1
|
Zhang X, Wu D, Zhang L, Zhang H, Yang L, Wei L, Mei H, Luo L, Jiang Z, Huang C. Predicting the potential mechanism of radix chimonanthi pracecocis in treating osteoarthritis by network pharmacology analysis combined with experimental validation. JOURNAL OF ETHNOPHARMACOLOGY 2024; 331:118231. [PMID: 38718891 DOI: 10.1016/j.jep.2024.118231] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Revised: 04/11/2024] [Accepted: 04/18/2024] [Indexed: 05/19/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Radix Chimonanthi Pracecocis (RCP), also known as Tiekuaizi, widely used by the Miao community in Guizhou, exhibits diverse biological activities and holds promise for the treatment of osteoarthritis (OA). However, there is a lack of contemporary pharmacological research in this area. AIMS OF THE STUDY This study aims to explore the potential of targets and mechanisms of RCP in the treatment of OA. MATERIALS AND METHODS The chemical components of RCP were identified using UPLC-MS/MS, and active components were determined based on the Lipinski rule. RCP and OA-related targets were retrieved from public databases such as TCMSP and GeneCards. Network pharmacology approaches were employed to identify key genes. The limma package (version 3.40.2) in R 4.3.2 was used to screen for differentially expressed genes (DEGs) between OA and healthy individuals in GSE82107. DEGs were analyzed using an independent sample t-test and receiver operating characteristic analysis in GraphPad Prism 9.5.1. Additionally, molecular docking (SYBYL2.1.1) was used to analyze the binding interactions between the active components and target proteins. Finally, we established a papain-induced osteoarthritis (OA) rat model and treated it with RCP aqueous extract by gavage. We validated relevant indicators using real-time fluorescence quantitative polymerase chain reaction, Western blot, immunohistochemistry, and enzyme-linked immunosorbent assays. RESULTS Seven active components and 53 targets were identified. The results of GO and KEGG enrichment analyses confirmed the significant role of RCP in the regulation of pyroptosis. Hypoxia-inducible factor-1α (HIF-1α) was identified as a key gene involved in the main biological functions. Molecular docking analysis revealed that Praecoxin, Isofraxidin, Esculin, and Naringenin can bind to the nucleotide-binding domain, leucine-rich repeat, and pyrin domain-containing protein 3 (NLRP3) (T-Score >5). Additionally, Praecoxin can bind to HIF-1α (T-Score >5). In vivo experiments demonstrated that RCP significantly affects the NLRP3 inflammasome, which is regulated by the HIF-1α pathway. RCP inhibited pyroptosis and reduced synovial inflammation. CONCLUSIONS This study confirmed the efficacy of RCP aqueous extract in the treatment of OA and identified seven active components (esculin, dihydrokaempferol, naringenin, praecoxin, carnosol, hydroxyvalerenic acid, isofraxidin) that may play an anti-pyroptosis role in the treatment of OA by downregulating the expression of HIF-1α and NLRP3 inflammasome.
Collapse
Affiliation(s)
- Xudong Zhang
- Guizhou University of Traditional Chinese Medicine, Guiyang, 550025, China
| | - Dongwen Wu
- Guizhou University of Traditional Chinese Medicine, Guiyang, 550025, China
| | - Lukai Zhang
- Hangzhou Xiaoshan District Chinese Medicine Hospital, Hangzhou, 311200, China
| | - Hongyan Zhang
- Guizhou University of Traditional Chinese Medicine, Guiyang, 550025, China
| | - Liping Yang
- Guizhou University of Traditional Chinese Medicine, Guiyang, 550025, China
| | - Li Wei
- Guizhou University of Traditional Chinese Medicine, Guiyang, 550025, China
| | - Huimin Mei
- Guizhou University of Traditional Chinese Medicine, Guiyang, 550025, China
| | - Liying Luo
- Guizhou University of Traditional Chinese Medicine, Guiyang, 550025, China
| | - Zong Jiang
- Guizhou University of Traditional Chinese Medicine, Guiyang, 550025, China
| | - Cong Huang
- Guizhou University of Traditional Chinese Medicine, Guiyang, 550025, China; Anshun Hospital of Traditional Chinese Medicine, Anshun, 561000, China.
| |
Collapse
|
2
|
Lin YY, Jbeily EH, Tjandra PM, Pride MC, Lopez-Torres M, Elmankabadi SB, Delman CM, Biris KK, Bang H, Silverman JL, Lee CA, Christiansen BA. Surgical restabilization reduces the progression of post-traumatic osteoarthritis initiated by ACL rupture in mice. Osteoarthritis Cartilage 2024; 32:909-920. [PMID: 38697509 DOI: 10.1016/j.joca.2024.04.013] [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: 04/26/2023] [Revised: 02/29/2024] [Accepted: 04/02/2024] [Indexed: 05/05/2024]
Abstract
OBJECTIVE People who sustain joint injuries such as anterior cruciate ligament (ACL) rupture often develop post-traumatic osteoarthritis (PTOA). In human patients, ACL injuries are often treated with ACL reconstruction. However, it is still unclear how effective joint restabilization is for reducing the progression of PTOA. The goal of this study was to determine how surgical restabilization of a mouse knee joint following non-invasive ACL injury affects PTOA progression. DESIGN In this study, 187 mice were subjected to non-invasive ACL injury or no injury. After injury, mice underwent restabilization surgery, sham surgery, or no surgery. Mice were then euthanized on day 14 or day 49 after injury/surgery. Functional analyses were performed at multiple time points to assess voluntary movement, gait, and pain. Knees were analyzed ex vivo with micro-computed tomography, RT-PCR, and whole-joint histology to assess articular cartilage degeneration, synovitis, and osteophyte formation. RESULTS Both ACL injury and surgery resulted in loss of epiphyseal trabecular bone (-27-32%) and reduced voluntary movement at early time points. Joint restabilization successfully lowered OA score (-78% relative to injured at day 14, p < 0.0001), and synovitis scores (-37% relative to injured at day 14, p = 0.042), and diminished the formation of chondrophytes/osteophytes (-97% relative to injured at day 14, p < 0.001, -78% at day 49, p < 0.001). CONCLUSIONS This study confirmed that surgical knee restabilization was effective at reducing articular cartilage degeneration and diminishing chondrophyte/osteophyte formation after ACL injury in mice, suggesting that these processes are largely driven by joint instability in this mouse model. However, restabilization was not able to mitigate the early inflammatory response and the loss of epiphyseal trabecular bone, indicating that these processes are independent of joint instability.
Collapse
Affiliation(s)
- Yu-Yang Lin
- University of California Davis Health, Department of Orthopaedic Surgery, Lawrence J. Ellison Musculoskeletal Research Center, 2700 Stockton Blvd, Suite 2301, Sacramento, CA 95817, USA
| | - Elias H Jbeily
- University of California Davis Health, Department of Orthopaedic Surgery, Lawrence J. Ellison Musculoskeletal Research Center, 2700 Stockton Blvd, Suite 2301, Sacramento, CA 95817, USA
| | - Priscilla M Tjandra
- University of California Davis Health, Department of Orthopaedic Surgery, Lawrence J. Ellison Musculoskeletal Research Center, 2700 Stockton Blvd, Suite 2301, Sacramento, CA 95817, USA
| | - Michael C Pride
- University of California Davis Health, Department of Psychiatry and Behavioral Sciences, 4625 2nd Ave, Sacramento, CA 95817, USA
| | - Michael Lopez-Torres
- University of California Davis Health, Department of Orthopaedic Surgery, Lawrence J. Ellison Musculoskeletal Research Center, 2700 Stockton Blvd, Suite 2301, Sacramento, CA 95817, USA
| | - Seif B Elmankabadi
- University of California Davis Health, Department of Orthopaedic Surgery, Lawrence J. Ellison Musculoskeletal Research Center, 2700 Stockton Blvd, Suite 2301, Sacramento, CA 95817, USA
| | - Connor M Delman
- University of California Davis Health, Department of Orthopaedic Surgery, Lawrence J. Ellison Musculoskeletal Research Center, 2700 Stockton Blvd, Suite 2301, Sacramento, CA 95817, USA
| | - Kristin K Biris
- University of California Davis Health, Department of Orthopaedic Surgery, Lawrence J. Ellison Musculoskeletal Research Center, 2700 Stockton Blvd, Suite 2301, Sacramento, CA 95817, USA
| | - Heejung Bang
- University of California Davis Health, Department of Public Health Sciences, Medical Sciences 1C, Davis, CA 95616, USA
| | - Jill L Silverman
- University of California Davis Health, Department of Psychiatry and Behavioral Sciences, 4625 2nd Ave, Sacramento, CA 95817, USA
| | - Cassandra A Lee
- University of California Davis Health, Department of Orthopaedic Surgery, Lawrence J. Ellison Musculoskeletal Research Center, 2700 Stockton Blvd, Suite 2301, Sacramento, CA 95817, USA
| | - Blaine A Christiansen
- University of California Davis Health, Department of Orthopaedic Surgery, Lawrence J. Ellison Musculoskeletal Research Center, 2700 Stockton Blvd, Suite 2301, Sacramento, CA 95817, USA.
| |
Collapse
|
3
|
Kraus VB, Hsueh MF. Molecular biomarker approaches to prevention of post-traumatic osteoarthritis. Nat Rev Rheumatol 2024; 20:272-289. [PMID: 38605249 DOI: 10.1038/s41584-024-01102-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/05/2024] [Indexed: 04/13/2024]
Abstract
Up to 50% of individuals develop post-traumatic osteoarthritis (PTOA) within 10 years following knee-joint injuries such as anterior cruciate ligament rupture or acute meniscal tear. Lower-extremity PTOA prevalence is estimated to account for ≥12% of all symptomatic osteoarthritis (OA), or approximately 5.6 million cases in the USA. With knowledge of the inciting event, it might be possible to 'catch PTOA in the act' with sensitive imaging and soluble biomarkers and thereby prevent OA sequelae by early intervention. Existing biomarker data in the joint-injury literature can provide insights into the pathogenesis and early risk trajectory related to PTOA and can help to elucidate a research agenda for preventing or slowing the onset of PTOA. Non-traumatic OA and PTOA have many clinical, radiological and genetic similarities, and efforts to understand early risk trajectories in PTOA might therefore contribute to the identification and classification of early non-traumatic OA, which is the most prevalent form of OA.
Collapse
Affiliation(s)
- Virginia Byers Kraus
- Duke Molecular Physiology Institute, Duke University, Durham, NC, USA.
- Department of Medicine, Duke University School of Medicine, Durham, NC, USA.
- Department of Orthopaedic Surgery, Duke University School of Medicine, Durham, NC, USA.
| | - Ming-Feng Hsueh
- Duke Molecular Physiology Institute, Duke University, Durham, NC, USA
- Department of Orthopaedic Surgery, Duke University School of Medicine, Durham, NC, USA
| |
Collapse
|
4
|
Han D, Wang W, Gong J, Ma Y, Li Y. Microbiota metabolites in bone: Shaping health and Confronting disease. Heliyon 2024; 10:e28435. [PMID: 38560225 PMCID: PMC10979239 DOI: 10.1016/j.heliyon.2024.e28435] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2023] [Revised: 02/16/2024] [Accepted: 03/19/2024] [Indexed: 04/04/2024] Open
Abstract
The intricate interplay between the gut microbiota and bone health has become increasingly recognized as a fundamental determinant of skeletal well-being. Microbiota-derived metabolites play a crucial role in dynamic interaction, specifically in bone homeostasis. In this sense, short-chain fatty acids (SCFAs), including acetate, propionate, and butyrate, indirectly promote bone formation by regulating insulin-like growth factor-1 (IGF-1). Trimethylamine N-oxide (TMAO) has been found to increase the expression of osteoblast genes, such as Runt-related transcription factor 2 (RUNX2) and bone morphogenetic protein-2 (BMP2), thus enhancing osteogenic differentiation and bone quality through BMP/SMADs and Wnt signaling pathways. Remarkably, in the context of bone infections, the role of microbiota metabolites in immune modulation and host defense mechanisms potentially affects susceptibility to infections such as osteomyelitis. Furthermore, ongoing research elucidates the precise mechanisms through which microbiota-derived metabolites influence bone cells, such as osteoblasts and osteoclasts. Understanding the multifaceted influence of microbiota metabolites on bone, from regulating homeostasis to modulating susceptibility to infections, has the potential to revolutionize our approach to bone health and disease management. This review offers a comprehensive exploration of this evolving field, providing a holistic perspective on the impact of microbiota metabolites on bone health and diseases.
Collapse
Affiliation(s)
- Dong Han
- Department of Trauma Orthopedics, Yantaishan Hospital, Yantai 264000, China
| | - Weijiao Wang
- Department of Otolaryngology, Yantaishan Hospital, Yantai 264000, China
| | - Jinpeng Gong
- Department of Trauma Orthopedics, Yantaishan Hospital, Yantai 264000, China
| | - Yupeng Ma
- Department of Trauma Orthopedics, Yantaishan Hospital, Yantai 264000, China
| | - Yu Li
- Department of Trauma Orthopedics, Yantaishan Hospital, Yantai 264000, China
| |
Collapse
|
5
|
Prinz E, Schlupp L, Dyson G, Barrett M, Szymczak A, Velasco C, Izda V, Dunn CM, Jeffries MA. OA susceptibility in mice is partially mediated by the gut microbiome, is transferrable via microbiome transplantation and is associated with immunophenotype changes. Ann Rheum Dis 2024; 83:382-393. [PMID: 37979958 PMCID: PMC10922159 DOI: 10.1136/ard-2023-224907] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Accepted: 11/03/2023] [Indexed: 11/20/2023]
Abstract
OBJECTIVES The Murphy Roths Large (MRL)/MpJ 'superhealer' mouse strain is protected from post-traumatic osteoarthritis (OA), although no studies have evaluated the microbiome in the context of this protection. This study characterised microbiome differences between MRL and wild-type mice, evaluated microbiome transplantation and OA and investigated microbiome-associated immunophenotypes. METHODS Cecal material from mixed sex C57BL6/J (B6) or female MRL/MpJ (MRL) was transplanted into B6 and MRL mice, then OA was induced by disruption of the medial meniscus surgery (DMM). In other experiments, transplantation was performed after DMM and transplantation was performed into germ-free mice. Transplanted mice were bred through F2. OARSI, synovitis and osteophyte scores were determined blindly 8 weeks after DMM. 16S microbiome sequencing was performed and metagenomic function was imputed. Immunophenotypes were determined using mass cytometry. RESULTS MRL-into-B6 transplant prior to DMM showed reduced OA histopathology (OARSI score 70% lower transplant vs B6 control), synovitis (60% reduction) and osteophyte scores (30% reduction) 8 weeks after DMM. When performed 48 hours after DMM, MRL-into-B6 transplant improved OA outcomes but not when performed 1-2 weeks after DMM. Protection was seen in F1 (60% reduction) and F2 progeny (30% reduction). Several cecal microbiome clades were correlated with either better (eg, Lactobacillus, R=-0.32, p=0.02) or worse (eg, Rikenellaceae, R=0.43, p=0.001) OA outcomes. Baseline immunophenotypes associated with MRL-into-B6 transplants and MRL included reduced double-negative T cells and increased CD25+CD4+ T cells. CONCLUSION The gut microbiome is responsible in part for OA protection in MRL mice and is transferrable by microbiome transplantation. Transplantation induces resting systemic immunophenotyping changes that correlate with OA protection.
Collapse
Affiliation(s)
- Emmaline Prinz
- Arthritis & Clinical Immunology Program, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma, USA
- Division of Rheumatology, Immunology, and Allergy, The University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, USA
| | - Leoni Schlupp
- Arthritis & Clinical Immunology Program, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma, USA
| | - Gabby Dyson
- Arthritis & Clinical Immunology Program, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma, USA
| | - Montana Barrett
- Arthritis & Clinical Immunology Program, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma, USA
| | - Aleksander Szymczak
- Arthritis & Clinical Immunology Program, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma, USA
| | - Cassandra Velasco
- Division of Rheumatology, Immunology, and Allergy, The University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, USA
| | - Vladislav Izda
- Arthritis & Clinical Immunology Program, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma, USA
| | - Christopher M Dunn
- Arthritis & Clinical Immunology Program, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma, USA
- Division of Rheumatology, Immunology, and Allergy, The University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, USA
| | - Matlock A Jeffries
- Arthritis & Clinical Immunology Program, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma, USA
- Division of Rheumatology, Immunology, and Allergy, The University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, USA
| |
Collapse
|
6
|
黄 婉, 涂 君, 乔 爱, 何 储. [Effects of VX765 on osteoarthritis and chondrocyte inflammation in rats]. ZHONGGUO XIU FU CHONG JIAN WAI KE ZA ZHI = ZHONGGUO XIUFU CHONGJIAN WAIKE ZAZHI = CHINESE JOURNAL OF REPARATIVE AND RECONSTRUCTIVE SURGERY 2024; 38:74-81. [PMID: 38225845 PMCID: PMC10796219 DOI: 10.7507/1002-1892.202308053] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Revised: 12/27/2023] [Indexed: 01/17/2024]
Abstract
Objective To investigate the effects and underlying mechanisms of VX765 on osteoarthritis (OA) and chondrocytes inflammation in rats. Methods Chondrocytes were isolated from the knee joints of 4-week-old Sprague Dawley (SD) rats. The third-generation cells were subjected to cell counting kit 8 (CCK-8) analysis to assess the impact of various concentrations (0, 1, 5, 10, 20, 50, 100 μmol/L) of VX765 on rat chondrocyte activity. An in vitro lipopolysaccharide (LPS) induced cell inflammation model was employed, dividing cells into control group, LPS group, VX765 concentration 1 group and VX765 concentration 2 group without obvious cytotoxicity. Western blot, real-time fluorescence quantitative PCR, and ELISA were conducted to measure the expression levels of inflammatory factors-transforming growth factor β 1 (TGF-β 1), interleukin 6 (IL-6), and tumor necrosis factor α (TNF-α). Additionally, Western blot and immunofluorescence staining were employed to assess the expressions of nuclear factor erythroid 2-related factor 2 (Nrf2) and heme oxygenase 1 (HO-1). Thirty-two SD rats were randomly assigned to sham surgery group (group A), OA group (group B), OA+VX765 (50 mg/kg) group (group C), and OA+VX765 (100 mg/kg) group (group D), with 8 rats in each group. Group A underwent a sham operation with a medial incision, while groups B to D underwent additional transverse incisions to the medial collateral ligament and anterior cruciate ligament, with removal of the medial meniscus. One week post-surgery, groups C and D were orally administered 50 mg/kg and 100 mg/kg VX765, respectively, while groups A and B received an equivalent volume of saline. Histopathological examination using HE and safranin-fast green staining was performed, and Mankin scoring was utilized for evaluation. Immunohistochemical staining technique was employed to analyze the expressions of matrix metalloproteinase 13 (MMP-13) and collagen type Ⅱ. Results The CCK-8 assay indicated a significant decrease in cell viability at VX765 concentrations exceeding 10 μmol/L ( P<0.05), so 4 μmol/L and 8 μmol/L VX765 without obvious cytotoxicity were selected for subsequent experiments. Following LPS induction, the expressions of TGF-β 1, IL-6, and TNF-α in cells significantly increased when compared with the control group ( P<0.05). However, intervention with 4 μmol/L and 8 μmol/L VX765 led to a significant decrease in expression compared to the LPS group ( P<0.05). Western blot and immunofluorescence staining demonstrated a significant upregulation of Nrf2 pathway-related molecules Nrf2 and HO-1 protein expressions by VX765 ( P<0.05), indicating Nrf2 pathway activation. Histopathological examination of rat knee joint tissues and immunohistochemical staining revealed that, compared to group B, treatment with VX765 in groups C and D improved joint structural damage in rat OA, alleviated inflammatory reactions, downregulated MMP-13 expression, and increased collagen type Ⅱ expression. Conclusion VX765 can improve rat OA and reduce chondrocyte inflammation, possibly through the activation of the Nrf2 pathway.
Collapse
Affiliation(s)
- 婉然 黄
- 温州医科大学附属第二医院育英儿童医院药学部(浙江温州 325000)Department of Pharmacy, Yuying Children’s Hospital, the Second Affiliated Hospital of Wenzhou Medical University, Wenzhou Zhejiang, 325000, P. R. China
| | - 君雪 涂
- 温州医科大学附属第二医院育英儿童医院药学部(浙江温州 325000)Department of Pharmacy, Yuying Children’s Hospital, the Second Affiliated Hospital of Wenzhou Medical University, Wenzhou Zhejiang, 325000, P. R. China
| | - 爱卿 乔
- 温州医科大学附属第二医院育英儿童医院药学部(浙江温州 325000)Department of Pharmacy, Yuying Children’s Hospital, the Second Affiliated Hospital of Wenzhou Medical University, Wenzhou Zhejiang, 325000, P. R. China
| | - 储君 何
- 温州医科大学附属第二医院育英儿童医院药学部(浙江温州 325000)Department of Pharmacy, Yuying Children’s Hospital, the Second Affiliated Hospital of Wenzhou Medical University, Wenzhou Zhejiang, 325000, P. R. China
| |
Collapse
|
7
|
Ren T, Yin N, Du L, Pan M, Ding L. Identification and validation of FPR1, FPR2, IL17RA and TLR7 as immunogenic cell death related genes in osteoarthritis. Sci Rep 2023; 13:16872. [PMID: 37803031 PMCID: PMC10558501 DOI: 10.1038/s41598-023-43440-z] [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: 04/03/2023] [Accepted: 09/24/2023] [Indexed: 10/08/2023] Open
Abstract
Immunogenic cell death (ICDs) has gained increasing attention for its significant clinical efficacy in various diseases. Similarly, more and more attention has been paid in the role of immune factors in the pathological process of osteoarthritis (OA). The objective of this study is to reveal the relationship between ICD-related genes and the process of OA at the gene level through bioinformatics analysis. In this study, Limma R package was applied to identify differentially expressed genes (DEG), and OA related module genes were determined by weighted gene co-expression network analysis. The ICD-related genes were extracted from a previous study. The module genes related to DEGs and ICD were overlapped. Then, hub genes were identified by a series of analyses using the Least absolute shrinkage and selection operator and random forest algorithm, the expression level and diagnostic value of hub genes were evaluated by Logistic regression. In addition, we used Spearman rank correlation analysis to clarify the relationship between hub genes and infiltrating immune cells and immune pathways. The expression levels of FPR1, FPR2, IL17RA, and TLR7 was verified in SD rat knee joint model of OA by immunohistochemistry. The expression levels of FPR1, FPR2, IL17RA, and TLR7 mRNA were detected in the IL-1β induced rat chondrocytes in qPCR experiment in vitro. Four hub genes (FPR1, FPR2, IL17RA, and TLR7) were ultimately identified as OA biomarkers associated with ICD. And knockdown of TLR7 reversed collagen II and ADAMTS-5 degradation in IL-1β-stimulated chondrocytes. This research may provide new immune related biomarkers for the diagnosis of OA and serve as a reference for disease treatment monitoring.
Collapse
Affiliation(s)
- Tingting Ren
- Department of Critical Care Medicine, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, 200127, China
| | - Nuo Yin
- Department of Orthopaedics, Shanghai Jiao Tong University Affiliated Sixth People's Hospital South Campus, Shanghai, 201400, China
| | - Li Du
- Department of Orthopaedics, Shanghai Jiao Tong University Affiliated Sixth People's Hospital South Campus, Shanghai, 201400, China
| | - Mingmang Pan
- Department of Orthopaedics, Shanghai Jiao Tong University Affiliated Sixth People's Hospital South Campus, Shanghai, 201400, China
| | - Liang Ding
- Department of Orthopaedics, Shanghai Jiao Tong University Affiliated Sixth People's Hospital South Campus, Shanghai, 201400, China.
| |
Collapse
|
8
|
Mendez ME, Murugesh DK, Christiansen BA, Loots GG. Antibiotic Treatment Prior to Injury Abrogates the Detrimental Effects of LPS in STR/ort Mice Susceptible to Osteoarthritis Development. JBMR Plus 2023; 7:e10759. [PMID: 37614305 PMCID: PMC10443070 DOI: 10.1002/jbm4.10759] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Revised: 04/08/2023] [Accepted: 04/12/2023] [Indexed: 08/25/2023] Open
Abstract
Post traumatic osteoarthritis (PTOA) is a form of secondary osteoarthritis (OA) that develops in ~50% of cases of severe articular joint injuries and leads to chronic and progressive degradation of articular cartilage and other joint tissues. PTOA progression can be exacerbated by repeated injury and systemic inflammation. Few studies have examined approaches for blunting or slowing down PTOA progression with emphasis on systemic inflammation; most arthritis studies focused on the immune system have been in the context of rheumatoid arthritis. To examine how the gut microbiome affects systemic inflammation during PTOA development, we used a chronic antibiotic treatment regimen starting at weaning for 6 weeks before anterior cruciate ligament (ACL) rupture in STR/ort mice combined with lipopolysaccharide (LPS)-induced systemic inflammation. STR/ort mice develop spontaneous OA as well as a more severe PTOA phenotype than C57Bl/6J mice. By 6 weeks post injury, histological examination showed a more robust cartilage staining in the antibiotic-treated (AB) STR/ort mice than in the untreated STR/ort controls. Furthermore, we also examined the effects of AB treatment on systemic inflammation and found that the effects of LPS administration before injury are also blunted by AB treatment in STR/ort mice. The AB- or AB+LPS-treated STR/ort injured joints more closely resembled the C57Bl/6J VEH OA phenotypes than the vehicle- or LPS-treated STR/ort, suggesting that antibiotic treatment has the potential to slow disease progression and should be further explored therapeutically as prophylactic post injury. © 2023 The Authors. JBMR Plus published by Wiley Periodicals LLC on behalf of American Society for Bone and Mineral Research.
Collapse
Affiliation(s)
- Melanie E Mendez
- Lawrence Livermore National Laboratories, Physical and Life Sciences DirectorateLivermoreCAUSA
| | - Deepa K Murugesh
- Lawrence Livermore National Laboratories, Physical and Life Sciences DirectorateLivermoreCAUSA
| | - Blaine A Christiansen
- Department of Orthopaedic SurgeryUniversity of California Davis HealthSacramentoCAUSA
| | - Gabriela G Loots
- Lawrence Livermore National Laboratories, Physical and Life Sciences DirectorateLivermoreCAUSA
- Department of Orthopaedic SurgeryUniversity of California Davis HealthSacramentoCAUSA
| |
Collapse
|
9
|
Dai W, Jin P, Li X, Zhao J, Lan Y, Li H, Zheng L. A carrier-free nano-drug assembled via π-π stacking interaction for the treatment of osteoarthritis. Biomed Pharmacother 2023; 164:114881. [PMID: 37210895 DOI: 10.1016/j.biopha.2023.114881] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Revised: 05/05/2023] [Accepted: 05/12/2023] [Indexed: 05/23/2023] Open
Abstract
Osteoarthritis (OA) is considered to be the most common joint disorder. Exogenous drug intervention is one of the effective means for OA treatment. Clinical applications of numerous drugs are restricted owing to the short retention as well as rapid clearance in the joint cavity. A wide variety of carrier-based nanodrugs have been developed, but additional carriers may bring unexpected side effects or even toxicity. Herein, by exploiting the spontaneous fluorescence of Curcumin, we designed a new carrier-free self-assembly nanomedicine Curcumin (Cur)/icariin (ICA) nanoparticles with adjustable particle size, which is composed of two small-molecule natural drugs assembled via π-π stacking interaction. Experimental results revealed that Cur/ICA NPs endowed with little cytotoxicity, high cellular uptake and sustained drug release, could inhibit secretion of inflammatory cytokines and reduce cartilage degeneration. Moreover, both the in vitro and in vivo experiments showed the NPs exerted superior synergism effects in anti-inflammatory and cartilage protection than either Cur or ICA alone, and self-monitored its retention by autofluorescence. Thus, the new self-assembly nano-drug combining Cur and ICA represents a new strategy for the treatment of osteoarthritis.
Collapse
Affiliation(s)
- Wanwu Dai
- Guangxi Engineering Center in Biomedical Materials for Tissue and Organ Regeneration & Collaborative Innovation Center of Regenerative Medicine and Medical BioResource Development and Application Co-constructed by the Province and Ministry, Life Sciences Institute, Guangxi Medical University, Nanning 530021, China; Department of Bone and Joint Surgery, The Third Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530031, China
| | - Pan Jin
- Guangxi Engineering Center in Biomedical Materials for Tissue and Organ Regeneration & Collaborative Innovation Center of Regenerative Medicine and Medical BioResource Development and Application Co-constructed by the Province and Ministry, Life Sciences Institute, Guangxi Medical University, Nanning 530021, China
| | - Xingyan Li
- Department of Bone and Joint Surgery, The Third Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530031, China
| | - Jinmin Zhao
- Guangxi Engineering Center in Biomedical Materials for Tissue and Organ Regeneration & Collaborative Innovation Center of Regenerative Medicine and Medical BioResource Development and Application Co-constructed by the Province and Ministry, Life Sciences Institute, Guangxi Medical University, Nanning 530021, China
| | - Ying Lan
- Guangxi Engineering Center in Biomedical Materials for Tissue and Organ Regeneration & Collaborative Innovation Center of Regenerative Medicine and Medical BioResource Development and Application Co-constructed by the Province and Ministry, Life Sciences Institute, Guangxi Medical University, Nanning 530021, China.
| | - Hongmian Li
- Department of Plastic and Reconstructive Surgery, The People's Hospital of Guangxi Zhuang Autonomous Region & Research Center of Medical Sciences, Guangxi Academy of Medical Sciences, Nanning 530021, China.
| | - Li Zheng
- Guangxi Engineering Center in Biomedical Materials for Tissue and Organ Regeneration & Collaborative Innovation Center of Regenerative Medicine and Medical BioResource Development and Application Co-constructed by the Province and Ministry, Life Sciences Institute, Guangxi Medical University, Nanning 530021, China; Guangxi Key Laboratory of Regenerative Medicine, Department of Orthopedics, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, China.
| |
Collapse
|
10
|
Feng Z, Huang Q, Zhang X, Xu P, Li S, Ma D, Meng Q. PPAR-γ Activation Alleviates Osteoarthritis through Both the Nrf2/NLRP3 and PGC-1α/Δψm Pathways by Inhibiting Pyroptosis. PPAR Res 2023; 2023:2523536. [PMID: 37020714 PMCID: PMC10070030 DOI: 10.1155/2023/2523536] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2022] [Revised: 02/10/2023] [Accepted: 02/27/2023] [Indexed: 03/30/2023] Open
Abstract
Osteoarthritis (OA) is a common degenerative joint disease with a gradually increasing morbidity in the aging and obese population. Emerging evidence has implicated pyroptosis in the etiology of OA and it may be recognized as a therapeutic target in OA. We have previously reported regarding another disease that peroxisome proliferator-activated receptor gamma (PPAR-γ) activation exerts an anti-inflammatory effect by suppressing the nucleotide-binding and oligomerization domain-like receptor containing protein (NLRP) 3 inflammasome. However, the relationship between PPAR-γ and NLRP3-mediated pyroptosis in OA cartilage and its underlying mechanisms is fully unclear. In this study, we found that the level of NLRP3-mediated pyroptosis in severe lateral femoral condyle cartilage wear in the knee of an OA patient was significantly higher than that in the mild lateral femoral condyle cartilage wear areas. Moreover, in lipopolysaccharide (LPS)/adenosine triphosphate (ATP)-induced primary chondrocytes and knee OA rat models, we demonstrated that activation of PPAR-γ by pioglitazone (Piog) attenuated LPS/ATP-induced chondrocyte pyroptosis and arthritis. These effects were partially counteracted by either blocking the nuclear factor erythroid-2-related factor (Nrf2)/NLRP3 or PGC1-α/Δψm signaling pathway. Simultaneous depression of these two signaling pathways can completely abrogate the protective effects of Piog on OA and chondrocytes. Taken together, Piog protects OA cartilage against pyroptosis-induced damage by simultaneously activating both the Nrf2/NLRP3 and PGC-1α/Δψm pathways, which enhances antioxidative and anti-inflammatory responses as well as mitochondrial biogenesis. Therefore, Piog may be a promising agent for human OA cartilage damage in future clinical treatments.
Collapse
|
11
|
Reddiar SB, de Veer M, Paterson BM, Sepehrizadeh T, Wai DCC, Csoti A, Panyi G, Nicolazzo JA, Norton RS. A Biodistribution Study of the Radiolabeled Kv1.3-Blocking Peptide DOTA-HsTX1[R14A] Demonstrates Brain Uptake in a Mouse Model of Neuroinflammation. Mol Pharm 2023; 20:255-266. [PMID: 36331024 DOI: 10.1021/acs.molpharmaceut.2c00614] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
The voltage-gated potassium channel Kv1.3 regulates the pro-inflammatory function of microglia and is highly expressed in the post-mortem brains of individuals with Alzheimer's and Parkinson's diseases. HsTX1[R14A] is a selective and potent peptide inhibitor of the Kv1.3 channel (IC50 ∼ 45 pM) that has been shown to decrease cytokine levels in a lipopolysaccharide (LPS)-induced mouse model of inflammation. Central nervous system exposure to HsTX1[R14A] was previously detected in this mouse model using liquid chromatography with tandem mass spectrometry, but this technique does not report on the spatial distribution of the peptide in the different brain regions or peripheral organs. Herein, the in vivo distribution of a [64Cu]Cu-labeled DOTA conjugate of HsTX1[R14A] was observed for up to 48 h by positron emission tomography (PET) in mice. After subcutaneous administration to untreated C57BL/6J mice, considerable uptake of the radiolabeled peptide was observed in the kidney, but it was undetectable in the brain. Biodistribution of a [68Ga]Ga-DOTA conjugate of HsTX1[R14A] was then investigated in the LPS-induced mouse model of neuroinflammation to assess the effects of inflammation on uptake of the peptide in the brain. A control peptide with very weak Kv1.3 binding, [68Ga]Ga-DOTA-HsTX1[R14A,Y21A,K23A] (IC50 ∼ 6 μM), was also tested. Significantly increased uptake of [68Ga]Ga-DOTA-HsTX1[R14A] was observed in the brains of LPS-treated mice compared to mice treated with control peptide, implying that the enhanced uptake was due to increased Kv1.3 expression rather than simply increased blood-brain barrier disruption. PET imaging also showed accumulation of [68Ga]Ga-DOTA-HsTX1[R14A] in inflamed joints and decreased clearance from the kidneys in LPS-treated mice. These biodistribution data highlight the potential of HsTX1[R14A] as a therapeutic for the treatment of neuroinflammatory diseases mediated by overexpression of Kv1.3.
Collapse
Affiliation(s)
- Sanjeevini Babu Reddiar
- Medicinal Chemistry, Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, Victoria 3052, Australia
| | - Michael de Veer
- Monash Biomedical Imaging, Monash University, Melbourne, Victoria 3800, Australia
| | - Brett M Paterson
- Monash Biomedical Imaging, Monash University, Melbourne, Victoria 3800, Australia.,School of Chemistry, Monash University, Clayton, Victoria 3800, Australia
| | - Tara Sepehrizadeh
- Monash Biomedical Imaging, Monash University, Melbourne, Victoria 3800, Australia
| | - Dorothy C C Wai
- Medicinal Chemistry, Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, Victoria 3052, Australia
| | - Agota Csoti
- Department of Biophysics and Cell Biology, Faculty of Medicine, University of Debrecen, Debrecen 4010, Hungary
| | - Gyorgy Panyi
- Department of Biophysics and Cell Biology, Faculty of Medicine, University of Debrecen, Debrecen 4010, Hungary
| | - Joseph A Nicolazzo
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, Victoria 3052, Australia
| | - Raymond S Norton
- Medicinal Chemistry, Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, Victoria 3052, Australia.,ARC Centre for Fragment-Based Design, Monash University, Parkville, Victoria 3052, Australia
| |
Collapse
|
12
|
Jurynec MJ, Gavile CM, Honeggar M, Ma Y, Veerabhadraiah SR, Novak KA, Hoshijima K, Kazmers NH, Grunwald DJ. NOD/RIPK2 signalling pathway contributes to osteoarthritis susceptibility. Ann Rheum Dis 2022; 81:1465-1473. [PMID: 35732460 PMCID: PMC9474725 DOI: 10.1136/annrheumdis-2022-222497] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Accepted: 06/07/2022] [Indexed: 12/21/2022]
Abstract
OBJECTIVES How inflammatory signalling contributes to osteoarthritis (OA) susceptibility is undetermined. An allele encoding a hyperactive form of the Receptor Interacting Protein Kinase 2 (RIPK2) proinflammatory signalling intermediate has been associated with familial OA. To test whether altered nucleotide-binding oligomerisation domain (NOD)/RIPK2 pathway activity causes heightened OA susceptibility, we investigated whether variants affecting additional pathway components are associated with familial OA. To determine whether the Ripk2104Asp disease allele is sufficient to account for the familial phenotype, we determined the effect of the allele on mice. METHODS Genomic analysis of 150 independent families with dominant inheritance of OA affecting diverse joints was used to identify coding variants that segregated strictly with occurrence of OA. Genome editing was used to introduce the OA-associated RIPK2 (p.Asn104Asp) allele into the genome of inbred mice. The consequences of the Ripk2104Asp disease allele on physiology and OA susceptibility in mice were measured by histology, immunohistochemistry, serum cytokine levels and gene expression. RESULTS We identified six novel variants affecting components of the NOD/RIPK2 inflammatory signalling pathway that are associated with familial OA affecting the hand, shoulder or foot. The Ripk2104Asp allele acts dominantly to alter basal physiology and response to trauma in the mouse knee. Whereas the knees of uninjured Ripk2Asp104 mice appear normal histologically, the joints exhibit a set of marked gene expression changes reminiscent of overt OA. Although the Ripk2104Asp mice lack evidence of chronically elevated systemic inflammation, they do exhibit significantly increased susceptibility to post-traumatic OA (PTOA). CONCLUSIONS Two types of data support the hypothesis that altered NOD/RIPK2 signalling confers susceptibility to OA.
Collapse
Affiliation(s)
- Michael J Jurynec
- Department of Orthopaedics, University of Utah Health, Salt Lake City, Utah, USA
- Department of Human Genetics, University of Utah Health, Salt Lake City, Utah, USA
| | - Catherine M Gavile
- Department of Orthopaedics, University of Utah Health, Salt Lake City, Utah, USA
| | - Matthew Honeggar
- Department of Orthopaedics, University of Utah Health, Salt Lake City, Utah, USA
| | - Ying Ma
- Department of Orthopaedics, University of Utah Health, Salt Lake City, Utah, USA
| | | | - Kendra A Novak
- Department of Orthopaedics, University of Utah Health, Salt Lake City, Utah, USA
| | - Kazuyuki Hoshijima
- Department of Human Genetics, University of Utah Health, Salt Lake City, Utah, USA
| | - Nikolas H Kazmers
- Department of Orthopaedics, University of Utah Health, Salt Lake City, Utah, USA
| | - David J Grunwald
- Department of Human Genetics, University of Utah Health, Salt Lake City, Utah, USA
| |
Collapse
|
13
|
Sebastian A, Hum NR, McCool JL, Wilson SP, Murugesh DK, Martin KA, Rios-Arce ND, Amiri B, Christiansen BA, Loots GG. Single-cell RNA-Seq reveals changes in immune landscape in post-traumatic osteoarthritis. Front Immunol 2022; 13:938075. [PMID: 35967299 PMCID: PMC9373730 DOI: 10.3389/fimmu.2022.938075] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Accepted: 07/06/2022] [Indexed: 12/13/2022] Open
Abstract
Osteoarthritis (OA) is the most common joint disease, affecting over 300 million people world-wide. Accumulating evidence attests to the important roles of the immune system in OA pathogenesis. Understanding the role of various immune cells in joint degeneration or joint repair after injury is vital for improving therapeutic strategies for treating OA. Post-traumatic osteoarthritis (PTOA) develops in ~50% of individuals who have experienced an articular trauma like an anterior cruciate ligament (ACL) rupture. Here, using the high resolution of single-cell RNA sequencing, we delineated the temporal dynamics of immune cell accumulation in the mouse knee joint after ACL rupture. Our study identified multiple immune cell types in the joint including neutrophils, monocytes, macrophages, B cells, T cells, NK cells and dendritic cells. Monocytes and macrophage populations showed the most dramatic changes after injury. Further characterization of monocytes and macrophages reveled 9 major subtypes with unique transcriptomics signatures, including a tissue resident Lyve1hiFolr2hi macrophage population and Trem2hiFcrls+ recruited macrophages, both showing enrichment for phagocytic genes and growth factors such as Igf1, Pdgfa and Pdgfc. We also identified several genes induced or repressed after ACL injury in a cell type-specific manner. This study provides new insight into PTOA-associated changes in the immune microenvironment and highlights macrophage subtypes that may play a role in joint repair after injury.
Collapse
Affiliation(s)
- Aimy Sebastian
- Physical and Life Sciences Directorate, Lawrence Livermore National Laboratory, Livermore, CA, United States
- *Correspondence: Aimy Sebastian, ; Gabriela G. Loots,
| | - Nicholas R. Hum
- Physical and Life Sciences Directorate, Lawrence Livermore National Laboratory, Livermore, CA, United States
| | - Jillian L. McCool
- Physical and Life Sciences Directorate, Lawrence Livermore National Laboratory, Livermore, CA, United States
- School of Natural Sciences, University of California Merced, Merced, CA, United States
| | - Stephen P. Wilson
- Physical and Life Sciences Directorate, Lawrence Livermore National Laboratory, Livermore, CA, United States
| | - Deepa K. Murugesh
- Physical and Life Sciences Directorate, Lawrence Livermore National Laboratory, Livermore, CA, United States
| | - Kelly A. Martin
- Physical and Life Sciences Directorate, Lawrence Livermore National Laboratory, Livermore, CA, United States
| | - Naiomy Deliz Rios-Arce
- Physical and Life Sciences Directorate, Lawrence Livermore National Laboratory, Livermore, CA, United States
| | - Beheshta Amiri
- Physical and Life Sciences Directorate, Lawrence Livermore National Laboratory, Livermore, CA, United States
| | - Blaine A. Christiansen
- Department of Orthopaedic Surgery, University of California Davis Health, Sacramento, CA, United States
| | - Gabriela G. Loots
- Physical and Life Sciences Directorate, Lawrence Livermore National Laboratory, Livermore, CA, United States
- School of Natural Sciences, University of California Merced, Merced, CA, United States
- *Correspondence: Aimy Sebastian, ; Gabriela G. Loots,
| |
Collapse
|
14
|
Cross-Tissue Analysis Using Machine Learning to Identify Novel Biomarkers for Knee Osteoarthritis. COMPUTATIONAL AND MATHEMATICAL METHODS IN MEDICINE 2022; 2022:9043300. [PMID: 35785145 PMCID: PMC9246600 DOI: 10.1155/2022/9043300] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/07/2021] [Revised: 04/28/2022] [Accepted: 05/13/2022] [Indexed: 11/18/2022]
Abstract
Background Knee osteoarthritis (KOA) is a common degenerative joint disease. In this study, we aimed to identify new biomarkers of KOA to improve the accuracy of diagnosis and treatment. Methods GSE98918 and GSE51588 were downloaded from the Gene Expression Omnibus database as training sets, with a total of 74 samples. Gene differences were analyzed by Gene Ontology, Kyoto Encyclopedia of Genes and Genomes pathway, and Disease Ontology enrichment analyses for the differentially expressed genes (DEGs), and GSEA enrichment analysis was carried out for the training gene set. Through least absolute shrinkage and selection operator regression analysis, the support vector machine recursive feature elimination algorithm, and gene expression screening, the range of DEGs was further reduced. Immune infiltration analysis was carried out, and the prediction results of the combined biomarker logistic regression model were verified with GSE55457. Results In total, 84 DEGs were identified through differential gene expression analysis. The five biomarkers that were screened further showed significant differences in cartilage, subchondral bone, and synovial tissue. The diagnostic accuracy of the model synthesized using five biomarkers through logistic regression was better than that of a single biomarker and significantly better than that of a single clinical trait. Conclusions CX3CR1, SLC7A5, ARL4C, TLR7, and MTHFD2 might be used as novel biomarkers to improve the accuracy of KOA disease diagnosis, monitor disease progression, and improve the efficacy of clinical treatment.
Collapse
|
15
|
Moore L, Pan Z, Brotto M. RNAseq of Osteoarthritic Synovial Tissues: Systematic Literary Review. FRONTIERS IN AGING 2022; 3:836791. [PMID: 35821799 PMCID: PMC9261452 DOI: 10.3389/fragi.2022.836791] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Accepted: 05/10/2022] [Indexed: 11/13/2022]
Abstract
Osteoarthritis (OA) is one of the most common causes of disability in aged people, and it is defined as a degenerative arthropathy, characterized by the disruption in joint tissue. The synovium plays a vital role in maintaining the health of the joint by supplying the nutrients to the surrounding tissues and the lubrication for joint movement. While it is well known that all the joint tissues are communicating and working together to provide a functioning joint, most studies on OA have been focused on bone and cartilage but much less about synovium have been reported. The purpose of this review was to investigate the current literature focused on RNA sequencing (RNAseq) of osteoarthritic synovial tissues to further understand the dynamic transcriptome changes occurring in this pivotal joint tissue. A total of 3 electronic databases (PubMed, CINHAL Complete, and Academic Complete) were systematically searched following PRISMA guidelines. The following criteria was used for inclusion: English language, free full text, between the period 2011–2022, size of sample (n > 10), study design being either retrospective or prospective, and RNAseq data of synovial tissue from OA subjects. From the initial search, 174 articles, 5 met all of our criteria and were selected for this review. The RNAseq analysis revealed several differentially expressed genes (DEGs) in synovial tissue. These genes are related to the inflammatory pathway and regulation of the extracellular matrix. The MMP family, particularly MMP13 was identified by three of the studies, indicating its important role in OA. IL6, a key contributor in the inflammation pathway, was also identified in 3 studies. There was a total of 8 DEGs, MMP13, MMP1, MMP2, APOD, IL6, TNFAIP6, FCER1G, and IGF1 that overlapped in 4 out of the 5 studies. One study focused on microbial RNA in the synovial tissue found that the microbes were differentially expressed in OA subjects too. These differentially expressed microbes have also been linked to the inflammatory pathway. Further investigation with more clinical gene profiling in synovial tissue of OA subjects is required to reveal the causation and progression, as well as aid in the development of new treatments.
Collapse
|
16
|
Yu L, Zhang X, Liu X, Li G, Chen M, Liu Z, Liu Q. CircTMOD3 promotes lipopolysaccharide-induced chondrocyte apoptosis in osteoarthritis by sponging miR-27a. J Bone Miner Metab 2022; 40:415-421. [PMID: 35103839 DOI: 10.1007/s00774-022-01310-0] [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: 08/21/2021] [Accepted: 01/06/2022] [Indexed: 10/19/2022]
Abstract
INTRODUCTION The progression of osteoarthritis (OA) requires the involvement of lipopolysaccharide (LPS)-induced inflammation, in which circTMOD3 plays an important role. We predicted that circTMOD3 could interact with miR-27a to inhibit LPS-induced chondrocyte apoptosis and explored the interaction between circTMOD3 and miR-27a in OA. MATERIALS AND METHODS Total RNAs were isolated from cartilage tissue samples from both OA patients (n = 62) and controls (n = 62) and subjected to RT-qPCRs to determine circTMOD3 and miR-27a (mature and premature) expression. Subcellular location of circTMOD3 and its interaction with premature miR-27a were analyzed using subcellular fractionation assay and RNA-RNA pulldown assay, respectively. CircTMOD3 was overexpressed in chondrocytes to study its role in miR-27a maturation. The roles of circTMOD3 and miR-27a in LPS-induced chondrocyte apoptosis were analyzed using cell apoptosis assay. RESULTS CircTMOD3 and premature miR-27a levels were increased while mature miR-27a level was decreased in OA. CircTMOD3 was located in both nuclear and cytoplasm fractions of chondrocytes. CircTMOD3 directly interacted with premature miR-27a and promoted LPS-induced chondrocyte apoptosis, while miR-27a inhibited LPS-induced chondrocyte apoptosis. Moreover, circTMOD3 overexpression suppressed miR-27a maturation and reduced the inhibitory effects of miR-27a on LPS-induced chondrocyte apoptosis. CONCLUSION CircTMOD3 suppresses miR-27a maturation in OA to promote chondrocyte apoptosis induced by LPS.
Collapse
Affiliation(s)
- Lu Yu
- Department of Orthopedics, Hebei Yanda Hospital, Yanjiao Economic and Technological Development Zone, No. 6, Sipulan Road, Sanhe City, Hebei Province, 065201, People's Republic of China
| | - Xiaogang Zhang
- Department of Orthopedics, Hebei Yanda Hospital, Yanjiao Economic and Technological Development Zone, No. 6, Sipulan Road, Sanhe City, Hebei Province, 065201, People's Republic of China
| | - Xingchao Liu
- Department of Orthopedics, Hebei Yanda Hospital, Yanjiao Economic and Technological Development Zone, No. 6, Sipulan Road, Sanhe City, Hebei Province, 065201, People's Republic of China
| | - Gang Li
- Department of Orthopedics, Hebei Yanda Hospital, Yanjiao Economic and Technological Development Zone, No. 6, Sipulan Road, Sanhe City, Hebei Province, 065201, People's Republic of China
| | - Mingliang Chen
- Department of Orthopedics, Hebei Yanda Hospital, Yanjiao Economic and Technological Development Zone, No. 6, Sipulan Road, Sanhe City, Hebei Province, 065201, People's Republic of China
| | - Zexin Liu
- Department of Orthopedics, Hebei Yanda Hospital, Yanjiao Economic and Technological Development Zone, No. 6, Sipulan Road, Sanhe City, Hebei Province, 065201, People's Republic of China
| | - Qinghe Liu
- Department of Orthopedics, Beijing Chaoyang Hospital, Capital Medical University, No. 8, Gongti Nan Road, Beijing, 100020, People's Republic of China.
| |
Collapse
|
17
|
Wang G, Li C, Zhang X, Tang L, Li Y. Long non-coding PRNCR1 regulates the proliferation and apoptosis of synoviocytes in osteoarthritis by sponging miR-377-3p. J Orthop Surg Res 2022; 17:238. [PMID: 35422021 PMCID: PMC9008967 DOI: 10.1186/s13018-022-03035-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Accepted: 02/23/2022] [Indexed: 11/21/2022] Open
Abstract
Background LncRNA PRNCR1 has been reported to be involved in LPS-induced inflammation, which contributes to osteoarthritis (OA). We predicted that miR-377-3p could bind to PRNCR1.MiR-377-3p can suppress OA development. We therefore analyzed the potential interaction between them in OA. Methods Expression of miR-377-3p and PRNCR1 in both OA (n = 40) and control (n = 40) samples were analyzed by RT-qPCR. MiR-377-3p or PRNCR1 were overexpressed in synoviocytes to explore their potential interaction. The subcellular location of PRNCR1 was analyzed by nuclear fractionation assay. The direct interaction between miR-377-3p and PRNCR1 was analyzed by RNA-pull down assay. The proliferation and apoptosis of synoviocytes were analyzed by BrdU and apoptosis assay, respectively. Results PRNCR1 was overexpressed in OA, while miR-377-3p was downexpressed in OA. PRNCR1 was detected in the cytoplasm and directly interacted with miR-377-3p. Interestingly, overexpression of PRNCR1 and miR-377-3p showed no regulatory role in each other’s expression. LPS treatment increased PRNCR1 expression and decreased miR-377-3p expression. PRNCR1 overexpression decreased LPS-induced synoviocyte proliferation and increased LPS-induced synoviocyte apoptosis. MiR-377-3p played opposite roles in cell proliferation and apoptosis. Moreover, PRNCR1 suppressed the role of miR-377-3p. Conclusions Therefore, PRNCR1 is was detected in cytoplasm and regulates synoviocyte proliferation and apoptosis in OA by sponging miR-377-3p. Supplementary Information The online version contains supplementary material available at 10.1186/s13018-022-03035-2.
Collapse
|
18
|
Engineering Closed-Loop, Autoregulatory Gene Circuits for Osteoarthritis Cell-Based Therapies. Curr Rheumatol Rep 2022; 24:96-110. [PMID: 35404006 DOI: 10.1007/s11926-022-01061-x] [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: 01/12/2022] [Indexed: 11/03/2022]
Abstract
PURPOSE OF REVIEW Genetic engineering offers the possibility to simultaneously target multiple cellular pathways in the joints affected by osteoarthritis (OA). The purpose of this review is to summarize the ongoing efforts to develop disease-modifying osteoarthritis drugs (DMOADs) using genetic engineering, including targeting approaches, genome editing techniques, and delivery methods. RECENT FINDINGS Several gene circuits have been developed that reprogram cells to autonomously target inflammation, and their efficacy has been demonstrated in chondrocytes and stem cells. Gene circuits developed for metabolic disorders, such as those targeting insulin resistance and obesity, also have the potential to mitigate the impact of these conditions on OA onset and/or progression. Despite the strides made in characterizing the inflammatory environment of the OA joint, our incomplete understanding of how the multiple regulators interact to control signal transduction, gene transcription, and translation to protein limits the development of targeted disease-modifying therapeutics. Continuous advances in targeted genome editing, combined with online toolkits that simplify the design and production of gene circuits, have the potential to accelerate the discovery and clinical application of multi-target gene circuits with disease-modifying properties for the treatment of OA.
Collapse
|
19
|
Rios‐Arce ND, Hum NR, Loots GG. Interactions between diabetes mellitus and osteoarthritis; from animal studies to clinical data. JBMR Plus 2022; 6:e10626. [PMID: 35509632 PMCID: PMC9059469 DOI: 10.1002/jbm4.10626] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Revised: 03/23/2022] [Accepted: 03/30/2022] [Indexed: 01/15/2023] Open
Abstract
Diabetes mellitus (DM) and osteoarthritis (OA) are commonly known metabolic diseases that affect a large segment of the world population. These two conditions share several risk factors such as obesity and aging; however, there is still no consensus regarding the direct role of DM on OA development and progression. Interestingly, both animal and human studies have yielded conflicting results, with some showing a significant role for DM in promoting OA, while others found no significant interactions between these conditions. In this review, we will discuss preclinical and clinical data that assessed the interaction between DM and OA. We will also discuss possible mechanisms associated with the effect of high glucose on the articular cartilage and chondrocytes. An emerging theme dominates the breath of published work in this area: most of the studies discussed in this review do not take into consideration the role of other factors such as the type of diabetes, age, biological sex, type of animal model, body mass index, and the use of pain medications when analyzing and interpreting data. Therefore, future studies should be more rigorous when designing experiments looking at DM and its effects on OA and should carefully account for these confounding factors, so that better approaches can be developed for monitoring and treating patients at risk of OA and DM. © 2022 The Authors. JBMR Plus published by Wiley Periodicals LLC on behalf of American Society for Bone and Mineral Research.
Collapse
Affiliation(s)
- Naiomy D. Rios‐Arce
- Physical and Life Sciences Directorate, Lawrence Livermore National Laboratories Livermore CA USA
| | - Nicholas R. Hum
- Physical and Life Sciences Directorate, Lawrence Livermore National Laboratories Livermore CA USA
| | - Gabriela G. Loots
- Physical and Life Sciences Directorate, Lawrence Livermore National Laboratories Livermore CA USA
- Molecular and Cell Biology, School of Natural Sciences University of California Merced Merced CA USA
| |
Collapse
|
20
|
Rios‐Arce ND, Murugesh DK, Hum NR, Sebastian A, Jbeily EH, Christiansen BA, Loots GG. Pre‐existing Type 1 Diabetes Mellitus Blunts the Development of
Post‐Traumatic
Osteoarthritis. JBMR Plus 2022; 6:e10625. [PMID: 35509635 PMCID: PMC9059474 DOI: 10.1002/jbm4.10625] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Revised: 02/26/2022] [Accepted: 03/09/2022] [Indexed: 11/12/2022] Open
Affiliation(s)
- Naiomy D. Rios‐Arce
- Physical and Life Sciences Directorate, Lawrence Livermore National Laboratories Livermore CA USA
| | - Deepa K. Murugesh
- Physical and Life Sciences Directorate, Lawrence Livermore National Laboratories Livermore CA USA
| | - Nicholas R. Hum
- Physical and Life Sciences Directorate, Lawrence Livermore National Laboratories Livermore CA USA
| | - Aimy Sebastian
- Physical and Life Sciences Directorate, Lawrence Livermore National Laboratories Livermore CA USA
| | - Elias H. Jbeily
- Department of Orthopedic Surgery UC Davis Medical Center Sacramento CA USA
| | | | - Gabriela G. Loots
- Physical and Life Sciences Directorate, Lawrence Livermore National Laboratories Livermore CA USA
- Molecular and Cell Biology School of Natural Sciences, UC Merced Merced CA USA
| |
Collapse
|
21
|
Zaki S, Blaker CL, Little CB. OA foundations - experimental models of osteoarthritis. Osteoarthritis Cartilage 2022; 30:357-380. [PMID: 34536528 DOI: 10.1016/j.joca.2021.03.024] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Revised: 03/01/2021] [Accepted: 03/10/2021] [Indexed: 02/02/2023]
Abstract
Osteoarthritis (OA) is increasingly recognised as a disease of diverse phenotypes with variable clinical presentation, progression, and response to therapeutic intervention. This same diversity is readily apparent in the many animal models of OA. However, model selection, study design, and interpretation of resultant findings, are not routinely done in the context of the target human (or veterinary) patient OA sub-population or phenotype. This review discusses the selection and use of animal models of OA in discovery and therapeutic-development research. Beyond evaluation of the different animal models on offer, this review suggests focussing the approach to OA-animal model selection on study objective(s), alignment of available models with OA-patient sub-types, and the resources available to achieve valid and translatable results. How this approach impacts model selection is discussed and an experimental design checklist for selecting the optimal model(s) is proposed. This approach should act as a guide to new researchers and a reminder to those already in the field, as to issues that need to be considered before embarking on in vivo pre-clinical research. The ultimate purpose of using an OA animal model is to provide the best possible evidence if, how, when and where a molecule, pathway, cell or process is important in clinical disease. By definition this requires both model and study outcomes to align with and be predictive of outcomes in patients. Keeping this at the forefront of research using pre-clinical OA models, will go a long way to improving the quality of evidence and its translational value.
Collapse
Affiliation(s)
- S Zaki
- Sydney School of Veterinary Science, Faculty of Science, University of Sydney, Australia; Raymond Purves Bone and Joint Research Laboratory, Australia.
| | - C L Blaker
- Raymond Purves Bone and Joint Research Laboratory, Australia; Murray Maxwell Biomechanics Laboratory, The Kolling Institute, University of Sydney Faculty of Medicine and Health, At Royal North Shore Hospital, Australia.
| | - C B Little
- Raymond Purves Bone and Joint Research Laboratory, Australia.
| |
Collapse
|
22
|
Contino KF, Yadav H, Shiozawa Y. The gut microbiota can be a potential regulator and treatment target of bone metastasis. Biochem Pharmacol 2022; 197:114916. [PMID: 35041811 PMCID: PMC8858876 DOI: 10.1016/j.bcp.2022.114916] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Revised: 01/10/2022] [Accepted: 01/10/2022] [Indexed: 02/08/2023]
Abstract
The gut microbiota, an often forgotten organ, have a tremendous impact on human health. It has long been known that the gut microbiota are implicated in cancer development, and more recently, the gut microbiota have been shown to influence cancer metastasis to distant organs. Although one of the most common sites of distant metastasis is the bone, and the skeletal system has been shown to be a subject of interactions with the gut microbiota to regulate bone homeostasis, little research has been done regarding how the gut microbiota control the development of bone metastasis. This review will discuss the mechanisms through which the gut microbiota and derived microbial compounds (i) regulate gastrointestinal cancer disease progression and metastasis, (ii) influence skeletal remodeling and potentially modulate bone metastasis, and (iii) affect and potentially enhance immunotherapeutic treatments for bone metastasis.
Collapse
Affiliation(s)
- Kelly F Contino
- Department of Cancer Biology and Comprehensive Cancer Center, Wake Forest University Health Sciences, Winston-Salem, NC 27157, USA
| | - Hariom Yadav
- Department of Neurosurgery and Brain Repair and Institute for Microbiome, University of South Florida, Tampa, FL 33612, USA
| | - Yusuke Shiozawa
- Department of Cancer Biology and Comprehensive Cancer Center, Wake Forest University Health Sciences, Winston-Salem, NC 27157, USA.
| |
Collapse
|
23
|
Zhang Q, Bai X, Wang R, Zhao H, Wang L, Liu J, Li M, Chen Z, Wang Z, Li L, Wang D. 4‐octyl Itaconate inhibits lipopolysaccharide (LPS)‐induced osteoarthritis via activating Nrf2 signalling pathway. J Cell Mol Med 2022; 26:1515-1529. [PMID: 35068055 PMCID: PMC8899168 DOI: 10.1111/jcmm.17185] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2021] [Revised: 12/18/2021] [Accepted: 01/03/2022] [Indexed: 12/23/2022] Open
Abstract
Small molecule drug intervention for chondrocytes is a valuable method for the treatment of osteoarthritis (OA). The 4‐octyl itaconate (OI) is a cellular derivative of itaconate with sound cell permeability and transformation rate. We attempted to confirm the protective role of OI in chondrocytes and its regulatory mechanism. We used lipopolysaccharide (LPS) to induce chondrocyte inflammation injury. After the OI treatment, the secretion and mRNA expression of Il‐6, Il‐10, Mcp‐1 and Tnf‐α were detected by ELISA and qPCR. The protective effect of OI on articular cartilage was further verified in surgical destabilization of the medial meniscus model of OA. Cell death and apoptosis were evaluated based on CCK8, LDH, Typan blue staining, Annexin V and TUNEL analyses. The small interfering RNAs were used to knockout the Nrf2 gene of chondrocytes to verify the OI‐mediated Nrf2 signalling pathway. The results revealed that OI protects cells from LPS‐induced inflammatory injury and attenuates cell death and apoptosis induced by LPS. Similar protective effects were also observed on articular cartilage in mice. The OI activated Nrf2 signalling pathway and promoted the stable expression and translocation of Nrf2 into the nucleus. When the Nrf2 signalling pathway was blocked, the protective effect of OI was significantly counteracted in chondrocytes and a mouse arthritis model. Both itaconate and its derivative (i.e., OI) showed important medical effects in the treatment of OA.
Collapse
Affiliation(s)
- Qingchen Zhang
- Department of Orthopaedics Shandong Provincial Hospital Cheeloo College of Medicine Shandong University Jinan China
- Department of Orthopaedics Shandong Provincial Hospital Affiliated to Shandong First Medical University Jinan China
| | - Xiaohui Bai
- Department of Clinical Laboratory Shandong Provincial Hospital, Cheeloo College of Medicine Shandong University Jinan China
| | - Rongrong Wang
- Department of Clinical Laboratory Shandong Provincial Hospital, Cheeloo College of Medicine Shandong University Jinan China
| | - Hao Zhao
- Department of Clinical Laboratory Shandong Provincial Hospital, Cheeloo College of Medicine Shandong University Jinan China
| | - Lili Wang
- Department of Clinical Laboratory Shandong Provincial Hospital, Cheeloo College of Medicine Shandong University Jinan China
| | - Jingwen Liu
- Department of Clinical Laboratory Shandong Provincial Hospital, Cheeloo College of Medicine Shandong University Jinan China
| | - Ming Li
- Department of Clinical Laboratory Shandong Provincial Hospital, Cheeloo College of Medicine Shandong University Jinan China
| | - Zheng Chen
- Department of Orthopaedics Shandong Provincial Hospital Cheeloo College of Medicine Shandong University Jinan China
- Department of Orthopaedics Shandong Provincial Hospital Affiliated to Shandong First Medical University Jinan China
| | - Zejun Wang
- Department of Clinical Laboratory Shandong Provincial Hospital, Cheeloo College of Medicine Shandong University Jinan China
| | - Lianxin Li
- Department of Orthopaedics Shandong Provincial Hospital Cheeloo College of Medicine Shandong University Jinan China
- Department of Orthopaedics Shandong Provincial Hospital Affiliated to Shandong First Medical University Jinan China
| | - Dawei Wang
- Department of Orthopaedics Shandong Provincial Hospital Cheeloo College of Medicine Shandong University Jinan China
- Department of Orthopaedics Shandong Provincial Hospital Affiliated to Shandong First Medical University Jinan China
| |
Collapse
|
24
|
Liu D, Liu W, Jiang L, Dong S, Ma W, Wang S, Wan C. Silencing of TLR7 protects against lipopolysaccharide-induced chondrocyte apoptosis and injury by blocking the p21-mediated JAK2/STAT3 pathway. Am J Transl Res 2021; 13:13555-13566. [PMID: 35035696 PMCID: PMC8748165] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Accepted: 08/06/2021] [Indexed: 06/14/2023]
Abstract
Involvement of toll-like receptor 7 (TLR7) in the immune response has been reported in diverse inflammatory diseases. However, the role of TLR7 in the pathogenesis of osteoarthritis (OA) is poorly understood. In this study, we sought to investigate the contribution of TLR7 in regulating chondrocyte apoptosis, inflammation, and degradation of the extracellula matrix (ECM), and its underlying mechanisms. We found that TLR7 expression was increased in cartilage tissues of OA patients and in lipopolysaccharide (LPS)-induced chondrocytes. Silencing of TLR7 alleviated LPS-induced chondrocyte apoptosis, inflammation, and ECM degradation. Mechanistically, TLR7 silencing inhibited the JAK2/STAT3 signaling pathway by inducing p21 expression. Moreover, p21 knockdown and colivein (an activator of JAK2/STAT3 signaling) partially rescued the suppressive role of TLR7 silencing on chondrocyte apoptosis, the inflammatory response, and ECM underproduction. Taken together, our data revealed that knockdown of TLR7 attenuated chondrocyte apoptosis and injury by blocking the p21-mediated JAK2/STAT3 pathway, suggesting that TLR7 may be a therapeutic target in OA.
Collapse
Affiliation(s)
- Dan Liu
- Department of The Joint and Bone Surgery, Yantaishan HospitalYantai 264001, Shandong, China
| | - Wei Liu
- Department of Pathophysiology, Binzhou Medical UniversityBinzhou 256603, Shandong, China
| | - Limin Jiang
- Department of The Joint and Bone Surgery, Yantaishan HospitalYantai 264001, Shandong, China
| | - Shengjie Dong
- Department of The Joint and Bone Surgery, Yantaishan HospitalYantai 264001, Shandong, China
| | - Weihua Ma
- Department of The Joint and Bone Surgery, Yantaishan HospitalYantai 264001, Shandong, China
| | - Shijun Wang
- Department of The Joint and Bone Surgery, Yantaishan HospitalYantai 264001, Shandong, China
| | - Chao Wan
- Department of The Joint and Bone Surgery, Yantaishan HospitalYantai 264001, Shandong, China
| |
Collapse
|
25
|
Rzeczycki P, Rasner C, Lammlin L, Junginger L, Goldman S, Bergman R, Redding S, Knights AJ, Elliott M, Maerz T. Cannabinoid receptor type 2 is upregulated in synovium following joint injury and mediates anti-inflammatory effects in synovial fibroblasts and macrophages. Osteoarthritis Cartilage 2021; 29:1720-1731. [PMID: 34537380 PMCID: PMC8883578 DOI: 10.1016/j.joca.2021.09.003] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Revised: 08/09/2021] [Accepted: 09/06/2021] [Indexed: 02/02/2023]
Abstract
OBJECTIVE Joint injury-induced perturbations to the endocannabinoid system (ECS), a regulator of both inflammation and nociception, remain largely uncharacterized. We employed a mouse model of ACL rupture to assess alterations to nociception, inflammation, and the ECS while using in vitro models to determine whether CB2 agonism can mitigate inflammatory signaling in macrophages and fibroblast-like synoviocytes (FLS). DESIGN Mice underwent noninvasive ACL rupture (ACLR) via tibial compression-based loading. Nociception was measured longitudinally using mechanical allodynia and knee hyperalgesia testing. Synovitis was assessed using histological scoring and histomorphometry. Gene and protein markers of inflammation were characterized in whole joints and synovium. Immunohistochemistry assessed injury-induced alterations to CB1+, CB2+, and F4/80+ cells in synovium. To assess whether CB2 agonism can inhibit pro-inflammatory macrophage polarization, murine bone marrow-derived macrophages (mBMDM) were stimulated with IL-1β or conditioned medium from IL-1β-treated FLS and treated with vehicle (DMSO), the CB2 agonist HU308, or cannabidiol (CBD). Macrophage polarization was assessed as the ratio of M1-associated (IL1b, MMP1b, and IL6) to M2-associated (IL10, IL4, and CD206) gene expression. Human FLS (hFLS) isolated from synovial tissue of OA patients were treated with vehicle (DMSO) or HU308 following TNF-α or IL-1β stimulation to assess inhibition of catabolic/inflammatory gene expression. RESULTS ACLR induces synovitis, progressively-worsening PTOA severity, and an immediate and sustained increase in both mechanical allodynia and knee hyperalgesia, which persist beyond the resolution of molecular inflammation. Enrichment of CB2, but not CB1, was observed in ACLR synovium at 3d, 14d, and 28d, and CB2 was found to be associated with F4/80 (+) cells, which are increased in number in ACLR synovium at all time points. The CB2 agonist HU308 strongly inhibited mBMDM M1-type polarization following stimulation with either IL-1β or conditioned medium from IL-1β-treated mFLS, which was characterized by reductions in Il1b, Mmp1b, and Il6 and increases in Cd206 gene expression. Cannabidiol similarly inhibited IL-1β-induced mBMDM M1 polarization via a reduction in Il1b and an increase in Cd206 and Il4 gene expression. Lastly, in OA hFLS, HU308 treatment inhibited IL-1β-induced CCL2, MMP1, MMP3, and IL6 expression and further inhibited TNF-α-induced CCL2, MMP1, and GMCSF expression, demonstrating human OA-relevant anti-inflammatory effects by targeting CB2. CONCLUSIONS Joint injury perturbs the intra-articular ECS, characterized by an increase in synovial F4/80(+) cells, which express CB2, but not CB1. Targeting CB2 in murine macrophages and human FLS induced potent anti-inflammatory and anti-catabolic effects, which indicates that the CB2 receptor plays a key role in regulating inflammatory signaling in the two primary effector cells in the synovium. The intraarticular ECS is therefore a potential therapeutic target for blocking pathological inflammation in future disease-modifying PTOA treatments.
Collapse
Affiliation(s)
- P Rzeczycki
- Orthopaedic Research Laboratories, Department of Orthopaedic Surgery, University of Michigan, Ann Arbor, MI, USA
| | - C Rasner
- Orthopaedic Research Laboratories, Department of Orthopaedic Surgery, University of Michigan, Ann Arbor, MI, USA
| | - L Lammlin
- Orthopaedic Research Laboratories, Department of Orthopaedic Surgery, University of Michigan, Ann Arbor, MI, USA
| | - L Junginger
- Orthopaedic Research Laboratories, Department of Orthopaedic Surgery, University of Michigan, Ann Arbor, MI, USA
| | - S Goldman
- Orthopaedic Research Laboratories, Department of Orthopaedic Surgery, University of Michigan, Ann Arbor, MI, USA
| | - R Bergman
- Orthopaedic Research Laboratories, Department of Orthopaedic Surgery, University of Michigan, Ann Arbor, MI, USA
| | - S Redding
- Orthopaedic Research Laboratories, Department of Orthopaedic Surgery, University of Michigan, Ann Arbor, MI, USA
| | - A J Knights
- Orthopaedic Research Laboratories, Department of Orthopaedic Surgery, University of Michigan, Ann Arbor, MI, USA
| | - M Elliott
- Orthopaedic Research Laboratories, Department of Orthopaedic Surgery, University of Michigan, Ann Arbor, MI, USA
| | - T Maerz
- Orthopaedic Research Laboratories, Department of Orthopaedic Surgery, University of Michigan, Ann Arbor, MI, USA.
| |
Collapse
|
26
|
Brochard S, Pontin J, Bernay B, Boumediene K, Conrozier T, Baugé C. The benefit of combining curcumin, bromelain and harpagophytum to reduce inflammation in osteoarthritic synovial cells. BMC Complement Med Ther 2021; 21:261. [PMID: 34649531 PMCID: PMC8515758 DOI: 10.1186/s12906-021-03435-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Accepted: 09/27/2021] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Osteoarthritis (OA) is the most common form of arthritis, affecting millions of people worldwide and characterised by joint pain and inflammation. It is a complex disease involving inflammatory factors and affecting the whole joint, including the synovial membrane. Since drug combination is widely used to treat chronic inflammatory diseases, a similar strategy of designing plant-derived natural products to reduce inflammation in OA joints may be of interest. In this study, we characterised the response of OA synovial cells to lipopolysaccharide (LPS) and investigated the biological action of the combination of curcumin, bromelain and harpagophytum in this original in vitro model of osteoarthritis. METHODS Firstly, human synovial cells from OA patients were stimulated with LPS and proteomic analysis was performed. Bioinformatics analyses were performed using Cytoscape App and SkeletalVis databases. Additionally, cells were treated with curcumin, bromelain and harpagophytum alone or with the three vegetal compounds together. The gene expression involved in inflammation, pain or catabolism was determined by RT-PCR. The release of the encoded proteins by these genes and of prostaglandin E2 (PGE2) were also assayed by ELISA. RESULTS Proteomic analysis demonstrated that LPS induces the expression of numerous proteins involved in the OA process in human OA synovial cells. In particular, it stimulates inflammation through the production of pro-inflammatory cytokines (Interleukin-6, IL-6), catabolism through an increase of metalloproteases (MMP-1, MMP-3, MMP-13), and the production of pain-mediating neurotrophins (Nerve Growth Factor, NGF). These increases were observed in terms of mRNA levels and protein release. LPS also increases the amount of PGE2, another inflammation and pain mediator. At the doses tested, vegetal extracts had little effect: only curcumin slightly counteracted the effects of LPS on NGF and MMP-13 mRNA, and PGE2, IL-6 and MMP-13 release. In contrast, the combination of curcumin with bromelain and harpagophytum reversed lots of effects of LPS in human OA synovial cells. It significantly reduced the gene expression and/or the release of proteins involved in catabolism (MMP-3 and -13), inflammation (IL-6) and pain (PGE2 and NGF). CONCLUSION We have shown that the stimulation of human OA synovial cells with LPS can induce protein changes similar to inflamed OA synovial tissues. In addition, using this model, we demonstrated that the combination of three vegetal compounds, namely curcumin, bromelain and harpagophytum, have anti-inflammatory and anti-catabolic effects in synovial cells and may thus reduce OA progression and related pain.
Collapse
Affiliation(s)
- Sybille Brochard
- EA7451 BioConnect, Université de Caen Normandie, UNICAEN, 14032, Caen, France
| | - Julien Pontin
- Proteogen platform, Normandie Univ, UNICAEN, Caen, France
| | - Benoit Bernay
- Proteogen platform, Normandie Univ, UNICAEN, Caen, France
| | - Karim Boumediene
- EA7451 BioConnect, Université de Caen Normandie, UNICAEN, 14032, Caen, France
| | - Thierry Conrozier
- Rheumatology Department, Nord Franche-Comté Hospital, Trevenans, France
| | - Catherine Baugé
- EA7451 BioConnect, Université de Caen Normandie, UNICAEN, 14032, Caen, France.
| |
Collapse
|
27
|
Lu J, Guan H, Wu D, Hu Z, Zhang H, Jiang H, Yu J, Zeng K, Li H, Zhang H, Pan C, Cai D, Yu X. Pseudolaric acid B ameliorates synovial inflammation and vessel formation by stabilizing PPARγ to inhibit NF-κB signalling pathway. J Cell Mol Med 2021; 25:6664-6678. [PMID: 34117708 PMCID: PMC8278075 DOI: 10.1111/jcmm.16670] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Revised: 04/10/2021] [Accepted: 05/08/2021] [Indexed: 12/16/2022] Open
Abstract
Synovial macrophage polarization and inflammation are essential for osteoarthritis (OA) development, yet the molecular mechanisms and regulation responsible for the pathogenesis are still poorly understood. Here, we report that pseudolaric acid B (PAB) attenuated articular cartilage degeneration and synovitis during OA. PAB, a diterpene acid, specifically inhibited NF-κB signalling and reduced the production of pro-inflammatory cytokines, which further decreased M1 polarization and vessel formation. We further provide in vivo and in vitro evidences that PAB suppressed NF-κB signalling by stabilizing PPARγ. Using PPARγ antagonist could abolish anti-inflammatory effect of PAB and rescue the activation of NF-κB signalling during OA. Our findings identify a previously unrecognized role of PAB in the regulation of OA and provide mechanisms by which PAB regulates NF-κB signalling through PPARγ, which further suggest targeting synovial inflammation or inhibiting vessel formation at early stage could be an effective preventive strategy for OA.
Collapse
Affiliation(s)
- Jiansen Lu
- Department of Immunology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China.,Department of Orthopedics, Orthopedic Hospital of Guangdong Province, Academy of Orthopedics·Guangdong Province, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China.,Department of Joint Surgery, the Fifth Affiliated Hospital of Southern Medical University Guangdong Province, Guangzhou, China
| | - Hong Guan
- Department of Orthopedics, Orthopedic Hospital of Guangdong Province, Academy of Orthopedics·Guangdong Province, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China.,The Third School of Clinical Medicine, Southern Medical University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, Guangzhou, China.,Department of Joint Surgery, Center for Orthopaedic Surgery, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China
| | - Dan Wu
- Department of Immunology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Zhiqiang Hu
- Department of Immunology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Hongbo Zhang
- Department of Orthopedics, Orthopedic Hospital of Guangdong Province, Academy of Orthopedics·Guangdong Province, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China.,The Third School of Clinical Medicine, Southern Medical University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, Guangzhou, China.,Department of Joint Surgery, Center for Orthopaedic Surgery, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China
| | - Huaji Jiang
- Department of Immunology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Jingyao Yu
- School of Basic Medical Sciences, Gannan Medical University, Ganzhou, China
| | - Ke Zeng
- Department of Immunology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Hongyu Li
- Department of Immunology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Haiyan Zhang
- Department of Orthopedics, Orthopedic Hospital of Guangdong Province, Academy of Orthopedics·Guangdong Province, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China.,The Third School of Clinical Medicine, Southern Medical University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, Guangzhou, China.,Department of Joint Surgery, Center for Orthopaedic Surgery, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China
| | - Chenglong Pan
- Department of Joint Surgery, the Fifth Affiliated Hospital of Southern Medical University Guangdong Province, Guangzhou, China
| | - Daozhang Cai
- Department of Orthopedics, Orthopedic Hospital of Guangdong Province, Academy of Orthopedics·Guangdong Province, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China.,The Third School of Clinical Medicine, Southern Medical University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, Guangzhou, China.,Department of Joint Surgery, Center for Orthopaedic Surgery, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China
| | - Xiao Yu
- Department of Immunology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China.,Department of Joint Surgery, the Fifth Affiliated Hospital of Southern Medical University Guangdong Province, Guangzhou, China.,Guangdong Provincial Key Lab of Single Cell Technology and Application, Southern Medical University, Guangzhou, China
| |
Collapse
|
28
|
Single-Cell RNA-Seq Reveals Transcriptomic Heterogeneity and Post-Traumatic Osteoarthritis-Associated Early Molecular Changes in Mouse Articular Chondrocytes. Cells 2021; 10:cells10061462. [PMID: 34200880 PMCID: PMC8230441 DOI: 10.3390/cells10061462] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2021] [Revised: 06/02/2021] [Accepted: 06/07/2021] [Indexed: 12/25/2022] Open
Abstract
Articular cartilage is a connective tissue lining the surfaces of synovial joints. When the cartilage severely wears down, it leads to osteoarthritis (OA), a debilitating disease that affects millions of people globally. The articular cartilage is composed of a dense extracellular matrix (ECM) with a sparse distribution of chondrocytes with varying morphology and potentially different functions. Elucidating the molecular and functional profiles of various chondrocyte subtypes and understanding the interplay between these chondrocyte subtypes and other cell types in the joint will greatly expand our understanding of joint biology and OA pathology. Although recent advances in high-throughput OMICS technologies have enabled molecular-level characterization of tissues and organs at an unprecedented resolution, thorough molecular profiling of articular chondrocytes has not yet been undertaken, which may be in part due to the technical difficulties in isolating chondrocytes from dense cartilage ECM. In this study, we profiled articular cartilage from healthy and injured mouse knee joints at a single-cell resolution and identified nine chondrocyte subtypes with distinct molecular profiles and injury-induced early molecular changes in these chondrocytes. We also compared mouse chondrocyte subpopulations to human chondrocytes and evaluated the extent of molecular similarity between mice and humans. This work expands our view of chondrocyte heterogeneity and rapid molecular changes in chondrocyte populations in response to joint trauma and highlights potential mechanisms that trigger cartilage degeneration.
Collapse
|
29
|
Groen WMGAC, Utomo L, Castilho M, Gawlitta D, Malda J, van Weeren PR, Levato R, Korthagen NM. Impact of Endotoxins in Gelatine Hydrogels on Chondrogenic Differentiation and Inflammatory Cytokine Secretion In Vitro. Int J Mol Sci 2020; 21:E8571. [PMID: 33202964 PMCID: PMC7696312 DOI: 10.3390/ijms21228571] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Revised: 11/02/2020] [Accepted: 11/11/2020] [Indexed: 12/31/2022] Open
Abstract
Gelatine methacryloyl (GelMA) hydrogels are widely used in studies aimed at cartilage regeneration. However, the endotoxin content of commercially available GelMAs and gelatines used in these studies is often overlooked, even though endotoxins may influence several cellular functions. Moreover, regulations for clinical use of biomaterials dictate a stringent endotoxin limit. We determined the endotoxin level of five different GelMAs and evaluated the effect on the chondrogenic differentiation of equine mesenchymal stromal cells (MSCs). Cartilage-like matrix production was evaluated by biochemical assays and immunohistochemistry. Furthermore, equine peripheral blood mononuclear cells (PBMCs) were cultured on the hydrogels for 24 h, followed by the assessment of tumour necrosis factor (TNF)-α and C-C motif chemokine ligand (CCL)2 as inflammatory markers. The GelMAs were found to have widely varying endotoxin content (two with >1000 EU/mL and three with <10 EU/mL), however, this was not a critical factor determining in vitro cartilage-like matrix production of embedded MSCs. PBMCs did produce significantly higher TNF-α and CCL2 in response to the GelMA with the highest endotoxin level compared to the other GelMAs. Although limited effects on chondrogenic differentiation were found in this study, caution with the use of commercial hydrogels is warranted in the translation from in vitro to in vivo studies because of regulatory constraints and potential inflammatory effects of the content of these hydrogels.
Collapse
Affiliation(s)
- Wilhelmina M. G. A. C. Groen
- Department of Clinical Sciences, Faculty of Veterinary Medicine, Utrecht University, 3584 CM Utrecht, The Netherlands; (J.M.); (R.L.); (N.M.K.)
- Department of Orthopaedics, Regenerative Medicine Center, University Medical Center Utrecht, P.O. Box 85500, 3508 GA Utrecht, The Netherlands;
| | - Lizette Utomo
- Department of Oral and Maxillofacial Surgery and Special Dental Care, Regenerative Medicine Center, University Medical Center Utrecht, P.O. Box 85500, 3508 GA Utrecht, The Netherlands; (L.U.); (D.G.)
| | - Miguel Castilho
- Department of Orthopaedics, Regenerative Medicine Center, University Medical Center Utrecht, P.O. Box 85500, 3508 GA Utrecht, The Netherlands;
| | - Debby Gawlitta
- Department of Oral and Maxillofacial Surgery and Special Dental Care, Regenerative Medicine Center, University Medical Center Utrecht, P.O. Box 85500, 3508 GA Utrecht, The Netherlands; (L.U.); (D.G.)
| | - Jos Malda
- Department of Clinical Sciences, Faculty of Veterinary Medicine, Utrecht University, 3584 CM Utrecht, The Netherlands; (J.M.); (R.L.); (N.M.K.)
- Department of Orthopaedics, Regenerative Medicine Center, University Medical Center Utrecht, P.O. Box 85500, 3508 GA Utrecht, The Netherlands;
| | - P. René van Weeren
- Department of Clinical Sciences, Faculty of Veterinary Medicine, Utrecht University, 3584 CM Utrecht, The Netherlands; (J.M.); (R.L.); (N.M.K.)
| | - Riccardo Levato
- Department of Clinical Sciences, Faculty of Veterinary Medicine, Utrecht University, 3584 CM Utrecht, The Netherlands; (J.M.); (R.L.); (N.M.K.)
- Department of Orthopaedics, Regenerative Medicine Center, University Medical Center Utrecht, P.O. Box 85500, 3508 GA Utrecht, The Netherlands;
| | - Nicoline M. Korthagen
- Department of Clinical Sciences, Faculty of Veterinary Medicine, Utrecht University, 3584 CM Utrecht, The Netherlands; (J.M.); (R.L.); (N.M.K.)
- Department of Orthopaedics, Regenerative Medicine Center, University Medical Center Utrecht, P.O. Box 85500, 3508 GA Utrecht, The Netherlands;
| |
Collapse
|
30
|
Mendez ME, Murugesh DK, Sebastian A, Hum NR, McCloy SA, Kuhn EA, Christiansen BA, Loots GG. Antibiotic Treatment Prior to Injury Improves Post-Traumatic Osteoarthritis Outcomes in Mice. Int J Mol Sci 2020; 21:E6424. [PMID: 32899361 PMCID: PMC7503363 DOI: 10.3390/ijms21176424] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2020] [Revised: 08/28/2020] [Accepted: 08/29/2020] [Indexed: 12/21/2022] Open
Abstract
Osteoarthritis (OA) is a painful and debilitating disease characterized by the chronic and progressive degradation of articular cartilage. Post-traumatic OA (PTOA) is a secondary form of OA that develops in ~50% of cases of severe articular injury. Inflammation and re-occurring injury have been implicated as contributing to the progression of PTOA after the initial injury. However, there is very little known about external factors prior to injury that could affect the risk of PTOA development. To examine how the gut microbiome affects PTOA development we used a chronic antibiotic treatment regimen starting at weaning for six weeks prior to ACL rupture, in mice. A six-weeks post-injury histological examination showed more robust cartilage staining on the antibiotic (AB)-treated mice than the untreated controls (VEH), suggesting slower disease progression in AB cohorts. Injured joints also showed an increase in the presence of anti-inflammatory M2 macrophages in the AB group. Molecularly, the phenotype correlated with a significantly lower expression of inflammatory genes Tlr5, Ccl8, Cxcl13, and Foxo6 in the injured joints of AB-treated animals. Our results indicate that a reduced state of inflammation at the time of injury and a lower expression of Wnt signaling modulatory protein, Rspo1, caused by AB treatment can slow down or improve PTOA outcomes.
Collapse
Affiliation(s)
- Melanie E. Mendez
- Lawrence Livermore National Laboratories, Physical and Life Sciences Directorate, Livermore, CA 94550, USA; (M.E.M.); (D.K.M.); (A.S.); (N.R.H.); (S.A.M.); (E.A.K.)
| | - Deepa K. Murugesh
- Lawrence Livermore National Laboratories, Physical and Life Sciences Directorate, Livermore, CA 94550, USA; (M.E.M.); (D.K.M.); (A.S.); (N.R.H.); (S.A.M.); (E.A.K.)
| | - Aimy Sebastian
- Lawrence Livermore National Laboratories, Physical and Life Sciences Directorate, Livermore, CA 94550, USA; (M.E.M.); (D.K.M.); (A.S.); (N.R.H.); (S.A.M.); (E.A.K.)
| | - Nicholas R. Hum
- Lawrence Livermore National Laboratories, Physical and Life Sciences Directorate, Livermore, CA 94550, USA; (M.E.M.); (D.K.M.); (A.S.); (N.R.H.); (S.A.M.); (E.A.K.)
- UC Merced, School of Natural Sciences, Merced, CA 95343, USA
| | - Summer A. McCloy
- Lawrence Livermore National Laboratories, Physical and Life Sciences Directorate, Livermore, CA 94550, USA; (M.E.M.); (D.K.M.); (A.S.); (N.R.H.); (S.A.M.); (E.A.K.)
| | - Edward A. Kuhn
- Lawrence Livermore National Laboratories, Physical and Life Sciences Directorate, Livermore, CA 94550, USA; (M.E.M.); (D.K.M.); (A.S.); (N.R.H.); (S.A.M.); (E.A.K.)
| | | | - Gabriela G. Loots
- Lawrence Livermore National Laboratories, Physical and Life Sciences Directorate, Livermore, CA 94550, USA; (M.E.M.); (D.K.M.); (A.S.); (N.R.H.); (S.A.M.); (E.A.K.)
- UC Merced, School of Natural Sciences, Merced, CA 95343, USA
| |
Collapse
|