1
|
Liu S, Zhang G, Li N, Wang Z, Lu L. The Interplay of Aging and PANoptosis in Osteoarthritis Pathogenesis: Implications for Novel Therapeutic Strategies. J Inflamm Res 2025; 18:1951-1967. [PMID: 39959642 PMCID: PMC11829118 DOI: 10.2147/jir.s489613] [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: 08/01/2024] [Accepted: 01/22/2025] [Indexed: 02/18/2025] Open
Abstract
Osteoarthritis (OA) is a common degenerative joint disease characterized by the progressive degradation of articular cartilage, synovial inflammation, and subchondral bone remodeling. This review explores the interplay between aging, PANoptosis, and inflammation in OA progression. Age-related cellular and immune dysfunctions, including cellular senescence, senescence-associated secretory phenotypes (SASPs), and immunosenescence, significantly contribute to joint degeneration. In OA, dysregulated apoptosis, necroptosis, and pyroptosis, particularly in chondrocytes, exacerbate cartilage damage. Apoptosis, mediated by the JNK pathway, reduces chondrocyte density, while necroptosis and pyroptosis, involving RIPK-1/RIPK-3 and the NLRP3 inflammasome, respectively, amplify inflammation and cartilage destruction. Inflammatory cytokines and damage-associated molecular patterns (DAMPs) further enhance these PANoptotic pathways. Current therapeutic strategies primarily focus on anti-inflammatory agents such as non-steroidal anti-inflammatory drugs (NSAIDs) and corticosteroids, with growing interest in anti-senescence drugs targeting cellular senescence and SASP. Additionally, exploring PANoptosis mechanisms offers potential for innovative OA treatments.
Collapse
Affiliation(s)
- Shaoshan Liu
- Department of Joint Surgery, Liaocheng Traditional Chinese Medicine Hospital, Liaocheng, 252000, People's Republic of China
| | - Guifeng Zhang
- Department of Neurology, Liaocheng People's Hospital and Liaocheng Hospital Affiliated to Shandong First Medical University, Liaocheng, 252000, People's Republic of China
| | - Nan Li
- Department of Trauma Orthopedics, Liaocheng Traditional Chinese Medicine Hospital, Liaocheng, 252000, People's Republic of China
| | - Zheng Wang
- Department of Neurosurgery, Liaocheng Traditional Chinese Medicine Hospital, Liaocheng, 252000, People's Republic of China
| | - Liaodong Lu
- Department of Joint Surgery, Liaocheng Traditional Chinese Medicine Hospital, Liaocheng, 252000, People's Republic of China
| |
Collapse
|
2
|
Cruz CA, Pruneski JA, McAllister RN, Riopelle D, Bottoni CR. Fifteen-Year Radiographic Follow-up Comparison of Early Versus Delayed ACL Reconstruction: A Retrospective Review of a Previous Prospective Randomized Clinical Trial. Orthop J Sports Med 2024; 12:23259671241298753. [PMID: 39669710 PMCID: PMC11635895 DOI: 10.1177/23259671241298753] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/27/2024] [Accepted: 10/10/2024] [Indexed: 12/14/2024] Open
Abstract
Background Posttraumatic osteoarthritis (PTOA) after anterior cruciate ligament injury and reconstruction (ACLR) is a prevalent cause of long-term disability. Few studies have compared the effect of ACLR timing on the development of PTOA. Purpose/Hypothesis The purpose of this study was to compare the rate of PTOA at a long-term follow-up between patients who underwent early ACLR (<21 days after injury) versus delayed ACLR (>6 weeks after injury). The authors hypothesized that patients who underwent early ACLR would have lower rates of PTOA compared with the delayed ACLR cohort. Study Design Cohort study; Level of evidence, 2. Methods The authors contacted patients from a previous prospective randomized controlled trial who were randomized to undergo either early (<21 days) or delayed (>6 weeks) ACLR with hamstring tendon autografts. Weightbearing radiographs were obtained at a minimum 15-year follow-up, and radiographic PTOA was evaluated using the Kellgren-Lawrence (K-L) classification system. The prevalence of pathologies was compared between the early and delayed groups using appropriate testing, and logistic regression was used to evaluate for associations with failure-a K-L grade of ≥2 or conversion to total knee arthroplasty (TKA). Results At a mean follow-up of 15.6 years, radiographs were obtained for 58 (28 early, 30 delayed) of the original 69 (84.1%) patients. High rates of PTOA (K-L grade ≥2) were observed in the early (82.1%) and delayed (86.7%) cohorts (P = .634). Two (7.1%) patients in the early cohort converted to TKA compared with 4 (13.3%) patients in the delayed cohort (P = .44). Surgical timing did not affect arthritis severity (P≥ .4), and no factors predicted developing radiographic PTOA in either cohort (P > .2). Increased time from injury decreased the odds of failure in the early ACLR cohort (odds ratio, 0.79; P = .041). Conclusion In this study, >80% of patients who underwent ACLR with hamstring tendon autografts had radiographic evidence of PTOA at a mean 15.6-year follow-up, with no difference in the prevalence or severity of PTOA between the early and delayed groups. In addition, 11% of patients had converted to TKA by the time of the final follow-up, and the conversion rate did not differ according to the timing of ACLR.
Collapse
Affiliation(s)
| | - James A. Pruneski
- Department of Orthopaedic Surgery, Tripler Army Medical Center, Honolulu, Hawaii, USA
| | - Rebecca N. McAllister
- Department of Orthopaedic Surgery, Tripler Army Medical Center, Honolulu, Hawaii, USA
| | - David Riopelle
- Department of Orthopaedic Surgery, University of Southern California, Los Angeles, California, USA. Presented at the AOSSM Annual Meeting, Washington, District of Columbia, July 2023
| | - Craig R. Bottoni
- Department of Orthopaedic Surgery, Tripler Army Medical Center, Honolulu, Hawaii, USA
| |
Collapse
|
3
|
Lo Presti M, Costa GG, Agrò G, Vasco C, Boffa A, Di Martino A, Andriolo L, Cenacchi A, Zaffagnini S, Filardo G. Platelet-Rich Plasma Injections Do Not Improve the Recovery After Arthroscopic Partial Meniscectomy: A Double-Blind Randomized Controlled Trial. Am J Sports Med 2024; 52:3198-3205. [PMID: 39425245 PMCID: PMC11542319 DOI: 10.1177/03635465241283052] [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: 04/12/2024] [Accepted: 08/13/2024] [Indexed: 10/21/2024]
Abstract
BACKGROUND Arthroscopic meniscectomy is one of the most performed surgical procedures in orthopaedics. Different approaches have been proposed to improve patient recovery but with unsatisfactory results. Platelet-rich plasma (PRP) augmentation has been proposed as a strategy to improve the recovery after meniscectomy. PURPOSE To investigate the clinical benefits of an intra-articular PRP injection after meniscectomy, in terms of faster and better patient recovery. STUDY DESIGN Randomized controlled trial; Level of evidence, 1. METHODS Ninety patients were randomized into a treatment group, with arthroscopic partial meniscectomy immediately followed by a 5-mL injection of autologous conditioned plasma, and a control group with partial meniscectomy alone. Patients were evaluated at baseline and at 15, 30, 60, and 180 days of follow-up with the visual analog scale (VAS) score for pain (primary outcome), as well as with International Knee Documentation Committee subjective score, Knee injury and Osteoarthritis Outcome Score subscales, Tegner score, and EuroQol-Visual Analog Scale score. Objective evaluation was performed analyzing knee range of motion and circumference and the International Knee Documentation Committee objective score. Complications, patient judgment, and satisfaction were documented as well. RESULTS No major complications and an overall significant improvement in the clinical scores were observed in both groups. Overall, the comparative analysis did not demonstrate significant between-group differences in absolute values or improvements of both subjective and objective scores, as well as activity level. The improvement in terms of VAS pain score for the treatment group was significant already at 15 days (from 4.3 ± 2.5 to 2.5 ± 2.5; P = .014), while in the control group it became significant at 30 days (from 3.7 ± 2.3 to 2.0 ± 2.4; P = .004). No significant differences were observed between the 2 groups in terms of judgment of treatment results and satisfaction. CONCLUSION A single postoperative injection of PRP was not able to significantly improve patient recovery after arthroscopic partial meniscectomy. PRP augmentation did not provide overall benefits at a short-term follow-up (6 months) in terms of pain relief, function, objective parameters, and return-to-sport activities. REGISTRATION NCT02872753 (ClinicalTrials.gov identifier).
Collapse
Affiliation(s)
- Mirco Lo Presti
- Clinica Ortopedica e Traumatologica 2, IRCCS Istituto Ortopedico Rizzoli, Bologna, Italy
| | | | | | - Cosimo Vasco
- U.O.C. Ortopedia e Traumatologia, Ospedale S. Maria della Scaletta, Imola, Italy
| | - Angelo Boffa
- Clinica Ortopedica e Traumatologica 2, IRCCS Istituto Ortopedico Rizzoli, Bologna, Italy
| | - Alessandro Di Martino
- Clinica Ortopedica e Traumatologica 2, IRCCS Istituto Ortopedico Rizzoli, Bologna, Italy
- Applied and Translational Research center (ATRc), IRCCS Istituto Ortopedico Rizzoli, Bologna, Italy
| | - Luca Andriolo
- Clinica Ortopedica e Traumatologica 2, IRCCS Istituto Ortopedico Rizzoli, Bologna, Italy
| | | | - Stefano Zaffagnini
- Clinica Ortopedica e Traumatologica 2, IRCCS Istituto Ortopedico Rizzoli, Bologna, Italy
| | - Giuseppe Filardo
- Applied and Translational Research center (ATRc), IRCCS Istituto Ortopedico Rizzoli, Bologna, Italy
- Faculty of Biomedical Sciences, Università della Svizzera Italiana, Lugano, Switzerland
| |
Collapse
|
4
|
Sun C, Chang K, Fleming BC, Owens BD, Beveridge JE, Zhao Y, Peng G, Wei L. Alpha-2-Macroglobulin Attenuates Posttraumatic Osteoarthritis Cartilage Damage by Inhibiting Inflammatory Pathways With Modified Intra-articular Drilling in a Yucatan Minipig Model. Am J Sports Med 2024; 52:2882-2892. [PMID: 39214071 DOI: 10.1177/03635465241272401] [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] [Indexed: 09/04/2024]
Abstract
BACKGROUND Posttraumatic osteoarthritis (PTOA) arises secondarily to joint trauma and is driven by catabolic inflammatory pathways. Alpha-2-macroglobulin (α2M) is a naturally occurring proteinase inhibitor found in human serum and synovial fluid that binds proteases as well as proinflammatory cytokines involved in the pathogenesis of PTOA. PURPOSE (1) To investigate the therapeutic potential of intra-articular α2M injections during the acute stages of PTOA by inhibiting inflammatory pathways driven by the cytokines expressed by the synovium in a large preclinical Yucatan minipig model and (2) to determine if 3 intra-articular α2M injections have greater chondroprotective effects compared with 1 intra-articular injection. STUDY DESIGN Controlled laboratory study. METHODS A total of 48 Yucatan minipigs were randomized into 4 groups (n = 12 each): (1) modified intra-articular drilling (mIAD) and saline (mIAD + saline), (2) mIAD and 1 intra-articular α2M injection (mIAD +α2M-1), (3) mIAD and 3 α2M injections (mIAD +α2M-3), and (4) sham control. Surgical hindlimbs were harvested at 15 weeks after surgery. Cartilage degeneration, synovial changes, inflammatory gene expression, and matrix metalloproteinase levels were evaluated. Gait asymmetry was measured before and after surgery using a pressure-sensing walkway system. RESULTS Macroscopic lesion areas and microscopic cartilage degeneration scores were lower in the mIAD +α2M-1 and mIAD +α2M-3 groups compared with the mIAD + saline group (P < .05) and similar to those in the sham group (P > .05). Synovial membrane scores of the mIAD +α2M-1 and mIAD +α2M-3 groups were lower than that of the mIAD + saline group (P < .05) and higher than that of the sham group (P < .05). Interleukin-1 beta, nuclear factor kappa B, and tumor necrosis factor alpha mRNA expression in the synovium and matrix metalloproteinase-1 levels in synovial fluid were significantly lower in the mIAD +α2M-1 and mIAD +α2M-3 groups compared with the mIAD + saline group (P < .05). No significant differences were observed between the mIAD +α2M-1 and mIAD +α2M-3 groups for all measured outcomes. There were early changes in gait (P < .05) between preoperative and postoperative time points for the mIAD + saline, mIAD +α2M-1, and mIAD +α2M-3 groups that normalized by 15 weeks. CONCLUSION Animals receiving early α2M treatment exhibited less cartilage damage, milder synovitis, and lower inflammation compared with animals with no α2M treatment. These results exemplify the early anti-inflammatory effects of α2M and provide evidence that intra-articular α2M injections may slow the progression of PTOA. CLINICAL RELEVANCE In patients presenting with an acute joint injury, an early intervention with α2M may have the potential to reduce cartilage degeneration from catabolic pathways and delay the development of PTOA.
Collapse
Affiliation(s)
- Changqi Sun
- Department of Orthopaedics, Rhode Island Hospital, Warren Alpert Medical School, Brown University, Providence, Rhode Island, USA
| | - Kenny Chang
- Department of Orthopaedics, Rhode Island Hospital, Warren Alpert Medical School, Brown University, Providence, Rhode Island, USA
| | - Braden C Fleming
- Department of Orthopaedics, Rhode Island Hospital, Warren Alpert Medical School, Brown University, Providence, Rhode Island, USA
| | - Brett D Owens
- Department of Orthopaedics, Rhode Island Hospital, Warren Alpert Medical School, Brown University, Providence, Rhode Island, USA
| | - Jillian E Beveridge
- Department of Orthopaedics, Rhode Island Hospital, Warren Alpert Medical School, Brown University, Providence, Rhode Island, USA
| | - Yu Zhao
- University of Rochester Medical Center, Rochester, New York, USA
| | - Guoxuan Peng
- Department of Orthopaedics, Rhode Island Hospital, Warren Alpert Medical School, Brown University, Providence, Rhode Island, USA
| | - Lei Wei
- Department of Orthopaedics, Rhode Island Hospital, Warren Alpert Medical School, Brown University, Providence, Rhode Island, USA
| |
Collapse
|
5
|
Betsch K, Martinez VG, Lyons LP, Weinberg JB, Wittstein JR, McNulty AL. Shedding light on the effects of blood on meniscus tissue: the role of mononuclear leukocytes in mediating meniscus catabolism. Osteoarthritis Cartilage 2024; 32:938-949. [PMID: 38782253 PMCID: PMC11254574 DOI: 10.1016/j.joca.2024.04.022] [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: 10/17/2023] [Revised: 04/12/2024] [Accepted: 04/23/2024] [Indexed: 05/25/2024]
Abstract
OBJECTIVE Traumatic meniscal injuries can cause acute pain, hemarthrosis (bleeding into the joint), joint immobility, and post-traumatic osteoarthritis (PTOA). However, the exact mechanism(s) by which PTOA develops following meniscal injuries is unknown. Since meniscus tears commonly coincide with hemarthrosis, investigating the direct effects of blood and its constituents on meniscus tissue is warranted. The goal of this study was to determine the direct effects of blood and blood components on meniscus tissue catabolism. METHODS Porcine meniscus explants or primary meniscus cells were exposed to whole blood or various fractions of blood for 3 days to simulate blood exposure following injury. Explants were then washed and cultured for an additional 3 days prior to collection for biochemical analyses. RESULTS Whole blood increased matrix metalloproteinase (MMP) activity. Fractionation experiments revealed blood-derived red blood cells did not affect meniscus catabolism. Conversely, viable mononuclear leukocytes induced MMP activity, nitric oxide (NO) production, and loss of tissue sulfated glycosaminoglycan (sGAG) content, suggesting that these cells are mediating meniscus catabolism. CONCLUSIONS These findings highlight the potential challenges of meniscus healing in the presence of hemarthrosis and the need for further research to elucidate the in vivo effects of blood and blood-derived mononuclear leukocytes due to both hemarthrosis and blood-derived therapeutics.
Collapse
Affiliation(s)
- Kevin Betsch
- Department of Orthopaedic Surgery, Duke University School of Medicine, Durham, NC, USA.
| | - Vianna G Martinez
- Department of Pathology, Duke University School of Medicine, Durham, NC, USA.
| | - Lucas P Lyons
- Department of Orthopaedic Surgery, Duke University School of Medicine, Durham, NC, USA.
| | - J Brice Weinberg
- Department of Medicine, VA Medical Center, Durham, NC, USA; Department of Medicine, Duke University School of Medicine, Durham, NC, USA.
| | - Jocelyn R Wittstein
- Department of Orthopaedic Surgery, Duke University School of Medicine, Durham, NC, USA.
| | - Amy L McNulty
- Department of Orthopaedic Surgery, Duke University School of Medicine, Durham, NC, USA; Department of Pathology, Duke University School of Medicine, Durham, NC, USA; Department of Biomedical Engineering, Duke University, Durham, NC, USA.
| |
Collapse
|
6
|
Nishida Y, Hashimoto Y, Orita K, Nishino K, Kinoshita T, Iida K, Nakamura H. Longitudinal measurement of serum cartilage oligomeric matrix protein can detect the progression of cartilage degeneration in anterior cruciate ligament reconstruction patients. Asia Pac J Sports Med Arthrosc Rehabil Technol 2024; 37:27-32. [PMID: 39091893 PMCID: PMC11292425 DOI: 10.1016/j.asmart.2024.06.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2024] [Revised: 04/16/2024] [Accepted: 06/20/2024] [Indexed: 08/04/2024] Open
Abstract
Background/objective Cartilage oligomeric matrix protein (COMP) has utility as a diagnostic marker for osteoarthritis (OA). Our previous study revealed that the serum COMP level can be used to detect early cartilage change in non-OA patients with anterior cruciate ligament (ACL)-deficiency. However, there are still no studies on detecting the progression of cartilage degeneration in early OA. The aim of present study was to investigate whether serum COMP can detect the progression of cartilage degeneration after ACL reconstruction in non-OA patients. Methods Patients without cartilage degeneration of early OA at ACL reconstruction and whose serum COMP levels could be measured were included in the study. Cartilage degeneration of early OA were defined as International Cartilage Repair Society (ICRS) grade 1 to 4 in more than 2 compartments or ICRS grade 2 to 4 in 1 compartment. The patients were divided into two groups: those who had cartilage degeneration of early OA at second-look arthroscopy (cartilage degeneration progression group) and those who did not (non-progression group), and the serum COMP values between the two groups were compared. Results Thirty-one patients were included. There were 8 cases (25.8 %) in progression group and 23 cases (74.2 %) in non-progression group. There were significant differences between the two groups regarding age and change in serum COMP level. In terms of the rate of change in COMP, an increase of more than 1.24-fold was the cut-off value for detecting the progression of cartilage degeneration. Conclusions In this study, the increase in serum COMP levels was significantly greater in progressed cartilage degeneration group than non-progression group after ACL reconstruction. Longitudinal serum COMP measurement could detect the progression of cartilage degeneration. Level of evidence Level Ⅲ, retrospective comparative study.
Collapse
Affiliation(s)
- Yohei Nishida
- Department of Orthopaedic Surgery, Osaka City General Hospital, Osaka, Japan
| | - Yusuke Hashimoto
- Department of Health and Sport Management, Osaka University of Health and Sports Science, Graduate School of Sport and Exercise Science, Osaka, Japan
- Department of Orthopaedic Surgery, Osaka Metropolitan University Graduate School of Medicine, Osaka, Japan
| | - Kumi Orita
- Department of Orthopaedic Surgery, Osaka Metropolitan University Graduate School of Medicine, Osaka, Japan
| | - Kazuya Nishino
- Department of Orthopaedic Surgery, Osaka Metropolitan University Graduate School of Medicine, Osaka, Japan
| | - Takuya Kinoshita
- Department of Orthopaedic Surgery, Osaka Saiseikai Nakatsu Hospital, Osaka, Japan
| | - Ken Iida
- Department of Orthopaedic Surgery, Osaka Metropolitan University Graduate School of Medicine, Osaka, Japan
| | - Hiroaki Nakamura
- Department of Orthopaedic Surgery, Osaka Metropolitan University Graduate School of Medicine, Osaka, Japan
| |
Collapse
|
7
|
Abdal Dayem A, Yan E, Do M, Kim Y, Lee Y, Cho SG, Kim DH. Engineering extracellular vesicles for ROS scavenging and tissue regeneration. NANO CONVERGENCE 2024; 11:24. [PMID: 38922501 PMCID: PMC11208369 DOI: 10.1186/s40580-024-00430-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Accepted: 05/22/2024] [Indexed: 06/27/2024]
Abstract
Stem cell therapy holds promise for tissue regeneration, yet significant challenges persist. Emerging as a safer and potentially more effective alternative, extracellular vesicles (EVs) derived from stem cells exhibit remarkable abilities to activate critical signaling cascades, thereby facilitating tissue repair. EVs, nano-scale membrane vesicles, mediate intercellular communication by encapsulating a diverse cargo of proteins, lipids, and nucleic acids. Their therapeutic potential lies in delivering cargos, activating signaling pathways, and efficiently mitigating oxidative stress-an essential aspect of overcoming limitations in stem cell-based tissue repair. This review focuses on engineering and applying EVs in tissue regeneration, emphasizing their role in regulating reactive oxygen species (ROS) pathways. Additionally, we explore strategies to enhance EV therapeutic activity, including functionalization and incorporation of antioxidant defense proteins. Understanding these molecular mechanisms is crucial for optimizing EV-based regenerative therapies. Insights into EV and ROS signaling modulation pave the way for targeted and efficient regenerative therapies harnessing the potential of EVs.
Collapse
Affiliation(s)
- Ahmed Abdal Dayem
- Department of Stem Cell and Regenerative Biotechnology, Molecular & Cellular Reprogramming Center, Institute of Advanced Regenerative Science, Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul, 05029, Republic of Korea
| | - Ellie Yan
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, 21205, USA
| | - Minjae Do
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, 21205, USA
| | - Yoojung Kim
- Department of Stem Cell and Regenerative Biotechnology, Molecular & Cellular Reprogramming Center, Institute of Advanced Regenerative Science, Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul, 05029, Republic of Korea
| | - Yeongseo Lee
- Department of Stem Cell and Regenerative Biotechnology, Molecular & Cellular Reprogramming Center, Institute of Advanced Regenerative Science, Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul, 05029, Republic of Korea
| | - Ssang-Goo Cho
- Department of Stem Cell and Regenerative Biotechnology, Molecular & Cellular Reprogramming Center, Institute of Advanced Regenerative Science, Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul, 05029, Republic of Korea.
- R&D Team, StemExOne Co., Ltd., 307 KU Technology Innovation Bldg, 120, Neungdong-ro, Gwangjin- gu, Seoul, 05029, Republic of Korea.
| | - Deok-Ho Kim
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, 21205, USA.
- Department of Mechanical Engineering, Johns Hopkins University, Baltimore, MD, 21205, USA.
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, 21205, USA.
- Center for Microphysiological Systems, Johns Hopkins University, Baltimore, MD, 21205, USA.
- Institute for NanoBiotechnology, Johns Hopkins University, Baltimore, MD, 21218, USA.
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA.
| |
Collapse
|
8
|
Hart DA. The Heterogeneity of Post-Menopausal Disease Risk: Could the Basis for Why Only Subsets of Females Are Affected Be Due to a Reversible Epigenetic Modification System Associated with Puberty, Menstrual Cycles, Pregnancy and Lactation, and, Ultimately, Menopause? Int J Mol Sci 2024; 25:3866. [PMID: 38612676 PMCID: PMC11011715 DOI: 10.3390/ijms25073866] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2024] [Revised: 03/19/2024] [Accepted: 03/28/2024] [Indexed: 04/14/2024] Open
Abstract
For much of human evolution, the average lifespan was <40 years, due in part to disease, infant mortality, predators, food insecurity, and, for females, complications of childbirth. Thus, for much of evolution, many females did not reach the age of menopause (45-50 years of age) and it is mainly in the past several hundred years that the lifespan has been extended to >75 years, primarily due to public health advances, medical interventions, antibiotics, and nutrition. Therefore, the underlying biological mechanisms responsible for disease risk following menopause must have evolved during the complex processes leading to Homo sapiens to serve functions in the pre-menopausal state. Furthermore, as a primary function for the survival of the species is effective reproduction, it is likely that most of the advantages of having such post-menopausal risks relate to reproduction and the ability to address environmental stresses. This opinion/perspective will be discussed in the context of how such post-menopausal risks could enhance reproduction, with improved survival of offspring, and perhaps why such risks are preserved. Not all post-menopausal females exhibit risk for this set of diseases, and those who do develop such diseases do not have all of the conditions. The diseases of the post-menopausal state do not operate as a unified complex, but as independent variables, with the potential for some overlap. The how and why there would be such heterogeneity if the risk factors serve essential functions during the reproductive years is also discussed and the concept of sets of reversible epigenetic changes associated with puberty, pregnancy, and lactation is offered to explain the observations regarding the distribution of post-menopausal conditions and their potential roles in reproduction. While the involvement of an epigenetic system with a dynamic "modification-demodification-remodification" paradigm contributing to disease risk is a hypothesis at this point, validation of it could lead to a better understanding of post-menopausal disease risk in the context of reproduction with commonalities may also lead to future improved interventions to control such risk after menopause.
Collapse
Affiliation(s)
- David A Hart
- Department of Surgery, Faculty of Kinesiology, and McCaig Institute for Bone and Joint Health, University of Calgary, Calgary, AB T2N 4N1, Canada
| |
Collapse
|
9
|
Nauth A, Haller J, Augat P, Anderson DD, McKee MD, Shearer D, Jenkinson R, Pape HC. Distal femur fractures: basic science and international perspectives. OTA Int 2024; 7:e320. [PMID: 38487402 PMCID: PMC10936154 DOI: 10.1097/oi9.0000000000000320] [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: 11/12/2023] [Revised: 12/21/2023] [Accepted: 12/26/2023] [Indexed: 03/17/2024]
Abstract
Distal femur fractures are challenging injuries to manage, and complication rates remain high. This article summarizes the international and basic science perspectives regarding distal femoral fractures that were presented at the 2022 Orthopaedic Trauma Association Annual Meeting. We review a number of critical concepts that can be considered to optimize the treatment of these difficult fractures. These include biomechanical considerations for distal femur fixation constructs, emerging treatments to prevent post-traumatic arthritis, both systemic and local biologic treatments to optimize nonunion management, the relative advantages and disadvantages of plate versus nail versus dual-implant constructs, and finally important factors which determine outcomes. A robust understanding of these principles can significantly improve success rates and minimize complications in the treatment of these challenging injuries.
Collapse
Affiliation(s)
- Aaron Nauth
- St. Michael's Hospital, University of Toronto, Toronto, ON, Canada
| | | | - Peter Augat
- Paracelsus Medical University in Salzburg, Austria and Institute of Biomechanics at Trauma Centre Murnau, Salzburg, Germany
| | - Donald D. Anderson
- Department of Orthopedics and Rehabilitation, University of Iowa, Iowa City, IA
| | - Michael D. McKee
- Department of Orthopaedic Surgery, University of Arizona College of Medicine, Phoenix, AZ
| | - David Shearer
- Department of Orthopaedic Surgery, University of California, San Francisco, CA
| | - Richard Jenkinson
- Sunnybrook Health Sciences Centre, University of Toronto, Toronto, ON, Canada
| | | |
Collapse
|
10
|
Wu D, Shen Z, Gou Y, Yu T, Hong J, Wang Y, Ni F, Qiqige N, Lu H, Xue E. PPAR γ activation in chondrocytes alleviates glucocorticoid-induced oxidative stress, mitochondrial impairment, and pyroptosis via autophagic flow enhancement. Chem Biol Interact 2024; 390:110877. [PMID: 38286393 DOI: 10.1016/j.cbi.2024.110877] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Revised: 01/03/2024] [Accepted: 01/16/2024] [Indexed: 01/31/2024]
Abstract
Osteoarthritis (OA) is a progressive age-related disease characterised by pathological changes in the synovium, articular cartilage, and subchondral bone, significantly reducing the patients' quality of life. This study investigated the role of glucocorticoids, specifically dexamethasone, in OA progression, with a particular focus on their effects on chondrocytes. Although glucocorticoids are commonly used for OA pain relief, our research demonstrated that high concentrations of dexamethasone may accelerate OA progression by enhancing the ability of reactive oxygen species to inhibit chondrocyte autophagy, resulting in cell death and accelerated cartilage degeneration. Despite reports on the acceleration of pathogenesis and cartilage damage in some patients of OA taking corticosteroids, the mechanism behind the same has not been investigated. This necessitates an investigation of the concentration-dependent changes in the cartilage cells upon dexamethasone administration. In addition, the protective effect of PPAR γ on chondrocytes can prevent the decrease in chondrocyte autophagy and delay cartilage degeneration. Therefore, our study suggests that the therapeutic use of glucocorticoids in OA treatment should be more nuanced considering their potential detrimental effects. Future investigations should focus on the mechanisms underlying the glucocorticoid-mediated modulation of cell death processes, which could provide insights into new therapeutic strategies for OA treatment.
Collapse
Affiliation(s)
- Dengying Wu
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Childrens Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province, China; Key Laboratory of Orthopaedics of Zhejiang Province, Wenzhou, Zhejiang Province, China
| | - Zhenyu Shen
- Key Laboratory of Orthopaedics of Zhejiang Province, Wenzhou, Zhejiang Province, China; The Second School of Medicine, Wenzhou Medical University, Wenzhou, Zhejiang Province, China
| | - Yong Gou
- Key Laboratory of Orthopaedics of Zhejiang Province, Wenzhou, Zhejiang Province, China; The Second School of Medicine, Wenzhou Medical University, Wenzhou, Zhejiang Province, China
| | - Tao Yu
- The Second School of Medicine, Wenzhou Medical University, Wenzhou, Zhejiang Province, China
| | - Jiaqian Hong
- The Second School of Medicine, Wenzhou Medical University, Wenzhou, Zhejiang Province, China
| | - Yitong Wang
- The Second School of Medicine, Wenzhou Medical University, Wenzhou, Zhejiang Province, China
| | - Feifei Ni
- The Second School of Medicine, Wenzhou Medical University, Wenzhou, Zhejiang Province, China
| | - Naren Qiqige
- The Second School of Medicine, Wenzhou Medical University, Wenzhou, Zhejiang Province, China
| | - Hongwei Lu
- Key Laboratory of Orthopaedics of Zhejiang Province, Wenzhou, Zhejiang Province, China; The Second School of Medicine, Wenzhou Medical University, Wenzhou, Zhejiang Province, China
| | - Enxing Xue
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Childrens Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province, China; Key Laboratory of Orthopaedics of Zhejiang Province, Wenzhou, Zhejiang Province, China.
| |
Collapse
|
11
|
Luo D, Zhu H, Li S, Wang Z, Xiao J. Mesenchymal stem cell-derived exosomes as a promising cell-free therapy for knee osteoarthritis. Front Bioeng Biotechnol 2024; 12:1309946. [PMID: 38292826 PMCID: PMC10824863 DOI: 10.3389/fbioe.2024.1309946] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Accepted: 01/05/2024] [Indexed: 02/01/2024] Open
Abstract
Osteoarthritis (OA), as a degenerative disease, leads to high socioeconomic burdens and disability rates. The knee joint is typically the most affected and is characterized by progressive destruction of articular cartilage, subchondral bone remodeling, osteophyte formation and synovial inflammation. The current management of OA mainly focuses on symptomatic relief and does not help to slow down the advancement of disease. Recently, mesenchymal stem cells (MSCs) and their exosomes have garnered significant attention in regenerative therapy and tissue engineering areas. Preclinical studies have demonstrated that MSC-derived exosomes (MSC-Exos), as bioactive factor carriers, have promising results in cell-free therapy of OA. This study reviewed the application of various MSC-Exos for the OA treatment, along with exploring the potential underlying mechanisms. Moreover, current strategies and future perspectives for the utilization of engineered MSC-Exos, alongside their associated challenges, were also discussed.
Collapse
Affiliation(s)
| | | | | | - Zhenggang Wang
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jun Xiao
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
12
|
Riggs KC, Sankar U. Inflammatory mechanisms in post-traumatic osteoarthritis: a role for CaMKK2. IMMUNOMETABOLISM (COBHAM, SURREY) 2023; 5:e00031. [PMID: 37849987 PMCID: PMC10578519 DOI: 10.1097/in9.0000000000000031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/14/2023] [Accepted: 08/23/2023] [Indexed: 10/19/2023]
Abstract
Post-traumatic osteoarthritis (PTOA) is a multifactorial disease of the cartilage, synovium, and subchondral bone resulting from direct joint trauma and altered joint mechanics after traumatic injury. There are no current disease-modifying therapies for PTOA, and early surgical interventions focused on stabilizing the joint do not halt disease progression. Chronic pain and functional disability negatively affect the quality of life and take an economic toll on affected patients. While multiple mechanisms are at play in disease progression, joint inflammation is a key contributor. Impact-induced mitochondrial dysfunction and cell death or altered joint mechanics after trauma culminate in inflammatory cytokine release from synoviocytes and chondrocytes, cartilage catabolism, suppression of cartilage anabolism, synovitis, and subchondral bone disease, highlighting the complexity of the disease. Current understanding of the cellular and molecular mechanisms underlying the disease pathology has allowed for the investigation of a variety of therapeutic strategies that target unique apoptotic and/or inflammatory processes in the joint. This review provides a concise overview of the inflammatory and apoptotic mechanisms underlying PTOA pathogenesis and identifies potential therapeutic targets to mitigate disease progression. We highlight Ca2+/calmodulin-dependent protein kinase kinase 2 (CaMKK2), a serine/threonine protein kinase that was recently identified to play a role in murine and human osteoarthritis pathogenesis by coordinating chondrocyte inflammatory responses and apoptosis. Given its additional effects in regulating macrophage inflammatory signaling and bone remodeling, CaMKK2 emerges as a promising disease-modifying therapeutic target against PTOA.
Collapse
Affiliation(s)
- Keegan C. Riggs
- Department of Anatomy, Cell Biology and Physiology, Indiana University School of Medicine, Indianapolis, IN, USA
- Indiana Center for Musculoskeletal Health, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Uma Sankar
- Department of Anatomy, Cell Biology and Physiology, Indiana University School of Medicine, Indianapolis, IN, USA
- Indiana Center for Musculoskeletal Health, Indiana University School of Medicine, Indianapolis, IN, USA
| |
Collapse
|
13
|
Anjiki K, Matsumoto T, Kuroda Y, Fujita M, Hayashi S, Nakano N, Tsubosaka M, Kamenaga T, Takashima Y, Kikuchi K, Ikuta K, Onoi Y, Tachibana S, Suda Y, Wada K, Matsushita T, Kuroda R. Heterogeneous Cells as well as Adipose-Derived Stromal Cells in Stromal Vascular Fraction Contribute to Enhance Anabolic and Inhibit Catabolic Factors in Osteoarthritis. Stem Cell Rev Rep 2023; 19:2407-2419. [PMID: 37477775 DOI: 10.1007/s12015-023-10589-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/07/2023] [Indexed: 07/22/2023]
Abstract
The stromal-vascular fraction (SVF), comprising heterogeneous cell populations and adipose-derived stromal cells (ADSCs), has therapeutic potential against osteoarthritis (OA); however, the underlying mechanism remains elusive. This study aimed to investigate the therapeutic effects of heterogeneous cells in rabbit SVF on rabbit chondrocytes. Rabbit SVF and ADSCs were autografted into knees at OA onset. The SVF (1 × 105) and low-dose ADSCs (lADSC; 1 × 104) groups adjusted for their stromal cell content were compared. Animals were euthanized 8 and 12 weeks after OA onset for macroscopic and histological analyses of OA progression and synovitis. Immunohistochemical and real-time polymerase chain reaction assessments were conducted. In vitro, immune-fluorescent double staining was performed for SVF to stain macrophages with F4/80, CD86(M1), and CD163(M2). OA progression was markedly suppressed, and synovitis was reduced in the SVF groups (OARSI histological score 8 W: 6.8 ± 0.75 vs. 3.8 ± 0.75, p = 0.001; 12 W: 8.8 ± 0.4 vs. 5.4 ± 0.49, p = 0.0002). The SVF groups had higher expression of collagen II and SOX9 in cartilage and TGF-β and IL-10 in the synovium, lower expression of MMP-13, and lower macrophage M1/M2 ratio than the lADSC groups. Immunofluorescent double staining revealed a markedly higher number of M2 than that of M1 macrophages in the SVF. The therapeutic effects of SVF on chondrocytes were superior than those of lADSCs, with enhanced anabolic and inhibited catabolic factors. Heterogeneous cells, mainly M2 macrophages in the SVF, enhanced growth factor secretion and chondrocyte-protective cytokines, thus benefiting chondrocytes and knee joint homeostasis. Overall, the SVF is a safe, relatively simple, and a useful treatment option for OA.
Collapse
Affiliation(s)
- Kensuke Anjiki
- Department of Orthopaedic Surgery, Kobe University Graduate School of Medicine, 7-5-1, Kusunoki-cho, Chuo-ku, Kobe, 650-0017, Japan
| | - Tomoyuki Matsumoto
- Department of Orthopaedic Surgery, Kobe University Graduate School of Medicine, 7-5-1, Kusunoki-cho, Chuo-ku, Kobe, 650-0017, Japan.
| | - Yuichi Kuroda
- Department of Orthopaedic Surgery, Kobe University Graduate School of Medicine, 7-5-1, Kusunoki-cho, Chuo-ku, Kobe, 650-0017, Japan
| | - Masahiro Fujita
- Department of Orthopaedic Surgery, Kobe University Graduate School of Medicine, 7-5-1, Kusunoki-cho, Chuo-ku, Kobe, 650-0017, Japan
| | - Shinya Hayashi
- Department of Orthopaedic Surgery, Kobe University Graduate School of Medicine, 7-5-1, Kusunoki-cho, Chuo-ku, Kobe, 650-0017, Japan
| | - Naoki Nakano
- Department of Orthopaedic Surgery, Kobe University Graduate School of Medicine, 7-5-1, Kusunoki-cho, Chuo-ku, Kobe, 650-0017, Japan
| | - Masanori Tsubosaka
- Department of Orthopaedic Surgery, Kobe University Graduate School of Medicine, 7-5-1, Kusunoki-cho, Chuo-ku, Kobe, 650-0017, Japan
| | - Tomoyuki Kamenaga
- Department of Orthopaedic Surgery, Kobe University Graduate School of Medicine, 7-5-1, Kusunoki-cho, Chuo-ku, Kobe, 650-0017, Japan
| | - Yoshinori Takashima
- Department of Orthopaedic Surgery, Kobe University Graduate School of Medicine, 7-5-1, Kusunoki-cho, Chuo-ku, Kobe, 650-0017, Japan
| | - Kenichi Kikuchi
- Department of Orthopaedic Surgery, Kobe University Graduate School of Medicine, 7-5-1, Kusunoki-cho, Chuo-ku, Kobe, 650-0017, Japan
| | - Kenmei Ikuta
- Department of Orthopaedic Surgery, Kobe University Graduate School of Medicine, 7-5-1, Kusunoki-cho, Chuo-ku, Kobe, 650-0017, Japan
| | - Yuma Onoi
- Department of Orthopaedic Surgery, Kobe University Graduate School of Medicine, 7-5-1, Kusunoki-cho, Chuo-ku, Kobe, 650-0017, Japan
| | - Shotaro Tachibana
- Department of Orthopaedic Surgery, Kobe University Graduate School of Medicine, 7-5-1, Kusunoki-cho, Chuo-ku, Kobe, 650-0017, Japan
| | - Yoshihito Suda
- Department of Orthopaedic Surgery, Kobe University Graduate School of Medicine, 7-5-1, Kusunoki-cho, Chuo-ku, Kobe, 650-0017, Japan
| | - Kensuke Wada
- Department of Orthopaedic Surgery, Kobe University Graduate School of Medicine, 7-5-1, Kusunoki-cho, Chuo-ku, Kobe, 650-0017, Japan
| | - Takehiko Matsushita
- Department of Orthopaedic Surgery, Kobe University Graduate School of Medicine, 7-5-1, Kusunoki-cho, Chuo-ku, Kobe, 650-0017, Japan
| | - Ryosuke Kuroda
- Department of Orthopaedic Surgery, Kobe University Graduate School of Medicine, 7-5-1, Kusunoki-cho, Chuo-ku, Kobe, 650-0017, Japan
| |
Collapse
|
14
|
Ferrero S, Louvois M, Barnetche T, Breuil V, Roux C. Impact of anterior cruciate ligament surgery on the development of knee osteoarthritis: A systematic literature review and meta-analysis comparing non-surgical and surgical treatments. OSTEOARTHRITIS AND CARTILAGE OPEN 2023; 5:100366. [PMID: 37252633 PMCID: PMC10209532 DOI: 10.1016/j.ocarto.2023.100366] [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: 11/18/2022] [Accepted: 04/24/2023] [Indexed: 05/31/2023] Open
Abstract
Introduction Context: The development of knee osteoarthritis (OA) after anterior cruciate ligament (ACL) injury is now widely recognized. The impact of surgical or non-surgical management on the development of post-traumatic osteoarthritis is still debated in the medical community.Here, we present a meta-analysis comparing the impact of surgical or non-surgical management of ACL injuries on the development of knee OA. Method A systematic literature review was conducted using data from the PubMed, EMBASE, Medline, and Cochrane libraries from February to May 2019. Only randomized clinical trials published between 2005 and 2019 with a non-surgical group and a surgical group were included to explore the onset or progression of knee OA after ACL injury. Trials had to have at least one radiographic endpoint (Kellgren-Lawrence scoring system). Heterogeneity was assessed using the Cochrane's Q and I2 statistical methods. Results Only three randomized controlled trials met the inclusion criteria and were selected for meta-analysis. Of the 343 injured knees included in the studies, 180 underwent ACL reconstruction and 163 underwent non-surgical treatment. The relative risk of knee osteoarthritis was higher after surgery than after non-surgical treatment (RR 1.72, CI 95% [1.18-2.53], I2 = 0%). Conclusion The results of this meta-analysis suggest a predisposition to knee osteoarthritis after ACL reconstruction surgery compared with non-surgical management. Due to the small number of good quality studies available, further well-conducted randomised studies are needed to confirm these findings.
Collapse
Affiliation(s)
- Stephanie Ferrero
- Department of Rheumatology, Pasteur Hospital, Nice University Hospital, Nice Sophia Antipolis University, F-06000, Nice, France
| | - Marion Louvois
- Department of Rheumatology, Pasteur Hospital, Nice University Hospital, Nice Sophia Antipolis University, F-06000, Nice, France
| | - Thomas Barnetche
- Department of Rheumatology, University Hospital of Bordeaux Pellegrin, France
| | - Veronique Breuil
- Department of Rheumatology, Pasteur Hospital, Nice University Hospital, Nice Sophia Antipolis University, F-06000, Nice, France
| | - Christian Roux
- Department Rheumatology, University of Cote D'Azur, Nice Hospital, Laboratory LAMHESS, EA6312, IBV CNRS IMR 7277 INSERM U1091 UNS, France
| |
Collapse
|
15
|
Li X, Li X, Yang J, Du Y, Chen L, Zhao G, Ye T, Zhu Y, Xu X, Deng L, Cui W. In Situ Sustained Macrophage-Targeted Nanomicelle-Hydrogel Microspheres for Inhibiting Osteoarthritis. RESEARCH (WASHINGTON, D.C.) 2023; 6:0131. [PMID: 37223475 PMCID: PMC10202383 DOI: 10.34133/research.0131] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/12/2023] [Accepted: 04/10/2023] [Indexed: 05/25/2023]
Abstract
There are still challenges in applying drug nanocarriers for in situ sustained macrophage targeting and regulation, due to the rapid clearance of nanocarriers and burst drug release in vivo. Herein, a nanomicelle-hydrogel microsphere, characterized by its macrophage-targeted nanosized secondary structure that allows it to accurately bind to M1 macrophages through active endocytosis, is employed for in situ sustained macrophage targeting and regulation, and addresses the insufficient osteoarthritis therapeutic efficacy caused by rapid clearance of drug nanocarriers. The 3-dimensional structure of a microsphere can prevent the rapid escape and clearance of a nanomicelle, thus keeping it in joints, while the ligand-guided secondary structure can carry drugs to accurately target and enter M1 macrophages, and release drugs via the transition from hydrophobicity to hydrophilicity of nanomicelles under inflammatory stimulation inside the macrophages. The experiments show that the nanomicelle-hydrogel microsphere can in situ sustainably target and regulate M1 macrophages for more than 14 days in joints, and attenuate local "cytokine storm" by continuous M1 macrophage apoptosis promotion and polarization inhibition. This micro/nano-hydrogel system shows excellent ability to sustainably target and regulate macrophage, realizes the improvement of drug utilization and efficacy inside the macrophage, and thereby can be a potential platform for treating macrophage-related diseases.
Collapse
Affiliation(s)
| | | | | | - Yawei Du
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai 200025, P. R. China
| | - Liang Chen
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai 200025, P. R. China
| | - Gang Zhao
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai 200025, P. R. China
| | - Tingjun Ye
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai 200025, P. R. China
| | - Yuan Zhu
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai 200025, P. R. China
| | - Xiangyang Xu
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai 200025, P. R. China
| | - Lianfu Deng
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai 200025, P. R. China
| | - Wenguo Cui
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai 200025, P. R. China
| |
Collapse
|
16
|
You B, Zhou C, Yang Y. MSC-EVs alleviate osteoarthritis by regulating microenvironmental cells in the articular cavity and maintaining cartilage matrix homeostasis. Ageing Res Rev 2023; 85:101864. [PMID: 36707035 DOI: 10.1016/j.arr.2023.101864] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 01/19/2023] [Accepted: 01/23/2023] [Indexed: 01/26/2023]
Abstract
Osteoarthritis (OA), a common cause of chronic articular cartilage degeneration, is the main cause of disability in older adults and severely affects quality of life. Multiple factors are involved in the pathogenesis of OA, resulting in imbalance in the homeostasis of the joint cavity microenvironment, which exacerbates the disease. Because of the deficiency of blood vessels and nerves in cartilage, existing therapies to promote cartilage healing are relatively ineffective. Mesenchymal stem cell (MSC)-related therapies have achieved positive outcomes for the treatment of OA, and these beneficial effects have been confirmed to be largely mediated by extracellular vesicles (EVs). MSC-derived EVs (MSC-EVs) have been demonstrated to participate in the regulation of chondrocyte function, to have anti-inflammatory and immunomodulatory effects, and to alleviate metabolic disorders of the extracellular matrix, thereby slowing the progression of OA. In addition, engineered MSC-EVs can enrich therapeutic molecules and optimize administration to enhance their therapeutic effects on OA. A thorough understanding of the endogenous properties of EVs and related engineering strategies could help researchers develop more precise control therapy for OA.
Collapse
Affiliation(s)
- Benshuai You
- Clinical Laboratory Center, The Affiliated Taizhou People's Hospital of Nanjing Medical University, Taizhou 225300, Jiangsu, China
| | - Chenglin Zhou
- Clinical Laboratory Center, The Affiliated Taizhou People's Hospital of Nanjing Medical University, Taizhou 225300, Jiangsu, China.
| | - Yang Yang
- Clinical Laboratory Center, The Affiliated Taizhou People's Hospital of Nanjing Medical University, Taizhou 225300, Jiangsu, China.
| |
Collapse
|
17
|
Salman LA, Ahmed G, Dakin SG, Kendrick B, Price A. Osteoarthritis: a narrative review of molecular approaches to disease management. Arthritis Res Ther 2023; 25:27. [PMID: 36800974 PMCID: PMC9938549 DOI: 10.1186/s13075-023-03006-w] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Accepted: 02/04/2023] [Indexed: 02/19/2023] Open
Abstract
Osteoarthritis (OA) is a chronic, progressive degenerative whole joint disease that affects the articular cartilage, subchondral bone, ligaments, capsule, and synovium. While it is still believed to be a mechanically driven disease, the role of underlying co-existing inflammatory processes and mediators in the onset of OA and its progression is now more appreciated. Post-traumatic osteoarthritis (PTOA) is a subtype of OA that occurs secondary to traumatic joint insults and is widely used in pre-clinical models to help understand OA in general. There is an urgent need to develop new treatments as the global burden is considerable and expanding. In this review, we focus on the recent pharmacological advances in the treatment of OA and summarize the most significant promising agents based on their molecular effects. Those are classified here into broad categories: anti-inflammatory, modulation of the activity of matrix metalloproteases, anabolic, and unconventional pleiotropic agents. We provide a comprehensive analysis of the pharmacological advances in each of these areas and highlight future insights and directions in the OA field.
Collapse
Affiliation(s)
- Loay A Salman
- Present Address: Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, Botnar Research Centre, University of Oxford, Windmill Road, Oxford, OX3 7LD, UK. .,Orthopedics Department, Hamad General Hospital, Hamad Medical Corporation, PO Box 3050, Doha, Qatar.
| | - Ghalib Ahmed
- Orthopedics Department, Hamad General Hospital, Hamad Medical Corporation, PO Box 3050, Doha, Qatar
| | - Stephanie G Dakin
- Present Address: Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, Botnar Research Centre, University of Oxford, Windmill Road, Oxford, OX3 7LD, UK
| | - Benjamin Kendrick
- Present Address: Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, Botnar Research Centre, University of Oxford, Windmill Road, Oxford, OX3 7LD, UK
| | - Andrew Price
- Present Address: Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, Botnar Research Centre, University of Oxford, Windmill Road, Oxford, OX3 7LD, UK
| |
Collapse
|
18
|
Use of Brain-Derived Stem/Progenitor Cells and Derived Extracellular Vesicles to Repair Damaged Neural Tissues: Lessons Learned from Connective Tissue Repair Regarding Variables Limiting Progress and Approaches to Overcome Limitations. Int J Mol Sci 2023; 24:ijms24043370. [PMID: 36834779 PMCID: PMC9958575 DOI: 10.3390/ijms24043370] [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: 12/28/2022] [Revised: 01/27/2023] [Accepted: 02/06/2023] [Indexed: 02/11/2023] Open
Abstract
Pluripotent neural stem or progenitor cells (NSC/NPC) have been reported in the brains of adult preclinical models for decades, as have mesenchymal stem/stromal cells (MSC) been reported in a variety of tissues from adults. Based on their in vitro capabilities, these cell types have been used extensively in attempts to repair/regenerate brain and connective tissues, respectively. In addition, MSC have also been used in attempts to repair compromised brain centres. However, success in treating chronic neural degenerative conditions such as Alzheimer's disease, Parkinson's disease, and others with NSC/NPC has been limited, as have the use of MSC in the treatment of chronic osteoarthritis, a condition affecting millions of individuals. However, connective tissues are likely less complex than neural tissues regarding cell organization and regulatory integration, but some insights have been gleaned from the studies regarding connective tissue healing with MSC that may inform studies attempting to initiate repair and regeneration of neural tissues compromised acutely or chronically by trauma or disease. This review will discuss the similarities and differences in the applications of NSC/NPC and MSC, where some lessons have been learned, and potential approaches that could be used going forward to enhance progress in the application of cellular therapy to facilitate repair and regeneration of complex structures in the brain. In particular, variables that may need to be controlled to enhance success are discussed, as are different approaches such as the use of extracellular vesicles from stem/progenitor cells that could be used to stimulate endogenous cells to repair the tissues rather than consider cell replacement as the primary option. Caveats to all these efforts relate to whether cellular repair initiatives will have long-term success if the initiators for neural diseases are not controlled, and whether such cellular initiatives will have long-term success in a subset of patients if the neural diseases are heterogeneous and have multiple etiologies.
Collapse
|
19
|
Dilley JE, Bello MA, Roman N, McKinley T, Sankar U. Post-traumatic osteoarthritis: A review of pathogenic mechanisms and novel targets for mitigation. Bone Rep 2023. [DOI: 10.1016/j.bonr.2023.101658] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
|
20
|
Yang B, Li X, Fu C, Cai W, Meng B, Qu Y, Kou X, Zhang Q. Extracellular vesicles in osteoarthritis of peripheral joint and temporomandibular joint. Front Endocrinol (Lausanne) 2023; 14:1158744. [PMID: 36950682 PMCID: PMC10025484 DOI: 10.3389/fendo.2023.1158744] [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: 02/04/2023] [Accepted: 02/21/2023] [Indexed: 03/08/2023] Open
Abstract
Osteoarthritis (OA) is a disabling disease with significant morbidity worldwide. OA attacks the large synovial joint, including the peripheral joints and temporomandibular joint (TMJ). As a representative of peripheral joint OA, knee OA shares similar symptoms with TMJ OA. However, these two joints also display differences based on their distinct development, anatomy, and physiology. Extracellular vesicles (EVs) are phospholipid bilayer nanoparticles, including exosomes, microvesicles, and apoptotic bodies. EVs contain proteins, lipids, DNA, micro-RNA, and mRNA that regulate tissue homeostasis and cell-to-cell communication, which play an essential role in the progression and treatment of OA. They are likely to partake in mechanical response, extracellular matrix degradation, and inflammatory regulation during OA. More evidence has shown that synovial fluid and synovium-derived EVs may serve as OA biomarkers. More importantly, mesenchymal stem cell-derived EV shows a therapeutic effect on OA. However, the different function of EVs in these two joints is largely unknown based on their distinct biological characteristic. Here, we reviewed the effects of EVs in OA progression and compared the difference between the knee joint and TMJ, and summarized their potential therapeutic role in the treatment of OA.
Collapse
Affiliation(s)
- Benyi Yang
- Guangdong Provincial Key Laboratory of Stomatology Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, South China Center of Craniofacial Stem Cell Research, Guangzhou, China
| | - Xin Li
- Department of Temporomandibular Joint, Affiliated Stomatology Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, China
- Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Guangdong Engineering Research Center of Oral Restoration and Reconstruction, Guangzhou, China
| | - Chaoran Fu
- Guangdong Provincial Key Laboratory of Stomatology Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, South China Center of Craniofacial Stem Cell Research, Guangzhou, China
| | - Wenyi Cai
- Department of Temporomandibular Joint, Affiliated Stomatology Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, China
- Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Guangdong Engineering Research Center of Oral Restoration and Reconstruction, Guangzhou, China
| | - Bowen Meng
- Guangdong Provincial Key Laboratory of Stomatology Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, South China Center of Craniofacial Stem Cell Research, Guangzhou, China
| | - Yan Qu
- Guangdong Provincial Key Laboratory of Stomatology Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, South China Center of Craniofacial Stem Cell Research, Guangzhou, China
| | - Xiaoxing Kou
- Guangdong Provincial Key Laboratory of Stomatology Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, South China Center of Craniofacial Stem Cell Research, Guangzhou, China
- *Correspondence: Qingbin Zhang, ; Xiaoxing Kou,
| | - Qingbin Zhang
- Department of Temporomandibular Joint, Affiliated Stomatology Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, China
- Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Guangdong Engineering Research Center of Oral Restoration and Reconstruction, Guangzhou, China
- *Correspondence: Qingbin Zhang, ; Xiaoxing Kou,
| |
Collapse
|
21
|
Hart DA. Osteoarthritis as an Umbrella Term for Different Subsets of Humans Undergoing Joint Degeneration: The Need to Address the Differences to Develop Effective Conservative Treatments and Prevention Strategies. Int J Mol Sci 2022; 23:ijms232315365. [PMID: 36499704 PMCID: PMC9736942 DOI: 10.3390/ijms232315365] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Revised: 11/30/2022] [Accepted: 12/04/2022] [Indexed: 12/12/2022] Open
Abstract
Osteoarthritis (OA) of joints such as the knee and hip are very prevalent, and the number of individuals affected is expected to continue to rise. Currently, conservative treatments after OA diagnosis consist of a series of increasingly invasive interventions as the degeneration and pain increase, leading very often to joint replacement surgery. Most interventions are focused on alleviating pain, and there are no interventions currently available that stop and reverse OA-associated joint damage. For many decades OA was considered a disease of cartilage, but it is now considered a disease of the whole multi-tissue joint. As pain is the usual presenting symptom, for most patients, it is not known when the disease process was initiated and what the basis was for the initiation. The exception is post-traumatic OA which results from an overt injury to the joint that elevates the risk for OA development. This scenario leads to very long wait lists for joint replacement surgery in many jurisdictions. One aspect of why progress has been so slow in addressing the needs of patients is that OA has been used as an umbrella term that does not recognize that joint degeneration may arise from a variety of mechanistic causes that likely need separate analysis to identify interventions unique to each subtype (post-traumatic, metabolic, post-menopausal, growth and maturation associated). A second aspect of the slow pace of progress is that the bulk of research in the area is focused on post-traumatic OA (PTOA) in preclinical models that likely are not clearly relevant to human OA. That is, only ~12% of human OA is due to PTOA, but the bulk of studies investigate PTOA in rodents. Thus, much of the research community is failing the patient population affected by OA. A third aspect is that conservative treatment platforms are not specific to each OA subset, nor are they integrated into a coherent fashion for most patients. This review will discuss the literature relevant to the issues mentioned above and propose some of the directions that will be required going forward to enhance the impact of the research enterprise to affect patient outcomes.
Collapse
Affiliation(s)
- David A Hart
- Department of Surgery, Faculty of Kinesiology, McCaig Institute for Bone & Joint Health, University of Calgary, Calgary, AB T2N 4N1, Canada
| |
Collapse
|
22
|
Yin H, Li M, Tian G, Ma Y, Ning C, Yan Z, Wu J, Ge Q, Sui X, Liu S, Zheng J, Guo W, Guo Q. The role of extracellular vesicles in osteoarthritis treatment via microenvironment regulation. Biomater Res 2022; 26:52. [PMID: 36199125 PMCID: PMC9532820 DOI: 10.1186/s40824-022-00300-7] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2022] [Accepted: 09/18/2022] [Indexed: 11/10/2022] Open
Abstract
Osteoarthritis (OA) is a degenerative joint disease that is common among the middle-aged and older populations, causes patients to experience recurrent pain in their joints and negatively affects their quality of life. Currently, therapeutic options for patients with OA consist of medications to alleviate pain and treat the symptoms; however, due to typically poor outcomes, patients with advanced OA are unlikely to avoid joint replacement. In recent years, several studies have linked disrupted homeostasis of the joint cavity microenvironment to the development of OA. Recently, extracellular vesicles (EVs) have received increasing attention in the field of OA. EVs are natural nano-microcarrier materials with unique biological activity that are produced by cells through paracrine action. They are composed of lipid bilayers that contain physiologically active molecules, such as nucleic acids and proteins. Moreover, EVs may participate in local and distal intercellular and intracellular communication. EVs have also recently been shown to influence OA development by regulating biochemical factors in the OA microenvironmental. In this article, we first describe the microenvironment of OA. Then, we provide an overview of EVs, summarize the main types used for the treatment of OA, and describe their mechanisms. Next, we review clinical studies using EVs for OA treatment. Finally, the specific mechanism underlying the application of miRNA-enriched EVs in OA therapy is described.
Collapse
Affiliation(s)
- Han Yin
- Institute of Orthopedics, The First Medical Center, Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma and War Injuries PLA, No. 28 Fuxing Road, Haidian District, Beijing, 100853, PR China
| | - Muzhe Li
- Institute of Orthopedics, The First Medical Center, Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma and War Injuries PLA, No. 28 Fuxing Road, Haidian District, Beijing, 100853, PR China
- Department of Orthopedics, The First Affiliated Hospital of University of South China, Hengyang, 421000, China
| | - Guangzhao Tian
- Institute of Orthopedics, The First Medical Center, Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma and War Injuries PLA, No. 28 Fuxing Road, Haidian District, Beijing, 100853, PR China
- School of Medicine, Nankai University, Tianjin, 300071, China
| | - Yang Ma
- Institute of Orthopedics, The First Medical Center, Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma and War Injuries PLA, No. 28 Fuxing Road, Haidian District, Beijing, 100853, PR China
| | - Chao Ning
- Institute of Orthopedics, The First Medical Center, Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma and War Injuries PLA, No. 28 Fuxing Road, Haidian District, Beijing, 100853, PR China
| | - Zineng Yan
- Institute of Orthopedics, The First Medical Center, Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma and War Injuries PLA, No. 28 Fuxing Road, Haidian District, Beijing, 100853, PR China
| | - Jiang Wu
- Institute of Orthopedics, The First Medical Center, Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma and War Injuries PLA, No. 28 Fuxing Road, Haidian District, Beijing, 100853, PR China
| | - Qian Ge
- Huaiyin People's Hospital of Huai'an, Huai'an, 223001, China
| | - Xiang Sui
- Institute of Orthopedics, The First Medical Center, Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma and War Injuries PLA, No. 28 Fuxing Road, Haidian District, Beijing, 100853, PR China
| | - Shuyun Liu
- Institute of Orthopedics, The First Medical Center, Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma and War Injuries PLA, No. 28 Fuxing Road, Haidian District, Beijing, 100853, PR China.
| | - Jinxuan Zheng
- Department of Orthodontics, Guanghua School of Stomatology, Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-Sen University, No.56 Linyuan Xi Road, Yuexiu District, Guangzhou, Guangdong, 510055, People's Republic of China.
| | - Weimin Guo
- Department of Orthopaedic Surgery, Guangdong Provincial Key Laboratory of Orthopedics and Traumatology, First Affiliated Hospital, Sun Yat-Sen University, No.58 Zhongshan Second Road, Yuexiu District, Guangzhou, 510080, Guangdong, China.
| | - Quanyi Guo
- Institute of Orthopedics, The First Medical Center, Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma and War Injuries PLA, No. 28 Fuxing Road, Haidian District, Beijing, 100853, PR China.
| |
Collapse
|
23
|
Zhuang Y, Jiang S, Yuan C, Lin K. The potential therapeutic role of extracellular vesicles in osteoarthritis. Front Bioeng Biotechnol 2022; 10:1022368. [PMID: 36185451 PMCID: PMC9523151 DOI: 10.3389/fbioe.2022.1022368] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2022] [Accepted: 09/01/2022] [Indexed: 11/13/2022] Open
Abstract
Osteoarthritis (OA) is a worldwide and disabling disease, which cause severe pain and heavy socioeconomic burden. However, pharmacologic or surgical therapies cannot mitigate OA progression. Mesenchymal stem cells (MSCs) therapy has emerged as potential approach for OA treatment, while the immunogenicity and ethical audit of cell therapy are unavoidable. Compared with stem cell strategy, EVs induce less immunological rejection, and they are more stable for storage and in vivo application. MSC-EVs-based therapy possesses great potential in regulating inflammation and promoting cartilage matrix reconstruction in OA treatment. To enhance the therapeutic effect, delivery efficiency, tissue specificity and safety, EVs can be engineered via different modification strategies. Here, the application of MSC-EVs in OA treatment and the potential underlying mechanism were summarized. Moreover, EV modification strategies including indirect MSC modification and direct EV modification were reviewed.
Collapse
Affiliation(s)
- Yu Zhuang
- Department of Oral and Cranio-Maxillofacial Surgery, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- College of Stomatology, Shanghai Jiao Tong University, Shanghai, China
- National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Research Unit of Oral and Maxillofacial Regenerative Medicine, Chinese Academy of Medical Sciences, Shanghai, China
| | - Shengjie Jiang
- Department of Oral and Cranio-Maxillofacial Surgery, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- College of Stomatology, Shanghai Jiao Tong University, Shanghai, China
- National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Research Unit of Oral and Maxillofacial Regenerative Medicine, Chinese Academy of Medical Sciences, Shanghai, China
| | - Changyong Yuan
- School of Stomatology, Xuzhou Medical University, Shanghai, China
- Department of Dental Implant, The Affiliated Stomatological Hospital of Xuzhou Medical University, Shanghai, China
- Shanghai Key Laboratory of Stomatology, Department of Oral and Cranio-Maxillofacial Surgery, Shanghai Ninth People’s Hospital, Shanghai Research Institute of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- *Correspondence: Changyong Yuan, ; Kaili Lin,
| | - Kaili Lin
- Department of Oral and Cranio-Maxillofacial Surgery, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- College of Stomatology, Shanghai Jiao Tong University, Shanghai, China
- National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Research Unit of Oral and Maxillofacial Regenerative Medicine, Chinese Academy of Medical Sciences, Shanghai, China
- *Correspondence: Changyong Yuan, ; Kaili Lin,
| |
Collapse
|
24
|
Hart DA, Nakamura N. Creating an Optimal In Vivo Environment to Enhance Outcomes Using Cell Therapy to Repair/Regenerate Injured Tissues of the Musculoskeletal System. Biomedicines 2022; 10:1570. [PMID: 35884875 PMCID: PMC9313221 DOI: 10.3390/biomedicines10071570] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Revised: 06/20/2022] [Accepted: 06/27/2022] [Indexed: 11/16/2022] Open
Abstract
Following most injuries to a musculoskeletal tissue which function in unique mechanical environments, an inflammatory response occurs to facilitate endogenous repair. This is a process that usually yields functionally inferior scar tissue. In the case of such injuries occurring in adults, the injury environment no longer expresses the anabolic processes that contributed to growth and maturation. An injury can also contribute to the development of a degenerative process, such as osteoarthritis. Over the past several years, researchers have attempted to use cellular therapies to enhance the repair and regeneration of injured tissues, including Platelet-rich Plasma and mesenchymal stem/medicinal signaling cells (MSC) from a variety of tissue sources, either as free MSC or incorporated into tissue engineered constructs, to facilitate regeneration of such damaged tissues. The use of free MSC can sometimes affect pain symptoms associated with conditions such as OA, but regeneration of damaged tissues has been challenging, particularly as some of these tissues have very complex structures. Therefore, implanting MSC or engineered constructs into an inflammatory environment in an adult may compromise the potential of the cells to facilitate regeneration, and neutralizing the inflammatory environment and enhancing the anabolic environment may be required for MSC-based interventions to fulfill their potential. Thus, success may depend on first eliminating negative influences (e.g., inflammation) in an environment, and secondly, implanting optimally cultured MSC or tissue engineered constructs into an anabolic environment to achieve the best outcomes. Furthermore, such interventions should be considered early rather than later on in a disease process, at a time when sufficient endogenous cells remain to serve as a template for repair and regeneration. This review discusses how the interface between inflammation and cell-based regeneration of damaged tissues may be at odds, and outlines approaches to improve outcomes. In addition, other variables that could contribute to the success of cell therapies are discussed. Thus, there may be a need to adopt a Precision Medicine approach to optimize tissue repair and regeneration following injury to these important tissues.
Collapse
Affiliation(s)
- David A. Hart
- Department of Surgery, Faculty of Kinesiology, McCaig Institute for Bone & Joint Health, University of Calgary, Calgary, AB T2N 4N1, Canada
- Bone & Joint Health Strategic Clinical Network, Alberta Health Services, Edmonton, AB T5J 3E4, Canada
| | - Norimasa Nakamura
- Institute of Medical Science in Sport, Osaka Health Science University, 1-9-27 Tenma, Kita-ku, Osaka 530-0043, Japan;
| |
Collapse
|
25
|
Bosser C, Auregan JC, Bensidhoum M, Hoc T, Bégué T, Vialle R, Nouguier-Lehon C, Bachy M. Early impairment of cartilage poroelastic properties in an animal model of ACL tear. Orthop Traumatol Surg Res 2022; 108:103116. [PMID: 34666200 DOI: 10.1016/j.otsr.2021.103116] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/26/2021] [Revised: 04/16/2021] [Accepted: 04/23/2021] [Indexed: 02/03/2023]
Abstract
BACKGROUND In more than 50% of cases, anterior cruciate ligament (ACL) lesions lead to post-traumatic osteoarthritis. Ligament reconstruction stabilizes the joint, but the tear seems to impair the poroelasticity of the cartilage: synovial membrane fluid inflammation is observed 3 weeks after tearing. There have been some descriptions of visible cartilage changes, but poroelasticity has never been analyzed at this early stage. The present animal study aimed to determine (1) whether cartilage showed early poroelastic deterioration after ACL tear; (2) whether an impairment correlated with macroscopic changes; and (3) whether cartilage poroelasticity deteriorated over time. HYPOTHESIS In the days following trauma, cartilage poroelasticity is greatly impaired, without macroscopically visible change. MATERIAL AND METHODS ACL tear was surgically induced in 18 New-Zealand rabbits. Cartilage poroelasticity was assessed on indentation-relaxation test in 3 groups: "early", at 2 weeks postoperatively (n=6), "mid-early" at 6 weeks (n=6) and in a non-operated control group ("non-op"). Macroscopic changes were scored in the same groups. RESULTS Poroelastic impairment was greatest at the early time-point (2 weeks). Permeability ranged from a mean 0.08±0.05×10-15 m4/Ns (range, 0.028-0.17) in the "non-op" group to 1.03±0.60×10-15 m4/Ns (range, 0.24-2.15) in the "early" group (p=0.007). Shear modulus ranged from 0.53±0.11MPa (range, 0.36-0.66) to 0.23±0.10MPa (range, 0.12-0.43), respectively (p=0.013). Macroscopic deterioration, on the other hand, differed significantly only between the "mid-early" and the "non-op" groups: p=0.011 for cartilage deterioration and p=0.008 for osteophyte formation. At the "mid-early" time point, poroelastic deterioration was less marked, with 0.33±0.33×10-15 m4/Ns permeability (range, 0.06-1.06) and shear modulus 0.30±0.10MPa (range, 0.13-0.41: respectively p=0.039 and p=0.023 compared to the "non-op" group. DISCUSSION The severe rapid deterioration in poroelasticity following ACL tear in an animal model, as notably seen in increased permeability, corresponds to changes in cartilage microstructure, with easier outflows of interstitial fluid. This mechanical degradation may underlie onset of microcracks within the cartilage, leading to physiological loading that the cartilage by its nature is unable to repair. Further investigations are needed to correlate these experimental data with clinical findings. LEVEL OF EVIDENCE III; comparative study with control group.
Collapse
Affiliation(s)
- Cartherine Bosser
- HealthDataSciences, 45, chemin du Barthélémy, 69260 Charbonnières-les-Bains, France
| | - Jean Charles Auregan
- Université de Paris, B3OA, UMR CNRS 7052, Inserm U1271, 10, avenue de Verdun, 75010 Paris, France; Service d'orthopédie, université Paris-Saclay, AP-HP, Antoine Béclère, 157, rue de la Porte-de-Trivaux, 92140 Clamart, France
| | - Morad Bensidhoum
- Université de Paris, B3OA, UMR CNRS 7052, Inserm U1271, 10, avenue de Verdun, 75010 Paris, France
| | - Thierry Hoc
- Université de Paris, B3OA, UMR CNRS 7052, Inserm U1271, 10, avenue de Verdun, 75010 Paris, France; École centrale de Lyon, département de mécanique, MSGMGC, 36, avenue Guy-de-Collongue, 69134 Ecully cedex, France
| | - Thierry Bégué
- Service d'orthopédie, université Paris-Saclay, AP-HP, Antoine Béclère, 157, rue de la Porte-de-Trivaux, 92140 Clamart, France
| | - Raphaël Vialle
- Service de chirurgie orthopédique et réparatrice de l'enfant, AP-HP-Sorbonne université, hôpital Trousseau, 26, avenue du Docteur-Arnold-Netter, 75012 Paris, France
| | - Cécile Nouguier-Lehon
- Université de Lyon, LTDS UMR CNRS 5513, école centrale de Lyon, 36, avenue Guy-de-Collongue, 69134 Ecully cedex, France
| | - Manon Bachy
- Université de Paris, B3OA, UMR CNRS 7052, Inserm U1271, 10, avenue de Verdun, 75010 Paris, France; Service de chirurgie orthopédique et réparatrice de l'enfant, AP-HP-Sorbonne université, hôpital Trousseau, 26, avenue du Docteur-Arnold-Netter, 75012 Paris, France.
| |
Collapse
|
26
|
Heard BJ, Barton KI, Abubacker S, Chung M, Martin CR, Schmidt TA, Shrive NG, Hart DA. Synovial and cartilage responsiveness to peri-operative hyaluronic acid ± dexamethasone administration following a limited injury to the rabbit stifle joint. J Orthop Res 2022; 40:838-845. [PMID: 34061360 DOI: 10.1002/jor.25108] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Revised: 04/03/2021] [Accepted: 05/25/2021] [Indexed: 02/04/2023]
Abstract
Posttraumatic osteoarthritis (PTOA) can develop after an injury to the knee. Previous studies have indicated that an intra-articular (IA) injection of the potent glucocorticoid dexamethasone (DEX) may significantly prevent induction of PTOA. The aim of the present study was to investigate the effectiveness of a single IA injection of hyaluronic acid (HA), alone and in combination with DEX following a localized intra-articular injury as a PTOA-preventing treatment option. An established rabbit model of surgical injury consisting of dual intra-articular (IA) drill holes in a non-cartilaginous area of the femoral notch near the origin of the anterior cruciate ligament (ACL) to allow for bleeding into the joint space was used. Immediately following surgery, subjects were treated with HA, HA + DEX, or received no treatment. An uninjured control group was used for comparison (N = 5/group). Rabbits were sacrificed and investigated at 9 weeks post-injury. At 9 weeks post-injury, there was a significant protective capacity of the single IA treatment of DEX + HA on the histological grade of the synovial tissue, and some variable location-specific effects of HA alone and HA + DEX interactions on cartilage damage. Thus, it is possible that co-treatment with HA may interfere with the effectiveness of the DEX. In vitro friction testing indicated that DEX did not interfere with the lubricating ability of HA or synovial fluid on cartilage. These results suggest that a single IA administration of HA in combination with DEX following an IA injury is not recommended for inhibition of PTOA progression in this model.
Collapse
Affiliation(s)
- B J Heard
- McCaig Institute for Bone and Joint Health, University of Calgary, Calgary, AB, Canada.,Department of Surgery, University of Calgary, Calgary, Alberta, Canada
| | - K I Barton
- McCaig Institute for Bone and Joint Health, University of Calgary, Calgary, AB, Canada.,Faculty of Kinesiology, University of Calgary, Calgary, Alberta, Canada
| | - S Abubacker
- McCaig Institute for Bone and Joint Health, University of Calgary, Calgary, AB, Canada.,Faculty of Kinesiology, University of Calgary, Calgary, Alberta, Canada
| | - M Chung
- McCaig Institute for Bone and Joint Health, University of Calgary, Calgary, AB, Canada
| | - C R Martin
- McCaig Institute for Bone and Joint Health, University of Calgary, Calgary, AB, Canada.,Department of Surgery, University of Calgary, Calgary, Alberta, Canada
| | - T A Schmidt
- McCaig Institute for Bone and Joint Health, University of Calgary, Calgary, AB, Canada.,Faculty of Kinesiology, University of Calgary, Calgary, Alberta, Canada.,Sculich School of Engineering, University of Calgary, Calgary, Alberta, Canada
| | - N G Shrive
- McCaig Institute for Bone and Joint Health, University of Calgary, Calgary, AB, Canada.,Sculich School of Engineering, University of Calgary, Calgary, Alberta, Canada
| | - D A Hart
- McCaig Institute for Bone and Joint Health, University of Calgary, Calgary, AB, Canada.,Department of Surgery, University of Calgary, Calgary, Alberta, Canada.,Faculty of Kinesiology, University of Calgary, Calgary, Alberta, Canada.,Bone & Joint Health Strategic Clinical Network, Edmonton, Alberta, Canada
| |
Collapse
|
27
|
Veloso C, Videira RA, Andrade PB, Cardoso C, Vitorino C. In vivo methodologies to assist preclinical development of topical fixed-dose combinations for pain management. Int J Pharm 2022; 616:121530. [PMID: 35121043 DOI: 10.1016/j.ijpharm.2022.121530] [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: 11/09/2021] [Revised: 01/18/2022] [Accepted: 01/27/2022] [Indexed: 11/24/2022]
Abstract
The combination in a fixed dose of two or more active pharmaceutical ingredients in the same pharmaceutical dosage form is an approach that has been used successfully in the treatment of several pathologies, including pain. In the preclinical development of a topical fixed-dose combination product with analgesic and anti-inflammatory activities for pain management, the main objective is to establish the nature of the interaction between the different active pharmaceutical ingredients while obtaining data on the medicinal product safety and efficacy. Despite the improvement of in vitro assays, animal models remain a fundamental strategy to characterise the interaction, efficacy and safety of active pharmaceutical ingredients at the physiological level, which cannot be reached by in vitro assays. Thus, the main goal of this review is to systematise the available animal models to evaluate the efficacy and safety of a new fixed-dose combination product for topical administration indicated for pain management. Particular emphasis is given to animal models that are accepted for regulatory purposes.
Collapse
Affiliation(s)
- Cláudia Veloso
- Faculty of Pharmacy, University of Coimbra, Pólo das Ciências da Saúde, Azinhaga de Santa Comba, 3000-548 Coimbra, Portugal; Coimbra Chemistry Center, Department of Chemistry, University of Coimbra, Rua Larga, 3004-535 Coimbra, Portugal
| | - Romeu A Videira
- REQUIMTE/LAQV, Laboratório de Farmacognosia, Departamento de Química, Faculdade de Farmácia, Universidade do Porto, R. Jorge Viterbo Ferreira, n° 228, 4050-313 Porto, Portugal.
| | - Paula B Andrade
- REQUIMTE/LAQV, Laboratório de Farmacognosia, Departamento de Química, Faculdade de Farmácia, Universidade do Porto, R. Jorge Viterbo Ferreira, n° 228, 4050-313 Porto, Portugal
| | - Catarina Cardoso
- Laboratórios Basi, Parque Industrial Manuel Lourenço Ferreira, lote 15, 3450-232 Mortágua, Portugal
| | - Carla Vitorino
- Faculty of Pharmacy, University of Coimbra, Pólo das Ciências da Saúde, Azinhaga de Santa Comba, 3000-548 Coimbra, Portugal; Coimbra Chemistry Center, Department of Chemistry, University of Coimbra, Rua Larga, 3004-535 Coimbra, Portugal.
| |
Collapse
|
28
|
Oladeji LO, Stoker AM, Stannard JP, Cook JL. A Hyperosmolar Saline Solution Fortified with Anti-Inflammatory Components Mitigates Articular Cartilage Pro-Inflammatory and Degradative Responses in an In Vitro Model of Knee Arthroscopy. Cartilage 2021; 13:1646S-1653S. [PMID: 33899552 PMCID: PMC8804770 DOI: 10.1177/19476035211011521] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
OBJECTIVE To evaluate differences in pro-inflammatory and degradative mediator production from osteoarthritic knee articular cartilage explants treated with a hyperosmolar saline solution supplemented with anti-inflammatory components (l-glutamine, ascorbic acid, sodium pyruvate, epigallocatechin gallate [EGCG], and dexamethasone) or normal saline using an in vitro model for knee arthroscopy. DESIGN Full-thickness 6 mm articular cartilage explants (n = 12/patient) were created from femoral condyle and tibial plateau samples collected from patients who received knee arthroplasty. One explant half was treated for 3 hours with hyperosmolar saline (600 mOsm/L) supplemented with anti-inflammatory components and the corresponding half with normal saline (308 mOsm/L). Explants were cultured for 3 days and then collected for biomarker analyses. Media biomarker concentrations were normalized to the wet weight of the tissue (mg) and were analyzed by a paired t test with significance set at P < 0.05. RESULTS Cartilage was collected from 9 females and 2 males (mean age = 68 years). Concentrations of MCP-1 (P < 0.001), IL-8 (P = 0.03), GRO-α (P = 0.02), MMP-1 (P < 0.001), MMP-2 (P < 0.001), and MMP-3 (P < 0.001) were significantly lower in explant halves treated with the enhanced hyperosmolar solution. When considering only those cartilage explants in the top tercile of tissue metabolism, IL-6 (P = 0.005), IL-8 (P = 0.0001), MCP-1 (P < 0.001), GRO-α (P = 0.0003), MMP-1 (P < 0.001), MMP-2 (P < 0.001), MMP-3 (P < 0.001), and GAG expression (P = 0.0001) was significantly lower in cartilage explant halves treated with the enhanced hyperosmolar solution. CONCLUSIONS Treatment of cartilage explants with a hyperosmolar saline arthroscopic irrigation solution supplemented with anti-inflammatory components was associated with significant decreases in inflammatory and degradative mediator production and mitigation of proteoglycan loss.
Collapse
Affiliation(s)
- Lasun O Oladeji
- Department of Orthopaedic Surgery, University of Missouri, Columbia, MO, USA.,Thompson Laboratory for Regenerative Orthopaedics, University of Missouri, Columbia, MO, USA
| | - Aaron M Stoker
- Department of Orthopaedic Surgery, University of Missouri, Columbia, MO, USA.,Thompson Laboratory for Regenerative Orthopaedics, University of Missouri, Columbia, MO, USA
| | - James P Stannard
- Department of Orthopaedic Surgery, University of Missouri, Columbia, MO, USA.,Thompson Laboratory for Regenerative Orthopaedics, University of Missouri, Columbia, MO, USA
| | - James L Cook
- Department of Orthopaedic Surgery, University of Missouri, Columbia, MO, USA.,Thompson Laboratory for Regenerative Orthopaedics, University of Missouri, Columbia, MO, USA
| |
Collapse
|
29
|
Arabiyat AS, Chen H, Erndt-Marino J, Burkhard K, Scola L, Fleck A, Wan LQ, Hahn MS. Hyperosmolar Ionic Solutions Modulate Inflammatory Phenotype and sGAG Loss in a Cartilage Explant Model. Cartilage 2021; 13:713S-721S. [PMID: 32975437 PMCID: PMC8804856 DOI: 10.1177/1947603520961167] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
OBJECTIVE The objective of this study was to compare the effects of hyperosmolar sodium (Na+), lithium (Li+) and potassium (K+) on catabolic and inflammatory osteoarthritis (OA) markers and sulfated glycosaminoglycan (sGAG) loss in TNF-α-stimulated cartilage explants. METHODS Explants from bovine stifle joints were stimulated with TNF-α for 1 day to induce cartilage degradation followed by supplementation with 50 mM potassium chloride (KCl), 50 mM lithium chloride (LiCl), 50 mM sodium chloride (NaCl), or 100 nM dexamethasone for an additional 6 days. We assessed the effect of TNF-α stimulation and hyperosmolar ionic treatment on sGAG loss and expression of OA-associated proteins: ADAMTS-5, COX-2, MMP-1, MMP-13, and VEGF. RESULTS TNF-α treatment increased sGAG loss (P < 0.001) and expression of COX-2 (P = 0.018), MMP-13 (P < 0.001), and VEGF (P = 0.017) relative to unstimulated controls. Relative to activated controls, LiCl and dexamethasone treatment attenuated sGAG loss (P = 0.008 and P = 0.042, respectively) and expression of MMP-13 (P = 0.005 and P = 0.036, respectively). In contrast, KCl treatment exacerbated sGAG loss (P = 0.032) and MMP-1 protein expression (P = 0.010). NaCl treatment, however, did not alter sGAG loss or expression of OA-related proteins. Comparing LiCl and KCl treatment shows a potent reduction (P < 0.05) in catabolic and inflammatory mediators following LiCl treatment. CONCLUSION These results suggest that these ionic species elicit varying responses in TNF-α-stimulated explants. Cumulatively, these findings support additional studies of hyperosmolar ionic solutions for potential development of novel intraarticular injections targeting OA.
Collapse
Affiliation(s)
- Ahmad S. Arabiyat
- Department of Biomedical Engineering,
Rensselaer Polytechnic Institute (RPI), Troy, NY, USA
- Center for Biotechnology and
Interdisciplinary Studies, Rensselaer Polytechnic Institute (RPI), Troy, NY,
USA
| | - Hongyu Chen
- Department of Biomedical Engineering,
Rensselaer Polytechnic Institute (RPI), Troy, NY, USA
- Center for Biotechnology and
Interdisciplinary Studies, Rensselaer Polytechnic Institute (RPI), Troy, NY,
USA
| | - Josh Erndt-Marino
- Department of Biomedical Engineering,
Tufts University, Medford, MA, USA
| | - Katie Burkhard
- Department of Biomedical Engineering,
Rensselaer Polytechnic Institute (RPI), Troy, NY, USA
| | - Lisa Scola
- Department of Biomedical Engineering,
Rensselaer Polytechnic Institute (RPI), Troy, NY, USA
| | - Allison Fleck
- Department of Biomedical Engineering,
Rensselaer Polytechnic Institute (RPI), Troy, NY, USA
- Center for Biotechnology and
Interdisciplinary Studies, Rensselaer Polytechnic Institute (RPI), Troy, NY,
USA
| | - Leo Q. Wan
- Department of Biomedical Engineering,
Rensselaer Polytechnic Institute (RPI), Troy, NY, USA
- Center for Biotechnology and
Interdisciplinary Studies, Rensselaer Polytechnic Institute (RPI), Troy, NY,
USA
| | - Mariah S. Hahn
- Department of Biomedical Engineering,
Rensselaer Polytechnic Institute (RPI), Troy, NY, USA
- Center for Biotechnology and
Interdisciplinary Studies, Rensselaer Polytechnic Institute (RPI), Troy, NY,
USA
| |
Collapse
|
30
|
Yaari L, Dolev A, Kittani M, Yassin M, Gbaren M, Feicht E, Haviv B. Preoperative pain sensitivity questionnaire helps customize pain management after arthroscopic partial meniscectomy. Knee Surg Sports Traumatol Arthrosc 2021; 29:4198-4204. [PMID: 33704517 DOI: 10.1007/s00167-021-06438-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/08/2020] [Accepted: 01/07/2021] [Indexed: 11/26/2022]
Abstract
PURPOSE To evaluate correlations between preoperative pain sensitivity and postoperative analgesic consumption together with pain perception shortly after arthroscopic partial meniscectomy in non-arthritic knees. METHODS Ninety-nine patients who underwent primary arthroscopic meniscectomy were prospectively divided into three postoperative treatment groups that were prescribed with betamethasone injection (at the end of surgery), oral celecoxib or rescue analgesia (control). Preoperative pain sensitivity was evaluated by pain sensitivity questionnaires (PSQ). Patients were followed for the first three postoperative weeks to evaluate knee injury and osteoarthritis outcome score (KOOS) pain scores and analgesics consumption. Statistical analysis included correlations among preoperative pain sensitivity, postoperative pain levels and analgesics consumption. A receiver operating characteristic curve was plotted to investigate the cutoff values of the PSQ score to predict insufficient postoperative pain reduction. RESULTS There were no differences at baseline among all study groups in age, sex, BMI, level of activity, comorbidities and surgical findings. At the final follow-up, KOOS pain scores improved in all groups (p < 0.001). Mean final KOOS pain scores were 76.1 ± 15.2 for the betamethasone group, 70.8 ± 12.6 for the celecoxib group and 78.7 ± 11.6 for the control group. No differences in scores were observed among groups (n.s.). In the control group, a negative correlation was observed between PSQ score and KOOS-pain scores at the end of the follow-up in addition to a positive correlation between PSQ score and rescue analgesia consumption at the first postoperative week. The optimal cutoff value for PSQ score to predict insufficient improvement in KOOS-pain subscale was 5.0 points. CONCLUSIONS A cutoff value of pain sensitivity questionnaire score above 5.0 points was determined to identify patients with higher sensitivity to pain who underwent arthroscopic partial meniscectomy. These patients reported relatively increased pain and consumed more rescue analgesics postoperatively unless treated with a single intraoperative corticosteroids injection or oral non-steroidal anti-inflammatories. Therefore, surgeons can use pain sensitivity questionnaire score as a preoperative tool to identify patients with high sensitivity to pain and customize their postoperative analgesics protocol to better fit their pain levels. LEVEL OF EVIDENCE II.
Collapse
Affiliation(s)
- Lee Yaari
- Arthroscopy and Sports Injuries Unit, Rabin Medical Center, Hasharon Hospital, 7 Keren Kayemet St, 49372, Petach-Tikva, Israel
- Orthopedic Department, Sackler Faculty of Medicine, Tel-Aviv University, Tel Aviv-Yafo, Israel
- Orthopedic Department, Rabin Medical Center, Hasharon Hospital, Petach-Tikva, Israel
| | - Amir Dolev
- Orthopedic Department, Sackler Faculty of Medicine, Tel-Aviv University, Tel Aviv-Yafo, Israel
- Orthopedic Department, Rabin Medical Center, Hasharon Hospital, Petach-Tikva, Israel
| | - Mohamed Kittani
- Arthroscopy and Sports Injuries Unit, Rabin Medical Center, Hasharon Hospital, 7 Keren Kayemet St, 49372, Petach-Tikva, Israel
- Orthopedic Department, Sackler Faculty of Medicine, Tel-Aviv University, Tel Aviv-Yafo, Israel
- Orthopedic Department, Rabin Medical Center, Hasharon Hospital, Petach-Tikva, Israel
| | - Mustafa Yassin
- Arthroscopy and Sports Injuries Unit, Rabin Medical Center, Hasharon Hospital, 7 Keren Kayemet St, 49372, Petach-Tikva, Israel
- Orthopedic Department, Sackler Faculty of Medicine, Tel-Aviv University, Tel Aviv-Yafo, Israel
- Orthopedic Department, Rabin Medical Center, Hasharon Hospital, Petach-Tikva, Israel
| | - Mahmod Gbaren
- Orthopedic Department, Rabin Medical Center, Hasharon Hospital, Petach-Tikva, Israel
| | - Elia Feicht
- Orthopedic Department, Rabin Medical Center, Hasharon Hospital, Petach-Tikva, Israel
| | - Barak Haviv
- Arthroscopy and Sports Injuries Unit, Rabin Medical Center, Hasharon Hospital, 7 Keren Kayemet St, 49372, Petach-Tikva, Israel.
- Orthopedic Department, Sackler Faculty of Medicine, Tel-Aviv University, Tel Aviv-Yafo, Israel.
- Orthopedic Department, Rabin Medical Center, Hasharon Hospital, Petach-Tikva, Israel.
| |
Collapse
|
31
|
Hart DA, Nakamura N, Shrive NG. Perspective: Challenges Presented for Regeneration of Heterogeneous Musculoskeletal Tissues that Normally Develop in Unique Biomechanical Environments. Front Bioeng Biotechnol 2021; 9:760273. [PMID: 34650964 PMCID: PMC8505961 DOI: 10.3389/fbioe.2021.760273] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Accepted: 09/13/2021] [Indexed: 11/30/2022] Open
Abstract
Perspective: Musculoskeletal (MSK) tissues such as articular cartilage, menisci, tendons, and ligaments are often injured throughout life as a consequence of accidents. Joints can also become compromised due to the presence of inflammatory diseases such as rheumatoid arthritis. Thus, there is a need to develop regenerative approaches to address such injuries to heterogeneous tissues and ones that occur in heterogeneous environments. Such injuries can compromise both the biomechanical integrity and functional capability of these tissues. Thus, there are several challenges to overcome in order to enhance success of efforts to repair and regenerate damaged MSK tissues. Challenges: 1. MSK tissues arise during development in very different biological and biomechanical environments. These early tissues serve as a template to address the biomechanical requirements evolving during growth and maturation towards skeletal maturity. Many of these tissues are heterogeneous and have transition points in their matrix. The heterogeneity of environments thus presents a challenge to replicate with regard to both the cells and the ECM. 2. Growth and maturation of musculoskeletal tissues occurs in the presence of anabolic mediators such as growth hormone and the IGF-1 family of proteins which decline with age and are low when there is a greater need for the repair and regeneration of injured or damaged tissues with advancing age. Thus, there is the challenge of re-creating an anabolic environment to enhance incorporation of implanted constructs. 3. The environments associated with injury or chronic degeneration of tissues are often catabolic or inflammatory. Thus, there is the challenge of creating a more favorable in vivo environment to facilitate the successful implantation of in vitro engineered constructs to regenerate damaged tissues. Conclusions: The goal of regenerating MSK tissues has to be to meet not only the biological requirements (components and structure) but also the heterogeneity of function (biomechanics) in vivo. Furthermore, for many of these tissues, the regenerative approach has to overcome the site of injury being influenced by catabolism/inflammation. Attempts to date using both endogenous cells, exogenous cells and scaffolds of various types have been limited in achieving long term outcomes, but progress is being made.
Collapse
Affiliation(s)
- David A Hart
- Department of Surgery, Faculty of Kinesiology, University of Calgary, Calgary, AB, Canada.,Bone and Joint Health Strategic Clinical Network, Alberta Health Services, Edmonton, AB, Canada.,McCaig Institute for Bone & Joint Health, University of Calgary, Calgary, AB, Canada
| | - Norimasa Nakamura
- Institute for Medical Science in Sport, Osaka Health Science University, Osaka, Japan
| | - Nigel G Shrive
- McCaig Institute for Bone & Joint Health, University of Calgary, Calgary, AB, Canada.,Biomedical Engineering Graduate Program, Department of Civil Engineering, University of Calgary, Calgary, AB, Canada
| |
Collapse
|
32
|
Belk JW, Keeling LE, Kraeutler MJ, Snow MG, Mei-Dan O, Scillia AJ, McCarty EC. Risk of Infection in Knee Arthroscopy Patients Undergoing Corticosteroid Injections in the Perioperative Period. Orthop J Sports Med 2021; 9:23259671211032941. [PMID: 34423063 PMCID: PMC8375342 DOI: 10.1177/23259671211032941] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Accepted: 04/14/2021] [Indexed: 02/04/2023] Open
Abstract
Background: Recent evidence suggests that there may be an increased risk of infection for patients undergoing a corticosteroid injection before, during, or after knee arthroscopy. Purpose: To systematically review the literature to evaluate the risk of postoperative infection in patients undergoing intra-articular corticosteroid injections (CSI) before, during, or after knee arthroscopy. Study Design: Systematic review; Level of evidence, 3. Methods: A systematic review was performed by searching the PubMed, Cochrane Library, and Embase databases to identify studies that evaluated the rate of postoperative infection in patients undergoing knee arthroscopy who received an intra-articular CSI during the perioperative period. The search phrase used was “knee AND arthroscopy AND injection AND (infection OR revision).” A subanalysis was also performed to analyze infection rates based on the timing of the corticosteroid injection in relation to arthroscopy. Results: Four studies met the inclusion criteria, representing 11,925 patients undergoing knee arthroscopy with an intra-articular CSI administered during the perioperative period (mean follow-up, 5.3 months) and 247,329 patients without a corticosteroid injection during the perioperative period (mean follow-up, 5.9 months). Patients who received an injection experienced a statistically significantly higher rate of postoperative infection (2.2%) when compared with patients who did not receive an injection (1.1%; P < .001). When analyzed by the timing of the injection, patients receiving an injection preoperatively or intraoperatively experienced a statistically significantly higher rate of postoperative infection (3% and 2.6%, respectively) when compared with patients receiving an injection postoperatively (1.4%; P = .001 for both). Conclusion: Patients undergoing knee arthroscopy who receive an intra-articular CSI during the perioperative period can be expected to experience significantly higher postoperative infection rates when compared with patients not receiving an injection. Furthermore, patients receiving a corticosteroid injection pre- or intraoperatively may experience significantly higher rates of postoperative infection when compared with patients receiving an injection postoperatively.
Collapse
Affiliation(s)
- John W Belk
- Department of Orthopaedics, University of Colorado School of Medicine, Aurora, Colorado, USA
| | - Laura E Keeling
- Department of Orthopaedic Surgery, Medstar Georgetown University Hospital, Washington DC, USA
| | - Matthew J Kraeutler
- Saint Joseph's Regional Medical Center, Department of Orthopaedic Surgery, Paterson, New Jersey, USA
| | - Michaela G Snow
- Department of Orthopaedics, University of Colorado School of Medicine, Aurora, Colorado, USA
| | - Omer Mei-Dan
- Department of Orthopaedics, University of Colorado School of Medicine, Aurora, Colorado, USA
| | - Anthony J Scillia
- Saint Joseph's Regional Medical Center, Department of Orthopaedic Surgery, Paterson, New Jersey, USA
| | - Eric C McCarty
- Department of Orthopaedics, University of Colorado School of Medicine, Aurora, Colorado, USA
| |
Collapse
|
33
|
Khella CM, Horvath JM, Asgarian R, Rolauffs B, Hart ML. Anti-Inflammatory Therapeutic Approaches to Prevent or Delay Post-Traumatic Osteoarthritis (PTOA) of the Knee Joint with a Focus on Sustained Delivery Approaches. Int J Mol Sci 2021; 22:8005. [PMID: 34360771 PMCID: PMC8347094 DOI: 10.3390/ijms22158005] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Revised: 07/20/2021] [Accepted: 07/22/2021] [Indexed: 12/11/2022] Open
Abstract
Inflammation plays a central role in the pathogenesis of knee PTOA after knee trauma. While a comprehensive therapy capable of preventing or delaying post-traumatic osteoarthritis (PTOA) progression after knee joint injury does not yet clinically exist, current literature suggests that certain aspects of early post-traumatic pathology of the knee joint may be prevented or delayed by anti-inflammatory therapeutic interventions. We discuss multifaceted therapeutic approaches that may be capable of effectively reducing the continuous cycle of inflammation and concomitant processes that lead to cartilage degradation as well as those that can simultaneously promote intrinsic repair processes. Within this context, we focus on early disease prevention, the optimal timeframe of treatment and possible long-lasting sustained delivery local modes of treatments that could prevent knee joint-associated PTOA symptoms. Specifically, we identify anti-inflammatory candidates that are not only anti-inflammatory but also anti-degenerative, anti-apoptotic and pro-regenerative.
Collapse
Affiliation(s)
| | | | | | | | - Melanie L. Hart
- G.E.R.N. Center for Tissue Replacement, Regeneration & Neogenesis, Department of Orthopedics and Trauma Surgery, Faculty of Medicine, Medical Center—Albert-Ludwigs—University of Freiburg, 79085 Freiburg im Breisgau, Germany; (C.M.K.); (J.M.H.); (R.A.); (B.R.)
| |
Collapse
|
34
|
Mason D, Englund M, Watt FE. Prevention of posttraumatic osteoarthritis at the time of injury: Where are we now, and where are we going? J Orthop Res 2021; 39:1152-1163. [PMID: 33458863 DOI: 10.1002/jor.24982] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Accepted: 01/11/2021] [Indexed: 02/04/2023]
Abstract
This overview of progress made in preventing post-traumatic osteoarthritis (PTOA) was delivered in a workshop at the Orthopaedics Research Society Annual Conference in 2019. As joint trauma is a major risk factor for OA, defining the molecular changes within the joint at the time of injury may enable the targeting of biological processes to prevent later disease. Animal models have been used to test therapeutic targets to prevent PTOA. A review of drug treatments for PTOA in rodents and rabbits between 2016 and 2018 revealed 11 systemic interventions, 5 repeated intra-articular or topical interventions, and 5 short-term intra-articular interventions, which reduced total Osteoarthritis Research Society International scores by 30%-50%, 20%-70%, and 0%-40%, respectively. Standardized study design, reporting of effect size, and quality metrics, alongside a "whole joint" approach to assessing efficacy, would improve the translation of promising new drugs. A roadblock to translating preclinical discoveries has been the lack of guidelines on the design and conduct of human trials to prevent PTOA. An international workshop addressing this in 2016 considered inclusion criteria and study design, and advocated the use of experimental medicine studies to triage candidate treatments and the development of early biological and imaging biomarkers. Human trials for the prevention of PTOA have tested anakinra after anterior cruciate ligament rupture and dexamethasone after radiocarpal injury. PTOA offers a unique opportunity for defining early mechanisms of OA to target therapeutically. Progress in trial design and high-quality preclinical research, and allegiance with patients, regulatory bodies, and the pharmaceutical industry, will advance this field.
Collapse
Affiliation(s)
- Deborah Mason
- Biomechanics and Bioengineeering Centre Versus Arthritis, School of Biosciences, Cardiff University, Cardiff, Wales, UK
| | - Martin Englund
- Faculty of Medicine, Department of Clinical Sciences Lund, Orthopedics, Clinical Epidemiology Unit, Lund Unversity, Lund, Sweden
| | - Fiona E Watt
- Centre for Osteoarthritis Pathogenesis Versus Arthritis, Kennedy Institute of Rheumatology, NDORMS, University of Oxford, Oxford, UK
| |
Collapse
|
35
|
Feltham T, Paudel S, Lobao M, Schon L, Zhang Z. Low-Intensity Pulsed Ultrasound Suppresses Synovial Macrophage Infiltration and Inflammation in Injured Knees in Rats. ULTRASOUND IN MEDICINE & BIOLOGY 2021; 47:1045-1053. [PMID: 33423862 DOI: 10.1016/j.ultrasmedbio.2020.12.019] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Revised: 12/14/2020] [Accepted: 12/16/2020] [Indexed: 06/12/2023]
Abstract
This study was designed to investigate how low-intensity pulsed ultrasound (LIPUS) suppresses traumatic joint inflammation and thereafter affects the progression of posttraumatic osteoarthritis. Intra-articular fracture (IAF) was created in the right knee of rats. LIPUS was applied to the knees with IAFs for 20 min/d for 2 wk-LIPUS(+) group. The study controls included rats that underwent sham surgery but no LIPUS treatment (control group) or underwent IAF surgery without LIPUS treatment-LIPUS(-) group. By histology, at 4 wk, leukocyte infiltration in the synovium was reduced in the LIPUS(+) group. Furthermore, LIPUS treatment reduced CD68+ macrophages in the synovium and limited their distribution mostly in the subintimal synovium. Measured with enzyme-linked immunosorbent assay, interleukin-1β (IL-1β) in the joint fluid of the LIPUS(+) group was reduced to about one-third that in the LIPUS(-) group. By reducing synovial macrophages and lowering IL-1β in the joint fluid, LIPUS is potentially therapeutic for posttraumatic osteoarthritis.
Collapse
Affiliation(s)
- Tyler Feltham
- Philadelphia College of Osteopathic Medicine-Georgia, Suwanee, Georgia, USA
| | - Sharada Paudel
- Frederick National Laboratory for Cancer Research, Frederick, Maryland, USA
| | - Mario Lobao
- Columbia Medical Center, Columbia University, New York, New York, USA
| | - Lew Schon
- Institute for Foot & Ankle Reconstruction, Mercy Medical Center, Baltimore, Maryland, USA
| | - Zijun Zhang
- Center for Orthopaedic Innovation, Mercy Medical Center, Baltimore, Maryland, USA.
| |
Collapse
|
36
|
Dolev A, Yaari L, Kittani M, Yassin M, Gbaren M, Feicht E, Shemesh S, Haviv B. Efficacy of Anti-inflammatory Treatment Versus Rescue Analgesia After Arthroscopic Partial Meniscectomy in Nonarthritic Knees: A 3-Arm Controlled Study. Orthop J Sports Med 2021; 9:2325967121991545. [PMID: 33796593 PMCID: PMC7983444 DOI: 10.1177/2325967121991545] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Accepted: 11/13/2020] [Indexed: 11/17/2022] Open
Abstract
Background There is currently no consensus regarding the appropriate treatment for postoperative pain after arthroscopic partial meniscectomy (APM). Prescribing a mild non-anti-inflammatory protocol of rescue analgesia may be sufficient to avoid the side effects of opioids or anti-inflammatories. Purpose/Hypothesis The purpose was to compare the efficacy of pain reduction after APM in nonarthritic knees using betamethasone or celecoxib as anti-inflammatory analgesics versus acetaminophen or tramadol as rescue analgesics. The hypothesis was that there is no advantage for anti-inflammatories in achieving postoperative immediate pain relief after APM in nonarthritic knees compared with a simple nonopioid treatment. Study Design Cohort study; Level of evidence, 2. Methods This 3-arm controlled study evaluated postoperative pain levels and analgesic consumption in patients who underwent primary APM (under general anesthesia) at a single institution from December 2018 to December 2019. Patients were prospectively divided into 3 treatment groups: (1) betamethasone injection at the end of the procedure, (2) oral celecoxib prescription, or (3) neither treatment (control). All groups were instructed to take supplementary acetaminophen as needed. Patients were also allowed to take tramadol as needed to evaluate the need for opioids. At postoperative weeks 1, 2, and 3, patients completed the Knee injury and Osteoarthritis Outcome Score (KOOS) Pain subscale, and results were compared between time points and groups. Results A total of 99 patients were included in the treatment groups: betamethasone group (32 patients), celecoxib group (30 patients), and control group (37 patients). At baseline, there were no statistically significant differences between the groups in age, sex, body mass index, level of activity, comorbidities, or surgical findings. KOOS Pain scores improved at every time point for all 3 groups (P < .001), and no differences in scores were observed among groups. The consumption of acetaminophen or tramadol as rescue analgesia throughout the follow-up period was negligible among groups. Conclusion During the first 3 postoperative weeks after APM in nonarthritic knees, pain was efficiently controlled by betamethasone or celecoxib; however, pain was also efficiently controlled by minimal consumption of acetaminophen with negligible use of tramadol. Therefore, acetaminophen could be prescribed as an effective first-line postoperative analgesic after APM.
Collapse
Affiliation(s)
- Amir Dolev
- Orthopedic Department, Hasharon Hospital, Rabin Medical Center, Petach Tikva, Israel.,Orthopedic Department, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Lee Yaari
- Orthopedic Department, Hasharon Hospital, Rabin Medical Center, Petach Tikva, Israel.,Orthopedic Department, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Mohamed Kittani
- Orthopedic Department, Hasharon Hospital, Rabin Medical Center, Petach Tikva, Israel.,Orthopedic Department, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Mustafa Yassin
- Orthopedic Department, Hasharon Hospital, Rabin Medical Center, Petach Tikva, Israel.,Orthopedic Department, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Mahmod Gbaren
- Orthopedic Department, Hasharon Hospital, Rabin Medical Center, Petach Tikva, Israel.,Orthopedic Department, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Elia Feicht
- Orthopedic Department, Hasharon Hospital, Rabin Medical Center, Petach Tikva, Israel.,Orthopedic Department, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Shai Shemesh
- Orthopedic Department, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Barak Haviv
- Orthopedic Department, Hasharon Hospital, Rabin Medical Center, Petach Tikva, Israel.,Orthopedic Department, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| |
Collapse
|
37
|
Zheng L, Wang Y, Qiu P, Xia C, Fang Y, Mei S, Fang C, Shi Y, Wu K, Chen Z, Fan S, He D, Lin X, Chen P. Primary chondrocyte exosomes mediate osteoarthritis progression by regulating mitochondrion and immune reactivity. Nanomedicine (Lond) 2020; 14:3193-3212. [PMID: 31855117 DOI: 10.2217/nnm-2018-0498] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Aim: We aimed to investigate the proteomics of primary chondrocyte exosomes and the effect of exosomes in osteoarthritis (OA) treatment. Materials & methods: We isolated exosomes from primary chondrocytes cultured in normal (D0) and inflammatory environments induced by IL-1β and determined the proteomics of these exosomes. Next, we investigated what effect and mechanism D0 chondrocytes exosomes have in OA treatment. Results: There were more proteins that belonged to mitochondrion and were involved in immune system processes in D0 exosomes. Notably, intra-articular administration of D0 exosomes successfully prevented the development of OA. D0 chondrocyte exosomes could restore mitochondrial dysfunction and polarize macrophage response toward an M2 phenotype. Conclusion: Our findings demonstrated that primary chondrocyte exosomes are efficient in OA treatment.
Collapse
Affiliation(s)
- Lin Zheng
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Medical College of Zhejiang University, Hangzhou, PR China.,Key Laboratory of Musculoskeletal System Degeneration & Regeneration Translational Research of Zhejiang Province, Hangzhou, PR China.,Department of Orthopedics, 5th Affiliated Hospital, Lishui Municipal Central Hospital, Wenzhou Medical University, Lishui, PR China
| | - Yiyun Wang
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Medical College of Zhejiang University, Hangzhou, PR China.,Key Laboratory of Musculoskeletal System Degeneration & Regeneration Translational Research of Zhejiang Province, Hangzhou, PR China
| | - Pengcheng Qiu
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Medical College of Zhejiang University, Hangzhou, PR China.,Key Laboratory of Musculoskeletal System Degeneration & Regeneration Translational Research of Zhejiang Province, Hangzhou, PR China
| | - Chen Xia
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Medical College of Zhejiang University, Hangzhou, PR China.,Key Laboratory of Musculoskeletal System Degeneration & Regeneration Translational Research of Zhejiang Province, Hangzhou, PR China
| | - Yifan Fang
- Hangzhou Foreign Languages School, Hangzhou, PR China
| | - Sheng Mei
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Medical College of Zhejiang University, Hangzhou, PR China.,Key Laboratory of Musculoskeletal System Degeneration & Regeneration Translational Research of Zhejiang Province, Hangzhou, PR China
| | - Chen Fang
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Medical College of Zhejiang University, Hangzhou, PR China.,Key Laboratory of Musculoskeletal System Degeneration & Regeneration Translational Research of Zhejiang Province, Hangzhou, PR China
| | - Yiling Shi
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Medical College of Zhejiang University, Hangzhou, PR China.,Key Laboratory of Musculoskeletal System Degeneration & Regeneration Translational Research of Zhejiang Province, Hangzhou, PR China
| | - Kaiwei Wu
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Medical College of Zhejiang University, Hangzhou, PR China.,Key Laboratory of Musculoskeletal System Degeneration & Regeneration Translational Research of Zhejiang Province, Hangzhou, PR China
| | - Zhijun Chen
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Medical College of Zhejiang University, Hangzhou, PR China.,Key Laboratory of Musculoskeletal System Degeneration & Regeneration Translational Research of Zhejiang Province, Hangzhou, PR China
| | - Shunwu Fan
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Medical College of Zhejiang University, Hangzhou, PR China.,Key Laboratory of Musculoskeletal System Degeneration & Regeneration Translational Research of Zhejiang Province, Hangzhou, PR China
| | - Dengwei He
- Department of Orthopedics, 5th Affiliated Hospital, Lishui Municipal Central Hospital, Wenzhou Medical University, Lishui, PR China
| | - Xianfeng Lin
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Medical College of Zhejiang University, Hangzhou, PR China.,Key Laboratory of Musculoskeletal System Degeneration & Regeneration Translational Research of Zhejiang Province, Hangzhou, PR China
| | - Pengfei Chen
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Medical College of Zhejiang University, Hangzhou, PR China.,Key Laboratory of Musculoskeletal System Degeneration & Regeneration Translational Research of Zhejiang Province, Hangzhou, PR China
| |
Collapse
|
38
|
Ebrahimi M, Turunen MJ, Finnilä MA, Joukainen A, Kröger H, Saarakkala S, Korhonen RK, Tanska P. Structure-Function Relationships of Healthy and Osteoarthritic Human Tibial Cartilage: Experimental and Numerical Investigation. Ann Biomed Eng 2020; 48:2887-2900. [PMID: 32648191 PMCID: PMC7723942 DOI: 10.1007/s10439-020-02559-0] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2020] [Accepted: 06/26/2020] [Indexed: 02/07/2023]
Abstract
Relationships between composition, structure and constituent-specific functional properties of human articular cartilage at different stages of osteoarthritis (OA) are poorly known. We established these relationships by comparison of elastic, viscoelastic and fibril-reinforced poroelastic mechanical properties with microscopic and spectroscopic analysis of structure and composition of healthy and osteoarthritic human tibial cartilage (n = 27). At a low frequency (0.005 Hz), proteoglycan content correlated negatively and collagen content correlated positively with the phase difference (i.e. tissue viscosity). At a high-frequency regime (> 0.05 Hz), proteoglycan content correlated negatively and collagen orientation angle correlated positively with the phase difference. Proteoglycans were lost in the early and advanced OA groups compared to the healthy group, while the superficial collagen orientation angle was greater only in the advanced OA group compared to the healthy group. Simultaneously, the initial fibril network modulus (fibril pretension) was smaller in the early and advanced OA groups compared to the healthy group. These findings suggest different mechanisms contribute to cartilage viscosity in low and high frequencies, and that the loss of superficial collagen pretension during early OA is due to lower tissue swelling (PG loss), while in advanced OA, both collagen disorganization and lower swelling modulate the collagen fibril pretension.
Collapse
Affiliation(s)
- Mohammadhossein Ebrahimi
- Department of Applied Physics, University of Eastern Finland, POB 1627, 70211, Kuopio, Finland.
- Research Unit of Medical Imaging, Physics and Technology, Faculty of Medicine, University of Oulu, Oulu, Finland.
| | - Mikael J Turunen
- Department of Applied Physics, University of Eastern Finland, POB 1627, 70211, Kuopio, Finland
- SIBlabs, University of Eastern Finland, Kuopio, Finland
| | - Mikko A Finnilä
- Research Unit of Medical Imaging, Physics and Technology, Faculty of Medicine, University of Oulu, Oulu, Finland
| | | | | | - Simo Saarakkala
- Research Unit of Medical Imaging, Physics and Technology, Faculty of Medicine, University of Oulu, Oulu, Finland
- Department of Diagnostic Radiology, Oulu University Hospital, Oulu, Finland
| | - Rami K Korhonen
- Department of Applied Physics, University of Eastern Finland, POB 1627, 70211, Kuopio, Finland
| | - Petri Tanska
- Department of Applied Physics, University of Eastern Finland, POB 1627, 70211, Kuopio, Finland
| |
Collapse
|
39
|
Bonnet CS, Gilbert SJ, Blain EJ, Williams AS, Mason DJ. AMPA/kainate glutamate receptor antagonists prevent posttraumatic osteoarthritis. JCI Insight 2020; 5:134055. [PMID: 32544091 DOI: 10.1172/jci.insight.134055] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2019] [Accepted: 05/29/2020] [Indexed: 01/10/2023] Open
Abstract
Musculoskeletal disorders represent the third greatest burden in terms of death and disability in the developed world. Osteoarthritis is the single greatest cause of chronic pain, has no cure, and affects 8.5 and 27 million people in the UK and US, respectively. Osteoarthritis is most prevalent in older people, but as it commonly occurs after joint injury, young people with such injuries are also susceptible. Painful joints are often treated with steroid or hyaluronic acid (HA) injections, but treatments to prevent subsequent joint degeneration remain elusive. In animals, joint injury increases glutamate release into the joint, acting on nerves to cause pain, and joint tissues to cause inflammation and degeneration. This study investigated synovial fluid glutamate concentrations and glutamate receptor (GluR) expression in injured human joints and compared the efficacy of GluR antagonists with current treatments in a mouse model of injury-induced osteoarthritis (ACL rupture). GluRs were expressed in the ligaments and meniscus after knee injury, and synovial fluid glutamate concentrations ranged from 19 to 129 μM. Intra-articular injection of NBQX (GluR antagonist) at the time of injury substantially reduced swelling and degeneration in the mouse ACL rupture model. HA had no effect, and Depo-Medrone reduced swelling for 1 day but increased degeneration by 50%. Intra-articular administration of NBQX modified both symptoms and disease to a greater extent than current treatments. There is an opportunity for repurposing related drugs, developed for CNS disorders and with proven safety in humans, to prevent injury-induced osteoarthritis. This could quickly reduce the substantial burden associated with osteoarthritis.
Collapse
Affiliation(s)
- Cleo S Bonnet
- School of Biosciences.,Biomechanics and Bioengineering Research Centre Versus Arthritis, and
| | - Sophie J Gilbert
- School of Biosciences.,Biomechanics and Bioengineering Research Centre Versus Arthritis, and
| | - Emma J Blain
- School of Biosciences.,Biomechanics and Bioengineering Research Centre Versus Arthritis, and
| | - Anwen S Williams
- Biomechanics and Bioengineering Research Centre Versus Arthritis, and.,School of Medicine, Cardiff University, Cardiff, United Kingdom
| | - Deborah J Mason
- School of Biosciences.,Biomechanics and Bioengineering Research Centre Versus Arthritis, and
| |
Collapse
|
40
|
Eom TH, Lim HR, Jeong SH, Park KS, Jang CH. Hearing Results Following Type 1 Tympanoplasty in Elderly Patients. In Vivo 2020; 34:1395-1398. [PMID: 32354936 DOI: 10.21873/invivo.11919] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2019] [Revised: 01/24/2020] [Accepted: 01/27/2020] [Indexed: 01/18/2023]
Abstract
BACKGROUND/AIM The outcomes of type 1 tympanoplasty in elderly patients remain controversial. Therefore, more studies are needed to clarify the prognosis of elderly patients after tympanoplasty. The purpose of this study was to evaluate the clinical outcomes of type 1 tympanoplasty in elderly patients. PATIENTS AND METHODS We retrospectively analyzed data from 116 patients who underwent type 1 tympanoplasty due to chronic otitis media. Seventy-one of the 116 patients were elderly individuals aged 65 years or older (study group). Forty-five patients were younger than 65 years (control group). Due to cochlear intolerance by aging in the study group, we used dexamethasone soaked gelfoam packing in the middle ear and intraoperative dexamethasone injection. To compare the outcomes between groups, we determined the mean hearing levels by averaging the hearing thresholds. The differences in the air-bone gaps before and after tympanoplasty were compared between groups. RESULTS In the study group, 54 patients had an underlying disease (76%). Hypertension was the most common underlying disease. The postoperative air conduction (AC) and bone conduction (BC) improved in both the study group and the control group. In the control group, postoperative air-bone gap (ABG) was significantly higher than preoperative ABG. Although the postoperative ABG improved in the study group, the improvement was insignificant. CONCLUSION Although significant improvement of ABG was not achieved, postoperative AC and BC were improved. Intraoperative dexamethasone injection and dexamethasone soaked gelfoam packing in the middle ear was effective to prevent deterioration of BC after operation.
Collapse
Affiliation(s)
- Tae-Ho Eom
- Suwan Best ENT Clinic, Gwangju, Republic of Korea
| | - Hye-Rin Lim
- Department of Otolaryngology, Chonnam National University Medical School, Gwangju, Republic of Korea
| | - Se-Hyun Jeong
- Department of Otolaryngology, Chonnam National University Medical School, Gwangju, Republic of Korea
| | - Kyeong-Suk Park
- Department of Otolaryngology, Chonnam National University Medical School, Gwangju, Republic of Korea
| | - Chul Ho Jang
- Department of Otolaryngology, Chonnam National University Medical School, Gwangju, Republic of Korea
| |
Collapse
|
41
|
Wang LJ, Zeng N, Yan ZP, Li JT, Ni GX. Post-traumatic osteoarthritis following ACL injury. Arthritis Res Ther 2020; 22:57. [PMID: 32209130 PMCID: PMC7092615 DOI: 10.1186/s13075-020-02156-5] [Citation(s) in RCA: 125] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2020] [Accepted: 03/16/2020] [Indexed: 02/08/2023] Open
Abstract
Post-traumatic osteoarthritis (PTOA) develops after joint injury. Specifically, patients with anterior cruciate ligament (ACL) injury have a high risk of developing PTOA. In this review, we outline the incidence of ACL injury that progresses to PTOA, analyze the role of ACL reconstruction in preventing PTOA, suggest possible mechanisms thought to be responsible for PTOA, evaluate current diagnostic methods for detecting early OA, and discuss potential interventions to combat PTOA. We also identify important directions for future research. Although much work has been done, the incidence of PTOA among patients with a history of ACL injury remains high due to the complexity of ACL injury progression to PTOA, the lack of sensitive and easily accessible diagnostic methods to detect OA development, and the limitations of current treatments. A number of factors are thought to be involved in the underlying mechanism, including structural factors, biological factors, mechanical factors, and neuromuscular factor. Since there is a clear "start point" for PTOA, early detection and intervention is of great importance. Currently, imaging modalities and specific biomarkers allow early detection of PTOA. However, none of them is both sensitive and easily accessible. After ACL injury, many patients undergo surgical reconstruction of ACL to restore joint stability and prevent excessive loading. However, convincing evidence is still lacking for the superiority of ACL-R to conservative management in term of the incidence of PTOA. As for non-surgical treatment such as anti-cytokine and chemokine interventions, most of them are investigated in animal studies and have not been applied to humans. A complete understanding of mechanisms to stratify the patients into different subgroups on the basis of risk factors is critical. And the improvement of standardized and quantitative assessment techniques is necessary to guide intervention. Moreover, treatments targeted toward different pathogenic pathways may be crucial to the management of PTOA in the future.
Collapse
Affiliation(s)
- Li-Juan Wang
- School of Sport Medicine and Rehabilitation, Beijing Sport University, Beijing, China
| | - Ni Zeng
- Department of Rehabilitation Medicine, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China
| | - Zhi-Peng Yan
- Department of Rehabilitation Medicine, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China
| | - Jie-Ting Li
- Department of Rehabilitation Medicine, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China
| | - Guo-Xin Ni
- School of Sport Medicine and Rehabilitation, Beijing Sport University, Beijing, China.
| |
Collapse
|
42
|
Yu W, Zhu Y, Li H, He Y. Injectable Quercetin-Loaded Hydrogel with Cartilage-Protection and Immunomodulatory Properties for Articular Cartilage Repair. ACS APPLIED BIO MATERIALS 2020; 3:761-771. [PMID: 35019280 DOI: 10.1021/acsabm.9b00673] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Articular cartilage plays an important role in human body. How to repair articular cartilage defects when they appear due to various factors has always been a major clinical challenge. Recently, studies have shown that slowing the degradation of cartilage extracellular matrix (ECM) and modulating the inflammatory response of the host thereby promoting cartilage tissue regeneration are important in the cartilage repair process. In this study, a drug-loaded injectable hydrogel was constructed for repairing articular cartilage. This hydrogel could not only maintain the phenotype of chondrocytes but also regulate the inflammatory response of the host. The injectable sodium alginate (SA)/bioglass (BG) hydrogel was mixed with the injectable thermal-responsive SA/agarose (AG)/quercetin (Que) hydrogel to obtain an injectable hydrogel containing both Que and BG (Que-BG hydrogel) for articular cartilage regeneration. The Que-BG hydrogel has a proper swelling ratio that can promote integration between the formed tissue and host tissue, and it allows Que to release slowly in situ to improve its bioavailability. The Que-BG hydrogel could upregulate SRY-box 9 (SOX9), aggrecan (ACAN), and collagen type II alpha 1 chain (COL2A1) of normal chondrocytes to maintain the normal chondrocyte phenotype. In addition, it could promote macrophage M2 polarization, reduce inflammation, and inhibit ECM degradation by downregulating the expression of inducible nitric oxide synthase (iNOS), matrix metalloproteinase-13 (MMP13), and matrix metalloproteinase-1 (MMP1) in degenerative chondrocytes. After injecting the Que-BG hydrogel into a rat cartilage defect model, the formed tissue was observed to be similar to the normal tissue and was highly integrated with the surrounding tissue. Therefore, the injectable Que-BG hydrogel improves Que bioavailability, maintains chondrocyte phenotype, inhibits ECM degradation, and reduces inflammatory response.
Collapse
Affiliation(s)
- Weihan Yu
- Department of Orthopedics, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, School of Biomedical Engineering, Shanghai Jiao Tong University, 600 Yishan Road, Shanghai 200233, China.,Department of Orthopedics, Shanghai General Hospital, Shanghai 200080, China
| | - Yanlun Zhu
- School of Biomedical Engineering, Shanghai Jiao Tong University, 1954 Huashan Road, Shanghai 200030, China.,School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China
| | - Haiyan Li
- Department of Orthopedics, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, School of Biomedical Engineering, Shanghai Jiao Tong University, 600 Yishan Road, Shanghai 200233, China.,School of Biomedical Engineering, Shanghai Jiao Tong University, 1954 Huashan Road, Shanghai 200030, China
| | - Yaohua He
- Department of Orthopedics, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, School of Biomedical Engineering, Shanghai Jiao Tong University, 600 Yishan Road, Shanghai 200233, China.,Department of Orthopedics, Shanghai Sixth People's Hospital, Jinshan Branch, Shanghai 201599, China
| |
Collapse
|
43
|
Stefani RM, Lee AJ, Tan AR, Halder SS, Hu Y, Guo XE, Stoker AM, Ateshian GA, Marra KG, Cook JL, Hung CT. Sustained low-dose dexamethasone delivery via a PLGA microsphere-embedded agarose implant for enhanced osteochondral repair. Acta Biomater 2020; 102:326-340. [PMID: 31805408 PMCID: PMC6956850 DOI: 10.1016/j.actbio.2019.11.052] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2019] [Revised: 11/26/2019] [Accepted: 11/27/2019] [Indexed: 12/16/2022]
Abstract
Articular cartilage defects are a common source of joint pain and dysfunction. We hypothesized that sustained low-dose dexamethasone (DEX) delivery via an acellular osteochondral implant would have a dual pro-anabolic and anti-catabolic effect, both supporting the functional integrity of adjacent graft and host tissue while also attenuating inflammation caused by iatrogenic injury. An acellular agarose hydrogel carrier with embedded DEX-loaded poly(lactic-co-glycolic) acid (PLGA) microspheres (DLMS) was developed to provide sustained release for at least 99 days. The DLMS implant was first evaluated in an in vitro pro-inflammatory model of cartilage degradation. The implant was chondroprotective, as indicated by maintenance of Young's modulus (EY) (p = 0.92) and GAG content (p = 1.0) in the presence of interleukin-1β insult. In a subsequent preliminary in vivo experiment, an osteochondral autograft transfer was performed using a pre-clinical canine model. DLMS implants were press-fit into the autograft donor site and compared to intra-articular DEX injection (INJ) or no DEX (CTL). Functional scores for DLMS animals returned to baseline (p = 0.39), whereas CTL and INJ remained significantly worse at 6 months (p < 0.05). DLMS knees were significantly more likely to have improved OARSI scores for proteoglycan, chondrocyte, and collagen pathology (p < 0.05). However, no significant improvements in synovial fluid cytokine content were observed. In conclusion, utilizing a targeted DLMS implant, we observed in vitro chondroprotection in the presence of IL-1-induced degradation and improved in vivo functional outcomes. These improved outcomes were correlated with superior histological scores but not necessarily a dampened inflammatory response, suggesting a primarily pro-anabolic effect. STATEMENT OF SIGNIFICANCE: Articular cartilage defects are a common source of joint pain and dysfunction. Effective treatment of these injuries may prevent the progression of osteoarthritis and reduce the need for total joint replacement. Dexamethasone, a potent glucocorticoid with concomitant anti-catabolic and pro-anabolic effects on cartilage, may serve as an adjuvant for a variety of repair strategies. Utilizing a dexamethasone-loaded osteochondral implant with controlled release characteristics, we demonstrated in vitro chondroprotection in the presence of IL-1-induced degradation and improved in vivo functional outcomes following osteochondral repair. These improved outcomes were correlated with superior histological cartilage scores and minimal-to-no comorbidity, which is a risk with high dose dexamethasone injections. Using this model of cartilage restoration, we have for the first time shown the application of targeted, low-dose dexamethasone for improved healing in a preclinical model of focal defect repair.
Collapse
Affiliation(s)
- Robert M Stefani
- Department of Biomedical Engineering, Columbia University, 351 Engineering Terrace, 1210 Amsterdam Avenue, New York 10027, NY United States
| | - Andy J Lee
- Department of Biomedical Engineering, Columbia University, 351 Engineering Terrace, 1210 Amsterdam Avenue, New York 10027, NY United States
| | - Andrea R Tan
- Department of Biomedical Engineering, Columbia University, 351 Engineering Terrace, 1210 Amsterdam Avenue, New York 10027, NY United States
| | - Saiti S Halder
- Department of Biomedical Engineering, Columbia University, 351 Engineering Terrace, 1210 Amsterdam Avenue, New York 10027, NY United States
| | - Yizhong Hu
- Department of Biomedical Engineering, Columbia University, 351 Engineering Terrace, 1210 Amsterdam Avenue, New York 10027, NY United States
| | - X Edward Guo
- Department of Biomedical Engineering, Columbia University, 351 Engineering Terrace, 1210 Amsterdam Avenue, New York 10027, NY United States
| | - Aaron M Stoker
- Missouri Orthopaedic Institute, University of Missouri, 1100 Virginia Avenue, Columbia 65212, MO, United States
| | - Gerard A Ateshian
- Department of Biomedical Engineering, Columbia University, 351 Engineering Terrace, 1210 Amsterdam Avenue, New York 10027, NY United States; Department of Mechanical Engineering, Columbia University, 500 West 120th Street, 220 S.W. Mudd, New York 10027, NY, United States
| | - Kacey G Marra
- University of Pittsburgh, Biomedical Science Tower, 200 Lothrop Street, Pittsburgh 15213, PA, United States
| | - James L Cook
- Missouri Orthopaedic Institute, University of Missouri, 1100 Virginia Avenue, Columbia 65212, MO, United States
| | - Clark T Hung
- Department of Biomedical Engineering, Columbia University, 351 Engineering Terrace, 1210 Amsterdam Avenue, New York 10027, NY United States.
| |
Collapse
|
44
|
He T, Zhang C, Vedadghavami A, Mehta S, Clark HA, Porter RM, Bajpayee AG. Multi-arm Avidin nano-construct for intra-cartilage delivery of small molecule drugs. J Control Release 2019; 318:109-123. [PMID: 31843642 DOI: 10.1016/j.jconrel.2019.12.020] [Citation(s) in RCA: 60] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2019] [Revised: 12/07/2019] [Accepted: 12/12/2019] [Indexed: 01/15/2023]
Abstract
Targeted drug delivery to joint tissues like cartilage remains a challenge that has prevented clinical translation of promising osteoarthritis (OA) drugs. Local intra-articular (IA) injections of drugs suffer from rapid clearance from the joint space and slow diffusive transport through the dense, avascular cartilage matrix comprised of negatively charged glycosaminoglycans (GAGs). Here we apply drug carriers that leverage electrostatic interactions with the tissue's high negative fixed charge density (FCD) for delivering small molecule drugs to cartilage cell and matrix sites. We demonstrate that a multi-arm cationic nano-construct of Avidin (mAv) with 28 sites for covalent drug conjugation can rapidly penetrate through the full thickness of cartilage in high concentration and have long intra-cartilage residence time in both healthy and arthritic cartilage via weak-reversible binding with negatively charged aggrecans. mAv's intra-cartilage mean uptake was found to be 112× and 33× the equilibration bath concentration in healthy and arthritic (50% GAG depleted) cartilage, respectively. mAv was conjugated with Dexamethasone (mAv-Dex), a broad-spectrum glucocorticoid, using a combination of hydrolysable ester linkers derived from succinic anhydride (SA), 3,3-dimethylglutaric anhydride (GA) and phthalic anhydride (PA) in 2:1:1 M ratio that enabled 50% drug release within 38.5 h followed by sustained release in therapeutic doses over 2 weeks. A single 10 μM low dose of controlled release mAv-Dex (2:1:1) effectively suppressed IL-1α-induced GAG loss, cell death and inflammatory response significantly better than unmodified Dex over 2 weeks in cartilage explant culture models of OA. With this multi-arm design, <1 μM Avidin was needed - a concentration which has been shown to be safe, preventing further GAG loss and cytotoxicity. A charge-based cartilage homing drug delivery platform like this can elicit disease modifying effects as well as facilitate long-term symptomatic pain and inflammation relief by enhancing tissue specificity and prolonging intra-cartilage residence time of OA drugs. This nano-construct thus has high translational potential for enabling intra-cartilage delivery of a broad array of small molecule OA drugs and their combinations to chondrocytes, enabling OA treatment with a single injection of low drug doses and eliminating toxicity issues associated with multiple high dose injections.
Collapse
Affiliation(s)
- Tengfei He
- Departments of Bioengineering, Northeastern University, Boston, MA 02115, USA.
| | - Chenzhen Zhang
- Departments of Bioengineering, Northeastern University, Boston, MA 02115, USA.
| | - Armin Vedadghavami
- Departments of Bioengineering, Northeastern University, Boston, MA 02115, USA.
| | - Shikhar Mehta
- Departments of Bioengineering, Northeastern University, Boston, MA 02115, USA.
| | - Heather A Clark
- Departments of Bioengineering, Northeastern University, Boston, MA 02115, USA; Chemistry and Chemical Biology, Northeastern University, Boston, MA 02115, USA.
| | - Ryan M Porter
- Departments of Internal Medicine and Orthopaedic Surgery, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA.
| | - Ambika G Bajpayee
- Departments of Bioengineering, Northeastern University, Boston, MA 02115, USA; Mechanical Engineering, Northeastern University, Boston, MA 02115, USA.
| |
Collapse
|
45
|
Allogeneic Versus Autologous Injectable Mesenchymal Stem Cells for Knee Osteoarthritis: Review and Current Status. Tech Orthop 2019. [DOI: 10.1097/bto.0000000000000357] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
|
46
|
Abstract
While glucocorticoids have been used for over 50 years to treat rheumatoid and osteoarthritis pain, the prescription of glucocorticoids remains controversial because of potentially harmful side effects at the molecular, cellular and tissue levels. One member of the glucocorticoid family, dexamethasone (DEX) has recently been demonstrated to rescue cartilage matrix loss and chondrocyte viability in animal studies and cartilage explant models of tissue injury and post-traumatic osteoarthritis, suggesting the possibility of DEX as a disease-modifying drug if used appropriately. However, the literature on the effects of DEX on cartilage reveals conflicting results on the drug's safety, depending on the dose and duration of DEX exposure as well as the model system used. Overall, DEX has been shown to protect against arthritis-related changes in cartilage structure and function, including matrix loss, inflammation and cartilage viability. These beneficial effects are not always observed in model systems using initially healthy cartilage or isolated chondrocytes, where many studies have reported significant increases in chondrocyte apoptosis. It is crucially important to understand under what conditions DEX may be beneficial or harmful to cartilage and other joint tissues and to determine potential for safe use of this glucocorticoid in the clinic as a disease-modifying drug.
Collapse
Affiliation(s)
- R. Black
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - A. J. Grodzinsky
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA,Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA,Department of Electrical Engineering and Computer Science, Massachusetts Institute of Technology, Cambridge, MA, USA,Address for correspondence: Prof. Al Grodzinsky, MIT, Centre for Biomedical Engineering, 500 Technology Square, Cambridge, MA, 02139, USA.
| |
Collapse
|
47
|
Rudnik‐Jansen I, Tellegen AR, Pouran B, Schrijver K, Meij BP, Emans PJ, de Gendt E, Thomas RE, Kik MJ, de Visser HM, Weinans H, Egas A, van Maarseveen E, Woike N, Mihov G, Thies J, Tryfonidou MA, Creemers LB. Local controlled release of corticosteroids extends surgically induced joint instability by inhibiting tissue healing. Br J Pharmacol 2019; 176:4050-4064. [PMID: 31378925 PMCID: PMC6811746 DOI: 10.1111/bph.14817] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2018] [Revised: 05/28/2019] [Accepted: 07/11/2019] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND AND PURPOSE Corticosteroids are intra-articularly injected to relieve pain in joints with osteoarthritis (OA) or acute tissue damage such as ligament or tendon tears, despite its unverified contraindication in unstable joints. Biomaterial-based sustained delivery may prolong reduction of inflammatory pain, while avoiding harmful peak drug concentrations. EXPERIMENTAL APPROACH The applicability of prolonged corticosteroid exposure was examined in a rat model of anterior cruciate ligament and medial meniscus transection (ACLT + pMMx) with ensuing degenerative changes. KEY RESULTS Intra-articular injection of a bolus of the corticosteroid triamcinolone acetonide (TAA) resulted in enhanced joint instability in 50% of the joints, but neither instability-induced OA cartilage degeneration, synovitis, nor the OA-related bone phenotype was affected. However, biomaterial microsphere-based extended TAA release enhanced instability in 94% of the animals and induced dystrophic calcification and exacerbation of cartilage degeneration. In healthy joints, injection with TAA releasing microspheres had no effect at all. In vitro, TAA inhibited cell migration out of joint tissue explants, suggesting inhibited tissue healing in vivo as mechanisms for enhanced instability and subsequent cartilage degeneration. CONCLUSIONS AND IMPLICATIONS We conclude that short-term TAA exposure has minor effects on surgically induced unstable joints, but its extended presence is detrimental by extending instability and associated joint degeneration through compromised healing. This supports a contraindication of prolonged corticosteroid exposure in tissue damage-associated joint instability, but not of brief exposure.
Collapse
Affiliation(s)
- Imke Rudnik‐Jansen
- Department of OrthopaedicsUniversity Medical Center UtrechtUtrechtThe Netherlands
| | - Anna R. Tellegen
- Department of Clinical Sciences of Companion Animals, Faculty of Veterinary MedicineUtrecht UniversityUtrechtThe Netherlands
| | - Behdad Pouran
- Department of OrthopaedicsUniversity Medical Center UtrechtUtrechtThe Netherlands
- Department of Biomechanical Engineering, Faculty of Mechanical Maritime and Materials EngineeringDelft University of TechnologyDelftThe Netherlands
| | - Karin Schrijver
- Department of OrthopaedicsUniversity Medical Center UtrechtUtrechtThe Netherlands
| | - Björn P. Meij
- Department of Clinical Sciences of Companion Animals, Faculty of Veterinary MedicineUtrecht UniversityUtrechtThe Netherlands
| | - Pieter J. Emans
- Department of OrthopaedicsMaastricht University Medical Center UtrechtUtrechtThe Netherlands
| | - Erin de Gendt
- Department of OrthopaedicsUniversity Medical Center UtrechtUtrechtThe Netherlands
| | - Rachel E. Thomas
- Department of Pathobiology, Faculty of Veterinary MedicineUtrecht UniversityUtrechtThe Netherlands
| | - Marja J.L. Kik
- Department of Pathobiology, Faculty of Veterinary MedicineUtrecht UniversityUtrechtThe Netherlands
| | - Huub M. de Visser
- Department of OrthopaedicsUniversity Medical Center UtrechtUtrechtThe Netherlands
| | - Harrie Weinans
- Department of OrthopaedicsUniversity Medical Center UtrechtUtrechtThe Netherlands
- Department of Biomechanical Engineering, Faculty of Mechanical Maritime and Materials EngineeringDelft University of TechnologyDelftThe Netherlands
| | - Annelies Egas
- Division Laboratory and Pharmacy, Clinical PharmacyUniversity Medical Center UtrechtUtrechtThe Netherlands
| | - Erik van Maarseveen
- Division Laboratory and Pharmacy, Clinical PharmacyUniversity Medical Center UtrechtUtrechtThe Netherlands
| | - Nina Woike
- Dept of Science and InnovationDSM BiomedicalGeleenThe Netherlands
| | - George Mihov
- Dept of Science and InnovationDSM BiomedicalGeleenThe Netherlands
| | - Jens Thies
- Dept of Science and InnovationDSM BiomedicalGeleenThe Netherlands
| | - Marianna A. Tryfonidou
- Department of Clinical Sciences of Companion Animals, Faculty of Veterinary MedicineUtrecht UniversityUtrechtThe Netherlands
| | - Laura B. Creemers
- Department of OrthopaedicsUniversity Medical Center UtrechtUtrechtThe Netherlands
| |
Collapse
|
48
|
Phen HM, Schenker ML. Minimizing Posttraumatic Osteoarthritis After High-Energy Intra-Articular Fracture. Orthop Clin North Am 2019; 50:433-443. [PMID: 31466660 DOI: 10.1016/j.ocl.2019.05.002] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
This article serves to provide an overview of molecular and surgical interventions to minimize the progression of posttraumatic arthritis following high-energy intra-articular fractures. The roles of cartilage and the microcellular environment are discussed, as well as the response of the joint and cartilage to injury. Molecular therapies, such as glucocorticoids, mesenchymal stem cells, and bisphosphonates, are presented as potential treatments to prevent progression to posttraumatic arthritis. High-energy intra-articular fractures of the elbow, hip, knee, and ankle are discussed, with emphasis on restoring anatomic alignment, articular reduction, and stability of the joint.
Collapse
Affiliation(s)
- Huai Ming Phen
- Emory Orthopaedic Trauma & Fracture, 49 Jesse Hill Jr. Drive South East, 3rd Floor, Atlanta, GA 30303, USA.
| | - Mara L Schenker
- Emory Orthopaedic Trauma & Fracture, 49 Jesse Hill Jr. Drive South East, 3rd Floor, Atlanta, GA 30303, USA
| |
Collapse
|
49
|
Heard BJ, Barton KI, Agbojo OM, Chung M, Sevick JL, Bader TJ, Martin CR, Shrive NG, Hart DA. Molecular Response of Rabbit Menisci to Surgically Induced Hemarthrosis and a Single Intra-Articular Dexamethasone Treatment. J Orthop Res 2019; 37:2043-2052. [PMID: 31095777 DOI: 10.1002/jor.24346] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/02/2019] [Accepted: 05/02/2019] [Indexed: 02/04/2023]
Abstract
Anterior cruciate ligament reconstructive surgery can restore biomechanical stability, however, such surgery cannot reliably prevent the onset of post-traumatic osteoarthritis. The aim of this study was to elucidate the molecular response that occurs within the menisci following a surgical injury that allows bleeding into the joint space, and then to investigate the effect of dexamethasone (DEX) on this molecular response. Cell viability studies following acute controlled exposure to blood and blood plus DEX were also conducted. Forty-eight New Zealand white rabbits were randomly allocated into control, sham, surgical, and surgical + DEX groups (each group n = 6). Animals were sacrificed at 48 h and 9 weeks, and menisci were harvested. The messenger RNA (mRNA) expression levels for key inflammatory, and degradative proteins, as well as mRNA levels for autophagy pathway molecules were quantified, and statistically significant changes were described. Meniscal cell viability was calculated by incubating groups of medial and lateral menisci in autologous blood, or autologous blood plus DEX for 48 h (each group n = 4; total of eight medial and eight lateral menisci), and then conducting a histological live/dead assay. Results indicated a significant reduction in only medial meniscal cell viability when the tissue was exposed to blood in combination with DEX. A single administration of DEX following surgery significantly suppresses the elevated molecular expression for key inflammatory and degradative markers within menisci at 48 h and 9 weeks post-surgery. In vitro, autologous blood did not affect cell viability, but addition of DEX uniquely impacted the medial menisci. © 2019 Orthopaedic Research Society. Published by Wiley Periodicals, Inc. J Orthop Res 37:2043-2052, 2019.
Collapse
Affiliation(s)
- Bryan J Heard
- Cumming School of Medicine, McCaig Institute for Bone and Joint Health, University of Calgary, Calgary, Alberta, Canada.,Department of Surgery, University of Calgary, Calgary, Alberta, Canada
| | - Kristen I Barton
- Cumming School of Medicine, McCaig Institute for Bone and Joint Health, University of Calgary, Calgary, Alberta, Canada
| | - Omokhowa M Agbojo
- Cumming School of Medicine, McCaig Institute for Bone and Joint Health, University of Calgary, Calgary, Alberta, Canada.,Schulich School of Engineering, University of Calgary, Calgary, Alberta, Canada
| | - May Chung
- Cumming School of Medicine, McCaig Institute for Bone and Joint Health, University of Calgary, Calgary, Alberta, Canada
| | - John L Sevick
- Cumming School of Medicine, McCaig Institute for Bone and Joint Health, University of Calgary, Calgary, Alberta, Canada
| | - Taylor J Bader
- Cumming School of Medicine, McCaig Institute for Bone and Joint Health, University of Calgary, Calgary, Alberta, Canada
| | - C Ryan Martin
- Cumming School of Medicine, McCaig Institute for Bone and Joint Health, University of Calgary, Calgary, Alberta, Canada.,Department of Surgery, University of Calgary, Calgary, Alberta, Canada
| | - Nigel G Shrive
- Cumming School of Medicine, McCaig Institute for Bone and Joint Health, University of Calgary, Calgary, Alberta, Canada.,Schulich School of Engineering, University of Calgary, Calgary, Alberta, Canada
| | - David A Hart
- Cumming School of Medicine, McCaig Institute for Bone and Joint Health, University of Calgary, Calgary, Alberta, Canada.,Department of Surgery, University of Calgary, Calgary, Alberta, Canada.,Faculty of Kinesiology, University of Calgary, Calgary, Alberta, Canada.,Alberta Health Services Bone and Joint Health Strategic Clinical Networks, Calgary, Alberta, Canada
| |
Collapse
|
50
|
Cancienne JM, Kew ME, Smith MK, Carson EW, Miller MD, Werner BC. The Timing of Corticosteroid Injections Following Simple Knee Arthroscopy Is Associated With Infection Risk. Arthroscopy 2019; 35:1688-1694. [PMID: 31027921 DOI: 10.1016/j.arthro.2019.01.025] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/11/2018] [Revised: 01/03/2019] [Accepted: 01/09/2019] [Indexed: 02/02/2023]
Abstract
PURPOSE To examine any association between the timing of ipsilateral postoperative corticosteroid injection following simple knee arthroscopy and infection. METHODS Private payer (PP) and Medicare (MC) national insurance databases were queried for patients who underwent simple arthroscopic knee procedures. Patients undergoing concomitant open or more complex procedures with grafts were excluded. Patients who underwent ipsilateral corticosteroid injections within 2, 4, 6, and 8 weeks postoperatively were then identified. Postoperative infection within 90 days after the injection was assessed using International Classification of Diseases, 9th Revision, and Current Procedural Terminology coding and compared using a multivariate binomial logistic regression analysis. RESULTS A total of 5,533 patients were identified, including 725 that received an injection within 2 weeks; 1,236 patients within 4 weeks; 1,716 patients within 6 weeks; and 1,856 patients that received an injection within 8 weeks postoperatively. In both the PP and MC datasets, the rate of infection was significantly higher in the 2-week group compared with the 6- (PP: odds ratio [OR] 3.81, P = .012; MC: OR 9.36, P = .001) and 8-week (PP: OR 8.59, P = .003; MC: OR 7.80, P = .001) groups. The rate of infection was also higher in the 4-week group compared with the 6- (PP: OR 2.54, P = .024; MC: OR 8.91, P = .001) and 8-week (PP: OR 5.64, P = .009; MC: OR 7.80, P = .001) groups. There was no difference in infection rates between the 2- and 4-week groups in either dataset (PP: P = .278; MC: P = .861). CONCLUSIONS There is a significant association between intra-articular knee corticosteroid injections within 4 weeks of surgery and an increased incidence of postoperative infection in both MC and PP patients after knee arthroscopy compared with patients with steroid injections more than 4 weeks postoperatively and matched controls who did not receive injections. LEVEL OF EVIDENCE Level III, retrospective comparative study.
Collapse
Affiliation(s)
- Jourdan M Cancienne
- Department of Orthopaedic Surgery, University of Virginia Health System, Charlottesville, Virginia, U.S.A
| | - Michelle E Kew
- Department of Orthopaedic Surgery, University of Virginia Health System, Charlottesville, Virginia, U.S.A
| | - Marvin K Smith
- Department of Orthopaedic Surgery, University of Virginia Health System, Charlottesville, Virginia, U.S.A
| | - Eric W Carson
- Department of Orthopaedic Surgery, University of Virginia Health System, Charlottesville, Virginia, U.S.A
| | - Mark D Miller
- Department of Orthopaedic Surgery, University of Virginia Health System, Charlottesville, Virginia, U.S.A
| | - Brian C Werner
- Department of Orthopaedic Surgery, University of Virginia Health System, Charlottesville, Virginia, U.S.A..
| |
Collapse
|