1
|
Scheike A, Plomp S, Fugazzola MC, Meurot C, Berenbaum F, van Weeren PR, Tryfonidou MA, von Hegedus JH. The Anti-Inflammatory Effects of Liraglutide in Equine Inflammatory Joint Models. J Orthop Res 2025; 43:893-903. [PMID: 39904754 PMCID: PMC11982606 DOI: 10.1002/jor.26050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Revised: 12/16/2024] [Accepted: 01/13/2025] [Indexed: 02/06/2025]
Abstract
This study investigates the anti-inflammatory properties of liraglutide, a glucagon-like peptide 1 receptor agonists, in equine in vitro models and in an in vivo acute synovitis model in Shetland ponies. The anti-inflammatory effect of liraglutide was assessed by measuring concentrations of inflammatory biomarker C-C Motif Chemokine Ligand 2 (CCL2) in culture media of equine whole blood, peripheral blood mononuclear cells (PBMCs), chondrocytes, and synoviocytes, with or without lipopolysaccharide (LPS) or interleukin-1β. In the in vivo experiment, acute synovitis was bilaterally induced with 0.25 ng LPS in the intercarpal joints of seven healthy Shetland ponies. The ponies were subsequently treated with either 6 mg liraglutide or a placebo as a paired control in each joint. The impact of liraglutide on biomarkers associated with inflammation (including white blood cell count, total protein, CCL2, and bradykinin) and cartilage metabolism (such as glycosaminoglycans, general matrix metalloproteinase activity, carboxypropeptide type II collagen, and collagen-cleavage neoepitope of type II collagen) was assessed across serial synovial fluid samples. Liraglutide was found to have an anti-inflammatory effect by reducing CCL2 concentrations in culture media of whole blood, PBMCs, chondrocytes, and synoviocytes. In contrast, no significant differences in synovial fluid inflammatory nor cartilage metabolism biomarker levels were found between joints treated with LPS and 6 mg liraglutide, versus LPS and placebo. In conclusion, liraglutide demonstrates the potential to attenuate inflammatory processes in joint cells. Additional research is necessary to validate its efficacy within the complex milieu of an inflamed joint.
Collapse
Affiliation(s)
- Ann‐Sofie Scheike
- Department of Clinical SciencesFaculty of Veterinary Medicine, Utrecht UniversityUtrechtThe Netherlands
| | - Saskia Plomp
- Department of Clinical SciencesFaculty of Veterinary Medicine, Utrecht UniversityUtrechtThe Netherlands
| | - Maria Carlotta Fugazzola
- Department of Clinical SciencesFaculty of Veterinary Medicine, Utrecht UniversityUtrechtThe Netherlands
| | | | - Francis Berenbaum
- 4Moving BiotechLilleFrance
- INSERM CRSA, AP‐HP Saint‐Antoine HospitalSorbonne UniversityParisFrance
| | - Paul René van Weeren
- Department of Clinical SciencesFaculty of Veterinary Medicine, Utrecht UniversityUtrechtThe Netherlands
| | | | | |
Collapse
|
2
|
Sun Y, Luo Z, Fu Y, Ngo T, Wang W, Wang Y, Kong Y. Primary cilia and inflammatory response: unveiling new mechanisms in osteoarthritis progression. Exp Biol Med (Maywood) 2025; 250:10490. [PMID: 40357414 PMCID: PMC12066368 DOI: 10.3389/ebm.2025.10490] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2025] [Accepted: 04/11/2025] [Indexed: 05/15/2025] Open
Abstract
Osteoarthritis (OA) is a common degenerative joint disease that can lead to chronic pain and disability. The pathogenesis of OA involves chronic low-grade inflammation, characterized by the degradation of chondrocytes, inflammation of the synovium, and systemic low-grade inflammation. This inflammatory response accelerates the progression of OA and contributes to pain and functional impairment. Primary cilia play a crucial role in cellular signal transduction and the maintenance of cartilage matrix homeostasis, and their dysfunction is closely linked to inflammatory responses. Given these roles, primary cilia may significantly contribute to the pathogenesis of OA. This review explores inflammation-associated signaling pathways in OA, including NF-κB, MAPK, JAK/STAT, and PI3K/AKT/mTOR signaling. In addition, we place particular emphasis on cilia-mediated inflammatory modulation in OA. Primary cilia mediate chondrocyte responses to mechanical loading and inflammatory cytokines via pathways including NF-κB, MAPK, TRPV4, and Hedgehog signaling. Notably, alterations in the length and incidence of primary cilia in chondrocytes during OA further underscore their potential role in disease pathogenesis. The identification of biomarkers and therapeutic targets related to primary cilia and inflammatory pathways offers new potential for the treatment and management of OA.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Ying Kong
- Department of Rehabilitation, The Second Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
3
|
Kim C, Li B, Nakamura S, Neely EJ, Rockel JS, Oussenko T, Zhang P, Kapoor M, Nagy A. Engineered mesenchymal stromal cells with interleukin-1beta sticky-trap attenuate osteoarthritis in knee joints. Front Cell Dev Biol 2025; 13:1559155. [PMID: 40264709 PMCID: PMC12011853 DOI: 10.3389/fcell.2025.1559155] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2025] [Accepted: 03/24/2025] [Indexed: 04/24/2025] Open
Abstract
Osteoarthritis (OA) is a common chronic inflammatory joint disease, in which innate immunity plays a pivotal role in pathogenesis. Anti-interleukin-1(IL-1) therapies have shown inconsistent results in clinical trials, potentially due to a mismatch in the spatial and temporal dynamics of interleukin-1beta (IL-1β) production and therapeutic interventions. To address this issue, we developed a novel IL-1β "sticky-trap" utilizing cell and gene-based technologies from our lab and evaluated its efficacy in reducing osteoarthritis progression using a murine destabilization of the medial meniscus (DMM) OA model and a compact bone-derived mesenchymal stromal cell (MSC)-based gene expression system. The extracellular domain of interleukin-1 receptor 2 (IL1R2) was employed to design the sticky IL1R2 trap (stkIL1R2). A murine compact bone-derived MSC line was engineered for gene delivery. Although stkIL1R2 was undetectable in the engineered MSC supernatants by enzyme-linked immunosorbent assay (ELISA) and Western blot, it was localized on the cell surface and extracellular matrix (ECM) and demonstrated specific binding to IL-1β using a fluorescent protein-fused binding assay. Doxycycline (Dox)-induced expression of stkIL1R2 significantly inhibited lipocalin-2 (LCN2) expression which is a biomarker of IL-1β activity. For in vivo experiments, 5 × 104 Dox-inducible stkIL1R2f expressing MSCs were injected into the knee joints of DMM mice. Bioluminescence imaging revealed MSC survival in the knee joints for up to 7 weeks post-injection. Histological analyses at 10 weeks post-injection, including Safranin-O and Masson trichrome staining, showed that stkIL1R2 treated joints exhibited significantly less cartilage degradation and synovitis compared to controls, as assessed by Osteoarthritis Research Society International (OARSI) scoring of the femur, tibia, and synovium. Moreover, stkIL1R2 treatment reduced matrix metalloproteinases-13 (MMP-13) positive cells and collagen type II degradation in the affected joints. In conclusion, we developed a MSC line expressing an inducible IL1 sticky-trap, which localized to the cell surface and ECM and specifically bound IL-1β. These engineered MSCs survived in normal and DMM knee joints for up to 7 weeks and significantly delayed OA progression and inflammation in the murine model. This study introduces a promising therapeutic approach to combat OA progression.
Collapse
Affiliation(s)
- Christopher Kim
- Lunenfeld Tanenbaum Research Institute, Sinai Health System, Toronto, ON, Canada
- Schroeder Arthritis Institute, Krembil Research Institute, University Health Network, Toronto, ON, Canada
| | - Biao Li
- Lunenfeld Tanenbaum Research Institute, Sinai Health System, Toronto, ON, Canada
- Schroeder Arthritis Institute, Krembil Research Institute, University Health Network, Toronto, ON, Canada
| | - Sayaka Nakamura
- Schroeder Arthritis Institute, Krembil Research Institute, University Health Network, Toronto, ON, Canada
| | - Eric J. Neely
- Lunenfeld Tanenbaum Research Institute, Sinai Health System, Toronto, ON, Canada
| | - Jason S. Rockel
- Schroeder Arthritis Institute, Krembil Research Institute, University Health Network, Toronto, ON, Canada
| | - Tatiana Oussenko
- Lunenfeld Tanenbaum Research Institute, Sinai Health System, Toronto, ON, Canada
| | - Puzheng Zhang
- Lunenfeld Tanenbaum Research Institute, Sinai Health System, Toronto, ON, Canada
| | - Mohit Kapoor
- Schroeder Arthritis Institute, Krembil Research Institute, University Health Network, Toronto, ON, Canada
| | - Andras Nagy
- Lunenfeld Tanenbaum Research Institute, Sinai Health System, Toronto, ON, Canada
- Department of Obstetrics and Gynaecology and Institute of Medical Science, University of Toronto, Toronto, ON, Canada
- Australian Regenerative Medicine Institute, Monash University, Melbourne, VIC, Australia
| |
Collapse
|
4
|
Hu Z, Wang C, Wang C, He J, Yan Y, Xu Z, Yu Y, Yu Y, Cheng H, Liu L, Tang M, Zhang C, Yu H, Jing J, Cheng W. The comparative efficacy of L-glutamine, celecoxib, and glucosamine sulfate in osteoarthritis management. Sci Rep 2025; 15:8992. [PMID: 40089639 PMCID: PMC11910618 DOI: 10.1038/s41598-025-93357-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2024] [Accepted: 03/06/2025] [Indexed: 03/17/2025] Open
Abstract
To explore the therapeutic efficacy of L-glutamine (L-Gln) on pathological progression and clinical symptoms of osteoarthritis (OA), and compare with glucosamine sulfate (GS), and celecoxib (CXB). Rats were administered sodium chloride, L-Gln, GS, or CXB via gavage for eight weeks starting from the fifth week after sham operation or Anterior Cruciate Ligament Transection (ACLT) + Medial Meniscectomy (MMx). Then the severity of knee OA in rats was evaluated by serological analysis, histological examination and imaging examination. In addition, patients with mild primary OA were administered L-Gln, GS, or CXB orally for 12 weeks in accordance with the randomization principle. The efficacy end points were the change from baseline to week 24 in the pain and physical function subscale scores of the Western Ontario and McMaster Universities OA Index (WOMAC), and Lequesne score. Treatment with L-Gln alleviated the increased concentration of serum cartilage degradation markers caused by OA in rats. Histological tests showed improvement in knee joint cartilage destruction after treatment. Three-dimensional CT scans and reconstructions revealed a reduction in osteophyte formation and subchondral bone loss. L-glutamine performed as well as or better than glucosamine sulfate and celecoxib in all comparative measures among the three treatment groups. In clinical trials, the WOMAC pain and physical function subscale scores, as well as the Lequesne score, decreased from baseline in all three patient groups during follow-up, with no significant differences observed between the groups. Our research indicates that L-Gln is comparable to GS and CXB in improving the pathological progression and clinical efficacy of OA, which makes it a promising drug for the treatment of osteoarthritis.
Collapse
Affiliation(s)
- Zhongyao Hu
- Department of Orthopedics, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, China
- Institute of Orthopedics, Research Center for Translational Medicine, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, China
| | | | - Chen Wang
- Department of Orthopedics, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, China
- Institute of Orthopedics, Research Center for Translational Medicine, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, China
| | - Junyan He
- Department of Orthopedics, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, China
- Institute of Orthopedics, Research Center for Translational Medicine, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, China
| | - Yiqun Yan
- Department of Orthopedics, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, China
- Institute of Orthopedics, Research Center for Translational Medicine, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, China
| | - Zelin Xu
- Department of Orthopedics, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, China
- Institute of Orthopedics, Research Center for Translational Medicine, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, China
| | - Yangmang Yu
- Department of Orthopedics, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, China
- Institute of Orthopedics, Research Center for Translational Medicine, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, China
| | - Ya Yu
- Huoshan County Hospital, Lu'an, 237299, Anhui, China
| | - Huan Cheng
- Huoshan County Hospital of Traditional Chinese Medicine, Lu'an, 237200, Anhui, China
| | - Lei Liu
- Suzhou Municipal Hospital in Anhui Province, Suzhou, 234000, Anhui, China
| | - Miao Tang
- Suzhou Municipal Hospital in Anhui Province, Suzhou, 234000, Anhui, China
| | - Chun Zhang
- Department of Orthopedics, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, China
- Institute of Orthopedics, Research Center for Translational Medicine, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, China
| | - Haoran Yu
- Department of Orthopedics, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, China.
- Institute of Orthopedics, Research Center for Translational Medicine, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, China.
| | - Juehua Jing
- Department of Orthopedics, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, China.
- Institute of Orthopedics, Research Center for Translational Medicine, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, China.
| | - Wendan Cheng
- Department of Orthopedics, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, China.
- Institute of Orthopedics, Research Center for Translational Medicine, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, China.
| |
Collapse
|
5
|
Chen H, Liu S, Xing J, Wen Y, Chen L. Orientin alleviates chondrocyte senescence and osteoarthritis by inhibiting PI3K/AKT pathway. Bone Joint Res 2025; 14:245-258. [PMID: 40085067 PMCID: PMC11908465 DOI: 10.1302/2046-3758.143.bjr-2023-0383.r2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 03/16/2025] Open
Abstract
AIMS Osteoarthritis (OA) is a common degenerative disease that leads to pain, disability, and reduced quality of life. Orientin exhibits considerable anti-inflammatory and antioxidative properties, but its role in chondrocyte senescence and OA progress has not yet been fully characterized. The aim of this study was to evaluate the protective effects of orientin on OA. METHODS The role of orientin in extracellular matrix (ECM) degradation, mitochondrial homeostasis, and chondrocyte senescence was investigated in vitro. Meanwhile, we used molecular docking, small molecular inhibitors, and RNA interference to screen and validate candidate proteins regulated by orientin. In an anterior cruciate ligament transection (ACLT) rat model, radiograph, micro-CT, and various histological examinations were applied to evaluate the therapeutic effects of orientin on OA. RESULTS We found that orientin inhibited ECM degradation and senescence-associated secretory phenotype (SASP) factor expression in interleukin (IL)-1β-treated chondrocytes. Additionally, orientin reduced the level of reactive oxygen species (ROS) and improved mitochondrial homeostasis. Furthermore, orientin suppressed IL-1β-induced activation of the nuclear factor kappa B (NF-κB) signalling pathway. We also found that orientin bound to phosphoinositide 3-kinase (PI3K) and inhibited NF-κB cascades via the PI3K/AKT pathway. In vivo, we demonstrated that orientin improved cartilage wear and reduced synovial inflammation and osteophyte in an ACLT rat model. CONCLUSION Orientin improves mitochondrial homeostasis, inhibits chondrocyte senescence, and alleviates OA progress via the PI3K/AKT/NF-κB axis, which suggests that orientin is a potential effective therapeutic agent for OA.
Collapse
Affiliation(s)
- Haitao Chen
- Division of Joint Surgery and Sports Medicine, Department of Orthopedic Surgery, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Siyi Liu
- Division of Joint Surgery and Sports Medicine, Department of Orthopedic Surgery, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Junwei Xing
- Division of Joint Surgery and Sports Medicine, Department of Orthopedic Surgery, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Yinxian Wen
- Division of Joint Surgery and Sports Medicine, Department of Orthopedic Surgery, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Liaobin Chen
- Division of Joint Surgery and Sports Medicine, Department of Orthopedic Surgery, Zhongnan Hospital of Wuhan University, Wuhan, China
| |
Collapse
|
6
|
Bai J, Lv T, Yu H, Ji Z, Gu X, Gao Y, Ma L. The combined impact of neutrophil-percentage-to-albumin ratio and depressive symptoms on mortality in US arthritis patients: insights from NHANES (2005-2018). Front Public Health 2025; 13:1545250. [PMID: 40115342 PMCID: PMC11922730 DOI: 10.3389/fpubh.2025.1545250] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2024] [Accepted: 02/24/2025] [Indexed: 03/23/2025] Open
Abstract
Background The neutrophil-to-albumin ratio (NPAR) reflects inflammation and nutritional status, while depression significantly impacts survival in chronic disease patients. This study examines the independent and combined effects of NPAR and depressive symptoms on all-cause and cardiovascular mortality in arthritis patients. Methods We analyzed a nationally representative sample of people with arthritisaged 40 and older from NHANES (2005-2018). NPAR assessed inflammation and nutritional status, while depressive symptoms were measured by PHQ-9. Weighted Cox regression examined the independent and joint associations of NPAR and PHQ-9 with all-cause and cardiovascular disease (CVD) mortality. Results Our analysis indicated that higher NPAR levels combined with lower depressive symptoms (PHQ-9 < 10) significantly increased all-cause and CVD mortality risks in arthritis patients. In this group, the hazard ratio (HR) for all-cause mortality was 2.087, with a similarly elevated CVD mortality risk (HR = 2.614), underscoring NPAR's predictive strength in non-depressed individuals. Among those with higher depressive symptoms, while elevated NPAR was still associated with increased mortality, its impact on CVD mortality was less marked, highlighting the need for further research into the NPAR-depression interaction. Conclusion This study identifies NPAR as a key predictor of mortality in arthritis patients, particularly those with fewer depressive symptoms. NPAR significantly predicts all-cause and CVD mortality, underscoring its value as an inflammation and nutrition biomarker. Integrating NPAR in clinical practice could enhance individualized risk assessment and intervention for arthritis patients.
Collapse
Affiliation(s)
- Jinyue Bai
- Department of General Practice, Aerospace Center Hospital, Beijing, China
| | - Taihong Lv
- Department of General Medicine, Beijing TianTan Hospital, Capital Medical University, Beijing, China
| | - Hanming Yu
- Department of Pulmonary and Critical Care Medicine, The Fourth Affiliated Hospital of China Medical University, Shenyang, China
| | - Zishuo Ji
- Department of Neurology, Beijing TianTan Hospital, Capital Medical University, Beijing, China
| | - Xiu Gu
- Department of Pulmonary and Critical Care Medicine, The Fourth Affiliated Hospital of China Medical University, Shenyang, China
| | - Yun Gao
- Department of General Practice, Aerospace Center Hospital, Beijing, China
| | - Li Ma
- Department of General Medicine, Beijing TianTan Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
7
|
Largo R, Mediero A, Villa-Gomez C, Bermejo-Alvarez I, Herrero-Beaumont G. Aberrant anabolism hinders constructive metabolism of chondrocytes by pharmacotherapy in osteoarthritis. Bone Joint Res 2025; 14:199-207. [PMID: 40042132 PMCID: PMC11881514 DOI: 10.1302/2046-3758.143.bjr-2024-0241.r1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 04/03/2025] Open
Abstract
Osteoarthritis (OA) is a highly prevalent and disabling disease with an unmet therapeutic need. The characteristic cartilage loss and alteration of other joint structures result from a complex interaction of multiple risk factors, with mechanical overload consistently playing a central role. This overload generates an inflammatory response in the cartilage due to the activation of the innate immune response in chondrocytes, which occurs through various cellular mechanisms. Moreover, risk factors associated with obesity, being overweight, and metabolic syndrome enhance the inflammatory response both locally and systemically. OA chondrocytes, the only cells present in articular cartilage, are therefore inflamed and initiate an anabolic process in an attempt to repair the damaged tissue, which ultimately results in an aberrant and dysfunctional process. Under these circumstances, where the cartilage continues to be subjected to chronic mechanical stress, proposing a treatment that stimulates the chondrocytes' anabolic response to restore tissue structure does not appear to be a therapeutic target with a high likelihood of success. In fact, anabolic drugs proposed for the treatment of OA have yet to demonstrate efficacy. By contrast, multiple therapeutic strategies focused on pharmacologically managing the inflammatory component, both at the joint and systemic levels, have shown promise. Therefore, prioritizing the control of chronic innate pro-inflammatory pathways presents the most viable and promising therapeutic strategy for the effective management of OA. As research continues, this approach may offer the best opportunity to alleviate the burden of this incapacitating disease.
Collapse
Affiliation(s)
- Raquel Largo
- Joint and Bone Research Unit, Service of Rheumatology, IIS Fundación Jiménez Díaz UAM, Madrid, Spain
| | - Aranzazu Mediero
- Joint and Bone Research Unit, Service of Rheumatology, IIS Fundación Jiménez Díaz UAM, Madrid, Spain
| | - Cristina Villa-Gomez
- Joint and Bone Research Unit, Service of Rheumatology, IIS Fundación Jiménez Díaz UAM, Madrid, Spain
| | - Ismael Bermejo-Alvarez
- Joint and Bone Research Unit, Service of Rheumatology, IIS Fundación Jiménez Díaz UAM, Madrid, Spain
| | - Gabriel Herrero-Beaumont
- Joint and Bone Research Unit, Service of Rheumatology, IIS Fundación Jiménez Díaz UAM, Madrid, Spain
| |
Collapse
|
8
|
Hlubek R, Kušnierová P, Walder P, Bystroňová I, Douša P. [Biomarkers and Their Role in Understanding Osteoarthritis]. ACTA CHIRURGIAE ORTHOPAEDICAE ET TRAUMATOLOGIAE CECHOSLOVACA 2025; 92:36-43. [PMID: 40145590 DOI: 10.55095/achot2024/051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/27/2024] [Accepted: 09/30/2024] [Indexed: 03/28/2025]
Abstract
Osteoarthritis (OA) is a degenerative joint disease characterized by progressive damage and loss of articular cartilage with concomitant structural and functional changes in the joint. It is the most common cause of joint pain globally and the resulting productivity loss to the economy. The clinical symptoms of osteoarthritis are mostly determined by the difficulties of patients related to the development of articular degenerative changes, which secondarily lead to joint stiffness and functional limitation. The diagnosis of this disease is currently based on typical clinical symptoms and radiographic findings (e.g. joint space narrowing, osteophytes, subchondral sclerosis, etc.). These parameters, however, are difficult to detect in the early stages of the disease and are most often recognized in the advanced stages. For these reasons, the diagnosis of osteoarthritis is often delayed until irreversible destruction of joint tissue occurs and conservative treatment is less effective. Despite recent scientific progress in understanding the genetic and molecular principles of joint degeneration, currently there is no reliable causal therapy for OA. This review aims to summarize current knowledge of osteoarthritis and possible future directions for diagnosis and early intervention. One of such directions is the study of the so-called biomarkers. A biomarker is defined as an indicator of biological processes and can include radiographic, histological, physiological, or molecular characteristics. In particular, molecular biomarkers are widely studied in knee OA. Attention of the research community is focused on the study of biomarkers as a method of detection and prediction of the early stages of osteoarthritis before irreversible joint damage occurs. Biomarkers help develop more effective and, above all, personalized treatment, thus improve the overall clinical approach to the patient.
Collapse
Affiliation(s)
- Rudolf Hlubek
- Klinika urazove chirurgie a ortopedie, Ortopedicke oddeleni, Fakultni nemocnice Ostrava
- Katedra chirurgickych oboru, Lekarska fakulta, Ostravska univerzita, Ostrava
| | - Pavlína Kušnierová
- Ustav laboratorni mediciny, Oddeleni klinicke biochemie, Fakultni nemocnice Ostrava
- Ustav laboratorni mediciny, Lekarska fakulta, Ostravska univerzita, Ostrava
| | - Pavel Walder
- Klinika urazove chirurgie a ortopedie, Ortopedicke oddeleni, Fakultni nemocnice Ostrava
- Katedra chirurgickych oboru, Lekarska fakulta, Ostravska univerzita, Ostrava
| | - Iveta Bystroňová
- Ustav laboratorni mediciny, Oddeleni klinicke biochemie, Fakultni nemocnice Ostrava
- Ustav laboratorni mediciny, Lekarska fakulta, Ostravska univerzita, Ostrava
| | - Pavel Douša
- Klinika urazove chirurgie a ortopedie, Ortopedicke oddeleni, Fakultni nemocnice Ostrava
- Katedra chirurgickych oboru, Lekarska fakulta, Ostravska univerzita, Ostrava
| |
Collapse
|
9
|
Figueroa-Valdés AI, Luz-Crawford P, Herrera-Luna Y, Georges-Calderón N, García C, Tobar HE, Araya MJ, Matas J, Donoso-Meneses D, de la Fuente C, Cuenca J, Parra E, Lillo F, Varela C, Cádiz MI, Vernal R, Ortloff A, Nardocci G, Castañeda V, Adasme-Vidal C, Kunze-Küllmer M, Hidalgo Y, Espinoza F, Khoury M, Alcayaga-Miranda F. Clinical-grade extracellular vesicles derived from umbilical cord mesenchymal stromal cells: preclinical development and first-in-human intra-articular validation as therapeutics for knee osteoarthritis. J Nanobiotechnology 2025; 23:13. [PMID: 39806427 PMCID: PMC11730155 DOI: 10.1186/s12951-024-03088-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Accepted: 12/30/2024] [Indexed: 01/16/2025] Open
Abstract
Osteoarthritis (OA) is a joint disease characterized by articular cartilage degradation. Persistent low-grade inflammation defines OA pathogenesis, with crucial involvement of pro-inflammatory M1-like macrophages. While mesenchymal stromal cells (MSC) and their small extracellular vesicles (sEV) hold promise for OA treatment, achieving consistent clinical-grade sEV products remains a significant challenge. This study aims to develop fully characterized, reproducible, clinical-grade batches of sEV derived from umbilical cord (UC)-MSC for the treatment of OA while assessing its efficacy and safety. Initially, a standardized, research-grade manufacturing protocol was established to ensure consistent sEV production. UC-MSC-sEV characterization under non-cGMP conditions showed consistent miRNA and protein profiles, suggesting their potential for standardized manufacturing. In vitro studies evaluated the efficacy, safety, and potency of sEV; animal studies confirmed their effectiveness and safety. In vitro, UC-MSC-sEV polarized macrophages to an anti-inflammatory M2b-like phenotype, through STAT1 modulation, indicating their potential to create an anti-inflammatory environment in the affected joints. In silico studies confirmed sEV's immunosuppressive signature through miRNA and proteome analysis. In an OA mouse model, sEV injected intra-articularly (IA) induced hyaline cartilage regeneration, validated by histological and μCT analyses. The unique detection of sEV signals within the knee joint over time highlights its safety profile by confirming the retention of sEV in the joint. The product development of UC-MSC-sEV involved refining, standardizing, and validating processes in compliance with GMP standards. The initial assessment of the safety of the clinical-grade product via IA administration in a first-in-human study showed no adverse effects after a 12 month follow-up period. These results support the progress of this sEV-based therapy in an early-phase clinical trial, the details of which are presented and discussed in this work. This study provides data on using UC-MSC-sEV as local therapy for OA, highlighting their regenerative and anti-inflammatory properties and safety in preclinical and a proof-of-principle clinical application.
Collapse
Affiliation(s)
- Aliosha I Figueroa-Valdés
- Laboratorio de Medicina Nano-Regenerativa, Centro de Investigación e Innovación Biomédica (CiiB), Universidad de los Andes, Santiago, Chile
- IMPACT, Center of Interventional Medicine for Precision and Advanced Cellular Therapy, Santiago, Chile
| | - Patricia Luz-Crawford
- Laboratorio de Inmunología Celular y Molecular, Centro de Investigación e Innovación Biomédica (CiiB), Universidad de los Andes, Santiago, Chile
- IMPACT, Center of Interventional Medicine for Precision and Advanced Cellular Therapy, Santiago, Chile
| | - Yeimi Herrera-Luna
- Laboratorio de Inmunología Celular y Molecular, Centro de Investigación e Innovación Biomédica (CiiB), Universidad de los Andes, Santiago, Chile
- IMPACT, Center of Interventional Medicine for Precision and Advanced Cellular Therapy, Santiago, Chile
| | - Nicolás Georges-Calderón
- Laboratorio de Medicina Nano-Regenerativa, Centro de Investigación e Innovación Biomédica (CiiB), Universidad de los Andes, Santiago, Chile
- Programa de Doctorado en Biomedicina, Facultad de Medicina, Universidad de los Andes, Santiago, Chile
- IMPACT, Center of Interventional Medicine for Precision and Advanced Cellular Therapy, Santiago, Chile
| | - Cynthia García
- Laboratorio de Inmunología Celular y Molecular, Centro de Investigación e Innovación Biomédica (CiiB), Universidad de los Andes, Santiago, Chile
- Programa de Doctorado en Biomedicina, Facultad de Medicina, Universidad de los Andes, Santiago, Chile
- IMPACT, Center of Interventional Medicine for Precision and Advanced Cellular Therapy, Santiago, Chile
| | - Hugo E Tobar
- Laboratorio de Medicina Nano-Regenerativa, Centro de Investigación e Innovación Biomédica (CiiB), Universidad de los Andes, Santiago, Chile
- IMPACT, Center of Interventional Medicine for Precision and Advanced Cellular Therapy, Santiago, Chile
| | - María Jesús Araya
- Laboratorio de Inmunología Celular y Molecular, Centro de Investigación e Innovación Biomédica (CiiB), Universidad de los Andes, Santiago, Chile
- Programa de Doctorado en Biomedicina, Facultad de Medicina, Universidad de los Andes, Santiago, Chile
- IMPACT, Center of Interventional Medicine for Precision and Advanced Cellular Therapy, Santiago, Chile
| | - José Matas
- Centro de Terapia Celular, Clínica Universidad de los Andes, Santiago, Chile
- Departmento de Cirugía Ortopédica, Clínica Universidad de los Andes, Santiago, Chile
| | - Darío Donoso-Meneses
- Laboratorio de Medicina Nano-Regenerativa, Centro de Investigación e Innovación Biomédica (CiiB), Universidad de los Andes, Santiago, Chile
- Programa de Doctorado en Biomedicina, Facultad de Medicina, Universidad de los Andes, Santiago, Chile
- IMPACT, Center of Interventional Medicine for Precision and Advanced Cellular Therapy, Santiago, Chile
| | | | - Jimena Cuenca
- Laboratorio de Medicina Nano-Regenerativa, Centro de Investigación e Innovación Biomédica (CiiB), Universidad de los Andes, Santiago, Chile
- Consorcio REGENERO, Chilean Consortium for Regenerative Medicine, Santiago, Chile
- IMPACT, Center of Interventional Medicine for Precision and Advanced Cellular Therapy, Santiago, Chile
- Cells for Cells, Santiago, Chile
| | - Eliseo Parra
- Consorcio REGENERO, Chilean Consortium for Regenerative Medicine, Santiago, Chile
| | - Fernando Lillo
- Consorcio REGENERO, Chilean Consortium for Regenerative Medicine, Santiago, Chile
| | - Cristóbal Varela
- Departmento de Radiología, Clínica Universidad de los Andes, Santiago, Chile
| | - María Ignacia Cádiz
- Laboratorio de Medicina Nano-Regenerativa, Centro de Investigación e Innovación Biomédica (CiiB), Universidad de los Andes, Santiago, Chile
- Consorcio REGENERO, Chilean Consortium for Regenerative Medicine, Santiago, Chile
- Cells for Cells, Santiago, Chile
| | - Rolando Vernal
- Laboratorio de Biología Periodontal, Facultad de Odontología, Universidad de Chile, Santiago, Chile
| | - Alexander Ortloff
- Departamento de Ciencias Veterinarias y Salud Pública, Facultad de Recursos Naturales, Universidad Católica de Temuco, Temuco, Chile
| | - Gino Nardocci
- Laboratorio de Biología Molecular y Bioinformática, Centro de Investigación e Innovación Biomédica (CiiB), Universidad de los Andes, Santiago, Chile
- IMPACT, Center of Interventional Medicine for Precision and Advanced Cellular Therapy, Santiago, Chile
- Escuela de Medicina, Facultad de Medicina, Universidad de los Andes, Santiago, Chile
| | - Verónica Castañeda
- Laboratorio de Biología Molecular y Bioinformática, Centro de Investigación e Innovación Biomédica (CiiB), Universidad de los Andes, Santiago, Chile
- Programa de Doctorado en Biomedicina, Facultad de Medicina, Universidad de los Andes, Santiago, Chile
- IMPACT, Center of Interventional Medicine for Precision and Advanced Cellular Therapy, Santiago, Chile
| | - Catalina Adasme-Vidal
- Laboratorio de Medicina Nano-Regenerativa, Centro de Investigación e Innovación Biomédica (CiiB), Universidad de los Andes, Santiago, Chile
- IMPACT, Center of Interventional Medicine for Precision and Advanced Cellular Therapy, Santiago, Chile
| | - Maximiliano Kunze-Küllmer
- Consorcio REGENERO, Chilean Consortium for Regenerative Medicine, Santiago, Chile
- Cells for Cells, Santiago, Chile
- EVast Bio, Miami, FL, USA
| | - Yessia Hidalgo
- Laboratorio de Medicina Nano-Regenerativa, Centro de Investigación e Innovación Biomédica (CiiB), Universidad de los Andes, Santiago, Chile
- IMPACT, Center of Interventional Medicine for Precision and Advanced Cellular Therapy, Santiago, Chile
| | - Francisco Espinoza
- Consorcio REGENERO, Chilean Consortium for Regenerative Medicine, Santiago, Chile
- IMPACT, Center of Interventional Medicine for Precision and Advanced Cellular Therapy, Santiago, Chile
- Centro de Terapia Celular, Clínica Universidad de los Andes, Santiago, Chile
- Departmento de Reumatología, Clínica Universidad de los Andes, Santiago, Chile
| | - Maroun Khoury
- Laboratorio de Medicina Nano-Regenerativa, Centro de Investigación e Innovación Biomédica (CiiB), Universidad de los Andes, Santiago, Chile.
- Consorcio REGENERO, Chilean Consortium for Regenerative Medicine, Santiago, Chile.
- IMPACT, Center of Interventional Medicine for Precision and Advanced Cellular Therapy, Santiago, Chile.
- Cells for Cells, Santiago, Chile.
- EVast Bio, Miami, FL, USA.
| | - Francisca Alcayaga-Miranda
- Laboratorio de Medicina Nano-Regenerativa, Centro de Investigación e Innovación Biomédica (CiiB), Universidad de los Andes, Santiago, Chile.
- Consorcio REGENERO, Chilean Consortium for Regenerative Medicine, Santiago, Chile.
- IMPACT, Center of Interventional Medicine for Precision and Advanced Cellular Therapy, Santiago, Chile.
- Cells for Cells, Santiago, Chile.
| |
Collapse
|
10
|
Zhang Z, Lian Y, He Y, Liu H, Meng K, Wang Y, Ma W. Genetic insights into the risk of hip osteoarthritis on stroke: A single-variable and multivariable Mendelian randomization. PLoS One 2025; 20:e0313032. [PMID: 39787159 PMCID: PMC11717317 DOI: 10.1371/journal.pone.0313032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2024] [Accepted: 10/17/2024] [Indexed: 01/12/2025] Open
Abstract
BACKGROUND Hip osteoarthritis has been identified as a potential risk factor for stroke, with previous studies have demonstrated an association between hip osteoarthritis and stroke. This study aims to further elucidate the causal relationship between the two, employing Two-Sample and Multivariable Mendelian randomization methods. METHODS SNPs, derived from two extensive GWAS, served as instruments in exploring the association between genetically predicted hip osteoarthritis and stroke risk, utilizing two-sample Mendelian randomization. In Multivariable Mendelian randomization, factors such as cigarettes per day, alcoholic drinks per week, hypertension, body mass index, type 2 diabetes, C-reactive protein, rheumatoid arthritis were incorporated to further account for the independent causal effects of multiple correlated exposures. RESULTS Two-sample Mendelian randomization analysis revealed that hip osteoarthritis exerts a potential causal effect on any stroke, any ischemic stroke, and cardioembolic stroke, while it did not influence large artery stroke and small vessel stroke. Multivariable MR analysis indicated that the causal effect of hip osteoarthritis on any ischemic stroke and cardioembolic stroke was no longer evident after adjusting for C-reactive protein, and similarly, the effect on any ischemic stroke was not observed after adjusting for type 2 diabetes. However, the effects on any stroke, any ischemic stroke, and cardioembolic stroke remained significant after adjustments for hypertension, alcoholic drinks per week, cigarettes per day, body mass index, and rheumatoid arthritis. CONCLUSION The study demonstrated that elevated hip osteoarthritis, as predicted by genetic factors, was potential associated with an increased risk of any stroke, any ischemic stroke, and cardioembolic stroke, but showed no correlation with hypertension, alcoholic drinks per week, cigarettes per day, type 2 diabetes, C-reactive protein, body mass index levels, and rheumatoid arthritis.
Collapse
Affiliation(s)
- Zhengze Zhang
- The First Clinical Medical School of Guangzhou University of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, PR China
- Department of Anesthesiology, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, PR China
| | - Yanan Lian
- Shandong Mental Health Center, Jinan, Shandong, PR China
| | - Yuewen He
- The First Clinical Medical School of Guangzhou University of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, PR China
- Department of Anesthesiology, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, PR China
| | - Hao Liu
- The First Clinical Medical School of Guangzhou University of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, PR China
- Department of Anesthesiology, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, PR China
| | - Kai Meng
- Department of Orthopaedics, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, Shandong, PR China
| | - Yong Wang
- Department of Anesthesiology, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, PR China
| | - Wuhua Ma
- Department of Anesthesiology, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, PR China
| |
Collapse
|
11
|
Wei K, Zhou C, Shu Z, Shang X, Zou Y, Zhou W, Xu H, Liang Y, Ma T, Sun X, Xiao J. MYSM1 attenuates osteoarthritis by recruiting PP2A to deubiquitinate and dephosphorylate RIPK2. Bone Res 2025; 13:3. [PMID: 39746943 PMCID: PMC11696715 DOI: 10.1038/s41413-024-00368-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Revised: 08/27/2024] [Accepted: 08/30/2024] [Indexed: 01/04/2025] Open
Abstract
Osteoarthritis (OA), the most prevalent degenerative joint disease, is marked by cartilage degradation and pathological alterations in surrounding tissues. Currently, no effective disease-modifying treatments exist. This study aimed to elucidate the critical roles of Myb-like, SWIRM, and MPN domains 1 (MYSM1) and its downstream effector, Receptor-interacting protein kinase 2 (RIPK2), in OA pathogenesis and the underlying mechanisms. Our findings revealed reduced MYSM1 levels in the cartilage of OA patients and mouse models. Genetic or adenovirus-induced MYSM1 knockout exacerbated OA progression in mice, whereas MYSM1 overexpression mitigated it. Mechanistically, MYSM1 inhibited the NF-κB and MAPK signaling pathways. Conversely, downstream RIPK2 significantly increased OA-like phenotypes and activated the NF-κB and MAPK pathways. The Ripk2S176D mutation accelerated OA pathogenesis, while Ripk2 silencing or Ripk2S176A mutation deactivated NF-κB and MAPK pathways, counteracting the role of MYSM1. MYSM1 deubiquitinates and dephosphorylates RIPK2S176 by recruiting protein phosphatase 2 A (PP2A). These results suggest that targeting MYSM1 or downstream RIPK2 offers promising therapeutic potential for OA.
Collapse
Affiliation(s)
- Kang Wei
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Department of Plastic Surgery, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Chuankun Zhou
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Zixing Shu
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Xingru Shang
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Yi Zou
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Wei Zhou
- Institute of Hepatobiliary Diseases, Transplant Center, Hubei Key Laboratory of Medical Technology on Transplantation, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Huanhuan Xu
- Department of Obstetrics and Gynecology, Wuhan Children's Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yulin Liang
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Tian Ma
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Xuying Sun
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China.
| | - Jun Xiao
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China.
| |
Collapse
|
12
|
Dyson G, Barrett M, Schlupp L, Prinz E, Hannebut N, Szymczak A, Brawner CM, Jeffries MA. Ketogenic Diet-Associated Worsening of Osteoarthritis Histologic Secerity, Increased Pain Sensitivity and Gut Microbiome Dysbiosis in Mice. ACR Open Rheumatol 2025; 7:e11794. [PMID: 39853943 PMCID: PMC11760994 DOI: 10.1002/acr2.11794] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2024] [Revised: 11/29/2024] [Accepted: 12/09/2024] [Indexed: 01/26/2025] Open
Abstract
OBJECTIVES Dietary interventions are a potentially powerful treatment option for knee osteoarthritis (OA). The objective of this study was to evaluate a well-formulated ketogenic diet (KD) in the context of knee OA histology and pain using the destabilization of the medial meniscus (DMM) mouse model and correlate with gut microbiome and systemic cytokine levels. METHODS Adult male mice underwent unilateral DMM or sham surgery and were then fed eight weeks of KD or chow. At baseline and every two weeks, mechanical allodynia of the operated and contralateral knees was assessed via analgesiometry. Knee joints were collected for histology, gut microbiome analysis was performed on cecal material via 16S sequencing, and serum cytokines were analyzed via Bio-Plex assay. RESULTS KD mice had worse histopathologic OA after DMM (mean ± SEM Osteoarthritis Research Society International score: KD-DMM: 4.0 ± 0.5 vs chow-DMM: 2.7 ± 0.08; P = 0.02). KD mice had increased mechanical allodynia postsurgery (P = 0.005 in mixed-effects model). The gut microbiome changed substantially with KD: 59 clades were altered by KD in DMM and 39 by KD in sham (36 were shared, 25 overlapped with previous murine OA studies). Several clades were correlated on an individual-mouse level with both histology and allodynia (eg, Lactobacillus histology P = 0.004, allodynia P = 1 × 10-4). Serum analysis showed four cytokines increased with KD (interleukin [IL]-1β, IL-2, IL-3, and IL-13). CONCLUSION KD started immediately after OA induction via DMM is associated with worsened histologic outcomes. KD also worsens mechanical allodynia after either DMM or sham surgery. KD induces significant gut microbiome dysbiosis in clades previously associated with murine OA.
Collapse
Affiliation(s)
- Gabby Dyson
- Oklahoma Medical Research Foundation, Arthritis & Clinical Immunology Program and the Oklahoma City Veterans Affairs Medical Center
| | - Montana Barrett
- Oklahoma Medical Research Foundation, Arthritis & Clinical Immunology Program and the Oklahoma City Veterans Affairs Medical Center
| | - Leoni Schlupp
- Oklahoma Medical Research Foundation, Arthritis & Clinical Immunology ProgramOklahoma City
| | - Emmaline Prinz
- Oklahoma Medical Research Foundation, Arthritis & Clinical Immunology ProgramOklahoma City
| | - Nicholas Hannebut
- Oklahoma Medical Research Foundation, Arthritis & Clinical Immunology Program and the Oklahoma City Veterans Affairs Medical Center
| | - Aleksander Szymczak
- Oklahoma Medical Research Foundation, Arthritis & Clinical Immunology ProgramOklahoma City
| | - Cindy Miranda Brawner
- Oklahoma Medical Research Foundation, Arthritis & Clinical Immunology Program and the Oklahoma City Veterans Affairs Medical Center
| | - Matlock A. Jeffries
- Oklahoma Medical Research Foundation, Arthritis & Clinical Immunology Program, the University of Oklahoma Health Sciences Center, and the Oklahoma City Veterans Affairs Medical Center
| |
Collapse
|
13
|
Turkiewicz A, Hellberg C, Dell'Isola A, Englund M. Antihistamine use and osteoarthritis or joint pain. Osteoarthritis Cartilage 2025; 33:176-179. [PMID: 39277027 DOI: 10.1016/j.joca.2024.09.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Revised: 08/13/2024] [Accepted: 09/07/2024] [Indexed: 09/17/2024]
Abstract
OBJECTIVES Antihistamines have been reported to be linked with less pain in osteoarthritis. We aimed to estimate associations between antihistamine use and three outcomes: prevalent osteoarthritis, current joint pain, and developing osteoarthritis. METHODS We included 25 003 participants of EpiHealth - a cohort of persons aged 45 to 75 from Malmö/Uppsala in Sweden. Participants self-reported the presence of allergy, joint pain and osteoarthritis at a study visit between years 2010 and 2016. Further, we obtained data about diagnoses of allergy and osteoarthritis from health-care registers (primary, specialist and inpatient care). Exposure was prescribed dispensed antihistamines (H1-antagonists) during ∼6 years preceding the EpiHealth visit retrieved from the Prescribed Drugs Register. The outcomes were osteoarthritis (any location), pain in knees/hips/hands-wrists at the examination (cross-sectional) and future incident diagnosis of osteoarthritis (longitudinal, in a cohort free of osteoarthritis at EpiHealth). We report risk ratios (95% confidence intervals [CI]) from logistic regression and hazard ratios (HR) from Cox regression, from models adjusted for age, sex, body mass index, allergy and use of healthcare. We used prescribed dispensed penicillin as negative control. RESULTS The associations between use of antihistamines and osteoarthritis/joint pain at EpiHealth were 1.13 (95%CI 1.06, 1.20) and 1.02 (0.99, 1.05), respectively. The HR of future incident osteoarthritis diagnosis with use of antihistamines was 1.15 (1.03, 1.28). The association (HR) between penicillin use and future incident osteoarthritis diagnosis was 1.16 (1.07, 1.25). CONCLUSIONS In a large population-based observational cohort, use of antihistamines was neither associated with less joint pain/osteoarthritis nor lower risk of future osteoarthritis.
Collapse
Affiliation(s)
- Aleksandra Turkiewicz
- Clinical Epidemiology Unit, Orthopedics, Clinical Sciences Lund, Lund University, Sweden.
| | - Clara Hellberg
- Clinical Epidemiology Unit, Orthopedics, Clinical Sciences Lund, Lund University, Sweden
| | - Andrea Dell'Isola
- Clinical Epidemiology Unit, Orthopedics, Clinical Sciences Lund, Lund University, Sweden
| | - Martin Englund
- Clinical Epidemiology Unit, Orthopedics, Clinical Sciences Lund, Lund University, Sweden
| |
Collapse
|
14
|
Wu X, Li H, Meng F, Lui TH, Pan X. iTRAQ proteomic analysis of exosomes derived from synovial fluid reveals disease patterns and potential biomarkers of Osteoarthritis. J Orthop Surg Res 2024; 19:849. [PMID: 39702169 DOI: 10.1186/s13018-024-05336-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Accepted: 12/03/2024] [Indexed: 12/21/2024] Open
Abstract
Exosomes extracted from synovial fluid (SF-exo) reflect the status of their originating cells. The proteomic profiles of SF-exo are important for the diagnosis of osteoarthritis (OA). To delineate the proteomic differences between SF-exo from OA patients and healthy individuals, a quantitative proteomic study based on iTRAQ technology was performed. In this study, a total of 439 proteins were identified, with 20 proteins exhibiting increased expression in the OA patient group, while 5 showed decreased expression levels. Bioinformatic analysis showed these differentially expressed proteins (DEPs) were involved in a variety of immune-related processes, including complement activation and antigen binding. For further screening, we downloaded a publicly available dataset of synovial fluid (PXD023708) and compared it with our dataset. The comparative Results identified that 5 DEPs overlapped in two datasets, and protein-protein interaction revealed that C3, C4B and APOM were key members of a tightly interactive network. Through receiver operating characteristic (ROC) curve analysis and enzyme-linked immunosorbent assay (ELISA), we confirmed 5 DEPs (C3, C4B, APOM, MMP3, DPYSL2) as potential diagnostic biomarkers for OA. And Pearson correlation analysis confirmed that most of these biomarkers had no significant linear correlation with age. Overall, our study provides the first comprehensive description of the proteomic landscape of SF-exo in OA and identifies several potential biomarkers. These findings are expected to provide valuable insights into the diagnosis and treatment of OA.
Collapse
Affiliation(s)
- Xiaomin Wu
- Department of Orthopaedics, The Second School of Clinical Medicine, Southern Medical University, The Second Affiliated Hospital of Shenzhen University, The Clinical Medical College of Guangdong Medical University, People's Hospital of Shenzhen Baoan District, Shenzhen, China
| | - Huaiming Li
- Department of Orthopaedics, The Second School of Clinical Medicine, Southern Medical University, The Second Affiliated Hospital of Shenzhen University, The Clinical Medical College of Guangdong Medical University, People's Hospital of Shenzhen Baoan District, Shenzhen, China
| | - Fengzhen Meng
- Department of Orthopaedics, The Second School of Clinical Medicine, Southern Medical University, The Second Affiliated Hospital of Shenzhen University, The Clinical Medical College of Guangdong Medical University, People's Hospital of Shenzhen Baoan District, Shenzhen, China
| | - Tun Hing Lui
- Department of Orthopaedic and Traumatology, North District Hospital, Sheung Shui, Hong Kong, China
| | - Xiaohua Pan
- Department of Orthopaedics, The Second School of Clinical Medicine, Southern Medical University, The Second Affiliated Hospital of Shenzhen University, The Clinical Medical College of Guangdong Medical University, People's Hospital of Shenzhen Baoan District, Shenzhen, China.
| |
Collapse
|
15
|
Mikaeeli Kangarshahi B, Naghib SM, Rabiee N. 3D printing and computer-aided design techniques for drug delivery scaffolds in tissue engineering. Expert Opin Drug Deliv 2024; 21:1615-1636. [PMID: 39323396 DOI: 10.1080/17425247.2024.2409913] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2024] [Revised: 09/19/2024] [Accepted: 09/24/2024] [Indexed: 09/27/2024]
Abstract
INTRODUCTION The challenge in tissue engineering lies in replicating the intricate structure of the native extracellular matrix. Recent advancements in AM, notably 3D printing, offer unprecedented capabilities to tailor scaffolds precisely, controlling properties like structure and bioactivity. CAD tools complement this by facilitating design using patient-specific data. AREA’S COVERED This review introduces additive manufacturing (AM) and computer-aided design (CAD) as pivotal tools in advancing tissue engineering, particularly cartilage regeneration. This article explores various materials utilized in AM, focusing on polymers and hydrogels for their advantageous properties in tissue engineering applications. Integrating bioactive molecules, including growth factors, into scaffolds to promote tissue regeneration is discussed alongside strategies involving different cell sources, such as stem cells, to enhance tissue development within scaffold matrices. EXPERT OPINION Applications of AM and CAD in addressing specific challenges like osteochondral defects and osteoarthritis in cartilage tissue engineering are highlighted. This review consolidates current research findings, offering expert insights into the evolving landscape of AM and CAD technologies in advancing tissue engineering, particularly in cartilage regeneration.
Collapse
Affiliation(s)
- Babak Mikaeeli Kangarshahi
- Nanotechnology Department, School of Advanced Technologies, Iran University of Science and Technology, Tehran, Iran
| | - Seyed Morteza Naghib
- Nanotechnology Department, School of Advanced Technologies, Iran University of Science and Technology, Tehran, Iran
| | - Navid Rabiee
- Department of Biomaterials, Saveetha Dental College and Hospitals, SIMATS, Saveetha University, Chennai, India
| |
Collapse
|
16
|
Jiang P, Zhou X, Yang Y, Bai L. Pectolinarigenin targeting FGFR3 alleviates osteoarthritis progression by regulating the NF-κB/NLRP3 inflammasome pyroptotic pathway. Int Immunopharmacol 2024; 140:112741. [PMID: 39094365 DOI: 10.1016/j.intimp.2024.112741] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Revised: 07/08/2024] [Accepted: 07/19/2024] [Indexed: 08/04/2024]
Abstract
OBJECTIVE Osteoarthritis (OA) is a chronic degenerative disease characterized by cartilage degeneration, involving inflammation, pyroptosis, and degeneration of the extracellular matrix (ECM). Pectolinarigenin (PEC) is a natural flavonoid with antioxidant, anti-inflammatory and anti-tumor properties. This study aims to explore the potential of PEC in ameliorating OA progression and its underlying mechanisms. METHODS Chondrocytes were exposed to 10 ng/mL IL-1β to simulate OA-like changes. The effect of PEC on IL-1β-treated chondrocytes was assessed using ELISA, western blot, and immunofluorescence. The mRNA sequencing (mRNA-seq) was employed to explore the possible targets of PEC in delaying OA progression. The OA mouse model was induced through anterior cruciate ligament transection (ACLT) and divided into sham, ACLT, ACLT+5 mg/kg PEC, and ACLT+10 mg/kg PEC groups. Micro-computed tomography and histological analysis were conducted to confirm the beneficial effects of PEC on OA in vivo. RESULTS PEC mitigated chondrocyte pyroptosis, as evidenced by reduced levels of pyroptosis-related proteins. Additionally, PEC attenuated IL-1β-mediated chondrocyte ECM degradation and inflammation. Mechanistically, mRNA-seq showed that FGFR3 was a downstream target of PEC. FGFR3 silencing reversed the beneficial effects of PEC on IL-1β-exposed chondrocytes. PEC exerted anti-pyroptotic, anti-ECM degradative, and anti-inflammatory effects through upregulating FGFR3 to inhibit the NF-κB/NLRP3 pyroptosis-related pathway. Consistently, in vivo experiments demonstrated the chondroprotective effects of PEC in OA mice. CONCLUSION PEC alleviate OA progression by FGFR3/NF-κB/NLRP3 pathway mediated chondrocyte pyroptosis, ECM degradation and inflammation, suggesting the potential of PEC as a therapeutic agent for OA.
Collapse
Affiliation(s)
- Peng Jiang
- Department of Orthopedics, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| | - Xiaonan Zhou
- Department of Orthopedics, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| | - Yue Yang
- Department of Orthopedics, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| | - Lunhao Bai
- Department of Orthopedics, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China.
| |
Collapse
|
17
|
Ulrich M, Lehnick D, Strobel K, Grünig H, Lima T, Iselin L, Bhure U. Quantitative bone single photon emission computed tomography/computed tomography in symptomatic and asymptomatic foot and ankle osteoarthritis. Nucl Med Commun 2024; 45:848-857. [PMID: 39011813 DOI: 10.1097/mnm.0000000000001878] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/17/2024]
Abstract
PURPOSE The purpose of this study was to evaluate and quantify the prevalence of increased uptake in SPECT/CT in symptomatic and asymptomatic foot and ankle joints in patients with osteoarthritis. METHODS In 63 patients with osteoarthritis (OA), the painful symptomatic foot (SF) and asymptomatic contralateral foot (AF) were imaged with bone SPECT/CT. Presence, localization, and maximum standardized uptake value (SUV max ) of the active joints were assessed for SF and AF. CT OA grade (grade 1: mild, grade 2: moderate, grade 3: severe) and presence of five morphological features of OA (joint space narrowing, subchondral sclerosis, subchondral cysts, irregular joint margins, and osteophytes) were evaluated. RESULTS In total 32 (51%) patients showed additional uptake in the AF, whereas 31 (49%) patients showed it only in the SF. SF showed more active joints than AF (106 vs. 43). CT OA grades positively correlated with SUV max (Kendall's tau b = 0.62, P < 0.001). SUV max values (per foot) in SF were higher in patients with uptake in bilateral feet (SF+, AF+) [median (IQR): 17.9 (10.7-23.3)] as compared with patients with active sites only in the SF (SF+, AF-) [10.4 (6.4-19.1); P < 0.001]. Number of active OA joints in SF was higher in patients with bilateral uptake ( P = 0.017). CONCLUSION In conclusion, half of the patients exhibited increased uptake in the contralateral asymptomatic foot. SUV max showed a significant correlation to CT osteoarthritis grade, in the symptomatic and asymptomatic foot. Future follow-up studies will provide further insights into the prognostic and therapeutic value of these findings.
Collapse
Affiliation(s)
- Martin Ulrich
- Department of Orthopaedics, University of Zurich, Stadtspital, Zurich,
| | - Dirk Lehnick
- Department of Health Sciences and Medicine, University of Lucerne,
- Department of Biostatistics & Methodology, Clinical Trial Unit Central Switzerland, University of Lucerne,
| | - Klaus Strobel
- Department of Nuclear Medicine and Radiology, Cantonal Hospital Lucerne and
| | - Hannes Grünig
- Department of Nuclear Medicine and Radiology, Cantonal Hospital Lucerne and
| | - Thiago Lima
- Department of Health Sciences and Medicine, University of Lucerne,
- Department of Nuclear Medicine and Radiology, Cantonal Hospital Lucerne and
| | - Lukas Iselin
- Department of Orthopaedics, Orthopädische Klinik, Klinik St.Anna, Lucerne, Switzerland
| | - Ujwal Bhure
- Department of Nuclear Medicine and Radiology, Cantonal Hospital Lucerne and
| |
Collapse
|
18
|
Sheng W, Yue Y, Qi T, Qin H, Liu P, Wang D, Zeng H, Yu F. The Multifaceted Protective Role of Nuclear Factor Erythroid 2-Related Factor 2 in Osteoarthritis: Regulation of Oxidative Stress and Inflammation. J Inflamm Res 2024; 17:6619-6633. [PMID: 39329083 PMCID: PMC11424688 DOI: 10.2147/jir.s479186] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Accepted: 09/05/2024] [Indexed: 09/28/2024] Open
Abstract
Osteoarthritis (OA) is a chronic degenerative joint disease characterized by the degradation of joint cartilage, subchondral bone sclerosis, synovitis, and structural changes in the joint. Recent research has highlighted the role of various genes in the pathogenesis and progression of OA, with nuclear factor erythroid 2-related factor 2 (NRF2) emerging as a critical player. NRF2, a vital transcription factor, plays a key role in regulating the OA microenvironment and slowing the disease's progression. It modulates the expression of several antioxidant enzymes, such as Heme oxygenase-1 (HO-1) and NAD(P)H oxidoreductase 1 (NQO1), among others, which help reduce oxidative stress. Furthermore, NRF2 inhibits the nuclear factor kappa-B (NF-κB) signaling pathway, thereby decreasing inflammation, joint pain, and the breakdown of cartilage extracellular matrix, while also mitigating cell aging and death. This review discusses NRF2's impact on oxidative stress, inflammation, cell aging, and various cell death modes (such as apoptosis, necroptosis, and ferroptosis) in OA-affected chondrocytes. The role of NRF2 in OA macrophages, and synovial fibroblasts was also discussed. It also covers NRF2's role in preserving the cartilage extracellular matrix and alleviating joint pain. The purpose of this review is to provide a comprehensive understanding of NRF2's protective mechanisms in OA, highlighting its potential as a therapeutic target and underscoring its significance in the development of novel treatment strategies for OA.
Collapse
Affiliation(s)
- Weibei Sheng
- Department of Bone & Joint Surgery, National & Local Joint Engineering Research Center of Orthopaedic Biomaterials, Shenzhen Key Laboratory of Orthopaedic Diseases and Biomaterials Research, Peking University Shenzhen Hospital, Shenzhen, 518036, People’s Republic of China
| | - Yaohang Yue
- Department of Bone & Joint Surgery, National & Local Joint Engineering Research Center of Orthopaedic Biomaterials, Shenzhen Key Laboratory of Orthopaedic Diseases and Biomaterials Research, Peking University Shenzhen Hospital, Shenzhen, 518036, People’s Republic of China
| | - Tiantian Qi
- Department of Bone & Joint Surgery, National & Local Joint Engineering Research Center of Orthopaedic Biomaterials, Shenzhen Key Laboratory of Orthopaedic Diseases and Biomaterials Research, Peking University Shenzhen Hospital, Shenzhen, 518036, People’s Republic of China
| | - Haotian Qin
- Department of Bone & Joint Surgery, National & Local Joint Engineering Research Center of Orthopaedic Biomaterials, Shenzhen Key Laboratory of Orthopaedic Diseases and Biomaterials Research, Peking University Shenzhen Hospital, Shenzhen, 518036, People’s Republic of China
| | - Peng Liu
- Department of Bone & Joint Surgery, National & Local Joint Engineering Research Center of Orthopaedic Biomaterials, Shenzhen Key Laboratory of Orthopaedic Diseases and Biomaterials Research, Peking University Shenzhen Hospital, Shenzhen, 518036, People’s Republic of China
| | - Deli Wang
- Department of Bone & Joint Surgery, National & Local Joint Engineering Research Center of Orthopaedic Biomaterials, Shenzhen Key Laboratory of Orthopaedic Diseases and Biomaterials Research, Peking University Shenzhen Hospital, Shenzhen, 518036, People’s Republic of China
| | - Hui Zeng
- Shenzhen Second People’s Hospital, The First Affiliated Hospital of Shenzhen University, Shenzhen, 518035, People’s Republic of China
| | - Fei Yu
- Department of Bone & Joint Surgery, National & Local Joint Engineering Research Center of Orthopaedic Biomaterials, Shenzhen Key Laboratory of Orthopaedic Diseases and Biomaterials Research, Peking University Shenzhen Hospital, Shenzhen, 518036, People’s Republic of China
| |
Collapse
|
19
|
Chatterjee P, Stevens HY, Kippner LE, Bowles-Welch AC, Drissi H, Mautner K, Yeago C, Gibson G, Roy K. Single-cell transcriptome and crosstalk analysis reveals immune alterations and key pathways in the bone marrow of knee OA patients. iScience 2024; 27:110827. [PMID: 39310769 PMCID: PMC11416684 DOI: 10.1016/j.isci.2024.110827] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Revised: 06/10/2024] [Accepted: 08/22/2024] [Indexed: 09/25/2024] Open
Abstract
Knee osteoarthritis (OA) is a significant medical and economic burden. To understand systemic immune effects, we performed deep exploration of bone marrow aspirate concentrates (BMACs) from knee-OA patients via single-cell RNA sequencing and proteomic analyses from a randomized clinical trial (MILES: NCT03818737). We found significant cellular and immune alterations in the bone marrow, specifically in MSCs, T cells and NK cells, along with changes in intra-tissue cellular crosstalk during OA progression. Unlike previous studies focusing on injury sites or peripheral blood, our probe into the bone marrow-an inflammation and immune regulation hub-highlights remote organ impact of OA, identifying cell types and pathways for potential therapeutic targeting. Our findings highlight increased cellular senescence and inflammatory pathways, revealing key upstream genes, transcription factors, and ligands. Additionally, we identified significant enrichment in key biological pathways like PI3-AKT-mTOR signaling and IFN responses, showing their potentially crucial role in OA onset and progression.
Collapse
Affiliation(s)
- Paramita Chatterjee
- Marcus Center for Therapeutic Cell Characterization and Manufacturing, The Parker H. Petit Institute for Bioengineering and Biosciences, Georgia Institute of Technology, Atlanta, GA, USA
- The Parker H. Petit Institute for Bioengineering and Biosciences Georgia Institute of Technology, Atlanta, GA, USA
| | - Hazel Y. Stevens
- Marcus Center for Therapeutic Cell Characterization and Manufacturing, The Parker H. Petit Institute for Bioengineering and Biosciences, Georgia Institute of Technology, Atlanta, GA, USA
- The Parker H. Petit Institute for Bioengineering and Biosciences Georgia Institute of Technology, Atlanta, GA, USA
| | - Linda E. Kippner
- Marcus Center for Therapeutic Cell Characterization and Manufacturing, The Parker H. Petit Institute for Bioengineering and Biosciences, Georgia Institute of Technology, Atlanta, GA, USA
- The Parker H. Petit Institute for Bioengineering and Biosciences Georgia Institute of Technology, Atlanta, GA, USA
| | - Annie C. Bowles-Welch
- Marcus Center for Therapeutic Cell Characterization and Manufacturing, The Parker H. Petit Institute for Bioengineering and Biosciences, Georgia Institute of Technology, Atlanta, GA, USA
- The Parker H. Petit Institute for Bioengineering and Biosciences Georgia Institute of Technology, Atlanta, GA, USA
| | - Hicham Drissi
- Department of Orthopaedics, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Kenneth Mautner
- Department of Orthopaedics, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Carolyn Yeago
- The Parker H. Petit Institute for Bioengineering and Biosciences Georgia Institute of Technology, Atlanta, GA, USA
| | - Greg Gibson
- School of Biological Sciences, Georgia Institute of Technology, Atlanta, GA 30332, USA
| | - Krishnendu Roy
- Marcus Center for Therapeutic Cell Characterization and Manufacturing, The Parker H. Petit Institute for Bioengineering and Biosciences, Georgia Institute of Technology, Atlanta, GA, USA
- Department of Biomedical Engineering, School of Engineering, Vanderbilt University, Nashville, TN, USA
- Department of Pathology, Microbiology and Immunology, School of Medicine, Vanderbilt University, Nashville, TN, USA
- Department of Chemical and Biomolecular Engineering, School of Engineering, Vanderbilt University, Nashville, TN, USA
| |
Collapse
|
20
|
Bergman RF, Lammlin L, Junginger L, Farrell E, Goldman S, Darcy R, Rasner C, Obeidat AM, Malfait AM, Miller RE, Maerz T. Sexual dimorphism of the synovial transcriptome underpins greater PTOA disease severity in male mice following joint injury. Osteoarthritis Cartilage 2024; 32:1060-1073. [PMID: 37716404 DOI: 10.1016/j.joca.2023.07.012] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Revised: 06/20/2023] [Accepted: 07/26/2023] [Indexed: 09/18/2023]
Abstract
OBJECTIVE Osteoarthritis (OA) is a disease with sex-dependent prevalence and severity in both human and animal models. We sought to elucidate sex differences in synovitis, mechanical sensitization, structural damage, bone remodeling, and the synovial transcriptome in the anterior cruciate ligament rupture (ACLR) mouse model of post-traumatic OA (PTOA). DESIGN Male and female 12-week-old C57/BL6J mice were randomized to Sham or noninvasive ACLR with harvests at 7d or 28d post-ACLR (n = 9 per sex in each group - Sham, 7d ACLR, 28d ACLR). Knee hyperalgesia, mechanical allodynia, and intra-articular matrix metalloproteinase (MMP) activity (via intravital imaging) were measured longitudinally. Trabecular and subchondral bone (SCB) remodeling and osteophyte formation were assessed by µCT. Histological scoring of PTOA, synovitis, and anti-MMP13 immunostaining were performed. NaV1.8-Cre;tdTomato mice were used to document localization and sprouting of nociceptors. Bulk RNA-seq of synovium in Sham, 7d, and 28d post-ACLR, and contralateral joints (n = 6 per group per sex) assessed injury-induced and sex-dependent gene expression. RESULTS Male mice exhibited more severe joint damage at 7d and 28d and more severe synovitis at 28d, accompanied by 19% greater MMP activity, 8% lower knee hyperalgesia threshold, and 43% lower hindpaw withdrawal threshold in injured limbs compared to female injured limbs. Females had injury-induced catabolic responses in trabecular and SCB, whereas males exhibited 133% greater normalized osteophyte volume relative to females and sclerotic remodeling of trabecular and SCB. NaV1.8+ nociceptor sprouting in SCB and medial synovium was induced by injury and comparable between sexes. RNA-seq of synovium demonstrated similar injury-induced transcriptomic programs between the sexes at 7d, but only female mice exhibited a transcriptomic signature indicative of synovial inflammatory resolution by 28d, whereas males had persistent pro-inflammatory, pro-fibrotic, pro-neurogenic, and pro-angiogenic gene expression. CONCLUSION Male mice exhibited more severe overall joint damage and pain behavior after ACLR, which was associated with persistent activation of synovial inflammatory, fibrotic, and neuroangiogenic processes, implicating persistent synovitis in driving sex differences in murine PTOA.
Collapse
Affiliation(s)
- Rachel F Bergman
- Orthopaedic Research Laboratories, Department of Orthopaedic Surgery, University of Michigan, Ann Arbor, MI, United States
| | - Lindsey Lammlin
- Orthopaedic Research Laboratories, Department of Orthopaedic Surgery, University of Michigan, Ann Arbor, MI, United States
| | - Lucas Junginger
- Orthopaedic Research Laboratories, Department of Orthopaedic Surgery, University of Michigan, Ann Arbor, MI, United States
| | - Easton Farrell
- Orthopaedic Research Laboratories, Department of Orthopaedic Surgery, University of Michigan, Ann Arbor, MI, United States
| | - Sam Goldman
- Orthopaedic Research Laboratories, Department of Orthopaedic Surgery, University of Michigan, Ann Arbor, MI, United States
| | - Rose Darcy
- Orthopaedic Research Laboratories, Department of Orthopaedic Surgery, University of Michigan, Ann Arbor, MI, United States
| | - Cody Rasner
- Orthopaedic Research Laboratories, Department of Orthopaedic Surgery, University of Michigan, Ann Arbor, MI, United States
| | - Alia M Obeidat
- Department of Internal Medicine, Division of Rheumatology, Rush University, Chicago, IL, United States
| | - Anne-Marie Malfait
- Department of Internal Medicine, Division of Rheumatology, Rush University, Chicago, IL, United States
| | - Rachel E Miller
- Department of Internal Medicine, Division of Rheumatology, Rush University, Chicago, IL, United States
| | - Tristan Maerz
- Orthopaedic Research Laboratories, Department of Orthopaedic Surgery, University of Michigan, Ann Arbor, MI, United States.
| |
Collapse
|
21
|
Zhu S, Qu W, He C. Evaluation and management of knee osteoarthritis. J Evid Based Med 2024; 17:675-687. [PMID: 38963824 DOI: 10.1111/jebm.12627] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Accepted: 06/18/2024] [Indexed: 07/06/2024]
Abstract
Knee osteoarthritis (KOA) significantly contributes to the global disability burden, with its incidence expected to escalate by 74.9% by 2050. The urgency to comprehend and tackle this condition is critical, necessitating an updated and thorough review of KOA. A systematic review up to February 26, 2024, has elucidated the principal aspects of KOA's pathogenesis, risk factors, clinical manifestations, and contemporary management paradigms. The origins of KOA are intricately linked to mechanical, inflammatory, and metabolic disturbances that impair joint function. Notable risk factors include age, obesity, and previous knee injuries. Diagnosis predominantly relies on clinical assessment, with radiographic evaluation reserved conditionally. The significance of rehabilitation assessments, informed by the International Classification of Functioning, Disability, and Health framework, is highlighted. Treatment strategies are diverse, prioritizing nonpharmacological measures such as patient education, exercise, and weight management, with pharmacological interventions considered adjuncts. Intra-articular injections and surgical options are contemplated for instances where conventional management is inadequate. KOA stands as a predominant disability cause globally, characterized by a complex etiology and profound effects on individuals' quality of life. Early, proactive management focusing on nonpharmacological interventions forms the cornerstone of treatment, aiming to alleviate symptoms and enhance joint function. This comprehensive review underscores the need for early diagnosis, individualized treatment plans, and the integration of rehabilitation assessments to optimize patient outcomes. Further research is needed to refine prevention strategies and improve management outcomes for KOA patients.
Collapse
Affiliation(s)
- Siyi Zhu
- Rehabilitation Medicine Center and Institute of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu, China
- Key Laboratory of Rehabilitation Medicine in Sichuan Province, West China Hospital, Sichuan University, Chengdu, China
| | - Wenchun Qu
- Department of Pain Medicine, Mayo Clinic, Jacksonville, Florida, USA
| | - Chengqi He
- Rehabilitation Medicine Center and Institute of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu, China
- Key Laboratory of Rehabilitation Medicine in Sichuan Province, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
22
|
Butarbutar JC, Basuki P, Sungono V, Riantho A, Fidiasrianto K. Burden of osteoarthritis in Indonesia: A Global Burden of Disease (GBD) study 2019. NARRA J 2024; 4:e884. [PMID: 39280323 PMCID: PMC11391984 DOI: 10.52225/narra.v4i2.884] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Accepted: 08/02/2024] [Indexed: 09/18/2024]
Abstract
Osteoarthritis (OA) is a complex and common condition, especially affecting the knees due to their weight-bearing role. Traditionally seen as a degenerative disease, OA is now understood to have both mechanical and inflammatory causes. Despite its increasing prevalence, there is limited data on OA in Indonesia, resulting in low awareness among clinicians and the public. The aim of this study was to describe the OA burden in Indonesia, focusing on its prevalence, incidence, and years lived with disability (YLD) from 1990 to 2019, using data from the Global Burden of Disease (GBD) study 2019. A descriptive cross-sectional study was conducted to examine the prevalence, incidence, and YLD of OA in Indonesia from the GBD study 2019. OA prevalence and YLD were compared to other countries according to similar social demographics and geographical proximity. OA YLD was also compared to the top causes of death and disability YLD in Indonesia. The study found that OA cases in Indonesia more than doubled from 1990 to 2019, with increases of 153.12% in males and 143.36% in females. Similar trends were observed for knee OA. The age-standardized prevalence rate in Indonesia increased by 11.03% in males and 8.42% in females, and these were higher compared to China, India, Singapore, and the global average. Younger people had a higher OA prevalence rate growth than older groups. The incidence rate for OA also rose significantly, with males seeing a 10.89% increase to 290 per 100,000 people and females with an 8.57% increase to 384 per 100,000 people. Despite lower overall burden rates compared to some countries, Indonesia experienced significant growth in YLD due to OA (12.16% in males and 9.65% in females) since 1990. Although OA was less burdensome than stroke, diabetes, low back pain, and chronic obstructive pulmonary disease (COPD), its YLD growth rate was higher. In conclusion, OA prevalence and incidence in Indonesia significantly increased from 1990 to 2019, with a notable rise among younger populations. OA had a higher YLD growth compared to several other major diseases in Indonesia, highlighting the need for early detection and preventive measures, particularly for the younger population.
Collapse
Affiliation(s)
- John Cp Butarbutar
- Department of Orthopedics and Traumatology, Faculty of Medicine, Universitas Pelita Harapan, Tangerang, Indonesia
- Department of Orthopedics and Traumatology, Siloam Hospitals Lippo Village, Tangerang, Indonesia
| | - Pamela Basuki
- Faculty of Medicine, Universitas Indonesia, Jakarta, Indonesia
| | - Veli Sungono
- Department of Public Health, Faculty of Medicine, Universitas Pelita Harapan, Tangerang, Indonesia
| | - Albert Riantho
- Department of Orthopedics and Traumatology, Faculty of Medicine, Universitas Pelita Harapan, Tangerang, Indonesia
- Department of Orthopedics and Traumatology, Siloam Hospitals Lippo Village, Tangerang, Indonesia
| | - Kevin Fidiasrianto
- Department of Orthopedics and Traumatology, Faculty of Medicine, Universitas Pelita Harapan, Tangerang, Indonesia
- Department of Orthopedics and Traumatology, Siloam Hospitals Lippo Village, Tangerang, Indonesia
| |
Collapse
|
23
|
Lawrence M, Goyal A, Pathak S, Ganguly P. Cellular Senescence and Inflammaging in the Bone: Pathways, Genetics, Anti-Aging Strategies and Interventions. Int J Mol Sci 2024; 25:7411. [PMID: 39000517 PMCID: PMC11242738 DOI: 10.3390/ijms25137411] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Revised: 06/22/2024] [Accepted: 07/03/2024] [Indexed: 07/16/2024] Open
Abstract
Advancing age is associated with several age-related diseases (ARDs), with musculoskeletal conditions impacting millions of elderly people worldwide. With orthopedic conditions contributing towards considerable number of patients, a deeper understanding of bone aging is the need of the hour. One of the underlying factors of bone aging is cellular senescence and its associated senescence associated secretory phenotype (SASP). SASP comprises of pro-inflammatory markers, cytokines and chemokines that arrest cell growth and development. The accumulation of SASP over several years leads to chronic low-grade inflammation with advancing age, also known as inflammaging. The pathways and molecular mechanisms focused on bone senescence and inflammaging are currently limited but are increasingly being explored. Most of the genes, pathways and mechanisms involved in senescence and inflammaging coincide with those associated with cancer and other ARDs like osteoarthritis (OA). Thus, exploring these pathways using techniques like sequencing, identifying these factors and combatting them with the most suitable approach are crucial for healthy aging and the early detection of ARDs. Several approaches can be used to aid regeneration and reduce senescence in the bone. These may be pharmacological, non-pharmacological and lifestyle interventions. With increasing evidence towards the intricate relationship between aging, senescence, inflammation and ARDs, these approaches may also be used as anti-aging strategies for the aging bone marrow (BM).
Collapse
Affiliation(s)
- Merin Lawrence
- School of Biological and Chemical Sciences, University of Galway, H91W2TY Galway, Ireland
| | - Abhishek Goyal
- RAS Life Science Solutions, Stresemannallee 61, 60596 Frankfurt, Germany
| | - Shelly Pathak
- Observational and Pragmatic Research Institute, 5 Coles Lane, Oakington, Cambridge CB24 3BA, UK
| | - Payal Ganguly
- Leeds Institute of Rheumatic and Musculoskeletal Medicine, University of Leeds, Leeds LS9 7JT, UK
| |
Collapse
|
24
|
Mariano A, Ammendola S, Migliorini A, Leopizzi M, Raimondo D, Scotto d'Abusco A. Intron retention in PI-PLC γ1 mRNA as a key mechanism affecting MMP expression in human primary fibroblast-like synovial cells. Cell Biochem Funct 2024; 42:e4091. [PMID: 38973151 DOI: 10.1002/cbf.4091] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Revised: 06/26/2024] [Accepted: 06/30/2024] [Indexed: 07/09/2024]
Abstract
The intron retention (IR) is a phenomenon utilized by cells to allow diverse fates at the same mRNA, leading to a different pattern of synthesis of the same protein. In this study, we analyzed the modulation of phosphoinositide-specific phospholipase C (PI-PLC) enzymes by Harpagophytum procumbens extract (HPE) in synoviocytes from joins of osteoarthritis (OA) patients. In some samples, the PI-PLC γ1 isoform mature mRNA showed the IR and, in these synoviocytes, the HPE treatment increased the phenomenon. Moreover, we highlighted that as a consequence of IR, a lower amount of PI-PLC γ1 was produced. The decrease of PI-PLC γ1 was associated with the decrease of metalloprotease-3 (MMP-3), and MMP-13, and ADAMTS-5 after HPE treatment. The altered expression of MMPs is a hallmark of the onset and progression of OA, thus substances able to decrease their expression are very desirable. The interesting outcomes of this study are that 35% of analyzed synovial tissues showed the IR phenomenon in the PI-PLC γ1 mRNA and that the HPE treatment increased this phenomenon. For the first time, we found that the decrease of PI-PLC γ1 protein in synoviocytes interferes with MMP production, thus affecting the pathways involved in the MMP expression. This finding was validated by the silencing of PI-PLC γ1 in synoviocytes where the IR phenomenon was not present. Our results shed new light on the biochemical mechanisms involved in the degrading enzyme production in the joint of OA patients, suggesting a new therapeutic target and highlighting the importance of personalized medicine.
Collapse
Affiliation(s)
- Alessia Mariano
- Department of Biochemical Sciences, Sapienza University of Rome, Roma, Italy
| | | | - Arianna Migliorini
- Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy
| | - Martina Leopizzi
- Department of Medico-Surgical Sciences and Biotechnologies, Polo Pontino-Sapienza University, Latina, Italy
| | - Domenico Raimondo
- Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy
| | | |
Collapse
|
25
|
Gusev E, Sarapultsev A. Exploring the Pathophysiology of Long COVID: The Central Role of Low-Grade Inflammation and Multisystem Involvement. Int J Mol Sci 2024; 25:6389. [PMID: 38928096 PMCID: PMC11204317 DOI: 10.3390/ijms25126389] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Revised: 06/05/2024] [Accepted: 06/07/2024] [Indexed: 06/28/2024] Open
Abstract
Long COVID (LC), also referred to as Post COVID-19 Condition, Post-Acute Sequelae of SARS-CoV-2 Infection (PASC), and other terms, represents a complex multisystem disease persisting after the acute phase of COVID-19. Characterized by a myriad of symptoms across different organ systems, LC presents significant diagnostic and management challenges. Central to the disorder is the role of low-grade inflammation, a non-classical inflammatory response that contributes to the chronicity and diversity of symptoms observed. This review explores the pathophysiological underpinnings of LC, emphasizing the importance of low-grade inflammation as a core component. By delineating the pathogenetic relationships and clinical manifestations of LC, this article highlights the necessity for an integrated approach that employs both personalized medicine and standardized protocols aimed at mitigating long-term consequences. The insights gained not only enhance our understanding of LC but also inform the development of therapeutic strategies that could be applicable to other chronic conditions with similar pathophysiological features.
Collapse
Affiliation(s)
| | - Alexey Sarapultsev
- Institute of Immunology and Physiology, Ural Branch of the Russian Academy of Science, 620049 Ekaterinburg, Russia;
| |
Collapse
|
26
|
Lee JM, Lim S, Kang G, Chung JY, Yun HW, Jin YJ, Park DY, Park JY. Synovial fluid monocyte-to-lymphocyte ratio in knee osteoarthritis patients predicts patient response to conservative treatment: a retrospective cohort study. BMC Musculoskelet Disord 2024; 25:379. [PMID: 38745277 PMCID: PMC11092220 DOI: 10.1186/s12891-024-07475-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/05/2023] [Accepted: 04/24/2024] [Indexed: 05/16/2024] Open
Abstract
BACKGROUND Biomarkers that predict the treatment response in patients with knee osteoarthritis are scarce. This study aimed to investigate the potential role of synovial fluid cell counts and their ratios as biomarkers of primary knee osteoarthritis. METHODS This retrospective study investigated 96 consecutive knee osteoarthritis patients with knee effusion who underwent joint fluid aspiration analysis and received concomitant intra-articular corticosteroid injections and blood tests. The monocyte-to-lymphocyte ratio (MLR) and neutrophil-to-lymphocyte ratio (NLR) were calculated. After 6 months of treatment, patients were divided into two groups: the responder group showing symptom resolution, defined by a visual analog scale (VAS) score of ≤ 3, without additional treatment, and the non-responder group showing residual symptoms, defined by a VAS score of > 3 and requiring further intervention, such as additional medication, repeated injections, or surgical treatment. Unpaired t-tests and univariate and multivariate logistic regression analyses were conducted between the two groups to predict treatment response after conservative treatment. The predictive value was calculated using the area under the receiver operating characteristic curve, and the optimal cutoff value was determined. RESULTS Synovial fluid MLR was significantly higher in the non-responder group compared to the responder group (1.86 ± 1.64 vs. 1.11 ± 1.37, respectively; p = 0.02). After accounting for confounding variables, odds ratio of non-responder due to increased MLR were 1.63 (95% confidence interval: 1.11-2.39). The optimal MLR cutoff value for predicting patient response to conservative treatment was 0.941. CONCLUSIONS MLR may be a potential biomarker for predicting the response to conservative treatment in patients with primary knee osteoarthritis.
Collapse
Affiliation(s)
- Jong Min Lee
- Department of Orthopaedic Surgery, School of Medicine, Ajou University, Suwon, South Korea
| | - Sumin Lim
- Department of Orthopaedic Surgery, School of Medicine, Ajou University, Suwon, South Korea
| | - Gunoo Kang
- Department of Orthopaedic Surgery, School of Medicine, Ajou University, Suwon, South Korea
| | - Jun Young Chung
- Department of Orthopaedic Surgery, School of Medicine, Ajou University, Suwon, South Korea
| | - Hee-Woong Yun
- Department of Orthopaedic Surgery, School of Medicine, Ajou University, Suwon, South Korea
- Cell Therapy Center, Ajou Medical Center, Suwon, Republic of Korea
| | - Yong Jun Jin
- Department of Orthopaedic Surgery, School of Medicine, Ajou University, Suwon, South Korea
- Cell Therapy Center, Ajou Medical Center, Suwon, Republic of Korea
| | - Do Young Park
- Department of Orthopaedic Surgery, School of Medicine, Ajou University, Suwon, South Korea.
- Cell Therapy Center, Ajou Medical Center, Suwon, Republic of Korea.
- Leading Convergence of Healthcare and Medicine, Ajou University, Institute of Science & Technology (ALCHeMIST), Suwon, Republic of Korea.
| | - Jae-Young Park
- Department of Orthopaedic Surgery, Uijeongbu Eulji Medical Center, Eulji University School of Medicine, Uijeongbu-si, Republic of Korea.
- Department of Orthopaedic Surgery, CHA University, CHA Bundang Medical Center, Seongnam-si, Gyeonggi-do, Republic of Korea.
| |
Collapse
|
27
|
Zhao J, Lai Y, Zeng L, Liang G, Jin X, Huang H, Luo M, Liu J. Inverse association of the systemic immune-inflammation index with serum anti-ageing protein Klotho levels in individuals with osteoarthritis: A cross-sectional study. PLoS One 2024; 19:e0300674. [PMID: 38713671 PMCID: PMC11075862 DOI: 10.1371/journal.pone.0300674] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Accepted: 03/01/2024] [Indexed: 05/09/2024] Open
Abstract
BACKGROUND The association between the systemic immune-inflammation index (SII) and the serum soluble-Klotho concentration (pg/ml) in osteoarthritis (OA) patients is unknown. This study aimed to investigate the relationship between the SII and serum soluble-Klotho levels in OA patients. METHODS All study data were obtained from the National Health and Nutrition Examination Survey (NHANES) database (n = 1852 OA patients; age range = 40-79 years). The SII and serum Klotho measurement data are from the NHANES mobile examination centre. The SII values were divided into quartiles (Q1-4: 0.02-3.36, 3.36-4.78, 4.79-6.70, and 6.70-41.75). A multivariate linear regression model was constructed to evaluate the association between the SII and serum Klotho levels in OA patients; interaction tests were conducted to test the stability of the statistical results. RESULTS Multivariate linear regression revealed a negative linear relationship between the SII and serum Klotho concentration in OA patients (β = -6.05; 95% CI: -9.72, -2.39). Compared to Q1, Q4 was associated with lower serum Klotho concentrations (β = -59.93; 95% CI: -96.57, -23.28). Compared with that of Q1, the β value of Q2-Q4 showed a downwards trend as the SII increased (Ptrend <0.001). The stratified analysis results indicated that the SII had a greater sensitivity in predicting serum Klotho concentrations in OA patients aged 60-79 years (Pinteraction = 0.028). CONCLUSIONS There was a significant negative linear correlation between the SII and serum Klotho concentration in OA patients. The SII can serve as a predictive indicator of serum Klotho concentrations in OA patients. Klotho may be a potential anti-inflammatory drug for OA treatment.
Collapse
Affiliation(s)
- Jinlong Zhao
- The Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, China
- The Second Affiliated Hospital of Guangzhou University of Chinese Medicine (Guangdong Provincial Hospital of Chinese Medicine), Guangzhou, China
- The Research Team on Bone and Joint Degeneration and Injury of Guangdong Provincial Academy of Chinese Medical Sciences, Guangzhou, China
| | - Yinhua Lai
- The Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Lingfeng Zeng
- The Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, China
- The Second Affiliated Hospital of Guangzhou University of Chinese Medicine (Guangdong Provincial Hospital of Chinese Medicine), Guangzhou, China
- The Research Team on Bone and Joint Degeneration and Injury of Guangdong Provincial Academy of Chinese Medical Sciences, Guangzhou, China
| | - Guihong Liang
- The Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, China
- The Second Affiliated Hospital of Guangzhou University of Chinese Medicine (Guangdong Provincial Hospital of Chinese Medicine), Guangzhou, China
- The Research Team on Bone and Joint Degeneration and Injury of Guangdong Provincial Academy of Chinese Medical Sciences, Guangzhou, China
| | - Xiao Jin
- The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Hetao Huang
- The Second Affiliated Hospital of Guangzhou University of Chinese Medicine (Guangdong Provincial Hospital of Chinese Medicine), Guangzhou, China
| | - Minghui Luo
- The Second Affiliated Hospital of Guangzhou University of Chinese Medicine (Guangdong Provincial Hospital of Chinese Medicine), Guangzhou, China
| | - Jun Liu
- The Research Team on Bone and Joint Degeneration and Injury of Guangdong Provincial Academy of Chinese Medical Sciences, Guangzhou, China
- Guangdong Second Chinese Medicine Hospital (Guangdong Province Engineering Technology Research Institute of Traditional Chinese Medicine), Guangzhou, China
| |
Collapse
|
28
|
Hao Y, Tang X, Xu F. Association between hyperuricemia and the risk of mortality in patients with osteoarthritis: A study based on the National Health and Nutrition Examination Survey database. PLoS One 2024; 19:e0302386. [PMID: 38713669 DOI: 10.1371/journal.pone.0302386] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Accepted: 04/02/2024] [Indexed: 05/09/2024] Open
Abstract
BACKGROUND The purpose of this study was to evaluate the relationship between hyperuricemia and the risks of all-cause mortality and cardiovascular disease (CVD) mortality in patients with osteoarthritis (OA). METHODS A retrospective cohort study was performed on 3,971 patients using data from the National Health and Nutrition Examination Survey database between 1999 and 2018. OA was diagnosed through specific questions and responses. The weighted COX regression models were used to explore the factors associated with all-cause mortality/CVD mortality in OA patients. Subgroup analyses were conducted based on age, gender, hypertension, dyslipidemia, CVD, and chronic kidney disease (CKD). Hazard ratio (HR) and 95% confidence interval (95% CI) were measured as the evaluation indexes. RESULTS During the duration of follow-up time (116.38 ± 2.19 months), 33.69% (1,338 patients) experienced all-cause mortality, and 11.36% (451 patients) died from CVD. Hyperuricemia was associated with higher risks of all-cause mortality (HR: 1.22, 95% CI: 1.06-1.41, P = 0.008) and CVD mortality (HR: 1.32, 95% CI: 1.02-1.72, P = 0.036) in OA patients. Subgroup analyses showed that hyperuricemia was related to the risk of all-cause mortality in OA patients aged >65 years (HR: 1.17, 95% CI: 1.01-1.36, P = 0.042), in all male patients (HR: 1.41, 95% CI: 1.10-1.80, P = 0.006), those diagnosed with hypertension (HR: 1.17, 95% CI: 1.01-1.37, P = 0.049), dyslipidemia (HR: 1.18, 95% CI: 1.01-1.39, P = 0.041), CVD (HR: 1.30, 95% CI: 1.09-1.55, P = 0.004), and CKD (HR: 1.31, 95% CI: 1.01-1.70, P = 0.046). The association between hyperuricemia and a higher risk of CVD mortality was found in OA patients aged ≤ 65 years (HR: 1.90, 95% CI: 1.06-3.41, P = 0.032), who did not suffer from diabetes (HR: 1.36, 95% CI: 1.01-1.86, P = 0.048), who did not suffer from hypertension (HR: 2.56, 95% CI: 1.12-5.86, P = 0.026), and who did not suffer from dyslipidemia (HR: 2.39, 95% CI: 1.15-4.97, P = 0.020). CONCLUSION These findings emphasize the importance of monitoring serum uric acid levels in OA patients for potentially reducing mortality associated with the disease.
Collapse
Affiliation(s)
- Ye Hao
- Articular Surgery, Beijing Shijingshan Hospital, Beijing, P.R. China
| | - Xin Tang
- Articular Surgery, Beijing Shijingshan Hospital, Beijing, P.R. China
| | - Feng Xu
- Articular Surgery, Beijing Shijingshan Hospital, Beijing, P.R. China
| |
Collapse
|
29
|
Abu-Awwad A, Tudoran C, Abu-Awwad SA, Tudoran M, Voita-Mekeres F, Faur C, Szilagyi G. Analogies Between Platelet-Rich Plasma Versus Hyaluronic Acid Intra-articular Injections in the Treatment of Advanced Knee Arthritis: A Single-Center Study. Cureus 2024; 16:e61163. [PMID: 38933627 PMCID: PMC11202161 DOI: 10.7759/cureus.61163] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/26/2024] [Indexed: 06/28/2024] Open
Abstract
Background Knee osteoarthritis (KOA), a degenerative joint disease, is a common cause of chronic knee pain and disability in adults. Conservative management options are the first-line approach, but intra-articular injections, such as platelet-rich plasma (PRP) and hyaluronic acid (HA), are considered for advanced cases. This study aims to compare the efficacy of PRP versus HA injections in patients with advanced KOA. Methods A retrospective study was conducted on 145 patients with advanced KOA. Seventy patients received PRP injections, while 75 patients received HA injections. The Visual Analog Scale (VAS), Western Ontario and McMaster Universities Osteoarthritis Index (WOMAC) score, and International Knee Documentation Committee (IKDC) score were employed to evaluate the treatment's efficacy. Adverse events associated with these injections were also recorded. Results Both PRP and HA injections significantly reduced pain and improved joint function in patients with advanced KOA. PRP injections were slightly more effective than HA injections in reducing pain scores. Both treatments showed similar improvements in functional outcomes. Adverse events were minimal and self-limiting for both treatments. Conclusions Both PRP and HA injections effectively ameliorate advanced KOA by reducing pain and improving function. PRP injections showed a slightly greater improvement in pain scores and functional outcomes. The choice between PRP and HA injections may depend on factors like cost, availability, and patient preference. Further research is needed to validate these findings and understand treatment suitability for different patient populations.
Collapse
Affiliation(s)
- Ahmed Abu-Awwad
- Orthopedics and Traumatology, Department XV-Discipline of Orthopedics-Traumatology, Victor Babes University of Medicine and Pharmacy Timisoara, Timisoara, ROU
- Orthopedics and Traumatology, "Pius Brinzeu" County Emergency Hospital, Timisoara, ROU
- Orthopedics and Traumatology, Research Center University Professor Doctor Teodor Șora, Victor Babes University of Medicine and Pharmacy, Timisoara, ROU
| | - Cristina Tudoran
- Department VII, Internal Medicine II, Discipline of Cardiology, Victor Babes University of Medicine and Pharmacy Timisoara, Timisoara, ROU
- Cardiology, Center of Molecular Research in Nephrology and Vascular Disease, Victor Babes University of Medicine and Pharmacy Timisoara, Timisoara, ROU
- Cardiology, "Pius Brinzeu" County Emergency Hospital, Timisoara, ROU
| | - Simona-Alina Abu-Awwad
- Gynecology, "Pius Brinzeu" County Emergency Hospital, Timisoara, ROU
- Gynecology, Department XII-Discipline of Obstetrics and Gynecology, Victor Babes University of Medicine and Pharmacy Timisoara, Timisoara, ROU
| | - Mariana Tudoran
- Cardiology, "Pius Brinzeu" County Emergency Hospital, Timisoara, ROU
| | - Florica Voita-Mekeres
- Morphological Disciplines, Faculty of Medicine and Pharmacy, University of Oradea, Oradea, ROU
| | - Cosmin Faur
- Orthopedics and Traumatology, Department XV-Discipline of Orthopedics-Traumatology, Victor Babes University of Medicine and Pharmacy Timisoara, Timisoara, ROU
- Orthopedics and Traumatology, "Pius Brinzeu" County Emergency Hospital, Timisoara, ROU
- Orthopedics and Traumatology, Research Center University Professor Doctor Teodor Șora, Victor Babes University of Medicine and Pharmacy, Timisoara, ROU
| | - Gheorghe Szilagyi
- Surgical Disciplines, Faculty of Medicine and Pharmacy, University of Oradea, Oradea, ROU
| |
Collapse
|
30
|
Li B, Yang Z, Li Y, Zhang J, Li C, Lv N. Exploration beyond osteoarthritis: the association and mechanism of its related comorbidities. Front Endocrinol (Lausanne) 2024; 15:1352671. [PMID: 38779455 PMCID: PMC11110169 DOI: 10.3389/fendo.2024.1352671] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/08/2023] [Accepted: 03/12/2024] [Indexed: 05/25/2024] Open
Abstract
Osteoarthritis is the most prevalent age-related degenerative joint disease and a leading cause of pain and disability in aged people. Its etiology is multifaceted, involving factors such as biomechanics, pro-inflammatory mediators, genetics, and metabolism. Beyond its evident impact on joint functionality and the erosion of patients' quality of life, OA exhibits symbiotic relationships with various systemic diseases, giving rise to various complications. This review reveals OA's extensive impact, encompassing osteoporosis, sarcopenia, cardiovascular diseases, diabetes mellitus, neurological disorders, mental health, and even cancer. Shared inflammatory processes, genetic factors, and lifestyle elements link OA to these systemic conditions. Consequently, recognizing these connections and addressing them offers opportunities to enhance patient care and reduce the burden of associated diseases, emphasizing the need for a holistic approach to managing OA and its complications.
Collapse
Affiliation(s)
| | | | | | | | | | - Naishan Lv
- The Second Affiliated Hospital of Shandong University of Traditional Chinese Medicine (Shandong Hospital of integrated traditional Chinese and Western medicine), Jinan, China
| |
Collapse
|
31
|
Korneva YS, Borisenko MV, Deev RV. Gut microbiota – a new link in the pathogenesis of osteoarthritis (literature review). THE SIBERIAN JOURNAL OF CLINICAL AND EXPERIMENTAL MEDICINE 2024; 39:38-43. [DOI: 10.29001/2073-8552-2024-39-1-38-43] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
Abstract
The intestinal microbiota (IMB) can indirectly affect the course of ostearthritis (OA) at the systemic level by stimulating a chronic nonspecific inflammatory reaction in the synovial membrane and subchondral bone, the cause of which is an increase in the amount of circulating lipopolysaccharides (LPS) of the bacterial wall, as well as provoke the development of metabolic syndrome, which links the two necessary components of the pathogenesis of OA. The result of direct exposure is the formation of leaky gut syndrome with the activation of LPS of the bacterial wall of mild inflammation, provoking the production of proinflammatory cytokines, the effect of which on synoviocytes and chondrocytes leads to their activation with subsequent production of IL-6 and IL-8, which contributes to the persistence of inflammation. By correlation analysis, the relationship of three taxa with OA joint damage was proved, namely, the order Desulfovibrionales and the genus Ruminiclostridium 5 – with knee joint OA, Methanobacteriaceae – with knee joint OA, and OA of any localization, and the appearance and degree of contamination with the genus Streptococcus correlated with the severity of pain syndrome. The metabolic syndrome itself can provoke the development of dysbiosis, so it can also be its consequence. A change in the composition of the microbiota in the form of the predominance of the genus Clostridium and the species Staphylococcus aureus with a decrease in the diversity of microorganisms is associated with an increase in the amount of adipose tissue in the body, dyslipidemia, insulin resistance with impaired carbohydrate metabolism. Low levels of LPS in the blood are found in obese patients even in the absence of an obvious focus of infection due to violations in the CMB, they signal TLR-4, triggering systemic inflammation. Studies have shown a positive effect of prescribing proand prebiotics on the course of OA, which makes IMB a promising target for the treatment and prevention of OA.
Collapse
Affiliation(s)
- Yu. S. Korneva
- Russian Scientific Research Institute of Traumatology and Orthopedics named after R.R. Vreden; North-Western State Medical University named after I.I. Mechnikov; Smolensk State Medical University of the Ministry of Health of the Russian Federation
| | - M. V. Borisenko
- North-Western State Medical University named after I.I. Mechnikov
| | - R. V. Deev
- Avtsyn Research Institute of Human Morphology of Federal state budgetary scientific institution “Petrovsky National Research Centre of Surgery”
| |
Collapse
|
32
|
Mukherjee A, Das B. The role of inflammatory mediators and matrix metalloproteinases (MMPs) in the progression of osteoarthritis. BIOMATERIALS AND BIOSYSTEMS 2024; 13:100090. [PMID: 38440290 PMCID: PMC10910010 DOI: 10.1016/j.bbiosy.2024.100090] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Revised: 12/04/2023] [Accepted: 02/20/2024] [Indexed: 03/06/2024] Open
Abstract
Osteoarthritis (OA) is a chronic musculoskeletal disorder characterized by an imbalance between (synthesis) and catabolism (degradation) in altered homeostasis of articular cartilage mediated primarily by the innate immune system. OA degenerates the joints resulting in synovial hyperplasia, degradation of articular cartilage with damage of the structural and functional integrity of the cartilage extracellular matrix, subchondral sclerosis, osteophyte formation, and is characterized by chronic pain, stiffness, and loss of function. Inflammation triggered by factors like biomechanical stress is involved in the development of osteoarthritis. In OA apart from catabolic effects, anti-inflammatory anabolic processes also occur continually. There is also an underlying chronic inflammation present, not only in cartilage tissue but also within the synovium, which perpetuates tissue destruction of the OA joint. The consideration of inflammation in OA considers synovitis and/or other cellular and molecular events in the synovium during the progression of OA. In this review, we have presented the progression of joint degradation that results in OA. The critical role of inflammation in the pathogenesis of OA is discussed in detail along with the dysregulation within the cytokine networks composed of inflammatory and anti-inflammatory cytokines that drive catabolic pathways, inhibit matrix synthesis, and promote cellular apoptosis. OA pathogenesis, fluctuation of synovitis, and its clinical impact on disease progression are presented here along with the role of synovial macrophages in promoting inflammatory and destructive responses in OA. The role of interplay between different cytokines, structure, and function of their receptors in the inter-cellular signaling pathway is further explored. The effect of cytokines in the increased synthesis and release of matrix-decomposing proteolytic enzymes, such as matrix metalloproteinase (MMPs) and a disintegrin-like and metalloproteinase with thrombospondin motif (ADAMTS), is elaborated emphasizing the potential impact of MMPs on the chondrocytes, synovial cells, articular and periarticular tissues, and other immune system cells migrating to the site of inflammation. We also shed light on the pathogenesis of OA via oxidative damage particularly due to nitric oxide (NO) via its angiogenic response to inflammation. We concluded by presenting the current knowledge about the tissue inhibitors of metalloproteinases (TIMPs). Synthetic MMP inhibitors include zinc binding group (ZBG), non-ZBG, and mechanism-based inhibitors, all of which have the potential to be therapeutically beneficial in the treatment of osteoarthritis. Improving our understanding of the signaling pathways and molecular mechanisms that regulate the MMP gene expression, may open up new avenues for the creation of therapies that can stop the joint damage associated with OA.
Collapse
Affiliation(s)
- Anwesha Mukherjee
- Department of Biomedical Engineering, Indian Institute of Technology Ropar, India
| | - Bodhisatwa Das
- Department of Biomedical Engineering, Indian Institute of Technology Ropar, India
| |
Collapse
|
33
|
Prinz E, Schlupp L, Dyson G, Barrett M, Szymczak A, Velasco C, Izda V, Dunn CM, Jeffries MA. OA susceptibility in mice is partially mediated by the gut microbiome, is transferrable via microbiome transplantation and is associated with immunophenotype changes. Ann Rheum Dis 2024; 83:382-393. [PMID: 37979958 PMCID: PMC10922159 DOI: 10.1136/ard-2023-224907] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Accepted: 11/03/2023] [Indexed: 11/20/2023]
Abstract
OBJECTIVES The Murphy Roths Large (MRL)/MpJ 'superhealer' mouse strain is protected from post-traumatic osteoarthritis (OA), although no studies have evaluated the microbiome in the context of this protection. This study characterised microbiome differences between MRL and wild-type mice, evaluated microbiome transplantation and OA and investigated microbiome-associated immunophenotypes. METHODS Cecal material from mixed sex C57BL6/J (B6) or female MRL/MpJ (MRL) was transplanted into B6 and MRL mice, then OA was induced by disruption of the medial meniscus surgery (DMM). In other experiments, transplantation was performed after DMM and transplantation was performed into germ-free mice. Transplanted mice were bred through F2. OARSI, synovitis and osteophyte scores were determined blindly 8 weeks after DMM. 16S microbiome sequencing was performed and metagenomic function was imputed. Immunophenotypes were determined using mass cytometry. RESULTS MRL-into-B6 transplant prior to DMM showed reduced OA histopathology (OARSI score 70% lower transplant vs B6 control), synovitis (60% reduction) and osteophyte scores (30% reduction) 8 weeks after DMM. When performed 48 hours after DMM, MRL-into-B6 transplant improved OA outcomes but not when performed 1-2 weeks after DMM. Protection was seen in F1 (60% reduction) and F2 progeny (30% reduction). Several cecal microbiome clades were correlated with either better (eg, Lactobacillus, R=-0.32, p=0.02) or worse (eg, Rikenellaceae, R=0.43, p=0.001) OA outcomes. Baseline immunophenotypes associated with MRL-into-B6 transplants and MRL included reduced double-negative T cells and increased CD25+CD4+ T cells. CONCLUSION The gut microbiome is responsible in part for OA protection in MRL mice and is transferrable by microbiome transplantation. Transplantation induces resting systemic immunophenotyping changes that correlate with OA protection.
Collapse
Affiliation(s)
- Emmaline Prinz
- Arthritis & Clinical Immunology Program, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma, USA
- Division of Rheumatology, Immunology, and Allergy, The University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, USA
| | - Leoni Schlupp
- Arthritis & Clinical Immunology Program, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma, USA
| | - Gabby Dyson
- Arthritis & Clinical Immunology Program, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma, USA
| | - Montana Barrett
- Arthritis & Clinical Immunology Program, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma, USA
| | - Aleksander Szymczak
- Arthritis & Clinical Immunology Program, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma, USA
| | - Cassandra Velasco
- Division of Rheumatology, Immunology, and Allergy, The University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, USA
| | - Vladislav Izda
- Arthritis & Clinical Immunology Program, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma, USA
| | - Christopher M Dunn
- Arthritis & Clinical Immunology Program, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma, USA
- Division of Rheumatology, Immunology, and Allergy, The University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, USA
| | - Matlock A Jeffries
- Arthritis & Clinical Immunology Program, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma, USA
- Division of Rheumatology, Immunology, and Allergy, The University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, USA
| |
Collapse
|
34
|
Magruder ML, Jacofsky D, Springer B, Scuderi GR, Hameed D, Mont MA. Semaglutide and Other GLP-1 Agonists: A Boon for the Arthroplasty Industry? J Arthroplasty 2024; 39:277-282. [PMID: 38182322 DOI: 10.1016/j.arth.2023.12.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Accepted: 12/08/2023] [Indexed: 01/07/2024] Open
Affiliation(s)
- Matthew L Magruder
- Department of Orthopaedic Surgery, Maimonides Medical Center, Brooklyn, New York
| | | | - Bryan Springer
- OrthoCarolina - Hip & Knee Center and Atrium Musculoskeletal Institute, Charlotte, North Carolina
| | - Giles R Scuderi
- Department of Orthopaedic Surgery, Lenox Hill Hospital, Northwell Orthopaedic Institute, New York, New York
| | - Daniel Hameed
- Rubin Institute for Advanced Orthopedics, Sinai Hospital of Baltimore, Baltimore, Maryland
| | - Michael A Mont
- Rubin Institute for Advanced Orthopedics, Sinai Hospital of Baltimore, Baltimore, Maryland
| |
Collapse
|
35
|
Schlupp L, Prinz E, Dyson G, Barrett M, Izda V, Dunn CM, Jeffries MA. Sex-Linked Discrepancies in C57BL6/J Mouse Osteoarthritis are Associated With the Gut Microbiome and are Transferrable by Microbiome Transplantation. Arthritis Rheumatol 2024; 76:231-237. [PMID: 37651283 PMCID: PMC10842085 DOI: 10.1002/art.42687] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Revised: 08/08/2023] [Accepted: 08/21/2023] [Indexed: 09/02/2023]
Abstract
OBJECTIVE Females have reduced osteoarthritis (OA) in surgical models. The objective of the current study was to evaluate a sex-linked gut microbiome in the pathogenesis of OA. METHODS We induced OA via destabilization of the medial meniscus surgery in adult male and female C57BL6/J mice with and without opposite-sex microbiome transplantation. Eight weeks later, animals were euthanized, and OA severity, synovitis, and osteophyte scores were determined. Serum lipopolysaccharide was measured chromogenically, and serum cytokines were quantified via multiplex immunoassay. Cecal microbiome profiles were generated using 16S deep sequencing. RESULTS Males had worse OA histology (3.5x, P = 6 × 10-7 ), synovitis (2.4x, P = 5 × 10-4 ), and osteophyte scores (3.7x, P = 3 × 10-4 ) than females. Male-into-female transplantation worsened all outcomes (histology 1.8x, P = 0.02; synovitis 2.0x, P = 3 × 10-5 ; osteophyte 2.1x, P = 0.01) compared to females, whereas female-into-male transplantation improved all outcomes except for synovitis (histology 0.53x, P = 2 × 10-4 ; osteophyte 0.28x, P = 5 × 10-4 ) compared to males. In the gut microbiome analysis, 44 clades were different in at least one group comparison; 5 clades were correlated with the Osteoarthritis Research Society International score (Lactobacillus R = -0.40, Aldercreutzia R = -0.40, rc4_4 R = -0.55, Sutterella R = -0.37, and Clostridiales R = 0.36). In the cytokine analysis, 10 analytes were different in at least one group comparison; 3 were different in two groups (female and female-into-male transplants vs male comparisons, all reduced in female and female-into-male transplants), including interleukin-12 (0.66x, P = 0.02; 0.66x, P = 0.02, respectively), eotaxin (0.74x, P = 5 × 10-6 ; 0.57x, P = 0.03), and tumor necrosis factor ⍺ (0.49x, P = 0.03; 0.52x, P = 0.009). CONCLUSION Sex-linked differences in the mouse gut microbiome are associated with OA outcomes, are reversible by opposite-sex microbiome transplantation, and are associated with serum cytokine changes.
Collapse
Affiliation(s)
- Leoni Schlupp
- Oklahoma Medical Research Foundation, Arthritis & Clinical Immunology Program, Oklahoma City, OK
| | - Emmaline Prinz
- Oklahoma Medical Research Foundation, Arthritis & Clinical Immunology Program, Oklahoma City, OK
- University of Oklahoma Health Sciences Center, Department of Internal Medicine, Division of Rheumatology, Immunology, and Allergy, Oklahoma City, OK
| | - Gabriella Dyson
- Oklahoma Medical Research Foundation, Arthritis & Clinical Immunology Program, Oklahoma City, OK
| | - Montana Barrett
- Oklahoma Medical Research Foundation, Arthritis & Clinical Immunology Program, Oklahoma City, OK
| | - Vladislav Izda
- Oklahoma Medical Research Foundation, Arthritis & Clinical Immunology Program, Oklahoma City, OK
| | - Christopher M. Dunn
- Oklahoma Medical Research Foundation, Arthritis & Clinical Immunology Program, Oklahoma City, OK
- University of Oklahoma Health Sciences Center, Department of Internal Medicine, Division of Rheumatology, Immunology, and Allergy, Oklahoma City, OK
| | - Matlock A. Jeffries
- Oklahoma Medical Research Foundation, Arthritis & Clinical Immunology Program, Oklahoma City, OK
- University of Oklahoma Health Sciences Center, Department of Internal Medicine, Division of Rheumatology, Immunology, and Allergy, Oklahoma City, OK
- Oklahoma City VA Medical Center, Oklahoma City, OK
| |
Collapse
|
36
|
Basak S, Hridayanka KSN, Duttaroy AK. Bioactives and their roles in bone metabolism of osteoarthritis: evidence and mechanisms on gut-bone axis. Front Immunol 2024; 14:1323233. [PMID: 38235147 PMCID: PMC10792057 DOI: 10.3389/fimmu.2023.1323233] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Accepted: 12/11/2023] [Indexed: 01/19/2024] Open
Abstract
Bioactives significantly modify and maintain human health. Available data suggest that Bioactives might play a beneficial role in chronic inflammatory diseases. Although promised, defining their mechanisms and opting to weigh their benefits and limitations is imperative. Detailed mechanisms by which critical Bioactives, including probiotics and prebiotics such as dietary lipids (DHA, EPA, alpha LA), vitamin D, polysaccharides (fructooligosaccharide), polyphenols (curcumin, resveratrol, and capsaicin) potentially modulate inflammation and bone metabolism is limited. Certain dietary bioactive significantly impact the gut microbiota, immune system, and pain response via the gut-immune-bone axis. This narrative review highlights a recent update on mechanistic evidence that bioactive is demonstrated demonstrated to reduce osteoarthritis pathophysiology.
Collapse
Affiliation(s)
- Sanjay Basak
- Molecular Biology Division, National Institute of Nutrition, Indian Council of Medical Research, Hyderabad, India
| | - Kota Sri Naga Hridayanka
- Molecular Biology Division, National Institute of Nutrition, Indian Council of Medical Research, Hyderabad, India
| | - Asim K. Duttaroy
- Department of Nutrition, Institute of Basic Medical Sciences, Faculty of Medicine, University of Oslo, Oslo, Norway
| |
Collapse
|
37
|
Zhou S, Zhao G, Chen R, Li Y, Huang J, Kuang L, Zhang D, Li Z, Xu H, Xiang W, Xie Y, Chen L, Ni Z. Lymphatic vessels: roles and potential therapeutic intervention in rheumatoid arthritis and osteoarthritis. Theranostics 2024; 14:265-282. [PMID: 38164153 PMCID: PMC10750203 DOI: 10.7150/thno.90940] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Accepted: 11/07/2023] [Indexed: 01/03/2024] Open
Abstract
Lymphatic vessel networks are a main part of the vertebrate cardiovascular system, which participate in various physiological and pathological processes via regulation of fluid transport and immunosurveillance. Targeting lymphatic vessels has become a potent strategy for treating various human diseases. The presence of varying degrees of inflammation in joints of rheumatoid arthritis (RA) and osteoarthritis (OA), characterized by heightened infiltration of inflammatory cells, increased levels of inflammatory factors, and activation of inflammatory signaling pathways, significantly contributes to the disruption of cartilage and bone homeostasis in arthritic conditions. Increasing evidence has demonstrated the pivotal role of lymphatic vessels in maintaining joint homeostasis, with their pathological alterations closely associated with the initiation and progression of inflammatory joint diseases. In this review, we provide a comprehensive overview of the evolving knowledge regarding the structural and functional aspects of lymphatic vessels in the pathogenesis of RA and OA. In addition, we summarized the potential regulatory mechanisms underlying the modulation of lymphatic function in maintaining joint homeostasis during inflammatory conditions, and further discuss the distinctions between RA and OA. Moreover, we describe therapeutic strategies for inflammatory arthritis based on lymphatic vessels, including the promotion of lymphangiogenesis, restoration of proper lymphatic vessel function through anti-inflammatory approaches, enhancement of lymphatic contractility and drainage, and alleviation of congestion within the lymphatic system through the elimination of inflammatory cells. At last, we envisage potential research perspectives and strategies to target lymphatic vessels in treating these inflammatory joint diseases.
Collapse
Affiliation(s)
- Siru Zhou
- War Trauma Medical Center, State Key Laboratory of Trauma and Chemical Poisoning, Army Medical Center, Daping Hospital, Army Medical University, Chongqing, 40038, People's Republic of China
- Department of Wound Repair and Rehabilitation Medicine, State Key Laboratory of Trauma and Chemical Poisoning, Army Medical Center, Daping Hospital, Army Medical University, Chongqing, 40038, People's Republic of China
| | - Guangyu Zhao
- Department of Emergency Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, People's Republic of China
- Rehabilitation Medicine Department, Army Medical Center, Daping Hospital, Army Medical University, Chongqing 400038, People's Republic of China
| | - Ran Chen
- War Trauma Medical Center, State Key Laboratory of Trauma and Chemical Poisoning, Army Medical Center, Daping Hospital, Army Medical University, Chongqing, 40038, People's Republic of China
- Department of Wound Repair and Rehabilitation Medicine, State Key Laboratory of Trauma and Chemical Poisoning, Army Medical Center, Daping Hospital, Army Medical University, Chongqing, 40038, People's Republic of China
| | - Yang Li
- War Trauma Medical Center, State Key Laboratory of Trauma and Chemical Poisoning, Army Medical Center, Daping Hospital, Army Medical University, Chongqing, 40038, People's Republic of China
| | - Junlan Huang
- Department of Wound Repair and Rehabilitation Medicine, State Key Laboratory of Trauma and Chemical Poisoning, Army Medical Center, Daping Hospital, Army Medical University, Chongqing, 40038, People's Republic of China
| | - Liang Kuang
- Department of Wound Repair and Rehabilitation Medicine, State Key Laboratory of Trauma and Chemical Poisoning, Army Medical Center, Daping Hospital, Army Medical University, Chongqing, 40038, People's Republic of China
| | - Dali Zhang
- Department of Wound Repair and Rehabilitation Medicine, State Key Laboratory of Trauma and Chemical Poisoning, Army Medical Center, Daping Hospital, Army Medical University, Chongqing, 40038, People's Republic of China
- The Department of Cardiology, General Hospital of Northern Theater Command, Shenyang 110015, People's Republic of China
| | - Zhijun Li
- Rehabilitation Medicine Department, Army Medical Center, Daping Hospital, Army Medical University, Chongqing 400038, People's Republic of China
| | - Haofeng Xu
- Rehabilitation Medicine Department, Army Medical Center, Daping Hospital, Army Medical University, Chongqing 400038, People's Republic of China
| | - Wei Xiang
- Rehabilitation Medicine Department, Army Medical Center, Daping Hospital, Army Medical University, Chongqing 400038, People's Republic of China
| | - Yangli Xie
- Department of Wound Repair and Rehabilitation Medicine, State Key Laboratory of Trauma and Chemical Poisoning, Army Medical Center, Daping Hospital, Army Medical University, Chongqing, 40038, People's Republic of China
| | - Lin Chen
- Department of Wound Repair and Rehabilitation Medicine, State Key Laboratory of Trauma and Chemical Poisoning, Army Medical Center, Daping Hospital, Army Medical University, Chongqing, 40038, People's Republic of China
| | - Zhenhong Ni
- Rehabilitation Medicine Department, Army Medical Center, Daping Hospital, Army Medical University, Chongqing 400038, People's Republic of China
| |
Collapse
|
38
|
Hildebrandt A, Dietrich T, Weber J, Günderoth MM, Zhou S, Fleckenstein FN, Jiang S, Winkler T, Duda GN, Tsitsilonis S, Keller J, Maleitzke T. The dual pro-inflammatory and bone-protective role of calcitonin gene-related peptide alpha in age-related osteoarthritis. Arthritis Res Ther 2023; 25:244. [PMID: 38102666 PMCID: PMC10722726 DOI: 10.1186/s13075-023-03215-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Accepted: 11/15/2023] [Indexed: 12/17/2023] Open
Abstract
BACKGROUND The vasoactive neuropeptide calcitonin gene-related peptide alpha (αCGRP) enhances nociception in primary knee osteoarthritis (OA) and has been shown to disrupt cartilage and joint integrity in experimental rheumatoid arthritis (RA). Little is known about how αCGRP may alter articular structures in primary OA. We investigated whether αCGRP modulates local inflammation and concomitant cartilage and bone changes in a murine model of age-dependent OA. METHODS Sixteen- to 18-month-old αCGRP-deficient mice (αCGRP-/-aged) were compared to, first, age-matched wild type (WTaged) and, second, young 4- to 5-month-old non-OA αCGRP-deficient (αCGRP-/-CTRL) and non-OA WT animals (WTCTRL). αCGRP levels were measured in serum. Knee and hip joint inflammation, cartilage degradation, and bone alterations were assessed by histology (OARSI histopathological grading score), gene expression analysis, and µ-computed tomography. RESULTS WTaged mice exhibited elevated αCGRP serum levels compared to young WTCTRL animals. Marked signs of OA-induced cartilage destruction were seen in WTaged animals, while αCGRP-/-aged mice were mostly protected from this effect. Age-dependent OA was accompanied by an increased gene expression of pro-inflammatory Tnfa, Il1b, and Il6 and catabolic Mmp13, Adamts5, Ctsk, Tnfs11 (Rankl), and Cxcl12/Cxcr4 in WTaged but not in αCGRP-/-aged mice. αCGRP-deficiency however further aggravated subchondral bone sclerosis of the medial tibial plateau and accelerated bone loss in the epi- and metaphyseal trabecular tibial bone in age-dependent OA. CONCLUSIONS Similar to its function in experimental RA, αCGRP exerts a dual pro-inflammatory and bone-protective function in murine primary OA. Although anti-CGRP treatment was previously not successful in reducing pain in OA clinically, these data underline a crucial pathophysiological role of αCGRP in age-related OA.
Collapse
Affiliation(s)
- Alexander Hildebrandt
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität Zu Berlin, Center for Musculoskeletal Surgery, Berlin, Germany
- Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Julius Wolff Institute, Berlin, Germany
| | - Tamara Dietrich
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität Zu Berlin, Center for Musculoskeletal Surgery, Berlin, Germany
- Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Julius Wolff Institute, Berlin, Germany
| | - Jérôme Weber
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität Zu Berlin, Center for Musculoskeletal Surgery, Berlin, Germany
- Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Julius Wolff Institute, Berlin, Germany
| | - Mara Meyer Günderoth
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität Zu Berlin, Center for Musculoskeletal Surgery, Berlin, Germany
- Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Julius Wolff Institute, Berlin, Germany
| | - Sijia Zhou
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität Zu Berlin, Center for Musculoskeletal Surgery, Berlin, Germany
- Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Julius Wolff Institute, Berlin, Germany
| | - Florian N Fleckenstein
- Department of Diagnostic and Interventional Radiology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität Zu Berlin, Berlin, Germany
- Berlin Institute of Health at Charité - Universitätsmedizin Berlin, BIH Biomedical Innovation Academy, BIH Charité Clinician Scientist Program, Berlin, Germany
| | - Shan Jiang
- Department of Trauma and Orthopedic Surgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Tobias Winkler
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität Zu Berlin, Center for Musculoskeletal Surgery, Berlin, Germany
- Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Julius Wolff Institute, Berlin, Germany
- Berlin Institute of Health Center for Regenerative Therapies, Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Georg N Duda
- Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Julius Wolff Institute, Berlin, Germany
| | - Serafeim Tsitsilonis
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität Zu Berlin, Center for Musculoskeletal Surgery, Berlin, Germany
- Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Julius Wolff Institute, Berlin, Germany
| | - Johannes Keller
- Department of Trauma and Orthopedic Surgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Tazio Maleitzke
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität Zu Berlin, Center for Musculoskeletal Surgery, Berlin, Germany.
- Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Julius Wolff Institute, Berlin, Germany.
- Berlin Institute of Health at Charité - Universitätsmedizin Berlin, BIH Biomedical Innovation Academy, BIH Charité Clinician Scientist Program, Berlin, Germany.
- Department of Orthopaedic Surgery, Copenhagen University Hospital - Amager and Hvidovre, Hvidovre, Denmark.
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark.
| |
Collapse
|
39
|
Qiao S, Zhao R, He S, Fu X, An J, Xia T. Quercitrin attenuates the progression of osteoarthritis via inhibiting NF-κB signaling pathways and enhance glucose transport capacity. Exp Cell Res 2023; 433:113854. [PMID: 37952573 DOI: 10.1016/j.yexcr.2023.113854] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Revised: 11/08/2023] [Accepted: 11/08/2023] [Indexed: 11/14/2023]
Abstract
Osteoarthritis (OA) is a common musculoskeletal disorder that impairs function and reduces the quality of life. Extracellular matrix (ECM) degradation and inflammatory mechanisms are crucial to the progression of OA. In this study, we aimed to investigate the anti-inflammatory activity, anti-ECM degradation property, and glucose transport capacity of quercitrin (QCT) on IL-1β-treated rat primary chondrocytes. Rat primary chondrocytes were treated with IL-1β to simulate inflammatory environmental conditions and OA in vitro. We examined the effects of QCT at concentrations ranging from 0 to 200 μM on the viability of rat chondrocytes and selected 5 μM for further study. Using qRT-PCR, immunofluorescent, immunocytochemistry, and western blotting techniques, we identified the potential molecular mechanisms and signaling pathways that are responsible for these effects. We established an OA rat model through anterior cruciate ligament transection (ACLT). The animals were then periodically injected with QCT into the knee articular cavity. Our in vivo and in vitro study showed that QCT could inhibit IL-1β-activated inflammation and ECM degradation in chondrocyte. Furthermore, QCT could inhibit the NF-κB signal pathway and enhance glucose transport capacity in the IL-1β-stimulated chondrocytes. In vivo study proved that QCT attenuates OA progression in rats. Overall, QCT inhibited the activation of NF-κB and enhanced glucose transport capacity to alleviate the progression of OA.
Collapse
Affiliation(s)
- Shigang Qiao
- Institute of Clinical Medicine Research, Suzhou Hospital, Affiliated Hospital of Medical School, Nanjing University, Suzhou, China
| | - Runze Zhao
- Department of Orthopedic Surgery, The First Affiliated Hospital, Orthopedic Institure, Medical College, Soochow University, Suzhou, China
| | - Shuangjian He
- Department of orthopedics, Suzhou Hospital, Affiliated Hospital of Medical School, Nanjing University, Suzhou, China
| | - Xuejie Fu
- Institute of Clinical Medicine Research, Suzhou Hospital, Affiliated Hospital of Medical School, Nanjing University, Suzhou, China
| | - Jianzhong An
- Institute of Clinical Medicine Research, Suzhou Hospital, Affiliated Hospital of Medical School, Nanjing University, Suzhou, China
| | - Tingting Xia
- Institute of Clinical Medicine Research, Suzhou Hospital, Affiliated Hospital of Medical School, Nanjing University, Suzhou, China.
| |
Collapse
|
40
|
Wang X, Cai W, Liu Y, Lu Y, Liu M, Cao X, Guo D. Exploring biomarkers associated with severity of knee osteoarthritis in Southern China using widely targeted metabolomics. BMC Musculoskelet Disord 2023; 24:953. [PMID: 38066443 PMCID: PMC10704822 DOI: 10.1186/s12891-023-07084-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/12/2023] [Accepted: 11/30/2023] [Indexed: 12/18/2023] Open
Abstract
BACKGROUND Metabolomics is a tool to study the pathogenesis of diseases and their associated metabolites, but there are still insufficient metabolomic studies on severe knee osteoarthritis.To investigate the differences in serum metabolites between healthy populations and knee osteoarthritis (KOA) patients in Southern China using widely targeted metabolomics, and to explore biomarkers and their metabolic pathways that could be associated with the severity of KOA. METHODS There were 10 healthy individuals in the control group and 32 patients with KOA. According to the Kellgren-Lawrence (KL) grading system, KOA was further divided into mild (n = 13, KL grade 1 and 2) and severe (n = 19, KL grade 3 and 4). Serum samples from all participants were collected and analyzed metabolomics based on ultra-performance liquid chromatography/electrospray ionization/tandem mass spectrometry. We screened for differential metabolites between patients and controls, and between mild and severe KOA. We explored the metabolic pathways involved in differential metabolism using the Kyoto Encyclopedia of Genes and Genomes database. RESULTS Sixty-one metabolites were differentially expressed in the sera of the patient group compared with the control group (45 upregulated and 16 downregulated). Analysis of the mild and severe KOA groups showed a total of 12 differential metabolites. Receiver operating characteristic curve analysis showed N-alpha-acetyl-L-asparagine was a good predictor of advanced osteoarthritis(OA).Differential metabolites are enriched in multiple pathways such as arachidonic acid metabolism. CONCLUSION Widely targeted metabolomics found that upregulation of the amino acid metabolite N-α-acetyl-L-asparagine was significantly associated with severe KOA and could be a biomarker for predicting severity of KOA. Arachidonic acid metabolism may play an important role in patients with severe KOA.
Collapse
Affiliation(s)
- Xiaochao Wang
- Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Wanling Cai
- Shuguang Hospital Affiliated to Shanghai University of Chinese Traditional Medicine, Shanghai, China
| | - Yihan Liu
- Department of Neurological Rehabilitation, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yaoming Lu
- Department of Orthopaedic Surgery, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, Guangdong, China
| | - Mange Liu
- Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Xuewei Cao
- Department of Orthopaedic Surgery, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, Guangdong, China
| | - Da Guo
- Department of Orthopaedic Surgery, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, Guangdong, China.
| |
Collapse
|
41
|
Li S, Ma L, Cui R. Identification of Novel Diagnostic Biomarkers and Classification Patterns for Osteoarthritis by Analyzing a Specific Set of Genes Related to Inflammation. Inflammation 2023; 46:2193-2208. [PMID: 37462886 DOI: 10.1007/s10753-023-01871-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2023] [Revised: 06/14/2023] [Accepted: 07/03/2023] [Indexed: 11/25/2023]
Abstract
Osteoarthritis (OA) is a prevalent joint disease globally. TNFA is recognized as a crucial inflammatory cytokine that plays a significant role in the pathophysiological mechanisms that occur during the progression of OA. However, the TNFA_SIGNALING_VIA_NFKB (TSVN)-related genes (TRGs) during the progression of OA remain unclear. By conducting a combinatory analysis of OA transcriptome data from three datasets, various differentially expressed TRGs were identified. The logistic regression model was used to mine hub TRGs for OA, and a nomogram prediction model was subsequently constructed using these TRGs. To identify new molecular subgroups, we performed consensus clustering. We then conducted functional analyses, including GO, KEGG, GSVA, and GSEA, to elucidate the underlying mechanisms. To determine the immune microenvironment, we applied xCell. The logistic regression analysis identified three hub TRGs (BHLHE40, BTG2, and CCNL1) as potential biomarkers for OA. Based on these TRGs, we constructed an OA predictive model. This model has demonstrated promising results in enhancing the accuracy of OA diagnosis, as evident from the ROC analysis (AUC merged dataset = 0.937, AUC validating dataset = 0.924). We identified two molecular subtypes, C1 and C2, and found that the C1 subtype showed activation of immune- and inflammation-related pathways. The involvement of TSVN in the development and progression of OA has been established. We identified several hub genes, such as BHLHE40, BTG2, and CCNL1, that may have a significant association with the progression of OA. Furthermore, our logistic regression model based on these genes has shown promising results in accurately diagnosing OA patients.
Collapse
Affiliation(s)
- Songsheng Li
- Orthopaedics Department III (Joint), The Fifth Clinical Medical College of Henan University of Chinese Medicine, Zhengzhou, China.
| | - Lige Ma
- Orthopaedics Department III (Joint), The Fifth Clinical Medical College of Henan University of Chinese Medicine, Zhengzhou, China
| | - Ruikai Cui
- Orthopaedics Department III (Joint), The Fifth Clinical Medical College of Henan University of Chinese Medicine, Zhengzhou, China
| |
Collapse
|
42
|
Yan Z, He Z, Jiang H, Zhang Y, Xu Y, Zhang Y. TRPV4-mediated mitochondrial dysfunction induces pyroptosis and cartilage degradation in osteoarthritis via the Drp1-HK2 axis. Int Immunopharmacol 2023; 123:110651. [PMID: 37506502 DOI: 10.1016/j.intimp.2023.110651] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Revised: 07/11/2023] [Accepted: 07/11/2023] [Indexed: 07/30/2023]
Abstract
Osteoarthritis (OA) is an age-related chronic degenerative disease with complex pathophysiological mechanisms. Accumulating evidence indicates that nod-like receptor pyrin domain 3 (NLRP3) inflammasome-mediated pyroptosis of chondrocytes plays a crucial role in the OA progression. Transient Receptor Potential Vanilloid 4 (TRPV4), described as a calcium-permeable cation channel, isassociated with proinflammatory factors and pyroptosis. In this study, we studied the potential functions of TRPV4 in chondrocyte pyroptosis and cartilage degradation. We found that lipopolysaccharides(LPS)-induced mitochondrial reactive oxygen species (mtROS) accumulation aggravated chondrocyte pyroptosis and cartilage degeneration. TRPV4 induces dynamin-related protein 1 (Drp1) mitochondrial translocation through the Ca2+-calmodulin-dependent protein kinase II (CaMKII) signaling pathway, which subsequently caused the mitochondrial dysfunction (e.g., mPTP over opening; Δψm depolarization; ATP production decreased; mtROS accumulation), pyroptosis and extracellular matrix (ECM) degradation through hexokinase 2 (HK2) dissociation from mitochondrial membrane. Moreover, TRPV4 inhibition reversed Drp1-involved chondrocyte pyroptosis and cartilage degeneration in the anterior cruciate ligament transection (ACLT) mouse model. Our findings revealed the internal mechanisms underlying TRPV4 regulation in chondrocytes and its intrinsic therapeutic efficacy for OA.
Collapse
Affiliation(s)
- Zijian Yan
- Department of Orthopaedics Surgery, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Zili He
- Department of Orthopaedics Surgery, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Hongyi Jiang
- Department of Orthopaedics Surgery, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Yu Zhang
- Molecular Pharmacology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Yitie Xu
- Molecular Pharmacology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Yingze Zhang
- Department of Orthopaedics Surgery, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325035, China; Department of Orthopedic Surgery of Hebei Province, Third Hospital of Hebei Medical University, 139 Ziqiang Road, Shijiazhuang 050051, Hebei, China; NHC Key Laboratory of Intelligent Orthopeadic Equipment, The Third Hospital of Hebei Medical University, Shijiazhuang, Hebei, China.
| |
Collapse
|
43
|
Xu X, Xu L, Xia J, Wen C, Liang Y, Zhang Y. Harnessing knee joint resident mesenchymal stem cells in cartilage tissue engineering. Acta Biomater 2023; 168:372-387. [PMID: 37481194 DOI: 10.1016/j.actbio.2023.07.024] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Revised: 06/26/2023] [Accepted: 07/17/2023] [Indexed: 07/24/2023]
Abstract
Osteoarthritis (OA) is a widespread clinical disease characterized by cartilage degeneration in middle-aged and elderly people. Currently, there is no effective treatment for OA apart from total joint replacement in advanced stages. Mesenchymal stem cells (MSCs) are a type of adult stem cell with diverse differentiation capabilities and immunomodulatory potentials. MSCs are known to effectively regulate the cartilage microenvironment, promote cartilage regeneration, and alleviate OA symptoms. As a result, they are promising sources of cells for OA therapy. Recent studies have revealed the presence of resident MSCs in synovial fluid, synovial membrane, and articular cartilage, which can be collected as knee joint-derived MSCs (KJD-MSC). Several preclinical and clinical studies have demonstrated that KJD-MSCs have great potential for OA treatment, whether applied alone, in combination with biomaterials, or as exocrine MSCs. In this article, we will review the characteristics of MSCs in the joints, including their cytological characteristics, such as proliferation, cartilage differentiation, and immunomodulatory abilities, as well as the biological function of MSC exosomes. We will also discuss the use of tissue engineering in OA treatment and introduce the concept of a new generation of stem cell-based tissue engineering therapy, including the use of engineering, gene therapy, and gene editing techniques to create KJD-MSCs or KJD-MSC derivative exosomes with improved functionality and targeted delivery. These advances aim to maximize the efficiency of cartilage tissue engineering and provide new strategies to overcome the bottleneck of OA therapy. STATEMENT OF SIGNIFICANCE: This research will provide new insights into the medicinal benefit of Joint resident Mesenchymal Stem Cells (MSCs), specifically on its cartilage tissue engineering ability. Through this review, the community will further realize promoting joint resident mesenchymal stem cells, especially cartilage progenitor/MSC-like progenitor cells (CPSC), as a preventive measure against osteoarthritis and cartilage injury. People and medical institutions may also consider cartilage derived MSC as an alternative approach against cartilage degeneration. Moreover, the discussion presented in this study will convey valuable information for future research that will explore the medicinal benefits of cartilage derived MSC.
Collapse
Affiliation(s)
- Xiao Xu
- Department of Joint Surgery and Sports Medicine, Affiliated Hospital of Jining Medical University, Jining Medical University, Jining, Shandong 272029, China; Department of Orthopedics, the First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen 518035, China
| | - Limei Xu
- Department of Hematology, Affiliated Hospital of Jining Medical University, Jining Medical University, Jining, Shandong 272029, China
| | - Jiang Xia
- Department of Chemistry, the Chinese University of Hong Kong, Shatin, Hong Kong SAR, China
| | - Caining Wen
- Department of Joint Surgery and Sports Medicine, Affiliated Hospital of Jining Medical University, Jining Medical University, Jining, Shandong 272029, China
| | - Yujie Liang
- Department of Joint Surgery and Sports Medicine, Affiliated Hospital of Jining Medical University, Jining Medical University, Jining, Shandong 272029, China; Department of Chemistry, the Chinese University of Hong Kong, Shatin, Hong Kong SAR, China.
| | - Yuanmin Zhang
- Department of Joint Surgery and Sports Medicine, Affiliated Hospital of Jining Medical University, Jining Medical University, Jining, Shandong 272029, China.
| |
Collapse
|
44
|
Vun J, Iqbal N, Jones E, Ganguly P. Anti-Aging Potential of Platelet Rich Plasma (PRP): Evidence from Osteoarthritis (OA) and Applications in Senescence and Inflammaging. Bioengineering (Basel) 2023; 10:987. [PMID: 37627872 PMCID: PMC10451843 DOI: 10.3390/bioengineering10080987] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2023] [Revised: 07/21/2023] [Accepted: 08/15/2023] [Indexed: 08/27/2023] Open
Abstract
Aging and age-related changes impact the quality of life (QOL) in elderly with a decline in movement, cognitive abilities and increased vulnerability towards age-related diseases (ARDs). One of the key contributing factors is cellular senescence, which is triggered majorly by DNA damage response (DDR). Accumulated senescent cells (SCs) release senescence-associated secretory phenotype (SASP), which includes pro-inflammatory cytokines, matrix metalloproteinases (MMPs), lipids and chemokines that are detrimental to the surrounding tissues. Chronic low-grade inflammation in the elderly or inflammaging is also associated with cellular senescence and contributes to ARDs. The literature from the last decade has recorded the use of platelet rich plasma (PRP) to combat senescence and inflammation, alleviate pain as an analgesic, promote tissue regeneration and repair via angiogenesis-all of which are essential in anti-aging and tissue regeneration strategies. In the last few decades, platelet-rich plasma (PRP) has been used as an anti-aging treatment option for dermatological applications and with great interest in tissue regeneration for orthopaedic applications, especially in osteoarthritis (OA). In this exploration, we connect the intricate relationship between aging, ARDs, senescence and inflammation and delve into PRP's properties and potential benefits. We conduct a comparative review of the current literature on PRP treatment strategies, paying particular attention to the instances strongly linked to ARDs. Finally, upon careful consideration of this interconnected information in the context of aging, we suggest a prospective role for PRP in developing anti-aging therapeutic strategies.
Collapse
Affiliation(s)
- James Vun
- Leeds Institute of Rheumatic and Musculoskeletal Medicine, University of Leeds, Leeds LS9 7TF, UK; (J.V.); (E.J.)
- Leeds Orthopaedic & Trauma Sciences, Leeds General Infirmary, University of Leeds, Leeds LS97TF, UK
| | - Neelam Iqbal
- School of Chemical and Process Engineering, University of Leeds, Leeds LS2 9JT, UK;
| | - Elena Jones
- Leeds Institute of Rheumatic and Musculoskeletal Medicine, University of Leeds, Leeds LS9 7TF, UK; (J.V.); (E.J.)
| | - Payal Ganguly
- Leeds Institute of Rheumatic and Musculoskeletal Medicine, University of Leeds, Leeds LS9 7TF, UK; (J.V.); (E.J.)
| |
Collapse
|
45
|
Wang H, Zhang C, Zhu S, Gao C, Gao Q, Huang R, Liu S, Wei X, Zhang H, Wei Q, He C. Low-frequency whole-body vibration can enhance cartilage degradation with slight changes in subchondral bone in mice with knee osteoarthritis and does not have any morphologic effect on normal joints. PLoS One 2023; 18:e0270074. [PMID: 37590222 PMCID: PMC10434961 DOI: 10.1371/journal.pone.0270074] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Accepted: 07/20/2023] [Indexed: 08/19/2023] Open
Abstract
PURPOSES To evaluate the effects of low frequency whole-body vibration (WBV) on degeneration of articular cartilage and subchondral bone in mice with destabilization of the medial meniscus (DMM)induced osteoarthritis(OA) and mice with normal knee. METHODS Ten-week-old C57BL/6J male mice received DMM on right knees, while the left knees performed sham operation. There were six groups: DMM, SHAM DMM, DMM+WBV,SHAM DMM+WBV, DMM+ NON-WBV and SHAM DMM+NON-WBV. After four weeks, the knees were harvested from the DMM and SHAM DMM group. The remaining groups were treated with WBV (10 Hz) or NON-WBV. Four weeks later, the knees were harvested. Genes, containing Aggrecan(Acan) and CollagenⅡ(Col2a1), Matrix Metalloproteinases 3 and 13(MMP3,13), TNFα and IL6, were measured and staining was also performed. OA was graded with OARSI scores, and tibial plateaubone volume to tissue volume ratio(BV/TV), bone surface area to bone volume ratio (BS/BV), trabecular number(Tb.N) and trabecular thickness separation(TS) between groups were analyzed. RESULTS Increased OARSI scores and cartilage degradation were observed after WBV. BV/TV, Tb.N and TS were not significant between the groups. Significant reductions were observed in MMP3, MMP13, Col2a1, Acan, TNFα and IL6 in the DMM+WBV compared to SHAM DMM+WBV group. BV/TV, BS/BV, Tb.N, TS and OARSI scores were not significantly changed in the left knees. IL6 expression in the SHAM DMM+WBV group was significantly increased compared with the SHAM DMM+ NON-WBV group, while Col2a1, Acan and MMP13 expression decreased. CONCLUSION WBV accelerated cartilage degeneration and caused slight changes in subchondral bone in a DMM-induced OA model. WBV had no morphologic effect on normal joints.
Collapse
Affiliation(s)
- Haiming Wang
- Rehabilitation Medicine Department, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Center of Rehabilitation Engineering Technology Research, Henan Province, Zhengzhou, Henan, China
| | - Chi Zhang
- Rehabilitation Medicine Department, The Affiliated Hospital Of Southwest Medical University, Luzhou, Sichuan, China
- Department of Rehabilitation Medicine, Southwest Medical University, Luzhou, Sichuan, China
- Nuclear Medicine and Molecular Imaging Key Laboratory of Sichuan Province, Luzhou, Sichuan, China
| | - Siyi Zhu
- Rehabilitation Medical Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Rehabilitation Medicine Key Laboratory of Sichuan Province, Sichuan University, Chengdu, Sichuan, China
| | - Chengfei Gao
- Rehabilitation Medicine Department, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
- Department of Biomedical Engineering, The Hong Kong Polytechnic University, Hong Kong, China
| | - Qiang Gao
- Rehabilitation Medical Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Rehabilitation Medicine Key Laboratory of Sichuan Province, Sichuan University, Chengdu, Sichuan, China
| | - Ridong Huang
- Department of Respiratory and Critical Care Medicine, Targeted Tracer Research and Development Laboratory, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Sijia Liu
- Rehabilitation Medical Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Rehabilitation Medicine Key Laboratory of Sichuan Province, Sichuan University, Chengdu, Sichuan, China
| | - Xiangyang Wei
- Rehabilitation Medicine Department, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Center of Rehabilitation Engineering Technology Research, Henan Province, Zhengzhou, Henan, China
| | - Huakai Zhang
- Medical College of Zhengzhou University of Industrial technology, Zhengzhou, Henan, China
| | - Quan Wei
- Rehabilitation Medical Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Rehabilitation Medicine Key Laboratory of Sichuan Province, Sichuan University, Chengdu, Sichuan, China
| | - Chengqi He
- Rehabilitation Medical Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Rehabilitation Medicine Key Laboratory of Sichuan Province, Sichuan University, Chengdu, Sichuan, China
| |
Collapse
|
46
|
Sohn R, Jenei-Lanzl Z. Role of the Sympathetic Nervous System in Mild Chronic Inflammatory Diseases: Focus on Osteoarthritis. Neuroimmunomodulation 2023; 30:143-166. [PMID: 37429263 PMCID: PMC10428144 DOI: 10.1159/000531798] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Accepted: 06/28/2023] [Indexed: 07/12/2023] Open
Abstract
The sympathetic nervous system (SNS) is a major regulatory mediator connecting the brain and the immune system that influences accordingly inflammatory processes within the entire body. In the periphery, the SNS exerts its effects mainly via its neurotransmitters norepinephrine (NE) and epinephrine (E), which are released by peripheral nerve endings in lymphatic organs and other tissues. Depending on their concentration, NE and E bind to specific α- and β-adrenergic receptor subtypes and can cause both pro- and anti-inflammatory cellular responses. The co-transmitter neuropeptide Y, adenosine triphosphate, or its metabolite adenosine are also mediators of the SNS. Local pro-inflammatory processes due to injury or pathogens lead to an activation of the SNS, which in turn induces several immunoregulatory mechanisms with either pro- or anti-inflammatory effects depending on neurotransmitter concentration or pathological context. In chronic inflammatory diseases, the activity of the SNS is persistently elevated and can trigger detrimental pathological processes. Recently, the sympathetic contribution to mild chronic inflammatory diseases like osteoarthritis (OA) has attracted growing interest. OA is a whole-joint disease and is characterized by mild chronic inflammation in the joint. In this narrative article, we summarize the underlying mechanisms behind the sympathetic influence on inflammation during OA pathogenesis. In addition, OA comorbidities also accompanied by mild chronic inflammation, such as hypertension, obesity, diabetes, and depression, will be reviewed. Finally, the potential of SNS-based therapeutic options for the treatment of OA will be discussed.
Collapse
Affiliation(s)
- Rebecca Sohn
- Department of Orthopedics (Friedrichsheim), University Hospital Frankfurt, Goethe University, Frankfurt, Germany
| | - Zsuzsa Jenei-Lanzl
- Department of Orthopedics (Friedrichsheim), University Hospital Frankfurt, Goethe University, Frankfurt, Germany
| |
Collapse
|
47
|
Alexander A, Herz J, Calvier L. Reelin through the years: From brain development to inflammation. Cell Rep 2023; 42:112669. [PMID: 37339050 PMCID: PMC10592530 DOI: 10.1016/j.celrep.2023.112669] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Revised: 05/09/2023] [Accepted: 06/02/2023] [Indexed: 06/22/2023] Open
Abstract
Reelin was originally identified as a regulator of neuronal migration and synaptic function, but its non-neuronal functions have received far less attention. Reelin participates in organ development and physiological functions in various tissues, but it is also dysregulated in some diseases. In the cardiovascular system, Reelin is abundant in the blood, where it contributes to platelet adhesion and coagulation, as well as vascular adhesion and permeability of leukocytes. It is a pro-inflammatory and pro-thrombotic factor with important implications for autoinflammatory and autoimmune diseases such as multiple sclerosis, Alzheimer's disease, arthritis, atherosclerosis, or cancer. Mechanistically, Reelin is a large secreted glycoprotein that binds to several membrane receptors, including ApoER2, VLDLR, integrins, and ephrins. Reelin signaling depends on the cell type but mostly involves phosphorylation of NF-κB, PI3K, AKT, or JAK/STAT. This review focuses on non-neuronal functions and the therapeutic potential of Reelin, while highlighting secretion, signaling, and functional similarities between cell types.
Collapse
Affiliation(s)
- Anna Alexander
- Department of Molecular Genetics, University of Texas (UT) Southwestern Medical Center, Dallas, TX, USA; Center for Translational Neurodegeneration Research, UT Southwestern Medical Center, Dallas, TX, USA
| | - Joachim Herz
- Department of Molecular Genetics, University of Texas (UT) Southwestern Medical Center, Dallas, TX, USA; Center for Translational Neurodegeneration Research, UT Southwestern Medical Center, Dallas, TX, USA; Department of Neuroscience, UT Southwestern Medical Center, Dallas, TX, USA; Department of Neurology and Neurotherapeutics, UT Southwestern Medical Center, Dallas, TX, USA
| | - Laurent Calvier
- Department of Molecular Genetics, University of Texas (UT) Southwestern Medical Center, Dallas, TX, USA; Center for Translational Neurodegeneration Research, UT Southwestern Medical Center, Dallas, TX, USA.
| |
Collapse
|
48
|
Lendoiro-Cino N, Rodríguez-Coello A, Saborido A, F-Burguera E, Fernández-Rodríguez JA, Meijide-Faílde R, Blanco FJ, Vaamonde-García C. Study of hydrogen sulfide biosynthesis in synovial tissue from diabetes-associated osteoarthritis and its influence on macrophage phenotype and abundance. J Physiol Biochem 2023:10.1007/s13105-023-00968-y. [PMID: 37335394 DOI: 10.1007/s13105-023-00968-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Accepted: 05/31/2023] [Indexed: 06/21/2023]
Abstract
Type 2 diabetes (DB) is an independent risk factor for osteoarthritis (OA). However, the mechanisms underlying the connection between both diseases remain unclear. Synovial macrophages from OA patients with DB present a marked pro-inflammatory phenotype. Since hydrogen sulphide (H2S) has been previously described to be involved in macrophage polarization, in this study we examined H2S biosynthesis in synovial tissue from OA patients with DB, observing a reduction of H2S-synthetizing enzymes in this subset of individuals. To elucidate these findings, we detected that differentiated TPH-1 cells to macrophages exposed to high levels of glucose presented a lower expression of H2S-synthetizing enzymes and an increased inflammatory response to LPS, showing upregulated expression of markers associated with M1 phenotype (i.e., CD11c, CD86, iNOS, and IL-6) and reduced levels of those related to M2 fate (CD206 and CD163). The co-treatment of the cells with a slow-releasing H2S donor, GYY-4137, attenuated the expression of M1 markers, but failed to modulate the levels of M2 indicators. GYY-4137 also reduced HIF-1α expression and upregulated the protein levels of HO-1, suggesting their involvement in the anti-inflammatory effects of H2S induction. In addition, we observed that intraarticular administration of H2S donor attenuated synovial abundance of CD68+ cells, mainly macrophages, in an in vivo model of OA. Taken together, the findings of this study seem to reinforce the key role of H2S in the M1-like polarization of synovial macrophages associated to OA and specifically its metabolic phenotype, opening new therapeutic perspectives in the management of this pathology.
Collapse
Affiliation(s)
- Natalia Lendoiro-Cino
- Grupo de Investigación en Reumatología, Instituto de Investigación Biomédica de A Coruña (INIBIC), Complexo Hospitalario Universitario de A Coruña (CHUAC), Sergas, Universidade da Coruña (UDC), 15006, A Coruña, Spain
| | - Arianna Rodríguez-Coello
- Grupo de Investigación en Reumatología, Instituto de Investigación Biomédica de A Coruña (INIBIC), Complexo Hospitalario Universitario de A Coruña (CHUAC), Sergas, Universidade da Coruña (UDC), 15006, A Coruña, Spain
| | - Anna Saborido
- Grupo de Investigación en Reumatología, Instituto de Investigación Biomédica de A Coruña (INIBIC), Complexo Hospitalario Universitario de A Coruña (CHUAC), Sergas, Universidade da Coruña (UDC), 15006, A Coruña, Spain
| | - Elena F-Burguera
- Grupo de Investigación en Reumatología, Instituto de Investigación Biomédica de A Coruña (INIBIC), Complexo Hospitalario Universitario de A Coruña (CHUAC), Sergas, Universidade da Coruña (UDC), 15006, A Coruña, Spain
- Centro de Investigación Biomédica en Red de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Madrid, Spain
| | - Jennifer A Fernández-Rodríguez
- Grupo Envejecimiento e Inflamación, Instituto de Investigación Biomédica de A Coruña (INIBIC), Complexo Hospitalario Universitario de A Coruña (CHUAC), Sergas, Universidade da Coruña (UDC), 15006, A Coruña, Spain
| | - Rosa Meijide-Faílde
- Grupo de Terapia Celular y Medicina Regenerativa, Centro Interdisciplinar de Química e Bioloxía (CICA), Departamento de Fisioterapia, Medicina y Ciencias Biomédicas, Facultad de Ciencias da Saúde, Universidade da Coruña (UDC), 15006, A Coruña, Spain
| | - Francisco J Blanco
- Grupo de Investigación en Reumatología, Instituto de Investigación Biomédica de A Coruña (INIBIC), Complexo Hospitalario Universitario de A Coruña (CHUAC), Sergas, Universidade da Coruña (UDC), 15006, A Coruña, Spain
- Grupo de Investigación en Reumatología y Salud, Centro Interdisciplinar de Química e Bioloxía (CICA), Departamento de Fisioterapia, Medicina y Ciencias Biomédicas, Facultad de Fisioterapia, Universidade da Coruña (UDC), 15006, A Coruña, Spain
| | - Carlos Vaamonde-García
- Grupo de Investigación en Reumatología, Instituto de Investigación Biomédica de A Coruña (INIBIC), Complexo Hospitalario Universitario de A Coruña (CHUAC), Sergas, Universidade da Coruña (UDC), 15006, A Coruña, Spain.
- Grupo de Investigación en Reumatología y Salud, Centro Interdisciplinar de Química e Bioloxía (CICA), Departamento de Bioloxía, Facultad de Ciencias, Universidade da Coruña (UDC), 15008, A Coruña, Spain.
| |
Collapse
|
49
|
Mobasheri A, Thudium CS, Bay-Jensen AC, Maleitzke T, Geissler S, Duda GN, Winkler T. Biomarkers for osteoarthritis: Current status and future prospects. Best Pract Res Clin Rheumatol 2023; 37:101852. [PMID: 37620236 DOI: 10.1016/j.berh.2023.101852] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2023] [Accepted: 06/14/2023] [Indexed: 08/26/2023]
Abstract
Osteoarthritis (OA) is the most common form of arthritis globally and a major cause of pain, physical disability, and loss of economic productivity, with currently no causal treatment available. This review article focuses on current research on OA biomarkers and the potential for using biomarkers in future clinical practice and clinical trials of investigational drugs. We discuss how biomarkers, specifically soluble ones, have a long path to go before reaching clinical standards of care. We also discuss how biomarkers can help in phenotyping and subtyping to achieve enhanced stratification and move toward better-designed clinical trials. We also describe how biomarkers can be used for molecular endotyping and for determining the clinical outcomes of investigational cell-based therapies. Biomarkers have the potential to be developed as surrogate end points in clinical trials and help private-public consortia and the biotechnology and pharmaceutical industries develop more effective and targeted personalized treatments and enhance clinical care for patients with OA.
Collapse
Affiliation(s)
- Ali Mobasheri
- Research Unit of Health Sciences and Technology, Faculty of Medicine, University of Oulu, Oulu, Finland; Department of Regenerative Medicine, State Research Institute Centre for Innovative Medicine, Vilnius, Lithuania; Department of Joint Surgery, First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China; World Health Organization Collaborating Center for Public Health Aspects of Musculoskeletal Health and Aging, Université de Liège, Belgium.
| | | | | | - Tazio Maleitzke
- Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Center for Musculoskeletal Surgery, Berlin, Germany; Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Julius Wolff Institute, Berlin, Germany; Berlin Institute of Health at Charité - Universitätsmedizin Berlin, BIH Biomedical Innovation Academy, BIH Charité Clinician Scientist Program, Berlin, Germany
| | - Sven Geissler
- Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Center for Musculoskeletal Surgery, Berlin, Germany; Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Julius Wolff Institute, Berlin, Germany; Berlin Institute of Health at Charité - Universitätsmedizin Berlin, BIH Biomedical Innovation Academy, BIH Charité Clinician Scientist Program, Berlin, Germany; Berlin Center for Advanced Therapies (BECAT), Charité - Universitätsmedizin Berlin, 13353 Berlin, Germany; Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Berlin Institute of Health Center for Regenerative Therapies, Berlin, Germany
| | - Georg N Duda
- Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Center for Musculoskeletal Surgery, Berlin, Germany; Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Julius Wolff Institute, Berlin, Germany; Berlin Center for Advanced Therapies (BECAT), Charité - Universitätsmedizin Berlin, 13353 Berlin, Germany; Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Berlin Institute of Health Center for Regenerative Therapies, Berlin, Germany
| | - Tobias Winkler
- Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Center for Musculoskeletal Surgery, Berlin, Germany; Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Julius Wolff Institute, Berlin, Germany; Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Berlin Institute of Health Center for Regenerative Therapies, Berlin, Germany
| |
Collapse
|
50
|
Mariano A, Bigioni I, Marchetti M, Scotto d'Abusco A, Superti F. Repositioned Natural Compounds and Nanoformulations: A Promising Combination to Counteract Cell Damage and Inflammation in Respiratory Viral Infections. Molecules 2023; 28:molecules28104045. [PMID: 37241786 DOI: 10.3390/molecules28104045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Revised: 05/02/2023] [Accepted: 05/11/2023] [Indexed: 05/28/2023] Open
Abstract
Respiratory viral diseases are among the most important causes of disability, morbidity, and death worldwide. Due to the limited efficacy or side effects of many current therapies and the increase in antiviral-resistant viral strains, the need to find new compounds to counteract these infections is growing. Since the development of new drugs is a time-consuming and expensive process, numerous studies have focused on the reuse of commercially available compounds, such as natural molecules with therapeutic properties. This phenomenon is generally called drug repurposing or repositioning and represents a valid emerging strategy in the drug discovery field. Unfortunately, the use of natural compounds in therapy has some limitations, due to their poor kinetic performance and consequently reduced therapeutic effect. The advent of nanotechnology in biomedicine has allowed this limitation to be overcome, showing that natural compounds in nanoform may represent a promising strategy against respiratory viral infections. In this narrative review, the beneficial effects of some promising natural molecules, curcumin, resveratrol, quercetin, and vitamin C, which have been already studied both in native form and in nanoform, against respiratory viral infections are presented and discussed. The review focuses on the ability of these natural compounds, analyzed in in vitro and in vivo studies, to counteract inflammation and cellular damage induced by viral infection and provide scientific evidence of the benefits of nanoformulations in increasing the therapeutic potential of these molecules.
Collapse
Affiliation(s)
- Alessia Mariano
- Department of Biochemical Sciences, Sapienza University of Rome, 00185 Rome, Italy
| | - Irene Bigioni
- Department of Biochemical Sciences, Sapienza University of Rome, 00185 Rome, Italy
| | - Magda Marchetti
- National Centre for Innovative Technologies in Public Health, National Institute of Health, Viale Regina Elena 299, 00161 Rome, Italy
| | - Anna Scotto d'Abusco
- Department of Biochemical Sciences, Sapienza University of Rome, 00185 Rome, Italy
| | - Fabiana Superti
- National Centre for Innovative Technologies in Public Health, National Institute of Health, Viale Regina Elena 299, 00161 Rome, Italy
| |
Collapse
|