351
|
Na HS, Kwon JY, Lee SY, Lee SH, Lee AR, Woo JS, Jung K, Cho KH, Choi JW, Lee DH, Min HK, Park SH, Kim SJ, Cho ML. Metformin Attenuates Monosodium-Iodoacetate-Induced Osteoarthritis via Regulation of Pain Mediators and the Autophagy-Lysosomal Pathway. Cells 2021; 10:681. [PMID: 33808727 PMCID: PMC8003384 DOI: 10.3390/cells10030681] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Revised: 03/15/2021] [Accepted: 03/16/2021] [Indexed: 12/19/2022] Open
Abstract
Osteoarthritis (OA) is the most common degenerative arthritis associated with pain and cartilage destruction in the elderly; it is known to be involved in inflammation as well. A drug called celecoxib is commonly used in patients with osteoarthritis to control pain. Metformin is used to treat type 2 diabetes but also exhibits regulation of the autophagy pathway. The purpose of this study is to investigate whether metformin can treat monosodium iodoacetate (MIA)-induced OA in rats. Metformin was administered orally every day to rats with OA. Paw-withdrawal latency and threshold were used to assess pain severity. Cartilage damage and pain mediators in dorsal root ganglia were evaluated by histological analysis and a scoring system. Relative mRNA expression was measured by real-time PCR. Metformin reduced the progression of experimental OA and showed both antinociceptive properties and cartilage protection. The combined administration of metformin and celecoxib controlled cartilage damage more effectively than metformin alone. In chondrocytes from OA patients, metformin reduced catabolic factor gene expression and inflammatory cell death factor expression, increased LC3Ⅱb, p62, and LAMP1 expression, and induced an autophagy-lysosome fusion phenotype. We investigated if metformin treatment reduces cartilage damage and inflammatory cell death of chondrocytes. The results suggest the potential for the therapeutic use of metformin in OA patients based on its ability to suppress pain and protect cartilage.
Collapse
Affiliation(s)
- Hyun Sik Na
- The Rheumatism Research Center, Catholic Reasearch Institute of Medical Science, The Catholic University of Korea, Seoul 06591, Korea; (H.S.N.); (J.Y.K.); (S.-Y.L.); (S.H.L.); (A.R.L.); (J.S.W.); (K.-H.C.); (J.-W.C.)
| | - Ji Ye Kwon
- The Rheumatism Research Center, Catholic Reasearch Institute of Medical Science, The Catholic University of Korea, Seoul 06591, Korea; (H.S.N.); (J.Y.K.); (S.-Y.L.); (S.H.L.); (A.R.L.); (J.S.W.); (K.-H.C.); (J.-W.C.)
| | - Seon-Yeong Lee
- The Rheumatism Research Center, Catholic Reasearch Institute of Medical Science, The Catholic University of Korea, Seoul 06591, Korea; (H.S.N.); (J.Y.K.); (S.-Y.L.); (S.H.L.); (A.R.L.); (J.S.W.); (K.-H.C.); (J.-W.C.)
| | - Seung Hoon Lee
- The Rheumatism Research Center, Catholic Reasearch Institute of Medical Science, The Catholic University of Korea, Seoul 06591, Korea; (H.S.N.); (J.Y.K.); (S.-Y.L.); (S.H.L.); (A.R.L.); (J.S.W.); (K.-H.C.); (J.-W.C.)
| | - A Ram Lee
- The Rheumatism Research Center, Catholic Reasearch Institute of Medical Science, The Catholic University of Korea, Seoul 06591, Korea; (H.S.N.); (J.Y.K.); (S.-Y.L.); (S.H.L.); (A.R.L.); (J.S.W.); (K.-H.C.); (J.-W.C.)
- Department of Biomedicine & Health Sciences, College of Medicine, The Catholic University of Korea, 222, Banpo-daero, Seocho-gu, Seoul 06591, Korea
| | - Jin Seok Woo
- The Rheumatism Research Center, Catholic Reasearch Institute of Medical Science, The Catholic University of Korea, Seoul 06591, Korea; (H.S.N.); (J.Y.K.); (S.-Y.L.); (S.H.L.); (A.R.L.); (J.S.W.); (K.-H.C.); (J.-W.C.)
| | - KyungAh Jung
- Impact Biotech, Korea 505 Banpo-Dong, Seocho-Ku, Seoul 06591, Korea;
| | - Keun-Hyung Cho
- The Rheumatism Research Center, Catholic Reasearch Institute of Medical Science, The Catholic University of Korea, Seoul 06591, Korea; (H.S.N.); (J.Y.K.); (S.-Y.L.); (S.H.L.); (A.R.L.); (J.S.W.); (K.-H.C.); (J.-W.C.)
| | - Jeong-Won Choi
- The Rheumatism Research Center, Catholic Reasearch Institute of Medical Science, The Catholic University of Korea, Seoul 06591, Korea; (H.S.N.); (J.Y.K.); (S.-Y.L.); (S.H.L.); (A.R.L.); (J.S.W.); (K.-H.C.); (J.-W.C.)
| | - Dong Hwan Lee
- Department of Orthopedic Surgery, Uijeongbu St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul 11765, Korea;
| | - Hong-Ki Min
- Division of Rheumatology, Department of Internal Medicine, Konkuk University Medical Center, Seoul 05030, Korea;
| | - Sung-Hwan Park
- Division of Rheumatology, Department of Internal Medicine, Seoul St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul 06591, Korea
| | - Seok Jung Kim
- Department of Orthopedic Surgery, Uijeongbu St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul 11765, Korea;
| | - Mi-La Cho
- The Rheumatism Research Center, Catholic Reasearch Institute of Medical Science, The Catholic University of Korea, Seoul 06591, Korea; (H.S.N.); (J.Y.K.); (S.-Y.L.); (S.H.L.); (A.R.L.); (J.S.W.); (K.-H.C.); (J.-W.C.)
- Department of Medical Lifescience, College of Medicine, The Catholic University of Korea, Seoul 06591, Korea
| |
Collapse
|
352
|
Extracellular Vesicles from Mesenchymal Stromal Cells for the Treatment of Inflammation-Related Conditions. Int J Mol Sci 2021; 22:ijms22063023. [PMID: 33809632 PMCID: PMC8002312 DOI: 10.3390/ijms22063023] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Revised: 03/06/2021] [Accepted: 03/12/2021] [Indexed: 12/14/2022] Open
Abstract
Over the past two decades, mesenchymal stromal cells (MSCs) have demonstrated great potential in the treatment of inflammation-related conditions. Numerous early stage clinical trials have suggested that this treatment strategy has potential to lead to significant improvements in clinical outcomes. While promising, there remain substantial regulatory hurdles, safety concerns, and logistical issues that need to be addressed before cell-based treatments can have widespread clinical impact. These drawbacks, along with research aimed at elucidating the mechanisms by which MSCs exert their therapeutic effects, have inspired the development of extracellular vesicles (EVs) as anti-inflammatory therapeutic agents. The use of MSC-derived EVs for treating inflammation-related conditions has shown therapeutic potential in both in vitro and small animal studies. This review will explore the current research landscape pertaining to the use of MSC-derived EVs as anti-inflammatory and pro-regenerative agents in a range of inflammation-related conditions: osteoarthritis, rheumatoid arthritis, Alzheimer's disease, cardiovascular disease, and preeclampsia. Along with this, the mechanisms by which MSC-derived EVs exert their beneficial effects on the damaged or degenerative tissues will be reviewed, giving insight into their therapeutic potential. Challenges and future perspectives on the use of MSC-derived EVs for the treatment of inflammation-related conditions will be discussed.
Collapse
|
353
|
Zhang X, He J, Wang W. Progress in the use of mesenchymal stromal cells for osteoarthritis treatment. Cytotherapy 2021; 23:459-470. [PMID: 33736933 DOI: 10.1016/j.jcyt.2021.01.008] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Revised: 12/20/2020] [Accepted: 01/29/2021] [Indexed: 12/26/2022]
Abstract
LITERATURE REVIEW OF MSCS IN THE TREATMENT OF OSTEOARTHRITIS IN THE PAST FIVE YEARS: Osteoarthritis (OA) is one of the most common chronic joint diseases, with prominent symptoms caused by many factors. However, current medical interventions for OA have resulted in poor clinical outcomes, demonstrating that there are huge unmet medical needs in this area. Cell therapy has opened new avenues of OA treatment. Different sources of mesenchymal stromal cells (MSCs) may have different phenotypes and cellular functions. Pre-clinical and clinical studies have demonstrated the feasibility, safety and efficacy of MSC therapy. Mitogen-activated protein kinase, Wnt and Notch signaling pathways are involved in the chondrogenesis of MSC-mediated treatments. MSCs may also exert effective immunoregulatory and paracrine effects to stimulate tissue repair. Therapy with extracellular vesicles containing cytokines, which are secreted by MSCs, might be a potential treatment for OA.
Collapse
Affiliation(s)
- Xiaotian Zhang
- Department of Orthopedics, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Jiyin He
- Department of Orthopedics, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Wen Wang
- Clinical Development, IASO Biotherapeutics Co., Ltd., Shanghai, China.
| |
Collapse
|
354
|
Liu J, Ting JP, Al-Azzam S, Ding Y, Afshar S. Therapeutic Advances in Diabetes, Autoimmune, and Neurological Diseases. Int J Mol Sci 2021; 22:ijms22062805. [PMID: 33802091 PMCID: PMC8001105 DOI: 10.3390/ijms22062805] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2021] [Revised: 03/02/2021] [Accepted: 03/06/2021] [Indexed: 02/08/2023] Open
Abstract
Since 2015, 170 small molecules, 60 antibody-based entities, 12 peptides, and 15 gene- or cell-therapies have been approved by FDA for diverse disease indications. Recent advancement in medicine is facilitated by identification of new targets and mechanisms of actions, advancement in discovery and development platforms, and the emergence of novel technologies. Early disease detection, precision intervention, and personalized treatments have revolutionized patient care in the last decade. In this review, we provide a comprehensive overview of current and emerging therapeutic modalities developed in the recent years. We focus on nine diseases in three major therapeutics areas, diabetes, autoimmune, and neurological disorders. The pathogenesis of each disease at physiological and molecular levels is discussed and recently approved drugs as well as drugs in the clinic are presented.
Collapse
Affiliation(s)
- Jinsha Liu
- Protein Engineering, Lilly Biotechnology Center, Eli Lilly and Company, San Diego, CA 92121, USA; (J.L.); (J.P.T.); (Y.D.)
| | - Joey Paolo Ting
- Protein Engineering, Lilly Biotechnology Center, Eli Lilly and Company, San Diego, CA 92121, USA; (J.L.); (J.P.T.); (Y.D.)
| | - Shams Al-Azzam
- Professional Scientific Services, Eurofins Lancaster Laboratories, Lancaster, PA 17605, USA;
| | - Yun Ding
- Protein Engineering, Lilly Biotechnology Center, Eli Lilly and Company, San Diego, CA 92121, USA; (J.L.); (J.P.T.); (Y.D.)
| | - Sepideh Afshar
- Protein Engineering, Lilly Biotechnology Center, Eli Lilly and Company, San Diego, CA 92121, USA; (J.L.); (J.P.T.); (Y.D.)
- Correspondence:
| |
Collapse
|
355
|
El-Bidawy MH, Omar Hussain AB, Al-Ghamdi S, Aldossari KK, Haidara MA, Al-Ani B. Resveratrol ameliorates type 2 diabetes mellitus-induced alterations to the knee joint articular cartilage ultrastructure in rats. Ultrastruct Pathol 2021; 45:92-101. [PMID: 33567949 DOI: 10.1080/01913123.2021.1882629] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2020] [Accepted: 01/25/2021] [Indexed: 02/08/2023]
Abstract
Diabetes-induced osteoarthritis (OA) is a chronic inflammatory disease that damages the cartilage in the joints and could lead to disability. The protective effect of the antioxidant and anti-inflammatory agent, resveratrol, against alterations to the knee articular cartilage ultrastructure induced by type 2 diabetes mellitus (T2DM) associated with the inhibition of dyslipidemia, oxidative stress, and inflammation has not been investigated before. Therefore, we modeled OA in rats 10 weeks post diabetic induction using a high carbohydrate and fat diet and a single injection of streptozotocin (50 mg/kg body weight), and the protective group of rats started resveratrol (30 mg/kg; orally) treatment 2 weeks before diabetic induction and continued on resveratrol until the end of the experiment at week 12. Blood chemistry analysis confirmed hyperglycemia (elevated glucose and glycated hemoglobin, HbA1c), dyslipidemia (elevated triglyceride, cholesterol, and low-density lipoprotein-cholesterol), and upregulation of oxidative stress (malondialdehyde) and inflammatory (C-reactive protein and tumor necrosis factor-α) biomarkers in the model group. In addition, using light and electron microscopy examinations, we also observed in the model group substantial damage to the articular cartilage and profound chondrocyte and territorial matrix ultrastructural alterations such as chondrocytes with degenerated nucleus and mitochondria, scarce cytoplasmic processes, and absence of the fine fibrillar appearance of territorial matrix. Resveratrol pretreatment significantly (p ≤ 0.0029) but not completely protected from T2DM-induced OA. We conclude that resveratrol protects against alterations to the articular cartilage ultrastructure induced secondary to T2DM in rats, which is associated with the inhibition of glycemia, hyperlipidemia, and biomarkers of oxidative stress and inflammation.
Collapse
Affiliation(s)
- Mahmoud H El-Bidawy
- Department of BMS, Division of Physiology, College of Medicine, Prince Sattam Ibin Abdulaziz University, Al-Kharj, Saudi Arabia
- Department of Physiology, Kasr Al-Aini Faculty of Medicine, Cairo University, Cairo, Egypt
| | - Abo Bakr Omar Hussain
- Department of Cardiology, College of Medicine, Prince Sattam Ibin Abdulaziz University, Al-Kharj,Saudi Arabia
| | - Sameer Al-Ghamdi
- Department of Family &community Medicine,College of Medicine, Prince Sattam Ibin Abdulaziz University, Al-Kharj, Saudi Arabia
| | - Khalid K Aldossari
- Department of Family &community Medicine, College of Medicine, Prince Sattam Ibin Abdulaziz University, Al-Kharj,Saudi Arabia
| | - Mohamed A Haidara
- Department of Physiology, Kasr Al-Aini Faculty of Medicine, Cairo University, Cairo, Egypt
| | - Bahjat Al-Ani
- Department of Physiology, College of Medicine, King Khalid University, Abha, Saudi Arabia
| |
Collapse
|
356
|
Rose BJ, Weyand JA, Liu B, Smith JF, Perez BR, Clark JC, Goodman M, Hirschi Budge KM, Eggett DL, Arroyo JA, Reynolds PR, Kooyman DL. Exposure to second-hand cigarette smoke exacerbates the progression of osteoarthritis in a surgical induced murine model. Histol Histopathol 2021; 36:347-353. [PMID: 33576000 DOI: 10.14670/hh-18-311] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Osteoarthritis (OA), formerly understood to be a result of passive wear, is now known to be associated with chronic inflammation. Cigarette smoking promotes systemic inflammation and has been implicated in increased joint OA incidence in some studies, though the recent observational data on the association are contradictory. We hypothesize that second-hand smoke (SHS) treatment will increase the incidence of OA in a mouse model that has been subjected to a surgical destabilization of the medial meniscus (DMM). To test this hypothesis, we applied either SHS treatment or room air (RA) to mice for 28 days post-DMM surgery. Histopathology findings indicated that the knees of SHS mice exhibited more severe OA than their control counterparts. Increased expression of matrix metalloprotease-13 (MMP-13), an important extracellular protease known to degrade articular cartilage, and nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB), an intracellular effector of inflammatory pathways, were observed in the SHS group. These findings provide greater understanding and evidence for a detrimental role of cigarette smoke on OA progression and systemic inflammation.
Collapse
Affiliation(s)
- Brandon J Rose
- Department of Physiology and Developmental Biology, Brigham Young University, Provo, Utah, United States
| | - Jeffery A Weyand
- Department of Physiology and Developmental Biology, Brigham Young University, Provo, Utah, United States
| | - Brady Liu
- Department of Physiology and Developmental Biology, Brigham Young University, Provo, Utah, United States
| | - Jacob F Smith
- Department of Physiology and Developmental Biology, Brigham Young University, Provo, Utah, United States
| | - Brian R Perez
- Department of Physiology and Developmental Biology, Brigham Young University, Provo, Utah, United States
| | - J Christian Clark
- Department of Physiology and Developmental Biology, Brigham Young University, Provo, Utah, United States
| | - Michael Goodman
- Department of Physiology and Developmental Biology, Brigham Young University, Provo, Utah, United States
| | - Kelsey M Hirschi Budge
- Department of Physiology and Developmental Biology, Brigham Young University, Provo, Utah, United States
| | - Dennis L Eggett
- Department of Physiology and Developmental Biology, Brigham Young University, Provo, Utah, United States
| | - Juan A Arroyo
- Department of Physiology and Developmental Biology, Brigham Young University, Provo, Utah, United States
| | - Paul R Reynolds
- Department of Physiology and Developmental Biology, Brigham Young University, Provo, Utah, United States
| | - David L Kooyman
- Department of Physiology and Developmental Biology, Brigham Young University, Provo, Utah, United States
| |
Collapse
|
357
|
Effects of tart cherry and its metabolites on aging and inflammatory conditions: Efficacy and possible mechanisms. Ageing Res Rev 2021; 66:101254. [PMID: 33434683 DOI: 10.1016/j.arr.2021.101254] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2020] [Revised: 12/15/2020] [Accepted: 12/30/2020] [Indexed: 01/16/2023]
Abstract
Inflammation is an underlying cause of or a contributing factor to a number of chronic conditions, including hypertension, insulin resistance, arthritis, and cognitive disorders. A chronic inflammatory state is also associated with aging. Tart cherry (TC) has been extensively studied for its ability to prevent or treat inflammatory diseases and their associated risk factors. TC contains active compounds, including polyphenols that may contribute to its antioxidant and anti-inflammatory effects. Inflammatory signaling pathways regulate the recruitment of inflammatory cells important for the pathogenesis of disease. Whole TC, individual compounds, and their metabolites may be viable treatment options because they can target molecules involved in inflammatory pathways. In this review, the effectiveness of TC in reducing inflammatory markers associated with chronic diseases and the effects of the active compounds in TC and their metabolites on inflammatory pathways are discussed. The main polyphenols present in TC include cyanidins, kaempferol, quercetin, melatonin, neochlorogenic acid, chlorogenic acid, and 3-coumaroylquinic acid. Evidence supports an association between TC intake and reduced risk for inflammatory disease, which may be due to the effects of active compounds in TC on inflammatory pathways, such as NF-κB and mitogen-activated protein kinase.
Collapse
|
358
|
Autologous protein solution as selective treatment for advanced patellofemoral osteoarthritis in the middle-aged female patient: 54% response rate at 1 year follow-up. Knee Surg Sports Traumatol Arthrosc 2021; 29:988-997. [PMID: 32451622 DOI: 10.1007/s00167-020-06064-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/12/2020] [Accepted: 05/13/2020] [Indexed: 12/20/2022]
Abstract
PURPOSE The study wanted to investigate the benefit, durability and safety of autologous protein solution (APS) injection(s) in a middle-aged female-only cohort suffering predominantly from patellofemoral osteoarthritis. METHODS Fifty females (aged 50.4 ± 6.5) with mainly moderate-severe (86%) patellofemoral cartilage wear (PFCW) were treated with a unilateral intra-articular APS injection. The KOOS, NRS, Kujala, UCLA and EQ-5D were assessed at baseline and 1, 3, 6, and 12 months post-injection. Therapeutic response rate (TRR) was based on KOOS pain improvement > 10 points. Absolute improvement for, respectively, therapy responders and non-responders was determined. Second APS injection was administered if improvement was deemed insufficient by the patient after 3 months. RESULTS The TRR remained stable averaging to 53.7% at final follow-up with subjects improving overall from 40.3 ± 18.7 to 57.3 ± 24.8 points on KOOS pain (p = 0.0002) and from 48.4 ± 13.0 to 56.3 ± 18.1 points on Kujala (p = 0.0203) at 12 months. Significant improvement was observed for the other KOOS subscales and NRS at each follow-up. In absolute values, APS responders improved with 30.5 ± 11.4 points on KOOS pain at 12 months. In contrast, non-responders deteriorated with 5.9 ± 8.9 points relative to baseline. A second APS injection was administered in 28 subjects. Patients with definite synovitis improved more on KOOS symptoms (p = 0.017) and KOOS ADL (p = 0.037) at 12 months compared to non-synovitis subjects. Mild-moderate arthralgia (46%) and effusion (29%) were commonly observed during the first month post-injection. CONCLUSION This study evidenced a 54% response rate at 12 months to a single or second APS injection in a middle-aged female population with advanced patellofemoral cartilage wear. Moderate temporary flares can be expected without affecting clinical outcomes. Second APS injection has low efficacy in initially poor responding patients after 3 months. Major synovitis on baseline MRI appeared to be a beneficial prognosticator for pain relief and functional improvement after APS. LEVEL OF EVIDENCE IV.
Collapse
|
359
|
Xu Z, He Z, Shu L, Li X, Ma M, Ye C. Intra-Articular Platelet-Rich Plasma Combined With Hyaluronic Acid Injection for Knee Osteoarthritis Is Superior to Platelet-Rich Plasma or Hyaluronic Acid Alone in Inhibiting Inflammation and Improving Pain and Function. Arthroscopy 2021; 37:903-915. [PMID: 33091549 DOI: 10.1016/j.arthro.2020.10.013] [Citation(s) in RCA: 53] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/04/2020] [Revised: 09/30/2020] [Accepted: 10/11/2020] [Indexed: 02/02/2023]
Abstract
PURPOSE To evaluate the effectiveness and explore the therapeutic mechanisms of platelet-rich plasma (PRP) combined with hyaluronic acid (HA) as a treatment for knee osteoarthritis (KOA). METHODS In total, 122 knees were randomly divided into HA (34 knees), PRP (40 knees), and PRP+HA (48 knees) groups. Platelet densities in whole blood and PRP were examined using Wright-Giemsa staining. Visual analogue scale, Lequesne, Western Ontario and McMaster Universities Osteoarthritis Index, Lysholm scores, and postoperative complications were evaluated. High-frequency color Doppler imaging was used to observe the synovium and cartilage. Enzyme-linked immunosorbent assays were used to quantify interleukin-1β, tumor necrosis factor-α, matrix metalloproteinase-3, and tissue inhibitor of metalloproteinase-1 levels in synovial fluid. RESULTS The platelet density in PRP was 5.13-times that in whole blood (P = .002). At 24 months, pain and function scores in the PRP+HA group were better than those in the HA-alone and PRP-alone groups (Ppain = .000; Pfunction = .000). At 6 and 12 months, synovial hyperplasia in the PRP and PRP+HA groups was improved (P < .05). After 6 and 12 months, the synovial peak systolic velocity, synovial end-diastolic velocity, systolic/diastolic ratio, and resistance index were improved in the PRP+HA group (P < .05). Complications were greatest in the PRP group (P = .008). After 6 and 12 months, interleukin-1β, tumor necrosis factor-α, matrix metalloproteinase-3, and tissue inhibitor of metalloproteinase-1 in the PRP and PRP+HA groups decreased (P < .05), with more apparent inhibition in the PRP+HA group (P < .05). CONCLUSIONS PRP combined with HA is more effective than PRP or HA alone at inhibiting synovial inflammation and can effectively improve pain and function and reduce adverse reactions. Its mechanism involves changes in the synovium and cytokine content. LEVEL OF EVIDENCE Level II, Prospective cohort study.
Collapse
Affiliation(s)
- Zhe Xu
- Department of Orthopaedics, The Affiliated Hospital of Guizhou Medical University, Guiyang, China; Key Laboratory of Adult Stem Cell Transformation Research, Chinese Academy of Medical Sciences, Guiyang, China; National-Local Joint Engineering Laboratory of Cell Engineering and Biomedicine, Guiyang, China; Center for Tissue Engineering and Stem Cell Research, Guizhou Medical University, Guiyang, China
| | - Zhixu He
- Key Laboratory of Adult Stem Cell Transformation Research, Chinese Academy of Medical Sciences, Guiyang, China
| | - Liping Shu
- Key Laboratory of Adult Stem Cell Transformation Research, Chinese Academy of Medical Sciences, Guiyang, China; National-Local Joint Engineering Laboratory of Cell Engineering and Biomedicine, Guiyang, China; Center for Tissue Engineering and Stem Cell Research, Guizhou Medical University, Guiyang, China
| | - Xuanze Li
- Department of Orthopaedics, The Affiliated Hospital of Guizhou Medical University, Guiyang, China; Center for Tissue Engineering and Stem Cell Research, Guizhou Medical University, Guiyang, China
| | - Minxian Ma
- Key Laboratory of Adult Stem Cell Transformation Research, Chinese Academy of Medical Sciences, Guiyang, China; National-Local Joint Engineering Laboratory of Cell Engineering and Biomedicine, Guiyang, China; Center for Tissue Engineering and Stem Cell Research, Guizhou Medical University, Guiyang, China
| | - Chuan Ye
- Department of Orthopaedics, The Affiliated Hospital of Guizhou Medical University, Guiyang, China; Key Laboratory of Adult Stem Cell Transformation Research, Chinese Academy of Medical Sciences, Guiyang, China; National-Local Joint Engineering Laboratory of Cell Engineering and Biomedicine, Guiyang, China; Center for Tissue Engineering and Stem Cell Research, Guizhou Medical University, Guiyang, China; China Orthopaedic Regenerative Medicine Group (CORMed), Hangzhou, China.
| |
Collapse
|
360
|
Xiao H, Chen J, Duan L, Li S. Role of emerging vitamin K‑dependent proteins: Growth arrest‑specific protein 6, Gla‑rich protein and periostin (Review). Int J Mol Med 2021; 47:2. [PMID: 33448308 PMCID: PMC7834955 DOI: 10.3892/ijmm.2020.4835] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2020] [Accepted: 10/21/2020] [Indexed: 01/27/2023] Open
Abstract
Vitamin K‑dependent proteins (VKDPs) are a group of proteins that need vitamin K to conduct carboxylation. Thus far, scholars have identified a total of 17 VKDPs in the human body. In this review, we summarize three important emerging VKDPs: Growth arrest‑specific protein 6 (Gas 6), Gla‑rich protein (GRP) and periostin in terms of their functions in physiological and pathological conditions. As examples, carboxylated Gas 6 and GRP effectively protect blood vessels from calcification, Gas 6 protects from acute kidney injury and is involved in chronic kidney disease, GRP contributes to bone homeostasis and delays the progression of osteoarthritis, and periostin is involved in all phases of fracture healing and assists myocardial regeneration in the early stages of myocardial infarction. However, periostin participates in the progression of cardiac fibrosis, idiopathic pulmonary fibrosis and airway remodeling of asthma. In addition, we discuss the relationship between vitamin K, VKDPs and cancer, and particularly the carboxylation state of VKDPs in cancer.
Collapse
Affiliation(s)
- Huiyu Xiao
- Department of Physiology, Dalian Medical University, Dalian, Liaoning 116044
| | - Jiepeng Chen
- Sungen Bioscience Co., Ltd., Shantou, Guangdong 515071, P.R. China
| | - Lili Duan
- Sungen Bioscience Co., Ltd., Shantou, Guangdong 515071, P.R. China
| | - Shuzhuang Li
- Department of Physiology, Dalian Medical University, Dalian, Liaoning 116044
| |
Collapse
|
361
|
Morouço P, Fernandes C, Lattanzi W. Challenges and Innovations in Osteochondral Regeneration: Insights from Biology and Inputs from Bioengineering toward the Optimization of Tissue Engineering Strategies. J Funct Biomater 2021; 12:17. [PMID: 33673516 PMCID: PMC7931100 DOI: 10.3390/jfb12010017] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Revised: 02/17/2021] [Accepted: 02/23/2021] [Indexed: 02/06/2023] Open
Abstract
Due to the extremely high incidence of lesions and diseases in aging population, it is critical to put all efforts into developing a successful implant for osteochondral tissue regeneration. Many of the patients undergoing surgery present osteochondral fissure extending until the subchondral bone (corresponding to a IV grade according to the conventional radiographic classification by Berndt and Harty). Therefore, strategies for functional tissue regeneration should also aim at healing the subchondral bone and joint interface, besides hyaline cartilage. With the ambition of contributing to solving this problem, several research groups have been working intensively on the development of tailored implants that could promote that complex osteochondral regeneration. These implants may be manufactured through a wide variety of processes and use a wide variety of (bio)materials. This review aimed to examine the state of the art regarding the challenges, advantages, and drawbacks of the current strategies for osteochondral regeneration. One of the most promising approaches relies on the principles of additive manufacturing, where technologies are used that allow for the production of complex 3D structures with a high level of control, intended and predefined geometry, size, and interconnected pores, in a reproducible way. However, not all materials are suitable for these processes, and their features should be examined, targeting a successful regeneration.
Collapse
Affiliation(s)
| | | | - Wanda Lattanzi
- Department of Life Science and Public Health, Università Cattolica del Sacro Cuore, 00168 Rome, Italy;
| |
Collapse
|
362
|
Liu M, Xie Z, Sun G, Chen L, Qi D, Zhang H, Xiong J, Furey A, Rahman P, Lei G, Zhai G. Macrophage migration inhibitory factor may play a protective role in osteoarthritis. Arthritis Res Ther 2021; 23:59. [PMID: 33610191 PMCID: PMC7896408 DOI: 10.1186/s13075-021-02442-w] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Accepted: 02/10/2021] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Osteoarthritis (OA) is the most prevalent form of arthritis and the major cause of disability and overall diminution of quality of life in the elderly population. Currently there is no cure for OA, partly due to the large gaps in our understanding of its underlying molecular and cellular mechanisms. Macrophage migration inhibitory factor (MIF) is a procytokine that mediates pleiotropic inflammatory effects in inflammatory diseases such as rheumatoid arthritis (RA) and ankylosing spondylitis (AS). However, data on the role of MIF in OA is limited with conflicting results. We undertook this study to investigate the role of MIF in OA by examining MIF genotype, mRNA expression, and protein levels in the Newfoundland Osteoarthritis Study. METHODS One hundred nineteen end-stage knee/hip OA patients, 16 RA patients, and 113 healthy controls were included in the study. Two polymorphisms in the MIF gene, rs755622, and -794 CATT5-8, were genotyped using polymerase chain reaction-restriction fragment length polymorphism (PCR-RFLP) and PCR followed by automated capillary electrophoresis, respectively. MIF mRNA levels in articular cartilage and subchondral bone were measured by quantitative polymerase chain reaction. Plasma concentrations of MIF, tumor necrosis factor-alpha (TNF-α), interleukin-6 (IL-6), and interleukin-1 beta (IL-1β) were measured by enzyme-linked immunosorbent assay. RESULTS rs755622 and -794 CATT5-8 genotypes were not associated with MIF mRNA or protein levels or OA (all p ≥ 0.19). MIF mRNA level in cartilage was lower in OA patients than in controls (p = 0.028) and RA patients (p = 0.004), while the levels in bone were comparable between OA patients and controls (p = 0.165). MIF protein level in plasma was lower in OA patients than in controls (p = 3.01 × 10-10), while the levels of TNF-α, IL-6 and IL-1β in plasma were all significantly higher in OA patients than in controls (all p ≤ 0.0007). Multivariable logistic regression showed lower MIF and higher IL-1β protein levels in plasma were independently associated with OA (OR per SD increase = 0.10 and 8.08; 95% CI = 0.04-0.19 and 4.42-16.82, respectively), but TNF-α and IL-6 became non-significant. CONCLUSIONS Reduced MIF mRNA and protein expression in OA patients suggested MIF might have a protective role in OA and could serve as a biomarker to differentiate OA from other joint disorders.
Collapse
Affiliation(s)
- Ming Liu
- Division of Biomedical Sciences (Genetics), Faculty of Medicine, Memorial University of Newfoundland, St. John's, NL, A1B 3V6, Canada
| | - Zikun Xie
- Division of Biomedical Sciences (Genetics), Faculty of Medicine, Memorial University of Newfoundland, St. John's, NL, A1B 3V6, Canada.,Department of Orthopaedics, Xiangya Hospital, Central South University, Changsha, China
| | - Guang Sun
- Discipline of Medicine, Faculty of Medicine, Memorial University of Newfoundland, St. John's, Canada
| | - Liujun Chen
- College of Pharmacy, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Dake Qi
- College of Pharmacy, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Hongwei Zhang
- Discipline of Medicine, Faculty of Medicine, Memorial University of Newfoundland, St. John's, Canada
| | - Jieying Xiong
- Division of Biomedical Sciences (Genetics), Faculty of Medicine, Memorial University of Newfoundland, St. John's, NL, A1B 3V6, Canada
| | - Andrew Furey
- Discipline of Surgery, Faculty of Medicine, Memorial University of Newfoundland, St. John's, Canada
| | - Proton Rahman
- Discipline of Medicine, Faculty of Medicine, Memorial University of Newfoundland, St. John's, Canada
| | - Guanghua Lei
- Department of Orthopaedics, Xiangya Hospital, Central South University, Changsha, China
| | - Guangju Zhai
- Division of Biomedical Sciences (Genetics), Faculty of Medicine, Memorial University of Newfoundland, St. John's, NL, A1B 3V6, Canada.
| |
Collapse
|
363
|
Khella CM, Asgarian R, Horvath JM, Rolauffs B, Hart ML. An Evidence-Based Systematic Review of Human Knee Post-Traumatic Osteoarthritis (PTOA): Timeline of Clinical Presentation and Disease Markers, Comparison of Knee Joint PTOA Models and Early Disease Implications. Int J Mol Sci 2021; 22:1996. [PMID: 33671471 PMCID: PMC7922905 DOI: 10.3390/ijms22041996] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Revised: 02/05/2021] [Accepted: 02/08/2021] [Indexed: 12/15/2022] Open
Abstract
Understanding the causality of the post-traumatic osteoarthritis (PTOA) disease process of the knee joint is important for diagnosing early disease and developing new and effective preventions or treatments. The aim of this review was to provide detailed clinical data on inflammatory and other biomarkers obtained from patients after acute knee trauma in order to (i) present a timeline of events that occur in the acute, subacute, and chronic post-traumatic phases and in PTOA, and (ii) to identify key factors present in the synovial fluid, serum/plasma and urine, leading to PTOA of the knee in 23-50% of individuals who had acute knee trauma. In this context, we additionally discuss methods of simulating knee trauma and inflammation in in vivo, ex vivo articular cartilage explant and in vitro chondrocyte models, and answer whether these models are representative of the clinical inflammatory stages following knee trauma. Moreover, we compare the pro-inflammatory cytokine concentrations used in such models and demonstrate that, compared to concentrations in the synovial fluid after knee trauma, they are exceedingly high. We then used the Bradford Hill Framework to present evidence that TNF-α and IL-6 cytokines are causal factors, while IL-1β and IL-17 are credible factors in inducing knee PTOA disease progresssion. Lastly, we discuss beneficial infrastructure for future studies to dissect the role of local vs. systemic inflammation in PTOA progression with an emphasis on early disease.
Collapse
Affiliation(s)
| | | | | | | | - Melanie L. Hart
- G.E.R.N. Center for Tissue Replacement, Regeneration & Neogenesis, Department of Orthopedics and Trauma Surgery, Faculty of Medicine, Medical Center—Albert-Ludwigs-University of Freiburg, 79085 Freiburg im Breisgau, Germany; (C.M.K.); (R.A.); (J.M.H.); (B.R.)
| |
Collapse
|
364
|
Koo HK, Song P, Lee JH. Novel association between asthma and osteoarthritis: a nationwide health and nutrition examination survey. BMC Pulm Med 2021; 21:59. [PMID: 33593334 PMCID: PMC7885236 DOI: 10.1186/s12890-021-01425-6] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2020] [Accepted: 02/05/2021] [Indexed: 02/07/2023] Open
Abstract
Background Asthma and osteoarthritis (OA) are medical conditions that inhibit physical activity and adversely affect quality of life. Despite the high prevalence, there are limited studies focusing on the comorbid condition and association between asthma and OA. The aim of this study was to assess the prevalence of OA co-occurring with asthma and to identify the relevant clinical considerations. Methods Adult participants aged over 40 years who completed questionnaire assessments and spirometry tests were enrolled from the Korean National Health and Nutrition Examination Survey. Asthma and OA were defined based on the medical history of a diagnosis made by a doctor. Radiographic severities of OA were measured using the Kellgren–Lawrence grading system. Chronic obstructive pulmonary disease (COPD), as a comparative respiratory disease, was diagnosed based on the spirometric results. Results A total of 9344 subjects were enrolled, and the prevalence of asthma and COPD were 4.6% ± 0.3% and 12.0% ± 0.5%, respectively. The prevalence of OA in the asthma group was 31.9% ± 2.8%, which was significantly higher than that in the COPD (17.8% ± 1.5%) or control (16.2% ± 0.6%) groups. OA was more prevalent in patients with asthma after adjusting for age, sex, body mass index, and smoking status (OR 1.65; 95% CI 1.27–2.13). Furthermore, after adjustment of this model for the prescription of OA medication, OA remained independently associated with asthma (OR 1.56; 95% CI 1.10–2.20). Conversely, the relationship of OA medication with asthma was not significant (P = 0.64). This relationship was evident in patients with asthma without airflow limitation measured by spirometry (OR 1.97; 95% CI 1.32–2.93). Moreover, the radiographic severity of knee OA correlated with asthma (OR 1.10; 95% CI 1.0–1.21). Conclusions OA shows a high prevalence in patients with asthma, higher than in patients with COPD or the controls. The comorbid characteristics of these two conditions need to be considered in clinical practice.
Collapse
Affiliation(s)
- Hyeon-Kyoung Koo
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Inje University Ilsan Paik Hospital, Goyang, Korea
| | - Pamela Song
- Department of Neurology, Inje University Ilsan Paik Hospital, Goyang, Korea
| | - Joo-Hyun Lee
- Division of Rheumatology, Department of Internal Medicine, Inje University Ilsan Paik Hospital, Goyang, 10380, Korea.
| |
Collapse
|
365
|
Molecular and Cellular Effects of Chemical Chaperone-TUDCA on ER-Stressed NHAC-kn Human Articular Chondrocytes Cultured in Normoxic and Hypoxic Conditions. Molecules 2021; 26:molecules26040878. [PMID: 33562298 PMCID: PMC7915106 DOI: 10.3390/molecules26040878] [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: 12/31/2020] [Revised: 02/01/2021] [Accepted: 02/05/2021] [Indexed: 12/28/2022] Open
Abstract
Osteoarthritis (OA) is considered one of the most common arthritic diseases characterized by progressive degradation and abnormal remodeling of articular cartilage. Potential therapeutics for OA aim at restoring proper chondrocyte functioning and inhibiting apoptosis. Previous studies have demonstrated that tauroursodeoxycholic acid (TUDCA) showed anti-inflammatory and anti-apoptotic activity in many models of various diseases, acting mainly via alleviation of endoplasmic reticulum (ER) stress. However, little is known about cytoprotective effects of TUDCA on chondrocyte cells. The present study was designed to evaluate potential effects of TUDCA on interleukin-1β (IL-1β) and tunicamycin (TNC)-stimulated NHAC-kn chondrocytes cultured in normoxic and hypoxic conditions. Our results showed that TUDCA alleviated ER stress in TNC-treated chondrocytes, as demonstrated by reduced CHOP expression; however, it was not effective enough to prevent apoptosis of NHAC-kn cells in either normoxia nor hypoxia. However, co-treatment with TUDCA alleviated inflammatory response induced by IL-1β, as shown by down regulation of Il-1β, Il-6, Il-8 and Cox2, and increased the expression of antioxidant enzyme Sod2. Additionally, TUDCA enhanced Col IIα expression in IL-1β- and TNC-stimulated cells, but only in normoxic conditions. Altogether, these results suggest that although TUDCA may display chondoprotective potential in ER-stressed cells, further analyses are still necessary to fully confirm its possible recommendation as potential candidate in OA therapy.
Collapse
|
366
|
Rahmadian R, Adly M, Dilogo IH, Revilla G. Clinical Application Prospect of Human Synovial Tissue Stem Cells from Osteoarthritis Grade IV Patients in Cartilage Regeneration. Open Access Maced J Med Sci 2021. [DOI: 10.3889/oamjms.2021.5627] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
Osteoarthritis (OA) is a joint problem that continues to increase in prevalence as life expectancy increases. OA can affect any joint, especially those that support body weight such as the knee and hip joint. Although both primary and secondary OA have the same clinical symptoms, it can be caused by different etiologies. OA is no longer considered a degenerative disease, although age is still a major factor. Various attempts have been made to regenerate joint cartilage damaged by OA. The use of stem cells in OA therapy is a very promising opportunity. Stem cells are undifferentiated biological cells and are multipotent to differentiate into specific cells. In principle, local stem cells are the best source of stem cells to regenerate the surrounding tissue. The synovial membrane is a tissue in the joint that can regenerate. After synovectomy surgery, repair, and growth of synovial tissue occur rapidly. Synovial tissue as a source of stem cells only provides a limited amount. One source of synovial tissue that can be used is tissue taken from the total knee replacement process in grade 4 OA patients. However, it is necessary to prove the potential of synovial tissue stem cells originating from old-age donors.
Collapse
|
367
|
Bollmann M, Pinno K, Ehnold LI, Märtens N, Märtson A, Pap T, Stärke C, Lohmann CH, Bertrand J. MMP-9 mediated Syndecan-4 shedding correlates with osteoarthritis severity. Osteoarthritis Cartilage 2021; 29:280-289. [PMID: 33246160 DOI: 10.1016/j.joca.2020.10.009] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Revised: 09/25/2020] [Accepted: 10/13/2020] [Indexed: 02/02/2023]
Abstract
OBJECTIVE Osteoarthritis (OA) is a degenerative joint disease inducing the degradation of the articular cartilage. Syndecan-4 (Sdc4) is a heparan sulfate proteoglycan, expressed under inflammatory conditions and by chondrocytes during OA. Little is known about Sdc4 shedding and its regulation in OA. Therefore, we investigated the regulation of Sdc4 shedding and underlying shedding mechanisms under OA conditions. DESIGN Articular cartilage, serum, synovial fluid and synovial membrane from OA patients with different radiological severity were analyzed. ELISA, RT-qPCR and IHC for Sdc4, MMP-2 and -9 were performed. MMP inhibitors and siRNA were evaluated for their effect on Sdc4 shedding by ELISA and on IL-1 signaling by western blot (pERK/ERK). RESULTS Shed Sdc4 was increased in synovial fluid of OA patients, but not in the serum and is a good predictor (AUC = 0.72) for OA severity with a sensitivity of 67.5% and specificity 65.2%. MMP-9, but not MMP-2, was increased in cartilage and synovial membrane at mRNA levels and in the synovial fluid at protein levels. Shed Sdc4 correlated with the amount of MMP-9 in synovial fluid. Further, the inhibition and knock-down of MMP-9 decreased the amount of shed Sdc4 in vitro. Increased Sdc4 shedding resulted in less phosphorylation of ERK upon IL-1β stimulation. CONCLUSION Shed Sdc4 might be a good prognostic biomarker for OA mediated cartilage degradation. MMP-9 seems to be the relevant sheddase for Sdc4 under OA conditions, desensitizing chondrocytes towards IL-1 signaling.
Collapse
Affiliation(s)
- M Bollmann
- Department of Orthopaedic Surgery, Otto-von-Guericke University, Magdeburg, Germany
| | - K Pinno
- Department of Orthopaedic Surgery, Otto-von-Guericke University, Magdeburg, Germany
| | - L I Ehnold
- Department of Orthopaedic Surgery, Otto-von-Guericke University, Magdeburg, Germany
| | - N Märtens
- Department of Orthopaedic Surgery, Otto-von-Guericke University, Magdeburg, Germany
| | - A Märtson
- Department of Traumatology and Orthopedics, University of Tartu, Tartu University Hospital, Tartu, Estonia
| | - T Pap
- Institute of Musculoskeletal Medicine, University Hospital Münster, Münster, Germany
| | - C Stärke
- Department of Orthopaedic Surgery, Otto-von-Guericke University, Magdeburg, Germany
| | - C H Lohmann
- Department of Orthopaedic Surgery, Otto-von-Guericke University, Magdeburg, Germany
| | - J Bertrand
- Department of Orthopaedic Surgery, Otto-von-Guericke University, Magdeburg, Germany.
| |
Collapse
|
368
|
Makarczyk MJ, Gao Q, He Y, Li Z, Gold MS, Hochberg MC, Bunnell BA, Tuan RS, Goodman SB, Lin H. Current Models for Development of Disease-Modifying Osteoarthritis Drugs. Tissue Eng Part C Methods 2021; 27:124-138. [PMID: 33403944 PMCID: PMC8098772 DOI: 10.1089/ten.tec.2020.0309] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2020] [Accepted: 12/18/2020] [Indexed: 12/12/2022] Open
Abstract
Osteoarthritis (OA) is a painful and disabling disease that affects millions of people worldwide. Symptom-alleviating treatments exist, although none with long-term efficacy. Furthermore, there are currently no disease-modifying OA drugs (DMOADs) with demonstrated efficacy in OA patients, which is, in part, attributed to a lack of full understanding of the pathogenesis of OA. The inability to translate findings from basic research to clinical applications also highlights the deficiencies in the available OA models at simulating the clinically relevant pathologies and responses to treatments in humans. In this review, the current status in the development of DMOADs will be first presented, with special attention to those in Phase II-IV clinical trials. Next, current in vitro, ex vivo, and in vivo OA models are summarized and the respective advantages and disadvantages of each are highlighted. Of note, the development and application of microphysiological or tissue-on-a-chip systems for modeling OA in humans are presented and the issues that need to be addressed in the future are discussed. Microphysiological systems should be given serious consideration for their inclusion in the DMOAD development pipeline, both for their ability to predict drug safety and efficacy in human clinical trials at present, as well as for their potential to serve as a test platform for personalized medicine. Impact statement At present, no disease-modifying osteoarthritis (OA) drugs (DMOADs) have been approved for widespread clinical use by regulatory bodies. The failure of developing effective DMOADs is likely owing to multiple factors, not the least of which are the intrinsic differences between the intact human knee joint and the preclinical models. This work summarizes the current OA models for the development of DMOADs, discusses the advantages/disadvantages of each, and then proposes future model development to aid in the discovery of effective and personalized DMOADs. The review also highlights the microphysiological systems, which are emerging as a new platform for drug development.
Collapse
Affiliation(s)
- Meagan J. Makarczyk
- Department of Orthopaedic Surgery, Center for Cellular and Molecular Engineering, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Department of Bioengineering, Swanson School of Engineering, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Qi Gao
- Department of Orthopaedic Surgery, Stanford University, California, USA
| | - Yuchen He
- Department of Orthopaedic Surgery, Center for Cellular and Molecular Engineering, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Zhong Li
- Department of Orthopaedic Surgery, Center for Cellular and Molecular Engineering, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Michael S. Gold
- Department of Neurobiology, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Mark C. Hochberg
- Department of Medicine and Epidemiology and Public Health, University of Maryland, Baltimore, Maryland, USA
| | - Bruce A. Bunnell
- Department of Microbiology, Immunology, and Genetics, University of North Texas Health Science Center, Fort Worth, Texas, USA
| | - Rocky S. Tuan
- Institute for Tissue Engineering and Regenerative Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Stuart B. Goodman
- Department of Orthopaedic Surgery, Stanford University, California, USA
- Department of Bioengineering, Stanford University, California, USA
| | - Hang Lin
- Department of Orthopaedic Surgery, Center for Cellular and Molecular Engineering, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Department of Bioengineering, Swanson School of Engineering, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- McGowan Institute for Regenerative Medicine, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
369
|
A molecular docking study: Cepharanthine protects articular cartilage against arthritis by Wnt/PI3K/TLR-3 signaling. ARAB J CHEM 2021. [DOI: 10.1016/j.arabjc.2020.10.030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
|
370
|
Abstract
Supplemental Digital Content is available in the text Osteoarthritis is a debilitating disease leading to joint degeneration, inflammation, pain, and disability. Despite efforts to develop a disease modifying treatment, the only accepted and available clinical approaches involve palliation. Although many factors contribute to the development of osteoarthritis, the gut microbiome has recently emerged as an important pathogenic factor in osteoarthritis initiation and progression. This review examines the literature to date regarding the link between the gut microbiome and osteoarthritis.
Collapse
|
371
|
Ren G, Al-Jezani N, Railton P, Powell JN, Krawetz RJ. CCL22 induces pro-inflammatory changes in fibroblast-like synoviocytes. iScience 2021; 24:101943. [PMID: 33490888 PMCID: PMC7809191 DOI: 10.1016/j.isci.2020.101943] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Revised: 11/18/2020] [Accepted: 12/10/2020] [Indexed: 01/23/2023] Open
Abstract
Synovitis is common in patients with osteoarthritis (OA) and is associated with pain and disease progression. We have previously demonstrated that the chemokine C-C motif chemokine 22 (CCL22) induces chondrocyte apoptosis in vitro; however, the effects of CCL22 on the synovium remain unknown. Therefore, our goal was to investigate the effect of CCL22 on fibroblast-like synoviocytes (FLS). CCL22 treatment suppressed expression of IL-4 and IL-10 and promoted expression of S100A12 in FLS. The response of FLS to CCL22 was not dependent on the disease state of the joint (e.g., normal versus OA), but was instead correlated with the individuals' synovial fluid level of CCL22. CCL22 induction of S100A12 in FLS was attenuated after knockdown of CCR3, yet ligands of CCR3 (CCL7, CCL11) did not induce S100A12 expression. In the presence of CCL22, CCR3-positive FLS upregulate CCL22 and S100A12 driving a potential feedforward pro-inflammatory mechanism distinct from canonical CCL22 and CCR3 pathways.
Collapse
Affiliation(s)
- Guomin Ren
- McCaig Institute for Bone & Joint Health, Faculty of Medicine, University of Calgary, 3330 Hospital Drive NW, Calgary, AB T2N 4N1, Canada
- University of Calgary, Biomedical Engineering Graduate Program, Calgary, AB T2N 4N1, Canada
| | - Nedaa Al-Jezani
- McCaig Institute for Bone & Joint Health, Faculty of Medicine, University of Calgary, 3330 Hospital Drive NW, Calgary, AB T2N 4N1, Canada
| | - Pamela Railton
- Charles Sturt University, School of Biomedical Science, Wagga Wagga, NSW 2650, Australia
| | - James N. Powell
- McCaig Institute for Bone & Joint Health, Faculty of Medicine, University of Calgary, 3330 Hospital Drive NW, Calgary, AB T2N 4N1, Canada
- University of Calgary, Department of Surgery, Calgary, AB T2N 4N1, Canada
| | - Roman J. Krawetz
- McCaig Institute for Bone & Joint Health, Faculty of Medicine, University of Calgary, 3330 Hospital Drive NW, Calgary, AB T2N 4N1, Canada
- University of Calgary, Biomedical Engineering Graduate Program, Calgary, AB T2N 4N1, Canada
- University of Calgary, Department of Surgery, Calgary, AB T2N 4N1, Canada
- University of Calgary, Department of Anatomy and Cell Biology, Calgary, AB T2N 4N1, Canada
| |
Collapse
|
372
|
Laplace-Builhé B, Bahraoui S, Jorgensen C, Djouad F. From the Basis of Epimorphic Regeneration to Enhanced Regenerative Therapies. Front Cell Dev Biol 2021; 8:605120. [PMID: 33585444 PMCID: PMC7873919 DOI: 10.3389/fcell.2020.605120] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2020] [Accepted: 12/14/2020] [Indexed: 01/01/2023] Open
Abstract
Current cell-based therapies to treat degenerative diseases such as osteoarthritis (OA) fail to offer long-term beneficial effects. The therapeutic effects provided by mesenchymal stem cell (MSC) injection, characterized by reduced pain and an improved functional activity in patients with knee OA, are reported at short-term follow-up since the improved outcomes plateau or, even worse, decline several months after MSC administration. This review tackles the limitations of MSC-based therapy for degenerative diseases and highlights the lessons learned from regenerative species to comprehend the coordination of molecular and cellular events critical for complex regeneration processes. We discuss how MSC injection generates a positive cascade of events resulting in a long-lasting systemic immune regulation with limited beneficial effects on tissue regeneration while in regenerative species fine-tuned inflammation is required for progenitor cell proliferation, differentiation, and regeneration. Finally, we stress the direct or indirect involvement of neural crest derived cells (NCC) in most if not all adult regenerative models studied so far. This review underlines the regenerative potential of NCC and the limitations of MSC-based therapy to open new avenues for the treatment of degenerative diseases such as OA.
Collapse
Affiliation(s)
| | | | - Christian Jorgensen
- IRMB, Univ Montpellier, INSERM, Montpellier, France.,CHU Montpellier, Montpellier, France
| | | |
Collapse
|
373
|
Regenerative Medicine for Equine Musculoskeletal Diseases. Animals (Basel) 2021; 11:ani11010234. [PMID: 33477808 PMCID: PMC7832834 DOI: 10.3390/ani11010234] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Revised: 01/08/2021] [Accepted: 01/15/2021] [Indexed: 01/15/2023] Open
Abstract
Simple Summary Lameness due to musculoskeletal disease is the most common diagnosis in equine veterinary practice. Many of these orthopaedic disorders are chronic problems, for which no clinically satisfactory treatment exists. Thus, high hopes are pinned on regenerative medicine, which aims to replace or regenerate cells, tissues, or organs to restore or establish normal function. Some regenerative medicine therapies have already made their way into equine clinical practice mainly to treat tendon injures, tendinopathies, cartilage injuries and degenerative joint disorders with promising but diverse results. This review summarises the current knowledge of commonly used regenerative medicine treatments and critically discusses their use. Abstract Musculoskeletal injuries and chronic degenerative diseases commonly affect both athletic and sedentary horses and can entail the end of their athletic careers. The ensuing repair processes frequently do not yield fully functional regeneration of the injured tissues but biomechanically inferior scar or replacement tissue, causing high reinjury rates, degenerative disease progression and chronic morbidity. Regenerative medicine is an emerging, rapidly evolving branch of translational medicine that aims to replace or regenerate cells, tissues, or organs to restore or establish normal function. It includes tissue engineering but also cell-based and cell-free stimulation of endogenous self-repair mechanisms. Some regenerative medicine therapies have made their way into equine clinical practice mainly to treat tendon injures, tendinopathies, cartilage injuries and degenerative joint disorders with promising results. However, the qualitative and quantitative spatiotemporal requirements for specific bioactive factors to trigger tissue regeneration in the injury response are still unknown, and consequently, therapeutic approaches and treatment results are diverse. To exploit the full potential of this burgeoning field of medicine, further research will be required and is ongoing. This review summarises the current knowledge of commonly used regenerative medicine treatments in equine patients and critically discusses their use.
Collapse
|
374
|
Lambova SN. Osteoarthritis - From New Insights into Disease Pathogenesis to Contemporary Personalized Therapeutic Strategy. Curr Rheumatol Rev 2021; 17:4-6. [PMID: 33423648 DOI: 10.2174/1573397116666210108095700] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Affiliation(s)
- Sevdalina N Lambova
- Department of Propaedeutics of Internal Diseases, Faculty of Medicine, Medical University - Plovdiv, Plovdiv, Bulgaria
| |
Collapse
|
375
|
Cartilage Targets of Knee Osteoarthritis Shared by Both Genders. Int J Mol Sci 2021; 22:ijms22020569. [PMID: 33430025 PMCID: PMC7827374 DOI: 10.3390/ijms22020569] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Revised: 12/31/2020] [Accepted: 01/05/2021] [Indexed: 12/13/2022] Open
Abstract
As the leading cause of disability, osteoarthritis (OA) affects people of all ages, sexes, and races. With the increasing understanding of OA, the sex differences have attracted specific attention as the burden of OA is greater in women. There is no doubt that gender-specific OA management has great potential for precision treatment. On the other hand, from the marketing aspect, a medication targeting the OA-responsive biomarker(s) shared by both genders is more favorable for drug development. Thus, in the current study, a published transcriptome dataset of knee articular cartilage was used to compare OA and healthy samples for identifying the genes with the same significantly different expression trend in both males and females. With 128 genes upregulated and 143 genes downregulated in both OA males and females, 9 KEGG pathways have been enriched based on the current knowledge, including 'renal cell carcinoma,' 'ECM-receptor interaction,' 'HIF-1 signaling pathway,' 'MicroRNAs in cancer,' 'focal adhesion,' 'Relaxin signaling pathway,' 'breast cancer,' 'PI3K-Akt signaling pathway,' and 'human papillomavirus infection.' Here, we explore the potential impacts of these clusters in OA. We also analyze the identified 'cell plasma membrane related genes' in-depth to identify the potential chondrocyte cell surface target(s) of OA management.
Collapse
|
376
|
Kouroupis D, Willman MA, Best TM, Kaplan LD, Correa D. Infrapatellar fat pad-derived mesenchymal stem cell-based spheroids enhance their therapeutic efficacy to reverse synovitis and fat pad fibrosis. Stem Cell Res Ther 2021; 12:44. [PMID: 33413649 PMCID: PMC7792122 DOI: 10.1186/s13287-020-02107-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2020] [Accepted: 12/15/2020] [Indexed: 12/13/2022] Open
Abstract
Background To investigate the in vitro and in vivo anti-inflammatory/anti-fibrotic capacity of IFP-MSC manufactured as 3D spheroids. Our hypothesis is that IFP-MSC do not require prior cell priming to acquire a robust immunomodulatory phenotype in vitro in order to efficiently reverse synovitis and IFP fibrosis, and secondarily delay articular cartilage damage in vivo. Methods Human IFP-MSC immunophenotype, tripotentiality, and transcriptional profiles were assessed in 3D settings. Multiplex secretomes were assessed in IFP-MSC spheroids [Crude (non-immunoselected), CD146+ or CD146− immunoselected cells] and compared with 2D cultures with and without prior inflammatory/fibrotic cell priming. Functionally, IFP-MSC spheroids were assessed for their immunopotency on human PBMC proliferation and their effect on stimulated synoviocytes with inflammation and fibrotic cues. The anti-inflammatory and anti-fibrotic spheroid properties were further evaluated in vivo in a rat model of acute synovitis/fat pad fibrosis. Results Spheroids enhanced IFP-MSC phenotypic, transcriptional, and secretory immunomodulatory profiles compared to 2D cultures. Further, CD146+ IFP-MSC spheroids showed enhanced secretory and transcriptional profiles; however, these attributes were not reflected in a superior capacity to suppress activated PBMC. This suggests that 3D culturing settings are sufficient to induce an enhanced immunomodulatory phenotype in both Crude and CD146-immunoselected IFP-MSC. Crude IFP-MSC spheroids modulated the molecular response of synoviocytes previously exposed to inflammatory cues. Therapeutically, IFP-MSC spheroids retained substance P degradation potential in vivo, while effectively inducing resolution of inflammation/fibrosis of the synovium and fat pad. Furthermore, their presence resulted in arrest of articular cartilage degradation in a rat model of progressive synovitis and fat pad fibrosis. Conclusions 3D spheroids confer IFP-MSC a reproducible and enhanced immunomodulatory effect in vitro and in vivo, circumventing the requirement of non-compliant cell priming or selection before administration and thereby streamlining cell products manufacturing protocols.
Collapse
Affiliation(s)
- Dimitrios Kouroupis
- Department of Orthopedics, UHealth Sports Medicine Institute, University of Miami, Miller School of Medicine, 1450 NW 10th Ave (3014), Miami, FL, 33136, USA.,Diabetes Research Institute & Cell Transplantation Center, University of Miami, Miller School of Medicine, 1450 NW 10th Ave (3014), Miami, FL, 33136, USA
| | - Melissa A Willman
- Diabetes Research Institute & Cell Transplantation Center, University of Miami, Miller School of Medicine, 1450 NW 10th Ave (3014), Miami, FL, 33136, USA
| | - Thomas M Best
- Department of Orthopedics, UHealth Sports Medicine Institute, University of Miami, Miller School of Medicine, 1450 NW 10th Ave (3014), Miami, FL, 33136, USA
| | - Lee D Kaplan
- Department of Orthopedics, UHealth Sports Medicine Institute, University of Miami, Miller School of Medicine, 1450 NW 10th Ave (3014), Miami, FL, 33136, USA
| | - Diego Correa
- Department of Orthopedics, UHealth Sports Medicine Institute, University of Miami, Miller School of Medicine, 1450 NW 10th Ave (3014), Miami, FL, 33136, USA. .,Diabetes Research Institute & Cell Transplantation Center, University of Miami, Miller School of Medicine, 1450 NW 10th Ave (3014), Miami, FL, 33136, USA.
| |
Collapse
|
377
|
Koyama T, Uchida K, Fukushima K, Ohashi Y, Uchiyama K, Inoue G, Takahira N, Takaso M. Elevated levels of TNF-α, IL-1β and IL-6 in the synovial tissue of patients with labral tear: a comparative study with hip osteoarthritis. BMC Musculoskelet Disord 2021; 22:33. [PMID: 33407301 PMCID: PMC7788943 DOI: 10.1186/s12891-020-03888-w] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/22/2020] [Accepted: 12/16/2020] [Indexed: 01/15/2023] Open
Abstract
BACKGROUND Labral tear can be the initiating factor in the onset of hip osteoarthritis (HOA). However, the physiopathology of labral tear is not fully understood. Our aim was to compare synovial tissue inflammatory cytokine levels between patients with labral tear and late-stage HOA. METHODS Synovial tissue from sites showing the greatest inflammation was harvested from 106 hips from 100 subjects during hip surgery. RNA was extracted, and levels of TNFA, IL1B, IL6 and COX2 mRNA were compared among all patients using real-time PCR. Additionally, we examined whether femoroacetabular impingement (FAI) was associated with elevated levels of inflammatory cytokines in patients with labral tear. To analyze the effects of TNF-α on inflammatory mediators in hip synovial tissue, synovial fibroblasts were extracted from hip synovial tissue of patients with labral tear and late-stage HOA (n = 5 each). Mononuclear cells were extracted from synovial tissue, cultured for 7 days, and stimulated with control or 10 ng/mL human recombinant TNF-α for 1 day. mRNA was extracted from stimulated cells and IL1B, IL6, and COX2 levels were determined using real-time PCR. RESULTS TNFA, IL1B, and COX2 expression in synovial tissue were significantly higher in patients with labral tear than late-stage HOA (TNFA, p < 0.001; IL1B, p < 0.001; COX2, p = 0.001). There were no differences in expression between patients with labral tear with and without FAI (TNFA, p = 0.546; IL1B, p = 0.559; IL6, p = 0.599; COX2, p = 0.124). Compared to vehicle control, TNF-α stimulation significantly elevated IL1B, IL6, and COX2 expression in synovial fibroblasts collected from patients with labral tear and late-stage HOA (IL1B, p = 0.043 and p = 0.043; IL6, p = 0.043 and 0.043; COX2, p = 0.043 and p = 0.080, respectively). CONCLUSIONS TNFA, IL1B, and COX2 expression were elevated in the synovial tissue of patients with labral tear. Further investigations are needed to reveal the relationship between inflammatory cytokine levels and various aspects of labral tear pathology, including pain and the onset and progression of OA.
Collapse
Affiliation(s)
- Tomohisa Koyama
- Department of Orthopedic Surgery, Kitasato University School of Medicine, 1-15-1 Minami-ku, Kitasato, Sagamihara City, Kanagawa, 252-0374, Japan
| | - Kentaro Uchida
- Department of Orthopedic Surgery, Kitasato University School of Medicine, 1-15-1 Minami-ku, Kitasato, Sagamihara City, Kanagawa, 252-0374, Japan.
| | - Kensuke Fukushima
- Department of Orthopedic Surgery, Kitasato University School of Medicine, 1-15-1 Minami-ku, Kitasato, Sagamihara City, Kanagawa, 252-0374, Japan
| | - Yoshihisa Ohashi
- Department of Orthopedic Surgery, Kitasato University School of Medicine, 1-15-1 Minami-ku, Kitasato, Sagamihara City, Kanagawa, 252-0374, Japan
| | - Katsufumi Uchiyama
- Department of Orthopedic Surgery, Kitasato University School of Medicine, 1-15-1 Minami-ku, Kitasato, Sagamihara City, Kanagawa, 252-0374, Japan
| | - Gen Inoue
- Department of Orthopedic Surgery, Kitasato University School of Medicine, 1-15-1 Minami-ku, Kitasato, Sagamihara City, Kanagawa, 252-0374, Japan
| | - Naonobu Takahira
- Department of Rehabilitation, Kitasato University School of Allied Health Sciences, 1-15-1 Minami-ku, Kitasato, Sagamihara City, Kanagawa, 252-0374, Japan
| | - Masashi Takaso
- Department of Orthopedic Surgery, Kitasato University School of Medicine, 1-15-1 Minami-ku, Kitasato, Sagamihara City, Kanagawa, 252-0374, Japan
| |
Collapse
|
378
|
Zhou B, Li LH, Tan LM, Luo WB, Xiong H, Lu XL, Liu D, Li WY, Guo YX, Tang Z, Zhu LG. Tanshinone IIA Ameliorates Inflammation Response in Osteoarthritis via Inhibition of miR-155/FOXO3 Axis. Pharmacology 2021; 106:20-28. [PMID: 33395681 DOI: 10.1159/000505493] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2019] [Accepted: 12/12/2019] [Indexed: 11/19/2022]
Abstract
BACKGROUND Osteoarthritis (OA) is the most common joint disorder characterized by degeneration of the articular cartilage and joint destruction with an associated risk of mobility disability in elderly people. Although a lot of achievements have been made, OA is still regarded as an incurable disease. Therefore, the pathological mechanisms and novel therapeutic strategies of OA need more investigation. METHODS MTT assay was conducted to measure the viability of chondrocytes after LPS treatment. Cell apoptosis was analyzed by annexin V/propidium iodide labeling. ELISA was used to determine the concentrations of interleukin (IL)-1β, IL-6, and tumor necrosis factor (TNF)-α in the culture supernatant of chondrocytes. The expression level of miR-155, IL-1β, FOXO3, TNF-α, IL-6, caspase-3, and caspase-9 in chondrocytes was analyzed by RT-qPCR or Western blot. RESULTS We found that LPS led to inflammatory responses, cell apoptosis, and increased miR-155 expression in human articular chondrocytes. Tanshinone IIA could inhibit LPS-induced inflammation and cell apoptosis of chondrocytes via regulating the expression of miR-155 and FOXO3. miR-155 directly targeted the 3'-UTR of FOXO3 to regulate its expression. CONCLUSIONS Taken together, our data suggest tanshinone IIA ameliorates inflammation response in OA via inhibition of the miR-155/FOXO3 axis, and provide some evidences that tanshinone IIA could be designed and developed as a new promising clinical therapeutic drug for OA patients.
Collapse
Affiliation(s)
- Biao Zhou
- Department of Orthopedics, Wangjing Hospital of Chinese Academy of Chinese Medical Science, Beijing, China.,Department of Orthopedics, Xiangtan Hospital Affiliated to Nanhua University, Xiangtan, China
| | - Lin-Hui Li
- Department of Orthopedics, Wangjing Hospital of Chinese Academy of Chinese Medical Science, Beijing, China
| | - Li-Ming Tan
- Department of Orthopedics, The Fourth Hospital of Changsha, Changsha, China.,Department of Orthopedics, Changsha Hospital of Tranditional Chinese Medicine, Changsha, China
| | - Wen-Bing Luo
- Department of Orthopedics, The Chinese Medicine Hospital of Linli County, Linli, China.,Department of Orthopedics, Hunan University of Chinese Medicine, Changsha, China
| | - Hui Xiong
- Department of Orthopedics, Hunan University of Chinese Medicine, Changsha, China
| | - Xiao-Long Lu
- Department of Orthopedics, The Second Affiliated Hospital of Hunan University of Chinese Medicine, Changsha, China
| | - Dan Liu
- Department of Rheumatology, The First Hospital of Hunan University of Chinese Medicine, Changsha, China
| | - Wang-Yang Li
- Department of Orthopedics, Hunan University of Chinese Medicine, Changsha, China
| | - Yu-Xing Guo
- Department of Orthopedics, Hunan University of Chinese Medicine, Changsha, China
| | - Zhi Tang
- Department of Orthopedics, Hunan University of Chinese Medicine, Changsha, China
| | - Li-Guo Zhu
- Department of Orthopedics, Wangjing Hospital of Chinese Academy of Chinese Medical Science, Beijing, China,
| |
Collapse
|
379
|
Hudek R, Brobeil A, Brüggemann H, Sommer F, Gattenlöhner S, Gohlke F. Cutibacterium acnes is an intracellular and intra-articular commensal of the human shoulder joint. J Shoulder Elbow Surg 2021; 30:16-26. [PMID: 32741563 DOI: 10.1016/j.jse.2020.04.020] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/27/2019] [Revised: 04/03/2020] [Accepted: 04/12/2020] [Indexed: 02/01/2023]
Abstract
BACKGROUND Cutibacterium acnes (C acnes) is a mysterious member of the shoulder microbiome and is associated with chronic postoperative complications and low-grade infections. Nevertheless, it is unclear whether it represents a contaminant or whether it accounts for true infections. Because it can persist intracellularly in macrophages at several body sites, it might in fact be an intra-articular commensal of the shoulder joint. METHODS In 23 consecutive, otherwise healthy patients (17 male, 6 female; 58 years) who had no previous injections, multiple specimens were taken from the intra-articular tissue during first-time arthroscopic and open shoulder surgery. The samples were investigated by cultivation, genetic phylotyping, and immunohistochemistry using C acnes-specific antibodies and confocal laser scanning microscopy. RESULTS In 10 patients (43.5%), cultures were C acnes-positive. Phylotype IA1 dominated the subcutaneous samples (71%), whereas type II dominated the deep tissue samples (57%). Sixteen of 23 patients (69.6%) were C acnes-positive by immunohistochemistry; in total, 25 of 40 samples were positive (62.5%). Overall, 56.3% of glenohumeral immunohistochemical samples, 62.5% of subacromial samples, and 75% of acromioclavicular (AC) joint samples were positive. In 62.5% of the tested patients, C acnes was detected immunohistochemically to reside intracellularly within stromal cells and macrophages. DISCUSSION These data indicate that C acnes is a commensal of the human shoulder joint, where it persists within macrophages and stromal cells. Compared with culture-based methods, immunohistochemical staining can increase C acnes detection. Phylotype II seems to be most prevalent in the deep shoulder tissue. The high detection rate of C acnes in osteoarthritic AC joints might link its intra-articular presence to the initiation of osteoarthritis.
Collapse
Affiliation(s)
- Robert Hudek
- Rhön-Klinikum Campus Bad Neustadt, Department for Shoulder and Elbow Surgery, Bad Neustadt a. d. Saale, Germany.
| | - Alexander Brobeil
- Justus-Liebig-University Gießen, Institute for Pathology, Gießen, Germany
| | | | - Frank Sommer
- Phillipps-University Marburg, Institute for Medical Microbiology and Hospital Hygiene, Marburg, Germany
| | | | - Frank Gohlke
- Rhön-Klinikum Campus Bad Neustadt, Department for Shoulder and Elbow Surgery, Bad Neustadt a. d. Saale, Germany
| |
Collapse
|
380
|
Park S, Moon BR, Kim JE, Kim HJ, Zhang T. Aqueous Extracts of Morus alba Root Bark and Cornus officinalis Fruit Protect against Osteoarthritis Symptoms in Testosterone-Deficient and Osteoarthritis-Induced Rats. Pharmaceutics 2020; 12:pharmaceutics12121245. [PMID: 33371279 PMCID: PMC7767081 DOI: 10.3390/pharmaceutics12121245] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2020] [Revised: 12/16/2020] [Accepted: 12/18/2020] [Indexed: 12/16/2022] Open
Abstract
Water extracts of both Morus alba L. root bark (MBW) and Cornus officinalis Siebold and Zucc fruit (CFW) have traditionally been used to promote men's health in the elderly in Asia. We determined that the 12-week consumption of MBW and CFW could alleviate testosterone-deficiency syndrome and osteoarthritis (OA) symptoms in testosterone-deficient rats, and the action mechanisms were explored. Rats with bilateral orchiectomy (ORX) were fed a 45% fat diet containing either 0.5% MBW (ORX-MBW), 0.5% CFW(ORX-CFW), or 0.5% dextrin (ORX-CON). Sham-operated rats also received 0.5% dextrin (Non-ORX-CON). After 8 weeks of treatment, all rats had an injection of monoiodoacetate (MIA) into the left knee, and they continued the same diet for the additional 4 weeks. ORX-CFW and ORX-MBW partially prevented the reduction of serum testosterone concentrations and decreased insulin resistance, compared to the ORX-CON. ORX-CFW and ORX-MBW protected against the reduction of bone mineral density (BMD) and lean body mass (LBM) compared to the ORX-CON. The limping and edema scores were lower in the order of the ORX-CON, ORX-CRF = ORX-MBW, and Non-ORX-CON (p < 0.05). The scores for pain behaviors, measured by weight-distribution on the OA leg and maximum running velocity on a treadmill, significantly decreased in the same order as limping scores. ORX-MBW protected against the increased expression of matrix metalloproteinase (MMP)-3 and MMP-13 and reduced the production of inflammatory markers such as TNF-α and IL-1β, by MIA in the articular cartilage, compared to the ORX-CON (p < 0.05). The cartilage damage near the tidemark of the knee and proteoglycan loss was significantly less in ORX-MBW than ORX-CON. In conclusion, MBW, possibly CFW, could be effective alternative therapeutic agents for preventing osteoarthritis in testosterone-deficient elderly men.
Collapse
Affiliation(s)
- Sunmin Park
- Department Food & Nutrition, Obesity/Diabetes Center, Hoseo University, Asan 31499, Korea; (B.R.M.); (J.E.K.); (H.J.K.); (T.Z.)
- Department of Bio-Convergence System, Hoseo University, Asan 31499, Korea
- Correspondence: ; Tel.: +82-41-540-5345; Fax: +82-41-548-0670
| | - Bo Reum Moon
- Department Food & Nutrition, Obesity/Diabetes Center, Hoseo University, Asan 31499, Korea; (B.R.M.); (J.E.K.); (H.J.K.); (T.Z.)
| | - Ji Eun Kim
- Department Food & Nutrition, Obesity/Diabetes Center, Hoseo University, Asan 31499, Korea; (B.R.M.); (J.E.K.); (H.J.K.); (T.Z.)
| | - Hyun Joo Kim
- Department Food & Nutrition, Obesity/Diabetes Center, Hoseo University, Asan 31499, Korea; (B.R.M.); (J.E.K.); (H.J.K.); (T.Z.)
| | - Ting Zhang
- Department Food & Nutrition, Obesity/Diabetes Center, Hoseo University, Asan 31499, Korea; (B.R.M.); (J.E.K.); (H.J.K.); (T.Z.)
- Department of Bio-Convergence System, Hoseo University, Asan 31499, Korea
| |
Collapse
|
381
|
Araújo N, Viegas CSB, Zubía E, Magalhães J, Ramos A, Carvalho MM, Cruz H, Sousa JP, Blanco FJ, Vermeer C, Simes DC. Amentadione from the Alga Cystoseira usneoides as a Novel Osteoarthritis Protective Agent in an Ex Vivo Co-Culture OA Model. Mar Drugs 2020; 18:E624. [PMID: 33297528 PMCID: PMC7762386 DOI: 10.3390/md18120624] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2020] [Revised: 11/27/2020] [Accepted: 11/27/2020] [Indexed: 12/21/2022] Open
Abstract
Osteoarthritis (OA) remains a prevalent chronic disease without effective prevention and treatment. Amentadione (YP), a meroditerpenoid purified from the alga Cystoseira usneoides, has demonstrated anti-inflammatory activity. Here, we investigated the YP anti-osteoarthritic potential, by using a novel OA preclinical drug development pipeline designed to evaluate the anti-inflammatory and anti-mineralizing activities of potential OA-protective compounds. The workflow was based on in vitro primary cell cultures followed by human cartilage explants assays and a new OA co-culture model, combining cartilage explants with synoviocytes under interleukin-1β (IL-1β) or hydroxyapatite (HAP) stimulation. A combination of gene expression analysis and measurement of inflammatory mediators showed that the proposed model mimicked early disease stages, while YP counteracted inflammatory responses by downregulation of COX-2 and IL-6, improved cartilage homeostasis by downregulation of MMP3 and the chondrocytes hypertrophic differentiation factors Col10 and Runx2. Importantly, YP downregulated NF-κB gene expression and decreased phosphorylated IkBα/total IkBα ratio in chondrocytes. These results indicate the co-culture as a relevant pre-clinical OA model, and strongly suggest YP as a cartilage protective factor by inhibiting inflammatory, mineralizing, catabolic and differentiation processes during OA development, through inhibition of NF-κB signaling pathways, with high therapeutic potential.
Collapse
Affiliation(s)
- Nuna Araújo
- Centre of Marine Sciences (CCMAR), University of Algarve, 8005-139 Faro, Portugal; (N.A.); (C.S.B.V.)
| | - Carla S. B. Viegas
- Centre of Marine Sciences (CCMAR), University of Algarve, 8005-139 Faro, Portugal; (N.A.); (C.S.B.V.)
- GenoGla Diagnostics, Centre of Marine Sciences (CCMAR), University of Algarve, 8005-139 Faro, Portugal
| | - Eva Zubía
- Department of Organic Chemistry, Faculty of Marine and Environmental Sciences, University of Cadiz, 11510 Puerto Real (Cádiz), Spain;
| | - Joana Magalhães
- Unidad de Medicina Regenerativa, Grupo de Investigación en Reumatología (GIR), Instituto de Investigación Biomédica de A Coruña (INIBIC), Complejo Hospitalario Universitario de A Coruña (CHUAC), Sergas, 15006 A Coruña, Spain; (J.M.); (F.J.B.)
- Agrupación Estratégica CICA-INIBIC, Universidade da Coruña (UDC), 15006 A Coruña, Spain
- Centro de Investigación Biomédica en Red (CIBER), 28029 Madrid, Spain
| | - Acácio Ramos
- Department of Orthopedics and Traumatology, Hospital Particular do Algarve (HPA), 8005-226 Gambelas-Faro, Portugal; (A.R.); (M.M.C.); (H.C.); (J.P.S.)
| | - Maria M. Carvalho
- Department of Orthopedics and Traumatology, Hospital Particular do Algarve (HPA), 8005-226 Gambelas-Faro, Portugal; (A.R.); (M.M.C.); (H.C.); (J.P.S.)
| | - Henrique Cruz
- Department of Orthopedics and Traumatology, Hospital Particular do Algarve (HPA), 8005-226 Gambelas-Faro, Portugal; (A.R.); (M.M.C.); (H.C.); (J.P.S.)
| | - João Paulo Sousa
- Department of Orthopedics and Traumatology, Hospital Particular do Algarve (HPA), 8005-226 Gambelas-Faro, Portugal; (A.R.); (M.M.C.); (H.C.); (J.P.S.)
| | - Francisco J. Blanco
- Unidad de Medicina Regenerativa, Grupo de Investigación en Reumatología (GIR), Instituto de Investigación Biomédica de A Coruña (INIBIC), Complejo Hospitalario Universitario de A Coruña (CHUAC), Sergas, 15006 A Coruña, Spain; (J.M.); (F.J.B.)
- Agrupación Estratégica CICA-INIBIC, Universidade da Coruña (UDC), 15006 A Coruña, Spain
| | - Cees Vermeer
- Cardiovascular Research Institute CARIM, Maastricht University, 6229 EV Maastricht, The Netherlands;
| | - Dina C. Simes
- Centre of Marine Sciences (CCMAR), University of Algarve, 8005-139 Faro, Portugal; (N.A.); (C.S.B.V.)
- GenoGla Diagnostics, Centre of Marine Sciences (CCMAR), University of Algarve, 8005-139 Faro, Portugal
| |
Collapse
|
382
|
Zhu Z, Liao H, Liu S, Zhang J, Chen Y, Wang W. Cross-sectional study of the association between age-related macular degeneration and arthritis in the National Health and Nutrition Examination Survey 2005-2008. BMJ Open 2020; 10:e035805. [PMID: 33293303 PMCID: PMC7722807 DOI: 10.1136/bmjopen-2019-035805] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
Abstract
OBJECTIVE To explore the association between age-related macular degeneration (AMD) and arthritis in a representative sample of the US population. DESIGN Population-based, cross-sectional study. SETTING The National Health and Nutrition Examination Survey (NHANES) 2005-2008. PARTICIPANTS A total of 4813 participants aged 40 years and older with available information on AMD and arthritis in the 2005-2008 NHANES. METHODS The status and types of arthritis were obtained from questionnaires. Non-mydriatic fundus photographs were collected. The types of AMD were assessed using the modified Wisconsin Age-Related Maculopathy Grading Classification Scheme. The association between arthritis and AMD was evaluated using logistic regression models. RESULTS After adjusting for covariates, participants with any or early AMD had significantly lower odds of having any type of arthritis (any AMD: OR=0.56, 95% CI: 0.36-0.86; early AMD: OR=0.55, 95% CI: 0.34-0.88) or osteoarthritis (OA) (any AMD: OR=0.43, 95% CI: 0.26-0.71; early AMD: OR=0.44, 95% CI: 0.25-0.76) compared with those without AMD. When considering AMD as the outcome, significant negative associations were also found between any arthritis or OA and any (any arthritis: OR=0.64, 95% CI: 0.43-0.94; OA: OR=0.52, 95% CI: 0.33-0.82) or early AMD (any arthritis: OR=0.61, 95% CI: 0.40-0.93; OA: OR=0.51, 95% CI: 0.31-0.86) in the multivariable logistic models. There was no significant association between different types of arthritis and late AMD. CONCLUSIONS People with arthritis, especially those with OA, were less likely to have AMD compared with those without arthritis and vice versa. Further studies are needed to confirm this potential protective effect of arthritis and/or arthritis treatment on AMD and to explore the underlying mechanisms.
Collapse
Affiliation(s)
- Zhuoting Zhu
- Guangdong Eye Institute, Department of Ophthalmology, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Huan Liao
- Neural Regeneration Group, Institute of Reconstructive Neurobiology, University of Bonn, Bonn, Germany
| | - Sen Liu
- School of Medicine, Sun Yat-Sen University, Guangzhou, China
| | - Jian Zhang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Yifan Chen
- Medical Sciences Division, University of Oxford, Oxford, UK
| | - Wei Wang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
383
|
Examining the role of transient receptor potential canonical 5 (TRPC5) in osteoarthritis. OSTEOARTHRITIS AND CARTILAGE OPEN 2020; 2:100119. [PMID: 33381767 PMCID: PMC7762818 DOI: 10.1016/j.ocarto.2020.100119] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Accepted: 11/02/2020] [Indexed: 01/09/2023] Open
Abstract
Introduction Osteo-arthritis (OA) involves joint degradation and usually pain; with mechanisms poorly understood and few treatment options. There is evidence that the transient receptor potential canonical 5 (TRPC5) mRNA expression is reduced in OA patients’ synovia. Here we examine the profile of TRPC5 in DRG and involvement in murine models of OA. Design TRPC5 KO mice were subjected to partial meniscectomy (PMNX) or injected with monoiodoacetate (MIA) and pain-related behaviours were determined. Knee joint pathological scores were analysed and gene expression changes in ipsilateral synovium and dorsal root ganglia (DRG) determined. c-Fos protein expression in the ipsilateral dorsal horn of the spinal cord was quantified. Results TRPC5 KO mice developed a discrete enhanced pain-related phenotype. In the MIA model, the pain-related phenotype correlated with c-Fos expression in the dorsal horn and increased expression of nerve injury markers ATF3, CSF1 and galanin in the ipsilateral DRG. There were negligible differences in the joint pathology between WT and TRPC5 KO mice, however detailed gene expression analysis determined increased expression of the mast cell marker CD117 as well as extracellular matrix remodelling proteinases MMP2, MMP13 and ADAMTS4 in MIA-treated TRPC5 KO mice. TRPC5 expression was defined to sensory subpopulations in DRG. Conclusions Deletion of TRPC5 receptor signalling is associated with exacerbation of pain-like behaviour in OA which correlates with increased expression of enzymes involved in extracellular remodelling, inflammatory cells in the synovium and increased neuronal activation and injury in DRG. Together, these results identify a modulating role for TRPC5 in OA-induced pain-like behaviours.
Collapse
|
384
|
Lim HJ, Park S, Bak SG, Cheong SH, Lee S, Baek Y, Lee C, Lee KM, Lee SW, Lee S, Rho M. Beneficial effects of Vigna angularis extract in osteoporosis and osteoarthritis. Food Sci Nutr 2020; 8:6550-6556. [PMID: 33312539 PMCID: PMC7723184 DOI: 10.1002/fsn3.1944] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Accepted: 09/29/2020] [Indexed: 12/17/2022] Open
Abstract
In Asia, Vigna angularis (azuki bean) has been used as a traditional medicine to treat various diseases because of its biological properties. Osteoarthritis (OA) and osteoporosis (OP) are common regenerative bone diseases that are characterized by deterioration of joint and bone structure. In this study, we evaluated the effects of Vigna angularis extract (VAE) on monosodium iodoacetate (MIA)-induced OA and ovariectomy (OVX)-induced OP models. In the MIA-induced OA results, severe OA was alleviated by the administration of VAE. Extensive local damage in the cartilage and hemorrhagic and edematous of surrounding tissues were decreased by VAE treatment. Articular cartilage was almost intact except for a focal mild abrasion, and the surface was glistening, similar to that of the normal joint. In the OVX-induced OP results, bone mineral content (BMC) and bone mineral density (BMD) were recovered by VAE treatment, and it improved the microstructures of bone. These results show that VAE could inhibit OA and OP symptoms.
Collapse
Affiliation(s)
- Hyung Jin Lim
- Immunoregulatory Material Research CenterKorea Research Institute of Bioscience and BiotechnologyJeongeup‐siKorea
| | - Sang‐Ik Park
- College of Veterinary MedicineChonnam National UniversityGwangju‐siKorea
| | - Seon Gyeong Bak
- Immunoregulatory Material Research CenterKorea Research Institute of Bioscience and BiotechnologyJeongeup‐siKorea
- Department of Marine Bio Food ScienceChonnam National UniversityYeosu‐siKorea
| | - Sun Hee Cheong
- Department of Marine Bio Food ScienceChonnam National UniversityYeosu‐siKorea
| | - Soyoung Lee
- Immunoregulatory Material Research CenterKorea Research Institute of Bioscience and BiotechnologyJeongeup‐siKorea
| | - Young‐Bin Baek
- College of Veterinary MedicineChonnam National UniversityGwangju‐siKorea
| | - Chang‐Min Lee
- College of Veterinary MedicineChonnam National UniversityGwangju‐siKorea
| | - Kang Min Lee
- Department of Molecular BiologyChonbuk National UniversityJeonju‐siKorea
| | - Seung Woong Lee
- Immunoregulatory Material Research CenterKorea Research Institute of Bioscience and BiotechnologyJeongeup‐siKorea
| | - Seung‐Jae Lee
- Immunoregulatory Material Research CenterKorea Research Institute of Bioscience and BiotechnologyJeongeup‐siKorea
| | - Mun‐Chual Rho
- Immunoregulatory Material Research CenterKorea Research Institute of Bioscience and BiotechnologyJeongeup‐siKorea
| |
Collapse
|
385
|
Upadhyay HC, Singh M, Prakash O, Khan F, Srivastava SK, Bawankule DU. QSAR, ADME and docking guided semi-synthesis and in vitro evaluation of 4-hydroxy-α-tetralone analogs for anti-inflammatory activity. SN APPLIED SCIENCES 2020. [DOI: 10.1007/s42452-020-03798-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022] Open
|
386
|
Methylprednisolone acetate mitigates IL1β induced changes in matrix metalloproteinase gene expression in skeletally immature ovine explant knee tissues. Inflamm Res 2020; 70:99-107. [PMID: 33226449 DOI: 10.1007/s00011-020-01421-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2020] [Revised: 11/01/2020] [Accepted: 11/04/2020] [Indexed: 12/19/2022] Open
Abstract
OBJECTIVE AND DESIGN This study aimed at evaluating the effect of methylprednisolone (MPA) on messenger ribonucleic acid (mRNA) expression levels in immature ovine knee joint tissue explants following interleukin (IL)1β induction and to assess responsiveness of the explants. MATERIAL OR SUBJECTS Explants were harvested from the articular cartilage, synovium, and infrapatellar fat pad (IPFP) from immature female sheep. TREATMENT Methylprednisolone. METHODS The samples were allocated into six groups: (1) control, (2) MPA (10-3 M), (3) MPA (10-4 M), (4) IL1β, (5) IL1β + 10-3 M MPA, or (6) IL1β + 10-4 M MPA. mRNA expression levels for molecules relevant to inflammation, cartilage degradation/anabolism, activation of innate immunity, and adipose tissue/hormones were quantified. Fold changes with MPA treatment were compared via the comparative CT method. RESULTS Methylprednisolone treatment significantly suppressed MMPs consistently across the cartilage (MMP1, MMP3, and MMP13), synovium (MMP1 and MMP3), and IPFP (MMP13) (all p < 0.05). Other genes that were less consistently suppressed include endogenous IL1β (cartilage) and IL6 (IPFP) (all p < 0.05), and others not affected either by IL-1 exposure or subsequent MPA include TGFβ1, TLR4, and adipose-related molecules. CONCLUSIONS Methylprednisolone significantly mitigated IL1β induced mRNA expression for MMPs in the immature cartilage, synovium, and IPFP, but the extent of the responsiveness was tissue-, location-, and gene-specific.
Collapse
|
387
|
Mehling B, Hric M, Salatkova A, Vetrak R, Santora D, Ovariova M, Mihalyova R, Manvelyan M. A Retrospective Study of Stromal Vascular Fraction Cell Therapy for Osteoarthritis. J Clin Med Res 2020; 12:747-751. [PMID: 33224377 PMCID: PMC7665865 DOI: 10.14740/jocmr4354] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2020] [Accepted: 10/08/2020] [Indexed: 01/11/2023] Open
Abstract
Background Osteoarthritis (OA) is progressive degenerative damage to articular cartilage. Current therapeutic options are reduced to control the OA-associated symptoms, leaving the degenerative changes to progress until a joint replacement becomes mandatory. Therefore, therapeutic alternatives are warranted to improve the patient’s quality of life. Cell-based therapy is a developing therapeutic modality, showing promising results in the regeneration of injured cartilage and reduction of on-going inflammation within the affected joint. The current retrospective chart review study was aimed to analyze changes in pain and mobility of subjects with OA after stromal vascular fraction (SVF) cell therapy. Methods Three hundred fifty subjects with hip and knee OA, treated with autologous SVF cells at the Malacky Hospital (Bratislava, Slovakia) in the period from 2015 to 2018, were included in the retrospective chart review study. Results Seven days after SVF cell therapy, 45.2% of subjects experienced improved pain levels and mobility. Three, 6, and 12 months after therapy, improvement in pain levels reached 75.3%, 84.4%, and 84.9%, and improvement in mobility reached 75.2%, 84.4%, and 84.9%. Conclusions Our study of 350 subjects with hip and knee OA showed a significant improvement in pain levels and mobility 3, 6, and 12 months compared to 7 days after autologous SVF cell administration. The treatment demonstrated a strong safety profile with no severe adverse events or complications reported. The results of the study are showing that SVF cell therapy was more effective in subjects with arthritis stage III compared to arthritis stages I, II, and IV.
Collapse
Affiliation(s)
- Brian Mehling
- BHI Therapeutic Sciences, 214 State Street, Hackensack, NJ 07601, USA
| | - Milan Hric
- Malacky Hospital, Nemocnicna, Duklianskych hrdinov 34, 901 22 Malacky, Slovakia
| | - Adriana Salatkova
- Malacky Hospital, Nemocnicna, Duklianskych hrdinov 34, 901 22 Malacky, Slovakia
| | - Robert Vetrak
- Malacky Hospital, Nemocnicna, Duklianskych hrdinov 34, 901 22 Malacky, Slovakia
| | - Doreen Santora
- BHI Therapeutic Sciences, 214 State Street, Hackensack, NJ 07601, USA
| | - Miriama Ovariova
- Blue Horizon International, Grosslingova 17, 801 29 Bratislava, Slovakia
| | - Renata Mihalyova
- Blue Horizon International, Grosslingova 17, 801 29 Bratislava, Slovakia
| | - Marine Manvelyan
- BHI Therapeutic Sciences, 214 State Street, Hackensack, NJ 07601, USA
| |
Collapse
|
388
|
Zhang C. Flare-up of cytokines in rheumatoid arthritis and their role in triggering depression: Shared common function and their possible applications in treatment (Review). Biomed Rep 2020; 14:16. [PMID: 33269077 PMCID: PMC7694594 DOI: 10.3892/br.2020.1392] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2020] [Accepted: 05/31/2020] [Indexed: 01/01/2023] Open
Abstract
Chronic illnesses are associated with an increased risk of depression and anxiety. Rheumatoid arthritis (RA) is a chronic autoimmune disease that typically causes damage to the joints. RA extensively impacts patients, both physically and psychologically. Depression is a common comorbid disorder with RA, which leads to worsened health outcomes. There are several cytokines that are active in the joints of patients with RA. Inflammatory cytokines serve important roles in the key processes in the joints, which usually cause inflammation, articular damage and other comorbidities associated with RA. The key role of inflammatory cytokines could be attributed to their interactions within signaling pathways. In RA, IL-1, and the cytokines of TNF-α, IL-6 and IL-18 are primarily involved. Furthermore, depression is hypothesized to be strongly associated with systemic inflammation, particularly with dysregulation of the cytokine network. The present review summarizes the current state of knowledge on these two diseases from the perspective of inflammation and cytokines, and emphasizes the possible bridge between them by exploring the involvement of systemic cytokines in both conditions.
Collapse
Affiliation(s)
- Chunhai Zhang
- Thyroid Surgery Department, China-Japan Union Hospital of Jilin University, Changchun, Jilin 1300332, P.R. China
| |
Collapse
|
389
|
Induced Pluripotent Stem Cell-Differentiated Chondrocytes Repair Cartilage Defect in a Rabbit Osteoarthritis Model. Stem Cells Int 2020; 2020:8867349. [PMID: 33224204 PMCID: PMC7671807 DOI: 10.1155/2020/8867349] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2020] [Revised: 10/08/2020] [Accepted: 10/19/2020] [Indexed: 12/14/2022] Open
Abstract
The aim of this study was to explore the therapeutic effect of iPSC-mesenchymal stem cell (MSC)-derived chondrocytes in a rabbit osteoarthritis (OA) model. The iPSCs were characterized by gene expressions, immunostaining of pluripotent markers, and in vivo teratoma formation. iPSC-differentiated MSCs were characterized by flow cytometry and trilineage differentiation. A rabbit OA model was established by the transection of the anterior cruciate ligament. The therapeutic effect of transplanted iPSC-MSC-chondrocytes on the OA was evaluated by the histology, immunostaining, and qPCR of defective cartilage. The results showed iPSC could express pluripotency markers such as OCT4, SOX2, and NANOG and form an embryoid body and a teratoma. After differentiation of iPSCs for 30 days, MSCs were established. The iPSC-MSC could express typical MSC markers such as CD29, CD44, CD90, CD105, and HLA-ABC. They could differentiate into adipocytes, osteocytes, and chondrocytes. In this model, iPSC-MSC-chondrocytes significantly improved the histology and ICRS (International Cartilage Repair Society) scores. The transplanted cartilage expressed less IL-1β, TNF-α, and MMP13 than control cartilage. In conclusion, the iPSCs we derived might represent an emerging source for differentiated MSC-chondrocyte and might rescue cartilage defects through its anti-inflammatory and anti-catabolic effects.
Collapse
|
390
|
Ngo L, Knothe Tate ML. Osteoarthritis: New Strategies for Transport and Drug Delivery Across Length Scales. ACS Biomater Sci Eng 2020; 6:6009-6020. [PMID: 33449636 DOI: 10.1021/acsbiomaterials.0c01081] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Osteoarthritis (OA) is the fourth leading cause of disability in adults. Yet, few viable pharmaceutical options exist for pain abatement and joint restoration, aside from joint replacement at late and irreversible stages of the disease. From the first onset of OA, as joint pain increases, individuals with arthritis increasingly reach for drug delivery solutions, from taking oral glycosaminoglycans (GAGs) bought over the counter from retail stores (e.g., Costco) to getting injections of viscous, GAG-containing synovial fluid supplement in the doctor's office. Little is known regarding the efficacy of delivery mode and/or treatment by such disease-modulating agents. This Review addresses the interplay of mechanics and biology on drug delivery to affected joints, which has profound implications for molecular transport in joint health and (patho)physiology. Multiscale systems biology approaches lend themselves to understand the relationship between the cell and joint health in OA and other joint (patho)physiologies. This Review first describes OA-related structural and functional changes in the context of the multilength scale anatomy of articular joints. It then summarizes and categorizes, by size and charge, published molecular transport studies, considering changes in permeability induced through inflammatory pathways. Finally, pharmacological interventions for OA are outlined in the context of molecular weights and modes of drug delivery. Taken together, the current state-of-the-art points to a need for new drug delivery strategies that harness systems-based interactions underpinning molecular transport and maintenance of joint structure and function at multiple length scales from molecular agents to cells, tissues, and tissue compartments which together make up articular joints. Cutting edge and cross-length and -time scale imaging represents a key discovery enabling technology in this process.
Collapse
Affiliation(s)
- Lucy Ngo
- Graduate School of Biomedical Engineering, University of New South Wales, Sydney, New South Wales 2052, Australia
| | - Melissa L Knothe Tate
- Inaugural Paul Trainor Chair of Biomedical Engineering, Graduate School of Biomedical Engineering, University of New South Wales, Sydney, New South Wales 2052, Australia
| |
Collapse
|
391
|
Min N, Ma J, Shi L, Wang L, Liu C, Zhang Y, Xue Q. miR-223 promotes cartilage differentiation of bone marrow-derived mesenchymal stem cells and protects against osteoarthritis by suppressing NLRP-3 expression. Arch Med Sci 2020; 20:2002-2008. [PMID: 39967931 PMCID: PMC11831357 DOI: 10.5114/aoms.2020.100640] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Accepted: 09/05/2020] [Indexed: 02/20/2025] Open
Abstract
Introduction The present investigation evaluates the role of miR-223 mimic in the treatment of osteoarthritis (OA) and postulates the possible molecular mechanism of its action. Material and methods Bone marrow-derived mesenchymal stem cells (BMSCs) were isolated from rats and cultured in chondrogenic medium to stimulate the differentiation of chondrocytes. Alcian blue staining was performed to determine the chondrogenic differentiation and expression of miR-223 in the BMSCs. Moreover, expression of NLR family pyrin domain containing 3 (NLRP-3), matrix metallopeptidase-13 (MMP-13) and collagen (Col II) was determined in miR-223 mimic and inhibitor treated BMSCs. OA was induced by injecting anterior cruciate ligament transection in rats followed by further treatment with the miR-223 mimic for the period of the treatment protocol. Level and expression of inflammatory cytokines were estimated in the cartilage tissue of OA rats. Moreover, immunohistochemical analysis and histopathology study were also performed. Results Data of the study reveal that expression of miR-223 was higher in chondrogenic differentiated BMSCs than normal. Expression of MMP-13 and NLRP-3 was lower, and expression of Col II was higher in miR-223 mimic treated BMSCs than normal. Moreover, data of the in-vivo study indicate that the expression level of cytokines was lower in the cartilage tissue of the miR-223 mimic treated group than the OA group. Treatment with the miR-223 mimic ameliorates the altered histopathology and expression of NLRP-3 in the cartilage tissue of OA rats. Conclusions Data of the study reveal that the miR-223 mimic enhances the chondrogenic differentiation of BMSCs by regulating the NLRP-3/IL-18/TGF-β pathway.
Collapse
Affiliation(s)
- Nan Min
- Department of Orthopedics, Beijing Hospital, Beijing, China
| | - Jie Ma
- Center of Biotherapy, Beijing Hospital, National Center of Gerontology; Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, China
| | - Lei Shi
- Department of Orthopedics, Beijing Hospital, Beijing, China
| | - Lin Wang
- Department of Orthopedics, Beijing Hospital, Beijing, China
| | - Chi Liu
- Department of Orthopedics, Beijing Hospital, Beijing, China
| | - Yaonan Zhang
- Department of Orthopedics, Beijing Hospital, Beijing, China
| | - Qingyun Xue
- Department of Orthopedics, Beijing Hospital, Beijing, China
| |
Collapse
|
392
|
Targeting miR-18a sensitizes chondrocytes to anticytokine therapy to prevent osteoarthritis progression. Cell Death Dis 2020; 11:947. [PMID: 33144571 PMCID: PMC7609664 DOI: 10.1038/s41419-020-03155-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2020] [Revised: 10/18/2020] [Accepted: 10/19/2020] [Indexed: 02/07/2023]
Abstract
Inflammation participates in the development of OA and targeting inflammatory signaling pathways is a potential strategy for OA treatment. IL-1β is one of the most important inflammatory factors to trigger the activation of NF-κB signaling and accelerate OA progression, whereas OA patients could hardly benefit from inhibiting IL-1β in clinic, suggesting the importance to further explore the details of OA inflammation. We here showed that expression of miR-18a in chondrocytes was specifically induced in response to IL-1β in vitro as well as in rat model of OA during which NF-κB signaling was involved, and that nuclear-translocated p65 directly upregulated miR-18a expression at transcriptional level. Further, increased miR-18a mediated hypertrophy of chondrocytes, resulting in OA degeneration, by targeting TGFβ1, SMAD2, and SMAD3 and subsequently leading to repression of TGF-β signaling. And the level of serum miR-18a was positively correlated to severity of OA. Interestingly, other than IL-1β, pro-inflammation cytokines involving TNFα could also remarkably upregulate miR-18a via activating NF-κB signaling and subsequently induce chondrocytes hypertrophy, suggesting a pivotal central role of miR-18a in inflammatory OA progression. Thus, our study revealed a novel convergence of NF-κB and TGF-β signaling mediated by miR-18a, and a novel mechanism underlying inflammation-regulated OA dependent of NF-κB/miR-18a/TGF-β axis. Notably, in vivo assay showed that targeting miR-18a sensitized OA chondrocytes to IL-1β inhibitor as targeting IL-1β and miR-18a simultaneously had much stronger inhibitory effects on OA progression than suppressing IL-1β alone. Therefore, the diagnostic and therapeutic potentials of miR-18a for OA were also revealed.
Collapse
|
393
|
Affiliation(s)
- Dong Gil Jang
- School of Life Sciences, Ulsan National Institute of Science and Technology (UNIST), Ulsan 44919, Korea
| | - Hyo Jung Sim
- School of Life Sciences, Ulsan National Institute of Science and Technology (UNIST), Ulsan 44919, Korea
| | - Eun Kyung Song
- School of Life Sciences, Ulsan National Institute of Science and Technology (UNIST), Ulsan 44919, Korea
| | - Taejoon Kwon
- School of Life Sciences, Ulsan National Institute of Science and Technology (UNIST), Ulsan 44919, Korea
- Center for Genomic Integrity, Institute for Basic Science (IBS), Ulsan 44919, Korea
| | - Tae Joo Park
- School of Life Sciences, Ulsan National Institute of Science and Technology (UNIST), Ulsan 44919, Korea
- Center for Genomic Integrity, Institute for Basic Science (IBS), Ulsan 44919, Korea
| |
Collapse
|
394
|
Preparation and characterization of amnion hydrogel and its synergistic effect with adipose derived stem cells towards IL1β activated chondrocytes. Sci Rep 2020; 10:18751. [PMID: 33127964 PMCID: PMC7603317 DOI: 10.1038/s41598-020-75921-w] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Accepted: 10/13/2020] [Indexed: 12/29/2022] Open
Abstract
Inflammation leads to chondrocyte senescence and cartilage degeneration, resulting in osteoarthritis (OA). Adipose‐derived stem cells (ADSCs) exert paracrine effects protecting chondrocytes from degenerative changes. However, the lack of optimum delivery systems for ADSCs limits its use in the clinic. The use of extracellular matrix based injectable hydrogels has gained increased attention due to their unique properties. In the present study, we developed hydrogels from amnion tissue as a delivery system for ADSCs. We investigated the potential of amnion hydrogel to maintain ADSC functions, the synergistic effect of AM with ADSC in preventing the catabolic responses of inflammation in stimulated chondrocytes. We also investigated the role of Wnt/β-catenin signaling pathway in IL-1β induced inflammation in chondrocytes and the ability of AM-ADSC to inhibit Wnt/β-catenin signaling. Our results showed that AM hydrogels supported cell viability, proliferation, and stemness. ADSCs, AM hydrogels and AM-ADSCs inhibited the catabolic responses of IL-1β and inhibited the Wnt/β-catenin signaling pathway, indicating possible involvement of Wnt/β-catenin signaling pathways in IL-1β induced inflammation. The results also showed that the synergistic effect of AM-ADSCs was more pronounced in preventing catabolic responses in activated chondrocytes. In conclusion, we showed that AM hydrogels can be used as a potential carrier for ADSCs, and can be developed as a potential therapeutic agent for treating OA.
Collapse
|
395
|
Assirelli E, Pulsatelli L, Dolzani P, Mariani E, Lisignoli G, Addimanda O, Meliconi R. Complement Expression and Activation in Osteoarthritis Joint Compartments. Front Immunol 2020; 11:535010. [PMID: 33193305 PMCID: PMC7658426 DOI: 10.3389/fimmu.2020.535010] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2020] [Accepted: 10/05/2020] [Indexed: 01/15/2023] Open
Abstract
Objective To investigate complement(C) factors(F) and their activation fragments expression in OA joint tissues. Design Immunohistochemistry and quantitative imaging were performed to analyze C3, C4, and CF (factor) B expression on osteochondral biopsies (43 patients) collected during arthroplasty. Isolated chondrocytes and synoviocytes, cartilage and synovial tissues obtained from surgical specimens of OA patients (15 patients) were cultured with or without IL-1β. Real time PCR for CFB, C3, and C4 was performed. Culture supernatants were analyzed for C3a, C5a, CFBa, and terminal complement complex (TCC) production. Results In osteochondral biopsies, C factor expression was located in bone marrow, in a few subchondral bone cells and chondrocytes. C3 was the most expressed while factor C4 was the least expressed factor. Gene expression showed that all C factors analyzed were expressed both in chondrocytes and synoviocytes. In chondrocyte cultures and cartilage explants, CFB expression was significantly higher than C3 and C4. Furthermore, CFB, but not C3 and C4 expression was significantly induced by IL-1β. As to C activation factors, C3a was the most produced and CFBa was induced by IL-1β in synovial tissue. TCC production was undetectable in isolated chondrocytes and synoviocytes cell culture supernatants, whereas it was significantly augmented in cartilage explants. Conclusion C factors were locally produced and activated in OA joint with the contribution of all tissues (cartilage, bone, and synovium). Our results support the involvement of innate immunity in OA and suggest an association between some C alternative pathway component and joint inflammation.
Collapse
Affiliation(s)
- Elisa Assirelli
- Laboratory of Immunorheumatology and Tissue Regeneration, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Istituto Ortopedico Rizzoli, Bologna, Italy
| | - Lia Pulsatelli
- Laboratory of Immunorheumatology and Tissue Regeneration, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Istituto Ortopedico Rizzoli, Bologna, Italy
| | - Paolo Dolzani
- Laboratory of Immunorheumatology and Tissue Regeneration, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Istituto Ortopedico Rizzoli, Bologna, Italy
| | - Erminia Mariani
- Laboratory of Immunorheumatology and Tissue Regeneration, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Istituto Ortopedico Rizzoli, Bologna, Italy
- Department of Medical and Surgical Sciences, Alma Mater Studiorum—University of Bologna, Bologna, Italy
| | - Gina Lisignoli
- Laboratory of Immunorheumatology and Tissue Regeneration, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Istituto Ortopedico Rizzoli, Bologna, Italy
| | - Olga Addimanda
- Medicine and Rheumatology Unit, IRCCS Istituto Ortopedico Rizzoli, Bologna, Italy
- Department of Biomedical and Neuromotor Sciences, Alma Mater Studiorum—University of Bologna, Bologna, Italy
| | - Riccardo Meliconi
- Medicine and Rheumatology Unit, IRCCS Istituto Ortopedico Rizzoli, Bologna, Italy
- Department of Biomedical and Neuromotor Sciences, Alma Mater Studiorum—University of Bologna, Bologna, Italy
| |
Collapse
|
396
|
Impacts of Green Tea on Joint and Skeletal Muscle Health: Prospects of Translational Nutrition. Antioxidants (Basel) 2020; 9:antiox9111050. [PMID: 33126483 PMCID: PMC7692648 DOI: 10.3390/antiox9111050] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2020] [Revised: 10/14/2020] [Accepted: 10/22/2020] [Indexed: 12/12/2022] Open
Abstract
Osteoarthritis and sarcopenia are two major joint and skeletal muscle diseases prevalent during aging. Osteoarthritis is a multifactorial progressive degenerative and inflammatory disorder of articular cartilage. Cartilage protection and pain management are the two most important strategies in the management of osteoarthritis. Sarcopenia, a condition of loss of muscle mass and strength, is associated with impaired neuromuscular innervation, the transition of skeletal muscle fiber type, and reduced muscle regenerative capacity. Management of sarcopenia requires addressing both skeletal muscle quantity and quality. Emerging evidence suggests that green tea catechins play an important role in maintaining healthy joints and skeletal muscle. This review covers (i) the prevalence and etiology of osteoarthritis and sarcopenia, such as excessive inflammation and oxidative stress, mitochondrial dysfunction, and reduced autophagy; (ii) the effects of green tea catechins on joint health by downregulating inflammatory signaling mediators, upregulating anabolic mediators, and modulating miRNAs expression, resulting in reduced chondrocyte death, collagen degradation, and cartilage protection; (iii) the effects of green tea catechins on skeletal muscle health via maintaining a dynamic balance between protein synthesis and degradation and boosting the synthesis of mitochondrial energy metabolism, resulting in favorable muscle homeostasis and mitigation of muscle atrophy with aging; and (iv) the current study limitations and future research directions.
Collapse
|
397
|
From classical to deep learning: review on cartilage and bone segmentation techniques in knee osteoarthritis research. Artif Intell Rev 2020. [DOI: 10.1007/s10462-020-09924-4] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
398
|
Down-regulated ciRS-7/up-regulated miR-7 axis aggravated cartilage degradation and autophagy defection by PI3K/AKT/mTOR activation mediated by IL-17A in osteoarthritis. Aging (Albany NY) 2020; 12:20163-20183. [PMID: 33099538 PMCID: PMC7655186 DOI: 10.18632/aging.103731] [Citation(s) in RCA: 72] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2020] [Accepted: 06/25/2020] [Indexed: 12/13/2022]
Abstract
Osteoarthritis (OA) is one of the most painful and widespread chronic degenerative joint diseases and is characterized by destructed articular cartilage and inflamed joints. Previously, our findings indicated that circular RNA ciRS-7 (ciRS-7)/microRNA 7 (miR-7) axis is abnormally expressed in OA, and regulates proliferation, inflammatory responses, and apoptosis of interleukin-1β (IL-1β)-stimulated chondrocytes. However, its underlying role in OA remains unknown. In this study, we first validated cartilage degradation and defection of autophagy in samples of OA patients. IL-1β initially stimulated autophagy of chondrocytes, and ultimately significantly suppressed autophagy. Upregulated ciRS-7/down-regulated miR-7 aggravated IL-1β-induced cartilage degradation, and restrained autophagy in vitro. Gene sequencing and bioinformatics analysis performed on a control group, IL-1β group, and IL-1β+miR-7-mimics group demonstrated that seven of the most significant mRNA candidates were enriched in the interleukin-17 (IL-17) signaling pathway. Increased IL-17A levels were also observed by qRT-PCR and ELISA. In addition, it was revealed that the ciRS-7/miR-7 axis ameliorated cartilage degradation and defection of autophagy by PI3K/AKT/mTOR activation in IL-1β-induced chondrocytes. Furthermore, an OA model was established in rats with medial meniscus destabilization. miR-7-siRNA-expressing lentiviruses alleviated surgical resection-induced cartilage destruction of OA mice, whereas miR-7 mimics worsened the effects. Thus, these findings revealed that the mechanism of the ciRS-7/miR-7 axis involved regulating OA progression and provided valuable directions for OA treatment.
Collapse
|
399
|
TWIST1-MicroRNA-10a-MAP3K7 Axis Ameliorates Synovitis of Osteoarthritis in Fibroblast-like Synoviocytes. MOLECULAR THERAPY-NUCLEIC ACIDS 2020; 22:1107-1120. [PMID: 33294296 PMCID: PMC7691159 DOI: 10.1016/j.omtn.2020.10.020] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Accepted: 10/17/2020] [Indexed: 11/21/2022]
Abstract
Synovitis refers to the inflammation of the synovial membrane and is commonly detected in patients with osteoarthritis (OA). Recent reports have suggested that microRNAs (miRNAs) could be a promising target for diagnosis and prognosis in OA. This study examines the effect of microRNA-10a (miR-10a) in fibroblast-like synoviocyte (FLS)-mediated synovitis obtained from patients with OA. Expression of miR-10a is negatively associated with the severity of synovitis. miR-10a inhibited proliferation, migration, and secretion of pro-inflammatory cytokines of OA-FLS that were obtained from OA patients in vitro. By using a patient-derived xenograft (PDX) model, miR-10a repressed proliferation of OA-FLSs and production of OA synovium-derived pro-inflammatory cytokines in vivo. Twist Family BHLH Transcription Factor 1 (TWIST1) and mitogen-activated protein kinase kinase kinase 7 (MAP3K7) were identified as an upstream regulator and direct target of miR-10a in OA-FLSs, respectively. Nuclear factor κB (NF-κB) signaling pathway, a downstream pathway of MAP3K7, was also repressed by miR-10a in OA-FLSs. To summarize, the TWIST1-miR-10a-MAP3K7-NF-κB pathway mediates the development of synovitis in OA. miR-10a functions as an anti-inflammatory mediator in OA-FLS.
Collapse
|
400
|
Abstract
Brucellosis is a bacterial disease of domestic animals and humans. The pathogenic ability of Brucella organisms relies on their stealthy strategy and their capacity to replicate within host cells and to induce long-lasting infections. Brucella organisms barely induce neutrophil activation and survive within these leukocytes by resisting microbicidal mechanisms. Very few Brucella-infected neutrophils are found in the target organs, except for the bone marrow, early in infection. Still, Brucella induces a mild reactive oxygen species formation and, through its lipopolysaccharide, promotes the premature death of neutrophils, which release chemokines and express "eat me" signals. This effect drives the phagocytosis of infected neutrophils by mononuclear cells that become thoroughly susceptible to Brucella replication and vehicles for bacterial dispersion. The premature death of the infected neutrophils proceeds without NETosis, necrosis/oncosis, or classical apoptosis morphology. In the absence of neutrophils, the Th1 response exacerbates and promotes bacterial removal, indicating that Brucella-infected neutrophils dampen adaptive immunity. This modulatory effect opens a window for bacterial dispersion in host tissues before adaptive immunity becomes fully activated. However, the hyperactivation of immunity is not without a price, since neutropenic Brucella-infected animals develop cachexia in the early phases of the disease. The delay in the immunological response seems a sine qua non requirement for the development of long-lasting brucellosis. This property may be shared with other pathogenic alphaproteobacteria closely related to Brucella We propose a model in which Brucella-infected polymorphonuclear neutrophils (PMNs) function as "Trojan horse" vehicles for bacterial dispersal and as modulators of the Th1 adaptive immunity in infection.
Collapse
|