedics, Qingdao Municipal Hospital, QingDao, China.
| |
Collapse
|
3
|
Lei P, Yu H, Jiang T, Ma J, Du J, Fang Y, Wang H, Chen R, Yang Q, Cheng Y, Wu W, Sun D. Development of a sodium hyaluronate-enriched therapeutic formulation with stevia glycoside and mogroside V for the comprehensive management of diabetes and its complications. Int J Biol Macromol 2025; 293:139487. [PMID: 39756763 DOI: 10.1016/j.ijbiomac.2025.139487] [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: 10/13/2024] [Revised: 12/27/2024] [Accepted: 01/02/2025] [Indexed: 01/07/2025]
Abstract
Diabetes prevalence continues to increase as a result of people's increasing sugar intake. Diabetes mellitus and its complications (dry skin, constipation, depression, and dental caries), as well as the prohibition of sweets ingestion, seriously affect patients' physical and mental health. Therefore, it is crucial to develop a long-term food for special medical purposes (FSMP) that aids in managing diabetes and its complications. To ensure effective biomedical function and taste, we developed a FSMP beverage formulation containing stevia glycoside, mogroside V, and sodium hyaluronate (SMH-B), each at a concentration of 0.1 mg/mL. Meanwhile, this study verified that SMH-B is an environmentally friendly and biocompatible formulation. Furthermore, both in vivo and in vitro studies have demonstrated that SMH-B significantly lowers blood glucose and lipid levels, enhances skin moisture and elasticity, prevents dental caries, alleviates constipation, reduces oxidative stress, and mitigates depressive symptoms. Notably, the SMH-B compound formula exhibits a more effective adjuvant therapeutic effect compared to single-ingredient formulation composed of stevia glycosides, mogroside V, and sodium hyaluronate. Moreover, SMH-B provides the sweetness desired by diabetic patients without affecting blood glucose levels, while also offering an auxiliary therapeutic role, making it a potential FSMP for diabetes management.
Collapse
Affiliation(s)
- Pengyu Lei
- Institute of Life Sciences & Biomedical Collaborative Innovation Center of Zhejiang Province, Wenzhou University, Wenzhou 325035, China
| | - Haiyang Yu
- Institute of Life Sciences & Biomedical Collaborative Innovation Center of Zhejiang Province, Wenzhou University, Wenzhou 325035, China
| | - Tao Jiang
- Institute of Life Sciences & Biomedical Collaborative Innovation Center of Zhejiang Province, Wenzhou University, Wenzhou 325035, China
| | - Jiahui Ma
- Institute of Life Sciences & Biomedical Collaborative Innovation Center of Zhejiang Province, Wenzhou University, Wenzhou 325035, China
| | - Jiao Du
- Institute of Life Sciences & Biomedical Collaborative Innovation Center of Zhejiang Province, Wenzhou University, Wenzhou 325035, China
| | - Yimeng Fang
- Institute of Life Sciences & Biomedical Collaborative Innovation Center of Zhejiang Province, Wenzhou University, Wenzhou 325035, China
| | - Hanbing Wang
- Department of Biotechnology, The University of Hong Kong, 999077, Hong Kong
| | - Rongbing Chen
- Department of Biomedical Engineering, City University of Hong Kong, 999077, Hong Kong
| | - Qinsi Yang
- Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou 325000, China
| | - Yongwei Cheng
- National Engineering Research Center of Cell Growth Factor Drugs and Protein Biologics, Wenzhou Medical University, Wenzhou 325000, China; MedTech (Wenzhou) Health Innovation Achievement Transformation Institute, Wenzhou Institute of Industry & Science, Wenzhou 325000, China.
| | - Wei Wu
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing 400044, China.
| | - Da Sun
- Institute of Life Sciences & Biomedical Collaborative Innovation Center of Zhejiang Province, Wenzhou University, Wenzhou 325035, China.
| |
Collapse
|
4
|
Pimenta I, Chaves Filho G, Silva EGB, Nogueira LFB, de Mendonça TS, Furtado TC, Ferreira GasparNeto PC, Dias LG, Fukada SY, Ciancaglini P, Ramos AP. Synthesis and Characterization of a Strontium-Quercetin Complex and Its In Vitro and In Vivo Potential for Application in Bone Regeneration. ACS OMEGA 2025; 10:4836-4846. [PMID: 39959088 PMCID: PMC11822716 DOI: 10.1021/acsomega.4c09949] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/05/2024] [Revised: 12/15/2024] [Accepted: 01/24/2025] [Indexed: 02/18/2025]
Abstract
Progress has been made toward developing therapies to treat bone-related diseases and defects caused by trauma. However, some of these therapies, such as administering strontium ranelate to treat osteoporosis, have significant side effects. In this context, designing new and safer strontium-based materials constitutes an important current challenge. Here, we have used quercetin as a platform to synthesize a new complex based on strontium and evaluate its activity in vitro and in vivo. First, we carried out strontium complexation with quercetin. Then, we employed Fourier transform infrared spectroscopy, nuclear magnetic resonance, and thermal gravimetric analysis to determine the chemical composition of the resulting complex as [(C15H7O7)Sr2]·6(H2O), which was also supported by theoretical calculations. This complex enhanced osteogenic differentiation of a preosteoblastic cell line in vitro, which increased alkaline phosphatase activity and extracellular matrix mineralization. By using a periapical lesion model in mice, we tested whether treatment with this complex could regenerate bone defects in vivo and found that the lesions decreased after 7 days. Together, our data showed that the strontium-quercetin complex synthesized herein is a potential candidate for developing new bone regeneration therapies.
Collapse
Affiliation(s)
- Israel
B. Pimenta
- Department
of Chemistry, Faculty of Philosophy, Science and Letters at Ribeirão
Preto, University of São Paulo, Ribeirão Preto 14040-901, São Paulo, Brazil
| | - Gildácio Chaves Filho
- Department
of Chemistry, Faculty of Philosophy, Science and Letters at Ribeirão
Preto, University of São Paulo, Ribeirão Preto 14040-901, São Paulo, Brazil
| | - Elias G. B. Silva
- Department
of Chemistry, Faculty of Philosophy, Science and Letters at Ribeirão
Preto, University of São Paulo, Ribeirão Preto 14040-901, São Paulo, Brazil
| | - Lucas F. B. Nogueira
- Department
of Chemistry, Faculty of Philosophy, Science and Letters at Ribeirão
Preto, University of São Paulo, Ribeirão Preto 14040-901, São Paulo, Brazil
| | - Tomaz Santana de Mendonça
- Department
of Biomolecular Sciences, School of Pharmaceutical Sciences of Ribeirao
Preto, University of Sao Paulo, Ribeirão Preto 14040-903, São Paulo, Brazil
| | - Taíssa C.
S. Furtado
- Department
of Biomolecular Sciences, School of Pharmaceutical Sciences of Ribeirao
Preto, University of Sao Paulo, Ribeirão Preto 14040-903, São Paulo, Brazil
| | - Paulo Cesar Ferreira GasparNeto
- Department
of Chemistry, Faculty of Philosophy, Science and Letters at Ribeirão
Preto, University of São Paulo, Ribeirão Preto 14040-901, São Paulo, Brazil
| | - Luis Gustavo Dias
- Department
of Chemistry, Faculty of Philosophy, Science and Letters at Ribeirão
Preto, University of São Paulo, Ribeirão Preto 14040-901, São Paulo, Brazil
| | - Sandra Yasuyo Fukada
- Department
of Biomolecular Sciences, School of Pharmaceutical Sciences of Ribeirao
Preto, University of Sao Paulo, Ribeirão Preto 14040-903, São Paulo, Brazil
| | - Pietro Ciancaglini
- Department
of Chemistry, Faculty of Philosophy, Science and Letters at Ribeirão
Preto, University of São Paulo, Ribeirão Preto 14040-901, São Paulo, Brazil
| | - Ana Paula Ramos
- Department
of Chemistry, Faculty of Philosophy, Science and Letters at Ribeirão
Preto, University of São Paulo, Ribeirão Preto 14040-901, São Paulo, Brazil
| |
Collapse
|
5
|
Carrillo-Norte JA, Gervasini-Rodríguez G, Santiago-Triviño MÁ, García-López V, Guerrero-Bonmatty R. Oral administration of hydrolyzed collagen alleviates pain and enhances functionality in knee osteoarthritis: Results from a randomized, double-blind, placebo-controlled study. Contemp Clin Trials Commun 2025; 43:101424. [PMID: 39839727 PMCID: PMC11745964 DOI: 10.1016/j.conctc.2024.101424] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Revised: 12/12/2024] [Accepted: 12/29/2024] [Indexed: 01/23/2025] Open
Abstract
Osteoarthritis (OA) is a major source of chronic pain and disability, representing a significant global health concern that affects 10-15 % of individuals aged over 60, with a higher prevalence among females than males. This investigation aimed to evaluate the impact of a dietary supplement containing collagen peptides (MW 1-3 kDa) on knee OA symptoms and inflammatory biomarkers such as C-reactive protein (CRP) and erythrocyte sedimentation rate (ESR). Adults aged 30-81 years (50 % female) with grade II or III OA and a minimum pain score of 40 on the 0 to 100 visual analogue scale (VAS) were enrolled. Participants were randomly assigned to receive either 10 g of the test product (verum group) or placebo and were assessed at baseline (T0, pre-treatment) and after a six-month follow-up period (T6). Baseline characteristics were comparable between groups. At T6, the verum group exhibited significant reductions in VAS pain scores, Lequesne algofunctional index (LAI) scores, CRP levels (mg/L), and ESR (mm/h) compared to placebo (p < 0.001). No adverse effects were reported during the study, and the supplement demonstrated good tolerability and yielded satisfactory safety and acceptability. These findings suggest that the dietary supplement may serve as a complement to drug therapy for knee OA by alleviating osteoarticular pain, improving locomotor function and potentially reducing reliance on analgesic and anti-inflammatory medications. This study provides valuable insights into the efficacy and safety of collagen peptides in managing knee OA symptoms.
Collapse
Affiliation(s)
- Juan Antonio Carrillo-Norte
- Department of Medical and Surgical Therapeutics, Division of Clinical Pharmacology, School of Medicine, University of Extremadura, Badajoz, Spain
| | - Guillermo Gervasini-Rodríguez
- Department of Medical and Surgical Therapeutics, Division of Clinical Pharmacology, School of Medicine, University of Extremadura, Badajoz, Spain
| | | | - Virginio García-López
- Department of Medical and Surgical Therapeutics, Division of Clinical Pharmacology, School of Medicine, University of Extremadura, Badajoz, Spain
| | | |
Collapse
|
6
|
Guo W, Gao J, Dawazhuoma, Mi X, Ciwang, Bianba. A meta-analysis of randomized controlled trials: evaluating the efficacy of isokinetic muscle strengthening training in improving knee osteoarthritis outcomes. J Orthop Surg Res 2025; 20:95. [PMID: 39856723 PMCID: PMC11762536 DOI: 10.1186/s13018-025-05495-8] [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: 11/05/2024] [Accepted: 01/13/2025] [Indexed: 01/27/2025] Open
Abstract
BACKGROUND Knee osteoarthritis (KOA) is a prevalent degenerative joint disease. The primary pathological manifestations of KOA include articular cartilage degeneration, joint space narrowing, and osteophyte formation, leading to a spectrum of symptoms, including joint pain, stiffness, reduced mobility, diminished muscle strength, and severe disability. We aimed to utilize a meta-analysis to evaluate the efficacy of isokinetic muscle strengthening training (IMST) as a rehabilitation treatment for KOA in lowland areas. METHODS The study conducted a comprehensive search of the CNKI, WanFang Data, VIP Database, PubMed, Ovid MEDLINE (1946-), Cochrane Library, Embase, and CBM databases. The databases were conducted from establishing each database to September 31, 2024. The studies included were randomized controlled trials (RCTs) with participants from the plains who met the diagnostic criteria for KOA as outlined in the 2019 edition, with no restrictions on gender, age, or disease course, and no patients with advanced disease; studies where in the control group was either a non-intervention group or a group receiving treatment, other than IMST, and the experimental group received IMST alone or in addition to the treatment administered to the control group; and studies with at least two of the following outcome indicators: (i) knee flexors (Flex)/extensors (Ext) peak torque (PT), (ii) knee Flex/Ext total work (TW), (iii) knee Flex/Ext max rep total work (MRTW), (iv) knee Flex/Ext average power (AP), (v) visual analogue scale (VAS) for pain, (vi) Lequesne index (LI), (vii) Western Ontario and McMaster University Osteoarthritis Index (WOMAC), (viii) Lysholm Knee Scoring Scale (LKSS), (ix) range of motion (ROM) of the knee joint, and (x) 6-min walk test. We systematically reviewed the RCTs in both Chinese and English and evaluated the quality of the included literature. Data were processed and analyzed using ROB 2, RevMan 5.4, Stata17, and GRADEpro. The study protocol was registered on PROSPERO (CRD42024607528). RESULTS Thirty-three (46 studies, 2,860 patients) had low-to-some concerns risk. IMST significantly improved physical therapy outcomes, including knee Flex PT and knee Ext PT at an angular velocity of 60°/second (standardized mean difference 13.19 [95% confidence interval 6.44, 19.94], P = 0.0001 and 16.34 [11.47, 21.22], P < 0.00001, respectively), and 180°/second (11.17 [2.86, 19.48], P = 0.008 and 12.62 [3.49, 21.75], P = 0.0077, respectively); knee Flex TW (79.77 [49.43, 110.10], P < 0.0001), Ext TW (86.27 [58.40, 114.15], P < 0.00001), knee Flex MRTW (9.38 [3.20, 15.56], P = 0.003), knee Ext MRTW (15.52 [8.96, 22.08], P < 0.0001), knee Flex AP (8.66 [0.70, 16.61], P = 0.03), knee Ext AP (7.27 [3.30, 11.23], P = 0.0003), knee Flex ROM (10.62 [7.94, 13.30], P < 0.00001), and LKSS scores (7.90 [5.91, 9.89], P < 0.00001). Additionally, it reduced VAS scores (- 0.70 [- 0.92, - 0.49], P < 0.00001), LI scores (- 1.24 [- 1.65, - 0.83], P < 0.00001), and WOMAC scores (- 6.05 [- 10.37, - 1.73], P = 0.006). Compared to the control group, superior clinical efficacy was noted in the experimental group. The quality of evidence the studies reported was poor, mainly due to original trials with high inter-study heterogeneity and imprecise results. The therapeutic effect of IMST on KOA remained significant after rigorous testing of subgroup and sensitivity analyses. CONCLUSIONS In patients with KOA, IMST improves muscle strength and relieves joint pain and stiffness. However, large-scale, high-quality, randomized controlled trials with extended observation periods are urgently needed to popularize the use of IMST in KOA patients.
Collapse
Affiliation(s)
- Wanqin Guo
- Medical College, Tibet University, Lhasa, Tibet, 850000, China
- High Altitude Medical Research Center, Medical College, Tibet University, Lhasa, Tibet, 850000, China
| | - Jingyang Gao
- Medical College, Tibet University, Lhasa, Tibet, 850000, China
- High Altitude Medical Research Center, Medical College, Tibet University, Lhasa, Tibet, 850000, China
| | - Dawazhuoma
- Medical College, Tibet University, Lhasa, Tibet, 850000, China
- High Altitude Medical Research Center, Medical College, Tibet University, Lhasa, Tibet, 850000, China
| | - Xiuling Mi
- Medical College, Tibet University, Lhasa, Tibet, 850000, China
- High Altitude Medical Research Center, Medical College, Tibet University, Lhasa, Tibet, 850000, China
| | - Ciwang
- The Second People's Hospital of Tibet Autonomous Region, Lhasa, Tibet, 850000, China
| | - Bianba
- Medical College, Tibet University, Lhasa, Tibet, 850000, China.
- High Altitude Medical Research Center, Medical College, Tibet University, Lhasa, Tibet, 850000, China.
- Key Laboratory of Plateau Sports and Health of Tibet Autonomous Region, Lhasa, Tibet, China.
| |
Collapse
|
7
|
Sajjad Khawaja A, Zafar M, Muhammad Zeeshan R, Ilyas MS, Aziz A, Zehra U. Non-steroidal anti-inflammatory drugs influence cartilage healing. Knee 2025; 52:121-130. [PMID: 39577110 DOI: 10.1016/j.knee.2024.10.021] [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: 06/05/2024] [Revised: 08/20/2024] [Accepted: 10/29/2024] [Indexed: 11/24/2024]
Abstract
BACKGROUND Retrograde intramedullary nailing is commonly performed to stabilize distal femoral shaft fractures which may lead to iatrogenic injuries of the knee articular cartilage. The limited regenerative capability of cartilage may further be hindered by intake of non-steroidal anti-inflammatory drugs (NSAIDs) which are usually advised for injuries of the musculoskeletal system. The current study was designed to evaluate the histological changes in the femoral articular cartilage of knee joint after retrograde femoral nailing of rats. METHODS Retrograde intramedullary nailing was performed in 36 adult male Wistar rats, divided into three groups of 12 each. Groups 1 and 2 were given nonselective and selective COX 2 inhibitors, respectively, while the third group was taken as control. Half of the animals from each group were sacrificed at the second week, and remainder on the seventh week, and samples of the femoral articular cartilage were assessed for cartilage regeneration according to the modified Mankin scoring on histology while BMP-2 expression was evaluated on immunohistochemistry. RESULTS Mean modified Mankin scores for cartilage degradation were increased in animals taking NSAIDs at the second and seventh weeks of healing (P = 0.02, P < 0.001 respectively). There was a significant decrease in chondrocytes at the second week (P = 0.001), along with the loss of proteoglycan content in these animals at both time points (P = 0.001). The BMP-2 expression was significantly enhanced in the control group at the second (P = 0.001) and seventh weeks (P = 0.001). CONCLUSION The results reveal that intake of NSAIDs hinders the process of cartilage healing by reducing the number of chondrocytes and loss of proteoglycan content and decreased expression of BMP-2.
Collapse
Affiliation(s)
| | - Maria Zafar
- Department of Anatomy, University of Health Sciences, Lahore, Pakistan
| | | | - Muhammad Saad Ilyas
- Department of Orthopaedic & Spine Surgery, Ghurki Trust Teaching Hospital, Lahore Pakistan
| | - Amer Aziz
- Department of Orthopaedic & Spine Surgery, Ghurki Trust Teaching Hospital, Lahore Pakistan
| | - Uruj Zehra
- Department of Anatomy, University of Health Sciences, Lahore, Pakistan.
| |
Collapse
|
8
|
Li R, Sun K. Regulation of chondrocyte apoptosis in osteoarthritis by endoplasmic reticulum stress. Cell Stress Chaperones 2024; 29:750-763. [PMID: 39515603 PMCID: PMC11626768 DOI: 10.1016/j.cstres.2024.11.001] [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: 08/05/2024] [Revised: 11/01/2024] [Accepted: 11/04/2024] [Indexed: 11/16/2024] Open
Abstract
Osteoarthritis (OA), a common degenerative joint disease, is characterized by the apoptosis of chondrocytes as a primary pathophysiological change, with endoplasmic reticulum stress (ERS) playing a crucial role. It has been demonstrated that an imbalance in endoplasmic reticulum (ER) homeostasis can lead to ERS, activating three cellular adaptive response pathways through the unfolded protein response to restore ER homeostasis. Mild ERS exerts a protective effect on cells, while prolonged ERS that disrupts the self-regulatory balance of the ER activates apoptotic signaling pathways, leading to chondrocyte apoptosis and hastening OA progression. Hence, controlling the ERS signaling pathway and its apoptotic factors has become a critical focus for preventing and treating OA. This review aims to elucidate the key mechanisms of ERS pathway-induced apoptosis, associated targets, and regulatory pathways, offering valuable insights to enhance the mechanistic understanding of OA. It also reviews the mechanisms studied for ERS-related drugs or compounds for the treatment of OA.
Collapse
Affiliation(s)
- Renzhong Li
- Taizhou Hospital of Traditional Chinese Medicine, Taizhou, Jiangsu Province, China; The Second Affiliated Hospital of Anhui University of Traditional Chinese Medicine, Hefei, Anhui Province, China
| | - Kui Sun
- The Second Affiliated Hospital of Anhui University of Traditional Chinese Medicine, Hefei, Anhui Province, China; Anhui Acupuncture Hospital, Hefei, Anhui Province, China.
| |
Collapse
|
9
|
Cheng G, Wang X, Zhang F, Wang K, Li Y, Guo T, Xu N, Wei W, Yan S. Reparative homing of bone mesenchymal stem cells induced by iMSCs via the SDF-1/CXCR4 axis for articular cartilage defect restoration. Biomed Pharmacother 2024; 181:117649. [PMID: 39536539 DOI: 10.1016/j.biopha.2024.117649] [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: 08/14/2024] [Revised: 10/17/2024] [Accepted: 10/28/2024] [Indexed: 11/16/2024] Open
Abstract
BACKGROUND The intrinsic healing ability of articular cartilage is poor after injury or illness, and untreated injury could lead to cartilage degeneration and ultimately osteoarthritis. iMSCs are derived from embryonic induced pluripotent stem cells and have strong therapeutic capabilities in the repair of cartilage defects, while the mechanism of action is unclear. The aim of this study is to clarify the repair mode of iMSCs on cartilage defects in rat knee joints, elucidate the chemotactic effect of iMSCs on autologous BMSCs in rats, and provide a basis for the treatment of cartilage defects and endogenous regeneration with iMSCs. METHODS Based on the establishment of the rat cartilage defect model, the reparative effect of iMSCs on the rat cartilage defect was evaluated. The cartilage repair was evaluated by quantitative score, H&E staining, Masson staining and Safranin-O staining, and the metabolic changes of iMSCs in the joint cavity were detected in vivo. The expression of SOX9, CD29, CD90, ColⅠ, ColⅡ, PCNA, SDF-1, and CXCR4 was detected by immunohistochemistry (IHC), IF, flow cytometry, respectively. After co-culturing iMSCs with BMSCs in vitro, the expression of CXCR4/SDF-1 on the cell membrane surface of BMSCs was detected by western blotting.; The level of p-Akt and p-Erk1/2 in total protein of BMSCs were detected by western blotting. SIGNIFICANCE Our research results provide experimental evidence for the treatment of cartilage defects and endogenous regeneration with iMSCs; This also provides new ideas for the clinical treatment of cartilage defects using iMSCs.
Collapse
Affiliation(s)
- Gang Cheng
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Hefei 230032, China; Collaborative Innovation Center of Anti-inflammatory and Immune Medicine, Hefei 230032, China
| | - Xulei Wang
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Hefei 230032, China; Collaborative Innovation Center of Anti-inflammatory and Immune Medicine, Hefei 230032, China; Laboratory Animal Center, Anhui Medical University, Hefei 230032, China
| | - Feng Zhang
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Hefei 230032, China; Collaborative Innovation Center of Anti-inflammatory and Immune Medicine, Hefei 230032, China
| | - Kang Wang
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Hefei 230032, China; Collaborative Innovation Center of Anti-inflammatory and Immune Medicine, Hefei 230032, China
| | - Ying Li
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Hefei 230032, China; Collaborative Innovation Center of Anti-inflammatory and Immune Medicine, Hefei 230032, China
| | - Tingting Guo
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Hefei 230032, China; Collaborative Innovation Center of Anti-inflammatory and Immune Medicine, Hefei 230032, China
| | - Nuo Xu
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Hefei 230032, China; Collaborative Innovation Center of Anti-inflammatory and Immune Medicine, Hefei 230032, China
| | - Wei Wei
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Hefei 230032, China; Collaborative Innovation Center of Anti-inflammatory and Immune Medicine, Hefei 230032, China.
| | - Shangxue Yan
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Hefei 230032, China; Collaborative Innovation Center of Anti-inflammatory and Immune Medicine, Hefei 230032, China; Laboratory Animal Center, Anhui Medical University, Hefei 230032, China.
| |
Collapse
|
10
|
Guan J, Li Z, Niu G, Li S, Li W, Song C, Leng H. Protective Effects of Vitamin D on Proteoglycans of Human Articular Chondrocytes through TGF-β1 Signaling. Nutrients 2024; 16:2991. [PMID: 39275306 PMCID: PMC11396982 DOI: 10.3390/nu16172991] [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: 08/04/2024] [Revised: 08/23/2024] [Accepted: 09/03/2024] [Indexed: 09/16/2024] Open
Abstract
The extracellular matrix of cartilage primarily constitutes of collagen and aggrecan. Cartilage degradation starts with aggrecan loss in osteoarthritis (OA). Vitamin D (VD) plays an essential role in several inflammation-related diseases and can protect the collagen in cartilage during OA. The present study focused on the role of VD in aggrecan turnover of human articular chondrocytes treated with tumor necrosis factor α (TNF-α) and the possible mechanism. Treatment with different doses of VD and different periods of intervention with TNF-α and TGF-β1 receptor (TGFβR1) inhibitor SB525334 were investigated. The viability of human chondrocytes and extracellular secretion of TGF-β1 were measured. The expression of intracellular TGFβR1 and VD receptor was examined. Transcriptional and translational levels of aggrecan and the related metabolic factors were analyzed. The results showed that TNF-α markedly reduced the viability, TGFβR1 expressions and aggrecan levels of human chondrocytes, and increased disintegrin and metalloproteinase with thrombospondin motifs. The alterations were partially inhibited by VD treatment. Furthermore, the effects of VD were blocked by the TGFβR1 inhibitor SB525334 in TNF-α-treated cells. VD may prevent proteoglycan loss due to TNF-α via TGF-β1 signaling in human chondrocytes.
Collapse
Affiliation(s)
- Jian Guan
- Department of Orthopedics, Peking University Third Hospital, Beijing 100191, China; (J.G.); (Z.L.); (G.N.); (S.L.)
- Department of Orthopedic Surgery, Beijing Luhe Hospital, Capital Medical University, Beijing 101100, China
| | - Zhuoxin Li
- Department of Orthopedics, Peking University Third Hospital, Beijing 100191, China; (J.G.); (Z.L.); (G.N.); (S.L.)
| | - Guodong Niu
- Department of Orthopedics, Peking University Third Hospital, Beijing 100191, China; (J.G.); (Z.L.); (G.N.); (S.L.)
| | - Siwei Li
- Department of Orthopedics, Peking University Third Hospital, Beijing 100191, China; (J.G.); (Z.L.); (G.N.); (S.L.)
| | - Weishi Li
- Engineering Research Center of Bone and Joint Precision Medicine, Ministry of Education, Beijing 100191, China;
| | - Chunli Song
- Beijing Key Lab of Spine Diseases, Beijing 100191, China;
| | - Huijie Leng
- Department of Orthopedics, Peking University Third Hospital, Beijing 100191, China; (J.G.); (Z.L.); (G.N.); (S.L.)
| |
Collapse
|
11
|
Yu S, Shu X, Wang X, Sheng Y, Li S, Wang Y, Zhang Y, Tao J, Jiang X, Wu C. The novel HSP90 monoclonal antibody 9B8 ameliorates articular cartilage degeneration by inhibiting glycolysis via the HIF-1 signaling pathway. Heliyon 2024; 10:e35603. [PMID: 39229534 PMCID: PMC11369415 DOI: 10.1016/j.heliyon.2024.e35603] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Revised: 07/04/2024] [Accepted: 07/31/2024] [Indexed: 09/05/2024] Open
Abstract
Osteoarthritis (OA) is a prevalent chronic degenerative disease that affects the bones and joints, particularly in middle-aged and elderly individuals. It is characterized by progressive joint pain, swelling, stiffness, and deformity. Notably, treatment with a heat shock protein 90 (HSP90) inhibitor has significantly curtailed cartilage destruction in a rat model of OA. Although the monoclonal antibody 9B8 against HSP90 is recognized for its anti-tumor properties, its potential therapeutic impact on OA remains uncertain. This study investigated the effects of 9B8 on OA and its associated signaling pathways in interleukin-1β (IL-1β)-stimulated human chondrocytes and a rat anterior cruciate ligament transection (ACLT) model. A specific concentration of 9B8 preserved cell viability against IL-1β-induced reduction. In vitro, 9B8 significantly reduced the expression of extracellular matrix-degrading enzyme such as disintegrin and metallopeptidase-4 (ADAMTS4) of thrombospondin motifs, matrix metalloproteinase-13 (MMP-13), as well as cellular inflammatory factors such as tumor necrosis factor-α (TNF-α) and interleukin-6 (IL-6), which were upregulated by IL-1β. In vivo, 9B8 effectively protected the articular cartilage and subchondral bone of the rat tibial plateau from ACLT-induced damage. Additionally, gene microarray analysis revealed that IL-1β substantially increased the expression of SLC2A1, PFKP, and ENO2 within the HIF-1 signaling pathway, whereas 9B8 suppressed the expression of these genes. Thus, 9B8 effectively mitigates ACLT-induced osteoarthritis in rats by modulating the HIF-1 signaling pathway, thereby inhibiting overexpression involved in glycolysis. These results collectively indicate that 9B8 is a promising novel drug for the prevention and treatment of OA.
Collapse
Affiliation(s)
- Shunan Yu
- Department of Molecular Orthopedics, Beijing Research Institute of Traumatology and Orthopedics, National Center for Orthopaedics, Beijing Jishuitan Hospital, Beijing, 100035, PR China
| | - Xiong Shu
- Department of Molecular Orthopedics, Beijing Research Institute of Traumatology and Orthopedics, National Center for Orthopaedics, Beijing Jishuitan Hospital, Beijing, 100035, PR China
| | - Xinyu Wang
- Department of Molecular Orthopedics, Beijing Research Institute of Traumatology and Orthopedics, National Center for Orthopaedics, Beijing Jishuitan Hospital, Beijing, 100035, PR China
| | - Yueyang Sheng
- Department of Molecular Orthopedics, Beijing Research Institute of Traumatology and Orthopedics, National Center for Orthopaedics, Beijing Jishuitan Hospital, Beijing, 100035, PR China
| | - Shan Li
- Department of Molecular Orthopedics, Beijing Research Institute of Traumatology and Orthopedics, National Center for Orthopaedics, Beijing Jishuitan Hospital, Beijing, 100035, PR China
| | - Ying Wang
- Department of Molecular Orthopedics, Beijing Research Institute of Traumatology and Orthopedics, National Center for Orthopaedics, Beijing Jishuitan Hospital, Beijing, 100035, PR China
| | - Yanzhuo Zhang
- Department of Molecular Orthopedics, Beijing Research Institute of Traumatology and Orthopedics, National Center for Orthopaedics, Beijing Jishuitan Hospital, Beijing, 100035, PR China
| | - Jiangfeng Tao
- Department of Molecular Orthopedics, Beijing Research Institute of Traumatology and Orthopedics, National Center for Orthopaedics, Beijing Jishuitan Hospital, Beijing, 100035, PR China
| | - Xu Jiang
- Department of Orthopaedics, Beijing Jishuitan Hospital, Capital Medical University, Beijing Research Institute of Traumatology and Orthopaedics, Beijing, 100035, PR China
| | - Chengai Wu
- Department of Molecular Orthopedics, Beijing Research Institute of Traumatology and Orthopedics, National Center for Orthopaedics, Beijing Jishuitan Hospital, Beijing, 100035, PR China
| |
Collapse
|
12
|
Mosaddad SA, Talebi S, Keyhan SO, Fallahi HR, Darvishi M, Aghili SS, Tavahodi N, Namanloo RA, Heboyan A, Fathi A. Dental implant considerations in patients with systemic diseases: An updated comprehensive review. J Oral Rehabil 2024; 51:1250-1302. [PMID: 38570927 DOI: 10.1111/joor.13683] [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: 03/30/2023] [Revised: 10/27/2023] [Accepted: 03/02/2024] [Indexed: 04/05/2024]
Abstract
BACKGROUND Various medical conditions and the drugs used to treat them have been shown to impede or complicate dental implant surgery. It is crucial to carefully monitor the medical status and potential post-operative complications of patients with systemic diseases, particularly elderly patients, to minimize the risk of health complications that may arise. AIM The purpose of this study was to review the existing evidence on the viability of dental implants in patients with systemic diseases and to provide practical recommendations to achieve the best possible results in the corresponding patient population. METHODS The information for our study was compiled using data from PubMed, Scopus, Web of Science and Google Scholar databases and searched separately for each systemic disease included in our work until October 2023. An additional manual search was also performed to increase the search sensitivity. Only English-language publications were included and assessed according to titles, abstracts and full texts. RESULTS In total, 6784 studies were found. After checking for duplicates and full-text availability, screening for the inclusion criteria and manually searching reference lists, 570 articles remained to be considered in this study. CONCLUSION In treating patients with systemic conditions, the cost-benefit analysis should consider the patient's quality of life and expected lifespan. The success of dental implants depends heavily on ensuring appropriate maintenance therapy, ideal oral hygiene standards, no smoking and avoiding other risk factors. Indications and contraindications for dental implants in cases of systemic diseases are yet to be more understood; broader and hardcore research needs to be done for a guideline foundation.
Collapse
Affiliation(s)
- Seyed Ali Mosaddad
- Department of Research Analytics, Saveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, Tamil Nadu, India
- Department of Conservative Dentistry and Bucofacial Prosthesis, Faculty of Odontology, Complutense University of Madrid, Madrid, Spain
- Student Research Committee, School of Dentistry, Shiraz University of Medical Sciences, Shiraz, Iran
- Maxillofacial Surgery & Implantology & Biomaterial Research Foundation, Tehran, Iran
| | - Sahar Talebi
- Research Committee, School of Dentistry, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Seied Omid Keyhan
- Maxillofacial Surgery & Implantology & Biomaterial Research Foundation, Tehran, Iran
- Department of Oral & Maxillofacial Surgery, Gangneung-Wonju National University, Gangneung, South Korea
- Department of Oral & Maxillofacial Surgery, College of Medicine, University of Florida, Jacksonville, FL, USA
- Iface Academy, Istanbul, Turkey
| | - Hamid Reza Fallahi
- Maxillofacial Surgery & Implantology & Biomaterial Research Foundation, Tehran, Iran
- Department of Oral & Maxillofacial Surgery, Gangneung-Wonju National University, Gangneung, South Korea
- Department of Oral & Maxillofacial Surgery, College of Medicine, University of Florida, Jacksonville, FL, USA
- Iface Academy, Istanbul, Turkey
| | - Mohammad Darvishi
- Faculty of Dentistry, Isfahan (Khorasgan) Branch, Islamic Azad University, Isfahan, Iran
| | - Seyedeh Sara Aghili
- Student Research Committee, School of Dentistry, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Narges Tavahodi
- Student Research Committee, Faculty of Dentistry, Mazandaran University of Medical Sciences, Sari, Iran
| | | | - Artak Heboyan
- Department of Research Analytics, Saveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, Tamil Nadu, India
- Department of Prosthodontics, Faculty of Stomatology, Yerevan State Medical University after Mkhitar Heratsi, Yerevan, Armenia
- Department of Prosthodontics, School of Dentistry, Tehran University of Medical Sciences, Tehran, Iran
| | - Amirhossein Fathi
- Department of Prosthodontics, Dental Materials Research Center, Dental Research Institute, School of Dentistry, Isfahan University of Medical Sciences, Isfahan, Iran
| |
Collapse
|
13
|
Mariano A, Ammendola S, Migliorini A, Leopizzi M, Raimondo D, Scotto d'Abusco A. Intron retention in PI-PLC γ1 mRNA as a key mechanism affecting MMP expression in human primary fibroblast-like synovial cells. Cell Biochem Funct 2024; 42:e4091. [PMID: 38973151 DOI: 10.1002/cbf.4091] [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: 03/27/2024] [Revised: 06/26/2024] [Accepted: 06/30/2024] [Indexed: 07/09/2024]
Abstract
The intron retention (IR) is a phenomenon utilized by cells to allow diverse fates at the same mRNA, leading to a different pattern of synthesis of the same protein. In this study, we analyzed the modulation of phosphoinositide-specific phospholipase C (PI-PLC) enzymes by Harpagophytum procumbens extract (HPE) in synoviocytes from joins of osteoarthritis (OA) patients. In some samples, the PI-PLC γ1 isoform mature mRNA showed the IR and, in these synoviocytes, the HPE treatment increased the phenomenon. Moreover, we highlighted that as a consequence of IR, a lower amount of PI-PLC γ1 was produced. The decrease of PI-PLC γ1 was associated with the decrease of metalloprotease-3 (MMP-3), and MMP-13, and ADAMTS-5 after HPE treatment. The altered expression of MMPs is a hallmark of the onset and progression of OA, thus substances able to decrease their expression are very desirable. The interesting outcomes of this study are that 35% of analyzed synovial tissues showed the IR phenomenon in the PI-PLC γ1 mRNA and that the HPE treatment increased this phenomenon. For the first time, we found that the decrease of PI-PLC γ1 protein in synoviocytes interferes with MMP production, thus affecting the pathways involved in the MMP expression. This finding was validated by the silencing of PI-PLC γ1 in synoviocytes where the IR phenomenon was not present. Our results shed new light on the biochemical mechanisms involved in the degrading enzyme production in the joint of OA patients, suggesting a new therapeutic target and highlighting the importance of personalized medicine.
Collapse
Affiliation(s)
- Alessia Mariano
- Department of Biochemical Sciences, Sapienza University of Rome, Roma, Italy
| | | | - Arianna Migliorini
- Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy
| | - Martina Leopizzi
- Department of Medico-Surgical Sciences and Biotechnologies, Polo Pontino-Sapienza University, Latina, Italy
| | - Domenico Raimondo
- Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy
| | | |
Collapse
|
14
|
Akpınar K, Şimşek EK, Özen ÖI, Haberal B. The effect of MSM in the treatment of ankle arthrosis: Is MSM as effective as methylprednisolone or hyaluronic acid? J Orthop Res 2024; 42:1527-1535. [PMID: 38316633 DOI: 10.1002/jor.25795] [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: 11/04/2023] [Revised: 01/09/2024] [Accepted: 01/19/2024] [Indexed: 02/07/2024]
Abstract
Posttraumatic ankle osteoarthritis (PTAO) causes severe ankle and adjacent joint morbidity. We aimed to compare the treatment efficacy of previously tried and still applied intra-articular injections and oral methylsulfonylmethane (MSM) at functional and histopathological level in PTAO animal model. Thirty-two adult female Sprague-Dawley rats were divided into four groups (Group 1: Control, Group 2: 0.06 g/kg/day MSM, Group 3: 0.04 mg/µL methylprednisolone [MP], Group 4: 0.04 mg/µL hyaluronic acid [HA]). MSM was started orally between Day 0 to the end of 8 weeks. Intra-articular injections were applied to the right ankles of the subjects after surgery. All subjects were killed after radiological evaluation at the 8th week. Subsequently, functional (range of motion) and histopathological evaluation was performed. Radiological evaluation showed better results of the MP (p < 0.001) and MSM (p < 0.001) groups than the control group. Severity of osteoarthritis (OA) in the MP group was significantly less than in the HA group (p = 0.032). When the total Osteoarthritis Research Society International score was compared, the severity of OA was higher in the KS and HA groups than in the control group (p < 0.001). No significant statistical difference was found in the histopathological comparison of MSM and control group (p = 0.466). There was no difference between the groups in range of motion measurement according to the contralateral ankle joint. The radiological progression of OA was slowed in the MSM and MP groups, but significant histopathological worsening was found in the MP and HA applied groups. We suggest that the treatment methods used in daily practice need to be reviewed.
Collapse
Affiliation(s)
- Koray Akpınar
- Department of Orthopaedic and Traumatology, Baskent University, Ankara, Turkey
| | - Ekin K Şimşek
- Department of Orthopaedics and Traumatology, Baskent University Hospital, Ankara, Turkey
| | | | - Bahtiyar Haberal
- Department of Orthopaedics and Traumatology, Baskent University Hospital, Ankara, Turkey
| |
Collapse
|
15
|
Bică G, Rogoveanu OC, Gherghina FL, Pisoschi CG, Buteică SA, Biță CE, Paliu IA, Mîndrilă I. The Histological and Biochemical Assessment of Monoiodoacetate-Induced Knee Osteoarthritis in a Rat Model Treated with Salicylic Acid-Iron Oxide Nanoparticles. BIOLOGY 2024; 13:331. [PMID: 38785813 PMCID: PMC11117951 DOI: 10.3390/biology13050331] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Revised: 05/04/2024] [Accepted: 05/07/2024] [Indexed: 05/25/2024]
Abstract
Iron oxide nanoparticles (IONPs) represent an important advance in the field of medicine with application in both diagnostic and drug delivery domains, offering a therapeutic approach that effectively overcomes physical and biological barriers. The current study aimed to assess whether oral administration of salicylic acid-functionalized iron oxide nanoparticles (SaIONPs) may exhibit beneficial effects in alleviating histological lesions in a murine monoiodoacetate (MIA) induced knee osteoarthritis model. In order to conduct our study, 15 Wistar male rats were randomly distributed into 3 work groups: Sham (S), MIA, and NP. At the end of the experiments, all animals were sacrificed for blood, knee, and liver sampling. Our results have shown that SaIONPs reached the targeted sites and also had a chondroprotective effect represented by less severe histological lesions regarding cellularity, altered structure morphology, and proteoglycan depletion across different layers of the knee joint cartilage tissue. Moreover, SaIONPs induced a decrease in malondialdehyde (MDA) and circulating Tumor Necrosis Factor-α (TNF-α) levels. The findings of this study suggest the therapeutic potential of SaIONPs knee osteoarthritis treatment; further studies are needed to establish a correlation between the administrated dose of SaIONPs and the improvement of the morphological and biochemical parameters.
Collapse
Affiliation(s)
- George Bică
- Department of Physical Medicine and Rehabilitation, University of Medicine and Pharmacy of Craiova, 2 Petru Rares Street, 200349 Craiova, Romania; (G.B.); (O.-C.R.); (F.-L.G.)
| | - Otilia-Constantina Rogoveanu
- Department of Physical Medicine and Rehabilitation, University of Medicine and Pharmacy of Craiova, 2 Petru Rares Street, 200349 Craiova, Romania; (G.B.); (O.-C.R.); (F.-L.G.)
| | - Florin-Liviu Gherghina
- Department of Physical Medicine and Rehabilitation, University of Medicine and Pharmacy of Craiova, 2 Petru Rares Street, 200349 Craiova, Romania; (G.B.); (O.-C.R.); (F.-L.G.)
| | - Cătălina-Gabriela Pisoschi
- Department of Biochemistry, University of Medicine and Pharmacy of Craiova, 2 Petru Rares Street, 200349 Craiova, Romania;
| | - Sandra-Alice Buteică
- Faculty of Pharmacy, University of Medicine and Pharmacy of Craiova, 2 Petru Rares Street, 200349 Craiova, Romania
| | - Cristina-Elena Biță
- Department of Rheumatology, University of Medicine and Pharmacy of Craiova, 2 Petru Rares Street, 200349 Craiova, Romania;
| | - Iulia-Alexandra Paliu
- Department of Pharmacology, University of Medicine and Pharmacy of Craiova, 2 Petru Rares Street, 200349 Craiova, Romania;
| | - Ion Mîndrilă
- Department of Anatomy, University of Medicine and Pharmacy of Craiova, 2 Petru Rares Street, 200349 Craiova, Romania;
| |
Collapse
|
16
|
Saha SK, Zhu Y, Murray P, Madden L. Future proofing of chondroitin sulphate production: Importance of sustainability and quality for the end-applications. Int J Biol Macromol 2024; 267:131577. [PMID: 38615853 DOI: 10.1016/j.ijbiomac.2024.131577] [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: 02/02/2024] [Revised: 04/10/2024] [Accepted: 04/11/2024] [Indexed: 04/16/2024]
Abstract
Chondroitin sulphates (CSs) are the most well-known glycosaminoglycans (GAGs) found in any living organism, from microorganisms to invertebrates and vertebrates (including humans), and provide several health benefits. The applications of CSs are numerous including tissue engineering, osteoarthritis treatment, antiviral, cosmetics, and skincare applications. The current commercial production of CSs mostly uses animal, bovine, porcine, and avian tissues as well as marine organisms, marine mammals, sharks, and other fish. The production process consists of tissue hydrolysis, protein removal, and purification using various methods. Mostly, these are chemical-dependent and are complex, multi-step processes. There is a developing trend for abandonment of harsh extraction chemicals and their substitution with different green-extraction technologies, however, these are still in their infancy. The quality of CSs is the first and foremost requirement for end-applications and is dependent on the extraction and purification methodologies used. The final products will show different bio-functional properties, depending on their origin and production methodology. This is a comprehensive review of the characteristics, properties, uses, sources, and extraction methods of CSs. This review emphasises the need for extraction and purification processes to be environmentally friendly and gentle, followed by product analysis and quality control to ensure the expected bioactivity of CSs.
Collapse
Affiliation(s)
- Sushanta Kumar Saha
- Shannon Applied Biotechnology Centre, LIFE Health and Biosciences Research Institute, Technological University of the Shannon: Midlands Midwest, Moylish Park, Limerick V94 E8YF, Ireland.
| | - Yin Zhu
- Shannon Applied Biotechnology Centre, LIFE Health and Biosciences Research Institute, Technological University of the Shannon: Midlands Midwest, Moylish Park, Limerick V94 E8YF, Ireland
| | - Patrick Murray
- Shannon Applied Biotechnology Centre, LIFE Health and Biosciences Research Institute, Technological University of the Shannon: Midlands Midwest, Moylish Park, Limerick V94 E8YF, Ireland
| | - Lena Madden
- Shannon Applied Biotechnology Centre, LIFE Health and Biosciences Research Institute, Technological University of the Shannon: Midlands Midwest, Moylish Park, Limerick V94 E8YF, Ireland
| |
Collapse
|
17
|
Chen X, Huang X, Liu Y, Zhang Z, Chen J. Assessing the causal associations of different types of statins use and knee/hip osteoarthritis: A Mendelian randomization study. PLoS One 2024; 19:e0297766. [PMID: 38648228 PMCID: PMC11034643 DOI: 10.1371/journal.pone.0297766] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Accepted: 01/11/2024] [Indexed: 04/25/2024] Open
Abstract
OBJECTIVE This study comprehensively evaluated the causal relationship between different types of statins use and knee/hip osteoarthritis (OA) using a two-sample and multivariate Mendelian randomization (MR) method. METHODS MR analysis was conducted using publicly available summary statistics data from genome-wide association studies (GWAS) to assess the causal associations between total statins use (including specific types) and knee/hip OA. The primary analysis utilized the inverse variance-weighted (IVW) method, with sensitivity analysis conducted to assess robustness. Multivariable MR (MVMR) analysis adjusted for low-density lipoprotein cholesterol (LDL-C), intermediate-density lipoprotein cholesterol (IDL-C), high-density lipoprotein cholesterol (HDL-C), and body mass index (BMI). RESULTS The MR analysis revealed a significant inverse association between genetically predicted total statins use and the risk of knee OA (OR = 0.950, 95%CI: 0.920-0.982, p = 0.002) as well as hip OA (OR = 0.932, 95%CI: 0.899-0.966, p <0.001). Furthermore, this study highlighted a reduced risk of knee/hip OA with the use of atorvastatin and simvastatin. Rosuvastatin use was associated with a decreased risk of hip OA but showed no association with knee OA. MVMR results indicated no correlation between exposure factors and outcomes after adjusting for LDL-C or IDL-C. HDL-C may not significantly contribute to statin-induced osteoarthritis, while BMI may play an important role. CONCLUSION This study provides compelling evidence of the close relationship between statin use and a reduced risk of knee/hip OA, particularly with atorvastatin and simvastatin. LDL-C and IDL-C may mediate these effects. These findings have important implications for the clinical prevention and treatment of knee/hip OA.
Collapse
Affiliation(s)
- Xin Chen
- Department of Urology, Mindong Hospital Affiliated to Fujian Medical University, Fuan, Fujian Province, People’s Republic of China
| | - Xin Huang
- Department of Orthopedics, Mindong Hospital Affiliated to Fujian Medical University, Fuan, Fujian Province, People’s Republic of China
| | - Youqun Liu
- Department of Nursing, Xiangan Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, People’s Republic of China
| | - Zhiwei Zhang
- Department of Orthopedics, Mindong Hospital Affiliated to Fujian Medical University, Fuan, Fujian Province, People’s Republic of China
| | - Jiliang Chen
- Department of Orthopedics, Mindong Hospital Affiliated to Fujian Medical University, Fuan, Fujian Province, People’s Republic of China
| |
Collapse
|
18
|
Yokota S, Ishizu H, Miyazaki T, Takahashi D, Iwasaki N, Shimizu T. Osteoporosis, Osteoarthritis, and Subchondral Insufficiency Fracture: Recent Insights. Biomedicines 2024; 12:843. [PMID: 38672197 PMCID: PMC11048726 DOI: 10.3390/biomedicines12040843] [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: 03/02/2024] [Revised: 03/31/2024] [Accepted: 04/08/2024] [Indexed: 04/28/2024] Open
Abstract
The increased incidence of osteoarthritis (OA), particularly knee and hip OA, and osteoporosis (OP), owing to population aging, have escalated the medical expense burden. Osteoarthritis is more prevalent in older women, and the involvement of subchondral bone fragility spotlights its association with OP. Notably, subchondral insufficiency fracture (SIF) may represent a more pronounced condition of OA pathophysiology. This review summarizes the relationship between OA and OP, incorporating recent insights into SIF. Progressive SIF leads to joint collapse and secondary OA and is associated with OP. Furthermore, the thinning and fragility of subchondral bone in early-stage OA suggest that SIF may be a subtype of OA (osteoporosis-related OA, OPOA) characterized by significant subchondral bone damage. The high bone mineral density observed in OA may be overestimated due to osteophytes and sclerosis and can potentially contribute to OPOA. The incidence of OPOA is expected to increase along with population aging. Therefore, prioritizing OP screening, early interventions for patients with early-stage OA, and fracture prevention measures such as rehabilitation, fracture liaison services, nutritional management, and medication guidance are essential.
Collapse
Affiliation(s)
| | | | | | | | | | - Tomohiro Shimizu
- Department of Orthopedic Surgery, Faculty of Medicine, Graduate School of Medicine, Hokkaido University, Sapporo 060-8638, Japan; (S.Y.); (H.I.); (T.M.); (D.T.); (N.I.)
| |
Collapse
|
19
|
Chen Z, Zhang S, Duan P, Yin Z, Dong S, Pang R, Tan H. Intra-articular injection of ascorbic acid enhances microfracture-mediated cartilage repair. Sci Rep 2024; 14:3811. [PMID: 38361039 PMCID: PMC10869716 DOI: 10.1038/s41598-024-54514-x] [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: 09/14/2023] [Accepted: 02/13/2024] [Indexed: 02/17/2024] Open
Abstract
Previous studies have confirmed that ascorbic acid (AA) can promote cartilage repair and improve cartilage differentiation in bone marrow mesenchymal stem cells. However, the use of microfracture (MFX) combined with AA to repair cartilage damage has not been studied. This study established a rabbit animal model and treated cartilage injury with different concentrations of AA combined with MFX. Macroscopic observations, histological analysis, immunohistochemical analysis and reverse transcription quantitative polymerase chain reaction analysis of TGF-β, AKT/Nrf2, and VEGF mRNA expression were performed. The results showed that intra-articular injection of AA had a positive effect on cartilage repair mediated by microfractures. Moreover, 10 mg/ml AA was the most effective at promoting cartilage repair mediated by microfractures. Intra-articular injection of AA promoted the synthesis of type II collagen and the formation of glycosaminoglycans by downregulating the mRNA expression of TGF-β and VEGF. In summary, this study confirmed that AA could promote cartilage repair after MFX surgery.
Collapse
Affiliation(s)
- Zhian Chen
- Graduate School, Kunming Medical University, Kunming City, Yunnan Province, China
| | - Sihe Zhang
- Department of Cell Biology, School of Medicine, Nankai University, Tianjin, China
| | - Peiya Duan
- Neurology Department, Longling County People's Hospital, Baoshan City, Yunnan Province, China
| | - Zhengbo Yin
- Graduate School, Kunming Medical University, Kunming City, Yunnan Province, China
| | - Shuangbin Dong
- Graduate School, Kunming Medical University, Kunming City, Yunnan Province, China
| | - Rongqing Pang
- Basic Medical Laboratory, People's Liberation Army Joint Logistic Support Force 920th Hospital, Kunming City, Yunnan Province, China.
| | - Hongbo Tan
- Department of Orthopaedics, People's Liberation Army Joint Logistic Support Force 920th Hospital, Kunming City, Yunnan Province, China.
| |
Collapse
|
20
|
Wang HH, Yii CY, Yong SB. Correspondence: Study Comparing Gutong Patch and NSAIDs in Treating Knee Osteoarthritis. Pharmacol Res 2024; 199:107026. [PMID: 38042357 DOI: 10.1016/j.phrs.2023.107026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/16/2023] [Accepted: 11/30/2023] [Indexed: 12/04/2023]
Affiliation(s)
- Hung-Hsuan Wang
- China Medical University Children's Hospital, Taichung, Taiwan
| | - Chin-Yuan Yii
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Landseed International Hospital, Taoyuan, Taiwan; Department of Biomedical Sciences and Engineering, National Central University, Taoyuan, Taiwan
| | - Su-Boon Yong
- Center for Allergy, Immunology, and Microbiome (A.I.M.), China Medical University Hospital, Taichung, Taiwan; Department of Allergy, Immunology, and Rheumatology (AIR), China Medical University Children's Hospital, Taichung, Taiwan; Department of Medicine, College of Medicine, China Medical University, Taichung, Taiwan
| |
Collapse
|
21
|
Wang Y, Li T, Weng X. Response letter to Hung-Hsuan Wang et al. on efficacy and safety of Gutong patch compared with NSAIDs for knee osteoarthritis: A real-world multicenter, prospective cohort study in China. Pharmacol Res 2024; 199:107035. [PMID: 38065296 DOI: 10.1016/j.phrs.2023.107035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Accepted: 12/05/2023] [Indexed: 01/13/2024]
Affiliation(s)
- Yingjie Wang
- Department of Orthopedic Surgery, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing 100730, China
| | - Tian Li
- School of Basic Medicine, Fourth Military Medical University, Xi'an 710032, China
| | - Xisheng Weng
- Department of Orthopedic Surgery, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing 100730, China.
| |
Collapse
|
22
|
Zhang YZ, Wei ZJ, Yu SN, Wang XY, Wang Y, Wu CA, Jiang X. Dihydrotanshinone I protects human chondrocytes and alleviates damage from spontaneous osteoarthritis in a guinea pig model. Sci Rep 2023; 13:21355. [PMID: 38049518 PMCID: PMC10696037 DOI: 10.1038/s41598-023-48902-y] [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: 09/20/2023] [Accepted: 11/30/2023] [Indexed: 12/06/2023] Open
Abstract
Osteoarthritis (OA) is the most common degenerative joint disease. Currently, no satisfactory pharmacological treatment exists for OA. The potential anti-inflammatory properties of Dihydrotanshinone I (DHT) have been reported, but its effects on OA are unclear. In this study, we assess the impact of DHT on the viability of human chondrocytes in vitro. We then use a guinea pig model to investigate the effects of DHT on knee osteoarthritis progression. Twelve-week-old Dunkin Hartley guinea pigs spontaneously developing OA were intraperitoneally injected with different doses of DHT for eight weeks. Micro-CT analysis was performed on the subchondral bone in the knee, and histological assessment of the knee joint was done using stained sections, the ratio of hyaline to calcified cartilage, and Mankin scores. DHT successfully restored IL-1β-induced decreases in cell viability in human primary chondrocytes. In the guinea pig model, intraperitoneal injections of DHT ameliorated age-induced OA, effectively reduced the expression level of two cartilage metabolism-related genes (ADAMTS4 and MMP13) and decreased the inflammatory biomarker IL-6 in the serum of guinea pigs developing spontaneous osteoarthritis. These findings demonstrate DHT's protective effects on chondrocytes and suggest that it alleviates cartilage degradation and proteoglycan loss in OA.
Collapse
Affiliation(s)
- Yan-Zhuo Zhang
- National Center for Orthopaedics, Department of Molecular Orthopaedics, Beijing Research Institute of Traumatology and Orthopaedics, Beijing Jishuitan Hospital, Capital Medical University, Beijing, 100035, People's Republic of China
| | - Zhen-Jie Wei
- National Center for Orthopaedics, Department of Molecular Orthopaedics, Beijing Research Institute of Traumatology and Orthopaedics, Beijing Jishuitan Hospital, Capital Medical University, Beijing, 100035, People's Republic of China
| | - Shu-Nan Yu
- National Center for Orthopaedics, Department of Molecular Orthopaedics, Beijing Research Institute of Traumatology and Orthopaedics, Beijing Jishuitan Hospital, Capital Medical University, Beijing, 100035, People's Republic of China
| | - Xin-Yu Wang
- National Center for Orthopaedics, Department of Molecular Orthopaedics, Beijing Research Institute of Traumatology and Orthopaedics, Beijing Jishuitan Hospital, Capital Medical University, Beijing, 100035, People's Republic of China
| | - Ying Wang
- National Center for Orthopaedics, Department of Molecular Orthopaedics, Beijing Research Institute of Traumatology and Orthopaedics, Beijing Jishuitan Hospital, Capital Medical University, Beijing, 100035, People's Republic of China
| | - Cheng-Ai Wu
- National Center for Orthopaedics, Department of Molecular Orthopaedics, Beijing Research Institute of Traumatology and Orthopaedics, Beijing Jishuitan Hospital, Capital Medical University, Beijing, 100035, People's Republic of China
| | - Xu Jiang
- National Center for OrthopaedicsDepartment of Orthopaedics, Beijing Research Institute of Traumatology and Orthopaedics, Beijing Jishuitan Hospital, Capital Medical University, Beijing, 100035, People's Republic of China.
| |
Collapse
|
23
|
Santos MM, Santos AM, Nascimento Júnior JAC, Andrade TDA, Rajkumar G, Frank LA, Serafini MR. The management of osteoarthritis symptomatology through nanotechnology: a patent review. J Microencapsul 2023; 40:475-490. [PMID: 37698545 DOI: 10.1080/02652048.2023.2258955] [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: 03/17/2023] [Accepted: 09/11/2023] [Indexed: 09/13/2023]
Abstract
Osteoarthritis is considered a degenerative joint disease that is characterised by inflammation, chronic pain, and functional limitation. The increasing development of nanotechnology in drug delivery systems has provided new ideas and methods for osteoarthritis therapy. This review aimed to evaluate patents that have developed innovations, therapeutic strategies, and alternatives using nanotechnology in osteoarthritis treatment. The results show patents deposited from 2015 to November 2021 in the online databases European Patent Office and World Intellectual Property Organisation. A total of 651 patents were identified for preliminary assessment and 16 were selected for full reading and discussion. The evaluated patents are focused on the intraarticular route, oral route, and topical route for osteoarthritis treatment. The intraarticular route presented a higher patent number, followed by the oral and topical routes, respectively. The development of new technologies allows us to envision a promising and positive future in osteoarthritis treatment.
Collapse
Affiliation(s)
| | | | | | | | - Gomathi Rajkumar
- Department of Botany, Sri Sarada College for Women (Autonomous), Affiliated to Periyar University, Salem, India
| | - Luiza Abrahão Frank
- Postgraduate Program in Pharmaceutical Sciences, Federal University of Rio Grande do Sul, Porto Alegre, Brazil
| | - Mairim Russo Serafini
- Postgraduate Program in Health Sciences, Federal University of Sergipe, Aracaju, Brazil
- Postgraduate Program in Pharmaceutical Sciences, Federal University of Sergipe, São Cristóvão, Brazil
| |
Collapse
|
24
|
Yamamoto N, Szymski D, Voss A, Ishikawa H, Muraki T, Cunha RA, Ejnisman B, Noack J, McCarty E, Mulcahey MK, Itoi E. Non-operative management of shoulder osteoarthritis: Current concepts. J ISAKOS 2023; 8:289-295. [PMID: 37321293 DOI: 10.1016/j.jisako.2023.06.002] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Revised: 03/26/2023] [Accepted: 06/08/2023] [Indexed: 06/17/2023]
Abstract
Glenohumeral osteoarthritis (OA) is one of the most common causes of shoulder pain. Conservative treatment options include physical therapy, pharmacological therapy, and biological therapy. Patients with glenohumeral OA present shoulder pain and decreased shoulder range of motion (ROM). Abnormal scapular motion is also seen in patients as adaptation to the restricted glenohumeral motion. Physical therapy is performed to (1) decrease pain, (2) increase shoulder ROM, and (3) protect the glenohumeral joint. To decrease pain, it should be assessed whether the pain appears at rest or during shoulder motion. Physical therapy may be effective for motion pain rather than rest pain. To increase shoulder ROM, the soft tissues responsible for the ROM loss need to be identified and targeted for intervention. To protect the glenohumeral joint, rotator cuff strengthening exercises are recommended. Administration of pharmacological agents is the major part next to physical therapy in the conservative treatment. The main aim of pharmacological treatment is the reduction of pain and diminution of inflammation in the joint. To achieve this aim, non-steroidal anti-inflammatory drugs are recommended as first-line therapy. Additionally, the supplementation of oral vitamin C and vitamin D can help to slow down cartilage degeneration. Depending on the individual comorbidities and contraindications, sufficient medication with good pain reduction is thus possible for each patient. This interrupts the chronic inflammatory state in the joint and, in turn, enables pain-free physical therapy. Biologics such as platelet-rich plasma, bone marrow aspirate concentrate, and mesenchymal stem cells have gathered increased attention. Good clinical outcomes have been reported, but we need to be aware that these options are helpful in decreasing shoulder pain but neither stopping the progression nor improving OA. Further evidence of biologics needs to be obtained to determine their effectiveness. In athletes, a combined approach of activity modification and physical therapy can be effective. Oral medications can provide patients with transient pain relief. Intra-articular corticosteroid injection, which provides longer-term effects, must be used cautiously in athletes. There is mixed evidence for the efficacy of hyaluronic acid injections. There is still limited evidence regarding the use of biologics.
Collapse
Affiliation(s)
- Nobuyuki Yamamoto
- Department of Orthopaedic Surgery, Tohoku University School of Medicine, Sendai 980-8574, Japan
| | - Dominik Szymski
- Department of Trauma Surgery, University Medical Center Regensburg, Regensburg 93053, Germany; Sporthopaedicum Regensburg, Regensburg 93053, Germany
| | - Andreas Voss
- Department of Trauma Surgery, University Medical Center Regensburg, Regensburg 93053, Germany; Sporthopaedicum Regensburg, Regensburg 93053, Germany
| | - Hiroaki Ishikawa
- Department of Physical Medicine and Rehabilitation, Tohoku University Graduate School of Medicine, Sendai 980-8574, Japan
| | - Takayuki Muraki
- Department of Physical Medicine and Rehabilitation, Tohoku University Graduate School of Medicine, Sendai 980-8574, Japan
| | - Ronaldo A Cunha
- Department of Sports Medicine, Federal University of São Paulo, São Paulo 05508-090, Brazil
| | - Benno Ejnisman
- Department of Sports Medicine, Federal University of São Paulo, São Paulo 05508-090, Brazil
| | - Joseph Noack
- Department of Orthopaedic Surgery, University of Colorado, CO 80309, USA
| | - Eric McCarty
- Department of Orthopaedic Surgery, University of Colorado, CO 80309, USA
| | - Mary K Mulcahey
- Department of Orthopaedic Surgery, Tulane University School of Medicine, LA 70112, USA
| | - Eiji Itoi
- Department of Orthopaedic Surgery, Tohoku Rosai Hospital, Sendai 981-8563, Japan.
| |
Collapse
|
25
|
Sun T, Wang M, Liang W, Gao P, Liu Q, Yan X. Revealing mechanism of Methazolamide for treatment of ankylosing spondylitis based on network pharmacology and GSEA. Sci Rep 2023; 13:15370. [PMID: 37717047 PMCID: PMC10505193 DOI: 10.1038/s41598-023-42721-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2023] [Accepted: 09/14/2023] [Indexed: 09/18/2023] Open
Abstract
Methazolamide is a carbonic anhydrase (CA) inhibitor with satisfactory safety. Our previous studies have demonstrated the elevation of CA1 expression and the therapeutic effect of Methazolamide in Ankylosing spondylitis (AS). In this study, we explored the pathogenic role of CA1 and the pharmacological mechanism of Methazolamide in AS through Gene Set Enrichment Analysis (GSEA) and network pharmacology. Seven out of twelve CA1 related gene sets were enriched in AS group. CA1 was core enriched in above seven gene sets involving zinc ion binding, arylesterase activity and one carbon metabolic process. Functional analysis of the candidate target genes obtained from the intersection of AS associated genes and Methazolamide target genes indicated that Methazolamide exerts therapeutic effects on AS mainly through inflammatory pathways which regulate the production of tumor necrosis factor, IL-6 and nitric oxide. PTGS2, ESR1, GSK3β, JAK2, NOS2 and CA1 were selected as therapeutic targets of Methazolamide in AS. Molecular docking and molecular dynamics simulations were performed successfully. In addition, we innovatively obtained the intersection of Gene Ontology (GO)/Kyoto Encyclopedia of Genes and Genomes (KEGG) analyses and GSEA results, and found that 18 GO terms and 5 KEGG terms were indicated in the pharmacological mechanism of Methazolamide in AS, involving bone mineralization, angiogenesis, inflammation, and chemokine signaling pathways. Nevertheless, validation for these mechanisms is needed in vivo/vitro experiments.
Collapse
Affiliation(s)
- Tao Sun
- Department of Orthopedic Surgery, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, No. 16766, Jingshi Road, Lixia District, Jinan City, Shandong Province, China
| | - Manzhi Wang
- Department of Hematology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, Shandong, China
| | - Weiqiang Liang
- Department of Orthopedic Surgery, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, No. 16766, Jingshi Road, Lixia District, Jinan City, Shandong Province, China
| | - Ping Gao
- Department of Orthopedic Surgery, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, No. 16766, Jingshi Road, Lixia District, Jinan City, Shandong Province, China
| | - Qiang Liu
- Department of Cardiology, Shandong Medicine and Health Key Laboratory of Cardiac Electrophysiology and Arrhythmia, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong First Medical University, Jinan, Shandong, China
| | - Xinfeng Yan
- Department of Orthopedic Surgery, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, No. 16766, Jingshi Road, Lixia District, Jinan City, Shandong Province, China.
| |
Collapse
|
26
|
Zhou L, Xu J, Schwab A, Tong W, Xu J, Zheng L, Li Y, Li Z, Xu S, Chen Z, Zou L, Zhao X, van Osch GJ, Wen C, Qin L. Engineered biochemical cues of regenerative biomaterials to enhance endogenous stem/progenitor cells (ESPCs)-mediated articular cartilage repair. Bioact Mater 2023; 26:490-512. [PMID: 37304336 PMCID: PMC10248882 DOI: 10.1016/j.bioactmat.2023.03.008] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Revised: 02/21/2023] [Accepted: 03/13/2023] [Indexed: 06/13/2023] Open
Abstract
As a highly specialized shock-absorbing connective tissue, articular cartilage (AC) has very limited self-repair capacity after traumatic injuries, posing a heavy socioeconomic burden. Common clinical therapies for small- to medium-size focal AC defects are well-developed endogenous repair and cell-based strategies, including microfracture, mosaicplasty, autologous chondrocyte implantation (ACI), and matrix-induced ACI (MACI). However, these treatments frequently result in mechanically inferior fibrocartilage, low cost-effectiveness, donor site morbidity, and short-term durability. It prompts an urgent need for innovative approaches to pattern a pro-regenerative microenvironment and yield hyaline-like cartilage with similar biomechanical and biochemical properties as healthy native AC. Acellular regenerative biomaterials can create a favorable local environment for AC repair without causing relevant regulatory and scientific concerns from cell-based treatments. A deeper understanding of the mechanism of endogenous cartilage healing is furthering the (bio)design and application of these scaffolds. Currently, the utilization of regenerative biomaterials to magnify the repairing effect of joint-resident endogenous stem/progenitor cells (ESPCs) presents an evolving improvement for cartilage repair. This review starts by briefly summarizing the current understanding of endogenous AC repair and the vital roles of ESPCs and chemoattractants for cartilage regeneration. Then several intrinsic hurdles for regenerative biomaterials-based AC repair are discussed. The recent advances in novel (bio)design and application regarding regenerative biomaterials with favorable biochemical cues to provide an instructive extracellular microenvironment and to guide the ESPCs (e.g. adhesion, migration, proliferation, differentiation, matrix production, and remodeling) for cartilage repair are summarized. Finally, this review outlines the future directions of engineering the next-generation regenerative biomaterials toward ultimate clinical translation.
Collapse
Affiliation(s)
- Liangbin Zhou
- Musculoskeletal Research Laboratory of Department of Orthopaedics and Traumatology & Innovative Orthopaedic Biomaterials and Drug Translational Research Laboratory of Li Ka Shing Institute of Health Sciences, Faculty of Medicine, The Chinese University of Hong Kong, 999077, Hong Kong SAR, China
- Department of Biomedical Engineering, Faculty of Engineering, The Hong Kong Polytechnic University, 999077, Hong Kong SAR, China
| | - Jietao Xu
- Department of Orthopaedics and Sports Medicine, Erasmus MC, University Medical Center Rotterdam, 3015 GD, Rotterdam, the Netherlands
| | - Andrea Schwab
- Department of Orthopaedics and Sports Medicine, Erasmus MC, University Medical Center Rotterdam, 3015 GD, Rotterdam, the Netherlands
- Department of Oral and Maxillofacial Surgery, Erasmus MC, University Medical Center Rotterdam, 3015 GD, Rotterdam, the Netherlands
| | - Wenxue Tong
- Musculoskeletal Research Laboratory of Department of Orthopaedics and Traumatology & Innovative Orthopaedic Biomaterials and Drug Translational Research Laboratory of Li Ka Shing Institute of Health Sciences, Faculty of Medicine, The Chinese University of Hong Kong, 999077, Hong Kong SAR, China
| | - Jiankun Xu
- Musculoskeletal Research Laboratory of Department of Orthopaedics and Traumatology & Innovative Orthopaedic Biomaterials and Drug Translational Research Laboratory of Li Ka Shing Institute of Health Sciences, Faculty of Medicine, The Chinese University of Hong Kong, 999077, Hong Kong SAR, China
| | - Lizhen Zheng
- Musculoskeletal Research Laboratory of Department of Orthopaedics and Traumatology & Innovative Orthopaedic Biomaterials and Drug Translational Research Laboratory of Li Ka Shing Institute of Health Sciences, Faculty of Medicine, The Chinese University of Hong Kong, 999077, Hong Kong SAR, China
- Centre for Regenerative Medicine and Health, Hong Kong Institute of Science & Innovation, Chinese Academy of Sciences - CRMH, 999077, Hong Kong SAR, China
| | - Ye Li
- Musculoskeletal Research Laboratory of Department of Orthopaedics and Traumatology & Innovative Orthopaedic Biomaterials and Drug Translational Research Laboratory of Li Ka Shing Institute of Health Sciences, Faculty of Medicine, The Chinese University of Hong Kong, 999077, Hong Kong SAR, China
| | - Zhuo Li
- Department of Biomedical Engineering, Faculty of Engineering, The Chinese University of Hong Kong, 999077, Hong Kong SAR, China
| | - Shunxiang Xu
- Musculoskeletal Research Laboratory of Department of Orthopaedics and Traumatology & Innovative Orthopaedic Biomaterials and Drug Translational Research Laboratory of Li Ka Shing Institute of Health Sciences, Faculty of Medicine, The Chinese University of Hong Kong, 999077, Hong Kong SAR, China
| | - Ziyi Chen
- Musculoskeletal Research Laboratory of Department of Orthopaedics and Traumatology & Innovative Orthopaedic Biomaterials and Drug Translational Research Laboratory of Li Ka Shing Institute of Health Sciences, Faculty of Medicine, The Chinese University of Hong Kong, 999077, Hong Kong SAR, China
| | - Li Zou
- Musculoskeletal Research Laboratory of Department of Orthopaedics and Traumatology & Innovative Orthopaedic Biomaterials and Drug Translational Research Laboratory of Li Ka Shing Institute of Health Sciences, Faculty of Medicine, The Chinese University of Hong Kong, 999077, Hong Kong SAR, China
| | - Xin Zhao
- Department of Biomedical Engineering, Faculty of Engineering, The Hong Kong Polytechnic University, 999077, Hong Kong SAR, China
| | - Gerjo J.V.M. van Osch
- Department of Orthopaedics and Sports Medicine, Erasmus MC, University Medical Center Rotterdam, 3015 GD, Rotterdam, the Netherlands
- Department of Otorhinolaryngology, Erasmus MC, University Medical Center Rotterdam, 3015 GD, Rotterdam, the Netherlands
- Department of Biomechanical Engineering, Faculty of Mechanical, Maritime and Materials Engineering, Delft University of Technology (TU Delft), 2600 AA, Delft, the Netherlands
| | - Chunyi Wen
- Department of Biomedical Engineering, Faculty of Engineering, The Hong Kong Polytechnic University, 999077, Hong Kong SAR, China
| | - Ling Qin
- Musculoskeletal Research Laboratory of Department of Orthopaedics and Traumatology & Innovative Orthopaedic Biomaterials and Drug Translational Research Laboratory of Li Ka Shing Institute of Health Sciences, Faculty of Medicine, The Chinese University of Hong Kong, 999077, Hong Kong SAR, China
- Centre for Translational Medicine Research and Development, Shenzhen Institute of Advanced Technology, The Chinese Academy of Sciences, 518000, Shenzhen, China
| |
Collapse
|
27
|
Jiang Q, Zhang S. Stimulus-Responsive Drug Delivery Nanoplatforms for Osteoarthritis Therapy. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2023; 19:e2206929. [PMID: 36905239 DOI: 10.1002/smll.202206929] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Revised: 02/16/2023] [Indexed: 06/08/2023]
Abstract
Osteoarthritis (OA) is one of the most prevalent age-related degenerative diseases. With an increasingly aging global population, greater numbers of OA patients are providing clear economic and societal burdens. Surgical and pharmacological treatments are the most common and conventional therapeutic strategies for OA, but often fall considerably short of desired or optimal outcomes. With the development of stimulus-responsive nanoplatforms has come the potential for improved therapeutic strategies for OA. Enhanced control, longer retention time, higher loading rates, and increased sensitivity are among the potential benefits. This review summarizes the advanced application of stimulus-responsive drug delivery nanoplatforms for OA, categorized by either those that depend on endogenous stimulus (reactive oxygen species, pH, enzyme, and temperature), or those that depend on exogenous stimulus (near-infrared ray, ultrasound, magnetic fields). The opportunities, restrictions, and limitations related to these various drug delivery systems, or their combinations, are discussed in areas such as multi-functionality, image guidance, and multi-stimulus response. The remaining constraints and potential solutions that are represented by the clinical application of stimulus-responsive drug delivery nanoplatforms are finally summarized.
Collapse
Affiliation(s)
- Qi Jiang
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cells and Regenerative Medicine, Department of Orthopedic Surgery of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310058, China
- Zhejiang University-University of Edinburgh Institute, Zhejiang University School of Medicine, Key Laboratory of Tissue Engineering and Regenerative Medicine of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, 310058, China
- China Orthopedic Regenerative Medicine Group (CORMed), Hangzhou, 310058, China
| | - Shufang Zhang
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cells and Regenerative Medicine, Department of Orthopedic Surgery of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310058, China
- Zhejiang University-University of Edinburgh Institute, Zhejiang University School of Medicine, Key Laboratory of Tissue Engineering and Regenerative Medicine of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, 310058, China
- China Orthopedic Regenerative Medicine Group (CORMed), Hangzhou, 310058, China
| |
Collapse
|
28
|
Toupet K, Jorgensen C, Noël D. An injectable copolymer of fatty acids (ARA 3000 BETA) as a promising treatment for osteoarthritis. Sci Rep 2023; 13:7783. [PMID: 37179423 PMCID: PMC10183038 DOI: 10.1038/s41598-023-34206-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Accepted: 04/26/2023] [Indexed: 05/15/2023] Open
Abstract
Osteoarthritis (OA) is the most prevalent rheumatic disease and a fast growing cause of disability. Current pharmacological treatments include antalgics and non-steroid anti-inflammatory drugs to control pain and inflammation as well as slow acting drugs such as intra-articular (IA) administration of hyaluronic acid. Oral supplementation or diet rich in polyunsaturated free fatty acids are proposed but evidence for benefit is still under debate. We here investigated the therapeutic potential of ARA 3000 BETA, an injectable copolymer of fatty acids, at the structural level in OA. Collagenase-induced osteoarthritis model was induced in C57BL/6 mice by collagenase injection into knee joint. Mice were treated with one or two IA or four intra-muscular injections (IM) of ARA 3000 BETA. At sacrifice, knee joints were recovered for cartilage analysis by confocal laser scanning microscopy (CLSM) and bone analysis by micro-computed tomography system. OA histological scoring was performed after safranin O/fast green staining. Histological analysis revealed a protective effect against cartilage degradation in treated knee joints after IM and IA administration. This was confirmed by CLSM with a significant improvement of all articular cartilage parameters, including thickness, volume and surface degradation whatever the administration route. A slight protective effect was also noticed on subchondral bone parameters and knee joint calcification after IM administration and to a lesser extent, two IA injections. We demonstrated the therapeutic efficacy of injectable ARA 3000 BETA in OA with a protection against cartilage and bone alterations providing the proof-of-concept that clinical translation might be envisioned to delay disease progression.
Collapse
Affiliation(s)
- Karine Toupet
- IRMB, University of Montpellier, INSERM, Montpellier, France
- ECELLFRANCE, University of Montpellier, INSERM, CHU Montpellier, Montpellier, France
| | - Christian Jorgensen
- IRMB, University of Montpellier, INSERM, Montpellier, France
- ECELLFRANCE, University of Montpellier, INSERM, CHU Montpellier, Montpellier, France
- Clinical Immunology and Osteoarticular Diseases Therapeutic Unit, Hôpital Lapeyronie, Montpellier, France
| | - Danièle Noël
- IRMB, University of Montpellier, INSERM, Montpellier, France.
- ECELLFRANCE, University of Montpellier, INSERM, CHU Montpellier, Montpellier, France.
- Clinical Immunology and Osteoarticular Diseases Therapeutic Unit, Hôpital Lapeyronie, Montpellier, France.
| |
Collapse
|
29
|
Shanmugasundaram S, Solanki K, Saseendar S, Chavada VK, D'Ambrosi R. Role of Doxycycline as an Osteoarthritis Disease-Modifying Drug. J Clin Med 2023; 12:jcm12082927. [PMID: 37109263 PMCID: PMC10145123 DOI: 10.3390/jcm12082927] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2023] [Revised: 04/03/2023] [Accepted: 04/06/2023] [Indexed: 04/29/2023] Open
Abstract
Doxycycline is a drug that has been proposed to modify osteoarthritis (OA) progression, in addition to its role as an antibiotic. However, available evidence thus far comprises sporadic reports, with no consensus on its benefits. Hence, this review attempts to analyze the evidence available thus far on the role of doxycycline as a disease-modifying osteoarthritis drug (DMOAD) in knee osteoarthritis. The earliest evidence of doxycycline in OA appeared in 1991 when doxycycline was found to inhibit the type XI collagenolytic activity of extracts from the human osteoarthritic cartilage, and gelatinase and tetracycline were found to inhibit this metalloproteinase activity in articular cartilage in vivo, which could modify cartilage breakdown in osteoarthritis. Apart from the inhibition of cartilage damage by metalloproteinases (MMPs) and other cartilage-related mechanisms, doxycycline also affects the bone and interferes with many enzyme systems. The most significant finding after reviewing various studies was that doxycycline has a definitive role in structural changes in osteoarthritis progression and radiological joint space width, but its role in the improvement of clinical outcomes as a DMOAD has not been established. However, there is much of a gap and lack of evidence in this regard. Doxycycline, as an MMP inhibitor, has theoretical advantages for clinical outcomes, but the present studies reveal only beneficial structural changes in osteoarthritis and very minimal or nonexistent advantages in clinical outcomes. Current evidence does not favor the regular use of doxycycline for the treatment of osteoarthritis as an individual treatment option or in combination with others. However, multicenter large cohort studies are warranted to determine the long-term benefits of doxycycline.
Collapse
Affiliation(s)
| | | | | | - Vijay K Chavada
- Department of Community Medicine, Indira Gandhi Medical College and Research Institute, Puducherry 605009, India
| | - Riccardo D'Ambrosi
- IRCCS Ospedale Galeazzi-Sant'Ambrogio, 20161 Milan, Italy
- Dipartimento di Scienze Biomediche per la Salute, Università degli Studi di Milano, 20133 Milan, Italy
| |
Collapse
|
30
|
Yao Q, Wu X, Tao C, Gong W, Chen M, Qu M, Zhong Y, He T, Chen S, Xiao G. Osteoarthritis: pathogenic signaling pathways and therapeutic targets. Signal Transduct Target Ther 2023; 8:56. [PMID: 36737426 PMCID: PMC9898571 DOI: 10.1038/s41392-023-01330-w] [Citation(s) in RCA: 432] [Impact Index Per Article: 216.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Revised: 01/06/2023] [Accepted: 01/17/2023] [Indexed: 02/05/2023] Open
Abstract
Osteoarthritis (OA) is a chronic degenerative joint disorder that leads to disability and affects more than 500 million population worldwide. OA was believed to be caused by the wearing and tearing of articular cartilage, but it is now more commonly referred to as a chronic whole-joint disorder that is initiated with biochemical and cellular alterations in the synovial joint tissues, which leads to the histological and structural changes of the joint and ends up with the whole tissue dysfunction. Currently, there is no cure for OA, partly due to a lack of comprehensive understanding of the pathological mechanism of the initiation and progression of the disease. Therefore, a better understanding of pathological signaling pathways and key molecules involved in OA pathogenesis is crucial for therapeutic target design and drug development. In this review, we first summarize the epidemiology of OA, including its prevalence, incidence and burdens, and OA risk factors. We then focus on the roles and regulation of the pathological signaling pathways, such as Wnt/β-catenin, NF-κB, focal adhesion, HIFs, TGFβ/ΒΜP and FGF signaling pathways, and key regulators AMPK, mTOR, and RUNX2 in the onset and development of OA. In addition, the roles of factors associated with OA, including MMPs, ADAMTS/ADAMs, and PRG4, are discussed in detail. Finally, we provide updates on the current clinical therapies and clinical trials of biological treatments and drugs for OA. Research advances in basic knowledge of articular cartilage biology and OA pathogenesis will have a significant impact and translational value in developing OA therapeutic strategies.
Collapse
Affiliation(s)
- Qing Yao
- Department of Biochemistry, School of Medicine, Shenzhen Key Laboratory of Cell Microenvironment, Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Southern University of Science and Technology, Shenzhen, 518055, China.
| | - Xiaohao Wu
- Department of Biochemistry, School of Medicine, Shenzhen Key Laboratory of Cell Microenvironment, Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Southern University of Science and Technology, Shenzhen, 518055, China
| | - Chu Tao
- Department of Biochemistry, School of Medicine, Shenzhen Key Laboratory of Cell Microenvironment, Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Southern University of Science and Technology, Shenzhen, 518055, China
| | - Weiyuan Gong
- Department of Biochemistry, School of Medicine, Shenzhen Key Laboratory of Cell Microenvironment, Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Southern University of Science and Technology, Shenzhen, 518055, China
| | - Mingjue Chen
- Department of Biochemistry, School of Medicine, Shenzhen Key Laboratory of Cell Microenvironment, Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Southern University of Science and Technology, Shenzhen, 518055, China
| | - Minghao Qu
- Department of Biochemistry, School of Medicine, Shenzhen Key Laboratory of Cell Microenvironment, Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Southern University of Science and Technology, Shenzhen, 518055, China
| | - Yiming Zhong
- Department of Biochemistry, School of Medicine, Shenzhen Key Laboratory of Cell Microenvironment, Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Southern University of Science and Technology, Shenzhen, 518055, China
| | - Tailin He
- Department of Biochemistry, School of Medicine, Shenzhen Key Laboratory of Cell Microenvironment, Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Southern University of Science and Technology, Shenzhen, 518055, China
| | - Sheng Chen
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Guozhi Xiao
- Department of Biochemistry, School of Medicine, Shenzhen Key Laboratory of Cell Microenvironment, Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Southern University of Science and Technology, Shenzhen, 518055, China.
| |
Collapse
|
31
|
Resveratrol Improves the Progression of Osteoarthritis by Regulating the SIRT1-FoxO1 Pathway-Mediated Cholesterol Metabolism. Mediators Inflamm 2023; 2023:2936236. [PMID: 36643587 PMCID: PMC9833897 DOI: 10.1155/2023/2936236] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Revised: 11/29/2022] [Accepted: 12/02/2022] [Indexed: 01/06/2023] Open
Abstract
Osteoarthritis (OA) is considered a metabolic disorder. This study investigated the effect of resveratrol (RES) on cholesterol accumulation in osteoarthritic articular cartilage via the silent information regulator 1 (SIRT1)/forkhead transcription factor (FoxO1) pathway. Interleukin (IL)-1β-treated chondrocytes that mimic OA chondrocytes were used in in vitro experiments. The optimal RES concentration was selected based on the results of chondrocyte proliferation in the Cell Counting Kit-8 assay. Western blotting, immunofluorescence, and reverse transcription-quantitative polymerase chain reaction were performed. For the animal experiments, mice were randomly divided into the RES group (n = 15), medial meniscus destabilization group (n = 15), and sham group (n = 15), and each group received the same dose of RES or saline. Articular cartilage tissue was obtained eight weeks after surgery for relevant histological analysis. Clinical tissue test results suggest that downregulation of the SIRT1/FoxO1 pathway is associated with cholesterol buildup in OA chondrocytes. For the in vitro studies, RES increased the expression of SIRT1 and phosphorylation of FoxO1 in IL-1β-treated chondrocytes, promoted the expression of cholesterol efflux factor liver X receptor alpha (LXRα), and inhibited the expression of cholesterol synthesis-associated factor sterol-regulatory element binding proteins 2 (SREBP2). This reduced IL-1β-induced chondrocytes cholesterol accumulation. SIRT1 inhibition prevented the RES-mediated reduction in cholesterol buildup. Inhibiting FoxO1 but not SIRT1 reduced FoxO1 phosphorylation and increased cholesterol buildup in cultured chondrocytes. Additionally, in vivo experiments have shown that RES can alleviate cholesterol buildup and pathological changes in OA cartilage. Our findings suggest that RES regulates cholesterol buildup in osteoarthritic articular cartilage via the SIRT1/FoxO1 pathway, thereby improving the progression of OA.
Collapse
|
32
|
Li G, Liu S, Xu H, Chen Y, Deng J, Xiong A, Wang D, Weng J, Yu F, Gao L, Ding C, Zeng H. Potential effects of teriparatide (PTH (1-34)) on osteoarthritis: a systematic review. Arthritis Res Ther 2023; 25:3. [PMID: 36609338 PMCID: PMC9817404 DOI: 10.1186/s13075-022-02981-w] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Accepted: 12/19/2022] [Indexed: 01/09/2023] Open
Abstract
Osteoarthritis (OA) is a common and prevalent degenerative joint disease characterized by degradation of the articular cartilage. However, none of disease-modifying OA drugs is approved currently. Teriparatide (PTH (1-34)) might stimulate chondrocyte proliferation and cartilage regeneration via some uncertain mechanisms. Relevant therapies of PTH (1-34) on OA with such effects have recently gained increasing interest, but have not become widespread practice. Thus, we launch this systematic review (SR) to update the latest evidence accordingly. A comprehensive literature search was conducted in PubMed, Web of Science, MEDLINE, the Cochrane Library, and Embase from their inception to February 2022. Studies investigating the effects of the PTH (1-34) on OA were obtained. The quality assessment and descriptive summary were made of all included studies. Overall, 307 records were identified, and 33 studies were included. In vivo studies (n = 22) concluded that PTH (1-34) slowed progression of OA by alleviating cartilage degeneration and aberrant remodeling of subchondral bone (SCB). Moreover, PTH (1-34) exhibited repair of cartilage and SCB, analgesic, and anti-inflammatory effects. In vitro studies (n = 11) concluded that PTH (1-34) was important for chondrocytes via increasing the proliferation and matrix synthesis but preventing apoptosis or hypertrophy. All included studies were assessed with low or unclear risk of bias in methodological quality. The SR demonstrated that PTH (1-34) could alleviate the progression of OA. Moreover, PTH (1-34) had beneficial effects on osteoporotic OA (OPOA) models, which might be a therapeutic option for OA and OPOA treatment.
Collapse
Affiliation(s)
- Guoqing Li
- grid.440601.70000 0004 1798 0578Department of Bone & Joint Surgery, Peking University Shenzhen Hospital, Shenzhen, 518036 People’s Republic of China ,grid.440601.70000 0004 1798 0578National & Local Joint Engineering Research Center of Orthopaedic Biomaterials, Peking University Shenzhen Hospital, Shenzhen, 518036 People’s Republic of China
| | - Su Liu
- grid.440601.70000 0004 1798 0578Department of Bone & Joint Surgery, Peking University Shenzhen Hospital, Shenzhen, 518036 People’s Republic of China ,grid.440601.70000 0004 1798 0578National & Local Joint Engineering Research Center of Orthopaedic Biomaterials, Peking University Shenzhen Hospital, Shenzhen, 518036 People’s Republic of China
| | - Huihui Xu
- grid.440601.70000 0004 1798 0578Department of Bone & Joint Surgery, Peking University Shenzhen Hospital, Shenzhen, 518036 People’s Republic of China ,grid.440601.70000 0004 1798 0578National & Local Joint Engineering Research Center of Orthopaedic Biomaterials, Peking University Shenzhen Hospital, Shenzhen, 518036 People’s Republic of China
| | - Yixiao Chen
- grid.440601.70000 0004 1798 0578Department of Bone & Joint Surgery, Peking University Shenzhen Hospital, Shenzhen, 518036 People’s Republic of China ,grid.440601.70000 0004 1798 0578National & Local Joint Engineering Research Center of Orthopaedic Biomaterials, Peking University Shenzhen Hospital, Shenzhen, 518036 People’s Republic of China
| | - Jiapeng Deng
- grid.440601.70000 0004 1798 0578Department of Bone & Joint Surgery, Peking University Shenzhen Hospital, Shenzhen, 518036 People’s Republic of China ,grid.440601.70000 0004 1798 0578National & Local Joint Engineering Research Center of Orthopaedic Biomaterials, Peking University Shenzhen Hospital, Shenzhen, 518036 People’s Republic of China
| | - Ao Xiong
- grid.440601.70000 0004 1798 0578Department of Bone & Joint Surgery, Peking University Shenzhen Hospital, Shenzhen, 518036 People’s Republic of China ,grid.440601.70000 0004 1798 0578National & Local Joint Engineering Research Center of Orthopaedic Biomaterials, Peking University Shenzhen Hospital, Shenzhen, 518036 People’s Republic of China
| | - Deli Wang
- grid.440601.70000 0004 1798 0578Department of Bone & Joint Surgery, Peking University Shenzhen Hospital, Shenzhen, 518036 People’s Republic of China ,grid.440601.70000 0004 1798 0578National & Local Joint Engineering Research Center of Orthopaedic Biomaterials, Peking University Shenzhen Hospital, Shenzhen, 518036 People’s Republic of China
| | - Jian Weng
- grid.440601.70000 0004 1798 0578Department of Bone & Joint Surgery, Peking University Shenzhen Hospital, Shenzhen, 518036 People’s Republic of China ,grid.440601.70000 0004 1798 0578National & Local Joint Engineering Research Center of Orthopaedic Biomaterials, Peking University Shenzhen Hospital, Shenzhen, 518036 People’s Republic of China
| | - Fei Yu
- grid.440601.70000 0004 1798 0578Department of Bone & Joint Surgery, Peking University Shenzhen Hospital, Shenzhen, 518036 People’s Republic of China ,grid.440601.70000 0004 1798 0578National & Local Joint Engineering Research Center of Orthopaedic Biomaterials, Peking University Shenzhen Hospital, Shenzhen, 518036 People’s Republic of China
| | - Liang Gao
- Center for Clinical Medicine, Huatuo Institute of Medical Innovation (HTIMI), Berlin, Germany. .,Sino Euro Orthopaedics Network (SEON), Berlin, Germany.
| | - Changhai Ding
- Clinical Research Centre, Zhujiang Hospital, Southern Medical University, Guangzhou, People's Republic of China.
| | - Hui Zeng
- Department of Bone & Joint Surgery, Peking University Shenzhen Hospital, Shenzhen, 518036, People's Republic of China. .,National & Local Joint Engineering Research Center of Orthopaedic Biomaterials, Peking University Shenzhen Hospital, Shenzhen, 518036, People's Republic of China.
| |
Collapse
|
33
|
Nie Y, Liu H, Wang J, Yang Y, Zhao W, Chen D, Li Y. Systemic evaluation of the relationship between asthma and osteoarthritis: Evidence from a meta-analysis and Mendelian randomization study. Digit Health 2023; 9:20552076231203648. [PMID: 37744746 PMCID: PMC10515552 DOI: 10.1177/20552076231203648] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/11/2023] [Indexed: 09/26/2023] Open
Abstract
Objective Osteoarthritis (OA) and asthma are two common chronic diseases with increasing incidence and prevalence, whereas there has been rare evidence to suggest the relationship between OA and asthma. This study aimed to analyze the causal relationship between OA and asthma. Methods Existing studies of the relationship between asthma and OA published till July 18, 2023, were identified from PubMed and Web of Science databases for meta-analysis. Subsequently, the causal relationship of all and site-specific OA with asthma was explored through a bidirectional two-sample Mendelian randomization (MR) analysis. Results A total of four eligible studies were included in the meta-analysis. In these studies, 80,550 participants were recruited, of whom 13,781 patients had OA. The asthma group had a significantly higher prevalence of OA than the control group (odds ratio (OR) = 2.08; 95% confidence intervals (CI): 1.42-3.03). However, MR analysis did not support a causal relationship between asthma and all OA and site-specific OA: knee and hip OA (OR = 1.03; 95% CI: 0.98-1.09), knee OA (OR = 1.02; 95% CI:0.96-1.08), and hip OA (OR = 1.04; 95% CI: 0.97-1.12). No causal relationship between OA and asthma was found through reverse MR analysis. Conclusions This meta-analysis suggests that patients with asthma are likely to have a greater prevalence of OA. However, the result of MR analysis reveals that asthma does not have a causal relationship to all OA or site-specific OA.
Collapse
Affiliation(s)
- Yaoyao Nie
- Department of Epidemiology and Health Statistics, School of Public Health, Hangzhou Medical College, Hangzhou, China
| | - Houpu Liu
- Department of Epidemiology and Health Statistics, School of Public Health, Hangzhou Medical College, Hangzhou, China
| | - Jing Wang
- Department of Epidemiology and Health Statistics, School of Public Health, Hangzhou Medical College, Hangzhou, China
| | - Ye Yang
- Department of Epidemiology and Health Statistics, School of Public Health, Hangzhou Medical College, Hangzhou, China
| | - Wenxia Zhao
- Department of Epidemiology and Health Statistics, School of Public Health, Hangzhou Medical College, Hangzhou, China
| | - Dingwan Chen
- Zhejiang Provincial Health Research Center, Hangzhou Medical College, Hangzhou, China
| | - Yingjun Li
- Department of Epidemiology and Health Statistics, School of Public Health, Hangzhou Medical College, Hangzhou, China
| |
Collapse
|
34
|
Bruno MC, Cristiano MC, Celia C, d'Avanzo N, Mancuso A, Paolino D, Wolfram J, Fresta M. Injectable Drug Delivery Systems for Osteoarthritis and Rheumatoid Arthritis. ACS NANO 2022; 16:19665-19690. [PMID: 36512378 DOI: 10.1021/acsnano.2c06393] [Citation(s) in RCA: 67] [Impact Index Per Article: 22.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/17/2023]
Abstract
Joint diseases are one of the most common causes of morbidity and disability worldwide. The main diseases that affect joint cartilage are osteoarthritis and rheumatoid arthritis, which require chronic treatment focused on symptomatic relief. Conventional drugs administered through systemic or intra-articular routes have low accumulation and/or retention in articular cartilage, causing dose-limiting toxicities and reduced efficacy. Therefore, there is an urgent need to develop improved strategies for drug delivery, in particular, the use of micro- and nanotechnology-based methods. Encapsulation of therapeutic agents in delivery systems reduces drug efflux from the joint and protects against rapid cellular and enzymatic clearance following intra-articular injection. Consequently, the use of drug delivery systems decreases side effects and increases therapeutic efficacy due to enhanced drug retention in the intra-articular space. Additionally, the frequency of intra-articular administration is reduced, as delivery systems enable sustained drug release. This review summarizes various advanced drug delivery systems, such as nano- and microcarriers, developed for articular cartilage diseases.
Collapse
Affiliation(s)
- Maria Chiara Bruno
- Department of Health Sciences, School of Pharmacy and Nutraceuticals, University "Magna Græcia" of Catanzaro, Campus Universitario "S. Venuta", Building of BioSciences, Viale S. Venuta, Germaneto-Catanzaro, I-88100, Italy
| | - Maria Chiara Cristiano
- Department of Experimental and Clinical Medicine, School of Pharmacy and Nutraceuticals, University "Magna Græcia" of Catanzaro, Campus Universitario "S. Venuta", Building of BioSciences, Viale S. Venuta, Germaneto-Catanzaro, I-88100, Italy
| | - Christian Celia
- Department of Pharmacy, University of Chieti - Pescara "G. d'Annunzio", Via dei Vestini 31, Chieti, I-66100, Italy
- Laboratory of Drug Targets Histopathology, Institute of Cardiology, Lithuanian University of Health Sciences, A. Mickeviciaus g. 9, LT-44307, Kaunas, Lithuania
| | - Nicola d'Avanzo
- Department of Health Sciences, School of Pharmacy and Nutraceuticals, University "Magna Græcia" of Catanzaro, Campus Universitario "S. Venuta", Building of BioSciences, Viale S. Venuta, Germaneto-Catanzaro, I-88100, Italy
- Department of Pharmacy, University of Chieti - Pescara "G. d'Annunzio", Via dei Vestini 31, Chieti, I-66100, Italy
| | - Antonia Mancuso
- Department of Experimental and Clinical Medicine, School of Pharmacy and Nutraceuticals, University "Magna Græcia" of Catanzaro, Campus Universitario "S. Venuta", Building of BioSciences, Viale S. Venuta, Germaneto-Catanzaro, I-88100, Italy
| | - Donatella Paolino
- Department of Experimental and Clinical Medicine, School of Pharmacy and Nutraceuticals, University "Magna Græcia" of Catanzaro, Campus Universitario "S. Venuta", Building of BioSciences, Viale S. Venuta, Germaneto-Catanzaro, I-88100, Italy
| | - Joy Wolfram
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, QLD 4072, Australia
- School of Chemical Engineering, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Massimo Fresta
- Department of Health Sciences, School of Pharmacy and Nutraceuticals, University "Magna Græcia" of Catanzaro, Campus Universitario "S. Venuta", Building of BioSciences, Viale S. Venuta, Germaneto-Catanzaro, I-88100, Italy
| |
Collapse
|
35
|
Liang X, Li SR, Zhang XX, He SH, Li SS, Li TF. Teriparatide prevented synovial inflammation and cartilage destruction in mice with DMM. Connect Tissue Res 2022; 64:274-284. [PMID: 36537662 DOI: 10.1080/03008207.2022.2157723] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
AIM Emerging data have demonstrated that low-grade inflammation in osteoarthritis, a long-held degenerative disease. The inflamed synovium produces various cytokines that induce cartilage destruction and joint pain. A previous study showed that teriparatide, an FDA approved anti-osteoporotic drug, may enhance cartilage repair. Our study focuses on its role in OA synovitis. MATERIALS AND METHODS Primary mouse articular chondrocytes were used to determine the most potent cytokines involved in OA inflammation and cartilage destruction. A destabilization of the medial meniscus mouse model was established to investigate the effect of teriparatide in OA, particularly, on synovial inflammation and cartilage degradation. RESULTS In vitro experiments showed that TNF-α was the most potent inducer of cartilage matrix-degrading enzymes, and that teriparatide antagonized the TNF-α of effect. Consistently, articular cartilage samples from TNF-α transgenic mice contained more MMP-13 positive chondrocytes than those from wild type mice. In addition, more type II collagen was cleaved in human OA cartilage than in normal cartilage samples. CONCLUSIONS Teriparatide can prevent synovitis and cartilage degradation by suppressing TNF-α mediated MMP-13 overexpression. Together with its chondroregenerative capability, teriparatide may be the first effective disease modifying osteoarthritis drug.
Collapse
Affiliation(s)
| | | | - Xin-Xin Zhang
- Department of Rheumatology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, People's Republic of China
| | - Shi-Hao He
- Department of Rheumatology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, People's Republic of China
| | - Shan-Shan Li
- Department of Rheumatology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, People's Republic of China
| | - Tian-Fang Li
- Department of Rheumatology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, People's Republic of China
| |
Collapse
|
36
|
Zeng L, Yang T, Yang K, Yu G, Li J, Xiang W, Chen H. Efficacy and Safety of Curcumin and Curcuma longa Extract in the Treatment of Arthritis: A Systematic Review and Meta-Analysis of Randomized Controlled Trial. Front Immunol 2022; 13:891822. [PMID: 35935936 PMCID: PMC9353077 DOI: 10.3389/fimmu.2022.891822] [Citation(s) in RCA: 43] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Accepted: 04/22/2022] [Indexed: 12/20/2022] Open
Abstract
BackgroundModern pharmacological research found that the chemical components of Curcuma longa L. are mainly curcumin and turmeric volatile oil. Several recent randomized controlled trials (RCT) have shown that curcumin improves symptoms and inflammation in patients with arthritis.MethodsPubmed, Cochran Library, CNKI, and other databases were searched to collect the randomized controlled trials (RCTs). Then, the risk of bias of RCTs were assessed and data of RCTs were extracted. Finally, RevMan 5.3 was utilized for meta-analysis.ResultsTwenty-nine (29) RCTs involving 2396 participants and 5 types of arthritis were included. The arthritis included Ankylosing Spondylitis (AS), Rheumatoid Arthritis (RA), Osteoarthritis (OA), Juvenile idiopathic arthritis (JIA) and gout/hyperuricemia. Curcumin and Curcuma longa Extract were administered in doses ranging from 120 mg to 1500 mg for a duration of 4-36 weeks. In general, Curcumin and Curcuma longa Extract showed safety in all studies and improved the severity of inflammation and pain levels in these arthritis patients. However, more RCTs are needed in the future to elucidate the effect of Curcumin and Curcuma longa Extract supplementation in patients with arthritis, including RA, OA, AS and JIA.ConclusionCurcumin and Curcuma longa Extract may improve symptoms and inflammation levels in people with arthritis. However, due to the low quality and small quantity of RCTs, the conclusions need to be interpreted carefully.
Collapse
Affiliation(s)
- Liuting Zeng
- Department of Rheumatology and Clinical Immunology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, National Clinical Research Center for Dermatologic and Immunologic Diseases (NCRC-DID), Key Laboratory of Rheumatology and Clinical Immunology, Ministry of Education, Beijing, China
- *Correspondence: Hua Chen, ; Liuting Zeng, ; Tiejun Yang, ; Kailin Yang,
| | - Tiejun Yang
- Department of Orthopedics, People’s Hospital of Ningxiang City, Ningxiang City, China
- *Correspondence: Hua Chen, ; Liuting Zeng, ; Tiejun Yang, ; Kailin Yang,
| | - Kailin Yang
- Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio-Cerebral Diseases, Hunan University of Chinese Medicine, Changsha City, China
- *Correspondence: Hua Chen, ; Liuting Zeng, ; Tiejun Yang, ; Kailin Yang,
| | - Ganpeng Yu
- Department of Orthopedics, People’s Hospital of Ningxiang City, Ningxiang City, China
| | - Jun Li
- Department of Orthopedics, People’s Hospital of Ningxiang City, Ningxiang City, China
| | - Wang Xiang
- Department of Rheumatology, The First People's Hospital Changde City, Changde City, China
| | - Hua Chen
- Department of Rheumatology and Clinical Immunology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, National Clinical Research Center for Dermatologic and Immunologic Diseases (NCRC-DID), Key Laboratory of Rheumatology and Clinical Immunology, Ministry of Education, Beijing, China
- *Correspondence: Hua Chen, ; Liuting Zeng, ; Tiejun Yang, ; Kailin Yang,
| |
Collapse
|
37
|
The effects of alendronate on the suppression of bone resorption and the promotion of cartilage formation in the human mosaicplasty donor site: A randomized, double-blind, placebo-controlled prospective study. J Orthop Sci 2022; 27:835-843. [PMID: 34167868 DOI: 10.1016/j.jos.2021.05.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/09/2021] [Revised: 05/03/2021] [Accepted: 05/19/2021] [Indexed: 11/22/2022]
Abstract
BACKGROUND We previously reported that early alendronate administration accelerated bone formation and improved the quality of repaired cartilage in the donor site in rabbits. To investigate whether alendronate administration has effects in humans similar to those observed in rabbits. METHODS The study cohort included 35 patients over the age of 12-years old who underwent mosaicplasty without osteoporotic therapy from March 2011 to October 2012. The donor sites were medial or lateral in the patellofemoral joint. Placebo (P) or Bonalon containing 35 mg of alendronate (A) was administered orally every week for 8 weeks. The cohort comprised 15 male and 20 female, including 14 right and 21 left knees. The mean age at the time of surgery was 57.1 years. Bone formation was examined using computer tomography and lateral knee radiography, and cartilage formation was examined using magnetic resonance imaging (MRI), second-look assessment, and intraoperative acoustic evaluation. The clinical outcomes were assessed using the Japanese Orthopaedic Association knee score and visual analog scale (VAS). Bone and cartilage formation in the donor site and clinical outcomes were assessed at 3, 6, and 12 months after mosaicplasty. RESULTS The ratio of TRAP-5b in group A was significantly smaller than that in group P at 2 and 8 weeks after mosaicplasty. The extent of bone formation in the donor sites in group A was significantly greater than that in group P at 3 and 6 months after mosaicplasty. Cartilage formation did not differ significantly between the two groups as determined by MRI, macroscopic assessment, and intraoperative acoustic evaluation. Clinical outcomes did not differ significantly between the two groups, and no negative clinical outcomes were observed. CONCLUSION Early alendronate administration accelerated bone formation but not cartilage formation in the mosaicplasty donor site in humans.
Collapse
|
38
|
Zheng CY, Chu XY, Gao CY, Hu HY, He X, Chen X, Yang K, Zhang DL. TAT&RGD Peptide-Modified Naringin-Loaded Lipid Nanoparticles Promote the Osteogenic Differentiation of Human Dental Pulp Stem Cells. Int J Nanomedicine 2022; 17:3269-3286. [PMID: 35924260 PMCID: PMC9342892 DOI: 10.2147/ijn.s371715] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Accepted: 07/15/2022] [Indexed: 11/25/2022] Open
Abstract
Background Naringin is a naturally occurring flavanone that promotes osteogenesis. Owing to the high lipophilicity, poor in vivo bioavailability, and extensive metabolic alteration upon administration, the clinical efficacy of naringin is understudied. Additionally, information on the molecular mechanism by which it promotes osteogenesis is limited. Methods In this study, we prepared TAT & RGD peptide-modified naringin-loaded nanoparticles (TAT-RGD-NAR-NPs), evaluated their potency on the osteogenic differentiation of human dental pulp stem cells (hDPSCs), and studied its mechanism of action through metabolomic analysis. Results The particle size and zeta potential of TAT-RGD-NAR-NPs were 160.70±2.05 mm and –20.77±0.47mV, respectively. The result of cell uptake assay showed that TAT-RGD-NAR-NPs could effectively enter hDPSCs. TAT-RGD-NAR-NPs had a more significant effect on cell proliferation and osteogenic differentiation promotion. Furthermore, in metabolomic analysis, naringin particles showed a strong influence on the glycerophospholipid metabolism pathway of hDPSCs. Specifically, it upregulated the expression of PLA2G3 and PLA2G1B (two isozymes of phospholipase A2, PLA2), increased the biosynthesis of lysophosphatidic acid (LPA). Conclusion These results suggested that TAT-RGD-NPs might be used for transporting naringin to hDPSCs for modulating stem cell osteogenic differentiation. The metabolomic analysis was used for the first time to elucidate the mechanism by which naringin promotes hDPSCs osteogenesis by upregulating PLA2G3 and PLA2G1B.
Collapse
Affiliation(s)
- Chun-Yan Zheng
- Department of Orthodontics, Beijing Stomatological Hospital, Capital Medical University School of Stomatology, Capital Medical University, Beijing, People’s Republic of China
| | - Xiao-Yang Chu
- Department of Stomatology, Fifth Medical Center of Chinese PLA General Hospital, Beijing, People’s Republic of China
| | - Chun-Yan Gao
- Department of Orthodontics, Beijing Stomatological Hospital, Capital Medical University School of Stomatology, Capital Medical University, Beijing, People’s Republic of China
| | - Hua-Ying Hu
- Birth Defects Prevention and Control Technology Research Center, Medical Innovation Research Division of Chinese PLA General Hospital, Beijing, People’s Republic of China
| | - Xin He
- Department of Orthodontics, Beijing Stomatological Hospital, Capital Medical University School of Stomatology, Capital Medical University, Beijing, People’s Republic of China
| | - Xu Chen
- Department of Orthodontics, Beijing Stomatological Hospital, Capital Medical University School of Stomatology, Capital Medical University, Beijing, People’s Republic of China
| | - Kai Yang
- Prenatal Diagnosis Center, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing, People’s Republic of China
| | - Dong-Liang Zhang
- Department of Orthodontics, Beijing Stomatological Hospital, Capital Medical University School of Stomatology, Capital Medical University, Beijing, People’s Republic of China
- Correspondence: Dong-Liang Zhang, Department of Orthodontics, Beijing Stomatological Hospital, Capital Medical University School of Stomatology, Capital Medical University, 11 Xilahutong Road, Beijing, 100040, People’s Republic of China, Email
| |
Collapse
|
39
|
Voina-Tonea A, Labunet A, Objelean A, Onisor F, Bran S, Mester A, Piciu A, Sava S. A Systematic Analysis of the Available Human Clinical Studies of Dental Implant Failure in Patients with Inflammatory Bowel Disease. Medicina (B Aires) 2022; 58:medicina58030343. [PMID: 35334519 PMCID: PMC8954657 DOI: 10.3390/medicina58030343] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Revised: 02/17/2022] [Accepted: 02/22/2022] [Indexed: 11/16/2022] Open
Abstract
Background and objectives: The aim was to evaluate the current literature on the influence of inflammatory bowel disease (ulcerative colitis/Crohn’s disease) in dental implant osseointegration in human clinical studies. Materials and methods: This review was conducted under the Preferred Reporting Items for Systematic Review and Meta-Analysis guidelines. PubMed, Scopus, and Web of Science databases were electronic screened to find relevant articles published until October 2021. The inclusion criteria consisted of human clinical studies that reported the use of dental implant in patients diagnosed with inflammatory bowel disease. Risk of bias was assessed according to The Strengthening the Reporting of Observational studies in Epidemiology criteria. Results: A total of 786 studies were identified from databases. Of these, six studies were included in the review and reported the use of implants in patients with Crohn’s disease. No articles were available for ulcerative colitis. Included articles indicated that Crohn’s disease may determine early and late implant failure. Besides Crohn’s disease, several patients presented associated risk factors and systemic disease that determined implant failure. Conclusions: The presence of clinical studies on the influence of IBD in implant therapy is low. When recommending an implant therapy to IBD patients, the multidisciplinary team should be aware of side effects and a close collaboration between members of this team is necessary. More data are needed to sustain the effect of IBD on implant therapy.
Collapse
Affiliation(s)
- Andrada Voina-Tonea
- Department of Dental Materials, Iuliu Hațieganu University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania; (A.V.-T.); (A.L.); (A.O.); (S.S.)
| | - Anca Labunet
- Department of Dental Materials, Iuliu Hațieganu University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania; (A.V.-T.); (A.L.); (A.O.); (S.S.)
| | - Adriana Objelean
- Department of Dental Materials, Iuliu Hațieganu University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania; (A.V.-T.); (A.L.); (A.O.); (S.S.)
| | - Florin Onisor
- Department of Maxillofacial Surgery and Implantology, Iuliu Hațieganu University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania;
- Correspondence: (F.O.); (A.M.)
| | - Simion Bran
- Department of Maxillofacial Surgery and Implantology, Iuliu Hațieganu University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania;
| | - Alexandru Mester
- Department of Oral Health, Iuliu Hațieganu University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania
- Correspondence: (F.O.); (A.M.)
| | - Andra Piciu
- Department of Medical Oncology, Iuliu Hațieganu University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania;
| | - Sorina Sava
- Department of Dental Materials, Iuliu Hațieganu University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania; (A.V.-T.); (A.L.); (A.O.); (S.S.)
| |
Collapse
|
40
|
Xu F, Zhao LJ, Liao T, Li ZC, Wang LL, Lin PY, Jiang R, Wei QJ. Ononin ameliorates inflammation and cartilage degradation in rat chondrocytes with IL-1β-induced osteoarthritis by downregulating the MAPK and NF-κB pathways. BMC Complement Med Ther 2022; 22:25. [PMID: 35086536 PMCID: PMC8793192 DOI: 10.1186/s12906-022-03504-5] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Accepted: 01/05/2022] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND Osteoarthritis (OA) treatment aims to improve inflammation and delay cartilage degeneration. However, there is no effective strategy presently available. Ononin, a representative isoflavone glycoside component extracted from natural Chinese herbs, exerts anti-inflammatory and proliferative effects. However, the therapeutic effect of ononin on chondrocyte inflammation remains unclear. METHODS In this study, we explored the therapeutic effect and potential mechanism of ononin in OA by establishing an interleukin-1 beta (IL-1β)-induced chondrocyte inflammation model. RESULTS Our results verified that ononin alleviated the IL-1β-induced decrease in chondrocyte viability, attenuated the overexpression of the inflammatory factors tumour necrosis factor α (TNF-α) and interleukin 6 (IL-6), and simultaneously inhibited the expression of cartilage extracellular matrix (ECM)-degrading enzymes such as matrix metalloproteinase-13 (MMP-13). Furthermore, the decomposition of Collagen II protein could be alleviated in the OA model by ononin. Finally, ononin improved chondrocyte inflammation by downregulating the mitogen-activated protein kinase (MAPK) and nuclear factor kappa-B (NF-κB) signalling pathways. CONCLUSION Our findings suggested that ononin could inhibit the IL-1β-induced proinflammatory response and ECM degradation in chondrocytes by interfering with the abnormal activation of the MAPK and NF-κB pathways, indicating its protective effect against OA.
Collapse
Affiliation(s)
- Fang Xu
- Department of Orthopaedics Trauma and Hand Surgery, The First Affiliated Hospital of Guangxi Medical University, Shuangyong Road No. 6, Nanning, 530021, China
- Guangxi Colleges and Universities Key Laboratory of Prevention and Control of Highly Prevalent Diseases, Guangxi Medical University, Shuangyong Road No. 22, Nanning, 530021, China
| | - Liang-Jun Zhao
- Department of Bone and Joint Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, China
| | - Ting Liao
- Department of Endocrinology, Liuzhou Municipal Liutie Central Hospital, Feie Road No. 22, Liuzhou, 545007, China
| | - Zhao-Cong Li
- Guangxi Colleges and Universities Key Laboratory of Prevention and Control of Highly Prevalent Diseases, Guangxi Medical University, Shuangyong Road No. 22, Nanning, 530021, China
- Department of Toxicology, School of Public Health, Guangxi Medical University, Shuangyong Road No. 22, Nanning, 530021, Guangxi, China
| | - Lei-Lei Wang
- Guangxi Colleges and Universities Key Laboratory of Prevention and Control of Highly Prevalent Diseases, Guangxi Medical University, Shuangyong Road No. 22, Nanning, 530021, China
- Department of Toxicology, School of Public Health, Guangxi Medical University, Shuangyong Road No. 22, Nanning, 530021, Guangxi, China
| | - Pan-Yu Lin
- Department of Orthopaedics Trauma and Hand Surgery, The First Affiliated Hospital of Guangxi Medical University, Shuangyong Road No. 6, Nanning, 530021, China
| | - Rui Jiang
- Department of Orthopaedics Trauma and Hand Surgery, The First Affiliated Hospital of Guangxi Medical University, Shuangyong Road No. 6, Nanning, 530021, China
| | - Qing-Jun Wei
- Department of Orthopaedics Trauma and Hand Surgery, The First Affiliated Hospital of Guangxi Medical University, Shuangyong Road No. 6, Nanning, 530021, China.
| |
Collapse
|
41
|
Hoffman JM, Tolbert MK, Promislow DEL. Demographic factors associated with joint supplement use in dogs from the Dog Aging Project. Front Vet Sci 2022; 9:906521. [PMID: 35958315 PMCID: PMC9361857 DOI: 10.3389/fvets.2022.906521] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Accepted: 07/04/2022] [Indexed: 01/29/2023] Open
Abstract
Osteoarthritis (OA) is one of the most prevalent age-related chronic conditions that afflict companion dogs, and multiple joint supplements are available to prevent or treat OA, though the efficacy of these treatments is controversial. While the demographic factors that are associated with OA diagnosis are well established, the factors that are associated with joint supplement use are not as well studied. Using data collected from the Dog Aging Project, we analyzed owner survey responses regarding joint supplement administration and OA diagnosis for 26,951 adult dogs. In this cross-sectional analysis, logistic regression models and odds-ratios (OR) were employed to determine demographic factors of dogs and their owners that were associated with joint supplement administration. Forty percent of adult dogs in our population were given some type of joint supplement. Perhaps not surprisingly, dogs of older age, larger size, and those that were ever overweight were more likely to receive a joint supplement. Younger owner age, urban living, owner education, and feeding commercial dry food were associated with a reduced likelihood of administration of joint supplements to dogs. Interestingly, mixed breed dogs were also less likely to be administered a joint supplement (OR: 0.73). Dogs with a clinical diagnosis of OA were more likely to receive a joint supplement than those without a reported OA diagnosis (OR: 3.82). Neutered dogs were more likely to have a diagnosis of OA, even after controlling for other demographic factors, yet their prevalence of joint supplement administration was the same as intact dogs. Overall, joint supplement use appears to be high in our large population of dogs in the United States. Prospective studies are needed to determine if joint supplements are more commonly administered as a preventative for OA or after an OA clinical diagnosis.
Collapse
Affiliation(s)
- Jessica M Hoffman
- Department of Biology, University of Alabama at Birmingham, Birmingham, AL, United States
| | - M Katherine Tolbert
- Department of Small Animal Clinical Sciences, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, TX, United States
| | - Daniel E L Promislow
- Department of Laboratory Medicine and Pathology, University of Washington School of Medicine, Seattle, WA, United States.,Department of Biology, University of Washington, Seattle, WA, United States
| | | |
Collapse
|
42
|
Evaluation of Human Bone Marrow Mesenchymal Stromal Cell (MSC) Functions on a Biomorphic Rattan-Wood-Derived Scaffold: A Comparison between Cultured and Uncultured MSCs. Bioengineering (Basel) 2021; 9:bioengineering9010001. [PMID: 35049710 PMCID: PMC8773042 DOI: 10.3390/bioengineering9010001] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Revised: 11/15/2021] [Accepted: 12/18/2021] [Indexed: 11/16/2022] Open
Abstract
The reconstruction of large bone defects requires the use of biocompatible osteoconductive scaffolds. These scaffolds are often loaded with the patient’s own bone marrow (BM) cells to facilitate osteoinductivity and biological potency. Scaffolds that are naturally sourced and fabricated through biomorphic transitions of rattan wood (B-HA scaffolds) offer a unique advantage of higher mechanical strength and bioactivity. In this study, we investigated the ability of a biomorphic B-HA scaffold (B-HA) to support the attachment, survival and gene expression profile of human uncultured BM-derived mesenchymal stromal cells (BMSCs, n = 6) and culture expanded MSCs (cMSCs, n = 7) in comparison to a sintered, porous HA scaffold (S-HA). B-HA scaffolds supported BMSC attachment (average 98%) and their survival up to 4 weeks in culture. Flow cytometry confirmed the phenotype of cMSCs on the scaffolds. Gene expression indicated clear segregation between cMSCs and BMSCs with MSC osteogenesis- and adipogenesis-related genes including RUNX2, PPARγ, ALP and FABP4 being higher expressed in BMSCs. These data indicated a unique transcriptional signature of BMSCs that was distinct from that of cMSCs regardless of the type of scaffold or time in culture. There was no statistical difference in the expression of osteogenic genes in BMSCs or cMSCs in B-HA compared to S-HA. VEGF release from cMSCs co-cultured with human endothelial cells (n = 4) on B-HA scaffolds suggested significantly higher supernatant concentration with endothelial cells on day 14. This indicated a potential mechanism for providing vasculature to the repair area when such scaffolds are used for treating large bone defects.
Collapse
|
43
|
Deng J, Zong Z, Su Z, Chen H, Huang J, Niu Y, Zhong H, Wei B. Recent Advances in Pharmacological Intervention of Osteoarthritis: A Biological Aspect. Front Pharmacol 2021; 12:772678. [PMID: 34887766 PMCID: PMC8649959 DOI: 10.3389/fphar.2021.772678] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Accepted: 10/04/2021] [Indexed: 12/27/2022] Open
Abstract
Osteoarthritis (OA) is a degenerative joint disease in the musculoskeletal system with a relatively high incidence and disability rate in the elderly. It is characterized by the degradation of articular cartilage, inflammation of the synovial membrane, and abnormal structure in the periarticular and subchondral bones. Although progress has been made in uncovering the molecular mechanism, the etiology of OA is still complicated and unclear. Nevertheless, there is no treatment method that can effectively prevent or reverse the deterioration of cartilage and bone structure. In recent years, in the field of pharmacology, research focus has shifted to disease prevention and early treatment rather than disease modification in OA. Biologic agents become more and more attractive as their direct or indirect intervention effects on the initiation or development of OA. In this review, we will discuss a wide spectrum of biologic agents ranging from DNA, noncoding RNA, exosome, platelet-rich plasma (PRP), to protein. We searched for key words such as OA, DNA, gene, RNA, exosome, PRP, protein, and so on. From the pharmacological aspect, stem cell therapy is a very special technique, which is not included in this review. The literatures ranging from January 2016 to August 2021 were included and summarized. In this review, we aim to help readers have a complete and precise understanding of the current pharmacological research progress in the intervention of OA from the biological aspect and provide an indication for the future translational studies.
Collapse
Affiliation(s)
- Jinxia Deng
- Affiliated Hospital of Guangdong Medical University, Guangdong Medical University, Zhanjiang, China
| | - Zhixian Zong
- Affiliated Hospital of Guangdong Medical University, Guangdong Medical University, Zhanjiang, China
| | - Zhanpeng Su
- Affiliated Hospital of Guangdong Medical University, Guangdong Medical University, Zhanjiang, China
| | - Haicong Chen
- Affiliated Hospital of Guangdong Medical University, Guangdong Medical University, Zhanjiang, China
| | - Jianping Huang
- College of Dentistry, Yonsei University, Seoul, South Korea.,Department of Stomatology, Guangdong Medical University, Zhanjiang, China
| | - Yanru Niu
- Affiliated Hospital of Guangdong Medical University, Guangdong Medical University, Zhanjiang, China
| | - Huan Zhong
- Affiliated Hospital of Guangdong Medical University, Guangdong Medical University, Zhanjiang, China
| | - Bo Wei
- Affiliated Hospital of Guangdong Medical University, Guangdong Medical University, Zhanjiang, China
| |
Collapse
|
44
|
Oppedisano F, Bulotta RM, Maiuolo J, Gliozzi M, Musolino V, Carresi C, Ilari S, Serra M, Muscoli C, Gratteri S, Palma E, Mollace V. The Role of Nutraceuticals in Osteoarthritis Prevention and Treatment: Focus on n-3 PUFAs. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:4878562. [PMID: 34925695 PMCID: PMC8683171 DOI: 10.1155/2021/4878562] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Revised: 10/30/2021] [Accepted: 11/18/2021] [Indexed: 12/14/2022]
Abstract
Osteoarthritis (OA) is a disease caused by joint degeneration with massive cartilage loss, and obesity is among the risk factors for its onset, though the pathophysiological mechanisms underlying the disease and better therapeutic approach still remain to be assessed. In recent years, several nutraceutical interventions have been investigated in order to define better solutions for preventing and treating OA. Among them, polyunsaturated fatty acids (n-3 PUFAs) appear to represent potential candidates in counteracting OA and its consequences, due to their anti-inflammatory, antioxidant, and chondroinductive effects. PUFAs have been found to counteract the onset and progression of OA by reducing bone and cartilage destruction, inhibiting proinflammatory cytokine release, reactive oxygen species (ROS) generation, and the NF-κB pathway's activation. Moreover, a diet rich in n-3 PUFAs and their derivatives (maresins and resolvins) demonstrates beneficial effects on associated pain reduction. Finally, it has been shown that together with the anti-inflammatory and antioxidant properties of eicosapentaenoic (EPA) and docosahexaenoic (DHA) acids, their antiapoptotic and antiangiogenic effects contribute in reducing OA development. The present review is aimed at assessing evidence suggesting the potential benefit of nutraceutical supplementation with PUFAs in OA management according to their efficacy in targeting relevant pathophysiological mechanisms responsible for inflammation and joint destruction processes, and this may represent a novel and potentially useful approach in OA prevention and treatment. For that purpose, a PubMed literature survey was conducted with a focus on some in vitro and in vivo studies and clinical trials from 2015 to 2020.
Collapse
Affiliation(s)
- Francesca Oppedisano
- Institute of Research for Food Safety & Health IRC-FSH, University Magna Graecia, 88100 Catanzaro, Italy
| | - Rosa Maria Bulotta
- Nutramed S.c.a.r.l., Complesso Ninì Barbieri, Roccelletta di Borgia, 88021 Catanzaro, Italy
| | - Jessica Maiuolo
- Institute of Research for Food Safety & Health IRC-FSH, University Magna Graecia, 88100 Catanzaro, Italy
| | - Micaela Gliozzi
- Institute of Research for Food Safety & Health IRC-FSH, University Magna Graecia, 88100 Catanzaro, Italy
| | - Vincenzo Musolino
- Institute of Research for Food Safety & Health IRC-FSH, University Magna Graecia, 88100 Catanzaro, Italy
| | - Cristina Carresi
- Institute of Research for Food Safety & Health IRC-FSH, University Magna Graecia, 88100 Catanzaro, Italy
| | - Sara Ilari
- Institute of Research for Food Safety & Health IRC-FSH, University Magna Graecia, 88100 Catanzaro, Italy
| | - Maria Serra
- Institute of Research for Food Safety & Health IRC-FSH, University Magna Graecia, 88100 Catanzaro, Italy
| | - Carolina Muscoli
- Institute of Research for Food Safety & Health IRC-FSH, University Magna Graecia, 88100 Catanzaro, Italy
| | - Santo Gratteri
- Institute of Research for Food Safety & Health IRC-FSH, University Magna Graecia, 88100 Catanzaro, Italy
| | - Ernesto Palma
- Institute of Research for Food Safety & Health IRC-FSH, University Magna Graecia, 88100 Catanzaro, Italy
- Nutramed S.c.a.r.l., Complesso Ninì Barbieri, Roccelletta di Borgia, 88021 Catanzaro, Italy
| | - Vincenzo Mollace
- Institute of Research for Food Safety & Health IRC-FSH, University Magna Graecia, 88100 Catanzaro, Italy
- Nutramed S.c.a.r.l., Complesso Ninì Barbieri, Roccelletta di Borgia, 88021 Catanzaro, Italy
- IRCCS San Raffaele Pisana, Via di Valcannuta, Rome, Italy
| |
Collapse
|
45
|
Liu Q, Zhai L, Han M, Shi D, Sun Z, Peng S, Wang M, Zhang C, Gao J, Yan W, Jiang Q, Chen D, Xu Q, Tan M, Sun Y. SHP2 inhibition attenuates osteoarthritis by maintaining homeostasis of cartilage metabolism via the DOK1/UPP1/uridine cascade. Arthritis Rheumatol 2021; 74:462-474. [PMID: 34569725 DOI: 10.1002/art.41988] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2021] [Revised: 09/10/2021] [Accepted: 09/23/2021] [Indexed: 11/08/2022]
Abstract
OBJECTIVE Protein tyrosine kinases (PTKs) regulate osteoarthritis (OA) progression by activating a series of signal transduction pathways. However, the roles of protein tyrosine phosphatases (PTPs) in OA remain obscure. METHODS The expression of 107 PTP genes in human OA cartilage was analyzed based on a single-cell sequencing dataset. The enzyme activity of the PTP SHP2 was detected in primary chondrocytes after interleukin (IL)-1β treatment and in human OA cartilage. Destabilized medial meniscus (DMM) model and IL-1β-stimulated primary mouse chondrocytes were treated with an SHP2 inhibitor and celecoxib (a clinical drug for the treatment of OA). The function of SHP2 in OA pathogenesis was further verified in Aggrecan-CreERT ; SHP2 flox/flox mice. The downstream protein expression profile and dephosphorylated substrate of SHP2 were examined by tandem mass tag (TMT) labeling-based global proteomic and stable isotope labeling using amino acids in cell culture (SILAC)-labeled tyrosine phosphoproteomic analysis, respectively. RESULTS SHP2 enzyme activity significantly increased in human OA samples with serious articular cartilage injury and in IL-1β-stimulated chondrocytes. Pharmacological inhibition or genetic deletion of SHP2 ameliorated OA progression. SHP2 inhibitors dramatically reduced the expression of cartilage degradation-related genes and simultaneously promoted the expression of cartilage synthesis-related genes. Mechanistically, SHP2 inhibition suppressed the dephosphorylation of DOK1 and subsequently reduced the expression of uridine phosphorylase 1 and increased uridine level, thereby contributing to the homeostasis of cartilage metabolism. CONCLUSIONS SHP2 is a novel accelerator of the imbalance in the cartilage homeostasis. Specific inhibition of SHP2 may ameliorate OA by maintaining the anabolic and catabolic balance.
Collapse
Affiliation(s)
- Qianqian Liu
- State Key Laboratory of Pharmaceutical Biotechnology, Chemistry and Biomedicine Innovation Center (ChemBIC), School of Life Sciences, Nanjing University, 163 Xianlin Avenue, 210023, Jiangsu, China
| | - Linhui Zhai
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zuchongzhi Road, Shanghai, 201203, China
| | - Mingrui Han
- State Key Laboratory of Pharmaceutical Biotechnology, Chemistry and Biomedicine Innovation Center (ChemBIC), School of Life Sciences, Nanjing University, 163 Xianlin Avenue, 210023, Jiangsu, China
| | - Dongquan Shi
- Department of Sports Medicine and Adult Reconstructive Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, 321 Zhongshan Road, 210008, Jiangsu, China
| | - Ziying Sun
- Department of Sports Medicine and Adult Reconstructive Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, 321 Zhongshan Road, 210008, Jiangsu, China
| | - Shuang Peng
- Institute of Pharmacology, University of Bern, Bern, Switzerland
| | - Meijing Wang
- State Key Laboratory of Pharmaceutical Biotechnology, Chemistry and Biomedicine Innovation Center (ChemBIC), School of Life Sciences, Nanjing University, 163 Xianlin Avenue, 210023, Jiangsu, China
| | - Chenyang Zhang
- State Key Laboratory of Pharmaceutical Biotechnology, Chemistry and Biomedicine Innovation Center (ChemBIC), School of Life Sciences, Nanjing University, 163 Xianlin Avenue, 210023, Jiangsu, China
| | - Jian Gao
- State Key Laboratory of Pharmaceutical Biotechnology, Chemistry and Biomedicine Innovation Center (ChemBIC), School of Life Sciences, Nanjing University, 163 Xianlin Avenue, 210023, Jiangsu, China
| | - Wenjin Yan
- Department of Sports Medicine and Adult Reconstructive Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, 321 Zhongshan Road, 210008, Jiangsu, China
| | - Qing Jiang
- Department of Sports Medicine and Adult Reconstructive Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, 321 Zhongshan Road, 210008, Jiangsu, China
| | - Dijun Chen
- State Key Laboratory of Pharmaceutical Biotechnology, Chemistry and Biomedicine Innovation Center (ChemBIC), School of Life Sciences, Nanjing University, 163 Xianlin Avenue, 210023, Jiangsu, China
| | - Qiang Xu
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, 221004, Jiangsu, China
| | - Minjia Tan
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zuchongzhi Road, Shanghai, 201203, China
| | - Yang Sun
- State Key Laboratory of Pharmaceutical Biotechnology, Chemistry and Biomedicine Innovation Center (ChemBIC), School of Life Sciences, Nanjing University, 163 Xianlin Avenue, 210023, Jiangsu, China.,Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, 221004, Jiangsu, China
| |
Collapse
|
46
|
Stem Cells in Autologous Microfragmented Adipose Tissue: Current Perspectives in Osteoarthritis Disease. Int J Mol Sci 2021; 22:ijms221910197. [PMID: 34638538 PMCID: PMC8508703 DOI: 10.3390/ijms221910197] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2021] [Revised: 09/09/2021] [Accepted: 09/14/2021] [Indexed: 12/13/2022] Open
Abstract
Osteoarthritis (OA) is a chronic debilitating disorder causing pain and gradual degeneration of weight-bearing joints with detrimental effects on cartilage volume as well as cartilage damage, generating inflammation in the joint structure. The etiology of OA is multifactorial. Currently, therapies are mainly addressing the physical and occupational aspects of osteoarthritis using pharmacologic pain treatment and/or surgery to manage the symptomatology of the disease with no specific regard to disease progression or prevention. Herein, we highlight alternative therapeutics for OA specifically considering innovative and encouraging translational methods with the use of adipose mesenchymal stem cells.
Collapse
|
47
|
Vijaykumar A, Mina M. Lithium Chloride Exerts Differential Effects on Dentinogenesis and Osteogenesis in Primary Pulp Cultures. FRONTIERS IN DENTAL MEDICINE 2021. [DOI: 10.3389/fdmed.2021.649500] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Wnt/β-catenin signaling is known to play essential roles in odontoblast differentiation and reparative dentin formation. Various Wnt activators including LiCl have been increasingly studied for their effectiveness to induce repair of the dentin-pulp complex. LiCl is a simple salt thought to activate Wnt/β-catenin signaling by inhibiting GSK3β. Previous in vitro and in vivo studies showed that LiCl increased odontoblast differentiation and enhanced reparative dentin formation. However, the underlying molecular and cellular mechanisms by which LiCl regulates odontoblast and osteoblast differentiation during reparative dentinogenesis are not well-understood. Our in vitro studies show that exposure of early dental pulp progenitors to LiCl increased the survival and the pool of αSMA+ progenitors, leading to enhanced odontoblast and osteoblast differentiation. The positive effects of LiCl in the differentiation of osteoblasts and odontoblasts from αSMA+ progenitors are mediated by Wnt/β-catenin signaling. Our results also showed that continuous and late exposure of dental pulp cells to LiCl increased the expression of odontoblast markers through Wnt/β-catenin signaling, and the number of odontoblasts expressing DMP1-Cherry and DSPP-Cerulean transgenes. However, unlike the early treatment, both continuous and late treatments decreased the expression of Bsp and the expression of BSP-GFPtpz transgene. These observations suggest that prolonged treatment with LiCl in more mature cells of the dental pulp has an inhibitory effect on osteoblast differentiation. The inhibitory effects of LiCl on osteogenesis and Bsp were not mediated through Wnt/β-catenin signaling. These observations suggest that the effects of LiCl, and GSK3β antagonists on reparative dentinogenesis involve multiple pathways and are not specific to Wnt/β-catenin signaling.
Collapse
|
48
|
Glucosamine and Chondroitin Sulfate: Is There Any Scientific Evidence for Their Effectiveness as Disease-Modifying Drugs in Knee Osteoarthritis Preclinical Studies?-A Systematic Review from 2000 to 2021. Animals (Basel) 2021; 11:ani11061608. [PMID: 34072407 PMCID: PMC8228516 DOI: 10.3390/ani11061608] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2021] [Revised: 05/19/2021] [Accepted: 05/27/2021] [Indexed: 12/19/2022] Open
Abstract
Simple Summary Osteoarthritis is the most common progressive joint disease diagnosed in companion animals and its management continues to be a significant challenge. Nutraceuticals have been widely investigated over the years in the treatment of osteoarthritis and among them, glucosamine and chondroitin sulfate treatments are probably the most common therapies used in veterinary management. However, heterogeneous results were obtained among animal studies and the evidence of their efficacy is still controversial. Animal models have a crucial role in studying the histological changes and evaluating the therapy efficacy of different drugs. Consequently, we consider it may be of interest to evaluate the effectiveness of the most representative nutraceuticals in experimental animal studies of osteoarthritis. In this systematic review, we found a large inconsistency among the experimental protocols, but a positive cartilage response and biochemical modulation were observed in half of the evaluated articles, mainly associated with pre-emptive administrations and with some therapies’ combinations. Even though some of these results were promising, additional data are needed to draw solid conclusions, and further studies evaluating their efficacy in the long term and focusing on other synovial components may be needed to clarify their function. Abstract Glucosamine and chondroitin sulfate have been proposed due to their physiological and functional benefits in the management of osteoarthritis in companion animals. However, the scientific evidence for their use is still controversial. The purpose of this review was to critically elucidate the efficacy of these nutraceutical therapies in delaying the progression of osteoarthritis, evaluating their impact on the synovial knee joint tissues and biochemical markers in preclinical studies by systematically reviewing the last two decades of peer-reviewed publications on experimental osteoarthritis. Three databases (PubMed, Scopus and, Web of Science) were screened for eligible studies. Twenty-two articles were included in the review. Preclinical studies showed a great heterogeneity among the experimental designs and their outcomes. Generally, the evaluated nutraceuticals, alone or in combination, did not seem to prevent the subchondral bone changes, the synovial inflammation or the osteophyte formation. However, further experimental studies may be needed to evaluate their effect at those levels. Regarding the cartilage status and biomarkers, positive responses were identified in approximately half of the evaluated articles. Furthermore, beneficial effects were associated with the pre-emptive administrations, higher doses and, multimodality approaches with some combined therapies. However, additional studies in the long term and with good quality and systematic design are required.
Collapse
|
49
|
Volpi N, Galeotti F, Maccari F, Capitani F, Mantovani V. Structural definition of terrestrial chondroitin sulfate of various origin and repeatability of the production process. J Pharm Biomed Anal 2020; 195:113826. [PMID: 33358299 DOI: 10.1016/j.jpba.2020.113826] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Revised: 12/02/2020] [Accepted: 12/03/2020] [Indexed: 10/22/2022]
Abstract
We report results on the structure, physicochemical characteristics and purity of chondroitin sulfate (CS) samples derived from three largely available and common biological sources such as bovine and porcine trachea and chicken keel bones with the aim to define their structural signatures. Many lots of CS produced by a manufacturer at industrial scale were characterized with a view to assess the reproducibility of the process as not controlled extractive procedures may produce final products with variable structure and biological contaminants as well as not constant clinical efficacy and safety. By using standardized source animal tissues and manufacturing procedure, highly pure CS (∼92 %) products with constant structure and characteristics were obtained. Bovine CS showed a lower molecular weight (MWw of ∼21,500 Da) than porcine (MWw of ∼26,000 Da) and chicken (MWw of ∼35,900 Da) products with a CV% of ∼2.0-7.5 and a polydispersity variability of 0.7-2.7 %. The ratio between the sulfate groups main located in position 4 and 6 of N-acetyl-galactosamine (4/6 ratio) was ∼1.70 for bovine CS versus a value of 3.60 for porcine and ∼2.70 for chicken samples with a overall charge density of 0.92-0.93 and a CV% of 2.1-2.5. The final products also showed the presence of a very low and constant content of other co-purified bio(macro)molecules (hyaluronic acid, keratan sulfate, dermatan sulfate, heparan sulfate, nucleic acids and proteins), calcium and sodium, and the absence of versican. Finally, a high reproducibility of molecular weight values, disaccharide composition, specific optical rotation and particle dimension was observed. The observed parameters are structural signatures useful to specifically identify the origin of CS and obtained by a standardized and highly reproducible manufacturing process. The compositional profile determined from this study provides a measure of the norm and range of variation in CS samples of terrestrial origin produced under standardized production protocol to which future pharmaceutical/nutraceutical final products can be compared. Moreover, the physicochemical properties including molecular weight, disaccharide composition, presence of natural contaminants and particle dimension were characterized to provide the basis of CS of high quality for application as pharmaceutical/nutraceutical active agents.
Collapse
Affiliation(s)
- Nicola Volpi
- Department of Life Sciences, University of Modena and Reggio Emilia, Modena, Italy.
| | - Fabio Galeotti
- Department of Life Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Francesca Maccari
- Department of Life Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Federica Capitani
- Department of Life Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Veronica Mantovani
- Department of Life Sciences, University of Modena and Reggio Emilia, Modena, Italy; Clinical and Experimental Medicine PhD Program, University of Modena and Reggio Emilia, Modena, Italy
| |
Collapse
|
50
|
Honvo G, Lengelé L, Charles A, Reginster JY, Bruyère O. Role of Collagen Derivatives in Osteoarthritis and Cartilage Repair: A Systematic Scoping Review With Evidence Mapping. Rheumatol Ther 2020; 7:703-740. [PMID: 33068290 PMCID: PMC7695755 DOI: 10.1007/s40744-020-00240-5] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Accepted: 09/23/2020] [Indexed: 12/17/2022] Open
Abstract
INTRODUCTION There is currently no disease-modifying drug for osteoarthritis (OA), and some safety concerns have been identified about the leading traditional drugs. Therefore, research efforts have focused on alternatives such as supplementation with collagen derivatives. The objective of this scoping review is to examine the extent, range, and nature of research, and to summarize and disseminate research findings on the effects of collagen derivatives in OA and cartilage repair. The purpose is to identify gaps in the current body of evidence in order to further help progress research in this setting. METHODS The databases Medline, Scopus, CENTRAL, TOXLINE, and CDSR were comprehensively searched from inception to search date. After studies selection against eligibility criteria, following recommended methods, data were charted from the retrieved articles and these were subsequently synthesized. Numerical and graphical descriptive statistical methods were used to show trends in publications and geographical distribution of studies. RESULTS The systematic literature search identified a total of 10,834 records. Forty-one published studies were ultimately included in the review, 16 of which were preclinical studies and 25 were clinical studies (including four systematic reviews/meta-analyses). Collagen hydrolysate (CH) and undenatured collagen (UC) were the two types of collagen derivatives studied, with a total of 28 individual studies on CH and nine on UC. More than a third of studies originated from Asia, and most of them have been published after 2008. Oral forms of collagen derivatives were mainly studied; three in vivo preclinical studies and three clinical trials investigated intra-articularly injected CH. In most of the clinical trials, treatment durations varied between 3 and 6 months, with the shortest being 1.4 months and the longest 11 months. All in vivo preclinical studies and clinical trials, regardless of their quality, concluded on beneficial effects of collagen derivatives in OA and cartilage repair, whether used as nutritional supplement or delivered intra-articularly, and whatever the manufacturers of the products, the doses and the outcomes considered in each study. CONCLUSIONS Although current evidence shows some potential for the use of CH and UC as an option for management of patients with OA, there is still room for progress in terms of laboratory and clinical research before any definitive conclusion can be made. Harmonization of outcomes in preclinical studies and longer randomized placebo-controlled trials in larger populations with the use of recommended and validated endpoints are warranted before collagen derivatives can be recommended by large scientific societies.
Collapse
Affiliation(s)
- Germain Honvo
- Division of Public Health, Epidemiology and Health Economics, University of Liège, Liège, Belgium.
- World Health Organization (WHO) Collaborating Center for Public Health Aspects of Musculoskeletal Health and Aging, University of Liège, Liège, Belgium.
| | - Laetitia Lengelé
- Division of Public Health, Epidemiology and Health Economics, University of Liège, Liège, Belgium
- World Health Organization (WHO) Collaborating Center for Public Health Aspects of Musculoskeletal Health and Aging, University of Liège, Liège, Belgium
| | - Alexia Charles
- Division of Public Health, Epidemiology and Health Economics, University of Liège, Liège, Belgium
- World Health Organization (WHO) Collaborating Center for Public Health Aspects of Musculoskeletal Health and Aging, University of Liège, Liège, Belgium
| | - Jean-Yves Reginster
- Division of Public Health, Epidemiology and Health Economics, University of Liège, Liège, Belgium
- World Health Organization (WHO) Collaborating Center for Public Health Aspects of Musculoskeletal Health and Aging, University of Liège, Liège, Belgium
- Chair for Biomarkers Research, Biochemistry Department, College of Science, King Saud University, Riyadh, Kingdom of Saudi Arabia
| | - Olivier Bruyère
- Division of Public Health, Epidemiology and Health Economics, University of Liège, Liège, Belgium
- World Health Organization (WHO) Collaborating Center for Public Health Aspects of Musculoskeletal Health and Aging, University of Liège, Liège, Belgium
| |
Collapse
|